Skip to main content

Biomaterial-based mechanical regulation facilitates scarless wound healing with functional skin appendage regeneration

Abstract

Scar formation resulting from burns or severe trauma can significantly compromise the structural integrity of skin and lead to permanent loss of skin appendages, ultimately impairing its normal physiological function. Accumulating evidence underscores the potential of targeted modulation of mechanical cues to enhance skin regeneration, promoting scarless repair by influencing the extracellular microenvironment and driving the phenotypic transitions. The field of skin repair and skin appendage regeneration has witnessed remarkable advancements in the utilization of biomaterials with distinct physical properties. However, a comprehensive understanding of the underlying mechanisms remains somewhat elusive, limiting the broader application of these innovations. In this review, we present two promising biomaterial-based mechanical approaches aimed at bolstering the regenerative capacity of compromised skin. The first approach involves leveraging biomaterials with specific biophysical properties to create an optimal scarless environment that supports cellular activities essential for regeneration. The second approach centers on harnessing mechanical forces exerted by biomaterials to enhance cellular plasticity, facilitating efficient cellular reprogramming and, consequently, promoting the regeneration of skin appendages. In summary, the manipulation of mechanical cues using biomaterial-based strategies holds significant promise as a supplementary approach for achieving scarless wound healing, coupled with the restoration of multiple skin appendage functions.

Background

Achieving scarless wound healing with skin appendage regeneration requires the creation of a harmonious scarless environment and the mobilization of resident cells through biochemical or biomechanical cues. Biomechanical cues refer to how cells and tissues respond to both internal and external mechanical forces. One area of research focus has been the utilization of biomaterials to promote cutaneous wound healing, and increasing evidence suggests that biomechanical cues play a significant role in how the skin interacts with these materials. The skin is a highly specialized mechanoresponsive organ that reacts to mechanical forces in various physiological contexts, including growth, obesity, and friction. Skin appendages, such as hair follicles (HFs), sebaceous glands, and sweat glands (SwGs), are essential for skin homeostasis and function [1]. These appendages are constantly exposed to dynamic mechanical cues, from rapid expansion during embryonic and early postnatal development to external forces throughout life. However, significant skin defects caused by burns, severe trauma, or chronic diseases often disrupt the native mechanical homeostasis of skin and lead to the loss of functional cells, including basal keratinocytes, dermal papilla cells, and sweat gland cells (SGCs). The role of mechanical cues in scar formation and wound healing has been gradually revealed [2], which has opened up promising strategies for scarless wound healing [3, 4]. Biomaterials with unique physical properties, such as stiffness, topography, magnetism, and conductivity, have gained attention. To date, biomaterials with specific physical properties can provide a structural framework to facilitate the attachment and migration of host stem and progenitor cells, and drive the differentiation of these cells into tissue-specific cell types by breaking through the epigenetic barrier, improve immune ecology so as to enhance the efficiency of neovascularization, restoration of innervation, and regeneration of skin appendages [5,6,7,8]. Exact mechanisms by which biomaterials influence cells and ways to use them to stimulate tissues are interesting. This review outlines recent developments in biomaterial-based mechanical regulation. It begins by discussing the mechanical cue-dependent scarring process, including key factors such as molecules [transforming growth factor-β (TGF-β), vascular endothelial growth factor (VEGF), neuropeptides, etc.] and cells (fibroblasts, keratinocytes, immunocytes, etc.) [9,10,11,12]. It then delves into the biophysical characteristics of biomaterials that modulate extracellular signaling, alter cellular epigenetic states, support cell migration, and allow tissue ingrowth. Finally, it explores the potential of biomaterial-based mechanical cues to create scarless environment and to stimulate stem and progenitor cells for regeneration of skin appendages. In particular, by combining the principles of developmental science and biomaterials, we anticipate that biomaterials can facilitate the regeneration of multiple skin appendages through sequential regulation. Overall, this review offers novel strategies based on the physical properties of biomaterials to drive scarless wound healing and skin appendage regeneration.

Mechanical signal-dependent scarring

Previous study shed light on the effect of mechanical forces on scar formation, and the mechano-transduction signaling pathway can be a potential target to reduce scarring and promote skin regeneration [3]. From clinical observations and laboratory evidences, mechanical forces are involved in the process of wound healing, and regulate scar formation-related factors, including TGF-β, epithelial-mesenchymal transition (EMT), neurovascular abnormalities as well as inflammation [10, 13,14,15,16]. A deeper understanding of the mechanical dependence of scarring is conducive to the development of biomaterials for mechanical regulation.

Clinical observation

Typically, human wounds heal by forming non-functional fibrotic tissue, commonly referred to as a scar. Among fibroproliferative conditions, keloid and hypertrophic scarring are well-known. These abnormal scars tend to develop in specific areas, such as the anterior chest, shoulder, scapula, and lower abdomen, which are frequently subjected to skin stretching and mechanical forces. Conversely, scars are less common on the scalp and the anterior lower leg due to the direct connection of skin to underlying bones [17]. Moreover, the presence of skin tension lines underscores the significance of anisotropic mechanical properties within the skin’s anatomy. These lines, delineated by the arrangement of dermal collagen and elastin fibers, guide surgeons in making incisions that minimize the likelihood of scar formation [18]. These clinical observations strongly indicate that mechanical forces play a primary role in the formation of scars on wounded skin.

Laboratory evidence

As research progressed, the molecular and cellular basis for mechanical force-induced scarring is gradually discovered in the laboratory. Mechanosensitive proteins, cells, and biological processes lose their original equilibrium state under the influence of mechanical force and become risk factors for scar formation.

Mechanosensitive TGF-β

Notably, there is a growing interest in mechanically sensitive proteins, both in in vivo and in vitro studies. Among these proteins, TGF-β stands out as a well-established key cytokine that regulates the response of fibroblasts to injury and the pathological development of fibrosis [19]. Brief mechanical stretching of tissues reduces the levels of soluble TGF-β and collagen I. In contrast, sustained mechanical stimuli increases TGF-β levels, promoting the transformation of fibroblasts into myofibroblasts that express higher levels of α-smooth muscle actin (α-SMA). This transition exacerbates wound contraction force. Moreover, fibroblasts within mechanically loaded wounds undergo a more pronounced fibroproliferative process due to tension, which inhibits the phosphatidylinositol-3-kinase (PI3K)/Akt pathway associated apoptosis [20]. Thus, a detrimental cycle forms within wounded skin where the mechanical force derived from myofibroblasts in turn activate more fibroblasts in a manner of positive feedback to exacerbate scarring [20, 21] (Fig. 1a).

Fig. 1
figure 1

Mechanical cue-dependent scarring. a TGF-β activates fibroblasts to transform into myofibroblasts and the latter give rise to α-SMA, endowing wound aberrant contraction force. b Epithelial-mesenchymal transition (EMT) in inflammatory environment causes fibrosis related gene expression through diverse pathways. RAS-responsive element binding protein 1 (RREB1) serves as a connector between MAPK and Smad pathways. c Inflammatory factors leak from damaged vascular endothelium; imbalance between growth and regression of neovascularization causes irregular vessel cluster; blood flow in irregular vessel produces aberrant shear stress. d Nerve damage within wound launches neurogenic inflammation where neuropeptides [calcitonin gene-related peptide (CGRP) and substance P] from nerve endings mediate harmful vascular events including abnormal vasodilation and plasm extravasation. e Neutrophiles and macrophages, as the main innate immunocytes within wound, upregulate inflammatory factors and provide fibroblasts with enduring activating signals. Created with BioRender.com. TGF-β transforming growth factor-β, α-SMA α-smooth muscle actin, ST2 suppression of tumorigenicity 2 receptor, PI3K/Akt phosphatidylinositol-3-kinase/Akt, MAPK mitogen-activated protein kinase, TFs transcriptional factors, ZEB1 zinc finger E-box binding homeobox 1, ZEB2 zinc finger E-box binding homeobox 2, IL11 interleukin-11, CTGF connective tissue growth factor, PDGF-B platelet-derived growth factor B, WISP1 Wnt1-inducible signaling pathway protein 1, NF-κB nuclear factor-κB, STAT3 signal transducer and activator or transcription 3, HIF-1α hypoxia-inducible factor-1α

Scar-associated cell phenotype

Mechanical forces profoundly influence cell phenotypes [22, 23]. EMT is a pivotal event in wound healing, wherein epithelial cells acquire fibroblast-like features characterized by reduced intercellular adhesion and increased motility. This process is largely stimulated by TGF-β within wounds, resulting in decreased E-cadherin and increased vimentin and α-SMA levels. Injury-induced EMT plays a crucial role in fibrosis processes, including renal, pulmonary, cardiac fibrosis, and cutaneous scar formation due to aberrant TGF-β signaling [24]. Key transcription factors involved in TGF-β-induced EMT include Slug, Twist, zinc finger E-box binding homeobox 1 (ZEB1), ZEB2, and Snail, with signaling pathways encompassing Smad, PI3K/Akt, and mitogen-activated protein kinase (MAPK) pathways [9, 15, 24]. Notably, RAS-responsive element binding protein 1 (RREB1) acts as a bridge between TGF-β-Smad and Ras-MAPK, promoting fibrosis and scar formation by increasing the deposition of fibrous connective tissue, including interleukin (IL)-11, connective tissue growth factor (CTGF), platelet-derived growth factor B (PDGF-B) and WNT-inducible signaling pathway protein 1 (WISP1) [9]. Additionally, IL-33, primarily expressed and stored in the nuclei of endothelial and epithelial cells, contributes to organ fibrosis via type-2 EMT through the IL-33-suppression of tumorigenicity 2 receptor (ST2)-NF-κB p65-CD146 axis [25, 26]. In a high glucose environment, signal transducer and activator of transcription 3 activation increases hypoxia-inducible factor-1α levels and EMT [27] (Fig. 1b). In animal models, insulin, insulin growth factor receptors, protein kinase B, VEGF, and VEGF receptors are upregulated following acute stretching [28]. While these factors enhance vascularity, they also lead to skin microcirculation dysfunction, which is detrimental to scarless wound healing [29]. Various factors contribute to this dysfunction, including high blood vessel permeability, a disrupted neurovascular network, irregular formation of vascular clusters [12] (Fig. 1c), and endothelial mesenchymal transition influenced by shear stress, where endothelial cells acquire a mesenchymal phenotype and express α-SMA and collagen I [30, 31]. Furthermore, the release of neuropeptides (calcitonin gene-related peptide, substance P) by sensory endings is affected by mechanical stress through mechanosensitive nociceptors [10], because nociceptors with unmyelinated axons (C-fibers) are sensitive to compression and tensile. Its pro-scarring essence is nerve-mediated vascular-centric inflammation (Fig. 1d).

Mechanical force-related inflammation

In addition to mechanosensitive proteins, immunocytes especially neutrophils and macrophages also respond to mechanical forces and increase the level of inflammatory factors during healing, which is seen as another culprits for fibrosis and scar formation [32] (Fig. 1e). Neutrophils are the first “warrior” migrating from blood to impaired sites, releasing chemokines and cytokines to modulate inflammatory links, killing pathogens via phagocytosis, degranulation, reactive oxygen species, and neutrophil extracellular traps (NETs) [33, 34]. NETs could promote the transition of fibroblasts to myofibroblasts through IL-17 [35]. Normally, by the time of inflammatory phase ending, neutrophils experience apoptosis and are engulfed by macrophages, providing prerequisite for subsequent healing process [36]. However, malapropos behaviors of neutrophils and macrophages contribute to high level of inflammatory factors. Accumulated neutrophils, can be triggered by over-released recruitment promoters such as tumour necrosis factor-α and leukotriene B4 [37], and the dysfunctional phenotype transition of macrophages from pro-inflammatory (M1) to anti-inflammatory (M2) [11]. Macrophage is considered as the main initiator of fibrosis [38]. Indirectly, macrophages produce profibrotic mediators that activate fibroblasts, including TGF-β, PDGF, etc. Directly, matrix metalloproteases and tissue inhibitors of matrix metalloproteinases from macrophages control the turnover of extracellular matrix (ECM). Collectively, overactivation of immune cells under the influence of mechanical forces is one of the important factors in scarring.

Mechanism of mechano-transduction

Mechano-transduction refers to the process in which cells transmit mechanical forces to intracellular biochemical signals. Mounting studies indicate improper mechano-transduction is an important link in mediating mechanical forces to promote scarring [39]. Cells rely on crucial mechanical components, including focal adhesions (FAs), cytoskeleton, as well as the linker between the nuclear skeleton and cytoskeleton to effectively respond to mechanical forces from sheer stress, matrix stiffness, contraction, etc. (Fig. 2a). This mechano-transduction path is the key to directing biomaterial-based mechanical regulation.

Fig. 2
figure 2

Mechano-transduction and YAP/TAZ. a Integrin-cytoskeleton-nesprin-SUN-domain protein serves as a mechanical axis passing mechanical force from extracellular matrix to nuclear lamina. Mechano-sensitive integrin mediates extracellular force induced fibrogenic gene (e.g., MCP-1) expression and cytoskeleton reorganization through focal adhesion kinase (FAK)-extracellular regulated protein kinases (ERK) signaling and Rho GTPase, respectively. Intracellular force likewise affects cytoskeletal rearrangement through regulating actin-binding proteins (ABPs), plakin protein family and modification of tubulin (e.g., methylation). Nuclear force regulates chromatin status through interacting with nuclear laminA/C and laminB1/2. b Physical factors in extracellular environment (e.g., shear stress, stiffness, cell geometry) produce mechanical forces. These mechanical forces are transmitted by mechanosensitive transmembrane proteins like TGF-β1 and integrins as well as intracellular proteins, like CCM3, to regulate translocation of YAP/TAZ directly and indirectly. YAP/TAZ is directly phosphorylated by LATS1/2 and retains in cytoplasm to combine with protein 14-3-3 and degrade. Mechanical forces inhibit the phosphorylation of YAP/TAZ to increase the translocation of YAP/TAZ into nucleus, which will induce fibrosis related gene (e.g., IL6, IL8, SELE) expression. Created with BioRender.com. YAP/TAZ Yes-associated protein and transcriptional coactivator with PDZ-binding motif, FAK focal adhesion kinase, ERK extracellular signal-regulated kinase, NPC nuclear pore complex, MCP-1 monocyte chemoattractant protein-1, Tβ4 thymosin β4, Arp2/3 actin-related protein2/3, ECM extracellular matrix, TGF-β1 transforming growth factor-β1, CCM3 cerebral cavernous malformation 3, LATS1/2 large tumor suppressor 1/2, PP1A protein phosphatase 1α, IL interleukin, SELE selectin E, CTGF connective tissue growth factor, PAI-1 plasminogen activator inhibitor-1, LOX lysyl oxidase, TFs transcriptional factors

Integrin-focal adhesion kinase (FAK) signaling

FAs, which connect the actin cytoskeleton to the ECM, consist of transmembrane integrin heterodimers, actin-linking proteins (e.g., vinculin and paxillin), signaling proteins, FAK and so on. The autophosphorylation of FAK plays a crucial role in regulating the assembly and maturation of FAs in response to mechanical forces from ECM perturbations. This process helps maintain local harmony and regulates cell behavior [40]. Specifically, integrin tails bind to cytoplasmic proteins like talin and kindlin, while their extracellular parts bind to ligand proteins in the ECM. When integrin receptors are activated, downstream effectors like Rho GTPases come into play, influencing the reorganization of the cytoskeleton. The expression of integrins is tissue-specific and there are 8 β subunits and 18 α subunits in vertebrates. β1 is expressed in most cell types and it binds collagen, RGD (tripeptide Arg-Gly-Asp sequence), laminin, and leukocyte-specific receptors. Epithelial cells utilize α3 and α6 to interact with laminin, while fibroblasts and endothelial cells rely on α5β3 for cell spreading, and β3 serves as a direct sensor for shear stress [41,42,43]. In the skin, specialized hemidesmosomes (HDs) anchor keratinocytes to the basal membrane via intermediate filaments (IFs), and α6β4, a transmembrane receptor for laminin-332, is vital for HD formation [44, 45]. Interestingly, integrin α6β4 reduces the size of FAs, cellular spreading, traction force, and interferes with mechano-transduction-related signaling pathways [46]. FAs and HDs appear to have opposite roles, where FAs dominate keratinocyte motility, while HDs guide their settlement. FAs exert significant influence on the biological behavior of various cell types, including differentiation and migration. For example, endothelial cells display distinct cellular morphologies when placed on planar bio-adhesive surfaces or micro-grooved topographic surfaces due to the assembly of FAs [47]. In fibroblasts, mechanical forces transmitted via FAK activate extracellular-related kinase and downstream pro-fibrotic targets, ultimately contributing to the formation of cutaneous scars [48]. Inhibition of FAK in the wound tissue of red duroc pigs resulted in the regeneration of HFs, subcutaneous glands, and peri-follicular adipose tissue [49]. Consequently, targeting the formation of FAs in a mechanical manner holds significant potential to promote scarless wound healing by regulating resident cells within the wound.

