| Home | E-Submission | Sitemap | Contact Us |  
top_img
Algae > Volume 40(3); 2025 > Article
Yu, Sung, Kim, and Kim: Bioactive nanovesicles derived from the red alga Pyropia yezoensis for wound healing

ABSTRACT

Nanovesicles (NVs) derived from Pyropia yezoensis, a red alga extensively cultivated in East Asian coastal regions, are emerging as a promising therapeutic tool for promoting wound healing. This study aimed to evaluate the physiological properties and wound healing efficacy of Pyropia yezoensis-derived nanovesicles (PyNVs). Using various cell culture models, we analyzed the effects of PyNVs on skin cell proliferation, migration, and angiogenesis. Nanoparticle tracking analysis revealed that PyNVs are stable NVs with an average diameter of approximately 140 nm. At a high concentration (7.5 × 109 particles mL−1), PyNVs significantly enhanced angiogenesis, promoted the migration and proliferation of skin cells, and accelerated the wound healing process. Furthermore, PyNVs modulated the expression of extracellular matrix-related genes, increasing collagen synthesis while suppressing excessive matrix metalloproteinase-1 activity. These findings suggest that PyNVs possess the necessary characteristics to serve as a natural, cost-effective, and scalable therapeutic agent for wound healing. The study highlights the potential of PyNVs as a sustainable source of bioactive substances that could transform wound care and tissue regeneration applications.

INTRODUCTION

The skin, as the body’s outermost organ, serves as a vital barrier against external threats, including chemical agents, microorganisms, and environmental stressors (Sparr et al. 2013, Nguyen and Soulika 2019). However, this protective barrier can be compromised by various factors such as climate change, environmental pollution, surgical procedures, trauma, and diabetic complications, leading to both acute and chronic skin damage (Parrado et al. 2019, Kim et al. 2021). The process of wound healing involves a complex cascade of biological events orchestrated through multiple cellular interactions, encompassing distinct yet overlapping phases of inflammation, proliferation, and remodeling (Gurtner et al. 2008, Wilkinson and Hardman 2020). With more than one billion people worldwide affected by acute and chronic skin conditions, the development of effective therapeutic approaches for wound healing represents a significant challenge in global healthcare (Garraud et al. 2017, Graves et al. 2022).
Animal-derived nanovesicles (ADNVs), including exosomes, have emerged as crucial mediators of intercellular communication and potential therapeutic agents in tissue repair. These nanoscale, lipid bilayer-enclosed particles, typically smaller than 200 nm, are naturally secreted by virtually all cell types (Yáñez-Mó et al. 2015, Minutti et al. 2017). Extracellular vesicles (EVs) serve as sophisticated delivery vehicles for diverse bioactive molecules, including nucleic acids, proteins, lipids, and metabolites, protecting their cargo from degradation while facilitating targeted delivery to recipient cells (Pegtel et al. 2010, Abels and Breakefield 2016). Their fundamental roles in various physiological processes, including stem cell maintenance, tissue repair, immune regulation, and blood coagulation, have positioned EVs as promising candidates for therapeutic applications (Robbins and Morelli 2014, Maas et al. 2017).
However, the clinical translation of ADNVs faces significant challenges, including limited scalability, high production costs, potential immunogenicity, and concerns regarding pathogen transmission (Dad et al. 2021, Li et al. 2023). In contrast, plant-derived nanovesicles (PDNVs) offer several compelling advantages: they can be produced at scale with consistent quality, demonstrate lower toxicity and immunogenicity, exhibit enhanced stability due to their low cholesterol content, and show excellent biocompatibility (You et al. 2021, Kim and Park 2022). Recent studies have highlighted the therapeutic potential of PDNVs derived from various plant sources, including ginger, ginseng, wheat, and grapefruit, particularly in the context of wound healing (Ju et al. 2013, Wang et al. 2014).
Among various marine resources, Pyropia yezoensis is a red alga native to the East Asian Pacific region, particularly along the coasts of Korea, Japan, China, and Russia. It is high-value cultivated plant, the growth and quality of which are significantly influenced by seawater temperature and light quality (He et al. 2023, Zhong et al. 2023, Ma et al. 2024b, Shin et al. 2024). Furthermore, its unique properties make it an excellent candidate for use as a functional food, health supplement, and pharmaceutical ingredient (Harada et al. 1997, Choi et al. 2016, Kim et al. 2023, Jung et al. 2024). With an annual production exceeding half a million tons (Kim et al. 2023), P. yezoensis is especially rich in mycosporine-like amino acids, which provide strong UV protection and antioxidant benefits, making it a promising natural ingredient for sunscreens and anti-aging cosmetics (Jung et al. 2024). This alga is also celebrated for its high protein content and diverse biological activities, including antioxidant, anti-inflammatory, and photoprotective effects (Han et al. 2022, Wang et al. 2024). While significant research has focused on its ability to alleviate UVA-induced oxidative stress and modulate inflammatory responses (Toyosaki and Iwabuchi 2009, Choi et al. 2015, Ha et al. 2020), the therapeutic potential of P. yezoensis-derived nanovesicles (PyNVs) in wound healing remains largely unexplored. This represents a crucial gap in current knowledge, especially considering the documented healing properties inherent to this marine organism.
The present study aims to investigate the therapeutic potential of PyNVs in wound healing through a comprehensive analysis of their biological characteristics and functional effects. We hypothesize that PyNVs inherit the beneficial properties of their source organism while offering the advantages of nano-scale delivery vehicles. Our investigation encompasses the isolation and characterization of PyNVs, evaluation of their effects on key cell types involved in wound healing, and assessment of their impact on critical wound healing processes, including cell proliferation, migration, and extracellular matrix (ECM) remodeling.
This research addresses the growing need for cost-effective, scalable therapeutic approaches in wound healing while potentially offering insights into novel applications of marine-derived nanovesicles (NVs) in regenerative medicine. Understanding the processes through which PyNVs influence wound healing could provide valuable insights for developing more effective therapeutic strategies for treating both acute and chronic wounds.

