Abstract Skin wound healing is a dynamic process, yet scaffolds enabling stage-specific modulation remain limited. We fabricated a nanofiber scaffold from FDA-approved materials, consisting of two outer layers of radially aligned and random poly(ε-caprolactone) fibers and a middle layer of electrosprayed phase-change microparticles loaded with platelet-derived growth factor–BB (PDGF-BB)/vascular endothelial growth factor (VEGF) in the periphery and PDGF-BB/epidermal growth factor (EGF) in the center. Near-infrared irradiation through a photomask enabled spatiotemporal control of growth factor release, aligning PDGF-BB, VEGF, and EGF delivery with specific phases of wound healing to promote vascularization, cell proliferation, and tissue remodeling. In a preclinical porcine model, it enhanced closure and modulated the microenvironment by activating PI3K-Akt, MAPK, and immune pathways, up-regulating genes for survival and repair while down-regulating those linked to apoptosis and inflammation. With scalable manufacturing and large-animal efficacy, this scaffold holds translational potential for skin wound healing. __________________________________________________________________ Nanofiber scaffolds enable spatiotemporal release of bioactive effectors to promote skin wound healing. INTRODUCTION Skin wounds represent a major clinical challenge, and rapid and effective healing is essential to reduce infection, minimize scarring, and restore function ([40]1, [41]2). Traditional treatments, such as autografts, face limitations including donor site morbidity and immune rejection ([42]3–[43]5). Synthetic scaffolds have emerged to address these issues ([44]6, [45]7). For example, Restrata, comprising electrospun poly(lactic-co-glycolic acid) and polydioxanone nanofibers, have shown efficacy in the clinical setting by providing a structural support ([46]8, [47]9). These scaffolds, however, primarily serve a passive protection role and lack the ability to dynamically regulate the complex biological processes involved in skin regeneration ([48]10, [49]11). Consequently, their use often results in suboptimal healing outcomes and substantial scar formation ([50]12). Addressing these limitations necessitates the development of multifunctional, bioactive scaffolds capable of dynamically adapting their functionality with the overlapping yet distinct phases of skin repair: hemostasis and inflammation, proliferation, and tissue remodeling ([51]13, [52]14). Each phase is characterized by specific cellular activities and extracellular matrix (ECM) interactions with growth factors, which collectively determine the success of skin regeneration ([53]15). In the early stage of wound healing, vascular regeneration plays a critical role, indirectly influencing neural network regeneration and modulating inflammatory responses ([54]16). Supplementing exogenous growth factors, such as platelet-derived growth factor (PDGF) and vascular endothelial growth factor (VEGF), is essential at this stage ([55]17, [56]18). During the proliferative phase, various types of cells, such as fibroblasts, epithelial cells, and keratinocytes, migrate and proliferate, as supported by ongoing vascular regeneration ([57]19). Growth factors like PDGF and epidermal growth factor (EGF), which enhance the migration and proliferation of these cells, are particularly important during this phase ([58]20, [59]21). In the tissue remodeling stage, the reorganization of collagen and deposition of ECM lead to the maturation of regenerated skin tissue ([60]22). Achieving a precise control over the timing and location of the release of these factors from the scaffolds is essential for effective skin tissue regeneration. As for the structure of the scaffold, electrospun nanofibers have gained considerable attention for their ECM-like properties, as demonstrated by the clinical products ([61]23–[62]26). Radially aligned fibers, in particular, can provide topographical cues that promote cell migration from the periphery to the center, holding great promise for wound healing ([63]27, [64]28). Now, bioactive substances are commonly loaded into the fibers using coaxial or multiple-axial electrospinning, with their release governed by diffusion and material degradation ([65]29, [66]30). This approach often results in an initial burst release, followed by minimal or no release in later stages, greatly undermining sustained tissue regeneration and raising concerns about potential side effects due to high initial dosages. Stimulus-responsive nanofiber scaffolds that respond to internal cues such as reactive oxygen species ([67]31, [68]32) or external triggers ([69]33–[70]35) offer a promising strategy for controlled release of bioactive molecules. For example, a chitosan-based nanofiber dressing responsive to pH and glucose levels promoted wound healing in diabetic rats ([71]32). Additionally, physical therapies such as electric fields ([72]34), ultrasound ([73]33), and light have been applied in combination with nanofiber scaffolds to trigger the release of growth factors. Typically, near-infrared (NIR) light, a light source used in clinical treatments, has been used in photothermal therapy, including both photothermally triggered release and mild hypothermal effect ([74]35–[75]37). In one study, the release of bone morphogenetic protein–4 from polydopamine nanoparticles was triggered using NIR light during the proliferative stage of skin wound healing to help inhibit scar formation ([76]38). In addition, photothermal therapy is commonly used to treat skin wounds infected with drug-resistant bacteria and diabetic ulcer treatment ([77]39). The mild hypothermal effect can effectively eliminate bacteria at a specific temperature, reduce inflammation, and promote wound healing ([78]40, [79]41). Among various exogenous stimuli, NIR light offers advantages for cutaneous applications, including noncontact operation and precise spatial activation, and deep tissue penetration without harming surrounding ([80]42). However, all these strategies are limited to partially controlled release for only one stage of skin repair, making it difficult to achieve stage-specific, targeted treatments. The integration of NIR light with thermosensitive materials provides a versatile platform for spatiotemporal drug delivery. Thermosensitive phase change materials (PCMs), such as fatty acid–based mixtures, undergo solid-to-liquid phase transitions upon photothermal activation, enabling the controlled release of encapsulated bioactive substances at specific temperatures ([81]43, [82]44). For instance, the eutectic mixture of lauric acid and stearic acid, with a melting point tailored to the physiological temperature (e.g., 39.5°C), offers a precise control of temperature-responsive release without causing thermal damage to the surrounding tissues ([83]45). Prior studies have used a combination of electrospraying and electrospinning techniques to fabricate nanofiber scaffolds embedded with PCM microparticles to efficiently encapsulate and release therapeutic agents ([84]46). NIR-induced photothermal activation, in conjunction with a photomask, triggered the localized melting of PCMs, enabling the targeted release of therapeutic agents and thereby enhancing cellular migration ([85]47). Despite these promising results, the in vivo efficacy of these systems remains to be explored. Additionally, the electrospraying process allows for a precise control of the spatial locations of various microparticles on specific regions of a scaffold using a physical mask. We hypothesize that such a system can dynamically regulate the release of different growth factors in response to NIR stimuli, thereby optimizing the microenvironment at various stages of the healing process. Here, we developed a nanofiber scaffold by incorporating multiple cues to spatiotemporally promote skin wound healing. These cues include an aligned topographical structure for directing cell migration, bioactive growth factors with defined concentrations to regulate cell behavior at specific stages, and mild photothermal effect to modulate the biological microenvironment. The scaffold was fabricated using a combination of electrospinning and masked coaxial electrospraying techniques ([86]Fig. 1). The resulting scaffold, with a diameter of 2.5 cm, consists of three layers: an inner layer of radially aligned poly(ε-caprolactone) (PCL) nanofibers, a middle layer of growth factor–encapsulated PCM microparticles, and an