Next Article in Journal
Tacrolimus-Associated Tremor in Renal Transplant Patients: Potential Impact of the Galenic Formulation
Previous Article in Journal
Flavonoid-Based Combination Therapies and Nano-Formulations: An Emerging Frontier in Breast Cancer Treatment
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Mechanotransduction-Epigenetic Coupling in Pulmonary Regeneration: Multifunctional Bioscaffolds as Emerging Tools

1
Department of Physiology, School of Medicine, Southeast University, Nanjing 210009, China
2
Jiangsu Provincial Key Laboratory of Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, China
3
Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, China
*
Author to whom correspondence should be addressed.
Pharmaceuticals 2025, 18(10), 1487; https://doi.org/10.3390/ph18101487
Submission received: 2 September 2025 / Revised: 27 September 2025 / Accepted: 30 September 2025 / Published: 2 October 2025
(This article belongs to the Section Pharmacology)

Abstract

Pulmonary fibrosis (PF) is a progressive and fatal lung disease characterized by irreversible alveolar destruction and pathological extracellular matrix (ECM) deposition. Currently approved agents (pirfenidone and nintedanib) slow functional decline but do not reverse established fibrosis or restore functional alveoli. Multifunctional bioscaffolds present a promising therapeutic strategy through targeted modulation of critical cellular processes, including proliferation, migration, and differentiation. This review synthesizes recent advances in scaffold-based interventions for PF, with a focus on their dual mechano-epigenetic regulatory functions. We delineate how scaffold properties (elastic modulus, stiffness gradients, dynamic mechanical cues) direct cell fate decisions via mechanotransduction pathways, exemplified by focal adhesion–cytoskeleton coupling. Critically, we highlight how pathological mechanical inputs establish and perpetuate self-reinforcing epigenetic barriers to regeneration through aberrant chromatin states. Furthermore, we examine scaffolds as platforms for precision epigenetic drug delivery, particularly controlled release of inhibitors targeting DNA methyltransferases (DNMTi) and histone deacetylases (HDACi) to disrupt this mechano-reinforced barrier. Evidence from PF murine models and ex vivo lung slice cultures demonstrate scaffold-mediated remodeling of the fibrotic niche, with key studies reporting substantial reductions in collagen deposition and significant increases in alveolar epithelial cell markers following intervention. These quantitative outcomes highlight enhanced alveolar epithelial plasticity and upregulating antifibrotic gene networks. Emerging integration of stimuli-responsive biomaterials, CRISPR/dCas9-based epigenetic editors, and AI-driven design to enhance scaffold functionality is discussed. Collectively, multifunctional bioscaffolds hold significant potential for clinical translation by uniquely co-targeting mechanotransduction and epigenetic reprogramming. Future work will need to resolve persistent challenges, including the erasure of pathological mechanical memory and precise spatiotemporal control of epigenetic modifiers in vivo, to unlock their full therapeutic potential.

1. Introduction

Pulmonary fibrosis (PF) is a chronic progressive lung disease that leads to respiratory failure. Its pathogenesis involves irreversible alveolar destruction, impaired capillary barrier function, pathological extracellular matrix (ECM) accumulation, and progressive tissue stiffening [1,2]. Current therapies are palliative, alleviating symptoms without halting disease progression or reversing fibrosis. Approved drugs including pirfenidone and nintedanib only slow functional decline but fail to reverse established fibrosis or regenerate functional alveoli [3,4,5]. This therapeutic limitation arises from a self-sustaining fibrotic microenvironment, where biochemical abnormalities (e.g., persistent profibrotic cytokines) converge with aberrant biomechanics such as elevated matrix stiffness [6,7]. Innovative approaches capable of co-targeting these dual drivers are therefore urgently needed. Therefore, clarifying how these biochemical and biomechanical imbalances act on key reparative cells is essential for designing therapies that can reverse fibrosis.
Given the limitations of current therapies, a deeper understanding of the cellular and molecular mechanisms driving PF progression is crucial. Alveolar type II epithelial Cells (AT2 Cells) serve as the major progenitor population responsible for lung repair and regeneration. Following lung injury, AT2 Cells proliferate and differentiate into AT1 Cells to restore gas-exchange surfaces [8,9]. However, chronic exposure to profibrotic mediators disrupts this process. Transforming growth factor-β1 (TGF-β1) plays a central role by driving fibroblast-to-myofibroblast transdifferentiation via Smad2/3 phosphorylation and by promoting epithelial–mesenchymal transition (EMT) [10,11]. In addition, TGF-β1 engages secondary pathways such as Wnt/β-catenin and Notch, which further amplify fibrogenesis and reinforce pathological ECM deposition [12,13]. These signaling perturbations converge with epigenetic dysregulation, forming a multifaceted barrier to AT2 regeneration. In addition to these signaling abnormalities, epigenetic dysregulation has emerged as a pivotal driver of persistent AT2 dysfunction. DNA hypermethylation and histone deacetylation maintain fibroblast activation and compromise AT2 cell plasticity [14,15]. The reversible nature of these epigenetic modifications enables viable therapeutic targeting. Emerging evidence indicates that mechanical signals are transmitted from focal adhesions through actomyosin and the linker of nucleoskeleton and cytoskeleton (LINC) complex to the nucleus, where they reshape chromatin and thereby alter epigenetic states that affect AT2 plasticity. Moreover, the effects of epigenetic dysregulation are closely intertwined with mechanical abnormalities in the fibrotic niche. Critically, increased tissue stiffness can trigger and stabilize pathological epigenetic alterations via mechanotransduction signaling in fibrotic microenvironments. This mechanical information is then converted into stable epigenetic marks that lock cells into a profibrotic state. This process establishes a mechanically self-reinforcing barrier specific to fibrotic disease. This barrier perpetuates fibrosis progression by maintaining fibroblasts in an activated state and restricting AT2 regenerative potential [16,17,18].
To effectively disrupt this self-reinforcing barrier, therapeutic strategies must simultaneously target both its mechanical and epigenetic components. Multifunctional bioscaffolds represent promising vehicles to reverse pathological remodeling of the fibrotic niche [19,20]. The mechanical properties of these scaffolds can be precisely engineered to replicate physiological stiffness gradients. Soft regions that mimic healthy alveoli generally inhibit YAP/TAZ activity, whereas stiff regions promote RhoA/ROCK signaling; the underlying mechanisms are detailed in Section 2.2 and Section 2.3 [21,22]. Moreover, these constructs enable spatiotemporal delivery of regenerative factors (such as keratinocyte growth factor) and antifibrotic compounds, thereby coordinating tissue repair with fibrosis suppression [23,24,25]. Scaffolds also function as precision carriers for epigenetic modulators. Biodegradable polymers delivering DNA methyltransferase (DNMT) inhibitors (designated DNMTi) reverse hypermethylation of antifibrotic genes including BMP7. Histone deacetylases (HDACs) are enzymes that regulate epigenetic modifications, while their inhibitors, termed histone deacetylase inhibitors (HDACi), exert therapeutic effects by blocking HDAC activity. Hydrogel-encapsulated HDACi restore histone acetylation to block myofibroblast persistence [26,27,28,29]. These integrated scaffold systems convert passive structural templates into bioactive matrices that actively direct tissue regeneration [30,31]. Crucially, by simultaneously providing physiological mechanical cues and delivering epigenetic modulators, scaffolds offer a unique strategy to co-target and disrupt the intertwined mechano-epigenetic drivers of fibrosis. Preclinical studies using bleomycin (BLM)-induced models, precision-cut lung slices and organoids demonstrate scaffold efficacy in reversing established fibrotic remodeling [32,33,34].
This review presents an integrated mechano-epigenetic framework for scaffold-based lung regeneration. We analyze scaffold-guided mechanical control of cell fate based on current evidence. Systematic examination of epigenetic reprogramming via targeted chromatin modifier delivery is then presented. Particular emphasis is placed on the demonstrated synergy between scaffold mechanics and epigenetic drug release. Finally, clinical translation pathways for stimuli-responsive systems are evaluated. This framework enables novel therapeutic strategies to disrupt the self-sustaining fibrotic cascade in PF.

2. Mechanotransduction via Scaffold Mechanics

This section explores how scaffold-mediated mechanical cues regulate cellular responses essential for lung regeneration (Figure 1). We first establish fundamental principles of scaffold mechanical design including stiffness gradients and dynamic strain simulation. Subsequent analysis explores mechanotransduction mechanisms from focal adhesion assembly to cytoskeletal tension transmission. Finally, we analyze how integrated signaling pathways coordinate cell fate decisions and matrix remodeling. Together, these mechanobiological strategies establish targeted guidance of tissue morphogenesis to restore functional lung architecture in PF.

2.1. Scaffold Elastic Modulus and Stiffness Gradient Design

In lung tissue engineering, scaffold mechanical properties critically determine cellular behavior. Physiological alveolar ECM stiffness ranges from 1 to 5 kPa [35,36], whereas fibrotic extracellular matrix exhibits pathological stiffening beyond 20 kPa [37,38]. Biocompatible hydrogels accurately replicate this stiffness spectrum through controlled crosslinking density modulation. These materials allow researchers to mimic both healthy and early fibrotic microenvironments, providing optimal conditions for epithelial cell function and progenitor cell maintenance [39,40].
To simulate advanced fibrotic pathology, polymeric networks and nanocomposite reinforcements enable scaffold stiffening exceeding 20 kPa [41,42]. This approach recapitulates disease progression. It shows how excessive stiffness drives profibrotic cellular programs. Understanding these mechanisms is directly relevant for designing interventions to halt or reverse fibrosis. Such stiffened microenvironments authentically recapitulate disease progression by activating fibroblast-to-myofibroblast transdifferentiation. This process enhances alpha-smooth muscle actin expression and increases contractile activity [43,44]. Stiff substrates additionally upregulate fibrosis-associated genes including collagen type I alpha 1 chain (Col1a1) and Actin alpha 2 (ACTA2) while simultaneously impairing endogenous ECM remodeling capacity [44,45]. Pathological stiffness further disrupts basement membrane integrity, altering epithelial cell polarity and inhibiting regenerative repair mechanisms [46,47]. These effects on fibroblast activation and epithelial regeneration are partly mediated by mechanotransduction pathways such as YAP/TAZ and RhoA/ROCK, as described in Section 2.2 and Section 2.3.
Spatially controlled stiffness gradients generated via 3D bioprinting or microfluidic technologies establish continuous transitions from soft to stiff regions [48,49]. These engineered gradients mimic the heterogeneous stiffness found in fibrotic lungs. They enable region-specific investigation of epithelial and fibroblast responses, which is clinically relevant. Within these platforms, scaffold regions of specific stiffness elicit distinct cellular responses. Compliant zones at 1–5 kPa modeled on healthy alveoli promote epithelial cell adhesion and proliferation [50,51,52]. This regenerative effect is partly mediated via YAP/TAZ signaling, as elaborated in Section 2.2. Conversely, rigid domains exceeding 20 kPa that emulate fibrotic foci drive profibrotic cellular differentiation. These effects involve mechanotransduction pathways, including RhoA/ROCK, discussed in Section 2.3.

2.2. Dynamic Stretch and Aberrant YAP Activation

Physiological stretch supports alveolar epithelial homeostasis, whereas pathological stretch drives aberrant YAP activation linked to fibrosis. Beyond static stiffness matching, recapitulating physiological breathing-induced dynamic stretch is essential for alveolar epithelial barrier integrity and surfactant secretion [53,54]. In vitro models apply cyclic tensile strain to elastomeric scaffolds that replicate physiological breathing patterns observed in vivo [55,56]. Dynamic stimulation enhances integrin-mediated cell–matrix coupling and promotes continuous reorganization of intracellular stress fibers, leading to increased actin turnover rates [57,58]. Such remodeling optimizes tension transmission through actin-myosin networks, thereby facilitating activation of mechanotransduction effectors and supporting alveolar morphogenesis [59,60]. This biomechanical priming promotes efficient activation of downstream mechanotransduction effectors (such as YAP/TAZ), facilitating alveolar morphogenesis. Physiological stretch maintains alveolar epithelial homeostasis. Pathological stretch triggers aberrant YAP activation, leading to maladaptive repair and fibrosis. These findings inform scaffold design. Biomaterials that replicate normal cyclic breathing support epithelial function. They also limit pathological YAP/TAZ activity and improve the therapeutic efficacy of scaffolds in fibrotic lungs.
In fibrotic lungs, excessive stretch disrupts homeostasis and induces pathological nuclear YAP accumulation [61,62]. Activation of the IL-6–SFK–YAP signaling axis compromises epithelial fluid balance and barrier function, ultimately promoting fibrotic remodeling [63]. Idiopathic pulmonary fibrosis (IPF) patients exhibit persistent pathological YAP activation in airway epithelial Cells. This dysregulated YAP signaling directly contributes to epithelial barrier disruption [64,65]. Sustained YAP/TAZ hyperactivity across diverse cell types further impairs regeneration and accelerates fibrosis progression [22,66,67].

2.3. Focal Adhesions and Cytoskeletal Tension Transmission

Cells transduce external mechanical cues into biochemical responses through focal adhesions and the cytoskeletal network. When scaffold stiffness exceeds physiological ranges, enhanced phosphorylation occurs in core focal adhesion components including focal adhesion kinase (FAK) and adaptor protein Paxillin [68,69,70]. FAK phosphorylation drives α-SMA expression and myofibroblast differentiation, orchestrating TGF-β–mediated cytoskeletal reorganization and fibrogenic gene programming [71,72,73,74]. Pharmacological blockade of FAK inhibits these fibrogenic responses, highlighting its essential regulatory role in fibrosis.
Parallel activation of the RhoA/ROCK signaling cascade stimulates extensive stress fiber polymerization, significantly amplifying cytoskeletal contractile forces [75,76]. Force-stabilized networks strengthen cell-scaffold integration via integrin microaggregates while propagating mechanical signals to nuclei through actomyosin contractility and LINC complex-mediated nuclear reshaping [27,77,78,79]. This mechanical signaling axis modulates chromatin architecture, establishing a force-sensitive epigenetic regulation mechanism [80,81].

2.4. Integrated Mechanosignaling in Fibrosis

In PF, the mechanical microenvironment undergoes pathological changes. Elevated matrix stiffness and aberrant cellular tension characterize this dysregulated state. These altered mechanical forces activate key mechanosensitive signaling pathways, thereby driving fibrogenesis [38,82]. Elevated matrix stiffness (>20 kPa) induces fibroblast autocrine TGF-β1 secretion. TGF-β1 signaling through SMAD2/3 phosphorylation then drives fibroblast-to-myofibroblast transdifferentiation and excessive collagen deposition, constituting the core fibrotic cascade [45,83]. Elevated matrix stiffness and cytoskeletal tension activate YAP/TAZ, driving their nuclear translocation. This triggers TEAD-dependent transcription of target genes such as PCNA, CTGF, and CYR61, which together enhance fibroblast proliferation, migration, survival, and myofibroblast differentiation [84,85,86]. Increased stiffness also potentiates Wnt/β-catenin signaling. Stabilized β-catenin translocates to the nucleus and upregulates profibrotic genes, including CCND1 (Cyclin D1) and MYC (c-Myc) [87,88]. Collectively, activation of these mechanosensitive pathways expands and activates fibroblasts within the stiffened microenvironment, driving ECM deposition and fibrogenesis. Pathological ECM stiffening converges on the TGF-β/SMAD and YAP/TAZ pathways, creating a self-reinforcing signaling network that drives progressive lung remodeling and functional decline [89,90,91].
Beyond mechanotransduction, mechanical stress has been shown to remodel epigenetic states. This induces persistent mechanical memory through modified histone states and concurrent chromatin architectural reorganization [92,93,94]. Such mechano-driven epigenetic reprogramming plays a critical role in maintaining fibrogenic cell phenotypes. Pathological tissue stiffness is associated with upregulation of epigenetic enzymes such as DNA methyltransferase 1 (DNMT1) and G9a (EHMT2), promoting promoter DNA hypermethylation and repressive histone modifications. Consequently, antifibrotic gene expression is suppressed, accelerating fibrogenesis. These epigenetic alterations induce chromatin compaction and transcriptional silencing, thereby maintaining fibroblast profibrotic activation [95,96]. In liver fibrosis models, combined G9a (EHMT2) and DNMT1 inhibition reverses these epigenetic aberrations [95,97]. This intervention reactivates protective gene expression and potently suppresses fibrogenesis, indicating strong therapeutic translatability. Mechanical stimuli orchestrate histone methylation states via cytoskeletal force transmission to the nucleus [98,99,100]. This mechano-sensitive epigenetic reprogramming strongly influences cellular phenotypic commitment and functional specialization. Collectively, these findings demonstrate the close interplay between mechanical forces and epigenetic regulation in fibrosis pathogenesis. This mechanistic hierarchy provides a foundation for scaffold-mediated epigenetic reprogramming strategies aimed at disrupting pathological mechanical memory, detailed in Section 3.