Cytoskeleton and associated proteins

The cytoskeleton consists of three main components: microfilaments, microtubules and IFs. Monomeric G-actin molecules polymerize into actin filaments (F-actin), which are dynamically involved in numerous cellular processes throughout a cell’s lifespan. They interact with a diverse array of actin-binding proteins (ABPs) to influence cellular behaviors [50]. Notably, actin networks, such as lamellipodial and filopodial structures, play pivotal roles in cell polarity during migration and division [51]. The RhoA-Rho kinase/myosin light chain (ROCK/MLC) signaling axis regulates actomyosin contractility and mechanical forces, which is crucial for migration and morphogenesis [52]. Applying forces to actin filaments lead to conformational changes and ABP binding, offering intriguing possibilities for artificial manipulation of cellular behaviors [53]. Profilin, thymosin-β4, formin and the Arp2/3 complex are potential ABPs involved in the mechanical regulation of scarless wound healing [54].

Microtubules exist within the cytoplasm and are highly dynamic. Tubulin and tubule-binding proteins form the hollow cylinder structure of microtubules, serving as the intracellular transport network. Together with accessory proteins, microtubules also play a central role in forming the mitotic spindle, cilia, and flagella [55]. Tubulin modification, such as acetylation, impacts cellular behaviors by promoting the turnover of FAs during cell migration because of mechanical forces from substrate stiffness [56, 57]. Except for the traditional identity as skeletal component which typically supports, maintains the cell shape, microtubule functioning as perceiver of strain is largely coupled with FAs and actin filament [58].

IFs constitute another fibrous component of the cytoskeleton. The molecular composition of IFs varies across cell types. For instance, vimentin is expressed in mesenchymal cells, fibroblasts, and endothelial cells, while keratin is expressed in keratinocytes [59]. Desmin is found in smooth, skeletal, and cardiac muscles [60]. The cell-specific distribution of IFs suggests diverse spatial and temporal biological functions. IFs interact with actin filaments and microtubules, participate in the formation of FAs, and serve as mechano-sensors. For example, keratin IFs in keratinocytes preferentially align on stiff substrates [61]. IFs are anchored to the cell surface by cyto-linker proteins, such as the plakin protein family [62]. Plectin, a member of this family, links keratin filaments and integrin to impact the level of stiffness-dependent laminA/C in keratinocytes [61]. Plectin also plays a vital role in the stable assembly of IFs in fibroblasts by interacting with vimentin [63]. The cytoskeleton’s sensitivity to mechanical cues and its critical role in cellular activities have profound significance for biomaterial design.

Linker of nucleoskeleton and cytoskeleton (LINC) complex

The LINC complex plays a pivotal role in bridging the connection between the nuclear skeleton and cytoskeleton. Spanning the nuclear envelope, the LINC complex serves as a delivery path for mechanical forces [64]. Its key components include nesprins, which extend across the outer nuclear membrane and interact with cytoskeletal components, and Sad, UNC-84 domain protein (SUN), which span the inner nuclear membrane, connecting nesprins to the nuclear lamina [65]. The LINC complex influences gene expression through interacting with perinuclear constituents [66]. Changes in tension within nesprins mechanically modulate cell polarity through β-catenin signaling during EMT [67]. These changes result from actomyosin-dependent perturbations, providing strong evidence of mechanical regulation of the LINC complex [68]. Targeted strategies have been explored to manipulate the LINC complex. For example, the progression of laminA-associated dilated cardiomyopathy involving laminA can be slowed down by disrupting the LINC complex using adenovirus associated virus-mediated delivery of DN-SUN1 to cardiomyocytes [69]. Notably, in epithelial cells, tension generated by integrins around keratinocytes, which then passes through the LINC complex, can be transmitted to the nuclear lamina. This transmission helps maintain the proliferative potential of basal keratinocytes and suggests an adjustable mechanical cue to enhance re-epithelialization in wound healing [70]. In addition, keratin 15, a component of the LINC complex, is found to play a significant role in wound repair by interacting with nuclear constituents [71]. In summary, mechanical links offer considerable potential for mechanical tuning in wound-resident cells.

Yes-associated protein (YAP) and transcriptional coactivator with PDZ-binding motif (TAZ)

The Hippo pathway and its downstream effectors, YAP and TAZ, emerge as key players in mechano-transduction (Fig. 2b) [72]. In the cytoplasm, the nuclear Dbf-2-related (NDR) family of kinases, primarily large tumor suppressor 1/2 (LATS1/2), directly regulate the phosphorylation of YAP/TAZ. Mechanical cues and signals transmitted through integrins inhibit the phosphorylation of YAP/TAZ, either directly or by inactivating LATS1/2 or activating protein phosphatase 1α. Phosphorylated YAP/TAZ (p-YAP/TAZ) in the cytoplasm binds 14-3-3 proteins, and is subsequently degraded by proteasomes. Hypo-phosphorylated YAP/TAZ enters the nucleus to regulate transcriptional factors, primarily the transcriptional enhanced associate domain family [73, 74]. YAP/TAZ target genes involved in fibrogenesis, such as CTGF, plasminogen activator inhibitor 1 (PAI-1), and the lysyl oxidase (LOX) family of collagen crosslinking enzymes [73]. In dermal fibroblasts, YAP1 can be activated by TGF-β1 and acts as a mediator driving the transition of fibroblasts into myofibroblasts [75]. Fibrogenesis mediated by YAP is also explored in lung and kidney [76, 77]. In the context of cutaneous lesions, YAP/TAZ shuttles between the cytoplasm and nucleus before or after engaging in different signaling cascades, exhibiting complex roles. ECM stiffness and cell geometry regulate the subcellular localization of YAP in mammary epithelial cells through Rho GTPases and cytoskeletal tension [78]. Blood flow, which exerts shear stress on endothelial cells, inhibits YAP/TAZ activity, thereby suppressing pro-inflammatory gene (IL6, IL8, and selectin E) expression related to JNK signaling [43]. Hierarchical regulation exists between FAs and YAP [79]. Study of anisotropic forces suggests that YAP translocation occurs after stretch parallel to the long axis of elongated cells but not perpendicular to the stretch [80]. In mouse skin, YAP activity may activate plasminogen activator, urokinase (PLAU) and TGF-β receptor III to promote keratinocyte proliferation [81]. YAP-mediated mechano-transduction also holds the potential for immune modulation [82]. In postnatal wound healing, regulated by mechanical tension and YAP signaling, Engrailed-1 lineage-negative fibroblasts transform into Engrailed-1 lineage-positive fibroblasts, which are known to play a role in scarring [74]. Therefore, modulating YAP signaling can facilitate scarless wound healing, which indicates a direction for investigating the mechanical interactions between biomaterials and cells.

Inspiration from mechano-transduction for biomaterial design

Biomaterial stiffness can orchestrate ordered tension, guiding the speed and direction of cell migration reliant on the expansion and contraction of mechanosensitive cytoskeletal elements. Varied shear forces, produced by liquids flowing over biomaterial surfaces of differing roughness, can regulate the fate of circulating blood cells, including vascular endothelial cells, monocytes, and macrophages. These cells naturally experience shear forces physiologically. Deformable biomaterials have the capacity to generate tensile forces, impacting traction-sensitive tissues and organs like neuronal axons and dermal tissues. The adaptability and plasticity of these biomaterials allow for the construction of an appropriate mechanical environment, influencing cell behaviors effectively. The ability to finely tune the mechanical properties of biomaterials offers a promising avenue for controlling and manipulating cell fates during wound healing, providing a framework to optimize tissue repair and regeneration.

Bioactivities of biomaterials due to biophysical characteristics

Biomaterials, owing to their physical properties such as shape, stiffness, elasticity, topography, and static strength (which endows biomaterials with the ability to withstand tension, shear stress, compression, etc.), play a crucial role in regulating mechanical signals during their interaction with cells and the wound environment (Fig. 3a, b). These biomaterials are able to influence cell plasticity through regulating cell adhesion, cytoskeleton, nuclear state, as well as epigenetic modifications, which offers new insights into the fate transformation of wound-resident cells. Furthermore, biomaterials influence the speed and direction of cell motility and regulate tissue ingrowth. Their responsiveness to both biomechanical and biochemical cues allow them to adapt to the wound healing process. Importantly, the physical properties of biomaterials are tunable through various methods, making it feasible to develop biomaterial-based strategies to facilitate wound healing by mechanical means. Bioactivities of biomaterials are summarized in Table 1 [6, 7, 47, 57, 78, 83,84,85,86,87,88,89,90,91,92,93,94,95,96,97,98,99,100,101].

Fig. 3
figure 3

Biomaterials mechanically interact with wound environment. a A wide variety of biomaterials (e.g., polymer, mesoporous, liposome, iron/silica) can be designed for wound healing. These materials actively or passively match wound characteristics to provide optimal condition for wound healing. b Adaptive materials: adaptive materials actively respond to internal or external stimuli (e.g., pH, temperature, light and magnetism) to change their characteristics (e.g., size, stiffness, porosity, topography) or degrade. c Biomaterials for cell motility: engineered biomaterials have the potential to enhance the motility of cells by offering suitable motility speed and direction within a three-dimensional (3D) movement environment. Creating different pore sizes within materials is an important means to achieve multi-dimensional cell movement. d Biomaterials for tissue ingrowth: precisely designing the degradation modes and 3D structure of biomaterials plays a pivotal role in allowing tissue ingrowth. Neogenic vessel and neurofiber could gradually grow as materials degrade. e Biomaterials for immune ecology: biomaterial surface, topography, wettability, and stiffness regulate phenotype of macrophages, neutrophils (innate immune) as well as T cell activation (adaptive immune). Created with BioRender.com

Table 1 Bioactivities of biomaterials with specific physical property

Modulating extracellular signaling

Biomaterials modulate extracellular signaling to profoundly impact cellular activities, which suggests the potential for in situ mechanical reprogramming [102]. It is increasingly recognized that optimal cell adhesion is strongly influenced by the physical characteristics of biomaterial surfaces, including electrical charge, topography, and roughness [103, 104]. Key molecules from the extracellular or provisional matrix, such as fibronectin, vitronectin, collagen, laminin, or fibrin, play crucial roles in mediating cell adhesion to artificial biomaterials [105]. Whether biomaterials are employed in vivo or in vitro settings, these extracellular components are naturally drawn to the biomaterials from culture media, blood, and other bodily fluids, or they are deposited by resident cells. This process is essential for cells to establish FAs by binding integrins. Generally, positively charged surface is more conducive to the formation of adhesion because adhesion-mediating ECM molecules are usually negatively charged. While, different cell types have different responses to surface morphology created by various biomaterials. Distinct spatial dimensions affect the maturation of FAs. When cultured on polymerized high internal phase emulsion foams with closed [two-dimensional (2D)] or open [three-dimensional (3D)] pore morphologies, human embryonic stem cell-derived mesenchymal progenitor cells (hES-MP) display enhanced adhesion and cell viability on those with open pores because this open pore structure provides 3D attachment which is better for cells to exert comprehensively interaction [83]. When in the same spatial dimension, anisotropic surfaces prepared through diverse techniques, e.g., lithographic, microfabrication, electrospinning, can induce cell reorientation following microgrooves or aligned bumps and indentations, the so-called contact guidance phenomenon [84]. Micropatterned or planar surface of fibronectin-coated polydimethylsiloxane (PDMS) substrate influence behaviors of C3H10T1/2 cells (murine mesenchymal stem cells). Clear differences between the FA formation and maturation patterns are observed: cells on micropatterned surface displayed more orderly, quicker, and greater FA formation [85]. Therefore, tissue regeneration with specific structure can be directed by specific patterned surface. Nanoscale structure seems to be superior to microscale because basement membranes of various tissues are composed of a complex mixture of nano size (5–200 nm) pits, pores, protrusions, and fibers [86]. Therefore, more diverse cell-specific effects occur on nanoscale surfaces, which is more resemble to physiological conditions. For human mesenchymal stem cells, surface nanopatterning controls the initial assembly of FAs [84], while primary human fibroblasts on nanoscale roughness surface display reduced formation of FA complexes, less spread and an elongated morphology [86]. Still, these biomaterial-based strategies can only provide static surface which are different from real dynamic conditions within organisms. Recent technology has pushed the limits of static control by using shape memory polymers (SMPs). Human cardiomyocytes display different formation of FAs on SMPs coated with polyelectrolyte multilayer which has achieved dynamic nano-wrinkle formation. Specifically, FA proteins like zyxin, vinculin, paxillin align along the wrinkle direction [106]. In addition to inducing novel formation of FAs, biomaterials can also induce FAs sliding thus influencing cell polarization [107]. All of the above are materials that come into contact with cells and thus affect the formation of FAs, non-contact method is developed to exert external mechanical forces through using microfabricated arrays of magnetic and non-magnetic silicone elastomeric posts, and as a result, enhanced FA formation is observed [107]. Biomaterial-based controllable formation of FAs dictates restoration of specific cellular functions. The effect of microenvironment stiffness on the cytoskeleton has been extensively studied. Primary human aortic smooth muscle cells within 3D poly(ethyleneglycol)-fibrinogen hydrogels increase their assembly of F-actin stress fibers and FAs when rigidity increases [104]. Chondrocytes on silicon-based elastomer PDMS substrates with different substrate stiffnesses show changes of secreted extracellular linkage protein, laminin β1, FA complex protein, FAK as well as cytoskeleton reorganization [108]. Evidences show that substrate stiffness mediates the formation of novel cytoskeletal structure within mouse embryonic fibroblast 3T3 cells and rat Schwann cells [109, 110]. Accelerated osteoclast differentiation through cytoskeleton rearrangement is observed on stiffer PDMS substrates which is firstly manifested in variations of their morphology and fusion/fission activities, then in the upregulation of canonical osteoclast markers as well as the activation of cytoskeleton-associated adhesion molecules, including fibronectin and integrin αvβ3, followed by biochemical signaling cascades of paxillin, FAK, and RhoA [111]. It seems that the stiffness that best suits the function of a cell often matches the stiffness of its internal environment. Collectively, the physical properties of biomaterials affect extracellular signaling through regulating cell adhesion.

Influencing cellular epigenetics

Leveraging mechanical perturbations, biomaterials hold promise as a strategy to overcome epigenetic barriers and control cell fate because mechanical signals from biomaterials are transmitted from the cell membrane to the nucleus, directly influencing chromatin structure and gene expression [112]. Epigenetics encompass heritable alterations in gene function that occur without modifying the DNA sequence, ultimately resulting in observable changes in an organism’s traits or characteristics. The epigenome plays a crucial role in maintaining the stability of cell identity, especially in situations such as stem cell differentiation and somatic cell reprogramming, where cellular states can become disrupted. Epigenetic regulation involves common mechanisms such as chromatin remodeling, DNA methylation, and histone protein modifications. For instance, the stiffness of hydrogels plays a key role in reprogramming mouse embryonic fibroblasts into induced pluripotent stem cells through mesenchymal-to-epithelial transition and the regulation of stemness markers. Stiff surfaces promote an open and transcriptionally active euchromatin structure, while soft biomaterials induce a dense and transcriptionally inactive heterochromatin structure [23]. Nanomaterials, like nano-hydroxyapatite, have the capability to influence osteoblast lineage commitment by promoting DNA methylation and regulating the expression of key osteoblastic marker genes [87]. When human embryonic stem cells are cultured on nano-topographical substrates, they exhibit increased expression of mesenchymal and stromal markers, along with elevated levels of early osteogenic progenitors, compared to cells cultured on flat substrates in the same basic media conditions. Additionally, during substrate-induced differentiation, these cells undergo epithelial-to-mesenchymal transition, and they display DNA methylation changes akin to those induced by chemical factors [88]. Micro-grooved substrates enhanced the directed differentiation of cardiac progenitors into cardiomyocyte-like cells through promoting histone H3 acetylation [89]. Increasing accessibility of DNA and epigenetic modification is the prerequisite for cell fate turnover.