outer layer of randomly oriented PCL nanofibers. The microparticles were deposited on the nanofibers by sequentially using circular and annular masks during electrospraying, creating two concentric regions within the middle layer: The inner circular region (1.875 cm in diameter) contained PCM microparticles loaded with PDGF-BB, EGF, and indocyanine green (ICG), while the outer annular region contained PCM microparticles loaded with PDGF-BB, VEGF, and ICG. ICG, a Food and Drug Administration (FDA)–approved photothermal agent, was included as the photothermal agent. This scaffold was denoted PCM-P/V/E to indicate the encapsulation of PDGF-BB, VEGF, and EGF. Spatiotemporally controlled release of these growth factors was achieved through periodic NIR irradiation using three size-adjustable photomasks, allowing for precise and localized delivery of the encapsulated effectors. Full-thickness skin injury models based on small animals (rabbit) were used to evaluate the scaffold’s in vivo efficacy, while large-animal (porcine) studies provided further insights into the mechanisms of skin tissue repair and critical data to support the promise for clinical translation. Notably, this study used FDA-approved, clinically validated raw materials, combined with a scalable one-step fabrication process, to develop a skin tissue regeneration scaffold optimized for clinical application. The used photothermal therapy features a lightweight, user-friendly device that can be self-administered by patients under medical guidance. The proposed scaffold offers a highly innovative therapeutic approach for patients with severe skin injuries, addressing the urgent need for advanced wound repair solutions. Fig. 1. Schematic illustration of a bioactive nanofiber scaffold combined with NIR irradiation for accelerated skin wound healing. [87]Fig. 1. [88]Open in a new tab The scaffold is fabricated using electrospinning and mask-assisted coaxial electrospraying. It consists of three layers: the inner layer, made of radially aligned nanofibers; the outer layer, composed of randomly oriented nanofibers; and a middle layer containing phase-change microparticles. These microparticles are loaded with different combinations of growth factors: The peripheral region contains PDGF-BB and VEGF, while the central region contains PDGF-BB and EGF. NIR light and a size-adjustable photomask are applied at specific time points, enabling spatiotemporally controlled release of growth factors, thereby creating a favorable environment for skin regeneration and functional recovery. D, the outer diameter of region B; d, the outer diameter of region A. RESULTS Characterizations of the nanofiber scaffolds The PCM-P/V/E scaffold composed of two layers of nanofibers sandwiched with PCM microparticles. The radially aligned nanofibers, with an average diameter of 489 ± 90 nm, extended from the center of the scaffold toward the periphery, providing topographical cues for cell migration and cytoskeletal alignment ([89]Fig. 2A and fig. S1A). The randomly oriented nanofibers, with an average diameter of 394 ± 72 nm, were added to enhance the mechanical integrity of the scaffold, allowing it to withstand the forces typically encountered during surgical suturing ([90]Fig. 2B and fig. S1B). The PCM microparticles were uniformly dispersed throughout the scaffold ([91]Fig. 2C). These microparticles, being larger than the pore size formed by the physically stacked fibers, were effectively entrapped between the two nanofiber layers. Following oxygen plasma treatment, the water contact angle of the scaffold was 35°, indicating a hydrophilic surface (fig. S2). In mechanical testing, the scaffold demonstrated a tensile strength of 11.4 MPa and an elongation at break of 105.3%, reflecting favorable mechanical properties ([92]Fig. 2D). During surgical handling, the scaffold maintained structural integrity and was able to endure the forces applied during suturing. Fig. 2. In vitro physicochemical characterization of the scaffold. [93]Fig. 2. [94]Open in a new tab Scanning electron microscopy (SEM) images of the (A) radially aligned nanofibers and (B) randomly oriented nanofibers at low and high magnifications, respectively. Insets: Corresponding two-dimensional fast Fourier transform images derived from the SEM images. (C) SEM image of PCM microparticles distributed on the radially aligned nanofibers. (D) Stress-strain curve of the scaffold. (E) Temperature changes of the scaffold upon irradiation with an 808-nm NIR laser at varying power densities. (F) Temperature changes of the scaffold over five cycles of 2-min NIR irradiation. (G) Temperature changes of the scaffold over four cycles of 10-min NIR irradiation. (H) Schematic illustration of the photomask strategy for selective NIR irradiation of designated regions of the scaffold. (I to K) Schematic illustrations and corresponding temperature profiles of the scaffold in exposed and photomask-covered regions during (I) round 1, (J) round 2, and (K) round 3 of NIR irradiation, respectively. Insets: Infrared thermal images showing the temperature distribution of exposed and masked regions. (L) Total amounts and (M) cumulative release profiles of PDGF-BB, VEGF, and EGF from the scaffold following sequentially, masked NIR irradiation. Stable photothermal effect of the nanofiber scaffold upon NIR irradiation The nanofiber scaffold exhibited a stable photothermal effect by encapsulating ICG within the PCM microparticles. Upon irradiation with an 808-nm NIR laser at varying power densities, we monitored the temperature changes using an infrared thermal imaging camera. As shown in [95]Fig. 2E, when irradiated at power densities of 0.6 and 0.7 W/cm^2, the temperatures of the scaffold increased and eventually stabilized at 42.6° and 45.0°C, respectively, both above the melting point (39°C) of the PCM, which could induce a solid-to-liquid phase transition (fig. S3). Therefore, we selected a power density in the range of 0.6 to 0.7 W/cm^2 for subsequent experiments to ensure that the temperature exceeded the melting point of PCM without causing heat-induced damage to the surrounding tissue. To assess the stability of the photothermal effect, we irradiated the scaffold for multiple cycles with varied durations. As shown in [96]Fig. 2F, the temperature rapidly increased from room temperature to 39.6° to 43.8°C upon irradiation by the NIR laser. After 2 min of irradiation, the laser was turned off, and the scaffold quickly returned to room temperature. This cycle was repeated for an additional four times, with the temperature response remaining consistent throughout. To better simulate the in vivo conditions, we extended the irradiation duration to 10 min per cycle and monitored the temperature changes over time. As shown in [97]Fig. 2G, upon laser irradiation, the temperature increased rapidly and stabilized within the range of 39.2° to 41.2°C. After 10 min of irradiation, the scaffold cooled down quickly once the laser was turned off. This process was repeated three additional cycles, during which the temperature remained within the desired range. These results demonstrate that the scaffold maintained a stable photothermal effect over multiple irradiation cycles. The good stability could be attributed to the temperature-regulating properties of PCM, which absorbed latent heat during the phase transition, thereby maintaining the scaffold temperature within the desired range. NIR laser–triggered, spatiotemporally controlled release of specific growth factors from the nanofiber scaffold We used a photomask strategy to selectively expose different regions of the scaffold to NIR laser irradiation, triggering the PCM microparticles to melt and thereby release the encapsulated growth factors in the irradiated regions ([98]Fig. 2H). During the first three rounds of irradiation, we used three circular photomasks with diameters of 2, 1.5, and 0.5 cm, respectively, to sequentially expose region I, regions I + II, and regions I + II + III to the laser. In the fourth round, the entire scaffold was exposed to the laser. During each round of irradiation, the temperature in the exposed region increased to 39° to 42°C, while the temperature in the photomask-covered regions only showed a slight increase ([99]Fig. 2, I to K). To validate the feasibility of spatiotemporally controlling the release of growth factors, we encapsulated two model substances, trypan blue and rhodamine B, within the PCM microparticles in the peripheral