3. Epigenetic Drug Delivery via Scaffolds

Mechanical cues regulate cellular behavior through canonical signaling pathways while modifying epigenetic programs in lung fibroblasts and epithelial Cells. This process reinforces profibrotic gene expression [46,101]. Accumulating evidence indicates that matrix stiffness and cyclic strain remodel DNA methylation profiles and histone modification patterns, driving persistent myofibroblast differentiation [99,102]. This creates a mechanically reinforced epigenetic barrier that sustains fibrosis. To circumvent this mechanically stabilized epigenetic barrier, scaffold-based delivery systems can be designed for localized administration of small-molecule inhibitors or nucleic acid therapeutics within fibrotic niches. The following section details strategies to deliver DNMT and HDACi via hydrogel and nanofiber scaffolds, alongside emerging approaches targeting non-coding RNA and methyl-binding proteins, for spatiotemporal control of epigenetic reprogramming in PF. These strategies aim to directly reverse the mechano-induced epigenetic dysregulation described in Section 2.4.

3.1. DNMT Inhibitor Carrier Design

Pathological hypermethylation of anti-fibrotic gene promoters, driven by aberrant DNMT upregulation, constitutes a well-established therapeutic target in PF [103,104]. Critically, this hypermethylation is amplified by pathological mechanical cues. To address this mechanism, localized delivery systems show significant promise, specifically gelatin methacryloyl (GelMA) hydrogels for the DNMTi 5-azacytidine (5-AZA). This approach utilizes GelMA’s enzyme-responsive biodegradation to achieve sustained drug release within fibrotic niches exhibiting elevated matrix metalloproteinase expression. The controlled release kinetics align with the prolonged exposure requirements for effective epigenetic modulation [105,106,107]. Supporting evidence from varied delivery vehicles demonstrates 5-AZA’s ability to suppress DNMT function and restore physiological methylation patterns [103,108]. Moreover, GelMA’s biomimetic properties such as RGD motifs facilitate cellular interactions that may synergize with epigenetic reprogramming [109,110,111].
GelMA hydrogels provide critical advantages as localized epigenetic therapy carriers. Their tunable physicochemical characteristics allow tailored control of drug release kinetics required for sustained epigenetic reprogramming [112,113]. The hydrophilic hydrogel matrix of GelMA enables effective encapsulation of hydrophilic therapeutics such as 5-AZA, concurrently preventing degradation of susceptible compounds during transit. This protective capability overcomes inherent stability limitations of nucleoside analogs in systemic circulation [114,115,116]. GelMA’s biomimetic ECM structure facilitates cell–matrix interactions extending beyond RGD-mediated adhesion. These interactions could potentiate cellular sensitivity to epigenetic agents through mechanotransduction pathway regulation, a process actively sustaining fibrotic progression [42,117]. Experimental validation confirms that microenvironmental reprogramming enhances therapeutic outcomes of epigenome-targeting agents in pathological tissues. Collectively, these attributes establish GelMA-mediated 5-AZA delivery as a viable strategy for targeted DNMT inhibition in fibrotic lung tissue. This approach directly counters stiffness-induced hypermethylation, resolving the core epigenetic barrier underlying fibrosis.

3.2. HDACi Carrier Design

The inherent biocompatibility and tunable degradation of electrospun polycaprolactone (PCL)/collagen nanofibers position them as viable substrates for sustained drug release [118,119]. Combining PCL’s mechanical robustness with collagen’s bioactivity, these scaffolds maintain drug release for weeks. Such sustained delivery supports the efficacy of diverse regenerative therapeutics, including antibiotics and growth factors [120,121]. For instance, PCL-based airway stents incorporating antitumor agents have shown clinical potential in reducing tissue hyperplasia through prolonged local drug exposure [122,123]. Collagen-enhanced nanofibrous scaffolds potentiate wound healing by confining therapeutic agents at disease loci with precise spatiotemporal control [124,125]. The HDACi trichostatin A (TSA) and suberoylanilide hydroxamic acid (SAHA) attenuate fibrotic remodeling and oncogenesis via selective epigenetic remodeling [126,127,128]. Importantly, they counteract the histone deacetylation driven, in part, by pathological mechanical stress. Despite their therapeutic potential, the clinical translation of HDAC inhibitors is hampered by challenges including short systemic circulation and dose-limiting toxicities. To address these limitations, nanoencapsulation strategies have been developed to improve HDACi pharmacokinetic profiles. Evidence suggests that polymer-based nano-delivery systems, in particular, offer advantages in enhancing drug stability and promoting tissue-specific accumulation.
The strategy of integrating HDACi within PCL/collagen scaffolds builds upon established foundations in drug delivery and antifibrotic therapy. Direct reports on this specific combination for fibrosis treatment remain limited. However, technical feasibility is supported by analogous drug-carrier systems. Electrospun PCL matrices have successfully encapsulated structurally similar hydrophobic compounds, demonstrating compatibility with the physicochemical properties of typical HDACi [129,130]. Collagen components provide cell-adhesive motifs including integrin-binding domains, which may enhance drug accumulation and cellular uptake of HDACi at pathological sites. This effect has been observed in tumor microenvironment studies where collagen-functionalized carriers promote drug internalization by stromal or tumor Cells [131,132]. Furthermore, the nanofibrous scaffold architecture delivers physical cues known to directly regulate fibroblast spreading behavior and phenotypic transition [133,134]. This modulation establishes a foundation for creating a microenvironment favorable for targeted epigenetic therapy in fibrotic lesions.
Emerging evidence suggests such combinatorial approaches could address critical limitations in chronic disease management. Localized HDACi delivery via biodegradable scaffolds may circumvent systemic toxicity while enabling sustained modulation of fibrotic cascades, a strategy aligned with current trends in precision nanomedicine. Further optimization of release kinetics through material engineering could enhance temporal control over epigenetic reprogramming events, positioning this platform as a promising frontier for antifibrotic intervention. This approach directly targets the mechano-induced histone hypoacetylation contributing to the fibrotic epigenetic barrier.

3.3. Novel Epigenetic Targets for Scaffold-Based Intervention

Novel epigenetic regulators beyond classical DNMTi and HDACi provide opportunities for precision antifibrotic therapy through engineered scaffold-based delivery systems [135,136]. For instance, let-7 miRNA delivery suppresses the BTB and CNC homology 1 (BACH1)-enhancer of zeste homolog 2 (EZH2)-MYC signaling axis in murine fibrotic lungs, inhibiting alveolar epithelial cell reprogramming toward pro-fibrotic phenotypes [137,138]. Genetic lineage tracing demonstrates that let-7 restoration suppresses EZH2 expression in AT2 Cells, with concomitant reduction in global H3K27me3 histone methylation in fibrotic lungs [137]. Chromatin immunoprecipitation analyses further reveal that this epigenetic remodeling reactivates transcriptional programs essential for cell differentiation, inhibiting transition toward pro-fibrotic intermediates [139,140]. Although these associations are established, the precise causal relationship between let-7 mediated EZH2 downregulation and site-specific H3K27me3 reduction requires further investigation. Scaffold systems enabling controlled nucleic acid release could deliver let-7 mimics to exploit this epigenetic cascade for coordinated editing in fibrotic lungs.
Regarding another target, pathological MeCP2 overexpression induces epigenetic silencing of WIF1 via promoter hypermethylation, driving fibroblast-to-myofibroblast transdifferentiation in human fibrotic tissues [141,142]. Nanofibrous scaffolds achieve sustained, localized siRNA delivery for long-term gene silencing and effective fibrotic response modulation [143,144]. However, experimental evidence remains unreported for MeCP2-targeted siRNA nanofiber scaffolds specifically acting through WIF1 demethylation and Wnt/β-catenin pathway suppression in high-impact studies. These findings validate combined MeCP2-targeted siRNA and advanced scaffold systems for precise epigenetic intervention in fibrotic diseases. Essential future work must resolve the molecular mechanisms governing this targeted strategy.
The therapeutic efficacy of these approaches depends on scaffold design parameters. Optimized nanofiber architectures provide controlled release kinetics for nucleic acid therapeutics, and surface modifications enhance cellular internalization efficiency at fibrotic lesions [145,146]. Notably, biomaterial physical properties directly influence epigenetic effector activity, since substrate stiffness modulates histone modification enzymes in mesenchymal Cells [80,147,148]. Dual-targeting scaffolds co-delivering two nucleic acid therapeutics such as miRNAs or siRNAs achieve synergistic modulation of fibrotic pathways. These systems demonstrate enhanced cellular reprogramming and gene silencing effects relative to single-agent delivery in preclinical models. Cardiac fibroblast reprogramming exemplifies this advantage with dual miRNA-loaded scaffolds outperforming single miRNA systems [149,150,151]. Collectively, scaffold-mediated epigenetic targeting of regulators like let-7 and MeCP2 demonstrates a precision antifibrotic strategy. Critical knowledge gaps persist regarding mechanical memory erasure efficiency and in vivo spatiotemporal control of epigenetic modifiers (such as DNMTs and siRNAs). Resolving these challenges through intelligent biomaterial design will accelerate clinical translation of mechano-epigenetic therapies.

3.4. Dual-Drug Synergy Strategy

Combined treatment with 5-AZA and TSA synergistically regulates epigenetic pathways including DNA methylation and histone acetylation, reducing fibrotic progression. Preclinical kidney fibrosis models demonstrate that 5-AZA/TSA co-treatment reactivates key antifibrotic genes and signaling pathways, reducing fibrotic progression [152,153]. This dual epigenetic modulation enhances antifibrotic efficacy by concurrently targeting DNA methylation and histone acetylation. While current scaffold systems lack mechanical gradient-enabled sequential release of 5-AZA/TSA, integration of spatiomechanical cues with epigenetic drug actions represents a strategic advancement for antifibrotic biomaterial design.
Current evidence indicates synergistic epigenetic regulation from combined DNMTi and HDACi treatment may enhance CpG island demethylation and histone acetylation effects [154,155,156]. Biomaterial scaffolds enable co-delivery of epigenetic agents, potentially enhancing synergistic efficacy through spatiotemporal control [30,157,158]. Supporting this concept, fibrotic lung models demonstrate substantially reduced collagen I deposition, reestablished E-cadherin expression and restored alveolar architecture. However, studies report limited direct evidence comparing these systems to single-drug approaches. Recent studies establish that temporally and spatially controlled release systems dynamically modulate cell fate and gene expression. Mechanical gradient integration further enhances this regulatory capacity [81,159,160,161]. These advances suggest promise for overcoming mechano-epigenetic barriers, though synergistic disruption mechanisms require further validation. This approach interrupts fibrotic feedback loops and remodels alveolar microenvironments via coordinated mechano-epigenetic reprogramming (Figure 2).

4. Selected Pulmonary Fibrosis Models for Scaffold Application

The selection of pathophysiologically relevant models is paramount for evaluating scaffold-based interventions in pulmonary fibrosis. While in vivo systems capture systemic complexity, ex vivo platforms offer unprecedented resolution for mechanistic dissection. This section examines complementary models that address translational challenges from target validation to clinical application. These models enable integrated assessment of scaffold biofunctionality across molecular, cellular and organ-level metrics.

4.1. Bleomycin-Induced Murine Model

The BLM-induced murine model remains the gold standard for preclinical evaluation of scaffold-based antifibrotic interventions. This model reliably reproduces core pathophysiological features of PF within a controlled time course. Specifically, it recapitulates alveolar epithelial injury, inflammatory cell infiltration and progressive ECM remodeling [162,163]. Decellularized lung matrix (DLM) scaffolds sourced from healthy or fibrotic lungs maintain native three-dimensional architecture. These scaffolds preserve critical ECM ligand composition and physiological biomechanical cues. This preservation establishes DLM as a reproducible platform for investigating cell–matrix interactions and directing targeted recellularization protocols [164,165,166].
Recellularization of DLM scaffolds with epithelial progenitors or fibroblasts promotes cellular adhesion and migration, providing a microenvironment for lung tissue regeneration. While short-term gas exchange improvements have been documented in animal models, long-term functional recovery including sustained pulmonary compliance and gas-exchange capacity remains challenging. Direct in vivo evidence for DLM scaffolds constraining myofibroblastic differentiation or releasing antifibrotic factors is still limited [31,167,168].
These protective outcomes are attributable to the scaffold’s dual functions: restitution of physiological mechanical microenvironments and localized delivery of biochemical signals that modulate profibrotic pathways. Recent work emphasizes the translational potential of ECM-derived hydrogels for attenuating fibrogenic remodeling in BLM models, while highlighting practical considerations for scaffold preparation sterility and recellularization efficiency [169,170,171].
Practical evaluation requires multimodal endpoints. Histomorphometry and collagen quantification such as hydroxyproline assays are essential for assessing matrix burden [172,173]. Respiratory mechanics measured by flexiVent systems and arterial blood-gas metrics provide functional corollaries [174,175]. Scaffold-mediated benefit correlates with reduced collagen deposition and reversal of cellular activation states. This includes decreased alpha-smooth muscle actin (α-SMA) expression and downregulated TGF-β/SMAD signalling, consistent with mechanobiological mechanisms [169,176,177]. To strengthen causal inference, experimental designs increasingly combine DLM implantation with lineage tracing, targeted mechanotransduction perturbations such as YAP/TAZ inhibition, and epigenetic readouts. This links restored mechanics to transcriptional reprogramming of resident cells [174,178,179,180].

4.2. Ex Vivo Lung Slices and Organoids

Precision-cut lung slices (PCLS) and lung organoids occupy a critical intermediate niche between cell culture and whole-animal studies. PCLS retain native multicellular architecture, alveolar–capillary juxtaposition, and local ECM context. This enables direct scaffold application and high-fidelity monitoring of epithelial, mesenchymal and immune responses [178,181,182].
When combined with high-dimensional readouts such as single-cell RNA sequencing, ex vivo perturbations reveal cell type-specific transcriptional trajectories. These permit rapid assessment of scaffold-induced modulation of profibrotic gene programmes including Col1a1, ACTA2 and MMPs alongside epigenetic regulators [101,181,183]. Recent methodologic advances include standardized PCLS preparation and improved cryopreservation workflows. These have expanded assay throughput and reproducibility for mechanistic dissection [178,181].
Lung organoids derived from pluripotent or adult stem cells complement PCLS by enabling longer-term culture. They allow manipulation of stem/progenitor lineage cues and systematic tuning of scaffold properties such as porosity and stiffness gradients [184,185]. Co-culture with multifunctional scaffolds reproduces key morphogenetic events including lumenogenesis and epithelial differentiation. This facilitates controlled screens to identify stiffness ranges and drug-release kinetics favoring regenerative outcomes [186,187,188].
These platforms are particularly valuable for personalized scaffold optimization when using patient-derived organoids. This links ex vivo response phenotypes to potential precision-medicine strategies. Collectively, PCLS and organoid systems accelerate lead selection, reduce animal usage, and provide mechanistic bridges informing in vivo scaffold deployment [189,190].

5. Discussion and Future Perspectives

The therapeutic potential of multifunctional scaffolds targeting mechanobiology and epigenetics for lung regeneration has been established in preceding sections. While the benefits of these approaches are substantial, several technical challenges remain. Moving forward, translating this potential into clinical practice requires addressing key technological challenges and exploring advanced strategies for scaffold design and functionality. To provide a coherent perspective, this section highlights three interrelated directions: stimuli-responsive scaffolds, CRISPR-based epigenetic editing, and AI-driven personalization. Rather than being considered separately, these strategies are emphasized in terms of their integration (Figure 3).

5.1. Stimuli-Responsive Scaffolds

Recent advances have shown that smart scaffolds, constructed from functional polymers and nanocomposites, can sense changes in the pulmonary microenvironment. These changes include factors like oxidative stress and mechanical stretch. Importantly, the scaffolds can dynamically adjust their behavior in response to these sensed changes [191,192,193]. For example, ROS-responsive linkers have been used to build hydrogels. These hydrogels selectively degrade under elevated ROS levels. This degradation enables the on-demand release of antioxidant or antifibrotic drugs specifically within inflamed tissues [194,195,196]. Independent studies demonstrate that ROS-responsive nanoparticles and liposomes can be formulated for inhalation delivery to fibrotic lungs. This strategy exploits the high ROS microenvironment characteristic of these diseased tissues. Consequently, local drug accumulation is improved [197,198,199]. Block copolymers and shape memory polymers have been designed to exhibit reversible deformation under cyclic stretch. This mechanical stimulus mimics the natural breathing cycle. Consequently, these materials can modulate local stiffness in a time-dependent manner [200,201,202]. Recent advances have yielded two-way and light-responsive shape memory polymers (SMPs). These materials offer practical actuation strategies. Such strategies are compatible with physiological strain regimes. Furthermore, they utilize milder activation methods. These features make them attractive for lung applications [203,204]. Triggerable microcapsules embedded in hydrogels can release large protein cargos. These cargos include enzymes or CRISPR effector proteins. Release occurs under specific triggers such as light, magnetic fields, or ROS. This capability demonstrates the feasibility of scaffold-enabled, on-demand epigenetic editing while highlighting the need to carefully optimize delivery systems to ensure efficacy and safety [205,206,207].
Building on these advances in responsive materials, the next section considers how CRISPR-based epigenetic editing provides complementary opportunities for precise regulation at the molecular level.