MATERIALS AND METHODS

Isolation of NVs from Pyropia yezoensis

Pyropia yezoensis were cultured in Provasoli enriched seawater (Provasoli 1968) medium at 15°C under irradiation of 50 μmol photons m−2 s−1 provided by cool-white fluorescent lighting with a 12 h light/12 h dark cycle. Following established protocols for PyNVs isolation (You et al. 2021, Kim and Park 2022), the samples were mixed with phosphate-buffered saline (PBS; pH 8.0) in a 1:1 (w/w) ratio and homogenized using a blender (Model 7011G; Waring Laboratory Science, Torrington, CT, USA) at 22,000 rpm for 5 min at room temperature. The homogenate was centrifuged at 3,000 ×g for 30 min at room temperature to remove large debris, followed by centrifugation at 10,000 ×g for 1 h at room temperature. The supernatant was filtered through a 0.45 μm filter (Sartorius AG, Göttingen, Germany). Final ultracentrifugation was performed at 100,000 ×g for 2 h at room temperature using an Optima XE-90 ultracentrifuge (Beckman Coulter, Brea, CA, USA), according to established EV isolation protocols (Théry et al. 2018). The isolated PyNVs were resuspended in PBS (pH 8.0) and stored at −80°C until further use. The homogenized P. yezoensis extract using a blender was filtered through 0.22 μm and used as a control for the isolated PyNVs. The concentration of P. yezoensis extracts used in the experiments was conducted according to the highest concentration of PyNVs based on protein.
The size distribution and concentration of PyNVs were measured using a NanoSight NS300 (Malvern Panalytical, Worcestershire, UK) equipped with a 488 nm laser. Samples were diluted 1:1,000 in PBS. The instrument was configured with a camera level of 13 and a detection threshold of 4. For each sample, three 60-s videos were recorded at 25°C. Data were analyzed using NTA 3.4 software (Malvern Panalytical), and particle concentrations were corrected for the dilution factor. Concentrations are reported as mean ± standard error of the mean from three technical replicates. Total protein concentration of PyNVs was measured using the Pierce BCA Protein Assay Kit (Thermo Fisher Scientific, Waltham, MA, USA) according to the manufacturer’s instructions. The morphology and structural details of PyNVs were examined through cryo-transmission electron microscopy (Cryo-TEM) using a Glacios microscope (Thermo Fisher Scientific) operated at 200 kV.

Cell culture

Human keratinocyte cells (HaCaT) and human dermal fibroblasts (HDF) were maintained in high-glucose Dulbecco’s modified Eagle medium (DMEM; Gibco, Thermo Fisher Scientific) supplemented with 10% fetal bovine serum (FBS; Gibco) and 1% penicillin-streptomycin-amphotericin mixture (Gibco) at 37°C in a humidified atmosphere containing 5% CO2, as described by Werner et al. (2007). Human umbilical vein endothelial cells (HUVECs) were cultured in endothelial cell growth medium-2 (Lonza, Basel, Switzerland) under the same conditions, following established protocols (DiPietro 2016).

Cell viability and proliferation assays

Cell viability was assessed using established protocols (Zhou et al. 2020). HaCaT cells (3 × 105 cells well−1), HDF cells (4 × 104 cells well−1), and HUVECs (6 × 104 cells well−1) were seeded in 24-well plates. After 24 h, cells were treated with PyNVs at concentrations ranging from 0.5 × 109 to 2.0 × 1010 particles mL−1 in serum-free medium and incubated for 24 h at 37°C. Cell viability was evaluated using the water-soluble tetrazolium salt-1 (WST-1) assay (EZ Cytox; DoGen Bio, Seoul, Korea). The absorbance was measured at 450 nm using a multi-mode microplate reader (PerkinElmer, Waltham, MA, USA) after 2 h of WST-1 reagent incubation, following methods described by Wang et al. (2019).
For proliferation assays, HaCaT cells (6 × 104 cells well−1) and HDF cells (1 × 104 cells well−1) were seeded in 24-well plates and treated with 2% exosome-depleted FBS and PyNVs (0.5–7.5 × 109 particles mL−1), as described by Zhang et al. (2015). Cell proliferation was measured at 12, 24, and 48 h using the WST-1 assay as detailed above.

Wound healing assay

Cell migration was evaluated using a wound healing assay following modified protocols from Cappiello et al. (2018). HaCaT cells (5 × 104 cells well−1) were seeded on silicone inserts (SPLScar Block; SPL Life Sciences, Pocheon, Korea) placed in 6-well plates and incubated at 37°C for 24 h in a 5% CO2 incubator. After cell attachment, the silicone inserts were removed using sterile forceps to create a defined wound area (500 μm width). Cells were washed twice with PBS (pH 8.0) and cultured in fresh medium containing 2% exosome-depleted FBS and PyNVs (0.5–7.5 × 109 particles mL−1). Wound closure was monitored for 48 h using a JuLI Stage live cell imager (NanoEnTek, Seoul, Korea), with images captured every 30 min. The wound area was quantified using ImageJ software (NIH, Bethesda, MD, USA), and the closure rate was calculated as described by Tracy et al. (2016).

Gene expression analysis

Total RNA from HDF cells was isolated using WelPrep Total RNA Isolation Reagent (Welgene, Gyeongsan, Korea) according to the manufacturer’s protocol. RNA purity and concentration were determined using a NanoDrop spectrophotometer (Thermo Fisher Scientific), following methods described by Kim et al. (2017). First-strand cDNA was synthesized from 1 μg of total RNA using the QuantiTect Reverse Transcription Kit (Qiagen, Hilden, Germany). Gene-specific primers were designed to amplify fibronectin, elastin, COL1A1 (collagen type I alpha 1 chain), MMP-1 (matrix metalloproteinase-1), and β-actin. Primer pairs are as follows: fibronectin (5′-ACA ACA CCG AGG TGA CTG AGA C-3′, 5′-GGA CAC AAC GAT GCT TCC TGA G-3′), Elastin (5′-GGT TGT GTC ACC AGA AGC AGC T-3′, 5′-CCG TAA GTA GGA ATG CCT CCA AC-3′), COL1A1 (5′-GAT TCC CTG GAC CTA AAG GTG-3′, 5′-AGC CTC TCC ATC TTT GCC AGC A-3′), MMP-1 (5′-ATG AAG CAG CCC AGA TGT GGA G-3′, 5′-TGG TCC ACA TCT GCT CTT GGC A-3′), and β-actin (5′-CAC TGT GCC CAT CTA CG-3′, 5′-CTT AAT GTC ACG CAC GAT TTC-3′). Quantitative polymerase chain reaction (qPCR) was performed using WizPure qPCR Master Mix (Wizbiosolutions, Seongnam, Korea) on a Rotor-Gene Q 2plex RT-PCR system (Qiagen). The polymerase chain reaction (PCR) conditions were optimized based on previous studies (Lee et al. 2015): initial denaturation at 95°C for 10 min, followed by 40 cycles of 95°C for 15 s and 60°C for 1 min. Melting curve analysis was performed to confirm specific amplification. Gene expression was normalized to β-actin using the 2-ΔΔCT method as described by Livak and Schmittgen (2001).