and central regions of the scaffold, respectively (fig. S4). As shown in fig. S4A, the microparticles were exactly deposited in the designated regions. Upon NIR irradiation with corresponding photomasks, both trypan blue and rhodamine B were sequentially released from the scaffold (figs. S5 to S7). We then used the masking strategy to irradiate different regions of the PCM-P/V/E scaffold for 10 min per week from week 0 to week 3 to evaluate the release profile of growth factors from the scaffold. In the first round of NIR irradiation, region I, which contained PCM microparticles loaded with PDGF-BB and VEGF, was exposed. The amounts of PDGF-BB and VEGF released were 36.1 ± 1.2 ng and 35.9 ± 1.1 ng, respectively ([100]Fig. 2L). In the second round, regions I + II (containing PDGF-BB, VEGF, and EGF) were exposed, resulting in the release of 29.0 ± 1.8 ng of PDGF-BB, 23.1 ± 1.5 ng of VEGF, and 20.9 ± 1.1 ng of EGF. During the third round, regions I + II + III were exposed, leading to the release of 32.0 ± 1.3 ng of PDGF-BB and 35.8 ± 1.5 ng of EGF. Last, when the entire scaffold was exposed to the laser, 10.4 ± 1.9 ng of PDGF-BB and 7.5 ± 0.4 ng of EGF were released. These results demonstrate that NIR laser irradiation combined with a photomask enabled spatiotemporal controls of growth factor release from specific regions of the scaffold, with sufficient amounts to induce a biological effect while avoiding excessive or uncontrolled delivery. We further quantified the cumulative release of the three growth factors over the course of the experiment. As shown in [101]Fig. 2M, the release of PDGF-BB showed a continuous upward trend, with a cumulative release amount of 107.5 ± 0.8 ng. VEGF was predominantly released from regions I + II during the initial first round and the first week, with a total release of 59.0 ± 1.8 ng. EGF began releasing from the first week and continued through week 3, with a cumulative release of 64.2 ± 0.9 ng. These results confirm the ability to achieve NIR laser–triggered, spatiotemporally controlled release of specific growth factors from the scaffold. Manipulation of cell migration and proliferation by the scaffolds To evaluate the impacts of the spatiotemporally delivered growth factors on cell behaviors, we fabricated additional control scaffolds: PCM-P (with only PDGF-BB and ICG), PCM-P/E (with PDGF-BB, EGF, and ICG), a pristine fiber scaffold (Pristine scaffold), and a scaffold only with PCM microparticles (PCM group). The biological activity of the released PDGF-BB and VEGF was evaluated through an in vitro angiogenesis assay by culturing human umbilical vein endothelial cells (HUVECs) on Matrigel matrix in the different culture medium. Growth factors released from the PCM-P, PCM-P/E, and PCM-P/V/E scaffolds were collected and added to the culture medium to induce angiogenesis in HUVECs. These groups were named Released-P, Released-P/E, and Released-P/V/E, respectively, with HUVECs cultured in plain medium on the tissue culture plate (TCP) serving as the Blank control group. For comparison, free PDGF-BB (100 ng/mL) or VEGF (60 ng/mL) was added at a concentration similar to the released amount to serve as controls, referred to as Free-P and Free-V, respectively. After culturing for 6 hours, as shown in [102]Fig. 3A, the Blank group exhibited limited branching, with HUVECs predominantly adopting a flattened morphology and displaying filopodia. In contrast, the addition of free PDGF-BB or VEGF significantly promoted vascular network formation, with increased numbers of branch points and tubules, indicating angiogenic activity ([103]Fig. 3, B and C). The Released-P and Released-P/E groups showed similar angiogenesis to the Free-P group, with comparable branch points and tubule formation. The Released-P/V/E group, however, demonstrated more extensive vascular structures, including well-formed walls and lumens, and showed significantly increased numbers of branch points and tubules compared to the Released-P and Released-P/E groups. These results confirmed that the growth factors released from the scaffolds retained the biological activity. Fig. 3. Manipulation of cell migration and proliferation by the scaffolds under NIR irradiation. [104]Fig. 3. [105]Open in a new tab (A) Fluorescence micrographs showing angiogenesis of HUVECs cultured in different media: plain culture medium (Blank), medium supplemented with free PDGF-BB (Free-P) or free VEGF (Free-V), and medium containing growth factors released from scaffolds (Released-P, Released-P/E, and Released-P/V/E). Angiogenic structures were stained with calcein-AM (green). Quantification of (B) branch points and (C) number of tubules, respectively, formed by HUVECs in each group. (D) Optical density (OD) values of HUVECs cultured on different scaffolds at days 1, 3, 5, and 7. *P < 0.05, the blank horizontal lines represent **P < 0.01. (E) Fluorescence micrographs of live/dead staining of HUVECs cultured on different scaffolds at days (d) 1 and 3. Live cells were stained with calcein-AM (green) and dead cells with propidium iodide (red), respectively. (F) Fluorescence micrographs showing the radial migration of L929, NIH-3T3, and HaCaT cells from the periphery toward the center of the scaffolds after 4 days. White dashed lines indicate the initial boundary at the periphery of the scaffold. F-actin was stained with Alexa Fluor 555 phalloidin (red) and nuclei with 4′,6-diamidino-2-phenylindole (DAPI) (blue), respectively. (G to I) Quantitative analysis of the average and the farthest migration distances of (G) L929, (H) NIH-3T3, and (I) HaCaT cells, respectively, in different groups. **P < 0.01 versus all other groups; ^##P < 0.01 versus other five groups. We further assessed the biocompatibility of the different scaffolds using the Cell Counting Kit-8 assay. NIR laser irradiation was applied to the PCM-P, PCM-P/E, and PCM-P/V/E scaffolds on days 0, 2, 4, and 6, using different photomasks to achieve spatially selective masked irradiation (MI). For comparison, a single irradiation (SI) was performed on the entire surface of the PCM and PCM-P scaffolds without masking. As shown in [106]Fig. 3D, the optical density (OD) values for all groups progressively increased from day 1 to day 7, confirming the good biocompatibility of all scaffolds. Notably, on day 7, cell proliferation in the PCM-P/V/E (MI) group was significantly higher than in the other groups, which can be attributed to the spatiotemporally delivered PDGF-BB, VEGF, and EGF. [107]Figure 3E presents the results of live/dead staining of HUVECs cultured on the scaffolds for 1 and 3 days. A predominance of live cells (green fluorescence) with minimal dead cells (red fluorescence) was observed across all groups. Notably, the PCM-P/V/E (MI) group exhibited the highest density of live cells, indicating superior cytocompatibility and cell-supportive properties. Following skin injury, the recruitment and directed migration of fibroblasts and epidermal cells from the periphery of the wound toward the center are essential processes for effective wound healing. To replicate this behavior in vitro, the migratory response of L929 and NIH-3 T3 fibroblasts, along with human keratinocyte HaCaT cells, was assessed on the scaffolds over a 4-day period. A “wound” region was created by placing a polydimethylsiloxane disc (2 cm in diameter) at the center of the scaffold, and cells were then seeded circumferentially around the scaffold’s periphery. In the MI groups, NIR laser exposure was performed on days 0, 1, 2, and 3 using photomasks to spatially control the regions being irradiated. As shown in [108]Fig. 3F and fig. S8, cells cultured on TCP exhibited disorganized morphologies and minimal migration into the central region. In contrast, on the nanofiber scaffolds, L929 and NIH-3T3 cells displayed elongated morphologies aligned with the fiber orientation, while HaCaT cells appeared more densely clustered. All three cell types displayed greater migratory capacity on the scaffolds relative to TCP. Notably, the PCM-P (MI) group exhibited a larger migration area than the PCM-P (SI) group, indicating that multiple cycles of spatially controlled PDGF-BB release effectively sustained the promigratory effect. Among all groups, the PCM-P/V/E (MI) group induced the most extensive cell migration within the wound region. Quantitative analysis of L929 cell migration distances ([109]Fig. 3G) revealed a consistent trend, where both the average and the farthest migration distances