Supporting cell migration

Engineered biomaterials have the potential to enhance the flexible motility of cells by offering suitable motility speed and direction within a 3D movement environment [113] (Fig. 3c). Cells are inclined to migrate towards regions that match their optimal stiffness, where they can generate maximal traction [114]. Skin rupture disrupts the harmonious cellular community. Wound-resident cells, including epidermal stem cells, fibroblasts, and endothelial cells, mobilize collaboratively to facilitate wound closure. It’s noteworthy that platelet spreading and activation intensify when adhered to fibrinogen-coated polyacrylamide (PA) gels with greater stiffness [90]. Endothelial cells are more active on stiffer PDMS [115]. Keratinocyte cytoskeleton adapts to matrix rigidity on PA hydrogels [61]. Fibroblasts influenced by stiffness may lead to over-recruitment and fibrosis [116]. Hence, materials featuring stiffness gradients hold the potential to guide the migration of wound-resident cells and enhance their motility, ultimately improving the efficacy of wound healing. Numerous techniques can be employed to create materials with stiffness gradients. One approach involves segmentally controlling the proximity of reactants to regulate the degree of crosslinking. To illustrate, by leveraging differential diffusion distances of unreacted cross-linkers and monomers within a pre-polymerized hydrogel matrix, PA hydrogels were successfully engineered. These hydrogels exhibit linear stiffness gradients ranging from 0.5 to 8.2 kPa/mm, covering the mechanical spectrum observed in both physiological and pathological in vivo conditions [117]. Moreover, another effective approach involves overlaying films with varying moduli. For instance, one can position a thin, low modulus PDMS film, atop a high modulus PDMS structure. This technique not only allows the creation of flat surfaces with distinct stiffness patterns but also provides a means to introduce surface undulations [118]. Detecting micro-level stiffness is challenging, prompting the application of computational modeling based on filopodia mechano-sensing to predict and understand directed cell migration [91]. While proper stiffness facilitates flexible 2D cell motility, porosity is essential for enabling cells to infiltrate various anatomical layers, thereby offering a more realistic stereoscopic environment. Engineered porous biomaterials are designed to segregate different cell types and govern their interactions. By controlling the size of the pores, for instance, one can confine fibroblast activity to the dermal layer, allowing precise control over cellular behavior at distinct anatomical levels. A recent innovation involves the development of 3D porous films composed of chitosan, pectin, and ZnO, featuring pore sizes ranging from 99 to 126 nm. These films have been designed to expedite scarless wound healing [92]. Porous substrates promote the migration of endothelial cells, thereby facilitating and enhancing the process of wound healing [93]. Engineered porous β-tricalcium phosphate scaffolds have been utilized to address bone defects by stimulating cell migration, proliferation, and angiogenesis. These scaffolds serve as potential supporters of wound-resident cells [6]. Therefore, biomaterial-based biomimetic scaffolds are interesting to regulate cell motility.

Allowing tissue ingrowth

Precisely designing the degradation modes, 3D structure, and electrical properties of biomaterials plays a pivotal role in allowing tissue ingrowth [119] (Fig. 3d). Timely degradation of these materials is crucial to avoid unnecessary clinical interventions which is detrimental to developing tissue. As biomaterials degrade into smaller components, they not only create space for tissue growth but also enable the controlled release of trophic factors, including growth factors, cytokines within specific spatial and temporal constraints of the wound site. Several factors influence the degradation of biomaterials, including environmental factors such as temperature, pH, and fluid dynamics, as well as material attributes like porosity, surface patterns, and shape. The mode of degradation varies depending on the type of materials used. Materials like polylactide, polyglycolide, and polycaprolactone rely on hydrolysis, but their degradation rates differ, and direct contact with water molecules significantly affects the degradation process. Consequently, during the early phase of wound healing, when there is a substantial amount of fluid effusion, the degradation of hydrolytically unstable materials can be accelerated [120]. Premature degradation compromises the biomaterials’ ability to serve as a barrier against external harmful factors. As the proliferative phase begins, it’s imperative that biomaterials do not occupy excessive space, as more room is needed for tissue ingrowth. Hence, creating a 3D environment through sequential degradation for flexible cell movement serves as the foundation for tissue growth. Zhao et al. [121] and Xu et al. [122] have focused on developing a silicone rubber membrane (SRM). They initially crafted a single-layer SRM with an adjustable microporous structure, achieved through solvent evaporation-induced phase separation. Then, they constructed a silicone rubber membrane bilayer (SRM-B) consisting of upper and lower layers differing in thickness, pore size, and pore density. The smaller pores serve to prevent bacterial infection, while the larger ones promote cell adhesion and proliferation, thereby expediting re-epithelialization [121, 122]. Nanoscale biomaterials are easier to degrade and be absorbed because of the small size effect, which provides a higher specific surface area. The nanoscale dimensions make biomaterials more adept at mimicking the in vivo physical environment, thereby enhancing their effectiveness in therapeutic applications [5, 123,124,125]. In recent years, the electrical properties of biomaterials have gained attention as a promising parameter to replicate native biological electric activity. The concept of an enzymatic biofuel cell was initially introduced in 2015 as a means to serve as an electrical reservoir, generating electric stimuli to support regeneration and repair processes. By employing enzymes like glucose oxidase and bilirubin oxidase, enzymatic biofuel cells have demonstrated remarkable performance in muscle tissue regeneration, facilitating cell proliferation, differentiation, and migration [126]. A similar device has been applied to create a wound dressing, which combines a porous PA hydrogel, referred to as an “electricity auto-generating glucose-responsive enzymatic-biofuel-cell skin patch”. When this innovative patch is employed on wounded rat skin, it significantly improves the efficiency of wound closure, neovascularization and matrix deposition, demonstrating its potential as a valuable tool for wound healing [127]. While cells exhibit specific behaviors, achieving tissue ingrowth necessitates the establishment of connections and orderly arrangements. A comprehensive understanding of skin anatomy through tomography serves as inspiration for designing biomaterials with stereoscopic structures, ensuring adequate space for the growth of various tissues. The exploration of multifunctional composite biomaterials that facilitate tissue growth should be a priority for further research and development.

Adapting to wound environment

Technological advancements have shown that biomaterials with favorable physical properties can be tailored through various methods to adjust and adapt to the wound environment, thereby optimizing the conditions for regenerative healing (Fig. 3b). When breaches occur, the continuity of the skin is disrupted, leading to physical and chemical changes such as unevenly distributed mechanical tension, alterations in electric fields due to injury-induced ion redistribution, shifts in temperature, pH levels, and metabolite concentrations. Hydrogels have consistently shone in the field of wound healing. They can be derived from various sources, including gelatin, collagen, fibrin, hyaluronic acid, chondroitin sulfate, alginate, chitosan, poly(ethylene glycol), and poly(vinyl alcohol) [128]. While hydrogels share similarities with the ECM, their inherent lack of stiffness limits their ability to withstand tension and support tissue ingrowth effectively. To address this challenge, researchers focus on modifying gelatin hydrogel, creating variants with diverse mechanical properties using various methods. One approach involves directly reacting gelatin with methacrylic anhydride, resulting in the synthesis of gelatin methacryloyl (GelMA). The degree of methacrylation plays a crucial role in determining the elastic modulus of GelMA. Additionally, by incorporating self-assembled chitin nanofibers, micropatterned GelMA hydrogels with enhanced stiffness and strain properties have been developed. These modified hydrogels show the capacity to induce cell differentiation and vasculo-genesis, simplifying their utility in regenerative applications [100]. Self-adaptivity of biomaterials can be triggered by factors such as temperature, pH, metabolite concentrations, or external stimuli like light of varying wavelengths, magnetism, and electrical stimulation. In particular, fibrous GelMA scaffolds show superior performance compared with GelMA hydrogels. This is attributed to their ECM-like nanofibrous structure, which provides a higher surface-to-volume ratio, facilitating closer interaction between the materials and the wound environment. To simplify the process, researchers combined radical photo-crosslinking with reactive methacryloyl groups. This approach achieves the temporal and spatial control of the physical properties of GelMA hydrogel, including water retention, stiffness, strength, elasticity, and degradation, simply by altering the exposure time to light [129]. Contact-free methods for handling materials are preferred to avoid disrupting the healing process. In addition, temperature plays a crucial role in wound healing. Considering the temperature difference between deep wounds (37 °C) and skin surface (25 °C), a hydrogel with dual characteristics of fluid-like and solid-like properties has been developed. This innovative hydrogel is designed to provide a soft setting in deep wounds, while also serving as a robust barrier for irregularly shaped skin wounds. The inspiration for this ingenious self-adaptive hydrogel is drawn from the temperature-dependent viscoelasticity of polymers formed by borate ester bonds [130]. The pH value undergoes frequent changes during the healing process, which can be detrimental to biological reactions. Healthy skin typically maintains a pH range of 4–6. In contrast, wounds with pus, necrotic tissue, and serum crusts often exhibit a mean pH value of around 6.1, while chronic or infected wounds have a pH of approximately 7–8. Interestingly, wound debridement appears to lead to an increase in pH levels [131]. A pH range of 4–6 is considered conducive to effective wound healing, and pH-sensitive biomaterials can be employed to maintain an optimal pH environment within wound. In the case of protein-based hydrogels, their pH sensitivity is attributed to the presence of ionizable amino acids. Elastin-like polypeptides (ELPs) can be designed to incorporate ionizable amino acids like lysine, tyrosine, and valine. These ELPs can then be crosslinked to create hydrogels with adjustable pH sensitivity through chemical modification [132]. In order to monitor pH fluctuation within wound, pH-sensitive PDMS optical fibers are integrated into carborxymethy chitosan-protocatechualdehyde (PA)@Fe hydrogels to create a smart visible monitor system [133]. Furthermore, in the case of chronic wounds, particularly those associated with diabetes, elevated sugar levels can be leveraged in the design of responsive biomaterials. This responsiveness is achieved through the competitive binding of sugar to poly(amidoamine) dendrimers bearing phenylboronic acid (PBA-PAMAM). Notably, layer-by-layer thin films that incorporate PBA-PAMAM can degrade in response to changes in sugar levels [134]. Moreover, responsive biomaterials can enhance advanced strategies for wound healing. For instance, somatic cells undergoing in situ differentiation into pluripotent cells endow wounded skin with remarkable regenerative potential. However, during the process of somatic cell reprogramming to pluripotent cells, several changes occur, including shifts in extracellular pH from alkaline to acidic, alterations in metabolic pathways from oxidative phosphorylation to glycolysis, and rearrangement of the cytoskeleton. To harness these changes, a cell reprogramming-responsive hydrogel is developed. This hydrogel is designed to facilitate the expansion of cell clusters by adjusting its mechanical properties in response to the aforementioned alterations, thereby promoting the transformation of cell fate and enhancing regenerative healing potential [135]. Biomaterials can likewise be engineered to be responsive to various factors associated with scarring such as EMT, contraction forces, and fibroblast activation. These responsive biomaterials can play a pivotal role in regulating collagen deposition, alleviating mechanical tension, and influencing the behavior of fibroblasts, ultimately contributing to improved wound healing outcomes by orchestrating these critical cellular and mechanical processes.

Potential of biomaterial-based mechanical regulation for skin appendage regeneration

Mechanical regulation has been demonstrated effective in tissue development and regeneration [136]. On the one hand, biomaterial-based mechanical regulation is expected to provide a scar-free environment which is an important prerequisite to regenerating skin appendages. On the other hand, biomaterials regulate cellular plasticity through mechanical means which will be an effective way to expand the precursor cell bank. Biomaterials for skin appendage regeneration are discussed below.

Scarless environment: a prerequisite for skin appendage regeneration

Post-injury tissue fibrosis repair often results in a reduction of functional tissue structure and cell diversity, accompanied by an increase in fibrous tissue formation. This shift can be detrimental to the restoration or regeneration of skin appendages [137]. Therefore, the improvement of the fibrotic environment is essential for successful appendage regeneration. From the perspective of embryogenesis, the formation of skin appendages depends on the crosstalk of the dermis with the epidermis. Specifically, scattered cells within the epidermal plane produce a higher WNT signal than their neighbors, and these cells cluster into placodes [138, 139]. If the underlying mesenchyme produces elevated levels of bone morphogenetic protein (BMP) inhibitors, the WNThigh cells will form an HF [140]. If the underlying mesenchyme produces a strong BMP signal, the WNThigh cells will make a SwG [137]. Postnatally, close intercellular interactions between epithelial and dermal cells maintain and control the hair growth. Signals or soluble factors from dermal cells in hair bulbs directly influence the adjacent epithelial cells [141, 142]. However, in the context of scarring, dermal fibroblasts which are the main producer of mesenchyme signals are activated by various stimuli to experience phenotypic transformation to myofibroblasts. This transformation rapidly creates a pro-fibrotic environment within wound in a cascading manner, thus completely destroying the mesenchymal environment in which appendages grow [143,144,145]. Therefore, we highly confirm the necessity of biomaterial-based scarless wound healing. Bioactivities of biomaterials with specific physical properties make biomaterials a promising avenue for preventing scarring by mechanical means. Specifically, elastomeric materials can be employed to regulate collagen deposition and biomaterials with specific surface patterns and topographies can facilitate functional neovascularization and the restoration of innervation. Given the diverse biological processes involved, biomaterials often need to be tailored to specific requirements and these can be translated into quantifiable parameters.

Elastomeric materials

Scars are characterized by excessive collagen deposition, which is a mechanical cue-dependent process. Elastomeric materials are reported to alter collagen deposition and arrangement mechanically, and thus, they have emerged as promising candidates to prevent scar formation [146] (Fig. 4a). Previous studies have revealed that collagen deposition and arrangement are influenced by the magnitude and direction of skin stretching tension-caused mechanical forces [147]. Collagen fibers prefer to align in the direction of mechanical forces, inspiring the development of elastomeric materials to regulate tension mechanically for scar prevention [147, 148]. Elastomeric materials are mainly fabricated based on various natural and synthetic polymers, and their deformability enable them to offload tension within the wound, or change tension direction [149], which provides the mechanism basis for applying elastomeric materials to regulate collagen. Currently, it has been reported that elastomeric materials (e.g., silicone gel, poly fibers and electro-spun micro-fibrous scaffolds) can regulate collagen deposition and arrangement through a diversity of molecular biology events. For instance, traditional silicone gel sheeting has been used for an extended period to prevent and treat hypertrophic and keloid scars [150, 151]. Silicone surfaces can interact with collagen and further lower the effective concentration of collagen monomers available to form fibrils. Besides, because the silicones can locally act as another “binding partner” in the fibrosis process in vivo, that interaction also leads to conformational changes in the collagen molecules [152]. Poly(3-hydroxybutyrate-co-3-hydroxyvalerate) (PHBV) fibers, as a widely used viscoelastic material, have the ability to regulate collagen expression level and increase the ratio of collagen I/collagen III, resulting in the inhibition of skin scar [153]. Electro-spun nanofibrous PHBV meshes, characterized with elastic fibrous networks and high porosity, reduce collagen deposition through blocking fibroblast-myofibroblast transition, lowering pro-fibrosis factor TGF-β1, and increasing anti-fibrosis factor TGF-β3. In addition, healed skin treated with electro-spun nanofibrous PHBV meshes is softer and more elastic. This highlights the versatility and effectiveness of elastomeric biomaterials in promoting scarless wound healing and skin regeneration [7]. Similar results are also observed in electro-spun micro-fibrous scaffolds crafted from the copolymer poly(L-lactide-co-ε-caprolactone), and that decreases scar formation [154]. The upgrade in fabrication methods of elastomeric material has expanded the possibilities for using elastic materials in scar treatment and prevention. One notable example is that photo-crosslinking technique endows elastomeric materials ultraviolet light-responsiveness. Utilizing riboflavin’s biophoton sensitivity, a photocurable silk fibroin hydrogel is fabricated. In this process, riboflavin is triggered by ultraviolet light to generate active oxygen radicals, which induces chemical crosslinking of amino, phenol, and other groups within the silk fibroin macromolecules, resulting in the formation of a photocurable hydrogel. These hydrogels exhibit remarkable resilience after compression and show satisfactory cytocompatibility, making them promising candidates to optimize collagen deposition [155]. Drawing inspiration from the nucleophilic Michael addition reaction, which involves the reaction between nucleophiles and activated alkynes, researchers have successfully created an easily processable elastomeric polyamide material with shape memory properties. This innovative material can mimic skin deformation well, so it has potential to regulate collagen deposition and arrangement [156]. By integrating 3D printing technology, researchers have developed a faster and more energy-efficient approach to producing a robust ordered mesoporous elastomer. This elastomer exhibits an ordered nano-microstructure and a molecular multinetwork, which together confers hierarchical toughening properties. Importantly, it maintains excellent stiffness and elongation, comparable to commonly used engineering elastomers like silicone and vulcanized rubber [157]. Furthermore, natural materials such as elastin and resilin, which are highly elastomeric, open up additional possibilities for developing elastomeric materials with unique properties and applications in various fields [158]. In summary, advances in elastic materials will facilitate better application in the regulation of collagen deposition and arrangement to prevent scar.