Protein expression analysis

For enzyme-linked immunosorbent assay (ELISA) analysis, HDF cells were seeded at 1.5 × 104 cells per well in 24-well plates and cultured for 24 h, following protocols adapted from Singh et al. (2023). Cells were then treated with PyNVs (0.5–7.5 × 109 particles mL−1) for 72 h. Culture supernatants were collected and centrifuged at 3,000 ×g for 10 min at 4°C. Collagen type I production was measured using the Procollagen Type I C-peptide (PIP) EIA Kit (Takara Bio, Kusatsu, Japan), and MMP-1 levels were determined using the Human Total MMP-1 DuoSet ELISA Kit (R&D Systems, Minneapolis, MN, USA), as described by Schuster et al. (2023). Standard curves were generated using serial dilutions (0–1,000 ng mL−1) of the provided standards. The absorbance was measured at 450 nm using a microplate reader (PerkinElmer).

In vitro angiogenesis assay

Angiogenic capacity was evaluated using a tube formation assay following methods established by DiPietro (2016) and Morbidelli et al. (2021). Growth factor-reduced Matrigel (Corning, Corning, NY, USA) was thawed overnight at 4°C and applied to 48-well plates (150 μL well−1) at 4°C. The plates were incubated at 37°C for 1 h to allow polymerization. HUVECs (5 × 104 cells well−1) were seeded onto the Matrigel layer and treated with PyNVs (1.0–7.5 × 109 particles mL−1). After 16 h of incubation, tube formation was observed using an inverted microscope (Olympus, Tokyo, Japan), and five random fields per well were photographed. Tube formation parameters (total tube length, number of nodes, meshes, and branches) were quantified using the Angiogenesis Analyzer plugin for ImageJ software, as described by Akhtari et al. (2024).

Sodium dodecyl sulfate polyacrylamide gel electrophoresis and bioinformatics analysis

To analyze the protein composition of nanovesicles (NVs) isolated from P. yezoensis, samples were collected at each step of the NV isolation process and subjected to sodium dodecyl sulfate polyacrylamide gel electrophoresis (SDS-PAGE). A total of 20 μg of each sample was mixed with 4× Laemmli sample buffer and heated at 95°C for 5 min. The samples were then loaded onto a 12% SDS-polyacrylamide gel and electrophoresed at 120 V for 90 min. After electrophoresis, the gel was stained with Coomassie brilliant blue R-250 and destained using a methanol:acetic acid:water (4:1:5, v/v/v) solution. A molecular weight marker (Precision plus protein; Bio-Rad, Hercules, CA, USA) was used to estimate the molecular weight of the protein bands. In-gel digestion with trypsin and liquid chromatography-tandem mass spectrometry (LC-MS/MS) analysis were analyzed using an LTQ Orbitrap XL mass spectrometer (Thermo Fisher Scientific). The resulting tandem mass spectra were searched using the Mascot search engine (version 2.7.0; Matrix Science, London, UK) for protein identification.

Statistical analysis

All experiments were performed in triplicate and repeated at least three times independently, following established statistical procedures for biological studies (Potekaev et al. 2021). Data are presented as mean ± standard deviation. Statistical analyses were performed using GraphPad Prism 6.0 software (GraphPad Software Inc., San Diego, CA, USA). Normal distribution was confirmed using the Shapiro-Wilk test as recommended by Tottoli et al. (2020). Differences between groups were analyzed using two-way analysis of variance (ANOVA) followed by Tukey’s post hoc test. p-values were considered statistically significant at three levels: *p < 0.05, **p < 0.01, and ***p < 0.001.

RESULTS

Isolation and characterization of PyNVs

P. yezoensis cultivated under optimized conditions was used to isolate NVs using differential ultracentrifugation method (Fig. 1A). Nanoparticle tracking analysis revealed that the isolated PyNVs exhibited a size distribution ranging from 95 to 195 nm, with an average diameter of 140.1 nm (Fig. 1B). The mean particle concentration was 2.77 × 1011 ± 4.71 × 109 particles mL−1. Cryo-TEM analysis confirmed the presence of spherical vesicles with intact lipid bilayer structure, approximately 110 nm in diameter (Fig. 1C). The production yield of PyNVs, determined by dividing the number of isolated PyNVs by the weight of P. yezoensis used, was 2.29 × 1011 particles g−1 of P. yezoensis (Fig. 1D). The purity of isolated PyNVs was validated by a particle-to-protein ratio of 1.43 × 108 particles μg−1, which falls within the acceptable range for high-quality EV preparations (Fig. 1E). The protein composition of PyNVs was analyzed through SDS-PAGE and proteomic profiling. SDS-PAGE was performed on protein fractions collected during the NV isolation procedure. As shown in Supplementary Fig. S1A, the purified PyNVs fraction (lane 4) exhibited multiple distinct protein bands ranging approximately from 50 to 250 kDa, indicating the successful enrichment of protein-containing NVs. To further characterize the PyNVs protein cargo, LC-MS/MS analysis was conducted. A total of 232 proteins were identified and annotated (Supplementary Table S1). These proteins were classified into six major categories based on their functional roles and known associations with EVs, as summarized in Supplementary Fig. S1B. A total of 232 proteins were categorized into six groups: enzymes (76 proteins), cytosolic proteins (4 proteins), membrane proteins (8 proteins), heat shock proteins (5 proteins), cytoskeletal proteins (5 proteins), and others (134 proteins).

Evaluation of PyNVs safety profile

To assess the potential cytotoxicity of PyNVs, three cell types critical to wound healing—HaCaT, HDF, and HUVECs—were exposed to increasing concentrations (1 × 108 to 2 × 1010 particles mL−1) of PyNVs for 24 h. WST-1 assay results demonstrated that PyNVs and P. yezoensis extracts (140 μg mL−1) exhibited no significant cytotoxicity across the tested concentration range in all three cell types (Fig. 2), supporting their safety for therapeutic applications.

PyNVs enhance angiogenic response in HUVECs

Angiogenesis represents a critical early phase of wound healing, providing essential oxygen and nutrients while promoting cell survival. Using an in vitro tube formation assay with HUVECs, we evaluated the angiogenic potential of PyNVs. Angiogenic activity was enhanced in a dose-dependent manner in the PyNV-treated group, and treatment with 7.5 × 109 particles mL−1 of PyNV significantly enhanced angiogenic activity (Fig. 3A). Quantitative analysis revealed significant increases in total tube length (Fig. 3B), number of nodes (Fig. 3C), mesh formation (Fig. 3D), and branch points (Fig. 3E) compared to untreated controls. However, P. yezoensis extracts (50 μg mL−1) did not affect angiogenic activity even when used at high concentrations.