followed the order: TCP < Pristine scaffold < PCM (SI) < PCM-P (SI) < PCM-P (MI) < PCM-P/E (MI) < PCM-P/V/E (MI). Specifically, the average migration distances of the PCM-P (MI) and PCM-P/V/E (MI) groups were 379.2 ± 9.8 and 502.5 ± 4.0 μm, corresponding to 2.2- and 4.3-fold increases, respectively, relative to the Pristine scaffold group. The farthest migration distances in these groups were 883.3 ± 54.4 and 1140.0 ± 35.6 μm, representing 1.6- and 2.4-fold increases, respectively. Similarly, both NIH-3T3 fibroblasts and HaCaT keratinocytes exhibited significantly enhanced migration in the PCM-P/V/E (MI) group. Specifically, the average migration distance increased by 3.3-fold for NIH-3T3 cells and 2.1-fold for HaCaT cells, respectively, compared to the Pristine scaffold group ([110]Fig. 3, H and I). Collectively, these results demonstrate that the integration of radially aligned nanofibers with spatiotemporally delivered PDGF-BB, VEGF, and EGF synergistically enhanced cell migration, a critical determinant for promoting efficient wound closure. The scaffold promotes rabbit full-thickness skin wound healing upon NIR irradiation In vivo experiments were conducted to evaluate the therapeutic efficacy of the scaffolds in promoting full-thickness skin wound healing. Circular wounds (2.5 cm in diameter) were created on the dorsal surface of New Zealand white rabbits, followed by suturing of the scaffolds into place (fig. S9). Six scaffold groups (i.e., those used in cell studies) were tested to evaluate the distinct functions of the cues: Pristine scaffold, PCM (SI), PCM-P (SI), PCM-P (MI), PCM-P/E (MI), and PCM-P/V/E (MI). An untreated defect group without scaffold implantation served as the Blank control. As shown in [111]Fig. 4A, for the MI groups, NIR irradiation was applied for 10 min once per week over a 4-week period using three distinct photomasks. During irradiation, the temperature changes were monitored using an infrared thermal camera. The temperature in the irradiated region increased above 39°C within 90 s and remained in the range of 39° to 42°C during all four rounds of irradiation (fig. S10). This controlled mild heating could also promote vascular regeneration and wound healing without causing thermal damage to the tissue, as supported by previous studies ([112]48, [113]49). Photographs of the wound site were taken at each time point to visually assess the wound closure, and, at week 4 postsurgery, skin tissue samples were collected ([114]Fig. 4B). Fig. 4. In vivo evaluation of full-thickness skin wound healing in rabbits under NIR irradiation. [115]Fig. 4. [116]Open in a new tab (A) Schematic illustration of the masked NIR irradiation protocol applied at weeks 0, 1, 2, and 3 postsurgery. The inset shows infrared thermal images of the scaffold, indicating a stable temperature range of 39° to 42°C during continuous irradiation. (B) Representative macroscopic images of wound healing in each group over a 4-week period. (C) Pie charts showing the percentage of wound closure in different groups. (D) Hematoxylin and eosin (H&E) staining and (E) Sirius red staining micrographs of regenerated skin tissues from different groups at 4 weeks postsurgery. At the early stage of healing (week 1), scab formation was observed in all groups. The Pristine scaffold group showed no significant improvement in wound closure compared to the Blank control, indicating limited bioactivity ([117]Fig. 4C and fig. S11). The PCM (SI) group exhibited a greater wound closure percentage of 33.6 ± 1.0%. In the PCM-P (SI) group, the percentage of wound closure was 1.3-fold higher than that of the PCM (SI) group, indicating the additional contribution of PDGF-BB delivery in promoting wound healing. The PCM-P (MI) group achieved a wound closure percentage of 63.6 ± 1.2%. However, no statistically significant differences in wound healing rates were observed between the PCM-P (MI), PCM-P/E (MI), and PCM-P/V/E (MI) groups at this early time point, suggesting that the effects of additional EGF and VEGF incorporation may require a longer period to manifest in measurable macroscopic wound closure. The second and third weeks postsurgery represented the peak phase of wound healing, characterized by rapid reduction in wound size. Specifically, at week 2, the Blank group exhibited minimal wound reduction, with signs of suppuration. The wound closure percentages in the Pristine scaffold, PCM (SI), PCM-P (SI), PCM-P (MI), PCM-P/E (MI), and PCM-P/V/E (MI) groups were 53.8 ± 1.0%, 84.3 ± 0.9%, 89.9 ± 1.1%, 90.3 ± 1.8%, 92.6 ± 0.9%, and 95.4 ± 0.9%, respectively. Notably, the PCM-P/V/E (MI) scaffold obviously accelerated wound healing, achieving a 1.8-fold improvement compared to the Pristine scaffold group, indicating faster healing of a large wound. In addition, scaffolds and regenerated tissue from the PCM-P (SI) and PCM-P (MI) groups were collected for scanning electron microscopy (SEM) analysis (fig. S12). The fibrous morphology of the scaffolds was still visible, and the surrounding tissue was well integrated with the scaffold. By week 3 postsurgery, notable wound size reduction was observed across all groups. The Blank, Pristine scaffold, and PCM (SI) groups still showed wound areas of 14.1 ± 1.3%, 9.8 ± 1.1%, and 3.7 ± 1.0%, respectively. For the PCM-P (SI) and PCM-P (MI) groups involving PDGF-BB, the SI group had a residual wound area of 2.8 ± 0.7%, while the MI group showed complete wound closure, indicating that multiple rounds of PDGF-BB delivery were more effective than a one-time delivery. Notably, the wounds in the three MI groups were fully healed, and the PCM-P/V/E (MI) group not only achieved a complete wound closure but also exhibited smooth, intact regenerated skin resembling healthy tissue, indicating a scarless healing process. By week 4, complete tissue coverage was achieved in all groups, with the multicue scaffold showing substantially enhanced regenerative outcomes. Hematoxylin and eosin (H&E) staining was performed on the regenerated skin tissue 4 weeks postsurgery to evaluate the morphology of the epidermis, dermis, and appendages ([118]Fig. 4D). In the Blank group, the epidermis was incomplete. The Pristine scaffold group demonstrated complete epidermal regeneration, although focal erosion was still evident. In the PCM (SI) group, an epidermal covering had formed, but partial separation between the epidermal and dermal layers was observed. In contrast, the four groups with growth factor delivery—PCM-P (SI), PCM-P (MI), PCM-P/E (MI), PCM-P/V/E (MI)—exhibited a four-layered epidermis, including stratum corneum, granular layer, spinous layer, and basal layer. Notably, in the MI groups, the epidermis was intact, and, beneath the epidermis, organized tissue structures were observed. The PCM-P/V/E (MI) group exhibited the most compact and well-ordered tissue arrangement. Dermal regeneration and appendage formation were next evaluated. In the Blank group, inflammatory cells were still present, and few appendages were observed. The Pristine scaffold group showed abundant proliferating fibrous connective tissue. The regenerated dermis in the PCM (SI) group was loosely arranged, reflecting incomplete tissue regeneration. In the PCM-P (SI) and PCM-P (MI) groups, the dermis was more organized, and a higher number of appendages were regenerated. The most prominent results were observed in the PCM-P/E (MI) and PCM-P/V/E (MI) groups, where the dermis was highly organized, and numerous skin appendages were present. These groups achieved not only structural integrity but also restored skin function. Collagen types I and III are essential for maintaining the structural integrity, strength, and elasticity of skin. In healthy tissue, the type I/III collagen ratio typically exceeds 3:1, but this ratio decreases following injury ([119]50). Type III collagen initially forms a flexible matrix for cell migration and proliferation, later transitioning to type I collagen during tissue remodeling to provide structural support ([120]51). Therefore, maintaining an optimal balance of collagen types is essential for effective wound healing. Sirius red staining was used to detect collagen types I (red) and III (green) in the regenerated tissue ([121]Fig. 4E). A quantitative analysis of the collagen I to III ratio is shown in fig. S13. For the PCM-P(SI) group, the ratio was 2.62 ± 0.15, 1.2 times higher than that in the Pristine scaffold group, suggesting that the delivery of PDGF-BB was beneficial to regulating