5.2. CRISPR-Based Epigenetic Editing

CRISPR/dCas9-based epigenome editors, such as dCas9–TET1 and dCas9–p300, have matured as powerful tools. These tools can write or erase locus-specific DNA methylation and histone acetylation marks. This capability enables precise control of gene regulation. Critically, it achieves this without altering the underlying genomic sequence [208,209,210]. Systematic epigenome editing studies demonstrate that targeted chromatin modification can produce robust transcriptional responses. Importantly, these responses are context-dependent. This evidence supports therapeutic strategies aimed at the locus-specific reactivation of antifibrotic genes [211,212]. However, efficient and safe delivery of CRISPR effectors to pulmonary tissue remains a significant challenge, limiting translation. Local delivery of CRISPR effectors from biomaterials reduces systemic exposure. It also helps confine editing activity to the intended tissue microenvironment. This targeted approach represents a major translational advantage over systemic vectors [213,214,215]. However, there are significant limitations for CRISPR delivery specifically to lung tissue. These include the dense extracellular matrix and mucus barrier in fibrotic lungs, poor penetration of viral or non-viral carriers, potential immune clearance, and limited retention time at the target site. Overcoming these obstacles requires the development of specialized delivery systems, such as inhalable nanoparticles, hydrogel-based depots, or scaffold-mediated local release, to enhance targeting efficiency and editing efficacy [216,217,218]. Safety considerations for CRISPR-based editing are critical. Key concerns include off-target epigenetic effects, immunogenicity of CRISPR components, and persistent unintended chromatin changes. These risks motivate the adoption of specific mitigation strategies. These strategies include the use of nonviral carriers, transient cargo formats, and inducible activation systems. Collectively, they aim to minimize potential hazards [219,220].
While these delivery and safety challenges remain, artificial intelligence offers a means to optimize scaffold design and integrate CRISPR-based interventions with patient-specific strategies, as discussed in the following section.

5.3. Artificial Intelligence-Driven Personalized Design

Artificial intelligence (AI) and machine learning (ML) methods are increasingly applied to key challenges in biomaterial development. These challenges include polymeric biomaterial design, scaffold architecture optimization, and multi-parameter trade-off problems. Significantly, these methods accelerate discovery and design cycles [221,222,223]. Data-efficient ML pipelines integrate key computational and experimental methods. Specifically, they couple finite element modeling, experimental screening, and neural network surrogates. This integration enables constrained multi-objective optimization. The optimization targets scaffold lattices and mechanical properties critical for achieving tissue-specific compliance [224,225]. Reviews and recent applied studies demonstrate the utility of specific machine learning approaches. These approaches include supervised learning, active learning, and Bayesian optimization. They can significantly reduce experimental iterations. Furthermore, they effectively guide patient-specific scaffold customization. In addition, AI-guided design considers key scaffold properties, including mechanical compliance, porosity, and drug-loading capacity, to optimize regenerative performance. Despite these advantages, practical implementation must address reproducibility, scale-up challenges, and integration with regulatory standards. This guidance relies on integration with high-throughput characterization and multi-omics readouts [226,227]. AI further supports the scale-up of scaffold manufacturing. Specifically, it aids in predicting key process parameters for additive manufacturing. Additionally, AI enables defect detection during three-dimensional printing. These capabilities collectively enhance reproducibility and batch quality control. Such improvements are essential for successful clinical translation [228,229]. Collectively, AI-driven approaches bridge computational design with clinical-scale manufacturing, accelerating the translation of mechano-epigenetic scaffolds.
In summary, stimuli-responsive scaffolds, CRISPR-based epigenetic editing, and AI-driven personalized design are best viewed as complementary rather than isolated strategies. Smart scaffolds can serve as delivery platforms for CRISPR effectors, while AI methods can refine scaffold properties and support patient-specific applications. Taken together, the integration of these approaches offers a more effective route toward precision therapies for lung regeneration than any single strategy alone.

6. Discussion

PF is a chronic progressive disease initiated by recurrent epithelial injury. This condition is defined by alveolar epithelial cell dysfunction combined with aberrant fibroblast activation, pathological ECM deposition, and sustained inflammatory cytokine release. Progressive destruction of lung architecture and declining compliance characterize advanced PF, ultimately leading to respiratory failure [2,230]. Although current antifibrotic drugs (e.g., pirfenidone, nintedanib) can slow disease progression, they cannot significantly reverse established structural damage [231,232]. Given these limitations, developing novel regenerative strategies that both repair tissue and counteract fibrosis remains a core objective in pulmonary medicine.
Multifunctional bioscaffolds deliver coordinated mechanical, biochemical, and pharmacologic interventions and show promising potential to advance lung regeneration. However, direct mechanistic evidence of scaffold-mediated mechano-epigenetic reprogramming in pulmonary fibrosis remains limited [27,233]. Structurally and physically, scaffold materials can precisely mimic the elasticity and topography of the native alveolar ECM within the physiological stiffness range (1–5 kPa). Modulating both stiffness and nanostructure within this range promotes alveolar epithelial cell proliferation and differentiation, thereby facilitating the regeneration of the gas-exchange interface [165,234]. Mechanical stresses transmitted by the scaffold activate key mechanosensitive pathways, including YAP/TAZ, RhoA/ROCK, and TGF-β/SMAD signaling. This activation regulates critical cellular processes such as cell fate determination and ECM synthesis. Importantly, inhibiting these activated pathways disrupts the pro-fibrotic positive feedback loop, thereby attenuating fibrosis [86,165,235].
As carriers for epigenetic regulation, scaffolds can achieve local, precise release of DNMTi and HDACi. For example, 5-Aza demethylates and reactivates antifibrotic genes such as BMP7 and PPARγ, thereby counteracting stiffness-induced hypermethylation [135,236]. By contrast, HDACi including TSA and SAHA increase H3K9 acetylation, restoring histone marks altered by pathological mechanical cues. This restoration blocks TGF-β1/SMAD signaling and induces myofibroblast apoptosis [237,238]. Coupling these release profiles with mechanical gradients enables spatiotemporal and multi-targeted epigenetic modulation. This combined approach could enhance the therapeutic impact beyond current pharmacological options by reshaping cell fate and transcriptional networks at the tissue level [239].
Three-dimensional scaffolds significantly enhance disease modeling in oncology and cardiovascular research through accurate replication of tissue-specific microenvironments. These platforms actively control cell phenotype specification, transcriptional programming, and cell-ECM signaling dynamics, substantially improving physiological relevance in vitro. Scaffold microarchitecture parameters, specifically fiber diameter, pore geometry, and surface wettability, critically regulate cellular adhesion kinetics, proliferative capacity, migratory patterns, and drug response profiles. Recent studies in glioblastoma and myocardial infarction models demonstrate the necessity for model-specific scaffold customization [240,241,242,243]. Although these findings are encouraging, evidence in pulmonary fibrosis models is still emerging, highlighting the need for further validation.
Despite these advances, direct mechanistic evidence of scaffold-mediated mechano-epigenetic reprogramming in pulmonary fibrosis remains limited. Current high-impact research prioritizes oncological and cardiovascular applications, revealing a critical need to investigate scaffold-driven fibrosis regression pathways. To accelerate clinical translation of scaffold-based therapies, we propose three essential actions. First, establish reproducible manufacturing protocols with robust quality control systems. Second, implement standardized epigenetic assays for quantitative analysis of locus-specific DNA methylation and histone acetylation. Third, complete Good Laboratory Practice compliant toxicology studies and large-animal feasibility assessments before initiating human trials. Addressing these steps will bridge the gap between preclinical promises and practical clinical applications.
Future progress will combine stimuli-responsive scaffolds incorporating photo-responsive polymers, pH-sensitive materials, and hydrolytically degradable systems with CRISPR-based epigenetic editing and single-cell omics-guided target selection. This integration will convert tissue engineering scaffolds from passive structural elements into multifunctional dynamic platforms that actively regulate biological processes. Such systems will disrupt pathological mechano-epigenetic crosstalk through unified sensing and therapeutic intervention modules. Machine learning-driven precision engineering could further optimize these strategies, ensuring scaffold designs are adapted to specific lesions, disease stages, and individual patients.
In summary, multifunctional scaffolds serve as complementary therapeutic tools linking mechanistic discoveries in lung regeneration to clinical implementation. The integrated co-targeting of mechanosignaling and epigenetic reprogramming offers a balanced perspective, combining promise with the recognition of current limitations, and provides a versatile approach for PF and terminal lung diseases. This revised view situates scaffold-based therapies within the broader context of current antifibrotic strategies, highlighting both opportunities and practical considerations for translation.

7. Conclusions

Multifunctional scaffolds offer a promising approach for lung regeneration by combining mechanical support with biochemical and epigenetic cues. By simultaneously addressing abnormal mechanotransduction and dysregulated gene expression, these scaffolds may help reverse fibrosis and support tissue repair. The continued development of adaptable, patient-tailored scaffolds could facilitate the translation of these strategies into precision therapies for pulmonary diseases.

Author Contributions

J.W. designed the study, wrote the manuscript, reviewed and edited the manuscript. A.X. edited the drafts and conceptualized the figures. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by grants from the National Natural Science Foundation of China (No.82373547) and also supported by Jiangsu Province Science and Technology Plan Project ‘Provincial Frontier Technology R&D Program’ (BF2024054).

Data Availability Statement

No new data were created or analyzed in this study.