PyNVs promote cell proliferation and migration

To investigate the regenerative potential of PyNVs, we examined their effects on cell proliferation and migration. Both HaCaT and HDF cells treated with PyNVs (5 × 108 to 7.5 × 109 particles mL−1) showed dose-dependent increases in proliferation. The most pronounced effect was observed at 7.5 × 109 particles mL−1, with significant enhancement in proliferation of both cells at 48 h (Fig. 4A & B).
In the wound healing assay, HaCaT cells treated with higher concentrations of PyNVs (1 × 109 and 7.5 × 109 particles mL−1) demonstrated markedly enhanced migration capabilities. The wound closure rates reached 96.9 and 97.6%, respectively at these concentrations within 12 h, significantly surpassing the control group’s 76.9% closure rate (Fig. 4C). In contrast, high concentration of P. yezoensis extract (50 μg mL−1) did not affect skin cell proliferation or migration compared to the control. These findings suggest that PyNVs effectively promote both proliferation and directional migration of skin cells.

PyNVs modulate ECM-related gene and protein expression

To elucidate the molecular mechanisms underlying PyNVs-mediated wound healing, we analyzed the expression of key ECM-related genes. Real-time PCR analysis revealed that high concentrations of PyNVs (1 × 109 and 7.5 × 109 particles mL−1) significantly upregulated multiple ECM genes. At 7.5 × 109 particles mL−1, we observed remarkable increases in expression: fibronectin (3.5-fold) (Fig. 5A), elastin (8.5-fold) (Fig. 5B), COL1A1 (10-fold) (Fig. 5C), and MMP-1 (2.6-fold) (Fig. 5D).
To validate these findings at the protein level, we examined the secretion of collagen and MMP-1 in HDF cells treated with various concentrations of PyNVs. ELISA analysis demonstrated a dose-dependent increase in collagen secretion (Fig. 5E), while MMP-1 levels remained stable (Fig. 5F). Importantly, the collagen/MMP-1 ratio increased approximately 1.6-fold in the high-concentration group compared to controls (Fig. 5G), indicating that PyNVs promote ECM production while maintaining balanced matrix remodeling.

DISCUSSION

The skin serves as a vital barrier protecting the human body from external threats, including environmental factors and pathogens (Walker 2022). Disruption of this protective barrier by various factors such as ultraviolet radiation, environmental pollution, or physical trauma can trigger inflammatory responses, leading to both acute and chronic wound conditions (Jiao et al. 2024). Impaired wound healing not only causes physical discomfort but also significantly impacts patients’ psychological well-being and quality of life (Bechert and Abraham 2009, Razjouyan et al. 2017). This has driven increasing interest in developing effective therapeutic strategies using bioactive materials for skin regeneration and wound healing.
Current wound healing agents, including antioxidants, anti-inflammatory compounds, growth factors, cytokines, and various NVs, demonstrate certain limitations in clinical applications (Barrientos et al. 2014, Merjaneh et al. 2017). Synthetic agents often present challenges such as skin incompatibility, cytotoxicity, and high production costs (Halim et al. 2010, van der Veen et al. 2010). Animal-derived products raise concerns regarding immunogenicity and pathogen transmission. In contrast, naturally derived bioactive substances, particularly from marine sources, often demonstrate superior biocompatibility, lower production costs, and reduced immunogenicity, making them attractive alternatives for skin therapeutic applications (Ma et al. 2024a).
P. yezoensis, our chosen source material, contains an impressive array of bioactive compounds. Beyond its rich nutritional profile of vitamins (A, B1, B2, C, and E) and minerals (calcium, potassium, magnesium, phosphorus), it possesses well-documented therapeutic properties (Watanabe et al. 2000, El-Beltagi et al. 2022). These include antioxidant activities through free radical scavenging, anti-inflammatory effects via cytokine modulation, and direct wound healing promotion through enhanced collagen synthesis and matrix remodeling (Kim et al. 2014, Isaka et al. 2015). The combination of these properties makes P. yezoensis an ideal candidate for therapeutic NVs isolation.
Our characterization of PyNVs revealed several technical advantages over conventional PDNVs. The isolation method yielded vesicles with consistent physical properties (average diameter 140.1 nm) and a well-defined lipid bilayer structure, aligning with established standards for small PDNVs (Théry et al. 2018). High particle yield (2.29 × 1011 particles g−1) and purity (1.43 × 108 particles μg−1) suggest an efficient and scalable isolation process, providing practical advantages and benefits for therapeutic applications of PDNVs.
The molecular mechanisms underlying wound healing involve complex, coordinated processes (Fig. 6). Our findings demonstrate that PyNVs positively influence multiple aspects of this cascade. In the early phase, PyNVs significantly enhanced angiogenesis in HUVECs at 7.5 × 109 particles mL−1, promoting the formation of new blood vessels essential for tissue regeneration (DiPietro 2016, Morbidelli et al. 2021). This pro-angiogenic effect likely facilitates oxygen and nutrient delivery to the wound site, supporting the metabolic demands of regenerating tissue.
During the proliferative phase, the coordinated activities of keratinocytes and fibroblasts drive wound closure through cell migration and ECM synthesis (Tracy et al. 2016, Plikus et al. 2021). Our molecular analyses revealed that PyNVs significantly upregulated key ECM components in a dose-dependent manner. The dramatic increases in fibronectin (3.5-fold), elastin (8.5-fold), and type I collagen (10-fold) expression suggest that PyNVs actively modulate the wound microenvironment. Particularly noteworthy is the balanced regulation of ECM remodeling, where increased collagen production occurs without excessive MMP-1 activation, potentially preventing aberrant scarring.
The remodeling phase involves the transformation of granulation tissue into mature scar tissue, characterized by the replacement of type III collagen with type I collagen and the reorganization of ECM components (Velnar et al. 2009, Potekaev et al. 2021). Our results indicate that PyNVs maintain an optimal balance between ECM production and degradation, as evidenced by the increased collagen/MMP-1 ratio. This balanced remodeling is crucial for restoring tissue tensile strength while preventing excessive scarring.
The exceptional safety profile of PyNVs, demonstrated by the absence of cytotoxicity even at high concentrations (2 × 1010 particles mL−1), combined with its effects on angiogenesis and ECM modulation, positions them as promising candidates for clinical applications. However, several aspects warrant further investigation. Future studies should focus on the validation of PyNVs’ therapeutic efficacy through various in vivo wound models to bridge the gap between laboratory findings and clinical applications. The development of optimal delivery systems will be crucial to enhance tissue penetration and bioavailability of PyNVs in the wound environment. Additionally, comprehensive proteomics and transcriptomics analyses are needed to elucidate the precise molecular mechanisms underlying PyNVs’ therapeutic effects. Of particular interest is the potential application of PyNVs in treating chronic wounds and other skin pathologies, which could significantly expand their therapeutic utility. Furthermore, optimization of large-scale production processes will be essential to maintain consistent quality while meeting commercial demands.
In conclusion, this study demonstrates that NVs from Pyropia yezoensis are a promising, naturally-derived therapeutic tool for wound healing. PyNVs enhance angiogenesis, cell proliferation, migration, and ECM remodeling while maintaining a strong safety profile, making them suitable for clinical development. With P. yezoensis already cultivated on a large scale—exceeding half a million tons annually—and widely consumed as a functional food (Kim et al. 2023), it presents a cost-effective resource for scalable PyNVs production. The existing cultivation infrastructure and its recognized safety further support its use in developing therapeutics and cosmetics (Jung et al. 2024). Utilizing established aquaculture practices could lower production costs and expedite commercialization. As research advances, PyNVs may offer a sustainable alternative to current wound healing therapies, transforming the field of regenerative medicine.