collagen deposition and transition. The PCM-P/V/E (MI) group exhibited the largest collagen fibers with more extended morphology, and the ratio of collagen I to III was 3.89 ± 0.24, which was consistent with healthy skin, indicating well-organized collagen and accelerated tissue remodeling. To further investigate the regenerative mechanisms, we performed immunohistochemical and immunofluorescence staining ([122]Fig. 5). Because vascular regeneration is the first step in skin wound healing, VEGF-B and CD34 staining were used to assess the efficacy of the scaffolds on vascularization. As shown in [123]Fig. 5 (A and G), the relative expressions of VEGF-B in the PCM-P (SI), PCM-P (MI), PCM-P/E (MI), and PCM-P/V/E (MI) groups were 1.2, 1.5, 1.7, and 1.9 times higher, respectively, compared to the Pristine scaffold group. Similar results were obtained by quantifying neovessels through CD34 immunofluorescence staining, where the numbers of vessels per unit area in the PCM-P (SI), PCM-P (MI), PCM-P/E (MI), and PCM-P/V/E (MI) groups were 2.0, 2.8, 2.8, and 4.0 times, respectively, greater than that in the Pristine scaffold group ([124]Fig. 5, B and H, and fig. S14). Additionally, the PCM-P/V/E (MI) group exhibited a 1.4-fold increase in neovascularization compared to the PCM-P/E (MI) group, demonstrating that the incorporation and phase-specific release of VEGF notably enhanced angiogenesis. To evaluate cell proliferation, KI-67 staining was performed. The relative expressions of KI-67 in the PCM-P (SI), PCM-P (MI), PCM-P/E (MI), and PCM-P/V/E (MI) groups were 1.3, 1.6, 1.9, and 2.2 times, respectively, higher than that in the Pristine scaffold group ([125]Fig. 5, C and I). Enhanced proliferation in these groups likely contributed to improved tissue regeneration. Epidermal regeneration was further assessed by CK-4 immunohistochemical analysis. As shown in the immunohistochemical images ([126]Fig. 5D), the group receiving spatiotemporally controlled delivery of multiple growth factors exhibited a marked increase in epidermal thickness. The relative expressions of CK-4 in the PCM-P (SI), PCM-P (MI), PCM-P/E (MI), and PCM-P/V/E (MI) groups were 1.4, 2.3, 2.8, and 3.5 times, respectively, higher than that in the Pristine scaffold group ([127]Fig. 5J). Notably, the relative expression of CK-4 in the PCM-P/E (MI) group was 1.2 times higher than that in the PCM-P (MI) group, indicating that the sustained release of EGF provided a targeted enhancement of epidermal regeneration. Last, we assessed skin tissue remodeling by evaluating collagen deposition and transformation. The relative expressions of collagen III in the PCM-P (SI), PCM-P (MI), PCM-P/E (MI), and PCM-P/V/E (MI) groups were 87.2, 79.5, 74.4, and 56.4% of that in the Pristine scaffold group, respectively, while the relative expressions of collagen I were 1.3, 1.4, 1.5, and 1.6 times higher in these groups ([128]Fig. 5, E, F, K, and L, and fig. S15). These findings are consistent with the Sirius Red staining results. Upon masked NIR irradiation to trigger the on-demand release of multiple growth factors, the nanofiber scaffold promoted vascular regeneration, cell proliferation, collagen deposition, and skin tissue remodeling. Fig. 5. Immunohistochemical and immunofluorescence staining analysis of regenerated rabbit skin tissues in different treatment groups. [129]Fig. 5. [130]Open in a new tab (A) Immunohistochemical staining of VEGF-B. (B) Immunofluorescence staining of CD34. Immunohistochemical staining of (C) KI-67, (D) CK-4, (E) collagen III, and (F) collagen I, respectively. (G) Quantification of relative average OD of VEGF-B. (H) Quantification of blood vessel density. Quantification of relative average OD of (I) KI-67, (J) CK-4, (K) collagen III, and (L) collagen I, respectively. *P < 0.05, **P < 0.01, ***P < 0.001, and ****P < 0.0001. The scaffold promotes porcine full-thickness skin wound healing upon NIR irradiation To further evaluate the therapeutic efficacy of the scaffolds, a large-animal model in porcine were established on the basis of the findings from small animal experiments. This allowed for a more accurate representation of the human physiological environment, thus increasing the translational relevance of the results. Full-thickness skin wounds, 2.5 cm in diameter and 5 mm in thickness, were created on the backs of pigs, and various scaffolds were applied (fig. S16). NIR irradiation was performed for 10 min every 2 weeks using photomasks ([131]Fig. 6A). To reduce the anesthesia duration, several NIR lasers were administered simultaneously to multiple wounds on each pig, with temperature changes monitored using an infrared thermal camera ([132]Fig. 6B). In all groups subjected to NIR irradiation, the temperature of the scaffolds increased from body temperature to the phase transition temperature of PCM within 1.5 min. During the subsequent 10 min of continuous irradiation, the temperature remained stable around 39° to 43°C ([133]Fig. 6C and fig. S17), and this elevated temperature was maintained across all four rounds of irradiation, demonstrating the long-term photothermal stability of the scaffold ([134]Fig. 6C and fig. S18). Fig. 6. In vivo evaluation of porcine full-thickness skin wound healing under NIR irradiation. [135]Fig. 6. [136]Open in a new tab (A) Schematic illustration of the masked NIR irradiation protocol applied at weeks 0, 2, 4, and 6 postsurgery. (B) Photograph showing simultaneous NIR laser application to multiple wound sites. (C) Infrared thermal images showing the temperature of the scaffold increasing from baseline and stabilizing within the range of 39° to 43°C during continuous irradiation. (D) Representative macroscopic images of wound healing in different groups over 8 weeks. (E) Pie charts depicting the percentage of remaining wound area in each group. (F) H&E staining and (G) Masson’s trichrome staining micrographs of regenerated skin tissue in different groups at 8 weeks postsurgery, shown at varying magnifications. I and II indicate the epidermal and dermal regions, respectively. Wound images were captured at 0, 2, 4, 6, and 8 weeks postsurgery, and the wound areas were quantitatively measured to assess healing rates. As shown in [137]Fig. 6 (D and E) and fig. S19, by week 2, wounds in the Blank, Pristine scaffold, and PCM (SI) groups exhibited limited contraction. In contrast, the PCM-P (SI), PCM-P (MI), PCM-P/E (MI), and PCM-P/V/E (MI) groups showed significant wound contraction, with remaining wound areas of 69.0 ± 3.6%, 73.9 ± 1.7%, 70.7 ± 2.2%, and 66.1 ± 1.4%, respectively. These groups demonstrated healing rates of 3.6, 3.0, 3.4, and 3.9 times higher than that of the Pristine scaffold group, due to the delivery of growth factor at this stage. By the fourth week, significant reductions in wound surface area were observed in the growth factor delivery groups. Specifically, the PCM-P/V/E(MI) group had a wound area of 52.1 ± 3.2%, reflecting the synergistic action of PDGF-BB, VEGF, and EGF in promoting skin repair. Weeks 4 to 6 marked a critical phase of wound healing, coinciding with the third round of NIR-triggered growth factor release. At the sixth week, wounds in all groups had significantly decreased in size, especially in the MI groups. Compared to the PCM-P (SI) group, which had a wound area of 31.2%, the wound area in the PCM-P (MI) group was reduced to 22.2%, demonstrating the efficacy of multiple cycles of PDGF-BB delivery. The wound healing rates for the PCM-P (MI), PCM-P/E (MI), and PCM-P/V/E (MI) groups were 1.4, 1.5, and 1.6 times higher than the Pristine scaffold group, respectively. Notably, in the PCM-P/V/E (MI) group, the remaining wound area was just 8.8 ± 3.2%, with no open wound bed observed, indicating sustained, phase-specific delivery of growth factors during this peak regenerative window played a pivotal role in accelerating wound closure and contributing to the improved final repair outcomes. As a comparison, in a recent study, a polythiourea-based polyurethane foam scaffold was compared with two commercial products, NovoSorb BTM and Integra, for the repair of porcine full-thickness skin wounds (2 cm by 1 cm) ([138]52). By day 31, the remaining unhealed wound area was ~50% (~1 cm^2) in the NovoSorb BTM group and about 43% (~0.9 cm^2) in both the polyurethane foam scaffold and Integra groups. In our porcine model, the surface area of the skin defects was 2.45 times larger than what was used in that study. By weeks 4 and 6, the wounds treated