Acknowledgments

Figures were partly created using icons from BioRender.com.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Moss, B.J.; Ryter, S.W.; Rosas, I.O. Pathogenic Mechanisms Underlying Idiopathic Pulmonary Fibrosis. Annu. Rev. Pathol. Mech. Dis. 2022, 17, 515–546. [Google Scholar] [CrossRef] [PubMed]
  2. Richeldi, L.; Collard, H.R.; Jones, M.G. Idiopathic pulmonary fibrosis. Lancet 2017, 389, 1941–1952. [Google Scholar] [CrossRef]
  3. Lettieri, S.; Bertuccio, F.R.; del Frate, L.; Perrotta, F.; Corsico, A.G.; Stella, G.M.; Madala, S.K. The Plastic Interplay between Lung Regeneration Phenomena and Fibrotic Evolution: Current Challenges and Novel Therapeutic Perspectives. Int. J. Mol. Sci. 2024, 25, 547. [Google Scholar] [CrossRef] [PubMed]
  4. Ma, J.; Li, G.; Wang, H.; Mo, C. Comprehensive review of potential drugs with anti-pulmonary fibrosis properties. Biomed. Pharmacother. 2024, 173, 116282. [Google Scholar] [CrossRef]
  5. Distler, J.H.W.; Gyoerfi, A.-H.; Ramanujam, M.; Whitfield, M.L.; Koenigshoff, M.; Lafyatis, R. Shared and distinct mechanisms of fibrosis. Nat. Rev. Rheumatol. 2019, 15, 705–730. [Google Scholar] [CrossRef]
  6. Long, Y.; Niu, Y.; Liang, K.; Du, Y. Mechanical communication in fibrosis progression. Trends Cell Biol. 2022, 32, 70–90. [Google Scholar] [CrossRef]
  7. Strippoli, R.; Sandoval, P.; Moreno-Vicente, R.; Rossi, L.; Battistelli, C.; Terri, M.; Pascual-Anton, L.; Loureiro, M.; Matteini, F.; Calvo, E.; et al. Caveolin1 and YAP drive mechanically induced mesothelial to mesenchymal transition and fibrosis. Cell Death Dis. 2020, 11, 647. [Google Scholar] [CrossRef]
  8. Zacharias, W.J.; Frank, D.B.; Zepp, J.A.; Morley, M.P.; Alkhaleel, F.A.; Kong, J.; Zhou, S.; Cantu, E.; Morrisey, E.E. Regeneration of the lung alveolus by an evolutionarily conserved epithelial progenitor. Nature 2018, 555, 251–255. [Google Scholar] [CrossRef]
  9. Ruaro, B.; Salton, F.; Braga, L.; Wade, B.; Confalonieri, P.; Volpe, M.C.; Baratella, E.; Maiocchi, S.; Confalonieri, M. The History and Mystery of Alveolar Epithelial Type II Cells: Focus on Their Physiologic and Pathologic Role in Lung. Int. J. Mol. Sci. 2021, 22, 2566. [Google Scholar] [CrossRef]
  10. Kim, J.-H.; Ham, S.; Lee, Y.; Suh, G.Y.; Lee, Y.-S. TTC3 contributes to TGF-β1-induced epithelial-mesenchymal transition and myofibroblast differentiation, potentially through SMURF2 ubiquitylation and degradation. Cell Death Dis. 2019, 10, 92. [Google Scholar] [CrossRef] [PubMed]
  11. Xu, R.; Wu, M.; Wang, Y.; Li, C.; Zeng, L.; Wang, Y.; Xiao, M.; Chen, X.; Geng, S.; Lai, P.; et al. Mesenchymal stem cells reversibly de-differentiate myofibroblasts to fibroblast-like cells by inhibiting the TGF-β-SMAD2/3 pathway. Mol. Med. 2023, 29, 59. [Google Scholar]
  12. Lv, Q.; Wang, J.; Xu, C.; Huang, X.; Ruan, Z.; Dai, Y. Pirfenidone alleviates pulmonary fibrosis in vitro and in vivo through regulating Wnt/GSK-3β/β-catenin and TGF-β1/Smad2/3 signaling pathways. Mol. Med. 2020, 26, 49. [Google Scholar] [CrossRef]
  13. Zhang, X.; Xu, Z.; Chen, Q.; Zhou, Z. Notch signaling regulates pulmonary fibrosis. Front. Cell Dev. Biol. 2024, 12, 1450038. [Google Scholar] [CrossRef] [PubMed]
  14. Benincasa, G.; DeMeo, D.L.; Glass, K.; Silverman, E.K.; Napoli, C. Epigenetics and pulmonary diseases in the horizon of precision medicine: A review. Eur. Respir. J. 2021, 57, 2003406. [Google Scholar] [CrossRef] [PubMed]
  15. Ren, L.; Chang, Y.-F.; Jiang, S.-H.; Li, X.-H.; Cheng, H.-P. DNA methylation modification in Idiopathic pulmonary fibrosis. Front. Cell Dev. Biol. 2024, 12, 1416325. [Google Scholar] [CrossRef]
  16. Garitano, N.; Aguado-Alvaro, L.P.; Pelacho, B. Emerging Epigenetic Therapies for the Treatment of Cardiac Fibrosis. Biomedicine 2025, 13, 1170. [Google Scholar] [CrossRef]
  17. Toscano-Marquez, F.; Romero, Y.; Espina-Ordonez, M.; Cisneros, J. Absence of HDAC3 by Matrix Stiffness Promotes Chromatin Remodeling and Fibroblast Activation in Idiopathic Pulmonary Fibrosis. Cells 2023, 12, 1020. [Google Scholar] [CrossRef] [PubMed]
  18. Liu, R.; Li, Y.; Zheng, Q.; Ding, M.; Zhou, H.; Li, X. Epigenetic modification in liver fibrosis: Promising therapeutic direction with significant challenges ahead. Acta Pharm. Sin. B 2024, 14, 1009–1029. [Google Scholar] [CrossRef]
  19. Xue, T.; Qiu, X.; Liu, H.; Gan, C.; Tan, Z.; Xie, Y.; Wang, Y.; Ye, T. Epigenetic regulation in fibrosis progress. Pharmacol. Res. 2021, 173, 105910. [Google Scholar] [CrossRef]
  20. Ligresti, G.; Caporarello, N.; Meridew, J.A.; Jones, D.L.; Tan, Q.; Choi, K.M.; Haak, A.J.; Aravamudhan, A.; Roden, A.C.; Prakash, Y.S.; et al. CBX5/G9a/H3K9me-mediated gene repression is essential to fibroblast activation during lung fibrosis. JCI Insight 2019, 4, e127111. [Google Scholar] [CrossRef]
  21. Panciera, T.; Azzolin, L.; Cordenonsi, M.; Piccolo, S. Mechanobiology of YAP and TAZ in physiology and disease. Nat. Rev. Mol. Cell Biol. 2017, 18, 758–770. [Google Scholar] [CrossRef]
  22. Sun, M.; Sun, Y.; Feng, Z.; Kang, X.; Yang, W.; Wang, Y.; Luo, Y. New insights into the Hippo/YAP pathway in idiopathic pulmonary fibrosis. Pharmacol. Res. 2021, 169, 105635. [Google Scholar] [CrossRef]
  23. Lu, X.; Jin, H.; Quesada, C.; Farrell, E.C.; Huang, L.; Aliabouzar, M.; Kripfgans, O.D.; Fowlkes, J.B.; Franceschi, R.T.; Putnam, A.J.; et al. Spatially-directed cell migration in acoustically-responsive scaffolds through the controlled delivery of basic fibroblast growth factor. Acta Biomater. 2020, 113, 217–227. [Google Scholar] [CrossRef] [PubMed]
  24. Huang, L.; Quesada, C.; Aliabouzar, M.; Fowlkes, J.B.; Franceschi, R.T.; Liu, Z.; Putnam, A.J.; Fabiilli, M.L. Spatially-directed angiogenesis using ultrasound-controlled release of basic fibroblast growth factor from acoustically-responsive scaffolds. Acta Biomater. 2021, 129, 73–83. [Google Scholar] [CrossRef] [PubMed]
  25. Gansevoort, M.; Wentholt, S.; Li Vecchi, G.; de Vries, M.; Versteeg, E.M.M.; Boekema, B.K.H.L.; Choppin, A.; Barritault, D.; Chiappini, F.; van Kuppevelt, T.H.; et al. Next-Generation Biomaterials for Wound Healing: Development and Evaluation of Collagen Scaffolds Functionalized with a Heparan Sulfate Mimic and Fibroblast Growth Factor 2. J. Funct. Biomater. 2025, 16, 51. [Google Scholar] [CrossRef]
  26. Sukpaita, T.; Chirachanchai, S.; Chanamuangkon, T.; Nampuksa, K.; Monmaturapoj, N.; Sumrejkanchanakij, P.; Pimkhaokham, A.; Ampornaramveth, R.S. Novel Epigenetic Modulation Chitosan-Based Scaffold as a Promising Bone Regenerative Material. Cells 2022, 11, 3217. [Google Scholar] [CrossRef]
  27. Han, P.; Gomez, G.A.; Duda, G.N.; Ivanovski, S.; Poh, P.S.P. Scaffold geometry modulation of mechanotransduction and its influence on epigenetics. Acta Biomater. 2023, 163, 259–274. [Google Scholar] [CrossRef]
  28. Swanson, W.B.; Omi, M.; Zhang, Z.; Nam, H.K.; Jung, Y.; Wang, G.; Ma, P.X.; Hatch, N.E.; Mishina, Y. Macropore design of tissue engineering scaffolds regulates mesenchymal stem cell differentiation fate. Biomaterials 2021, 272, 120769. [Google Scholar] [CrossRef] [PubMed]
  29. Koushik, T.M.; Miller, C.M.; Antunes, E. Bone Tissue Engineering Scaffolds: Function of Multi-Material Hierarchically Structured Scaffolds. Adv. Healthc. Mater. 2023, 12, e2202766. [Google Scholar]
  30. Muzzio, N.; Moya, S.; Romero, G. Multifunctional Scaffolds and Synergistic Strategies in Tissue Engineering and Regenerative Medicine. Pharmaceutics 2021, 13, 792. [Google Scholar] [CrossRef]
  31. Zhang, X.; Chen, X.; Hong, H.; Hu, R.; Liu, J.; Liu, C. Decellularized extracellular matrix scaffolds: Recent trends and emerging strategies in tissue engineering. Bioact. Mater. 2022, 10, 15–31. [Google Scholar] [CrossRef]
  32. Chu, K.-A.; Wang, S.-Y.; Yeh, C.-C.; Fu, T.-W.; Fu, Y.-Y.; Ko, T.-L.; Chiu, M.-M.; Chen, T.-H.; Tsai, P.-J.; Fu, Y.-S. Reversal of bleomycin-induced rat pulmonary fibrosis by a xenograft of human umbilical mesenchymal stem cells from Wharton’s jelly. Theranostics 2019, 9, 6646–6664. [Google Scholar] [CrossRef]
  33. Bailey, K.E.; Pino, C.; Lennon, M.L.; Lyons, A.; Jacot, J.G.; Lammers, S.R.; Konigshoff, M.; Magin, C.M. Embedding of Precision-Cut Lung Slices in Engineered Hydrogel Biomaterials Supports Extended Ex Vivo Culture. Am. J. Respir. Cell Mol. Biol. 2020, 62, 14–22. [Google Scholar] [CrossRef]
  34. Valdoz, J.C.; Franks, N.A.; Cribbs, C.G.; Jacobs, D.J.; Dodson, E.L.; Knight, C.J.; Poulson, P.D.; Garfield, S.R.; Johnson, B.C.; Hemeyer, B.M.; et al. Soluble ECM promotes organotypic formation in lung alveolar model. Biomaterials 2022, 283, 121464. [Google Scholar] [CrossRef]
  35. Monaghan-Benson, E.; Aureille, J.; Guilluy, C. ECM stiffness regulates lung fibroblast survival through RasGRF1-dependent signaling. J. Biol. Chem. 2025, 301, 108161. [Google Scholar] [CrossRef] [PubMed]
  36. Lampi, M.C.; Reinhart-King, C.A. Targeting extracellular matrix stiffness to attenuate disease: From molecular mechanisms to clinical trials. Sci. Transl. Med. 2018, 10, eaao0475. [Google Scholar] [CrossRef]
  37. Jandl, K.; Kwapiszewska, G. Stiffness of the Extracellular Matrix: A Regulator of Prostaglandins in Pulmonary Fibrosis? Am. J. Respir. Cell Mol. Biol. 2020, 63, 721–722. [Google Scholar] [CrossRef]
  38. 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] [PubMed]
  39. Wang, Y.; Zhang, R.; Qiao, Z.; Dou, B.; Xu, H.; Meng, F.; Huang, J. Polyacrylamide-Based Hydrogel with Biocompatibility and Tunable Stiffness for Three-Dimensional Cell Culture. ACS Appl. Bio Mater. 2025, 8, 2356–2364. [Google Scholar] [CrossRef] [PubMed]
  40. Chen, Y.; Xue, D.; Huang, D.; Li, X.; Duan, Y.; Chen, B. Biofabrication of Tunable 3D Hydrogel for Investigating the Matrix Stiffness Impact on Breast Cancer Chemotherapy Resistance. ACS Biomater. Sci. Eng. 2025, 11, 1417–1431. [Google Scholar] [CrossRef]
  41. Liu, Z.; Zhang, H.; Zhou, R.; Gao, H.; Wu, Y.; Wang, Y.; Wu, H.; Guan, C.; Wang, L.; Tang, L.; et al. Thermoplastic Elastomer-Reinforced Hydrogels with Excellent Mechanical Properties, Swelling Resistance, and Biocompatibility. Adv. Sci. 2025, 12, e2414339. [Google Scholar] [CrossRef] [PubMed]
  42. Chen, Z.; Ezzo, M.; Zondag, B.; Rakhshani, F.; Ma, Y.; Hinz, B.; Kumacheva, E. Intrafibrillar Crosslinking Enables Decoupling of Mechanical Properties and Structure of a Composite Fibrous Hydrogel. Adv. Mater. 2024, 36, e2305964. [Google Scholar] [CrossRef]
  43. Ibanez, R.I.R.; do Amaral, R.J.F.C.; Reis, R.L.; Marques, A.P.; Murphy, C.M.; O’Brien, F.J. 3D-Printed Gelatin Methacrylate Scaffolds with Controlled Architecture and Stiffness Modulate the Fibroblast Phenotype towards Dermal Regeneration. Polymers 2021, 13, 2510. [Google Scholar] [CrossRef]
  44. Farrell, E.; Aliabouzar, M.; Quesada, C.; Baker, B.M.; Franceschi, R.T.; Putnam, A.J.; Fabiilli, M.L. Spatiotemporal control of myofibroblast activation in acoustically-responsive scaffolds via ultrasound-induced matrix stiffening. Acta Biomater. 2022, 138, 133–143. [Google Scholar] [CrossRef] [PubMed]
  45. Ragazzini, S.; Scocozza, F.; Bernava, G.; Auricchio, F.; Colombo, G.I.; Barbuto, M.; Conti, M.; Pesce, M.; Garoffolo, G. Mechanosensor YAP cooperates with TGF- β 1 signaling to promote myofibroblast activation and matrix stiffening in a 3D model of human cardiac fibrosis. Acta Biomater. 2022, 152, 300–312. [Google Scholar] [CrossRef] [PubMed]
  46. Younesi, F.S.; Miller, A.E.; Barker, T.H.; Rossi, F.M.V.; Hinz, B. Fibroblast and myofibroblast activation in normal tissue repair and fibrosis. Nat. Rev. Mol. Cell Biol. 2024, 25, 617–638. [Google Scholar] [CrossRef]
  47. Tiskratok, W.; Chuinsiri, N.; Limraksasin, P.; Kyawsoewin, M.; Jitprasertwong, P. Extracellular Matrix Stiffness: Mechanotransduction and Mechanobiological Response-Driven Strategies for Biomedical Applications Targeting Fibroblast Inflammation. Polymers 2025, 17, 822. [Google Scholar] [CrossRef]
  48. Soliman, B.G.; Chin, I.L.; Li, Y.; Ishii, M.; Ho, M.H.; Doan, V.K.; Cox, T.R.; Wang, P.Y.; Lindberg, G.C.J.; Zhang, Y.S.; et al. Droplet-based microfluidics for engineering shape-controlled hydrogels with stiffness gradient. Biofabrication 2024, 16, 045026. [Google Scholar] [CrossRef]
  49. Kuzucu, M.; Vera, G.; Beaumont, M.; Fischer, S.; Wei, P.; Shastri, V.P.; Forget, A. Extrusion-Based 3D Bioprinting of Gradients of Stiffness, Cell Density, and Immobilized Peptide Using Thermogelling Hydrogels. ACS Biomater. Sci. Eng. 2021, 7, 2192–2197. [Google Scholar] [CrossRef]
  50. Kersey, A.L.; Cheng, D.Y.; Deo, K.A.; Dubell, C.R.; Wang, T.-C.; Jaiswal, M.K.; Kim, M.H.; Murali, A.; Hargett, S.E.; Mallick, S.; et al. Stiffness assisted cell-matrix remodeling trigger 3D mechanotransduction regulatory programs. Biomaterials 2024, 306, 122473. [Google Scholar] [CrossRef]
  51. Chu, G.; Yuan, Z.; Zhu, C.; Zhou, P.; Wang, H.; Zhang, W.; Cai, Y.; Zhu, X.; Yang, H.; Li, B. Substrate stiffness- and topography-dependent differentiation of annulus fibrosus-derived stem cells is regulated by Yes-associated protein. Acta Biomater. 2019, 92, 254–264. [Google Scholar]
  52. Zhang, X.; Cao, D.; Xu, L.; Xu, Y.; Gao, Z.; Pan, Y.; Jiang, M.; Wei, Y.; Wang, L.; Liao, Y.; et al. Harnessing matrix stiffness to engineer a bone marrow niche for hematopoietic stem cell rejuvenation. Cell Stem Cell 2023, 30, 378–395. [Google Scholar] [CrossRef]
  53. Chen, G.-L.; Li, J.-Y.; Chen, X.; Liu, J.-W.; Zhang, Q.; Liu, J.-Y.; Wen, J.; Wang, N.; Lei, M.; Wei, J.-P.; et al. Mechanosensitive channels TMEM63A and TMEM63B mediate lung inflation-induced surfactant secretion. J. Clin. Investig. 2024, 134, e174508. [Google Scholar] [CrossRef]
  54. Kumar, V.; Madhurakkat Perikamana, S.K.; Tata, A.; Hoque, J.; Gilpin, A.; Tata, P.R.; Varghese, S. An In Vitro Microfluidic Alveolus Model to Study Lung Biomechanics. Front. Bioeng. Biotechnol. 2022, 10, 848699. [Google Scholar] [CrossRef]
  55. Sharifpoor, S.; Simmons, C.A.; Labow, R.S.; Santerre, J.P. A study of vascular smooth muscle cell function under cyclic mechanical loading in a polyurethane scaffold with optimized porosity. Acta Biomater. 2010, 6, 4218–4228. [Google Scholar] [CrossRef]
  56. Sencadas, V.; Sadat, S.; Silva, D.M. Mechanical performance of elastomeric PGS scaffolds under dynamic conditions. J. Mech. Behav. Biomed. Mater. 2020, 102, 103474. [Google Scholar] [CrossRef] [PubMed]