Notes

ACKNOWLEDGEMENTS

This work was supported by the Korea Health Technology R&D Project through the Korea Health Industry Development Institute (KHIDI), funded by the Ministry of Health & Welfare, Republic of Korea (RS-2023-KH135936). It was also supported by the Korea Institute of Marine Science & Technology Promotion (KIMST), funded by the Ministry of Oceans and Fisheries, Republic of Korea (RS-2025-02303933), and by a research grant from Kongju National University in 2022.

CONFLICTS OF INTEREST

The authors declare that they have no potential conflicts of interest.

SUPPLEMENTARY MATERIALS

Supplementary Table S1
Protein profile of PyNVs (https://e-algae.org).
algae-2025-40-8-6-Supplementary-Table-S1.xlsx
Supplementary Fig. S1
Proteomic analysis of PyNVs (https://e-algae.org).
algae-2025-40-8-6-Supplementary-Fig-S1.pdf

Fig. 1
Isolation and characterization of Pyropia yezoensis-derived nanovesicles (PyNVs). (A) Schematic diagram of the optimized PyNVs isolation protocol using differential ultracentrifugation. (B) Size distribution profile analyzed by nanoparticle tracking analysis (NTA), indicating a major peak between 95–195 nm with an average diameter of 140.1 nm. (C) Overviews and higher magnification (white square) images from cryo-transmission electron microscopy of morphology of freshly isolated PyNVs. High-magnification images show the spherical shape and intact lipid bilayer structure of PyNVs. (D) Production yield of PyNVs calculated by dividing the total number of isolated PyNVs by P. yezoensis mass. (E) Assessment of PyNVs quality, demonstrating a particle-to-protein ratio of 1.11 × 108 particles μg−1, indicative of high purity. Scale bars represent: C, 1 μm (overview); 100 nm (white square).
algae-2025-40-8-6f1.jpg
Fig. 2
Safety profile assessment of Pyropia yezoensis-derived nanovesicles (PyNVs). Cell viability of HaCaT (A), human dermal fibroblasts (HDF) (B), and human umbilical vein endothelial cell (HUVEC) cells (C) after 24 h of exposure to P. yezoensis extracts (140 μg mL−1) and PyNVs (1 × 109 to 2 × 1010 particles mL−1) was measured using the water-soluble tetrazolium salt-1 assay. Results are shown as the percentage of viability relative to untreated controls (mean ± standard deviation from three independent experiments). No significant cytotoxicity was observed for either the high-concentration extract or PyNVs.
algae-2025-40-8-6f2.jpg
Fig. 3
Pyropia yezoensis-derived nanovesicles (PyNVs) enhance angiogenesis in human umbilical vein endothelial cells (HUVECs). Representative images of tube formation in HUVECs treated with P. yezoensis extracts (50 μg mL−1) and PyNVs (A). Quantitative analysis of angiogenic parameters: total tube length (B), number of nodes (C), mesh formation (D), and branch points (E). Data are presented as mean ± standard deviation from three independent experiments. *p < 0.05, **p < 0.01, ***p < 0.001 compared to control, indicating significant enhancement in angiogenic activity.
algae-2025-40-8-6f3.jpg
Fig. 4
Pyropia yezoensis-derived nanovesicles (PyNVs) promote cell proliferation and wound healing. Cell proliferation kinetics in HaCaT cells (A) and human dermal fibroblasts (HDF) cells (B) treated with P. yezoensis extracts (140 μg mL−1) and PyNVs (0.5 to 7.5 × 109 particles mL−1) over 48 h, as measured by the water-soluble tetrazolium salt-1 assay. Scratch wound healing assay showing quantitative analysis of wound closure in HaCaT cells treated with increasing concentrations of PyNVs, demonstrating accelerated healing (C). P. yezoensis extract did not affect cell proliferation or migration. Data are shown as mean ± standard deviation from three independent experiments. *p < 0.05, **p < 0.01, ***p < 0.001 compared to control.
algae-2025-40-8-6f4.jpg
Fig. 5
Modulation of extracellular matrix (ECM)-related gene and protein expression by Pyropia yezoensis-derived nanovesicles (PyNVs). Relative mRNA expression levels of fibronectin (A), elastin (B), COL1A1 (collagen type I alpha 1 chain) (C), and MMP-1 (matrix metalloproteinase-1) (D) were normalized to β-actin in human dermal fibroblasts (HDF) cells after PyNVs treatment. Protein levels measured by enzyme-linked immunosorbent assay: type I collagen secretion (E), MMP-1 expression (F), and collagen/MMP-1 ratio (G) in HDF cells. Data represent mean ± standard deviation from three independent experiments. *p < 0.05, **p < 0.01, ***p < 0.001 compared to control, indicating significant modulation of ECM components.
algae-2025-40-8-6f5.jpg
Fig. 6
Mechanistic overview of Pyropia yezoensis-derived nanovesicles (PyNVs)-mediated wound healing. Schematic illustration summarizing the mechanisms by which PyNVs promote wound healing. PyNVs enhance angiogenesis, stimulate cell proliferation and migration, and modulate extracellular matrix (ECM) protein expression, contributing to improved tissue regeneration. This highlights the therapeutic potential of PyNVs for wound healing applications.
algae-2025-40-8-6f6.jpg