with PCM-P/V/E (MI) reached 47.9% (~2.4 cm^2) and 91.2% (~4.5 cm^2) of repair, respectively. Compared to NovoSorb BTM, the polyurethane foam scaffold, and Integra at 31 days, the repair efficiency of PCM-P/V/E (MI) was approximately increased by 1.4-, 1.2-, and 1.2-fold at week 4 and by 3.5-, 3.1- and 3.1-fold at week 6, respectively. These results underscore the effectiveness of spatiotemporal regulation of the injury microenvironment in accelerating wound healing. By the eighth week, wounds in the PCM-P (MI), PCM-P/E (MI), and PCM-P/V/E (MI) groups were reduced to less than 10%, while wounds in the Blank and Pristine scaffold groups remained at 32.9 ± 5.1% and 26.3 ± 4.4%, respectively. Notably, the PCM-P/V/E (MI) group exhibited complete wound healing, demonstrating that the sustained, on-demand delivery of multiple growth factors at different stages notably accelerated wound healing. Pathological analysis of regenerated skin tissue at 8 weeks postsurgery was performed using H&E staining ([139]Fig. 6F). In the Blank and Pristine scaffold groups, epidermal coverage was incomplete, with signs of crusting and erosion. The PCM (SI) group showed a better healing, but the epidermal structure remained irregular. Complete epidermal regeneration with notable thickness was observed in the PCM-P (SI) group, supported by PDGF-BB delivery. Upon masked NIR irradiation, the PCM-P (MI), PCM-P/E (MI), and PCM-P/V/E (MI) groups exhibited complete, well-organized epidermal structures. Among them, the PCM-P/V/E (MI) group showed the most organized dermal fibrous tissue, closely resembling the morphology of normal pig skin. Dermal regeneration was also evaluated. In the Blank group, the dermis was loosely arranged with immature collagen. The Pristine scaffold, PCM (SI), and PCM-P (SI) groups showed denser dermis, although less organized. In the groups subjected to MI, the presence of skin appendages indicated not only structural regeneration but also functional recovery. Notably, the PCM-P/E (MI) and PCM-P/V/E (MI) groups exhibited well-organized, compact collagen fibers in the dermis, with the PCM-P/V/E (MI) group closely resembling normal tissue. Masson staining was used to assess overall collagen deposition in the regenerated skin tissue ([140]Fig. 6G). The Blank group showed minimal collagen deposition, while the Pristine scaffold and PCM (SI) groups had increased collagen content, although the fibers were loosely and irregularly arranged. The PCM-P (SI) group exhibited higher collagen content with more orderly fiber arrangement. Upon MI, collagen secretion was significantly increased, and the newly formed collagen fibers became more organized and thicker. In the PCM-P/V/E (MI) group, collagen content, fiber organization, and thickness were comparable to those found in normal tissue. To further evaluate key aspects of skin wound healing, immunofluorescence staining of various markers was performed ([141]Fig. 7). CD31 immunofluorescence staining was used to visualize the number of newly formed blood vessels. As shown in [142]Fig. 7A and fig. S20, the numbers of vessels per unit area in the PCM-P (SI), PCM-P (MI), PCM-P/E (MI), and PCM-P/V/E (MI) groups were 2.5, 3.4, 3.3, and 5.4 times, respectively, higher than that in the Pristine scaffold group ([143]Fig. 7E). Cytokeratin-pan immunofluorescence staining was performed to assess epidermal regeneration ([144]Fig. 7B and fig. S21). The epidermal thickness and fluorescence intensity were progressively greater in the following order: Blank group, Pristine scaffold, PCM (SI), PCM-P (SI), PCM-P (MI), PCM-P/E (MI), and PCM-P/V/E (MI). The relative expression of cytokeratin-pan in the PCM-P/V/E (MI) group was 3.3 times higher than that in the Pristine scaffold group ([145]Fig. 7F). In evaluating the dermal regeneration, vimentin immunofluorescence staining was used ([146]Fig. 7C and fig. S22). The relative expressions of vimentin in the PCM-P (SI), PCM-P (MI), PCM-P/E (MI), and PCM-P/V/E (MI) groups were 1.2, 1.9, 2.0, and 3.4 times, respectively, higher than that in the Pristine scaffold group ([147]Fig. 7G). Dual immunofluorescence staining of collagen I and collagen III was performed to examine tissue remodeling ([148]Fig. 7D). The ratio of collagen type I to III in the PCM-P/V/E (MI) group was 3.17 ± 0.16, which was within the typical range found in healthy skin tissue and 1.4 times higher than that in the Pristine scaffold group ([149]Fig. 7H) ([150]53). These results collectively demonstrate that the nanofiber scaffold, combined with masked NIR irradiation therapy, could spatiotemporally trigger the on-demand delivery of PDGF-BB, VEGF, and EGF. This sequential and synergistic effect promoted vascular regeneration, epidermal formation, dermal regeneration, and, ultimately, tissue remodeling, emphasizing the potential of this approach to accelerate skin wound healing. Fig. 7. Immunofluorescence staining analysis of regenerated porcine skin tissues in different treatment groups. [151]Fig. 7. [152]Open in a new tab Representative images showing the expression of (A) CD31, (B) cytokeratin-pan, (C) vimentin, and (D) collagen I and collagen III, respectively. (E) Quantification of blood vessel density. Relative average OD of (F) cytokeratin-pan and (G) vimentin, respectively. (H) Ratio of collagen I to collagen III. *P < 0.05, **P < 0.01, ***P < 0.001, and ****P < 0.0001. The scaffold promotes porcine skin wound healing by comprehensively modulating the microenvironment To investigate the activation of signaling pathways and gene regulation, transcriptomic sequencing was conducted on regenerated porcine skin tissues from the Blank and PCM-P/V/E (MI) groups. Principal components analysis ([153]Fig. 8A) and hierarchical clustering heatmap ([154]Fig. 8B) revealed distinct transcriptional profiles between the two groups. The volcano plot ([155]Fig. 8C) demonstrated a large number of differentially expressed genes, while the Venn diagram ([156]Fig. 8D) showed 1881 coexpressed genes shared between the Blank and PCM-P/V/E (MI) groups. As show in [157]Fig. 8E, Gene Ontology enrichment analysis revealed significant alterations in genes associated with biological processes including cell adhesion, migration, ECM remodeling, and positive regulation of inflammatory response. In the cellular component category, notable changes were observed in extracellular space, cell membrane, extracellular region, exosome, cell surface components, and ECM, indicating the role of the scaffold in modulating cellular behaviors and promoting tissue remodeling. [158]Figure 8F shows the Kyoto Encyclopedia of Genes and Genomes pathway enrichment analysis, highlighting that the scaffold primarily promoted skin wound healing by regulating key signaling pathways, including phosphatidylinositol 3-kinase (PI3K)–Akt, cytokine-cytokine receptor interaction, and mitogen-activated protein kinase (MAPK). These pathways are crucial for tissue regeneration and work together to enhance the healing process. The activation of the cytokine-cytokine receptor interaction pathway indicates that the delivery of growth factors such as PDGF-BB, VEGF, and EGF enhanced cell migration, blood vessel formation, and epidermal regeneration, all of which are critical for effective wound healing. Additionally, pathways related to chemokine signaling, cell adhesion, and hematopoietic cell lineage were activated. Moreover, the incorporation of PCM in the scaffold activated fatty acid metabolism, further aiding tissue regeneration. Fig. 8. Transcriptome sequencing analysis of regenerated porcine skin tissues from the Blank and PCM-P/V/E (MI) groups. [159]Fig. 8. [160]Open in a new tab (A) Principal components analysis (PCA) revealing distinct gene expression patterns between the two groups. (B) Hierarchical clustering heatmap showing significantly up-regulated and down-regulated genes after PCM-P/V/E (MI) treatment (fold change > 1.5, P < 0.05). (C) Volcano plot indicating the distribution of differentially expressed genes. (D) Venn diagram displaying the overlap of expressed genes between the two groups. (E) Gene Ontology term enrichment analysis for differentially expressed genes. BP, biological process; CC, cellular component; MF, molecular function. (F) Kyoto Encyclopedia of Genes and Genomes pathway enrichment analysis showing the top 10 significantly enriched signaling pathways. (G) Heatmap and (H) protein-protein interaction (PPI) network of key genes involved in the PI3K-Akt signaling pathway (fold