  57. Geiger, B.; Boujemaa-Paterski, R.; Winograd-Katz, S.E.; Venghateri, J.B.; Chung, W.-L.; Medalia, O. The Actin Network Interfacing Diverse Integrin-Mediated Adhesions. Biomolecules 2023, 13, 294. [Google Scholar] [CrossRef] [PubMed]
  58. Zhang, Z.; Zhu, H.; Zhao, G.; Miao, Y.; Zhao, L.; Feng, J.; Zhang, H.; Miao, R.; Sun, L.; Gao, B.; et al. Programmable and Reversible Integrin-Mediated Cell Adhesion Reveals Hysteresis in Actin Kinetics that Alters Subsequent Mechanotransduction. Adv. Sci. 2023, 10, 2302421. [Google Scholar] [CrossRef] [PubMed]
  59. Katsuta, H.; Okuda, S.; Nagayama, K.; Machiyama, H.; Kidoaki, S.; Kato, M.; Sokabe, M.; Miyata, T.; Hirata, H. Actin crosslinking by α-actinin averts viscous dissipation of myosin force transmission in stress fibers. iScience 2023, 26, 106090. [Google Scholar] [CrossRef]
  60. Amiri, S.; Muresan, C.; Shang, X.; Huet-Calderwood, C.; Schwartz, M.A.; Calderwood, D.A.; Murrell, M. Intracellular tension sensor reveals mechanical anisotropy of the actin cytoskeleton. Nat. Commun. 2023, 14, 8011. [Google Scholar] [CrossRef]
  61. Yang, Y.; Santos, D.M.; Pantano, L.; Knipe, R.; Abe, E.; Logue, A.; Pronzati, G.; Black, K.E.; Spinney, J.J.; Giacona, F.; et al. Screening for Inhibitors of YAP Nuclear Localization Identifies Aurora Kinase A as a Modulator of Lung Fibrosis. Am. J. Respir. Cell Mol. Biol. 2022, 67, 36–49. [Google Scholar] [CrossRef] [PubMed]
  62. Blokland, K.E.C.; Nizamoglu, M.; Habibie, H.; Borghuis, T.; Schuliga, M.; Melgert, B.N.; Knight, D.A.; Brandsma, C.-A.; Pouwels, S.D.; Burgess, J.K. Substrate stiffness engineered to replicate disease conditions influence senescence and fibrotic responses in primary lung fibroblasts. Front. Pharmacol. 2022, 13, 989169. [Google Scholar] [CrossRef] [PubMed]
  63. Stancil, I.T.; Michalski, J.E.; Hennessy, C.E.; Hatakka, K.L.; Yang, I.V.; Kurche, J.S.; Rincon, M.; Schwartz, D.A. Interleukin-6-dependent epithelial fluidization initiates fibrotic lung remodeling. Sci. Transl. Med. 2022, 14, eabo5254. [Google Scholar] [CrossRef] [PubMed]
  64. Stancil, I.T.; Michalski, J.E.; Davis-Hall, D.; Chu, H.W.; Park, J.-A.; Magin, C.M.; Yang, I.V.; Smith, B.J.; Dobrinskikh, E.; Schwartz, D.A. Pulmonary fibrosis distal airway epithelia are dynamically and structurally dysfunctional. Nat. Commun. 2021, 12, 4566. [Google Scholar] [CrossRef]
  65. Papavassiliou, K.A.; Sofianidi, A.A.; Spiliopoulos, F.G.; Gogou, V.A.; Gargalionis, A.N.; Papavassiliou, A.G. YAP/TAZ Signaling in the Pathobiology of Pulmonary Fibrosis. Cells 2024, 13, 1519. [Google Scholar] [CrossRef]
  66. Digiovanni, G.T.; Han, W.; Sherrill, T.P.; Taylor, C.J.; Nichols, D.S.; Geis, N.M.; Singha, U.K.; Calvi, C.L.; McCall, A.S.; Dixon, M.M.; et al. Epithelial Yap/Taz are required for functional alveolar regeneration following acute lung injury. JCI Insight 2023, 8, e173374. [Google Scholar] [CrossRef]
  67. Haak, A.J.; Kostallari, E.; Sicard, D.; Ligresti, G.; Choi, K.M.; Caporarello, N.; Jones, D.L.; Tan, Q.; Meridew, J.; Espinosa, A.M.D.; et al. Selective YAP/TAZ inhibition in fibroblasts via dopamine receptor D1 agonism reverses fibrosis. Sci. Transl. Med. 2019, 11, eaau6296. [Google Scholar] [CrossRef]
  68. Xiao, J.; Ang, J.W.; Zhong, X.; Wong, D.C.P.; Thivakar, T.; Yow, I.; Lee, C.J.M.; Foo, R.S.Y.; Kanchanawong, P.; Low, B.C. Coordination of Focal Adhesion Nanoarchitecture and Dynamics in Mechanosensing for Cardiomyoblast Differentiation. ACS Appl. Mater. Interfaces 2025, 17, 4463–4479. [Google Scholar] [CrossRef]
  69. Bachmann, M.; Skripka, A.; Weissenbruch, K.; Wehrle-Haller, B.; Bastmeyer, M. Phosphorylated paxillin and phosphorylated FAK constitute subregions within focal adhesions. J. Cell Sci. 2022, 135, jcs258764. [Google Scholar] [CrossRef]
  70. Ripamonti, M.; Wehrle-Haller, B.; de Curtis, I. Paxillin: A Hub for Mechano-Transduction from the β3 Integrin-Talin-Kindlin Axis. Front. Cell Dev. Biol. 2022, 10, 852016. [Google Scholar] [CrossRef]
  71. Cruz-Soca, M.; Faundez-Contreras, J.; Cordova-Casanova, A.; Gallardo, F.S.; Bock-Pereda, A.; Chun, J.; Carlos Casar, J.; Brandan, E. Activation of skeletal muscle FAPs by LPA requires the Hippo signaling via the FAK pathway. Matrix Biol. 2023, 119, 57–81. [Google Scholar] [CrossRef]
  72. Ren, Z.; Pan, X.; Li, J.; Dong, X.; Tu, X.; Pan, L.L.; Sun, J. G protein coupled receptor 41 regulates fibroblast activation in pulmonary fibrosis via Gαi/o and downstream Smad2/3 and ERK1/2 phosphorylation. Pharmacol. Res. 2023, 191, 106754. [Google Scholar] [CrossRef] [PubMed]
  73. Yeung, V.; Sriram, S.; Tran, J.A.; Guo, X.; Hutcheon, A.E.K.; Zieske, J.D.; Karamichos, D.; Ciolino, J.B. FAK Inhibition Attenuates Corneal Fibroblast Differentiation In Vitro. Biomolecules 2021, 11, 1682. [Google Scholar] [CrossRef] [PubMed]
  74. Thannickal, V.J.; Lee, D.Y.; White, E.S.; Cui, Z.; Larios, J.M.; Chacon, R.; Horowitz, J.C.; Day, R.M.; Thomas, P.E. Myofibroblast differentiation by transforming growth factor-β1 is dependent on cell adhesion and integrin signaling via focal adhesion kinase. J. Biol. Chem. 2003, 278, 12384–12389. [Google Scholar] [CrossRef]
  75. Oakes, P.W.; Wagner, E.; Brand, C.A.; Probst, D.; Linke, M.; Schwarz, U.S.; Glotzer, M.; Gardel, M.L. Optogenetic control of RhoA reveals zyxin-mediated elasticity of stress fibres. Nat. Commun. 2017, 8, 15817. [Google Scholar] [CrossRef] [PubMed]
  76. Pardo-Pastor, C.; Rubio-Moscardo, F.; Vogel-Gonzalez, M.; Serra, S.A.; Afthinos, A.; Mrkonjic, S.; Destaing, O.; Abenza, J.F.; Fernandez-Fernandez, J.M.; Trepat, X.; et al. Piezo2 channel regulates RhoA and actin cytoskeleton to promote cell mechanobiological responses. Proc. Natl. Acad. Sci. USA 2018, 115, 1925–1930. [Google Scholar] [CrossRef]
  77. Jo, M.H.; Li, J.; Jaumouille, V.; Hao, Y.; Coppola, J.; Yan, J.; Waterman, C.M.; Springer, T.A.; Ha, T. Single-molecule characterization of subtype-specific β1 integrin mechanics. Nat. Commun. 2022, 13, 7471. [Google Scholar] [CrossRef]
  78. De Belly, H.; Yan, S.; da Rocha, H.B.; Ichbiah, S.; Town, J.P.; Zager, P.J.; Estrada, D.C.; Meyer, K.; Turlier, H.; Bustamante, C.; et al. Cell protrusions and contractions generate long-range membrane tension propagation. Cell 2023, 186, 3049–3061. [Google Scholar] [CrossRef]
  79. Fan, H.; Zhao, H.; Hou, Y.; Meng, D.; Jiang, J.; Lee, E.-B.; Fu, Y.; Zhang, X.; Chen, R.; Wang, Y. Heterogeneous focal adhesion cytoskeleton nanoarchitectures from microengineered interfacial curvature to oversee nuclear remodeling and mechanotransduction of mesenchymal stem cells. Cell. Mol. Biol. Lett. 2025, 30, 10. [Google Scholar] [CrossRef]
  80. Dupont, S.; Wickstrom, S.A. Mechanical regulation of chromatin and transcription. Nat. Rev. Genet. 2022, 23, 624–643. [Google Scholar] [CrossRef]
  81. Stephens, R.K.; Miroshnikova, Y.A. Nuclear periphery and its mechanical regulation in cell fate transitions. Curr. Opin. Struct. Biol. 2024, 87, 102867. [Google Scholar] [CrossRef]
  82. Tschumperlin, D.J.; Lagares, D. Mechano-therapeutics: Targeting Mechanical Signaling in Fibrosis and Tumor Stroma. Pharmacol. Ther. 2020, 212, 107575. [Google Scholar] [CrossRef] [PubMed]
  83. Khalil, H.; Kanisicak, O.; Prasad, V.; Correll, R.N.; Fu, X.; Schips, T.; Vagnozzi, R.J.; Liu, R.; Thanh, H.; Lee, S.-J.; et al. Fibroblast-specific TGF-β-Smad2/3 signaling underlies cardiac fibrosis. J. Clin. Investig. 2017, 127, 3770–3783. [Google Scholar] [CrossRef] [PubMed]
  84. Mason, D.E.; Collins, J.M.; Dawahare, J.H.; Trung Dung, N.; Lin, Y.; Voytik-Harbin, S.L.; Zorlutuna, P.; Yoder, M.C.; Boerckel, J.D. YAP and TAZ limit cytoskeletal and focal adhesion maturation to enable persistent cell motility. J. Cell Biol. 2019, 218, 1369–1389. [Google Scholar] [CrossRef] [PubMed]
  85. Dupont, S.; Morsut, L.; Aragona, M.; Enzo, E.; Giulitti, S.; Cordenonsi, M.; Zanconato, F.; Le Digabel, J.; Forcato, M.; Bicciato, S.; et al. Role of YAP/TAZ in mechanotransduction. Nature 2011, 474, 179–183. [Google Scholar] [CrossRef]
  86. Szeto, S.G.; Narimatsu, M.; Lu, M.; He, X.; Sidiqi, A.M.; Tolosa, M.F.; Chan, L.; De Freitas, K.; Bialik, J.F.; Majunnder, S.; et al. YAP/TAZ Are Mechanoregulators of TGF-β-Smad Signaling and Renal Fibrogenesis. J. Am. Soc. Nephrol. 2016, 27, 3117–3128. [Google Scholar] [CrossRef]
  87. Astudillo, P. Extracellular matrix stiffness and Wnt/β-catenin signaling in physiology and disease. Biochem. Soc. Trans. 2020, 48, 1187–1198. [Google Scholar] [CrossRef]
  88. 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]
  89. Dabaghi, M.; Singer, R.; Noble, A.; Arizpe Tafoya, A.V.; Gonzalez-Martinez, D.A.; Gupta, T.; Formosa-Dague, C.; Rosas, I.O.; Kolb, M.R.; Shargall, Y.; et al. Influence of lung extracellular matrix from non-IPF and IPF donors on primary human lung fibroblast biology. Biomater. Sci. 2025, 13, 1721–1741. [Google Scholar] [CrossRef]
  90. 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]
  91. Jandl, K.; Radic, N.; Zeder, K.; Kovacs, G.; Kwapiszewska, G. Pulmonary vascular fibrosis in pulmonary hypertension-The role of the extracellular matrix as a therapeutic target. Pharmacol. Ther. 2023, 247, 108438. [Google Scholar] [CrossRef] [PubMed]
  92. Matera, D.L.; DiLillo, K.M.; Smith, M.R.; Davidson, C.D.; Parikh, R.; Said, M.; Wilke, C.A.; Lombaert, I.M.; Arnold, K.B.; Moore, B.B.; et al. Microengineered 3D pulmonary interstitial mimetics highlight a critical role for matrix degradation in myofibroblast differentiation. Sci. Adv. 2020, 6, eabb5069. [Google Scholar] [CrossRef]
  93. Yanagihara, T.; Guignabert, C.; Kolb, M.R.J. Endothelial cells in pulmonary fibrosis: More than a bystander. Eur. Respir. J. 2023, 61, 2300407. [Google Scholar] [CrossRef]
  94. Balestrini, J.L.; Chaudhry, S.; Sarrazy, V.; Koehler, A.; Hinz, B. The mechanical memory of lung myofibroblasts. Integr. Biol. 2012, 4, 410–421. [Google Scholar] [CrossRef]
  95. Barcena-Varela, M.; Paish, H.; Alvarez, L.; Uriarte, I.; Latasa, M.U.; Santamaria, E.; Recalde, M.; Garate, M.; Claveria, A.; Colyn, L.; et al. Epigenetic mechanisms and metabolic reprogramming in fibrogenesis: Dual targeting of G9a and DNMT1 for the inhibition of liver fibrosis. Gut 2021, 70, 388–400. [Google Scholar] [CrossRef] [PubMed]
  96. Park, S.-M.; Lee, J.-H.; Ahn, K.S.; Shim, H.W.; Yoon, J.-Y.; Hyun, J.; Lee, J.H.; Jang, S.; Yoo, K.H.; Jang, Y.-K.; et al. Cyclic Stretch Promotes Cellular Reprogramming Process through Cytoskeletal-Nuclear Mechano-Coupling and Epigenetic Modification. Adv. Sci. 2023, 10, e2303395. [Google Scholar] [CrossRef] [PubMed]
  97. Barcena-Varela, M.; Caruso, S.; Llerena, S.; Alvarez-Sola, G.; Uriarte, I.; Latasa, M.U.; Urtasun, R.; Rebouissou, S.; Alvarez, L.; Jimenez, M.; et al. Dual Targeting of Histone Methyltransferase G9a and DNA-Methyltransferase 1 for the Treatment of Experimental Hepatocellular Carcinoma. Hepatology 2019, 69, 587–603. [Google Scholar] [CrossRef] [PubMed]
  98. Chen, L.-J.; Li, J.Y.-S.; Nguyen, P.; Norwich, G.; Wang, Y.; Teng, D.; Shiu, Y.-T.; Shyy, J.Y.J.; Chien, S. Pulsatile flow induces chromatin interaction with lamin-associated proteins to enrich H3K9 methylation in endothelial cells. Proc. Natl. Acad. Sci. USA 2025, 122, e2424566122. [Google Scholar] [CrossRef]
  99. Song, Y.; Soto, J.; Li, S. Mechanical regulation of histone modifications and cell plasticity. Curr. Opin. Solid State Mater. Sci. 2020, 24, 100872. [Google Scholar] [CrossRef]
  100. Alisafaei, F.; Jokhun, D.S.; Shivashankar, G.V.; Shenoy, V.B. Regulation of nuclear architecture, mechanics, and nucleocytoplasmic shuttling of epigenetic factors by cell geometric constraints. Proc. Natl. Acad. Sci. USA 2019, 116, 13200–13209. [Google Scholar] [CrossRef]
  101. Habermann, A.C.; Gutierrez, A.J.; Bui, L.T.; Yahn, S.L.; Winters, N.I.; Calvi, C.L.; Peter, L.; Chung, M.-I.; Taylor, C.J.; Jetter, C.; et al. Single-cell RNA sequencing reveals profibrotic roles of distinct epithelial and mesenchymal lineages in pulmonary fibrosis. Sci. Adv. 2020, 6, eaba1972. [Google Scholar] [CrossRef] [PubMed]
  102. Zhao, X.-B.; Chen, Y.-P.; Tan, M.; Zhao, L.; Zhai, Y.-Y.; Sun, Y.-L.; Gong, Y.; Feng, X.-Q.; Du, J.; Fan, Y.-B. Extracellular Matrix Stiffness Regulates DNA Methylatioan by PKCα-Dependent Nuclear Transport of DNMT3L. Adv. Healthc. Mater. 2021, 10, 2100821. [Google Scholar] [CrossRef]
  103. Yang, S.; Sun, Y.; Luo, Y.; Liu, Y.; Jiang, M.; Li, J.; Zhang, Q.; Bai, J. Hypermethylation of PPARG-encoding gene promoter mediates fine particulate matter-induced pulmonary fibrosis by regulating the HMGB1/NLRP3 axis. Ecotoxicol. Environ. Saf. 2024, 272, 116068. [Google Scholar] [CrossRef]
  104. Ting, L.; Feng, Y.; Zhou, Y.; Tong, Z.; Dong, Z. IL-27 induces autophagy through regulation of the DNMT1/lncRNA MEG3/ERK/p38 axis to reduce pulmonary fibrosis. Respir. Res. 2023, 24, 67. [Google Scholar] [CrossRef]
  105. Chen, S.; Wang, Y.; Lai, J.; Tan, S.; Wang, M. Structure and Properties of Gelatin Methacryloyl (GelMA) Synthesized in Different Reaction Systems. Biomacromolecules 2023, 24, 2928–2941. [Google Scholar] [CrossRef]
  106. Wang, Y.; Cao, Z.; Wei, Q.; Ma, K.; Hu, W.; Huang, Q.; Su, J.; Li, H.; Zhang, C.; Fu, X. VH298-loaded extracellular vesicles released from gelatin methacryloyl hydrogel facilitate diabetic wound healing by HIF-1a-me diate d enhancement of angiogenesis. Acta Biomater. 2022, 147, 342–355. [Google Scholar] [CrossRef] [PubMed]
  107. Mamidi, N.; Velasco Delgadillo, R.M.; Barrera, E.V. Covalently Functionalized Carbon Nano-Onions Integrated Gelatin Methacryloyl Nanocomposite Hydrogel Containing γ-Cyclodextrin as Drug Carrier for High-Performance pH-Triggered Drug Release. Pharmaceuticals 2021, 14, 291. [Google Scholar] [CrossRef] [PubMed]
  108. Zhang, Z.L.; Wang, J.; Liu, F.; Yuan, L.; Ding, M.J.; Chen, L.D.; Yuan, J.L.; Yang, K.; Qian, J.; Lu, W.J. Non-inflammatory emphysema induced by NO2 chronic exposure and intervention with demethylation 5-Azacytidine. Life Sci. 2019, 221, 121–129. [Google Scholar] [CrossRef]
  109. Brembilla, N.-C.; El-Harane, S.; Durual, S.; Krause, K.-H.; Preynat-Seauve, O. Adipose-Derived Stromal Cells Exposed to RGD Motifs Enter an Angiogenic Stage Regulating Endothelial Cells. Int. J. Mol. Sci. 2025, 26, 867. [Google Scholar] [CrossRef]
  110. Kim, H.J.; You, S.J.; Yang, D.H.; Chun, H.J.; Kim, M.S. Preparation of novel RGD-conjugated thermosensitive mPEG-PCL composite hydrogels and in vitro investigation of their impacts on adhesion-dependent cellular behavior. J. Ind. Eng. Chem. 2020, 84, 226–235. [Google Scholar] [CrossRef]