REFERENCES

Abels, E. R. & Breakefield, X. O. 2016. Introduction to extracellular vesicles: biogenesis, RNA cargo selection, content, release, and uptake. Cell. Mol. Neurobiol. 36:301–312. doi.org/10.1007/s10571-016-0366-z
crossref pmid pmc pdf
Akhtari, N., Ahmadi, M., Kiani Doust Vaghe, Y., et al. 2024. Natural agents as wound-healing promoters. Inflammopharmacology. 32:101–125. doi.org/10.1007/s10787-023-01318-6
crossref pmid pdf
Barrientos, S., Brem, H., Stojadinovic, O. & Tomic-Canic, M. 2014. Clinical application of growth factors and cytokines in wound healing. Wound Repair Regen. 22:569–578. doi.org/10.1111/wrr.12205
crossref pmid pmc
Bechert, K. & Abraham, S. E. 2009. Pain management and wound care. J. Am. Coll. Certif Wound Spec. 1:65–71. doi.org/10.1016/j.jcws.2008.12.001
crossref pmid pmc
Cappiello, F., Casciaro, B. & Mangoni, M. L. 2018. A novel in vitro wound healing assay to evaluate cell migration. J. Vis. Exp. 133:e56825. doi.org/10.3791/56825

Choi, J.-W., Kim, I.-H., Kim, Y.-M., Lee, M.-K. & Nam, T.-J. 2016. Pyropia yezoensis glycoprotein regulates antioxidant status and prevents hepatotoxicity in a rat model of D-galactosamine/lipopolysaccharide-induced acute liver failure. Mol. Med. Rep. 13:3110–3114. doi.org/10.3892/mmr.2016.4932
crossref pmid
Choi, J.-W., Kim, Y.-M., Park, S.-J., Kim, I.-H. & Nam, T.-J. 2015. Protective effect of Porphyra yezoensis glycoprotein on D-galactosamine-;induced cytotoxicity in Hepa 1c1c7 cells. Mol. Med. Rep. 11:3914–3919. doi.org/10.3892/mmr.2015.3244
crossref pmid
Dad, H. A., Gu, T.-W., Zhu, A.-Q., Huang, L.-Q. & Peng, L.-H. 2021. Plant exosome-like nanovesicles: emerging therapeutics and drug delivery nanoplatforms. Mol. Ther. 29:13–31. doi.org/10.1016/j.ymthe.2020.11.030
crossref pmid pmc
DiPietro, L. A. 2016. Angiogenesis and wound repair: when enough is enough. J. Leukoc. Biol. 100:979–984. doi.org/10.1189/jlb.4MR0316-102R
crossref pmid pmc pdf
El-Beltagi, H. S., Mohamed, A. A., Mohamed, H. I., Ramadan, K. M. A., Barqawi, A. A. & Mansour, A. T. 2022. Phytochemical and potential properties of seaweeds and their recent applications: a review. Mar. Drugs. 20:342. doi.org/10.3390/md20060342
crossref pmid pmc
Garraud, O., Hozzein, W. N. & Badr, G. 2017. Wound healing: time to look for intelligent, ‘natural’ immunological approaches? BMC Immunol. 18:23. doi.org/10.1186/s12865-017-0207-y
crossref pmid pmc pdf
Graves, N., Phillips, C. J. & Harding, K. 2022. A narrative review of the epidemiology and economics of chronic wounds. Br. J. Dermatol. 187:141–148. doi.org/10.1111/bjd.20692
crossref pmid pdf
Gurtner, G. C., Werner, S., Barrandon, Y. & Longaker, M. T. 2008. Wound repair and regeneration. Nature. 453:314–321. doi.org/10.1038/nature07039
crossref pmid pdf
Ha, Y., Lee, W.-H., Jeong, J., et al. 2020. Pyropia yezoensis extract suppresses IFN-gamma- and TNF-alpha-induced proinflammatory chemokine production in HaCaT cells via the down-regulation of NF-kappaB. Nutrients. 12:1238. doi.org/10.3390/nu12051238
crossref pmid pmc
Halim, A. S., Khoo, T. L. & Mohd Yussof, S. J. 2010. Biologic and synthetic skin substitutes: an overview. Indian J. Plast Surg. 43(Suppl):S23–S28. doi.org/10.4103/0970-0358.70712
crossref pmid pmc
Han, S., Park, J.-S., Umanzor, S., Yarish, C. & Kim, J. K. 2022. Effects of extraction methods for a new source of biostimulant from Sargassum horneri on the growth of economically important red algae, Neopyropia yezoensis. Sci. Rep. 12:11878. doi.org/10.1038/s41598-022-16197-0
crossref pmid pmc pdf
Harada, H., Noro, T. & Kamei, Y. 1997. Selective antitumor activity in vitro from marine algae from Japan coasts. Biol. Pharm. Bull. 20:541–546. doi.org/10.1248/bpb.20.541
crossref pmid
He, B., Zheng, Z., Niu, J., Xie, X. & Wang, G. 2023. Transcriptome analysis revealed regulatory mechanisms of light and culture density on free-living sporangial filaments of Neopyropia yezoensis (Rhodophyta). Algae. 38:283–294. doi.org/10.4490/algae.2023.38.5.25
crossref pdf
Isaka, S., Cho, K., Nakazono, S., et al. 2015. Antioxidant and anti-inflammatory activities of porphyran isolated from discolored nori (Porphyra yezoensis). Int. J. Biol. Macromol. 74:68–75. doi.org/10.1016/j.ijbiomac.2014.11.043
crossref pmid
Jiao, Q., Zhi, L., You, B., Wang, G., Wu, N. & Jia, Y. 2024. Skin homeostasis: mechanism and influencing factors. J. Cosmet. Dermatol. 23:1518–1526. doi.org/10.1111/jocd.16155
crossref pmid
Ju, S., Mu, J., Dokland, T., et al. 2013. Grape exosome-like nanoparticles induce intestinal stem cells and protect mice from DSS-induced colitis. Mol. Ther. 21:1345–1357. doi.org/10.1038/mt.2013.64
crossref pmid pmc
