Skip to main content

Advertisement

ADVERTISEMENT

Peer Review

Peer Reviewed

Review

Diving Deep Into Healing: The Promising Role of Fish Skin in Wound Recovery

August 2024
1943-2704
Wounds. 2024;36(8):274-280. doi:10.25270/wnds/23134
© 2024 HMP Global. All Rights Reserved.
Any views and opinions expressed are those of the author(s) and/or participants and do not necessarily reflect the views, policy, or position of Wounds or HMP Global, their employees, and affiliates.

Abstract

Fish skin has emerged as a potential candidate for improving wound healing due to its notable results in human trials, in which it has been directly applied as a dressing on wounds. The current review explores the mechanisms by which fish skin can boost the wound healing process. The natural wound healing process involves inflammation at the wound site to initiate tissue repair. The body balances this inflammation through interleukin signaling, and imbalances can cause chronic wounds or scarring. The wound site also secretes epidermal growth factor, which activates the Ras/Raf/MEK/ERK and PI3K/Akt pathways. These pathways promote angiogenesis (ie, replacing injured blood vessels) and epithelialization (ie, replacing injured skin). Delays in these pathways increase the healing time. The rich contents of omega-3, collagen, and selenium in fish skin boost wound healing by inhibiting compounds that can cause over-inflammation during interleukin signaling. They also upregulate the Ras/Raf/MEK/ERK and PI3K pathways by altering lipid composition (via omega-3), binding with collagen receptors (via collagen), and modulating selenoproteins (via selenium). The mechanisms discussed in this review support the finding that fish skin is a promising candidate with a strong potential to naturally boost the wound healing process in clinical settings. Continued investigation into the application of fish skin as a practical and commercial wound healing agent is warranted. Future study of additional wound healing properties of fish skin, such as microbial protection of open wounds, is recommended.

Abbreviations

5-LOX, 5-lipoxygenase; AA, arachidonic acid; COX-2, cyclooxygenase-2; DDR, discoidin domain receptor; DHA, docosahexaenoic acid; ECM, extracellular matrix; EGF, epidermal growth factor; EGFR, EGF receptor; EPA, eicosapentaenoic acid; GPx, glutathione peroxidase; IL, interleukin; MAPK, mitogen-activated protein kinase; mTORC, mammalian target of rapamycin complex; NFκB, nuclear factor-κB; PI3K, phosphoinositide 3-kinase; PIP2, phosphatidylinositol 4,5-bisphosphate; PIP3, phosphatidylinositol 3,4,5-trisphosphate; PTP, protein tyrosine phosphatases; Ras/Raf/MEK/ERK, ras protein, raf protein, MEK (a dual threonine and tyrosine recognition kinase), extracellular signal-regulated kinase; RTK, receptor tyrosine kinase; SelK, selenoprotein K; SOS, Son of Sevenless; TGF-β, transforming growth factor β; TNF-α, tumor necrosis factor α; Treg, regulatory T cell; TrxR, thioredoxin reductase; VEGF, vascular endothelial growth factor; VEGFR, VEGF receptor.

Introduction

Wound healing involves a series of physiological actions in the body that aim to restore the function and integrity of damaged skin.1,2 Failure of the natural wound healing process can lead to chronic wound complications, such as infection and amputation, that significantly affect quality of life.3-5 The key events of the wound healing process are inflammation, angiogenesis, and epithelialization, and various biochemical pathways are responsible for regulating these events.5 A disturbance in these events, which may be due to a weak immune system, comorbidities, or advanced age (ie, being 65 years or more in age), can lead to uncontrolled inflammation (resulting in excessive scarring), poor angiogenesis or growth of new blood vessels (resulting in delayed healing), and poor epithelialization or growth of new skin (resulting in increased risk of infection).6-8 To counter these risks, research is underway on substances that can support normal wound healing and reduce the risk of wound complications.5,6,9,10 One such substance that should be given due consideration is fish skin. Previous studies have determined that fish skin contains high amounts of omega-3, collagen, and selenium, and that applying fish skin on wounds markedly improves wound healing parameters.

The possible mechanisms of action behind these successful findings are discussed in this review, in the interest of promoting continued study of fish skin and the development of a practically beneficial and commercially available healing agent. The pathways in the normal wound healing process that regulate inflammation, angiogenesis, and epithelialization are also discussed, as are the means by which the omega-3, collagen, and selenium content in fish skin positively influences each pathway, and in turn, the wound healing process.