Fig. 4
figure 4

Biomaterial-based mechanical facilitation for scarless wound healing. Mechanical signals from biomaterials with specific physical properties are transformed to computable parameters by biosensor system. a Elastomer generates mechanical forces of different magnitudes and directions by stretching and contracting. b The hollow design of the microtubule structure allows for the mimicking of blood vessels and the inner wall simultaneously detects the velocity of the liquid as it flows through and the magnitude of the shear stress. c Patterned surface directs the growth of elongated tissue and it can mimic various two-dimensional (2D) structure of the matrix in vivo. d Surface with different stiffness renders cells different abilities to move. This is related to the stromal environment in which cells are physiologically located. e Viscoelasticity gives biomaterials solid-like properties such as elasticity, strength, and consequential stability as well as liquid-like properties such as flow properties that vary with time, temperature, load magnitude, and rate. This trait is more similar to the physiological situation. f Electrical conductivity is prevalent in living organisms, such as the resting and action potential of membrane potential. Biomaterials with electrical conductivity can be used to detect and simulate the required electrical signals within body. g Biomaterials and mechanical signals for cells: biosensor systems translate biomaterial-based mechanical signals into computable parameters like electricity stimulation, mechanical tension, magnetic force and friction. These quantitative parameters are then correlated with cell behaviors of interest (e.g., orienting traction, fibroblast activation, nerve growth, cell adhesion) through mathematical models. hi Finally, precise regulation of cellular behavior can be achieved through biomaterial-based mechanical regulation whether in vitro (e.g., multidimensional culture of cells) or in situ (e.g., activating endogenous cells). Created with BioRender.com

Tubular biomaterials

Considering that aberrant neovascularization can hinder scarless healing, there is a growing interest in achieving a well-organized vascular network rather than a disorganized vascular cluster through tubular biomaterials [159]. The organization and alignment of endothelial cells are pivotal for the formation of lumen-like structures and the development of well-structured blood vessels. A previous study has devised biodegradable conduits for the reconstruction of lymphatic vessels by facilitating the alignment and migration of lymphatic endothelial cells [94]. The formation of vessels is influenced by the balance between the magnitude of cell traction and the apparent stiffness of surrounding gel. In addition, vessel formation is less likely to occur in a very soft environment, highlighting the significance of the mechanical properties of surrounding tissue in regulating vascular network development [160]. Tubular biomaterials with suitable stiffness play a role in regulating the delicate balance between the growth and regression of blood vessels, and therefore, indicate a viable strategy to direct the well-organized neovascularization (Fig. 4b, d, e). In this strategy, shear stress is a crucial mechanical parameter which refers to the frictional force exerted by the flowing blood on the vascular endothelium, occurring parallel to the inner wall of blood vessels. The mechanical stimulus by shear stress can promote the development and remodeling of blood vessels, enhancing the process of neovascularization in tissues and contributing to their overall health and functionality [161,162,163]. An example is the use of a tubular collagen scaffold with shape memory for repairing tubular organs. This scaffold exhibits reversible expansion in response to increased luminal pressure and shear stress which further provides suitable attaching surface for endothelial cells [159]. This shear stress-responsive tubular collagen scaffold holds promise for well-organized neovascularization within wound. In addition, pre-implantation of cells into a specific tubular microfluidic system can promote the formation of blood vessels in vitro and normal vascular function [164]. In this setup, blood circulation flows through the lumen of these microtubes and shear stress occurs. Simultaneously, endothelial cells are carried by the blood flow and settle randomly along the inner surface of the microtube. These endothelial cells sense the shear stress generated by the flowing blood, leading to enhanced neovascularization. Tubular structures can be produced through various fabrication methods. Polymers such as polylactide-polycaprolactone, polylactide-co-glycolide, and polyhydroxyethyl methacrylate can be electro-spun to create tubular vascular grafts to promote angiogenesis and tissue regeneration [165]. Compared with synthetic materials, natural biopolymers are more conducive to cell attachment, migration, and proliferation when used as scaffolds. A novel biomimetic silk fibroin tubular scaffold crosslinked by poly(ethylene glycol) diglycidyl ether (PEG-DE) consists of three-layer structure: a regenerated SF intima, a silk braided media, and a regenerated SF adventitia. This silk fibroin tubular scaffold forms a porous layered tube with superior mechanical, permeable and cell adhesion properties that are beneficial to vascular regeneration [166]. Other forms of tubular materials include chitosan-carbon nanotube in the form of tubular hydrogel based on electrodeposition, tubular scaffold with circumferentially aligned nanofibers and so on [167, 168]. Collectively, various novel tubular biomaterials can be utilized to direct neovascularization through well-organized endothelial cells.

Specific-patterned biomaterials

In order to improve nerve restoration for better skin functional regeneration, specific-patterned nerve guidance conduits (NGCs) are employed to bridge the gap between the proximal and distal nerve stumps within lesions [169, 170]. These NGCs can be designed with diverse topographic features and provide appropriate mechanical environment for the growth of nerve tissue [171] (Fig. 4c–f). Plenty of evidences show that nerve outgrowth is mechanosensitive. Axon elongation can be induced by stretch forces [172]. Schwann cells sensing magnetic force-based mechanical stimulation accelerates peripheral nerve regeneration [8]. Electrospinning and micropatterning are commonly used technologies to create specific topographical features in NGCs. Electro-spun fibrous scaffolds are particularly appealing due to their high surface area-to-volume ratios, which promote cell attachment and growth. Micropatterning NGCs are characterized by smooth inner surfaces and longitudinal nanogrooves on the fiber surface. Schwann cells on anisotropic PA hydrogel micropatterned with aligned ridge/groove structures show obvious alignment growth and elongated neurites than on flat hydrogel [95, 173]. In addition, silk protein and polycaprolactone have also been used to construct composite anisotropic topography in the repair of peripheral nerve injury in adult rats [101, 169]. The height and angle of the groove could also influence neurites elongation. When culturing murine embryonic cortical neurons on micro-grooved platforms with a depth of 2.5–69.0 μm, axons are found to cross over the shallow grooves of 2.5 or 4.6 μm, whereas grooves of 22.0 or 69.0 μm cause the axons to turn after contact [174]. Except for aligned nanofibers and ridge/groove structures, pillars/posts, nanorough surfaces and surface stiffness are all common strategies used to modify the surface of NGCs [175]. For example, surface stiffness affects dorsal root ganglion (DRG) proliferation and differentiation in that soft hydrogel promotes the elongation of DRG’s axon by upregulating gene expression of Ntn4 and Unc5B [176]. Despite the positive effect of topography on nerve tissue, it is difficult to achieve the desired results by solely relying on the topographic features of NGCs. Therefore, numerous biomolecules such as fibronectin, laminin, peptide, and growth factor have been immobilized in NGCs via physical adsorption, covalent grafting, or chemical crosslinking for accelerating nerve regeneration [101]. To sum up, specific patterned biomaterials act in concert with biochemical cues to enhance or improve the efficiency of nerve restoration.

Immunomodulatory biomaterials

Macrophages and neutrophils play a prominent role in responding to biomaterials and this inspire the development of immunomodulatory biomaterials for scarless wound healing [177]. Biomaterial surface, topography, wettability, and stiffness have been proved to regulate phenotype of macrophages and neutrophils. Rough surface produces more friction which promotes monocytes migration from the bloodstream into the wound area [178, 179] (Fig. 3e). Substrates with varying stiffness can exert mechanical forces on cells. For instance, when exposed to PA gels coated with fibronectin, macrophages on stiffer substrates exhibit an eightfold larger spreading area and a faster proliferation rate compared to those on softer substrates [96]. Similarly, using engineered 3D fibrillar matrices with natural biopolymers collagen I and glycosaminoglycans to mimic matrix with changing stiffness, macrophages preferentially turn to pro-inflammatory phenotype on stiffer substrate [97]. Ion channel Piezo1 can also be activated by cyclic stress to regulate macrophage polarization [180, 181]. As for the surface roughness and wettability, increased surface wettability had a stronger macrophage activation effect than increases in roughness [182]. Desirable cell and tissue response to an implanted biomaterial with specific topography can be achieved. For example, IL-1β produced by macrophages on expanded polytetrafluoroethylene with 4.4 μm pore (4.4-ePTFE) is 15 times higher than that from macrophages on non-porous PTFE [183]. Nanoscale surface topography of titanium (Ti) enhances the inflammation-related M1 phenotype in J774.A1 macrophages [184]. Fiber diameter of electro-spun poly(L-lactic) (PLLA) scaffolds influences the activation and protein secretion of RAW 264.7 macrophages in that nanofibrous PLLA scaffolds impair the inflammatory response mediated by macrophages when compared with films and micro-fibrous scaffolds [185]. Random fiber orientation topography promotes a pro-inflammatory signature in macrophages where higher percentage of CCR7+ cells appear on disorganized polycaprolactone nanofiber substrates, indicating a polarization shift towards a pro-inflammatory (M1-like) phenotype [186]. In a septic lung injury study, inhibiting Rho-kinase signaling with Y-27632 decrease the accumulation of neutrophils [187]. When exposed on smooth, rough, and rough-hydrophilic modified Ti surfaces, neutrophils show differences in protein secretion and NET formation. Specifically, anti-inflammatory cytokine (IL-4, IL-10) secretion is enhanced by rough hydrophilic Ti surfaces, and larger NETs are formed on rough surface [188]. Except for innate immunity, Griffin et al. [189] interestingly found that microporous annealed particle scaffolds could hasten the regenerative healing process by activating adaptive immune response. Therefore, immunomodulatory biomaterials show great potential in promoting scarless wound healing.

Expanding stem or progenitor cell bank

Biomaterials regulate cellular plasticity through mechanical means, which will be an effective way to expand the stem and precursor cell bank [112, 190]. Various stem or progenitor cells are involved in the development of HF and SwG and they are influenced by mechanical forces from biomaterials [23, 191, 192], which is summarized in Table 2 [98, 193,194,195,196,197,198,199,200,201,202].

Table 2 Biomaterials for skin appendage regeneration

The activation of hair follicle stem cells (HFSCs) is enhanced by decreasing mechanical forces from stiff niche in the context of both wound-induced HF neogenesis and aging [203, 204]. Due to the mechanosensitive characterization of HFSCs, an HFSC organoid culture system based on hydrogel has been modified to replicate the properties of young and aged basal membranes, with stiffness levels of 1–3 kPa and 5–6 kPa, respectively. This modification resulted in a noticeable increase in the activation of bivalent promoters, which controls crucial genes responsible for HFSC activation and self-renewal. It’s evident that mechanical forces originating from the niche can effectively suppress the activation of aged HFSCs [204]. Self-assembly peptides hydrogel enhances the proliferation of Akp2+Bmp6+ skin-derived precursors, and more importantly de novo hair genesis in mice [193]. Layer-by-layer self-assembly with gelatin/alginate is used to fabricate nanoscale biomimetic ECM for CD200+α6+ HFSCs population [194]. Natural minerals like silica, ceramic, are also used to fabricate 2D/3D support for HFSCs [195, 196]. Polymer biomaterials used to support hair follicular cells are poly(ethylene-co-vinyl alcohol) (EVAL) and PHBV. Compared with common PHBV medium, hair follicular epithelial cells (outer root sheath) and dermal cells (dermal sheath) cocultured with patterned PHBV nanofiber matrices exhibit greater HF regenerative and repair ability due to increased expression of collagen I, elastin, and α-SMA as well as more dermal papilla produced from dermal sheath [205]. EVAL membranes facilitate dermal papilla self-assembly into many compact spheroidal microtissues that are able to induce new HFs because adhesive surface is needed for quick cell expansion and a biomaterial with a lower adhesivity is required for self-aggregation [197].

Due to the presence of hair cycles, the above potential cells have the innate abundance within skin because they are reservoir for restoring the hair cycle. However, loss of SGCs, including luminal cells and myoepithelial cells, is hard to restore as they are terminally differentiated cells [206]. Autologous SwG-derived progenitors, induced pluripotent stem cells derived from somatic cells as well as fate conversed cells from different lineages are potential bank for forming SwGs [207]. The differentiation of mesenchymal stem cells to SGCs can be enhanced on 3D-bioprinted constructs made from alginate-gelatin blends with high stiffness [98]. Similarly, biomaterials with tunable mechanical properties are used to construct 3D even 4D (time) bionic ECM to induce differentiation of multiple stem cells (e.g., bone marrow-derived mesenchymal stem cells) toward SGCs thus increasing the abundance and quality of SwG progenitor cells [198, 199]. This illuminates a new direction for SwG regeneration based on the mechanical regulation of biomaterials. In addition, according to lineage-tracing, multipotent progenitors may include cells with high expression of keratin 15, CD29, or CD49f [207]. Although the expression of these markers has not yet been confirmed to be influenced by mechanical signals, it is a promising research direction. Biomaterial-based mechanical reprogramming of cell fate provides broader thinking. Self-adaptive biomaterials loaded with SwG regeneration-related signaling molecules (such as ectodysplasin-A, BMP-related molecules) can be used to achieve continuously release to activate endogenous cells like myoepithelial cells and coiled ductal cells thus promoting SwG regeneration in vivo [207]. To enhance the safety and efficiency of cell delivery, SGCs are cultivated on gelatin microspheres, and then this complex is implanted into an engineering skin construct to form SwGs [200]. What’s more, better biological reactor for SwG organoids formation in vitro is of great interest where mechanical signals from biomaterials are exerted to change cell fates. In the meantime, the stereoscopic structure provides cells with better living space and structural guidance. Collectively, biomaterials play an interesting role in SwG regeneration which deserve more explorations from micro mechanism to macro pro-regeneration efficiency.

Supporting multiple appendages

Current in vivo or in vitro strategies for skin appendage regeneration can only restore single type of appendages (Fig. 5a–c). However, considering the native structure of skin where multiple appendages coexist, simultaneous regeneration of various skin appendages is attractive. The perfect repair of the skin must be accompanied by the regeneration of multiple appendages including HFs, SwGs, etc. Various plights hamper this ideal conceive. Biological problems trace back to the development of appendages. There are spatiotemporal antagonism signaling between SwGs and hair fate decision. Specifically, mesenchymal-derived BMP signal suppresses epithelial-derived sonic hedgehog (SHH) signal. BMP signal is for SwGs genesis while SHH signal is for hair. Therefore, hair genesis happens only when BMP is blocked, but it may be detrimental to SwG genesis. Naturally, starting signals for SwG genesis precede that for hair [208]. This chronological difference in timing limits artificial multiple-appendage regeneration. Booming biomaterials science may imply novel strategies. For example, biomaterials with separated chambers are expected to coculture SwG-like cells and hair follicular skin derived precursors in vitro through providing spatial freedom. Strategy that BMP signal is released first, followed by SHH signal can be achieved by smart biomaterials with sequential responsiveness to internal and external stimuli thus solving temporal conflicts. We propose a concept based on biomaterials called “co-culture and multiple skin appendage regeneration” where various appendages coexist (Fig. 5d). In the meantime, constructing highly biomimetic tissue engineering skin needs targeting dermal microcirculation, vascularized organoids as well as 3D bioprinting.