Jung, S., Lim, J.-M., Min, S.-R., Kim, G. H. & Jeong, W.-J. 2024. Increased shinorine production through the introduction of mycosporine-like amino acids biosynthetic genes from Pyropia yezoensis and Nostoc punctiforme into Nannochloropsis gaditana. Plant Biotechnol. Rep. 18:871–880. doi.org/10.1007/s11816-024-00938-w
crossref pdf
Kim, B. E., Kim, J., Goleva, E., et al. 2021. Particulate matter causes skin barrier dysfunction. JCI Insight. 6:e145185. doi.org/10.1172/jci.insight.145185
crossref pmid pmc
Kim, C.-R., Kim, Y.-M., Lee, M.-K., Kim, I.-H., Choi, Y.-H. & Nam, T.-J. 2017. Pyropia yezoensis peptide promotes collagen synthesis by activating the TGF-β/Smad signaling pathway in the human dermal fibroblast cell line Hs27. Int. J. Mol. Med. 39:31–38. doi.org/10.3892/ijmm.2016.2807
crossref pmid pmc
Kim, M. & Park, J. H. 2022. Isolation of Aloe saponaria-derived extracellular vesicles and investigation of their potential for chronic wound healing. Pharmaceutics. 14:1905. doi.org/10.3390/pharmaceutics14091905
crossref pmid pmc
Kim, S., You, D. H., Han, T. & Choi, E.-M. 2014. Modulation of viability and apoptosis of UVB-exposed human keratinocyte HaCaT cells by aqueous methanol extract of laver (Porphyra yezoensis). J. Photochem. Photobiol. B. 141:301–307. doi.org/10.1016/j.jphotobiol.2014.10.012
crossref pmid
Kim, Y. T., Kim, R.-W., Shim, E., Park, H., Klochkova, T. A. & Kim, G. H. 2023. Control of oomycete pathogens during Pyropia farming and processing using calcium propionate. Algae. 38:71–80. doi.org/10.4490/algae.2023.38.3.8
crossref pdf
Lee, H.-A., Kim, I.-H. & Nam, T.-J. 2015. Bioactive peptide from Pyropia yezoensis and its anti-inflammatory activities. Int. J. Mol. Med. 36:1701–1706. doi.org/10.3892/ijmm.2015.2386
crossref pmid
Li, A., Li, D., Gu, Y., et al. 2023. Plant-derived nanovesicles: further exploration of biomedical function and application potential. Acta Pharm. Sin. B. 13:3300–3320. doi.org/10.1016/j.apsb.2022.12.022
crossref pmid pmc
Livak, K. J. & Schmittgen, T. D. 2001. Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) method. Methods. 25:402–408. doi.org/10.1006/meth.2001.1262
crossref pmid
Ma, J., Fang, Y., Yu, H., et al. 2024a. Recent advances in living algae seeding wound dressing: focusing on diabetic chronic wound healing. Adv. Funct. Mater. 34:2308387. doi.org/10.1002/adfm.202308387
crossref
Ma, L.-H., Tian, L., Wang, Y.-Q., Xie, C.-Y. & Du, G.-Y. 2024b. Physiological and transcriptome analysis of acclimatory response to cold stress in marine red alga Pyropia yezoensis. Algae. 39:17–30. doi.org/10.4490/algae.2024.39.3.7
crossref pdf
Maas, S. L. N., Breakefield, X. O. & Weaver, A. M. 2017. Extracellular vesicles: unique intercellular delivery vehicles. Trends Cell Biol. 27:172–188. doi.org/10.1016/j.tcb.2016.11.003
crossref pmid pmc
Merjaneh, M., Langlois, A., Larochelle, S., Cloutier, C. B., Ricard-Blum, S. & Moulin, V. J. 2017. Pro-angiogenic capacities of microvesicles produced by skin wound myofibroblasts. Angiogenesis. 20:385–398. doi.org/10.1007/s10456-017-9554-9
crossref pmid pdf
Minutti, C. M., Knipper, J. A., Allen, J. E. & Zaiss, D. M. W. 2017. Tissue-specific contribution of macrophages to wound healing. Semin. Cell Dev. Biol. 61:3–11. doi.org/10.1016/j.semcdb.2016.08.006
crossref pmid
Morbidelli, L., Genah, S. & Cialdai, F. 2021. Effect of microgravity on endothelial cell function, angiogenesis, and vessel remodeling during wound healing. Front. Bioeng. Biotechnol. 9:720091. doi.org/10.3389/fbioe.2021.720091
crossref pmid pmc
Nguyen, A. V. & Soulika, A. M. 2019. The dynamics of the skin’s immune system. Int. J. Mol. Sci. 20:1811. doi.org/10.3390/ijms20081811
crossref pmid pmc
Parrado, C., Mercado-Saenz, S., Perez-Davo, A., Gilaberte, Y., Gonzalez, S. & Juarranz, A. 2019. Environmental stressors on skin aging: mechanistic insights. Front. Pharmacol. 10:759. doi.org/10.3389/fphar.2019.00759
crossref pmid pmc
Pegtel, D. M., Cosmopoulos, K., Thorley-Lawson, D. A., et al. 2010. Functional delivery of viral miRNAs via exosomes. Proc. Natl. Acad. Sci. U. S. A. 107:6328–6333. doi.org/10.1073/pnas.0914843107
crossref pmid pmc
Plikus, M. V., Wang, X., Sinha, S., et al. 2021. Fibroblasts: origins, definitions, and functions in health and disease. Cell. 184:3852–3872. doi.org/10.1016/j.cell.2021.06.024
crossref pmid pmc
Potekaev, N. N., Borzykh, O. B., Medvedev, G. V., et al. 2021. The role of extracellular matrix in skin wound healing. J. Clin. Med. 10:5947. doi.org/10.3390/jcm10245947
crossref pmid pmc
Provasoli, L. 1968. Media and prospects for the cultivation of algae. In : In : Watanabe A., Hattori A., editors Cultures and Collections of Algae. In: Proceedings of the US-Japan Conference, Japanese Society of Plant Physiology Press; Hakone. 63–75.