The normal mechanism of wound healing

Inflammation and the IL-10 signaling pathway. Inflammation is the body’s innate response to injury or infection, designed to eliminate pathogens, remove cellular debris, and initiate tissue repair.4,5,11,12 However, prolonged or excessive inflammation can impede wound healing and lead to chronic wounds and excessive scarring.4 Thus, the timely activation of anti-inflammatory mechanisms is crucial for efficient wound healing.13-15 Pro- and anti-inflammatory cytokines in the interleukin signaling pathway are balanced during the anti-inflammatory response.13

To ensure that cytokine-driven inflammation does not overwhelm a wound site, M2 macrophages secrete IL-10 and TGF-β.16-18 TGF-β induces the differentiation of Tregs, which suppress excessive immune responses by reducing the activity of effector T cells. TGF-β also produces IL-10.10 IL-10 reduces pro-inflammatory cytokines IL-1β, IL-6, and TNF-α by inhibiting activation of MAPK and nuclear factor-κB.19,20

In summary, the body requires pro-inflammatory cytokines to cause inflammation at a wound site so that the body can become aware of the wound and start its natural healing process. However, anti-inflammatory cytokines also need to be activated so that pro-inflammatory cytokines do not cause unchecked inflammation, a common finding in delayed wound healing. The components of fish skin increase the activation of anti-inflammatory cytokines, as discussed in “The Role of Fish Skin in Improving Wound Healing” section.

Angiogenesis and the Ras/Raf/MEK/ERK pathway. Upon wound injury, EGF is released by platelets, macrophages, and keratinocytes.21 EGF binds and activates EGFR, which begins a conformational change resulting in trans-autophosphorylation.22,23 The autophosphorylation event creates docking sites for signaling proteins, including Src homology 2 and growth factor receptor-bound protein 2, thereby initiating 2 important biochemical pathways.24-26

When growth factor receptor-bound protein 2 binds to EGFR, SOS is activated. SOS catalyzes the exchange of a guanosine diphosphate molecule for a guanosine triphosphate molecule to activate Ras, which then causes Raf, MEK (a dual threonine and tyrosine recognition kinase), and ERK (part of the MAPK family), to be activated in a specific order.27-29 In the nucleus, ERK moves and phosphorylates transcription factors like c-Jun. These elements enhance the expression of genes essential for the migration and proliferation of new epithelial cells in the wound bed.30

Similar to EGFR, VEGF activates its receptor (ie, VEGFR) to initiate the Ras/Raf/MEK/ERK pathway, thereby activating the transcription factors responsible for the migration and proliferation of endothelial cells, as well as tube formation, a crucial element of angiogenesis in the wound bed.31-34

Every time an EGFR or VEGFR molecule is activated, a series of biochemical steps, collectively known as the Ras/Raf/MEK/ERK pathway, can successfully occur at the wound site to eventually allow new blood vessels to replace injured ones. The new blood vessels also provide nutrients that enable faster wound recovery. As discussed in “The Role of Fish Skin in Improving Wound Healing” section, the components of fish skin increase the rate of activation of EGFR and VEGFR, thereby increasing the rate at which new healthy blood vessels are formed.

Epithelialization and the PI3K/Akt pathway. Simultaneously, EGFR and VEGFR trigger the activation of PI3K, which phosphorylates PIP2 to generate PIP3.35,36 PIP3 then activates Akt (also known as protein kinase B) by phosphorylating it at the threonine 308 residue by phosphoinositide-dependent kinase-1 and the serine 473 residue by mTORC2.37,38 Akt creates an environment that is favorable for new cell growth in 2 ways: it inactivates proapoptotic proteins (ie, BAD and Bcl-2) and proapoptotic enzymes (ie, caspases).39 It also activates mTORC1, which modulates the eukaryotic translation initiation factor 4E-binding protein 1 and ribosomal protein S6 kinase.40,41 This cascade promotes endothelial cell proliferation and stimulates protein synthesis for new cell growth.

Akt also boosts angiogenesis. Endothelial nitric oxide synthase and glycogen synthase kinase-3 are phosphorylated and modulated by Akt to increase endothelial cell proliferation and tube formation.38,42-44

Increasing the rate of activation of EGFR and VEGFR also increases the rate by which new cells grow and cover injured skin. Because fish skin components upregulate EGFR and VEGFR, they also upregulate the resulting series of biochemical steps, collectively known as the PI3K/Akt pathway, which increases the amount of healthy skin cells entering the wound site and the amount of protein being made available to synthesize new cells.