Fig. 5
figure 5

Biomaterials and regeneration of multiple appendages-an ideal skin unit. a, b Traditionally, biomaterials are used as carriers (diverse scales from nano- to macro-, e.g., tubes, rods, wires) to deliver bioactive factors (e.g., drugs, nucleic acid, peptides) or engineered cells (iPSCs) and organoids for tissue repair and regeneration. c In the field of skin wound repair, different combinations of biomaterials and bioactive ingredients have been used to regenerate sweat glands, hair follicles, nerve tissue, and vessel. However, a truly functional artificial skin requires a combination of in vitro and in vivo culture techniques for different cells and tissues. d It is promising to combine these assemblies into an integral construct to form a highly bionic skin unit. Biomaterials due to their variable physical properties can be used to create co-culture scaffolds for skin components, including vessel, nerve, hair follicles, sebaceous glands and sweat glands. In such a bionic skin, suitable conditions are separately established for the survival of various precursor cells (e.g., hair follicular SKPs, Schwann cell precursor, SwGC-like cells) with different regenerative signals to achieve co-culture of multiple skin appendages. Created with BioRender.com. iPSCs induced pluripotent stem cells, SKP skin derived precursor, SwGC sweat gland cell

Conclusion and outlook

In previous studies, in order to regenerate skin appendages, chemical method is always preferred. But accumulating evidences underscores the potential of targeted modulation of mechanical cues to enhance skin regeneration, promoting scarless repair by influencing the extracellular microenvironment and driving phenotypic transitions. In the meanwhile, the field of skin repair and skin appendage regeneration has witnessed remarkable advancements in the utilization of biomaterials with distinct physical properties. Therefore, we propose a global concept for scarless wound healing and skin appendage regeneration based on mechanical regulation combing with biomaterials. Notably, many studies remain confined to in vitro experiments, and even when transitioning to in vivo experiments, the subjects are typically limited to animals, which limits the translational application of biomaterials, making it less efficient. Therefore, it is crucial to shift more attention towards understanding the underlying mechanisms and principles. In the meantime, a difficult point in mechanical regulation is how to quantify the mechanical forces in biological effects (Fig. 4g–i). With the rise of biosensor technology, this problem is becoming increasingly soluble. Förster resonance energy transfer-based tension sensors have been extensively employed to measure forces and assess ECM stiffness at a small scale. This paves the way for more effective and targeted interventions in tissue regeneration and wound healing [209,210,211]. A low frequency Love wave sensor is used to detect 0.89–3.3 cP viscous change [212]. Machine learning-based image restoration and traction force microscopy are combined to understand relationship between cytoskeletal organization and ECM [213]. Biosensors play a crucial role in translating mechanical signals into computational models, offering a broader understanding of parameter quantification. Moreover, as we delve into the mechano-transduction pathway from FA to the nuclear lamina, it becomes evident that key players include, but are not limited to, integrins, the cytoskeleton, the LINC complex, and various associated proteins. In this intricate ‘relay race’, these components form a complex regulatory network that deserves further investigations. Scarless wound healing necessitates several essential factors, including high-quality re-epithelialization, reduced fibrosis, neovascularization, the restoration of innervation, and the regeneration of skin appendages. By combining computable mechanical parameters and the modification of key players in mechano-transduction, precise application of biomaterials can be achieved in scarless wound healing and skin appendage regeneration. This integrated approach holds promise for more effective interventions in tissue repair and regeneration.

Availability of data and materials

Not applicable.

Abbreviations

2D:

Two-dimensional

3D:

Three-dimensional

ABPs:

Actin-binding proteins;

α-SMA:

α-Smooth muscle actin

BMP:

Bone morphogenetic protein

CTGF:

Connective tissue growth factor

CCM3:

Cerebral cavernous malformations 3

CMCS:

Carborxymethy chitosan

DRG:

Dorsal root ganglion

ECM:

Extracellular matrix

ECs:

Endothelial cells

ELPs:

Elastin-like polypeptides

EMT:

Epithelial-mesenchymal transition

FAs:

Focal adhesions

FAK:

Focal adhesion kinase

GelMA:

Gelatin methacryloyl

HDs:

Hemidesmosomes

hES-MP:

Human embryonic stem cell-derived mesenchymal progenitor cells

HFSCs:

Hair follicle stem cells

HFs:

Hair follicles

IFs:

Intermediate filaments

IL:

Interleukin

LATS1/2:

Large tumor suppressor 1/2

LOX:

Lysyl oxidase

LINC:

Linker between nuclear-skeleton and cytoskeleton

MAPK:

Mitogen-activated protein kinase

NETs:

Neutrophil extracellular traps

NGCs:

Nerve guidance conduits

PA:

Polyacrylamide

PAI-1:

Plasminogen activator inhibitor 1

PBA-PAMAM:

Phenylboronic acid-bearing poly(amidoamine) dendrimer

PDMS:

Polydimethylsiloxane

PDGF-B:

Platelet-derived growth factor B

PEG-DE:

Poly(ethylene glycol) diglycidyl ether

PHBV:

Poly(3-hydroxybutyrate-co-3-hydroxyvalerate)

PI3K:

Phosphatidylinositol-3-kinase

PLLA:

Poly(L-lactic)

ROCK/MLC:

RhoA-Rho kinase/myosin light chain

RREB1:

RAS-responsive element binding protein 1

SGCs:

Sweat gland cells

SHH:

Sonic hedgehog

SMP:

Shape memory polymer

SRM:

Silicone rubber membrane

SRM-B:

Silicone rubber membrane bilayer

ST2:

Suppression of tumorigenicity 2 receptor

SwGs:

Sweat glands

TGF:

Transforming growth factor

Ti:

Titanium

VEGF:

Vascular endothelial growth factor

WISP1:

WNT-inducible signaling pathway protein 1

YAP/TAZ:

Yes-associated protein and transcriptional co-activator with PDZ-binding motif

ZEB1:

Zinc finger E-box binding homeobox 1

ZEB2:

Zinc finger E-box binding homeobox 2

References

  1. Hong ZX, Zhu ST, Li H, Luo JZ, Yang Y, An Y, et al. Bioengineered skin organoids: from development to applications. Mil Med Res. 2023;10(1):40.

    PubMed  PubMed Central  Google Scholar 

  2. Agha R, Ogawa R, Pietramaggiori G, Orgill DP. A review of the role of mechanical forces in cutaneous wound healing. J Surg Res. 2011;171(2):700–8.

    Article  PubMed  Google Scholar 

  3. Barnes LA, Marshall CD, Leavitt T, Hu MS, Moore AL, Gonzalez JG, et al. Mechanical forces in cutaneous wound healing: emerging therapies to minimize scar formation. Adv Wound Care (New Rochelle). 2018;7(2):47–56.

    Article  PubMed  Google Scholar 

  4. Lim CG, Jang J, Kim C. Cellular machinery for sensing mechanical force. BMB Rep. 2018;51(12):623–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Xie Y, Liao X, Zhang J, Yang F, Fan Z. Novel chitosan hydrogels reinforced by silver nanoparticles with ultrahigh mechanical and high antibacterial properties for accelerating wound healing. Int J Biol Macromol. 2018;119:402–12.

    Article  CAS  PubMed  Google Scholar 

  6. Wang X, Lin M, Kang Y. Engineering porous β-tricalcium phosphate (β-TCP) scaffolds with multiple channels to promote cell migration, proliferation, and angiogenesis. ACS Appl Mater Interfaces. 2019;11(9):9223–32.

    Article  CAS  PubMed  Google Scholar 

  7. Kim HS, Chen J, Wu LP, Wu J, Xiang H, Leong KW, et al. Prevention of excessive scar formation using nanofibrous meshes made of biodegradable elastomer poly(3-hydroxybutyrate-co-3-hydroxyvalerate). J Tissue Eng. 2020;11:2041731420949332.

    Article  PubMed  PubMed Central  Google Scholar 

  8. Xia B, Gao J, Li S, Huang L, Zhu L, Ma T, et al. Mechanical stimulation of Schwann cells promote peripheral nerve regeneration via extracellular vesicle-mediated transfer of microRNA 23b–3p. Theranostics. 2020;10(20):8974–95.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Su J, Morgani SM, David CJ, Wang Q, Er EE, Huang YH, et al. TGF-β orchestrates fibrogenic and developmental EMTs via the RAS effector RREB1. Nature. 2020;577(7791):566–71.

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  10. Akaishi S, Ogawa R, Hyakusoku H. Keloid and hypertrophic scar: neurogenic inflammation hypotheses. Med Hypotheses. 2008;71(1):32–8.

    Article  CAS  PubMed  Google Scholar 

  11. Krzyszczyk P, Schloss R, Palmer A, Berthiaume F. The role of macrophages in acute and chronic wound healing and interventions to promote pro-wound healing phenotypes. Front Physiol. 2018;9:419.

    Article  PubMed  PubMed Central  Google Scholar 

  12. Ogawa R, Akaishi S. Endothelial dysfunction may play a key role in keloid and hypertrophic scar pathogenesis—keloids and hypertrophic scars may be vascular disorders. Med Hypotheses. 2016;96:51–60.

    Article  CAS  PubMed  Google Scholar 

  13. Kimura S, Tsuji T. Mechanical and immunological regulation in wound healing and skin reconstruction. Int J Mol Sci. 2021;22(11):5474.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Lichtman MK, Otero-Vinas M, Falanga V. Transforming growth factor beta (TGF-β) isoforms in wound healing and fibrosis. Wound Repair Regen. 2016;24(2):215–22.

    Article  PubMed  Google Scholar 

  15. Ji Y, Dou YN, Zhao QW, Zhang JZ, Yang Y, Wang T, et al. Paeoniflorin suppresses TGF-β mediated epithelial-mesenchymal transition in pulmonary fibrosis through a Smad-dependent pathway. Acta Pharmacol Sin. 2016;37(6):794–804.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Huang C, Liu L, You Z, Zhao Y, Dong J, Du Y, et al. Endothelial dysfunction and mechanobiology in pathological cutaneous scarring: lessons learned from soft tissue fibrosis. Br J Dermatol. 2017;177(5):1248–55.

    Article  CAS  PubMed  Google Scholar 

  17. Ogawa R, Mitsuhashi K, Hyakusoku H, Miyashita T. Postoperative electron-beam irradiation therapy for keloids and hypertrophic scars: retrospective study of 147 cases followed for more than 18 months. Plast Reconstr Surg. 2003;111(2):547–53 (discussion 554-5).

    PubMed  Google Scholar 

  18. Laiacona D, Cohen JM, Coulon K, Lipsky ZW, Maiorana C, Boltyanskiy R, et al. Non-invasive in vivo quantification of human skin tension lines. Acta Biomater. 2019;88:141–8.

    Article  CAS  PubMed  Google Scholar 

  19. Bouffard NA, Cutroneo KR, Badger GJ, White SL, Buttolph TR, Ehrlich HP, et al. Tissue stretch decreases soluble TGF-beta1 and type-1 procollagen in mouse subcutaneous connective tissue: evidence from ex vivo and in vivo models. J Cell Physiol. 2008;214(2):389–95.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Aarabi S, Bhatt KA, Shi Y, Paterno J, Chang EI, Loh SA, et al. Mechanical load initiates hypertrophic scar formation through decreased cellular apoptosis. FASEB J. 2007;21(12):3250–61.

    Article  CAS  PubMed  Google Scholar 

  21. Talbott HE, Mascharak S, Griffin M, Wan DC, Longaker MT. Wound healing, fibroblast heterogeneity, and fibrosis. Cell Stem Cell. 2022;29(8):1161–80.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Przybyla L, Muncie JM, Weaver VM. Mechanical control of epithelial-to-mesenchymal transitions in development and cancer. Annu Rev Cell Dev Biol. 2016;32:527–54.

    Article  CAS  PubMed  Google Scholar 

  23. Choi B, Park KS, Kim JH, Ko KW, Kim JS, Han DK, et al. Stiffness of hydrogels regulates cellular reprogramming efficiency through mesenchymal-to-epithelial transition and stemness markers. Macromol Biosci. 2016;16(2):199–206.

    Article  CAS  PubMed  Google Scholar 

  24. Stone RC, Pastar I, Ojeh N, Chen V, Liu S, Garzon KI, et al. Epithelial-mesenchymal transition in tissue repair and fibrosis. Cell Tissue Res. 2016;365(3):495–506.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Kotsiou OS, Gourgoulianis KI, Zarogiannis SG. IL-33/ST2 axis in organ fibrosis. Front Immunol. 2018;9:2432.

    Article  PubMed  PubMed Central  Google Scholar 

  26. Sun Z, Ji N, Ma Q, Zhu R, Chen Z, Wang Z, et al. Epithelial-mesenchymal transition in asthma airway remodeling is regulated by the IL-33/CD146 axis. Front Immunol. 2020;11:1598.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Yang X, Bao M, Fang Y, Yu X, Ji J, Ding X. STAT3/HIF-1α signaling activation mediates peritoneal fibrosis induced by high glucose. J Transl Med. 2021;19(1):283.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Shrader CD, Ressetar HG, Luo J, Cilento EV, Reilly FD. Acute stretch promotes endothelial cell proliferation in wounded healing mouse skin. Arch Dermatol Res. 2008;300(9):495–504.

    Article  PubMed  Google Scholar 

  29. Korntner S, Lehner C, Gehwolf R, Wagner A, Grutz M, Kunkel N, et al. Limiting angiogenesis to modulate scar formation. Adv Drug Deliv Rev. 2019;146:170–89.

    Article  CAS  PubMed  Google Scholar 

  30. Zhang L, He J, Wang J, Liu J, Chen Z, Deng B, et al. Knockout RAGE alleviates cardiac fibrosis through repressing endothelial-to-mesenchymal transition (EndMT) mediated by autophagy. Cell Death Dis. 2021;12(5):470.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Testai L, Brancaleone V, Flori L, Montanaro R, Calderone V. Modulation of EndMT by hydrogen sulfide in the prevention of cardiovascular fibrosis. Antioxidants (Basel). 2021;10(6):910.

    Article  CAS  PubMed  Google Scholar 

  32. Eming SA, Wynn TA, Martin P. Inflammation and metabolism in tissue repair and regeneration. Science. 2017;356(6342):1026–30.

    Article  ADS  CAS  PubMed  Google Scholar 

  33. Kaur T, Dumoga S, Koul V, Singh N. Modulating neutrophil extracellular traps for wound healing. Biomater Sci. 2020;8(11):3212–23.

    Article  CAS  PubMed  Google Scholar 

  34. Hu Q, Zhang S, Yang Y, Yao JQ, Tang WF, Lyon CJ, et al. Extracellular vesicles in the pathogenesis and treatment of acute lung injury. Mil Med Res. 2022;9(1):61.

    PubMed  PubMed Central  Google Scholar 

  35. Chrysanthopoulou A, Mitroulis I, Apostolidou E, Arelaki S, Mikroulis D, Konstantinidis T, et al. Neutrophil extracellular traps promote differentiation and function of fibroblasts. J Pathol. 2014;233(3):294–307.

    Article  CAS  PubMed  Google Scholar 

  36. Wilgus TA, Roy S, McDaniel JC. Neutrophils and wound repair: positive actions and negative reactions. Adv Wound Care (New Rochelle). 2013;2(7):379–88.

    Article  PubMed  PubMed Central  Google Scholar 

  37. Li JL, Lim CH, Tay FW, Goh CC, Devi S, Malleret B, et al. Neutrophils self-regulate immune complex-mediated cutaneous inflammation through CXCL2. J Invest Dermatol. 2016;136(2):416–24.