Razjouyan, J., Grewal, G. S., Talal, T. K., Armstrong, D. G., Mills, J. L. & Najafi, B. 2017. Does physiological stress slow down wound healing in patients with diabetes? J. Diabetes Sci. Technol. 11:685–692. doi.org/10.1177/1932296817705397
crossref pmid pmc pdf
Robbins, P. D. & Morelli, A. E. 2014. Regulation of immune responses by extracellular vesicles. Nat. Rev. Immunol. 14:195–208. doi.org/10.1038/nri3622
crossref pmid pmc pdf
Schuster, R., Younesi, F., Ezzo, M. & Hinz, B. 2023. The role of myofibroblasts in physiological and pathological tissue repair. Cold Spring Harb. Perspect. Biol. 15:a041231. doi.org/10.1101/cshperspect.a041231
crossref pmid pmc
Shin, S. K., Xing, Q., Park, J.-S., Yarish, C., Kong, F. & Kim, J. K. 2024. Effects of biostimulants, AMPEP and Kelpak on the growth and asexual reproduction of Pyropia yezoensis (Bangiales, Rhodophyta) at different temperatures. Algae. 39:31–41. doi.org/10.4490/algae.2024.39.3.11
crossref pdf
Singh, D., Rai, V. & Agrawal, D. K. 2023. Regulation of collagen I and collagen III in tissue injury and regeneration. Cardiol. Cardiovasc. Med. 7:5–16. doi.org/10.26502/fccm.92920302
crossref pmid pmc
Sparr, E., Millecamps, D., Isoir, M., Burnier, V., Larsson, A. & Cabane, B. 2013. Controlling the hydration of the skin though the application of occluding barrier creams. J. R. Soc. Interface. 10:20120788. doi.org/10.1098/rsif.2012.0788
crossref pmid pmc pdf
Théry, C., Witwer, K. W., Aikawa, E., et al. 2018. Minimal information for studies of extracellular vesicles 2018 (MISEV2018): a position statement of the International Society for Extracellular Vesicles and update of the MISEV2014 guidelines. J. Extracell. Vesicles. 7:1535750. doi.org/10.1080/20013078.2018.1535750
pmid pmc
Tottoli, E. M., Dorati, R., Genta, I., Chiesa, E., Pisani, S. & Conti, B. 2020. Skin wound healing process and new emerging technologies for skin wound care and regeneration. Pharmaceutics. 12:735. doi.org/10.3390/pharmaceutics12080735
crossref pmid pmc
Toyosaki, T. & Iwabuchi, M. 2009. New antioxidant protein in seaweed (Porphyra yezoensis Ueda). Int. J. Food Sci. Nutr. 60(Suppl 2):46–56. doi.org/10.1080/09637480802345591
crossref pmid
Tracy, L. E., Minasian, R. A. & Caterson, E. J. 2016. Extracellular matrix and dermal fibroblast function in the healing wound. Adv. Wound Care (New Rochelle). 5:119–136. doi.org/10.1089/wound.2014.0561
crossref pmid pmc
van der Veen, V. C., van der Wal, M. B. A., van Leeuwen, M. C. E., Ulrich, M. M. W. & Middelkoop, E. 2010. Biological background of dermal substitutes. Burns. 36:305–321. doi.org/10.1016/j.burns.2009.07.012
crossref pmid
Velnar, T., Bailey, T. & Smrkolj, V. 2009. The wound healing process: an overview of the cellular and molecular mechanisms. J. Int. Med. Res. 37:1528–1542. doi.org/10.1177/147323000903700531
crossref pmid pdf
Walker, M. 2022. Human skin through the ages. Int. J. Pharm. 622:121850. doi.org/10.1016/j.ijpharm.2022.121850
crossref pmid
Wang, B., Zhuang, X., Deng, Z.-B., et al. 2014. Targeted drug delivery to intestinal macrophages by bioactive nanovesicles released from grapefruit. Mol. Ther. 22:522–534. doi.org/10.1038/mt.2013.190
crossref pmid pmc
Wang, H., Luan, F., Shi, Y., et al. 2024. Extraction, structural features, and pharmacological effects of the polysaccharides from Porphyra yezoensis: a review. Int. J. Biol. Macromol. 279(Pt 1):134745. doi.org/10.1016/j.ijbiomac.2024.134745
crossref pmid
Wang, T., Zheng, S.-L., Liu, L. & Voglmeir, J. 2019. Development of a colorimetric PNGase activity assay. Carbohydr. Res. 472:58–64. doi.org/10.1016/j.carres.2018.11.007
crossref pmid
Watanabe, F., Takenaka, S., Katsura, H., et al. 2000. Characterization of a vitamin B12 compound in the edible purple laver, Porphyra yezoensis. Biosci. Biotechnol. Biochem. 64:2712–2715. doi.org/10.1271/bbb.64.2712
crossref pmid
Werner, S., Krieg, T. & Smola, H. 2007. Keratinocyte-fibroblast interactions in wound healing. J. Invest. Dermatol. 127:998–1008. doi.org/10.1038/sj.jid.5700786
crossref pmid
Wilkinson, H. N. & Hardman, M. J. 2020. Wound healing: cellular mechanisms and pathological outcomes. Open Biol. 10:200223. doi.org/10.1098/rsob.200223
crossref pmid pmc pdf
Yáñez-Mó, M., Siljander, P. R.-M., Andreu, Z., et al. 2015. Biological properties of extracellular vesicles and their physiological functions. J. Extracell. Vesicles. 4:27066. doi.org/10.3402/jev.v4.27066
pmid
You, J. Y., Kang, S. J. & Rhee, W. J. 2021. Isolation of cabbage exosome-like nanovesicles and investigation of their biological activities in human cells. Bioact. Mater. 6:4321–4332. doi.org/10.1016/j.bioactmat.2021.04.023
crossref pmid pmc
Zhang, J., Guan, J., Niu, X., et al. 2015. Exosomes released from human induced pluripotent stem cells-derived MSCs facilitate cutaneous wound healing by promoting collagen synthesis and angiogenesis. J. Transl. Med. 13:49. doi.org/10.1186/s12967-015-0417-0
crossref pmid pmc pdf
Zhong, X., Che, S., Xie, C., et al. 2023. Physiological response of red macroalgae Pyropia yezoensis (Bangiales, Rhodophyta) to light quality: a short-term adaptation. Algae. 38:141–150. doi.org/10.4490/algae.2023.38.5.25
crossref pdf
Zhou, X., Brown, B. A., Siegel, A. P., et al. 2020. Exosome-mediated crosstalk between keratinocytes and macrophages in cutaneous wound healing. ACS Nano. 14:12732–12748. doi.org/10.1021/acsnano.0c03064
pmid pmc
Editorial Office
[14348] A-1716, Gwangmyeong Trade Center, 72 Iljik-ro Gwangmyeong-si. Gyeonggi-do, Korea
Tel: +82-2-899-5980  Fax: +82-2-899-5922    E-mail: editalgae@gmail.com
About |  Browse Articles |  Current Issue |  For Authors and Reviewers
Copyright © The Korean Society of Phycology.                 Developed in M2PI