  111. Avsec, Z.; Weilert, M.; Shrikumar, A.; Krueger, S.; Alexandari, A.; Dalal, K.; Fropf, R.; McAnany, C.; Gagneur, J.; Kundaje, A.; et al. Base-resolution models of transcription-factor binding reveal soft motif syntax. Nat. Genet. 2021, 53, 354–366. [Google Scholar] [CrossRef]
  112. Ruan, H.; Hu, Q.; Wen, D.; Chen, Q.; Chen, G.; Lu, Y.; Wang, J.; Cheng, H.; Lu, W.; Gu, Z. A Dual-Bioresponsive Drug-Delivery Depot for Combination of Epigenetic Modulation and Immune Checkpoint Blockade. Adv. Mater. 2019, 31, e1806957. [Google Scholar] [CrossRef]
  113. Gong, Y.; Chen, W.; Chen, X.; He, Y.; Jiang, H.; Zhang, X.; Pan, L.; Ni, B.; Yang, F.; Xu, Y.; et al. An Injectable Epigenetic Autophagic Modulatory Hydrogel for Boosting Umbilical Cord Blood NK Cell Therapy Prevents Postsurgical Relapse of Triple-Negative Breast Cancer. Adv. Sci. 2022, 9, e2201271. [Google Scholar] [CrossRef]
  114. Chakraborty, A.; Pacelli, S.; Alexander, S.; Huayamares, S.; Rosenkrans, Z.; Vergel, F.E.; Wu, Y.; Chakravorty, A.; Paul, A. Nanoparticle-Reinforced Tough Hydrogel as a Versatile Platform for Pharmaceutical Drug Delivery: Preparation and in Vitro Characterization. Mol. Pharm. 2023, 20, 767–774. [Google Scholar] [CrossRef]
  115. Zhu, S.; Yu, C.; Liu, N.; Zhao, M.; Chen, Z.; Liu, J.; Li, G.; Huang, H.; Guo, H.; Sun, T.; et al. Injectable conductive gelatin methacrylate/oxidized dextran hydrogel encapsulating umbilical cord mesenchymal stem cells for myocardial infarction treatment. Bioact. Mater. 2022, 13, 119–134. [Google Scholar] [CrossRef] [PubMed]
  116. Jiang, X.; Li, D.; Tassey, J.; Li, J.; Liu, J.; Li, G.; Sun, Y.; Zhao, X.; Wang, T.; Zhang, Y.; et al. Complex hydrogel for cartilage regeneration and anti-inflammation. Compos. Part B Eng. 2024, 280, 111481. [Google Scholar] [CrossRef]
  117. Saraswathibhatla, A.; Indana, D.; Chaudhuri, O. Cell-extracellular matrix mechanotransduction in 3D. Nat. Rev. Mol. Cell Biol. 2023, 24, 495–516. [Google Scholar] [CrossRef]
  118. Zhang, Q.; Li, Y.; Lin, Z.Y.; Wong, K.K.Y.; Lin, M.; Yildirimer, L.; Zhao, X. Electrospun polymeric micro/nanofibrous scaffolds for long-term drug release and their biomedical applications. Drug Discov. Today 2017, 22, 1351–1366. [Google Scholar] [CrossRef]
  119. Xing, L.; Chang, X.; Shen, L.; Zhang, C.; Fan, Y.; Cho, C.; Zhang, Z.; Jiang, H. Progress in drug delivery system for fibrosis therapy. Asian J. Pharm. Sci. 2021, 16, 47–61. [Google Scholar] [CrossRef] [PubMed]
  120. Liu, C.; Wang, Z.; Wei, X.; Chen, B.; Luo, Y. 3D printed hydrogel/PCL core/shell fiber scaffolds with NIR-triggered drug release for cancer therapy and wound healing. Acta Biomater. 2021, 131, 314–325. [Google Scholar] [CrossRef]
  121. Miszuk, J.; Liang, Z.; Hu, J.; Sanyour, H.; Hong, Z.; Fong, H.; Sun, H. Elastic Mineralized 3D Electrospun PCL Nanofibrous Scaffold for Drug Release and Bone Tissue Engineering. ACS Appl. Bio Mater. 2021, 4, 3639–3648. [Google Scholar] [CrossRef] [PubMed]
  122. Li, Z.; Tian, C.; Jiao, D.; Li, J.; Li, Y.; Zhou, X.; Zhao, H.; Zhao, Y.; Han, X. Synergistic effects of silver nanoparticles and cisplatin in combating inflammation and hyperplasia of airway stents. Bioact. Mater. 2022, 9, 266–280. [Google Scholar] [CrossRef] [PubMed]
  123. Li, Z.; Zhang, W.; Jiao, D.; Tian, C.; Xu, K.; Zhu, H.; Han, X. All-in-one properties of an anticancer-covered airway stent for the prevention of malignant central airway obstruction. APL Bioeng. 2023, 7, 036116. [Google Scholar] [CrossRef]
  124. Sun, L.; Li, J.; Gao, W.; Shi, M.; Tang, F.; Fu, X.; Chen, X. Coaxial nanofibrous scaffolds mimicking the extracellular matrix transition in the wound healing process promoting skin regeneration through enhancing immunomodulation. J. Mater. Chem. B 2021, 9, 1395–1405. [Google Scholar] [CrossRef]
  125. Ghorbani, M.; Nezhad-Mokhtari, P.; Ramazani, S. Aloe vera-loaded nanofibrous scaffold based on Zein/Polycaprolactone/Collagen for wound healing. Int. J. Biol. Macromol. 2020, 153, 921–930. [Google Scholar] [CrossRef] [PubMed]
  126. Mancino, S.; Boraso, M.; Galmozzi, A.; Serafini, M.M.; De Fabiani, E.; Crestani, M.; Viviani, B. Dose-dependent dual effects of HDAC inhibitors on glial inflammatory response. Sci. Rep. 2025, 15, 12262. [Google Scholar] [CrossRef]
  127. Dushanan, R.; Weerasinghe, S.; Dissanayake, D.P.; Senthilnithy, R. Driving the new generation histone deacetylase inhibitors in cancer therapy; manipulation of the histone abbreviation at the epigenetic level: An in-silico approach. Can. J. Chem. 2022, 100, 880–890. [Google Scholar] [CrossRef]
  128. Hebbel, R.P.; Vercellotti, G.M.; Pace, B.S.; Solovey, A.N.; Kollander, R.; Abanonu, C.F.; Nguyen, J.; Vineyard, J.V.; Belcher, J.D.; Abdulla, F.; et al. The HDAC inhibitors trichostatin A and suberoylanilide hydroxamic acid exhibit multiple modalities of benefit for the vascular pathobiology of sickle transgenic mice. Blood 2010, 115, 2483–2490. [Google Scholar] [CrossRef]
  129. Hassan, A.A.; Radwan, H.A.; Abdelaal, S.A.; Al-Radadi, N.S.; Ahmed, M.K.; Shoueir, K.R.; Hady, M.A. Polycaprolactone based electrospun matrices loaded with Ag/hydroxyapatite as wound dressings: Morphology, cell adhesion, and antibacterial activity. Int. J. Pharm. 2021, 593, 120143. [Google Scholar] [CrossRef]
  130. Aly, A.A.; Ahmed, M.K. Fibrous scaffolds of Ag/Fe co-doped hydroxyapatite encapsulated into polycaprolactone: Morphology, mechanical and in vitro cell adhesion. Int. J. Pharm. 2021, 601, 120557. [Google Scholar] [CrossRef]
  131. Baltes, F.; Caspers, J.; Henze, S.; Schlesinger, M.; Bendas, G. Targeting Discoidin Domain Receptor 1 (DDR1) Signaling and Its Crosstalk with β1-Integrin Emerges as a Key Factor for Breast Cancer Chemosensitization upon Collagen Type 1 Binding. Int. J. Mol. Sci. 2020, 21, 4956. [Google Scholar] [CrossRef]
  132. Sasaki, K.; Ishihara, J.; Ishihara, A.; Miura, R.; Mansurov, A.; Fukunaga, K.; Hubbell, J.A. Engineered collagen-binding serum albumin as a drug conjugate carrier for cancer therapy. Sci. Adv. 2019, 5, eaaw6081. [Google Scholar] [CrossRef] [PubMed]
  133. Tu, Z.; Han, F.; Zhu, Z.; Yu, Q.; Liu, C.; Bao, Y.; Li, B.; Zhou, F. Sustained release of basic fibroblast growth factor in micro/nanofibrous scaffolds promotes annulus fibrosus regeneration. Acta Biomater. 2023, 166, 241–253. [Google Scholar] [CrossRef] [PubMed]
  134. Qian, M.; Li, S.; Xi, K.; Tang, J.; Shen, X.; Liu, Y.; Guo, R.; Zhang, N.; Gu, Y.; Xu, Y.; et al. ECM-engineered electrospun fibers with an immune cascade effect for inhibiting tissue fibrosis. Acta Biomater. 2023, 171, 308–326. [Google Scholar] [CrossRef] [PubMed]
  135. Ho, T.-C.; Kim, H.S.; Chen, Y.; Li, Y.; LaMere, M.W.; Chen, C.; Wang, H.; Gong, J.; Palumbo, C.D.; Ashton, J.M.; et al. Scaffold-mediated CRISPR-Cas9 delivery system for acute myeloid leukemia therapy. Sci. Adv. 2021, 7, eabg3217. [Google Scholar] [CrossRef]
  136. Gao, Y.; Wang, K.; Wu, S.; Wu, J.; Zhang, J.; Li, J.; Lei, S.; Duan, X.; Men, K. Injectable and Photocurable Gene Scaffold Facilitates Efficient Repair of Spinal Cord Injury. ACS Appl. Mater. Interfaces 2024, 16, 4375–4394. [Google Scholar] [CrossRef]
  137. Seasock, M.J.; Shafiquzzaman, M.; Ruiz-Echartea, M.E.; Kanchi, R.S.; Tran, B.T.; Simon, L.M.; Meyer, M.D.; Erice, P.A.; Lotlikar, S.L.; Wenlock, S.C.; et al. Let-7 restrains an oncogenic circuit in AT2 cells to prevent fibrogenic cell intermediates in pulmonary fibrosis. bioRxiv 2025. [Google Scholar] [CrossRef]
  138. Elliot, S.; Periera-Simon, S.; Xia, X.; Catanuto, P.; Rubio, G.; Shahzeidi, S.; El Salem, F.; Shapiro, J.; Briegel, K.; Korach, K.S.; et al. MicroRNA let-7 Downregulates Ligand-Independent Estrogen Receptor-mediated Male-Predominant Pulmonary Fibrosis. Am. J. Respir. Crit. Care Med. 2019, 200, 1246–1257. [Google Scholar] [CrossRef]
  139. Chelladurai, P.; Kuenne, C.; Bourgeois, A.; Guenther, S.; Valasarajan, C.; Cherian, A.V.; Rottier, R.J.; Romanet, C.; Weigert, A.; Boucherat, O.; et al. Epigenetic reactivation of transcriptional programs orchestrating fetal lung development in human pulmonary hypertension. Sci. Transl. Med. 2022, 14, eabe5407. [Google Scholar] [CrossRef]
  140. Jolly, A.J.; Lu, S.; Dubner, A.M.; Strand, K.A.; Mutryn, M.F.; Pilotti-Riley, A.; Danis, E.P.; Nemenoff, R.A.; Moulton, K.S.; Majesky, M.W.; et al. Redistribution of the chromatin remodeler Brg1 directs smooth muscle-derived adventitial progenitor-to-myofibroblast differentiation and vascular fibrosis. JCI Insight 2023, 8, e164862. [Google Scholar] [CrossRef]
  141. Sha, J.-M.; Zhang, R.-Q.; Wang, X.-C.; Zhou, Y.; Song, K.; Sun, H.; Tu, B.; Tao, H. Epigenetic reader MeCP2 repressed WIF1 boosts lung fibroblast proliferation, migration and pulmonary fibrosis. Toxicol. Lett. 2023, 381, 1–12. [Google Scholar] [CrossRef] [PubMed]
  142. Mann, J.; Oakley, F.; Akiboye, F.; Elsharkawy, A.; Thorne, A.W.; Mann, D.A. Regulation of myofibroblast transdifferentiation by DNA methylation and MeCP2: Implications for wound healing and fibrogenesis. Cell Death Differ. 2007, 14, 275–285. [Google Scholar] [CrossRef]
  143. Pinese, C.; Lin, J.; Milbreta, U.; Li, M.; Wang, Y.; Leong, K.W.; Chew, S.Y. Sustained delivery of siRNA/mesoporous silica nanoparticle complexes from nanofiber scaffolds for long-term gene silencing. Acta Biomater. 2018, 76, 164–177. [Google Scholar] [CrossRef] [PubMed]
  144. Ding, L.; Tang, S.; Tang, W.; Mosley, D.D.; Yu, A.; Sil, D.; Romanova, S.; Bailey, K.L.; Knoell, D.L.; Wyatt, T.A.; et al. Perfluorocarbon Nanoemulsions Enhance Therapeutic siRNA Delivery in the Treatment of Pulmonary Fibrosis. Adv. Sci. 2022, 9, 2103676. [Google Scholar] [CrossRef]
  145. Qu, D.; Zhu, J.P.; Childs, H.R.; Lu, H.H. Nanofiber-based transforming growth factor-β3 release induces fibrochondrogenic differentiation of stem cells. Acta Biomater. 2019, 93, 111–122. [Google Scholar] [CrossRef]
  146. Wang, Z.; Song, X.; Cui, Y.; Cheng, K.; Tian, X.; Dong, M.; Liu, L. Silk fibroin H-fibroin/poly(ε-caprolactone) core-shell nanofibers with enhanced mechanical property and long-term drug release. J. Colloid Interface Sci. 2021, 593, 142–151. [Google Scholar] [CrossRef]
  147. Killaars, A.R.; Grim, J.C.; Walker, C.J.; Hushka, E.A.; Brown, T.E.; Anseth, K.S. Extended Exposure to Stiff Microenvironments Leads to Persistent Chromatin Remodeling in Human Mesenchymal Stem Cells. Adv. Sci. 2019, 6, 1801483. [Google Scholar] [CrossRef]
  148. Ma, H.; Zhang, T. Histone demethylase KDM3B mediates matrix stiffness-induced osteogenic differentiation of adipose-derived stem cells. Arch. Biochem. Biophys. 2024, 757, 110028. [Google Scholar] [CrossRef]
  149. Muniyandi, P.; Palaninathan, V.; Mizuki, T.; Mohamed, M.S.; Hanajiri, T.; Maekawa, T. Scaffold mediated delivery of dual miRNAs to transdifferentiate cardiac fibroblasts. Mater. Sci. Eng. C 2021, 128, 112323. [Google Scholar] [CrossRef]
  150. Yuan, T.; Wu, M.; Zhu, C.; Yu, H.; Pham, M.D.; Bottermann, K.; Mao, Y.; Wang, Y.; Langner, M.; Peitzsch, M.; et al. Targeting miRNA-1a and miRNA-15b: A Novel Combinatorial Strategy to Drive Adult Cardiac Regeneration. Adv. Sci. 2025, 12, e2414455. [Google Scholar] [CrossRef] [PubMed]
  151. Wang, Q.; Song, Y.; Chen, J.; Li, Q.; Gao, J.; Tan, H.; Zhu, Y.; Wang, Z.; Li, M.; Yang, H.; et al. Direct in vivo reprogramming with non-viral sequential targeting nanoparticles promotes cardiac regeneration. Biomaterials 2021, 276, 121028. [Google Scholar] [CrossRef]
  152. Acharya, N.; Kandel, R.; Roy, P.; Warraich, I.; Singh, K.P. Epigenetic therapeutics attenuate kidney injury and fibrosis by restoring the expression of epigenetically reprogrammed fibrogenic genes and signaling pathways. Eur. J. Pharm. Sci. 2025, 204, 106977. [Google Scholar] [CrossRef] [PubMed]
  153. Xiao, X.; Wang, W.; Guo, C.; Wu, J.; Zhang, S.; Shi, H.; Kwon, S.; Chen, J.; Dong, Z. Hypermethylation leads to the loss of HOXA5, resulting in JAG1 expression and NOTCH signaling contributing to kidney fibrosis. Kidney Int. 2024, 106, 98–114. [Google Scholar] [CrossRef] [PubMed]
  154. Blagitko-Dorfs, N.; Schlosser, P.; Greve, G.; Pfeifer, D.; Meier, R.; Baude, A.; Brocks, D.; Plass, C.; Luebbert, M. Combination treatment of acute myeloid leukemia cells with DNMT and HDAC inhibitors: Predominant synergistic gene downregulation associated with gene body demethylation. Leukemia 2019, 33, 945–956. [Google Scholar] [CrossRef] [PubMed]
  155. Brocks, D.; Schmidt, C.R.; Daskalakis, M.; Jang, H.S.; Shah, N.M.; Li, D.; Li, J.; Zhang, B.; Hou, Y.; Laudato, S.; et al. DNMT and HDAC inhibitors induce cryptic transcription start sites encoded in long terminal repeats. Nat. Genet. 2017, 49, 1052–1060. [Google Scholar] [CrossRef] [PubMed]
  156. Yuan, Z.; Sun, Q.; Li, D.; Miao, S.; Chen, S.; Song, L.; Gao, C.; Chen, Y.; Tan, C.; Jiang, Y. Design, synthesis and anticancer potential of NSC-319745 hydroxamic acid derivatives as DNMT and HDAC inhibitors. Eur. J. Med. Chem. 2017, 134, 281–292. [Google Scholar] [CrossRef]
  157. Wang, Y.; Bruggeman, K.F.; Franks, S.; Gautam, V.; Hodgetts, S.I.; Harvey, A.R.; Williams, R.J.; Nisbet, D.R. Is Viral Vector Gene Delivery More Effective Using Biomaterials? Adv. Healthc. Mater. 2021, 10, e2001238. [Google Scholar] [CrossRef]
  158. Xiang, Y.; Zhong, Z.; Yao, E.J.; Kiratitanaporn, W.; Suy, M.T.; Chen, S. 3D bioprinting of gene delivery scaffolds with controlled release. Bioprinting 2023, 31, e00270. [Google Scholar] [CrossRef]
  159. Eltaher, H.M.; Yang, J.; Shakesheff, K.M.; Dixon, J.E. Highly efficient intracellular transduction in three-dimensional gradients for programming cell fate. Acta Biomater. 2016, 41, 181–192. [Google Scholar] [CrossRef]
  160. Santorelli, M.; Bhamidipati, P.S.; Courte, J.; Swedlund, B.; Jain, N.; Poon, K.; Schildknecht, D.; Kavanagh, A.; Mackrell, V.A.; Sondkar, T.; et al. Control of spatio-temporal patterning via cell growth in a multicellular synthetic gene circuit. Nat. Commun. 2024, 15, 9867. [Google Scholar] [CrossRef]
  161. Chan, C.J.; Heisenberg, C.-P.; Hiiragi, T. Coordination of Morphogenesis and Cell-Fate Specification in Development. Curr. Biol. 2017, 27, R1024–R1035. [Google Scholar] [CrossRef] [PubMed]
  162. Kolb, P.; Upagupta, C.; Vierhout, M.; Ayaub, E.; Bellaye, P.S.; Gauldie, J.; Shimbori, C.; Inman, M.; Ask, K.; Kolb, M.R.J. The importance of interventional timing in the bleomycin model of pulmonary fibrosis. Eur. Respir. J. 2020, 55, 1901105. [Google Scholar] [CrossRef]