    Article  CAS  PubMed  Google Scholar 

  38. Wynn TA, Barron L. Macrophages: master regulators of inflammation and fibrosis. Semin Liver Dis. 2010;30(3):245–57.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Kim J, Han S, Lei A, Miyano M, Bloom J, Srivastava V, et al. Characterizing cellular mechanical phenotypes with mechano-node-pore sensing. Microsyst Nanoeng. 2018;4:17091.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Duperret EK, Ridky TW. Focal adhesion complex proteins in epidermis and squamous cell carcinoma. Cell Cycle. 2013;12(20):3272–85.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Seetharaman S, Etienne-Manneville S. Integrin diversity brings specificity in mechanotransduction. Biol Cell. 2018;110(3):49–64.

    Article  CAS  PubMed  Google Scholar 

  42. Slack RJ, MacDonald SJF, Roper JA, Jenkins RG, Hatley RJD. Emerging therapeutic opportunities for integrin inhibitors. Nat Rev Drug Discov. 2022;21(1):60–78.

    Article  CAS  PubMed  Google Scholar 

  43. Wang L, Luo JY, Li B, Tian XY, Chen LJ, Huang Y, et al. Integrin-YAP/TAZ-JNK cascade mediates atheroprotective effect of unidirectional shear flow. Nature. 2016;540(7634):579–82.

    Article  ADS  CAS  PubMed  Google Scholar 

  44. Georges-Labouesse E, Messaddeq N, Yehia G, Cadalbert L, Dierich A, Le Meur M. Absence of integrin alpha 6 leads to epidermolysis bullosa and neonatal death in mice. Nat Genet. 1996;13(3):370–3.

    Article  CAS  PubMed  Google Scholar 

  45. Dowling J, Yu QC, Fuchs E. Beta4 integrin is required for hemidesmosome formation, cell adhesion and cell survival. J Cell Biol. 1996;134(2):559–72.

    Article  CAS  PubMed  Google Scholar 

  46. Wang W, Zuidema A, Te Molder L, Nahidiazar L, Hoekman L, Schmidt T, et al. Hemidesmosomes modulate force generation via focal adhesions. J Cell Biol. 2020;219(2):e201904137.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Natale CF, Lafaurie-Janvore J, Ventre M, Babataheri A, Barakat AI. Focal adhesion clustering drives endothelial cell morphology on patterned surfaces. J R Soc Interface. 2019;16(158):20190263.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Rustad KC, Wong VW, Gurtner GC. The role of focal adhesion complexes in fibroblast mechanotransduction during scar formation. Differentiation. 2013;86(3):87–91.

    Article  CAS  PubMed  Google Scholar 

  49. Chen K, Kwon SH, Henn D, Kuehlmann BA, Tevlin R, Bonham CA, et al. Disrupting biological sensors of force promotes tissue regeneration in large organisms. Nat Commun. 2021;12(1):5256.

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  50. Tokuraku K, Kuragano M, Uyeda TQP. Long-range and directional allostery of actin filaments plays important roles in various cellular activities. Int J Mol Sci. 2020;21(9):3209.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Xie C, Jiang Y, Zhu Z, Huang S, Li W, Ou G. Actin filament debranching regulates cell polarity during cell migration and asymmetric cell division. Proc Natl Acad Sci U S A. 2021;118(37):e2100805118.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Hayakawa K, Tatsumi H, Sokabe M. Actin filaments function as a tension sensor by tension-dependent binding of cofilin to the filament. J Cell Biol. 2011;195(5):721–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Harris AR, Jreij P, Belardi B, Joffe AM, Bausch AR, Fletcher DA. Biased localization of actin binding proteins by actin filament conformation. Nat Commun. 2020;11(1):5973.

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  54. Lappalainen P, Kotila T, Jégou A, Romet-Lemonne G. Biochemical and mechanical regulation of actin dynamics. Nat Rev Mol Cell Biol. 2022;23(12):836–52.

    Article  CAS  PubMed  Google Scholar 

  55. Goodson HV, Jonasson EM. Microtubules and microtubule-associated proteins. Cold Spring Harb Perspect Biol. 2018;10(6):a022608.

    Article  PubMed  PubMed Central  Google Scholar 

  56. Bance B, Seetharaman S, Leduc C, Boëda B, Etienne-Manneville S. Microtubule acetylation but not detyrosination promotes focal adhesion dynamics and astrocyte migration. J Cell Sci. 2019;132(7):jcs225805.

    Article  CAS  PubMed  Google Scholar 

  57. Seetharaman S, Vianay B, Roca V, Farrugia AJ, De Pascalis C, Boëda B, et al. Microtubules tune mechanosensitive cell responses. Nat Mater. 2022;21(3):366–77.

    Article  ADS  CAS  PubMed  Google Scholar 

  58. Rafiq NBM, Nishimura Y, Plotnikov SV, Thiagarajan V, Zhang Z, Shi S, et al. A mechano-signalling network linking microtubules, myosin IIA filaments and integrin-based adhesions. Nat Mater. 2019;18(6):638–49.

    Article  ADS  CAS  PubMed  Google Scholar 

  59. Ndiaye AB, Koenderink GH, Shemesh M. Intermediate filaments in cellular mechanoresponsiveness: mediating cytoskeletal crosstalk from membrane to nucleus and back. Front Cell Dev Biol. 2022;10:882037.

    Article  PubMed  PubMed Central  Google Scholar 

  60. Nagle I, Delort F, Hénon S, Wilhelm C, Batonnet-Pichon S, Reffay M. The importance of intermediate filaments in the shape maintenance of myoblast model tissues. Elife. 2022;11:e76409.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Laly AC, Sliogeryte K, Pundel OJ, Ross R, Keeling MC, Avisetti D, et al. The keratin network of intermediate filaments regulates keratinocyte rigidity sensing and nuclear mechanotransduction. Sci Adv. 2021;7(5):eabd6187.

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  62. Daday C, Kolšek K, Gräter F. The mechano-sensing role of the unique SH3 insertion in plakin domains revealed by molecular dynamics simulations. Sci Rep. 2017;7(1):11669.

    Article  ADS  PubMed  PubMed Central  Google Scholar 

  63. Spurny R, Gregor M, Castañón MJ, Wiche G. Plectin deficiency affects precursor formation and dynamics of vimentin networks. Exp Cell Res. 2008;314(19):3570–80.

    Article  CAS  PubMed  Google Scholar 

  64. Chambliss AB, Khatau SB, Erdenberger N, Robinson DK, Hodzic D, Longmore GD, et al. The LINC-anchored actin cap connects the extracellular milieu to the nucleus for ultrafast mechanotransduction. Sci Rep. 2013;3:1087.

    Article  ADS  PubMed  PubMed Central  Google Scholar 

  65. Maurer M, Lammerding J. The driving force: nuclear mechanotransduction in cellular function, fate, and disease. Annu Rev Biomed Eng. 2019;21:443–68.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Alam SG, Zhang Q, Prasad N, Li Y, Chamala S, Kuchibhotla R, et al. The mammalian LINC complex regulates genome transcriptional responses to substrate rigidity. Sci Rep. 2016;6:38063.

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  67. Déjardin T, Carollo PS, Sipieter F, Davidson PM, Seiler C, Cuvelier D, et al. Nesprins are mechanotransducers that discriminate epithelial-mesenchymal transition programs. J Cell Biol. 2020;219(10):e201908036.

    Article  PubMed  PubMed Central  Google Scholar 

  68. Arsenovic PT, Ramachandran I, Bathula K, Zhu R, Narang JD, Noll NA, et al. Nesprin-2G, a component of the nuclear LINC complex, is subject to myosin-dependent tension. Biophys J. 2016;110(1):34–43.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Chai RJ, Werner H, Li PY, Lee YL, Nyein KT, Solovei I, et al. Disrupting the LINC complex by AAV mediated gene transduction prevents progression of Lamin induced cardiomyopathy. Nat Commun. 2021;12(1):4722.

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  70. Carley E, Stewart RM, Zieman A, Jalilian I, King DE, Zubek A, et al. The LINC complex transmits integrin-dependent tension to the nuclear lamina and represses epidermal differentiation. Elife. 2021;10:e58541.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Sawaya AP, Uchiyama A, Hope E, Bajpai D, Worrell S, Cross M, et al. Keratin 75 is a component of the LINC complex and has an essential role in mediating the SOX2 rapid healing response during wound repair. J Invest Dermatol. 2023;143(3):494–8.

    Article  CAS  PubMed  Google Scholar 

  72. Moya IM, Halder G. Hippo-YAP/TAZ signalling in organ regeneration and regenerative medicine. Nat Rev Mol Cell Biol. 2019;20(4):211–26.

    Article  CAS  PubMed  Google Scholar 

  73. Dey A, Varelas X, Guan KL. Targeting the Hippo pathway in cancer, fibrosis, wound healing and regenerative medicine. Nat Rev Drug Discov. 2020;19(7):480–94.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Mascharak S, desJardins-Park HE, Davitt MF, Griffin M, Borrelli MR, Moore AL, et al. Preventing Engrailed-1 activation in fibroblasts yields wound regeneration without scarring. Science. 2021;372(6540):eaba2374.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Piersma B, de Rond S, Werker PMN, Boo S, Hinz B, Van Beuge MM, et al. YAP1 is a driver of myofibroblast differentiation in normal and diseased fibroblasts. Am J Pathol. 2015;185(12):3326–37.

    Article  CAS  PubMed  Google Scholar 

  76. Liu F, Lagares D, Choi KM, Stopfer L, Marinković A, Vrbanac V, et al. Mechanosignaling through YAP and TAZ drives fibroblast activation and fibrosis. Am J Physiol Lung Cell Mol Physiol. 2015;308(4):L344–57.

    Article  CAS  PubMed  Google Scholar 

  77. Szeto SG, Narimatsu M, Lu M, He X, Sidiqi AM, Tolosa MF, et al. YAP/TAZ are mechanoregulators of TGF-β-smad signaling and renal fibrogenesis. J Am Soc Nephrol. 2016;27(10):3117–28.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Dupont S, Morsut L, Aragona M, Enzo E, Giulitti S, Cordenonsi M, et al. Role of YAP/TAZ in mechanotransduction. Nature. 2011;474(7350):179–83.

    Article  CAS  PubMed  Google Scholar 

  79. Wang S, Englund E, Kjellman P, Li Z, Ahnlide JK, Rodriguez-Cupello C, et al. CCM3 is a gatekeeper in focal adhesions regulating mechanotransduction and YAP/TAZ signalling. Nat Cell Biol. 2021;23(7):758–70.

    Article  CAS  PubMed  Google Scholar 

  80. Wen SM, Wen WC, Chao PHG. Zyxin and actin structure confer anisotropic YAP mechanotransduction. Acta Biomater. 2022;152:313–20.

    Article  CAS  PubMed  Google Scholar 

  81. Corley SM, Mendoza-Reinoso V, Giles N, Singer ES, Common JE, Wilkins MR, et al. Plau and Tgfbr3 are YAP-regulated genes that promote keratinocyte proliferation. Cell Death Dis. 2018;9(11):1106.

    Article  PubMed  PubMed Central  Google Scholar 

  82. Meli VS, Atcha H, Veerasubramanian PK, Nagalla RR, Luu TU, Chen EY, et al. YAP-mediated mechanotransduction tunes the macrophage inflammatory response. Sci Adv. 2020;6(49):8471.

    Article  ADS  Google Scholar 

  83. Viswanathan P, Ondeck MG, Chirasatitsin S, Ngamkham K, Reilly GC, Engler AJ, et al. 3D surface topology guides stem cell adhesion and differentiation. Biomaterials. 2015;52:140–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  84. González-García C, Sousa SR, Moratal D, Rico P, Salmerón-Sánchez M. Effect of nanoscale topography on fibronectin adsorption, focal adhesion size and matrix organisation. Colloids Surf B Biointerfaces. 2010;77(2):181–90.

    Article  PubMed  Google Scholar 

  85. Seo CH, Furukawa K, Montagne K, Jeong H, Ushida T. The effect of substrate microtopography on focal adhesion maturation and actin organization via the RhoA/ROCK pathway. Biomaterials. 2011;32(36):9568–75.

    Article  CAS  PubMed  Google Scholar 

  86. Lim JY, Dreiss AD, Zhou Z, Hansen JC, Siedlecki CA, Hengstebeck RW, et al. The regulation of integrin-mediated osteoblast focal adhesion and focal adhesion kinase expression by nanoscale topography. Biomaterials. 2007;28(10):1787–97.

    Article  CAS  PubMed  Google Scholar 

  87. Ha SW, Jang HL, Nam KT, Beck GR Jr. Nano-hydroxyapatite modulates osteoblast lineage commitment by stimulation of DNA methylation and regulation of gene expression. Biomaterials. 2015;65:32–42.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  88. Kingham E, White K, Gadegaard N, Dalby MJ, Oreffo RO. Nanotopographical cues augment mesenchymal differentiation of human embryonic stem cells. Small. 2013;9(12):2140–51.

    Article  CAS  PubMed  Google Scholar 

  89. Morez C, Noseda M, Paiva MA, Belian E, Schneider MD, Stevens MM. Enhanced efficiency of genetic programming toward cardiomyocyte creation through topographical cues. Biomaterials. 2015;70:94–104.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  90. Qiu Y, Brown AC, Myers DR, Sakurai Y, Mannino RG, Tran R, et al. Platelet mechanosensing of substrate stiffness during clot formation mediates adhesion, spreading, and activation. Proc Natl Acad Sci U S A. 2014;111(40):14430–5.

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  91. Kim MC, Silberberg YR, Abeyaratne R, Kamm RD, Asada HH. Computational modeling of three-dimensional ECM-rigidity sensing to guide directed cell migration. Proc Natl Acad Sci U S A. 2018;115(3):E390–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  92. Soubhagya AS, Moorthi A, Prabaharan M. Preparation and characterization of chitosan/pectin/ZnO porous films for wound healing. Int J Biol Macromol. 2020;157:135–45.

    Article  CAS  PubMed  Google Scholar 

  93. Chung HH, Casillo SM, Perry SJ, Gaborski TR. Porous substrates promote endothelial migration at the expense of fibronectin fibrillogenesis. ACS Biomater Sci Eng. 2018;4(1):222–30.

    Article  CAS  PubMed  Google Scholar 

  94. Bouta EM, McCarthy CW, Keim A, Wang HB, Gilbert RJ, Goldman J. Biomaterial guides for lymphatic endothelial cell alignment and migration. Acta Biomater. 2011;7(3):1104–13.

    Article  CAS  PubMed  Google Scholar 

  95. Huang C, Ouyang Y, Niu H, He N, Ke Q, Jin X, et al. Nerve guidance conduits from aligned nanofibers: improvement of nerve regeneration through longitudinal nanogrooves on a fiber surface. ACS Appl Mater Interfaces. 2015;7(13):7189–96.

    Article  CAS  PubMed  Google Scholar 

  96. Adlerz KM, Aranda-Espinoza H, Hayenga HN. Substrate elasticity regulates the behavior of human monocyte-derived macrophages. Eur Biophys J. 2016;45(4):301–9.

    Article  CAS  PubMed  Google Scholar 

  97. Friedemann M, Kalbitzer L, Franz S, Moeller S, Schnabelrauch M, Simon JC, et al. Instructing human macrophage polarization by stiffness and glycosaminoglycan functionalization in 3D collagen networks. Adv Healthc Mater. 2017. https://0-doi-org.brum.beds.ac.uk/10.1002/adhm.201600967.

    Article  PubMed  Google Scholar 

  98. Liu Y, Li J, Yao B, Wang Y, Wang R, Yang S, et al. The stiffness of hydrogel-based bioink impacts mesenchymal stem cells differentiation toward sweat glands in 3D-bioprinted matrix. Mater Sci Eng C Mater Biol Appl. 2021;118:111387.

    Article  CAS  PubMed  Google Scholar 

  99. Li X, Chen S, Li J, Wang X, Zhang J, Kawazoe N, et al. 3D culture of chondrocytes in gelatin hydrogels with different stiffness. Polymers. 2016;8(8):269.