change ≥ 2, P < 0.05). (I) Heatmap and (J) PPI network of differentially expressed genes involved in the MAPK signaling pathway (fold change ≥ 2, P < 0.05). Heatmaps of genes in the (K) nuclear factor κB (NF-κB) and (L) AMPK and Hedgehog signaling pathways, respectively (fold change > 1.5, P < 0.05). (M) Schematic illustration summarizing the proposed mechanism of scaffold-assisted therapeutic modulation to promote coordinated skin wound healing. A detailed analysis of the PI3K-Akt and MAPK signaling pathways revealed specific changes in gene expression. As show in [161]Fig. 8G, in the PI3K-Akt pathway, inflammation-related genes such as NFKB1, IL6, OSMR, and JAK3 were significantly down-regulated, suggesting that the sustained delivery of growth factors by the scaffold promoted tissue regeneration while suppressing the inflammatory response. Conversely, genes associated with metabolism (e.g., GYS2, CREB3L4, and FGF21) and cell survival, migration, and proliferation (e.g., AKT2, BCL2, FGF10, EFNA4, EGFR, and ERBB3) were significantly up-regulated. Additionally, genes involved in angiogenesis (e.g., EFNA1) and nerve regeneration (e.g., NGFR, NTF4, and EFNA5) were also up-regulated. These results indicate that the scaffold facilitated vascular regeneration and the formation of neural networks, contributing to the restoration of skin function. Protein-protein interaction (PPI) networks were constructed on the basis of the differentially expressed genes ([162]Fig. 8H), revealing that key proteins such as AKT2, EGFR, and ERBB3—central regulators of cell survival, migration, and proliferation—were highly interconnected, underscoring their critical roles in coordinating skin wound healing. In the MAPK signaling pathway ([163]Fig. 8I), genes related to immune response and apoptosis—such as TAOK2, MAX, STK4, and RELB—were down-regulated, indicating that the scaffold helped suppress inflammation and inhibit apoptosis. Conversely, genes involved in cell migration, proliferation, and tissue morphogenesis (RAC3, AKT2, EFNA3, FGF5, and DUSP8), as well as lipid metabolism (PLA2G4B), were up-regulated. Notably, nerve regeneration-associated genes including NGRF, PPP3CC, and EFNA4, were significantly up-regulated, indicating active neurogenesis alongside skin tissue regeneration, an essential aspect for restoring the sensory function of skin. The PPI network of MAPK-related genes ([164]Fig. 8J) further confirmed their central roles in orchestrating skin wound healing. In addition to the PI3K-Akt and MAPK signaling pathways, several other pathways were activated by the scaffold. Inflammation-related pathways, including nuclear factor κB (NF-κB), tumor necrosis factor (TNF), and chemokine signaling, exhibited widespread down-regulation of gene expression, particularly in proinflammatory genes, indicating an anti-inflammatory effect ([165]Fig. 8K and figs. S23 to S25). In contrast, the Hedgehog signaling pathway was significantly up-regulated, supporting tissue patterning and scarless skin regeneration, including hair follicle neogenesis (fig. S26) ([166]54). Several metabolism-related pathways were also activated, such as the adenosine 5′-monophosphate-activated protein kinase (AMPK) pathway and the alanine, aspartate, and glutamate metabolism pathway (figs. S27 and S28). Notably, the peroxisome signaling pathway, which is involved in cellular detoxification and lipid metabolism, was up-regulated, suggesting enhanced fatty acid metabolism, reduced oxidative stress, and promotion of skin cell proliferation (fig. S29). Collectively, gene expression changes in the Hedgehog and AMPK pathways suggest that modulation of cellular metabolism enhanced keratinocyte proliferation, fibroblast activity, and overall tissue remodeling, thereby facilitating effective skin wound healing ([167]Fig. 8L). Together, the scaffold, integrating a radially aligned topology, NIR light irradiation, and spatiotemporally delivered growth factors, promoted skin wound healing and functional recovery by establishing a favorable regenerative microenvironment ([168]Fig. 8M). It synergistically down-regulated genes associated with inflammation and apoptosis while up-regulating those involved in angiogenesis, nerve regeneration, cell survival, migration, proliferation, and ECM remodeling. These effects were mediated through the activation and cross-regulation of key signaling pathways, including PI3K-Akt, MAPK, NF-κB, TNF, Hedgehog, and AMPK, thereby comprehensively enhancing the wound healing process. DISCUSSION The skin, as the body’s largest organ and first line of defense, plays a crucial role in various physiological functions. Although the skin has innate regenerative capabilities, extensive wounds often require artificial therapeutic interventions to accelerate healing. Clinical products like allografts, xenografts, and synthetic matrices have advanced wound care ([169]8). Allografts, such as Apligraf and Dermagraft, offer bioactivity that reduces healing time but face challenges with production and storage due to the presence of live cells. Xenografts, like Integra and Oasis Wound Matrix, address these issues but may cause immune rejection. Synthetic polymer–based scaffolds, such as Restrata, Hyalomatrix, and NovoSorb BTM, offer nonimmunogenicity and biocompatibility but lack bioactivity, limiting their ability to modulate the healing process. With the improved understanding of the mechanisms underlying skin wound healing, scaffolds that precisely control biophysical and biochemical cues are increasingly recognized, which requires the dynamic manipulation of key healing stages, such as inflammation, cell proliferation, and tissue remodeling, to create a microenvironment that supports effective skin wound healing ([170]55). Our study presents an innovative bioactive scaffold that incorporates multiple guidance cues and leverages NIR irradiation for the spatiotemporally controlled delivery of specific growth factors, thereby actively regulating each stage of the wound healing process. The scaffold features a hierarchical structure, with radially aligned nanofibers facilitating directed cell migration and a randomly oriented outer layer providing mechanical support. Constructed from FDA-approved materials through scalable electrospinning and electrospraying techniques, the scaffold holds strong potential for clinical translation. This approach directly addresses the limitations of current therapeutic options. In contrast to existing synthetic matrices, the scaffold integrates topographical cues, a tunable photothermal effect, and sustained biochemical signaling to establish a dynamic microenvironment tailored to the distinct phases of wound healing. A key innovation is its capacity for on-demand, localized delivery of growth factors in response to NIR irradiation. The photothermal response, enabled by ICG-encapsulated phase-change microparticles, maintains temperatures above 40°C under clinical conditions. A photomask allows for spatially and temporally precise release of PDGF-BB, VEGF, and EGF, synchronized with specific phases of the healing process, which is a great challenge. In vitro, the released factors (107.45 ± 0.75 ng of PDGF-BB, 59.02 ± 1.81 ng of VEGF, and 64.15 ± 0.85 ng of EGF) significantly enhance angiogenesis and cell migration and proliferation. Furthermore, the synergistic effect of topographical and biochemical cues facilitates radial migration of L929, NIH-3T3, and HaCaT cells, improving cellular distribution across the wound bed. In a rabbit full-thickness wound model with a diameter of 2.5 cm, our scaffold combined with photomask-assisted NIR irradiation achieves an impressive healing rate of 95% (~4.7 cm^2) within 2 weeks, notably outperforming conventional scaffolds. Notably, the wounds heal without visible scarring, and the newly regenerated skin is indistinguishable from native tissue. Histological analysis confirms organized collagen deposition and a collagen I/III ratio comparable to that of healthy skin, reflecting the capacity of the scaffold to dynamically modulate the regenerative microenvironment. This modulation effectively supports vascular regeneration, epidermal proliferation, and dermal remodeling. Compared with studies using ECM-mimetic coatings from natural sources, our scaffold demonstrates a 5.7-fold improvement in healing efficiency, highlighting its transformative potential in wound care ([171]56). Moreover, the controlled release of growth factors mitigates risks associated