  163. Zheng, Q.; Tong, M.; Ou, B.; Liu, C.; Hu, C.; Yang, Y. Isorhamnetin protects against bleomycin-induced pulmonary fibrosis by inhibiting endoplasmic reticulum stress and epithelial-mesenchymal transition. Int. J. Mol. Med. 2019, 43, 117–126. [Google Scholar] [CrossRef]
  164. Uhl, F.E.; Zhang, F.; Pouliot, R.A.; Uriarte, J.J.; Enes, S.R.; Han, X.; Ouyang, Y.; Xia, K.; Westergren-Thorsson, G.; Malmstrom, A.; et al. Functional role of glycosaminoglycans in decellularized lung extracellular matrix. Acta Biomater. 2020, 102, 231–246. [Google Scholar] [CrossRef]
  165. Antczak, L.-A.M.; Moore, K.N.; Hendrick, T.E.; Heise, R.L. Binary fabrication of decellularized lung extracellular matrix hybridgels for in vitro chronic obstructive pulmonary disease modeling. Acta Biomater. 2024, 185, 190–202. [Google Scholar] [CrossRef] [PubMed]
  166. Balestrini, J.L.; Gard, A.L.; Gerhold, K.A.; Wilcox, E.C.; Liu, A.; Schwan, J.; Le, A.V.; Baevova, P.; Dimitrievska, S.; Zhao, L.; et al. Comparative biology of decellularized lung matrix: Implications of species mismatch in regenerative medicine. Biomaterials 2016, 102, 220–230. [Google Scholar] [CrossRef] [PubMed]
  167. Zhang, Y.; Zhang, C.; Li, Y.; Zhou, L.; Dan, N.; Min, J.; Chen, Y.; Wang, Y. Evolution of biomimetic ECM scaffolds from decellularized tissue matrix for tissue engineering: A comprehensive review. Int. J. Biol. Macromol. 2023, 246, 125672. [Google Scholar] [CrossRef]
  168. Bilodeau, C.; Shojaie, S.; Goltsis, O.; Wang, J.; Luo, D.; Ackerley, C.; Rogers, I.; Cox, B.; Post, M. TP63 basal cells are indispensable during endoderm differentiation into proximal airway cells on acellular lung scaffolds. NPJ Regen. Med. 2021, 6, 12. [Google Scholar] [CrossRef]
  169. Evangelista-Leite, D.; Carreira, A.C.O.; Nishiyama, M.Y.; Gilpin, S.E.; Miglino, M.A. The molecular mechanisms of extracellular matrix-derived hydrogel therapy in idiopathic pulmonary fibrosis models. Biomaterials 2023, 302, 122338. [Google Scholar] [CrossRef]
  170. Savitri, C.; Ha, S.S.; Kwon, J.W.; Kim, S.H.; Kim, Y.-M.; Park, H.M.; Kwon, H.; Ji, M.J.; Park, K. Human Fibroblast-Derived Matrix Hydrogel Accelerates Regenerative Wound Remodeling Through the Interactions with Macrophages. Adv. Sci. 2024, 11, e2305852. [Google Scholar] [CrossRef]
  171. Zhou, Z.; Bu, Z.; Wang, S.; Yu, J.; Liu, W.; Huang, J.; Hu, J.; Xu, S.; Wu, P. Extracellular matrix hydrogels with fibroblast growth factor 2 containing exosomes for reconstructing skin microstructures. J. Nanobiotechnol. 2024, 22, 438. [Google Scholar] [CrossRef]
  172. Rosin, N.L.; Winstone, T.M.L.; Kelley, M.; Biernaskie, J.; Dufour, A.; Orton, D.J. Targeted proteomic approach for quantification of collagen type I and type III in formalin-fixed paraffin-embedded tissue. Sci. Rep. 2024, 14, 17769. [Google Scholar] [CrossRef]
  173. Lipton, A.; Leitzel, K.; Ali, S.M.; Polimera, H.V.; Nagabhairu, V.; Marks, E.; Richardson, A.E.; Krecko, L.; Ali, A.; Koestler, W.; et al. High turnover of extracellular matrix reflected by specific protein fragments measured in serum is associated with poor outcomes in two metastatic breast cancer cohorts. Int. J. Cancer 2018, 143, 3027–3034. [Google Scholar] [CrossRef]
  174. Milman Krentsis, I.; Zheng, Y.; Rosen, C.; Shin, S.Y.; Blagdon, C.; Shoshan, E.; Qi, Y.; Wang, J.; Yadav, S.K.; Bachar Lustig, E.; et al. Lung cell transplantation for pulmonary fibrosis. Sci. Adv. 2024, 10, eadk2524. [Google Scholar] [CrossRef] [PubMed]
  175. Bai, X.; Zhao, G.; Chen, Q.; Li, Z.; Gao, M.; Ho, W.; Xu, X.; Zhang, X.-Q. Inhaled siRNA nanoparticles targeting IL11 inhibit lung fibrosis and improve pulmonary function post-bleomycin challenge. Sci. Adv. 2022, 8, eabn7162. [Google Scholar] [CrossRef]
  176. Duran, P.; Sesillo, F.B.; Cook, M.; Burnett, L.; Menefee, S.A.; Do, E.; French, S.; Zazueta-Damian, G.; Dzieciatkowska, M.; Saviola, A.J.; et al. Proregenerative extracellular matrix hydrogel mitigates pathological alterations of pelvic skeletal muscles after birth injury. Sci. Transl. Med. 2023, 15, eabj3138. [Google Scholar] [CrossRef] [PubMed]
  177. Enomoto, Y.; Katsura, H.; Fujimura, T.; Ogata, A.; Baba, S.; Yamaoka, A.; Kihara, M.; Abe, T.; Nishimura, O.; Kadota, M.; et al. Autocrine TGF-β-positive feedback in profibrotic AT2-lineage cells plays a crucial role in non-inflammatory lung fibrogenesis. Nat. Commun. 2023, 14, 4956. [Google Scholar] [CrossRef] [PubMed]
  178. Mascharak, S.; Guo, J.L.; Griffin, M.; Berry, C.E.; Wan, D.C.; Longaker, M.T. Modelling and targeting mechanical forces in organ fibrosis. Nat. Rev. Bioeng. 2024, 2, 305–323. [Google Scholar] [CrossRef]
  179. Jones, D.L.; Hallstroem, G.F.; Jiang, X.; Locke, R.C.; Evans, M.K.; Bonnevie, E.D.; Srikumar, A.; Leahy, T.P.; Nijsure, M.P.; Boerckel, J.D.; et al. Mechanoepigenetic regulation of extracellular matrix homeostasis via Yap and Taz. Proc. Natl. Acad. Sci. USA 2023, 120, e2211947120. [Google Scholar] [CrossRef]
  180. Wagner, D.E.; Alsafadi, H.N.; Mitash, N.; Justet, A.; Hu, Q.; Pineda, R.; Staab-Weijnitz, C.; Korfei, M.; Gvazava, N.; Wannemo, K.; et al. Inhibition of epithelial cell YAP-TEAD/LOX signaling attenuates pulmonary fibrosis in preclinical models. Nat. Commun. 2025, 16, 7099. [Google Scholar] [CrossRef]
  181. Lang, N.J.; Gote-Schniering, J.; Porras-Gonzalez, D.; Yang, L.; De Sadeleer, L.J.; Jentzsch, R.C.; Shitov, V.A.; Zhou, S.; Ansari, M.; Agami, A.; et al. Ex vivo tissue perturbations coupled to single-cell RNA-seq reveal multilineage cell circuit dynamics in human lung fibrogenesis. Sci. Transl. Med. 2023, 15, eadh0908. [Google Scholar] [CrossRef] [PubMed]
  182. Li, C.; Yu, Y.; Wan, Z.; Chiu, M.C.; Huang, J.; Zhang, S.; Zhu, X.; Lan, Q.; Deng, Y.; Zhou, Y.; et al. Human respiratory organoids sustained reproducible propagation of human rhinovirus C and elucidation of virus-host interaction. Nat. Commun. 2024, 15, 10772. [Google Scholar] [CrossRef] [PubMed]
  183. Toth, A.; Kannan, P.; Snowball, J.; Kofron, M.; Wayman, J.A.; Bridges, J.P.; Miraldi, E.R.; Swarr, D.; Zacharias, W.J. Alveolar epithelial progenitor cells require Nkx2-1 to maintain progenitor-specific epigenomic state during lung homeostasis and regeneration. Nat. Commun. 2023, 14, 8452. [Google Scholar] [CrossRef] [PubMed]
  184. Dye, B.R.; Youngblood, R.L.; Oakes, R.S.; Kasputis, T.; Clough, D.W.; Spence, J.R.; Shea, L.D. Human lung organoids develop into adult airway-like structures directed by physico-chemical biomaterial properties. Biomaterials 2020, 234, 119757. [Google Scholar] [CrossRef]
  185. Chan, L.L.Y.; Anderson, D.E.; Cheng, H.S.; Ivan, F.X.; Chen, S.; Kang, A.E.Z.; Foo, R.; Gamage, A.M.; Tiew, P.Y.; Koh, M.S.; et al. The establishment of COPD organoids to study host-pathogen interaction reveals enhanced viral fitness of SARS-CoV-2 in bronchi. Nat. Commun. 2022, 13, 7635. [Google Scholar] [CrossRef]
  186. Ritzau-Reid, K.I.; Callens, S.J.P.; Xie, R.; Cihova, M.; Reumann, D.; Grigsby, C.L.; Prados-Martin, L.; Wang, R.; Moore, A.C.; Armstrong, J.P.K.; et al. Microfibrous Scaffolds Guide Stem Cell Lumenogenesis and Brain Organoid Engineering. Adv. Mater. 2023, 35, e2300305. [Google Scholar] [CrossRef]
  187. Nikolaev, M.; Mitrofanova, O.; Broguiere, N.; Geraldo, S.; Dutta, D.; Tabata, Y.; Elci, B.; Brandenberg, N.; Kolotuev, I.; Gjorevski, N.; et al. Homeostatic mini-intestines through scaffold-guided organoid morphogenesis. Nature 2020, 585, 574–578. [Google Scholar] [CrossRef]
  188. Brassard, J.A.; Nikolaev, M.; Huebscher, T.; Hofer, M.; Lutolf, M.P. Recapitulating macro-scale tissue self-organization through organoid bioprinting. Nat. Mater. 2021, 20, 22–29. [Google Scholar] [CrossRef]
  189. Li, H.; Chen, Z.; Chen, N.; Fan, Y.; Xu, Y.; Xu, X. Applications of lung cancer organoids in precision medicine: From bench to bedside. Cell Commun. Signal. 2023, 21, 350. [Google Scholar] [CrossRef]
  190. Lehmann, M.; Krishnan, R.; Sucre, J.; Kulkarni, H.S.; Pineda, R.H.; Anderson, C.; Banovich, N.E.; Behrsing, H.P.; Dean, C.H.; Haak, A.; et al. Precision-Cut Lung Slices: Emerging Tools for Preclinical and Translational Lung Research An Official American Thoracic Society Workshop Report. Am. J. Respir. Cell Mol. Biol. 2025, 72, 16–31. [Google Scholar] [CrossRef]
  191. Eom, S.; Lee, S.Y.; Park, J.T.; Choi, I. Alveoli-Like Multifunctional Scaffolds for Optical and Electrochemical In Situ Monitoring of Cellular Responses from Type II Pneumocytes. Adv. Sci. 2023, 10, 2301395. [Google Scholar] [CrossRef]
  192. Gao, H.; Peng, W.; Zhou, Y.; Ding, Z.; Su, M.; Wu, Z.; Yu, C. Flexible and multi-functional three-dimensional scaffold based on enokitake-like Au nanowires for real-time monitoring of endothelial mechanotransduction. Biosens. Bioelectron. 2024, 263, 116610. [Google Scholar] [CrossRef]
  193. Di Gregorio, E.; Bitonto, V.; Baroni, S.; Stefania, R.; Aime, S.; Broche, L.M.; Senn, N.; Ross, P.J.; Lurie, D.J.; Geninatti Crich, S. Monitoring tissue implants by field-cycling 1H-MRI via the detection of changes in the 14N-quadrupolar-peak from imidazole moieties incorporated in a “smart” scaffold material. J. Mater. Chem. B 2021, 9, 4863–4872. [Google Scholar] [CrossRef]
  194. Huang, Q.; Qu, Y.; Tang, M.; Lan, K.; Zhang, Y.; Chen, S.; Li, W.; Gu, L. ROS-responsive hydrogel for bone regeneration: Controlled dimethyl fumarate release to reduce inflammation and enhance osteogenesis. Acta Biomater. 2025, 195, 183–200. [Google Scholar] [CrossRef]
  195. Yang, H.; Wang, W.; Xiao, J.; Yang, R.; Feng, L.; Xu, H.; Xu, L.; Xing, Y. ROS-responsive injectable hydrogels loaded with exosomes carrying miR-4500 reverse liver fibrosis. Biomaterials 2025, 314, 122887. [Google Scholar] [CrossRef]
  196. Kim, Y.E.; Kim, J. ROS-Scavenging Therapeutic Hydrogels for Modulation of the Inflammatory Response. ACS Appl. Mater. Interfaces 2022, 14, 23002–23021. [Google Scholar] [CrossRef] [PubMed]
  197. Liu, J.; Wu, Z.; Liu, Y.; Zhan, Z.; Yang, L.; Wang, C.; Jiang, Q.; Ran, H.; Li, P.; Wang, Z. ROS-responsive liposomes as an inhaled drug delivery nanoplatform for idiopathic pulmonary fibrosis treatment via Nrf2 signaling. J. Nanobiotechnol. 2022, 20, 213. [Google Scholar] [CrossRef]
  198. Wang, B.; Gao, Y.; Sun, L.; Xue, M.; Wang, M.; Zhang, Z.; Zhang, L.; Zhang, H. Inhaled pulmonary surfactant biomimetic liposomes for reversing idiopathic pulmonary fibrosis through synergistic therapeutic strategy. Biomaterials 2023, 303, 122404. [Google Scholar] [CrossRef] [PubMed]
  199. Li, X.; Li, Y.; Yu, C.; Bao, H.; Cheng, S.; Huang, J.; Zhang, Z. ROS-Responsive Janus Au/Mesoporous Silica Core/Shell Nanoparticles for Drug Delivery and Long-Term CT Imaging Tracking of MSCs in Pulmonary Fibrosis Treatment. ACS Nano 2023, 17, 6387–6399. [Google Scholar] [CrossRef] [PubMed]
  200. Xia, Y.; He, Y.; Zhang, F.; Liu, Y.; Leng, J. A Review of Shape Memory Polymers and Composites: Mechanisms, Materials, and Applications. Adv. Mater. 2021, 33, e2000713. [Google Scholar] [CrossRef]
  201. Lai, S.-M.; Jiang, S.-Y.F.; Chou, H.-C.; Lin, T.-Y.; Wei, Y.-E.; Yu, B.-Y. Novel two-way multiple shape memory effects of olefin block copolymer (OBC)/polycaprolactone (PCL) blends. Polym. Test. 2021, 102, 107333. [Google Scholar] [CrossRef]
  202. Gopinath, S.; Adarsh, N.N.; Nair, P.R.; Mathew, S. Shape-Memory Polymer Nanocomposites of Poly(ε-caprolactone) with the Polystyrene-block-polybutadiene-block-polystyrene-tri-block Copolymer Encapsulated with Metal Oxides. ACS Omega 2021, 6, 6261–6273. [Google Scholar] [CrossRef] [PubMed]
  203. Qi, X.; Pan, C.; Zhang, L.; Yue, D. Bio-Based, Self-Healing, Recyclable, Reconfigurable Multifunctional Polymers with Both One-Way and Two-Way Shape Memory Properties. ACS Appl. Mater. Interfaces 2023, 15, 3497–3506. [Google Scholar] [CrossRef] [PubMed]
  204. Zhou, J.; Yue, H.; Huang, M.; Hao, C.; He, S.; Liu, H.; Liu, W.; Zhu, C.; Dong, X.; Wang, D. Arbitrarily Reconfigurable and Thermadapt Reversible Two-Way Shape Memory Poly(thiourethane) Accomplished by Multiple Dynamic Covalent Bonds. ACS Appl. Mater. Interfaces 2021, 13, 43426–43437. [Google Scholar] [CrossRef]
  205. Chen, G.; Abdeen, A.A.; Wang, Y.; Shahi, P.K.; Robertson, S.; Xie, R.; Suzuki, M.; Pattnaik, B.R.; Saha, K.; Gong, S. A biodegradable nanocapsule delivers a Cas9 ribonucleoprotein complex for in vivo genome editing. Nat. Nanotechnol. 2019, 14, 974–980. [Google Scholar] [CrossRef]
  206. Wang, C.; Vazquez-Gonzalez, M.; Fadeev, M.; Sohn, Y.S.; Nechushtai, R.; Willner, I. Thermoplasmonic-Triggered Release of Loads from DNA-Modified Hydrogel Microcapsules Functionalized with Au Nanoparticles or Au Nanorods. Small 2020, 16, e2000880. [Google Scholar] [CrossRef]
  207. Metzger, J.M.; Wang, Y.; Neuman, S.S.; Snow, K.J.; Murray, S.A.; Lutz, C.M.; Bondarenko, V.; Felton, J.; Gimse, K.; Xie, R.; et al. Efficient in vivo neuronal genome editing in the mouse brain using nanocapsules containing CRISPR-Cas9 ribonucleoproteins. Biomaterials 2023, 293, 121959. [Google Scholar] [CrossRef]
  208. Seem, K.; Kaur, S.; Kumar, S.; Mohapatra, T. Epigenome editing for targeted DNA (de)methylation: A new perspective in modulating gene expression. Crit. Rev. Biochem. Mol. Biol. 2024, 59, 69–98. [Google Scholar] [CrossRef] [PubMed]
  209. Sung, C.K.; Yim, H. CRISPR-mediated promoter de/methylation technologies for gene regulation. Arch. Pharmacal Res. 2020, 43, 705–713. [Google Scholar] [CrossRef]
  210. O’Geen, H.; Tomkova, M.; Combs, J.A.; Tilley, E.K.; Segal, D.J. Determinants of heritable gene silencing for KRAB-dCas9+DNMT3 and Ezh2-dCas9+DNMT3 hit-and-run epigenome editing. Nucleic Acids Res. 2022, 50, 3239–3253. [Google Scholar] [CrossRef]
  211. Policarpi, C.; Munafo, M.; Tsagkris, S.; Carlini, V.; Hackett, J.A. Systematic epigenome editing captures the context-dependent instructive function of chromatin modifications. Nat. Genet. 2024, 56, 1168–1180. [Google Scholar] [CrossRef]
  212. McCutcheon, S.R.; Rohm, D.; Iglesias, N.; Gersbach, C.A. Epigenome editing technologies for discovery and medicine. Nat. Biotechnol. 2024, 42, 1199–1217. [Google Scholar] [CrossRef]
  213. Roth, G.V.; Gengaro, I.R.; Qi, L.S. Precision epigenetic editing: Technological advances, enduring challenges, and therapeutic applications. Cell Chem. Biol. 2024, 31, 1422–1446. [Google Scholar] [CrossRef]
  214. Li, M.; Chen, F.; Yang, Q.; Tang, Q.; Xiao, Z.; Tong, X.; Zhang, Y.; Lei, L.; Li, S. Biomaterial-Based CRISPR/Cas9 Delivery Systems for Tumor Treatment. Biomater. Res. 2024, 28, 0023. [Google Scholar] [CrossRef] [PubMed]