    Article  PubMed  PubMed Central  Google Scholar 

  100. Hassanzadeh P, Kazemzadeh-Narbat M, Rosenzweig R, Zhang X, Khademhosseini A, Annabi N, et al. Ultrastrong and flexible hybrid hydrogels based on solution self-assembly of chitin nanofibers in gelatin methacryloyl (GelMA). J Mater Chem B. 2016;4(15):2539–43.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  101. Li G, Zheng T, Wu L, Han Q, Lei Y, Xue L, et al. Bionic microenvironment-inspired synergistic effect of anisotropic micro-nanocomposite topology and biology cues on peripheral nerve regeneration. Sci Adv. 2021;7(28):i5812.

    Article  ADS  Google Scholar 

  102. Kim SH, Turnbull J, Guimond S. Extracellular matrix and cell signalling: the dynamic cooperation of integrin, proteoglycan and growth factor receptor. J Endocrinol. 2011;209(2):139–51.

    Article  CAS  PubMed  Google Scholar 

  103. Natale CF, Ventre M, Netti PA. Tuning the material-cytoskeleton crosstalk via nanoconfinement of focal adhesions. Biomaterials. 2014;35(9):2743–51.

    Article  CAS  PubMed  Google Scholar 

  104. Peyton SR, Kim PD, Ghajar CM, Seliktar D, Putnam AJ. The effects of matrix stiffness and RhoA on the phenotypic plasticity of smooth muscle cells in a 3-D biosynthetic hydrogel system. Biomaterials. 2008;29(17):2597–607.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  105. Bacakova L, Filova E, Parizek M, Ruml T, Svorcik V. Modulation of cell adhesion, proliferation and differentiation on materials designed for body implants. Biotechnol Adv. 2011;29(6):739–67.

    Article  CAS  PubMed  Google Scholar 

  106. Shi H, Wu X, Sun S, Wang C, Vangelatos Z, Ash-Shakoor A, et al. Profiling the responsiveness of focal adhesions of human cardiomyocytes to extracellular dynamic nano-topography. Bioact Mater. 2022;10:367–77.

    CAS  PubMed  Google Scholar 

  107. Sniadecki NJ, Anguelouch A, Yang MT, Lamb CM, Liu Z, Kirschner SB, et al. Magnetic microposts as an approach to apply forces to living cells. Proc Natl Acad Sci U S A. 2007;104(37):14553–8.

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  108. Zhou C, Duan M, Guo D, Du X, Zhang D, Xie J. Microenvironmental stiffness mediates cytoskeleton re-organization in chondrocytes through laminin-FAK mechanotransduction. Int J Oral Sci. 2022;14(1):15.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  109. Adamczyk O, Baster Z, Szczypior M, Rajfur Z. Substrate stiffness mediates formation of novel cytoskeletal structures in fibroblasts during cell-microspheres interaction. Int J Mol Sci. 2021;22(2):960.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  110. Gu Y, Ji Y, Zhao Y, Liu Y, Ding F, Gu X, et al. The influence of substrate stiffness on the behavior and functions of Schwann cells in culture. Biomaterials. 2012;33(28):6672–81.

    Article  CAS  PubMed  Google Scholar 

  111. Wang Q, Xie J, Zhou C, Lai W. Substrate stiffness regulates the differentiation profile and functions of osteoclasts via cytoskeletal arrangement. Cell Prolif. 2022;55(1):e13172.

    Article  CAS  PubMed  Google Scholar 

  112. Lv L, Tang Y, Zhang P, Liu Y, Bai X, Zhou Y. Biomaterial cues regulate epigenetic state and cell functions-a systematic review. Tissue Eng Part B Rev. 2018;24(2):112–32.

    Article  PubMed  Google Scholar 

  113. Huang X, Qu R, Peng Y, Yang Y, Fan T, Sun B, et al. Mechanical sensing element PDLIM5 promotes osteogenesis of human fibroblasts by affecting the activity of microfilaments. Biomolecules. 2021;11(5):759.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  114. Isomursu A, Park KY, Hou J, Cheng B, Mathieu M, Shamsan GA, et al. Directed cell migration towards softer environments. Nat Mater. 2022;21(9):1081–90.

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  115. Zhao D, Xue C, Li Q, Liu M, Ma W, Zhou T, et al. Substrate stiffness regulated migration and angiogenesis potential of A549 cells and HUVECs. J Cell Physiol. 2018;233(4):3407–17.

    Article  CAS  PubMed  Google Scholar 

  116. Wang A, Cao S, Stowe JC, Valdez-Jasso D. Substrate stiffness and stretch regulate profibrotic mechanosignaling in pulmonary arterial adventitial fibroblasts. Cells. 2021;10(5):1000.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  117. Hadden WJ, Young JL, Holle AW, Mcfetridge ML, Kim DY, Wijesinghe P, et al. Stem cell migration and mechanotransduction on linear stiffness gradient hydrogels. Proc Natl Acad Sci U S A. 2017;114(22):5647–52.

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  118. Pham JT, Xue L, Del Campo A, Salierno M. Guiding cell migration with microscale stiffness patterns and undulated surfaces. Acta Biomater. 2016;38:106–15.

    Article  CAS  PubMed  Google Scholar 

  119. Hiraki HL, Matera DL, Wang WY, Prabhu ES, Zhang Z, Midekssa F, et al. Fiber density and matrix stiffness modulate distinct cell migration modes in a 3D stroma mimetic composite hydrogel. Acta Biomater. 2023;163:378–91.

    Article  CAS  PubMed  Google Scholar 

  120. Yildirimer L, Seifalian AM. Three-dimensional biomaterial degradation—material choice, design and extrinsic factor considerations. Biotechnol Adv. 2014;32(5):984–99.

    Article  CAS  PubMed  Google Scholar 

  121. Zhao J, Luo G, Wu J, Xia H. Preparation of microporous silicone rubber membrane with tunable pore size via solvent evaporation-induced phase separation. ACS Appl Mater Interfaces. 2013;5(6):2040–6.

    Article  CAS  PubMed  Google Scholar 

  122. Xu R, Luo G, Xia H, He W, Zhao J, Liu B, et al. Novel bilayer wound dressing composed of silicone rubber with particular micropores enhanced wound re-epithelialization and contraction. Biomaterials. 2015;40:1–11.

    Article  PubMed  Google Scholar 

  123. Chu Y, Yu D, Wang P, Xu J, Li D, Ding M. Nanotechnology promotes the full-thickness diabetic wound healing effect of recombinant human epidermal growth factor in diabetic rats. Wound Repair Regen. 2010;18(5):499–505.

    Article  PubMed  Google Scholar 

  124. Khorasani MT, Joorabloo A, Adeli H, Milan PB, Amoupour M. Enhanced antimicrobial and full-thickness wound healing efficiency of hydrogels loaded with heparinized ZnO nanoparticles: in vitro and in vivo evaluation. Int J Biol Macromol. 2021;166:200–12.

    Article  CAS  PubMed  Google Scholar 

  125. Samadian H, Salehi M, Farzamfar S, Vaez A, Ehterami A, Sahrapeyma H, et al. In vitro and in vivo evaluation of electrospun cellulose acetate/gelatin/hydroxyapatite nanocomposite mats for wound dressing applications. Artif Cells Nanomed Biotechnol. 2018;46(sup1):964–74.

    Article  CAS  PubMed  Google Scholar 

  126. Lee JH, Jeon WY, Kim HH, Lee EJ, Kim HW. Electrical stimulation by enzymatic biofuel cell to promote proliferation, migration and differentiation of muscle precursor cells. Biomaterials. 2015;53:358–69.

    Article  CAS  PubMed  Google Scholar 

  127. Kim TH, Jeon WY, Ji Y, Park EJ, Yoon DS, Lee NH, et al. Electricity auto-generating skin patch promotes wound healing process by activation of mechanosensitive ion channels. Biomaterials. 2021;275:120948.

    Article  CAS  PubMed  Google Scholar 

  128. Li X, Sun Q, Li Q, Kawazoe N, Chen G. Functional hydrogels with tunable structures and properties for tissue engineering applications. Front Chem. 2018;6:499.

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  129. Zhao X, Sun X, Yildirimer L, Lang Q, Lin ZYW, Zheng R, et al. Cell infiltrative hydrogel fibrous scaffolds for accelerated wound healing. Acta Biomater. 2017;49:66–77.

    Article  CAS  PubMed  Google Scholar 

  130. Zhou W, Duan Z, Zhao J, Fu R, Zhu C, Fan D. Glucose and MMP-9 dual-responsive hydrogel with temperature sensitive self-adaptive shape and controlled drug release accelerates diabetic wound healing. Bioact Mater. 2022;17:1–17.

    PubMed  PubMed Central  Google Scholar 

  131. Schneider LA, Korber A, Grabbe S, Dissemond J. Influence of pH on wound-healing: a new perspective for wound-therapy? Arch Dermatol Res. 2007;298(9):413–20.

    Article  PubMed  Google Scholar 

  132. Hollingshead S, Liu JC. pH-sensitive mechanical properties of elastin-based hydrogels. Macromol Biosci. 2020;20(4):e1900369.

    Article  PubMed  Google Scholar 

  133. Min Y, Han R, Li G, Wang X, Chen S, Xie M, et al. The pH-sensitive optical fiber integrated CMCS-PA@Fe hydrogels for photothermal therapy and real-time monitoring of infected wounds. Adv Funct Mater. 2023;33(16):202212803.

    Article  Google Scholar 

  134. Sato F, Anzai JI. Sugar-sensitive dendrimer films as a sacrificial layer for the preparation of freestanding multilayer films. Mater Sci Eng C Mater Biol Appl. 2017;72:118–22.

    Article  CAS  PubMed  Google Scholar 

  135. Zhu F, Yan N, Lu X, Xu J, Gu H, Liang J, et al. Cell reprogramming-inspired dynamically responsive hydrogel boosts the induction of pluripotency via phase-separated biomolecular condensates. Adv Mater. 2023:e2211609.

  136. Vining KH, Mooney DJ. Mechanical forces direct stem cell behaviour in development and regeneration. Nat Rev Mol Cell Biol. 2017;18(12):728–42.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  137. Gonzales KAU, Fuchs E. Skin and its regenerative powers: an alliance between stem cells and their niche. Dev Cell. 2017;43(4):387–401.

    Article  CAS  PubMed  Google Scholar 

  138. Asare A, Levorse J, Fuchs E. Coupling organelle inheritance with mitosis to balance growth and differentiation. Science. 2017;355(6324):eaah4701.

    Article  PubMed  PubMed Central  Google Scholar 

  139. Ahtiainen L, Lefebvre S, Lindfors PH, Renvoisé E, Shirokova V, Vartiainen MK, et al. Directional cell migration, but not proliferation, drives hair placode morphogenesis. Dev Cell. 2014;28(5):588–602.

    Article  CAS  PubMed  Google Scholar 

  140. Noramly S, Morgan BA. BMPs mediate lateral inhibition at successive stages in feather tract development. Development. 1998;125(19):3775–87.

    Article  CAS  PubMed  Google Scholar 

  141. Gharzi A, Reynolds AJ, Jahoda CA. Plasticity of hair follicle dermal cells in wound healing and induction. Exp Dermatol. 2003;12(2):126–36.

    Article  CAS  PubMed  Google Scholar 

  142. Limat A, Breitkreutz D, Hunziker T, Boillat C, Wiesmann U, Klein E, et al. Restoration of the epidermal phenotype by follicular outer root sheath cells in recombinant culture with dermal fibroblasts. Exp Cell Res. 1991;194(2):218–27.

    Article  CAS  PubMed  Google Scholar 

  143. Plikus MV, Guerrero-Juarez CF, Ito M, Li YR, Dedhia PH, Zheng Y, et al. Regeneration of fat cells from myofibroblasts during wound healing. Science. 2017;355(6326):748–52.

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  144. Ko UH, Choi J, Choung J, Moon S, Shin JH. Physicochemically tuned myofibroblasts for wound healing strategy. Sci Rep. 2019;9(1):16070.

    Article  ADS  PubMed  PubMed Central  Google Scholar 

  145. Teng YY, Zou ML, Zhou XJ, Wu JJ, Liu SY, Yuan ZD, et al. Novel prospects for scarless wound healing: the roles of myofibroblasts and adipocytes. J Cell Mol Med. 2022;26(20):5113–21.

    Article  PubMed  PubMed Central  Google Scholar 

  146. Dunn MG, Silver FH, Swann DA. Mechanical analysis of hypertrophic scar tissue: structural basis for apparent increased rigidity. J Invest Dermatol. 1985;84(1):9–13.

    Article  CAS  PubMed  Google Scholar 

  147. Witte M, Rübhausen M, Jaspers S, Wenck H, Fischer F. A method to analyze the influence of mechanical strain on dermal collagen morphologies. Sci Rep. 2021;11(1):7565.

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  148. Janson IA, Putnam AJ. Extracellular matrix elasticity and topography: material-based cues that affect cell function via conserved mechanisms. J Biomed Mater Res A. 2015;103(3):1246–58.

    Article  PubMed  Google Scholar 

  149. Vizesi F, Jones C, Lotz N, Gianoutsos M, Walsh WR. Stress relaxation and creep: viscoelastic properties of common suture materials used for flexor tendon repair. J Hand Surg Am. 2008;33(2):241–6.

    Article  PubMed  Google Scholar 

  150. O’Brien L, Jones DJ. Silicone gel sheeting for preventing and treating hypertrophic and keloid scars. Cochrane Database Syst Rev. 2013;2013(9):CD003826.

    PubMed  PubMed Central  Google Scholar 

  151. Mustoe TA. Evolution of silicone therapy and mechanism of action in scar management. Aesthet Plast Surg. 2008;32(1):82–92.

    Article  Google Scholar 

  152. Kadziński L, Prokopowicz M, Jakóbkiewicz-Banecka J, Gabig-Cimińska M, Łukasiak J, Banecki B. Effect of silicone on the collagen fibrillogenesis and stability. J Pharm Sci. 2015;104(4):1275–81.

    Article  PubMed  PubMed Central  Google Scholar 

  153. Kuppan P, Vasanthan KS, Sundaramurthi D, Krishnan UM, Sethuraman S. Development of poly(3-hydroxybutyrate-co-3-hydroxyvalerate) fibers for skin tissue engineering: effects of topography, mechanical, and chemical stimuli. Biomacromol. 2011;12(9):3156–65.

    Article  CAS  Google Scholar 

  154. Lorden ER, Miller KJ, Bashirov L, Ibrahim MM, Hammett E, Jung Y, et al. Mitigation of hypertrophic scar contraction via an elastomeric biodegradable scaffold. Biomaterials. 2015;43:61–70.

    Article  CAS  PubMed  Google Scholar 

  155. Kuang D, Jiang F, Wu F, Kaur K, Ghosh S, Kundu SC, et al. Highly elastomeric photocurable silk hydrogels. Int J Biol Macromol. 2019;134:838–45.

    Article  CAS  PubMed  Google Scholar 

  156. Worch JC, Weems AC, Yu J, Arno MC, Wilks TR, Huckstepp RTR, et al. Elastomeric polyamide biomaterials with stereochemically tuneable mechanical properties and shape memory. Nat Commun. 2020;11(1):3250.

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  157. Rajasekharan AK, Gyllensten C, Blomstrand E, Liebi M, Andersson M. Tough ordered mesoporous elastomeric biomaterials formed at ambient conditions. ACS Nano. 2020;14(1):241–54.

    Article  CAS  PubMed  Google Scholar 

  158. Aghaei-Ghareh-Bolagh B, Mithieux SM, Weiss AS. Elastic proteins and elastomeric protein alloys. Curr Opin Biotechnol. 2016;39:56–60.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  159. Versteegden LR, van Kampen KA, Janke HP, Tiemessen DM, Hoogenkamp HR, Hafmans TG, et al. Tubular collagen scaffolds with radial elasticity for hollow organ regeneration. Acta Biomater. 2017;52:1–8.

    Article  CAS  PubMed  Google Scholar 

  160. Sieminski AL, Hebbel RP, Gooch KJ. The relative magnitudes of endothelial force generation and matrix stiffness modulate capillary morphogenesis in vitro. Exp Cell Res. 2004;297(2):574–84.