with unregulated delivery, such as aberrant tissue formation and potential carcinogenesis. Porcine models, which closely resemble human skin in terms of physiology and wound healing dynamics, further validate the translational potential of our scaffold. In full-thickness wounds (2.5 cm in diameter, 5 mm in depth), the scaffold achieves 47.9% healing by the fourth week, corresponding to a wound closure area of 2.4 cm^2. In comparison, a recent study using a reactive oxygen species-responsive polyurethane foam for similar wounds (2-cm length by 1-cm width), equivalent to 40.8% of the wound surface area in our model, reports 57% closure after 31 days, with a surface area of ~1.1 cm^2 ([172]52). In addition to outperforming this experimental material, our scaffold exhibits faster and more complete healing than commercially available products such as NovoSorb BTM and Restrata ([173]12). By the eighth week, wounds treated with our scaffold achieve near-complete closure. Unlike conventional treatments that often result in fibrotic scarring, our scaffold supports regeneration characterized by restored skin architecture and function. Histological analysis of regenerated tissue reveals a higher density of neovasculature, increased epidermal thickness, enhanced dermal matrix formation, and a collagen type I/III ratio within the typical range observed in healthy skin. To elucidate the underlying mechanisms, we conducted in vivo transcriptomic sequencing, which reveals that the scaffold activates key regenerative signaling pathways, including PI3K-Akt and MAPK, while also modulating immune responses and metabolic reprogramming. Genes involved in angiogenesis, neurogenesis, cell migration, proliferation, and cellular metabolism are significantly up-regulated, whereas those associated with apoptosis and inflammation are down-regulated. Notably, this coordinated molecular response enables the simultaneous initiation of angiogenesis and neurogenesis within the same ECM microenvironment. Neovascularization not only enhances nutrient and oxygen delivery but also creates a permissive niche for neural network development, further supporting functional restoration. Therefore, the favorable outcome stems from the scaffold’s capacity to dynamically modulate the wound microenvironment. The early release of PDGF-BB and VEGF effectively suppresses acute inflammation and initiates robust angiogenesis, laying the groundwork for regeneration. As healing progresses, the sustained release of PDGF-BB and EGF, in conjunction with the scaffold’s radially aligned fibers and fatty acid–rich matrix, promotes cell migration, proliferation, and tissue remodeling. Concurrently, fatty acid metabolism is regulated to further support cellular energy demands during tissue repair. By establishing a sequentially optimized and spatially structured microenvironment, the scaffold orchestrates a comprehensive series of regenerative events, ranging from vascularization and immune regulation to neural integration and collagen remodeling. As a result, it not only accelerates wound closure but also ensures the regeneration of functionally and structurally complete skin tissue, with minimal risk of adverse effects. The proposed scaffold provides a comprehensive solution to several persistent challenges in wound care by integrating photothermal-responsive technology with an advanced, adaptable scaffold design. Constructed from FDA-approved and clinically validated materials, the scaffold is compatible with scalable manufacturing techniques, which lowers regulatory hurdles and reduces production costs, enhancing its potential for broad clinical adoption. Its activation via a handheld NIR light emitter allows for convenient, noninvasive treatment under medical guidance, making it especially well-suited for outpatient care and resource-limited settings. By dynamically responding to the wound healing process, the scaffold minimizes risks such as chronic inflammation and excessive scarring, thereby promoting sustained tissue regeneration and long-term recovery. Its customizable structure supports use across a diverse range of wound types and sizes, from acute injuries to complex chronic wounds. The scaffold’s capacity to sustain and precisely regulate growth factor delivery addresses a critical need in managing chronic wounds, including diabetic ulcers and burn injuries. While the proposed scaffold shows great potential, future modifications may further enhance its suitability for clinical applications. One area of improvement lies in the precise regulation of NIR-triggered growth factor release, which depends on accurately monitoring the wound microenvironment to identify the factors needed at specific healing stages, enabling timely and targeted activation. Another important consideration is thermal control, as excessive local heating may compromise tissue viability and therapeutic consistency. To address this, future strategies could focus on developing smart, feedback-controlled NIR systems that integrate real-time thermal sensors to automatically adjust irradiation parameters. In parallel, PCMs could be further engineered with thermally responsive molecular regulators that become optically inactive upon reaching their melting point, thereby limiting further NIR absorption and preventing overheating. Collectively, these advances would contribute to a next-generation therapeutic paradigm for skin wound repair, one that enables precise, spatiotemporal regulation of regenerative cues across all stages of healing. MATERIALS AND METHODS Study design This study aimed to develop a nanofiber scaffold that enables the spatiotemporally controlled delivery of multiple growth factors, specifically designed to address the unique requirements of each phase in skin tissue regeneration for effective wound repair. The scaffold’s morphology was examined via SEM. Key properties such as chemical composition, mechanical strength, and photothermal performance were thoroughly evaluated. To validate the NIR-triggered controlled release system, an in vitro release simulation was set up to observe color changes corresponding to the release of differently colored dyes. The quantities of released growth factors were quantified using enzyme-linked immunosorbent assay kits, and their bioactivity was assessed. Various cell types relevant to skin tissue regeneration, including HUVECs, L929 fibroblasts, NIH-3T3 fibroblasts, and HaCaT cells, were used as in vitro models to assess the scaffold’s ability to regulate vascularization, promote cell proliferation, and enhance cell migration. For in vivo validation, full-thickness skin wound models were established in both small animals (rabbits) and large animals (pigs) to evaluate the efficacy of the scaffold combined with NIR irradiation in promoting skin wound healing. The rabbit skin wound repair experiment was approved by the Animal Ethics and Welfare Committee of Institute of Radiation Medicine, Chinese Academy of Medical Sciences and followed ethical guidelines (IRM-DWLL-2021210). The porcine skin wound repair experiment was approved by the Animal Ethics and Welfare Committee of Peking University People’s Hospital and followed ethical guidelines (2025PHE013). During these experiments, photomasks of adjustable sizes and regular NIR light interventions were applied, and photographs were taken to track wound healing progress. After the wound healing process, regenerated skin tissues were harvested and subjected to H&E staining, Masson’s trichrome staining, Sirius red staining, and a series of immunohistochemistry and immunofluorescence assays to assess key regenerative factors, including vascularization, cell proliferation, epidermal regeneration, dermal regeneration, and collagen deposition. To investigate the underlying molecular mechanisms, transcriptomic gene sequencing was performed on the regenerated skin tissue from pigs to identify pathway changes and gene expression regulation during skin repair. Statistical analysis All experiments were performed with at least three replicates, and data were expressed as means ± SD. Statistical analysis was conducted using Origin software and GraphPad Prism 8.0 software. Differences between two groups were analyzed using a t test. Comparisons among more than two groups were performed using one-way analysis of variance (ANOVA). A P value of less than 0.05 was considered statistically significant (*P < 0.05, **P < 0.01, ***P < 0.001, and ****P < 0.0001). Acknowledgments