  215. Du, Y.; Liu, Y.; Hu, J.; Peng, X.; Liu, Z. CRISPR/Cas9 systems: Delivery technologies and biomedical applications. Asian J. Pharm. Sci. 2023, 18, 100854. [Google Scholar] [CrossRef]
  216. Sun, L.; Fan, M.; Huang, D.; Li, B.; Xu, R.; Gao, F.; Chen, Y. Clodronate-loaded liposomal and fibroblast-derived exosomal hybrid system for enhanced drug delivery to pulmonary fibrosis. Biomaterials 2021, 271, 120761. [Google Scholar] [CrossRef] [PubMed]
  217. Wan, Q.; Zhang, X.; Zhou, D.; Xie, R.; Cai, Y.; Zhang, K.; Sun, X. Inhaled nano-based therapeutics for pulmonary fibrosis: Recent advances and future prospects. J. Nanobiotechnol. 2023, 21, 215. [Google Scholar] [CrossRef]
  218. Chang, X.; Xing, L.; Wang, Y.; Yang, C.-X.; He, Y.-J.; Zhou, T.-J.; Gao, X.-D.; Li, L.; Hao, H.-P.; Jiang, H.L. Monocyte-derived multipotent cell delivered programmed therapeutics to reverse idiopathic pulmonary fibrosis. Sci. Adv. 2020, 6, eaba3167. [Google Scholar] [CrossRef] [PubMed]
  219. Manghwar, H.; Li, B.; Ding, X.; Hussain, A.; Lindsey, K.; Zhang, X.; Jin, S. CRISPR/Cas Systems in Genome Editing: Methodologies and Tools for sgRNA Design, Off-Target Evaluation, and Strategies to Mitigate Off-Target Effects. Adv. Sci. 2020, 7, 1902312. [Google Scholar] [CrossRef]
  220. Merlin, J.P.J.; Abrahamse, H. Optimizing CRISPR/Cas9 precision: Mitigating off-target effects for safe integration with photodynamic and stem cell therapies in cancer treatment. Biomed. Pharmacother. 2024, 180, 117516. [Google Scholar] [CrossRef]
  221. McDonald, S.M.; Augustine, E.K.; Lanners, Q.; Rudin, C.; Brinson, L.C.; Becker, M.L. Applied machine learning as a driver for polymeric biomaterials design. Nat. Commun. 2023, 14, 4838. [Google Scholar] [CrossRef]
  222. Suwardi, A.; Wang, F.K.; Xue, K.; Han, M.Y.; Teo, P.L.; Wang, P.; Wang, S.J.; Liu, Y.; Ye, E.Y.; Li, Z.B.; et al. Machine Learning-Driven Biomaterials Evolution. Adv. Mater. 2022, 34, 2102703. [Google Scholar] [CrossRef]
  223. Webb, M.A.; Patel, R.A. Data-Driven Design of Polymer-Based Biomaterials: High-throughput Simulation, Experimentation, and Machine Learning. ACS Appl. Bio Mater. 2023, 7, 510–527. [Google Scholar]
  224. Kappe, K.; Hoschke, K.; Riedel, W.; Hiermaier, S. Multi-objective optimization of additive manufactured functionally graded lattice structures under impact. Int. J. Impact Eng. 2024, 183, 104789. [Google Scholar] [CrossRef]
  225. Peng, B.; Wei, Y.; Qin, Y.; Dai, J.B.; Li, Y.; Liu, A.B.; Tian, Y.; Han, L.L.; Zheng, Y.F.; Wen, P. Machine learning-enabled constrained multi-objective design of architected materials. Nat. Commun. 2023, 14, 6630. [Google Scholar] [CrossRef]
  226. Reel, P.S.; Reel, S.; Pearson, E.; Trucco, E.; Jefferson, E. Using machine learning approaches for multi-omics data analysis: A review. Biotechnol. Adv. 2021, 49, 107739. [Google Scholar] [CrossRef]
  227. Briscik, M.; Tazza, G.; Vidács, L.; Dillies, M.A.; Déjean, S. Supervised multiple kernel learning approaches for multi-omics data integration. BioData Min. 2024, 17, 53. [Google Scholar] [CrossRef] [PubMed]
  228. Virijevicc, K.; Zivanovic, M.N.; Nikolic, D.; Milivojevic, N.; Pavic, J.; Moric, I.; Scenerovic, L.; Dragacevic, L.; Thurner, P.J.; Rufin, M.; et al. AI-Driven Optimization of PCL/PEG Electrospun Scaffolds for Enhanced In Vivo Wound Healing. ACS Appl. Mater. Interfaces 2024, 16, 22989–23002. [Google Scholar]
  229. Rafieyan, S.; Ansari, E.; Vasheghani-Farahani, E. A practical machine learning approach for predicting the quality of 3D (bio)printed scaffolds. Biofabrication 2024, 16, 045014. [Google Scholar] [CrossRef]
  230. Koudstaal, T.; Funke-Chambour, M.; Kreuter, M.; Molyneaux, P.L.; Wijsenbeek, M.S. Pulmonary fibrosis: From pathogenesis to clinical decision-making. Trends Mol. Med. 2023, 29, 1076–1087. [Google Scholar] [CrossRef]
  231. Chianese, M.; Screm, G.; Salton, F.; Confalonieri, P.; Trotta, L.; Barbieri, M.; Ruggero, L.; Mari, M.; Reccardini, N.; Geri, P.; et al. Pirfenidone and Nintedanib in Pulmonary Fibrosis: Lights and Shadows. Pharmaceuticals 2024, 17, 709. [Google Scholar] [CrossRef]
  232. Vancheri, C.; Kreuter, M.; Richeldi, L.; Ryerson, C.J.; Valeyre, D.; Grutters, J.C.; Wiebe, S.; Stansen, W.; Quaresma, M.; Stowasser, S.; et al. Nintedanib with Add-on Pirfenidone in Idiopathic Pulmonary Fibrosis Results of the INJOURNEY Trial. Am. J. Respir. Crit. Care Med. 2018, 197, 356–363. [Google Scholar] [CrossRef]
  233. Ma, J.; Zhang, L.; Lei, B. Multifunctional MXene-Based Bioactive Materials for Integrated Regeneration Therapy. ACS Nano 2023, 17, 19526–19549. [Google Scholar] [CrossRef] [PubMed]
  234. Doryab, A.; Schmid, O. Bioactive Cell-Derived ECM Scaffold Forms a Unique Cellular Microenvironment for Lung Tissue Engineering. Biomedicine 2022, 10, 1791. [Google Scholar] [CrossRef] [PubMed]
  235. Hsieh, W.-T.; Liu, Y.-S.; Lee, Y.-h.; Rimando, M.G.; Lin, K.-h.; Lee, O.K. Matrix dimensionality and stiffness cooperatively regulate osteogenesis of mesenchymal stromal cells. Acta Biomater. 2016, 32, 210–222. [Google Scholar] [CrossRef] [PubMed]
  236. Raftery, R.M.; Walsh, D.P.; Ferreras, L.B.; Castano, I.M.; Chen, G.; LeMoine, M.; Osman, G.; Shakesheff, K.M.; Dixon, J.E.; O’Brien, F.J. Highly versatile cell-penetrating peptide loaded scaffold for efficient and localised gene delivery to multiple cell types: From development to application in tissue engineering. Biomaterials 2019, 216, 119277. [Google Scholar] [CrossRef]
  237. Zhang, W.; Zhang, Y.; Tu, T.; Schmull, S.; Han, Y.; Wang, W.; Li, H. Dual inhibition of HDAC and tyrosine kinase signaling pathways with CUDC-907 attenuates TGFβ1 induced lung and tumor fibrosis. Cell Death Dis. 2020, 11, 765. [Google Scholar] [CrossRef]
  238. Liang, H.; Wang, Q.; Wang, D.; Zheng, H.; Kalvakolanu, D.V.; Lu, H.; Wen, N.; Chen, X.; Xu, L.; Ren, J.; et al. RGFP966, a histone deacetylase 3 inhibitor, promotes glioma stem cell differentiation by blocking TGF-β signaling via SMAD7 (vol 180, 114118, 2020). Biochem. Pharmacol. 2020, 182, 114223. [Google Scholar] [CrossRef]
  239. Liu, C.-H.; Lee, H.-S.; Liou, J.-P.; Hua, H.-S.; Cheng, W.-H.; Yuliani, F.S.; Chen, B.-C.; Lin, C.-H. MPT0E028, a novel pan-HDAC inhibitor, prevents pulmonary fibrosis through inhibition of TGF-β-induced CTGF expression in human lung fibroblasts: Involvement of MKP-1 activation. Eur. J. Pharmacol. 2024, 977, 176711. [Google Scholar] [CrossRef]
  240. Mehdizadeh, H.; Sumo, S.; Bayrak, E.S.; Brey, E.M.; Cinar, A. Three-dimensional modeling of angiogenesis in porous biomaterial scaffolds. Biomaterials 2013, 34, 2875–2887. [Google Scholar] [CrossRef]
  241. Pupiute, A.; Ciuzas, D.; Baniukaitiene, O.; Tichonovas, M.; Martuzevicius, D.; Petrikaite, V.; Krugly, E. Development of two layer fibrous scaffolds for 3D in vitro modelling: Effects of morphology and surface properties on cell proliferation, adhesion and drug sensitivity. J. Drug Deliv. Sci. Technol. 2024, 101, 106213. [Google Scholar] [CrossRef]
  242. Jing, L.; Wang, X.; Leng, B.; Zhan, N.; Liu, H.; Wang, S.; Lu, Y.; Sun, J.; Huang, D. Engineered Nanotopography on the Microfibers of 3D-Printed PCL Scaffolds to Modulate Cellular Responses and Establish an In Vitro Tumor Model. ACS Appl. Bio Mater. 2021, 4, 1381–1394. [Google Scholar] [CrossRef] [PubMed]
  243. Redmond, J.; McCarthy, H.O.; Buchanan, P.; Levingstone, T.J.; Dunne, N.J. Development and characterisation of 3D collagen-gelatin based scaffolds for breast cancer research. Biomater. Adv. 2022, 142, 213157. [Google Scholar] [CrossRef] [PubMed]
Figure 1. The schematic of the transition from a healthy, compliant matrix to persistent fibrotic remodelling in the lung driven by mechanical cues. Quiescent Cells reside within a soft extracellular matrix (ECM). Progressive collagen deposition and crosslinking elevate tissue stiffness. Cells sense mechanical changes via integrin-mediated adhesions. This triggers the activation of mechanosensitive regulators such as YAP and TAZ, as well as profibrotic TGF-β and SMAD signalling. These factors translocate to the nucleus and drive transcriptional reprogramming. Accompanying epigenetic modifications such as DNA methylation and histone alterations stabilize profibrotic gene expression and promote fibroblast and myofibroblast activation. This leads to increased proliferation, migration and collagen production. Activated Cells further remodel and contract the ECM, increasing stiffness and establishing a self-sustaining positive feedback loop that perpetuates fibrosis.
Figure 1. The schematic of the transition from a healthy, compliant matrix to persistent fibrotic remodelling in the lung driven by mechanical cues. Quiescent Cells reside within a soft extracellular matrix (ECM). Progressive collagen deposition and crosslinking elevate tissue stiffness. Cells sense mechanical changes via integrin-mediated adhesions. This triggers the activation of mechanosensitive regulators such as YAP and TAZ, as well as profibrotic TGF-β and SMAD signalling. These factors translocate to the nucleus and drive transcriptional reprogramming. Accompanying epigenetic modifications such as DNA methylation and histone alterations stabilize profibrotic gene expression and promote fibroblast and myofibroblast activation. This leads to increased proliferation, migration and collagen production. Activated Cells further remodel and contract the ECM, increasing stiffness and establishing a self-sustaining positive feedback loop that perpetuates fibrosis.
Pharmaceuticals 18 01487 g001
Figure 2. Dual Drug Synergy Strategy for Epigenetic Reprogramming in Pulmonary Fibrosis (PF). The top panel depicts fibrotic lung tissue containing activated fibroblasts and aberrant epigenetic alterations, such as disrupted histone acetylation and DNA hypermethylation. The central panel presents biomaterial-based delivery systems, including gelatin methacryloyl (GelMA) hydrogels for the delivery of DNMT inhibitors (DNMTi), polycaprolactone (PCL)/collagen nanofibers for HDAC inhibitors (HDACi), as well as composite scaffolds engineered for co-delivery. The bottom panel demonstrates how combined treatment with DNMTi and HDACi reverses disease-associated epigenetic modifications, reactivates silenced antifibrotic genes, and facilitates both functional and structural restoration of lung tissue toward a physiological state. The figure highlights the promise of biomaterial-mediated co-delivery systems for synergistic epigenetic therapy in PF.
Figure 2. Dual Drug Synergy Strategy for Epigenetic Reprogramming in Pulmonary Fibrosis (PF). The top panel depicts fibrotic lung tissue containing activated fibroblasts and aberrant epigenetic alterations, such as disrupted histone acetylation and DNA hypermethylation. The central panel presents biomaterial-based delivery systems, including gelatin methacryloyl (GelMA) hydrogels for the delivery of DNMT inhibitors (DNMTi), polycaprolactone (PCL)/collagen nanofibers for HDAC inhibitors (HDACi), as well as composite scaffolds engineered for co-delivery. The bottom panel demonstrates how combined treatment with DNMTi and HDACi reverses disease-associated epigenetic modifications, reactivates silenced antifibrotic genes, and facilitates both functional and structural restoration of lung tissue toward a physiological state. The figure highlights the promise of biomaterial-mediated co-delivery systems for synergistic epigenetic therapy in PF.
Pharmaceuticals 18 01487 g002
Figure 3. Artificial intelligence (AI)-driven patient-specific scaffold design and epigenetic reprogramming strategy. Clinical and multi-omics data from patients are used to generate tailored scaffold designs, with AI optimizing architectural features, mechanical compliance, and drug loading parameters. The design incorporates stimuli-responsive scaffolds capable of sensing local microenvironmental cues and releasing therapeutics in a feedback-controlled manner. These platforms also facilitate CRISPR-based epigenetic editing alongside pharmacological intervention to achieve targeted reprogramming of disease-associated epigenetic signatures. Subsequent panels illustrate the recovery of cellular functionality and tissue integrity following treatment, highlighting the potential of combining AI-guided biomaterial design, stimuli-responsive delivery, and epigenetic editing for personalized antifibrotic therapy.
Figure 3. Artificial intelligence (AI)-driven patient-specific scaffold design and epigenetic reprogramming strategy. Clinical and multi-omics data from patients are used to generate tailored scaffold designs, with AI optimizing architectural features, mechanical compliance, and drug loading parameters. The design incorporates stimuli-responsive scaffolds capable of sensing local microenvironmental cues and releasing therapeutics in a feedback-controlled manner. These platforms also facilitate CRISPR-based epigenetic editing alongside pharmacological intervention to achieve targeted reprogramming of disease-associated epigenetic signatures. Subsequent panels illustrate the recovery of cellular functionality and tissue integrity following treatment, highlighting the potential of combining AI-guided biomaterial design, stimuli-responsive delivery, and epigenetic editing for personalized antifibrotic therapy.
Pharmaceuticals 18 01487 g003
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

Wang, J.; Xu, A. Mechanotransduction-Epigenetic Coupling in Pulmonary Regeneration: Multifunctional Bioscaffolds as Emerging Tools. Pharmaceuticals 2025, 18, 1487. https://doi.org/10.3390/ph18101487

AMA Style

Wang J, Xu A. Mechanotransduction-Epigenetic Coupling in Pulmonary Regeneration: Multifunctional Bioscaffolds as Emerging Tools. Pharmaceuticals. 2025; 18(10):1487. https://doi.org/10.3390/ph18101487

Chicago/Turabian Style

Wang, Jing, and Anmin Xu. 2025. "Mechanotransduction-Epigenetic Coupling in Pulmonary Regeneration: Multifunctional Bioscaffolds as Emerging Tools" Pharmaceuticals 18, no. 10: 1487. https://doi.org/10.3390/ph18101487

APA Style

Wang, J., & Xu, A. (2025). Mechanotransduction-Epigenetic Coupling in Pulmonary Regeneration: Multifunctional Bioscaffolds as Emerging Tools. Pharmaceuticals, 18(10), 1487. https://doi.org/10.3390/ph18101487

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

Article Metrics

Back to TopTop