    Article  CAS  PubMed  Google Scholar 

  161. Rivilis I, Milkiewicz M, Boyd P, Goldstein J, Brown MD, Egginton S, et al. Differential involvement of MMP-2 and VEGF during muscle stretch- versus shear stress-induced angiogenesis. Am J Physiol Heart Circ Physiol. 2002;283(4):H1430–8.

    Article  CAS  PubMed  Google Scholar 

  162. Wu SH, Zhang F, Yao S, Tang L, Zeng HT, Zhu LP, et al. Shear stress triggers angiogenesis of late endothelial progenitor cells via the PTEN/Akt/GTPCH/BH4 pathway. Stem Cells Int. 2020;2020:5939530.

    Article  PubMed  PubMed Central  Google Scholar 

  163. Egginton S, Hussain A, Hall-Jones J, Chaudhry B, Syeda F, Glen KE. Shear stress-induced angiogenesis in mouse muscle is independent of the vasodilator mechanism and quickly reversible. Acta Physiol (Oxf). 2016;218(3):153–66.

    Article  CAS  PubMed  Google Scholar 

  164. Virumbrales-Muñoz M, Ayuso JM, Gong MM, Humayun M, Livingston MK, Lugo-Cintron KM, et al. Microfluidic lumen-based systems for advancing tubular organ modeling. Chem Soc Rev. 2020;49(17):6402–42.

    Article  PubMed  PubMed Central  Google Scholar 

  165. Dorati R, Pisani S, Chiesa E, Genta I, Bruni G, Modena T, et al. Electrospun tubular vascular grafts to replace damaged peripheral arteries: a preliminary formulation study. Int J Pharm. 2021;596:120198.

    Article  CAS  PubMed  Google Scholar 

  166. Wang J, Wei Y, Yi H, Liu Z, Sun D, Zhao H. Cytocompatibility of a silk fibroin tubular scaffold. Mater Sci Eng C Mater Biol Appl. 2014;34:429–36.

    Article  PubMed  Google Scholar 

  167. Wang Y, Shi H, Qiao J, Tian Y, Wu M, Zhang W, et al. Electrospun tubular scaffold with circumferentially aligned nanofibers for regulating smooth muscle cell growth. ACS Appl Mater Interfaces. 2014;6(4):2958–62.

    Article  CAS  PubMed  Google Scholar 

  168. Nawrotek K, Tylman M, Rudnicka K, Gatkowska J, Balcerzak J. Tubular electrodeposition of chitosan-carbon nanotube implants enriched with calcium ions. J Mech Behav Biomed Mater. 2016;60:256–66.

    Article  CAS  PubMed  Google Scholar 

  169. Huang W, Begum R, Barber T, Ibba V, Tee NC, Hussain M, et al. Regenerative potential of silk conduits in repair of peripheral nerve injury in adult rats. Biomaterials. 2012;33(1):59–71.

    Article  PubMed  Google Scholar 

  170. Huang S, Kuri P, Aubert Y, Brewster M, Li N, Farrelly O, et al. Lgr6 marks epidermal stem cells with a nerve-dependent role in wound re-epithelialization. Cell Stem Cell. 2021;28(9):1582-96.e6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  171. Li GN, Hoffman-Kim D. Evaluation of neurite outgrowth anisotropy using a novel application of circular analysis. J Neurosci Methods. 2008;174(2):202–14.

    Article  PubMed  PubMed Central  Google Scholar 

  172. Song Y, Li D, Farrelly O, Miles L, Li F, Kim SE, et al. The mechanosensitive ion channel piezo inhibits axon regeneration. Neuron. 2019;102(2):373-89.e6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  173. Li G, Li S, Zhang L, Chen S, Sun Z, Li S, et al. Construction of biofunctionalized anisotropic hydrogel micropatterns and their effect on Schwann cell behavior in peripheral nerve regeneration. ACS Appl Mater Interfaces. 2019;11(41):37397–410.

    Article  CAS  PubMed  Google Scholar 

  174. Li N, Folch A. Integration of topographical and biochemical cues by axons during growth on microfabricated 3-D substrates. Exp Cell Res. 2005;311(2):307–16.

    Article  CAS  PubMed  Google Scholar 

  175. Hoffman-Kim D, Mitchel JA, Bellamkonda RV. Topography, cell response, and nerve regeneration. Annu Rev Biomed Eng. 2010;12:203–31.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  176. Zhang L, Han Q, Chen S, Suo D, Zhang L, Li G, et al. Soft hydrogel promotes dorsal root ganglion by upregulating gene expression of Ntn4 and Unc5B. Colloids Surf B Biointerfaces. 2021;199:111503.

    Article  CAS  PubMed  Google Scholar 

  177. Abaricia JO, Farzad N, Heath TJ, Simmons J, Morandini L, Olivares-Navarrete R. Control of innate immune response by biomaterial surface topography, energy, and stiffness. Acta Biomater. 2021;133:58–73.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  178. Son H, Choi HS, Baek SE, Kim YH, Hur J, Han JH, et al. Shear stress induces monocyte/macrophage-mediated inflammation by upregulating cell-surface expression of heat shock proteins. Biomed Pharmacother. 2023;161:114566.

    Article  CAS  PubMed  Google Scholar 

  179. Cohen HC, Joyce EJ, Kao WJ. Biomaterials selectively modulate interactions between human blood-derived polymorphonuclear leukocytes and monocytes. Am J Pathol. 2013;182(6):2180–90.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  180. Atcha H, Jairaman A, Holt JR, Meli VS, Nagalla RR, Veerasubramanian PK, et al. Mechanically activated ion channel Piezo1 modulates macrophage polarization and stiffness sensing. Nat Commun. 2021;12(1):3256.

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  181. Solis AG, Bielecki P, Steach HR, Sharma L, Harman CCD, Yun S, et al. Mechanosensation of cyclical force by PIEZO1 is essential for innate immunity. Nature. 2019;573(7772):69–74.

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  182. Hotchkiss KM, Reddy GB, Hyzy SL, Schwartz Z, Boyan BD, Olivares-Navarrete R. Titanium surface characteristics, including topography and wettability, alter macrophage activation. Acta Biomater. 2016;31:425–34.

    Article  CAS  PubMed  Google Scholar 

  183. Bota PC, Collie AMB, Puolakkainen P, Vernon RB, Sage EH, Ratner BD, et al. Biomaterial topography alters healing in vivo and monocyte/macrophage activation in vitro. J Biomed Mater Res A. 2010;95(2):649–57.

    Article  PubMed  PubMed Central  Google Scholar 

  184. Park JW, Han SH, Hanawa T. Effects of surface nanotopography and calcium chemistry of titanium bone implants on early blood platelet and macrophage cell function. Biomed Res Int. 2018;2018:1362958.

    Article  PubMed  PubMed Central  Google Scholar 

  185. Saino E, Focarete ML, Gualandi C, Emanuele E, Cornaglia AI, Imbriani M, et al. Effect of electrospun fiber diameter and alignment on macrophage activation and secretion of proinflammatory cytokines and chemokines. Biomacromol. 2011;12(5):1900–11.

    Article  CAS  Google Scholar 

  186. Schoenenberger AD, Tempfer H, Lehner C, Egloff J, Mauracher M, Bird A, et al. Macromechanics and polycaprolactone fiber organization drive macrophage polarization and regulate inflammatory activation of tendon in vitro and in vivo. Biomaterials. 2020;249:120034.

    Article  CAS  PubMed  Google Scholar 

  187. Palani K, Rahman M, Hasan Z, Zhang S, Qi Z, Jeppsson B, et al. Rho-kinase regulates adhesive and mechanical mechanisms of pulmonary recruitment of neutrophils in abdominal sepsis. Eur J Pharmacol. 2012;682(1–3):181–7.

    Article  CAS  PubMed  Google Scholar 

  188. Abaricia JO, Shah AH, Musselman RM, Olivares-Navarrete R. Hydrophilic titanium surfaces reduce neutrophil inflammatory response and NETosis. Biomater Sci. 2020;8(8):2289–99.

    Article  CAS  PubMed  Google Scholar 

  189. Griffin DR, Archang MM, Kuan CH, Weaver WM, Weinstein JS, Feng AC, et al. Activating an adaptive immune response from a hydrogel scaffold imparts regenerative wound healing. Nat Mater. 2021;20(4):560–9.

    Article  ADS  CAS  PubMed  Google Scholar 

  190. Downing TL, Soto J, Morez C, Houssin T, Fritz A, Yuan F, et al. Biophysical regulation of epigenetic state and cell reprogramming. Nat Mater. 2013;12(12):1154–62.

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  191. Song Y, Soto J, Li S. Mechanical regulation of histone modifications and cell plasticity. Curr Opin Solid State Mater Sci. 2020;24(6):100872.

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  192. Chu SY, Chou CH, Huang HD, Yen MH, Hong HC, Chao PH, et al. Mechanical stretch induces hair regeneration through the alternative activation of macrophages. Nat Commun. 2019;10(1):1524.

    Article  ADS  PubMed  PubMed Central  Google Scholar 

  193. Wang X, Wang J, Guo L, Wang X, Chen H, Wang X, et al. Self-assembling peptide hydrogel scaffolds support stem cell-based hair follicle regeneration. Nanomedicine. 2016;12(7):2115–25.

    Article  CAS  PubMed  Google Scholar 

  194. Chen P, Zhang F, Fan Z, Shen T, Liu B, Chen R, et al. Nanoscale microenvironment engineering for expanding human hair follicle stem cell and revealing their plasticity. J Nanobiotechnol. 2021;19(1):94.

    Article  Google Scholar 

  195. Zhang Z, Li W, Liu Y, Yang Z, Ma L, Zhuang H, et al. Design of a biofluid-absorbing bioactive sandwich-structured Zn-Si bioceramic composite wound dressing for hair follicle regeneration and skin burn wound healing. Bioact Mater. 2020;6(7):1910–20.

    PubMed  PubMed Central  Google Scholar 

  196. Zhang Z, Li W, Chang D, Wei Z, Wang E, Yu J, et al. A combination therapy for androgenic alopecia based on quercetin and zinc/copper dual-doped mesoporous silica nanocomposite microneedle patch. Bioact Mater. 2022;24:81–95.

    PubMed  PubMed Central  Google Scholar 

  197. Young TH, Lee CY, Chiu HC, Hsu CJ, Lin SJ. Self-assembly of dermal papilla cells into inductive spheroidal microtissues on poly(ethylene-co-vinyl alcohol) membranes for hair follicle regeneration. Biomaterials. 2008;29(26):3521–30.

    Article  CAS  PubMed  Google Scholar 

  198. Li X, Li H, Zhang M, Chen L, Zhang B. Cell proliferation and differentiation during the three dimensional reconstitution of eccrine sweat glands. J Mol Histol. 2017;48(2):113–20.

    Article  PubMed  Google Scholar 

  199. Li H, Li X, Zhang M, Chen L, Zhang B, Tang S, et al. Three-dimensional co-culture of BM-MSCs and eccrine sweat gland cells in Matrigel promotes transdifferentiation of BM-MSCs. J Mol Histol. 2015;46(4–5):431–8.

    Article  CAS  PubMed  Google Scholar 

  200. Huang S, Xu Y, Wu C, Sha D, Fu X. In vitro constitution and in vivo implantation of engineered skin constructs with sweat glands. Biomaterials. 2010;31(21):5520–5.

    Article  CAS  PubMed  Google Scholar 

  201. Liu X, Liu H, Qu X, Lei M, Zhang C, Hong H, et al. Electrical signals triggered controllable formation of calcium-alginate film for wound treatment. J Mater Sci Mater Med. 2017;28(10):146.

    Article  PubMed  Google Scholar 

  202. Yao G, Jiang D, Li J, Kang L, Chen S, Long Y, et al. Self-activated electrical stimulation for effective hair regeneration via a wearable omnidirectional pulse generator. ACS Nano. 2019;13(11):12345–56.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  203. Harn HI, Chiu PY, Lin CH, Chen HY, Lai YC, Yang FS, et al. Topological distribution of wound stiffness modulates wound-induced hair follicle neogenesis. Pharmaceutics. 2022;14(9):1926.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  204. Koester J, Miroshnikova YA, Ghatak S, Chacon-Martinez CA, Morgner J, Li X, et al. Niche stiffening compromises hair follicle stem cell potential during ageing by reducing bivalent promoter accessibility. Nat Cell Biol. 2021;23(7):771–81.

    Article  CAS  PubMed  Google Scholar 

  205. Han I, Shim KJ, Kim JY, Im SU, Sung YK, Kim M, et al. Effect of poly(3-hydroxybutyrate-co-3-hydroxyvalerate) nanofiber matrices cocultured with hair follicular epithelial and dermal cells for biological wound dressing. Artif Organs. 2007;31(11):801–8.

    Article  CAS  PubMed  Google Scholar 

  206. Lu C, Fuchs E. Sweat gland progenitors in development, homeostasis, and wound repair. Cold Spring Harb Perspect Med. 2014;4(2):a015222.

    Article  PubMed  PubMed Central  Google Scholar 

  207. Chen R, Zhu Z, Ji S, Geng Z, Hou Q, Sun X, et al. Sweat gland regeneration: current strategies and future opportunities. Biomaterials. 2020;255:120201.

    Article  CAS  PubMed  Google Scholar 

  208. Lu CP, Polak L, Keyes BE, Fuchs E. Spatiotemporal antagonism in mesenchymal-epithelial signaling in sweat versus hair fate decision. Science. 2016;354(6319):6102.

    Article  Google Scholar 

  209. Price AJ, Cost AL, Ungewiß H, Waschke J, Dunn AR, Grashoff C. Mechanical loading of desmosomes depends on the magnitude and orientation of external stress. Nat Commun. 2018;9(1):5284.

    Article  ADS  PubMed  PubMed Central  Google Scholar 

  210. Meng F, Sachs F. Orientation-based FRET sensor for real-time imaging of cellular forces. J Cell Sci. 2012;125(Pt 3):743–50.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  211. Meng F, Sachs F. Visualizing dynamic cytoplasmic forces with a compliance-matched FRET sensor. J Cell Sci. 2011;124(Pt 2):261–9.

    Article  CAS  PubMed  Google Scholar 

  212. Segura Chávez PA, Bonhomme J, Bellaredj MLF, Olive L, Beyssen D, Oudich M, et al. Love wave sensor with high penetration depth for potential application in cell monitoring. Biosensors (Basel). 2022;12(2):61.

    Article  PubMed  Google Scholar 

  213. Yoon S, Windoffer R, Kozyrina AN, Piskova T, Di Russo J, Leube RE. Combining image restoration and traction force microscopy to study extracellular matrix-dependent keratin filament network plasticity. Front Cell Dev Biol. 2022;10:901038.

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

Not applicable.

Funding

This work was supported in part by the National Nature Science Foundation of China (92268206, 81830064), the CAMS Innovation Fund for Medical Sciences (CIFMS, 2019-I2M-5-059), the Military Medical Research Projects (145AKJ260015000X, 2022-JCJQ-ZB-09600), the Military Key Basic Research of Foundational Strengthening Program (2020-JCJQ-ZD-256-021), the Science Foundation of National Defense Science and Technology for Excellent Young (2022-JCJQ-ZQ-017), the Military Medical Research and Development Projects (AWS17J005, 2019-126), and the Specific Research Fund of The Innovation Platform for Academicians of Hainan Province (YSPTZX202317).

Author information

Authors and Affiliations

Authors

Contributions

YYL and SFJ wrote the original draft, and arranged figures and tables of the manuscript. YFJ participated in literature search. YYL, SFJ, YFJ, and XYS reviewed and revised the manuscript. XBF and XYS received funding support. All authors contributed to the manuscript and approved the submitted version.

Corresponding authors

Correspondence to Xiao-Bing Fu, Yu-Feng Jiang or Xiao-Yan Sun.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare no competing interests.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Li, YY., Ji, SF., Fu, XB. et al. Biomaterial-based mechanical regulation facilitates scarless wound healing with functional skin appendage regeneration. Military Med Res 11, 13 (2024). https://0-doi-org.brum.beds.ac.uk/10.1186/s40779-024-00519-6

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://0-doi-org.brum.beds.ac.uk/10.1186/s40779-024-00519-6

Keywords