The Role of Fish Skin in Improving Wound Healing

Fish skin has emerged as a promising candidate for boosting the natural wound healing process due to its uniquely rich levels of omega-3, collagen, and selenium.45-48 This section discusses how these 3 components interact with the pathways involved in wound recovery (ie, IL-10 signaling, Ras/Raf/MEK/ERK, and PI3K/Akt) to improve healing.

Omega-3

Omega-3 fatty acid content, in particular EPA and DHA, can vary in fish skin depending on fish diet and species. Fatty fish species, such as salmon, mackerel, and sardines, are widely recognized as rich sources of omega-3 fatty acids.48-50

Role in the IL-10 signaling pathway: inhibition of 3 pro-inflammatory compounds and creation of 4 anti-inflammatory compounds. EPA and DHA can competitively inhibit cyclooxygenase-2 (COX-2), which is responsible for converting AA into pro-inflammatory prostaglandins51,52 (Figure 1). Additionally, EPA competes with AA as a substrate for 5-LOX. 5-LOX converts AA into leukotrienes, which are potent inflammatory mediators.53,54 EPA substitution results in the formation of anti-inflammatory resolvins and protectins instead. Both EPA and DHA also suppress NFκB activation by inhibiting the degradation of its inhibitory protein, inhibitor kappa B-alpha, thus preventing its translocation into the nucleus and subsequent expression of pro-inflammatory genes.55 Moreover, EPA and DHA serve as a starting point for the creation of specific pro-resolving lipid mediators, including lipoxins and maresins, which are responsible for suppressing IL-1β and IL-6.56,57

Figure 1

Role in the Ras/Raf/MEK/ERK and PI3K/Akt pathways: activation of Ras and PI3K by altering lipid composition of cell membranes. EPA and DHA can increase epithelialization and angiogenesis by increasing the activation of EGFR and VEGFR, respectively, which can then activate both pathways.58,59 This effect is exerted by replacing the fatty acids of the existing phospholipids in cell membranes with omega-3 fatty acids via acyltransferases and phospholipases.60-62 Because omega-3 fatty acids have longer and more unsaturated fatty acid tails, the increased unsaturation creates a more fluid and flexible membrane structure.62,63 This structure increases the clustering of transmembrane receptors and enhances receptor-receptor interactions, resulting in increased activity of VEGFR and EGFR.62-66

Collagen

The ECM of fish skin is laden with collagen, particularly type I collagen that is notably similar in structure to human collagen.67 Fish collagen has a high degree of purity (approximately 70%), depending on the season and the age and species of the fish.67,68 Different fish species and even different portions of the same species may have varying total collagen contents. Some of the highest content (54.2% w/w) has been found in the skin of Nile tilapia (Oreochromis niloticus).69,70

Role in the IL-10 signaling pathway: inhibition of 5 pro-inflammatory compounds and activation of 2 anti-inflammatory compounds. Collagen can sequester pro-inflammatory TNF-α and IL-1β by binding them to collagen fibers, thereby blocking their interaction with cellular receptors of pro-inflammatory signaling71-73 (Figure 2). Matrix metalloproteinases, which are responsible for breaking down ECM and producing pro-inflammatory cytokines, are naturally inhibited by collagen.74-76 Similar to omega-3 fatty acids, collagen-derived peptides inhibit COX-2 expression.77 Additionally, collagen-derived peptides, including glycine-proline-hydroxyproline, inhibit NFκB.71 Collagen also influences macrophage polarization to shift the balance from pro-inflammatory M1 to anti-inflammatory M2 macrophages.78,79 Instead, collagen promotes the activation of Tregs.80 Both activities upregulate the production of IL-10, as discussed previously.78-80

Figure 2

Role in the Ras/Raf/MEK/ERK and PI3K/Akt pathways: activation of Ras and PI3K by binding to collagen receptors. Collagen binds to integrins, a family of transmembrane collagen receptors, on both epithelial and endothelial cells.81 This activates focal adhesion kinase and Src family kinases, which phosphorylate and activate downstream effectors, including Ras and PI3K.81,82 Similarly, in epithelial cells, collagen binds to integrins and DDRs, which are non-integrin collagen receptors that are members of the RTK family and includes EGFR.83,84 Both integrins (eg, α1β1, α2β1) and DDRs mediate the attachment of endothelial cells to the postinjury ECM.84-87

Selenium

Selenium is an essential trace element present in fish skin and can exist in different chemical forms, including selenocysteine, selenomethionine, and various selenoproteins.88 Selenocysteine and selenomethionine are amino acid analogues of cysteine and methionine, respectively, with a selenium atom replacing the sulfur atom in the side chain. These selenium-containing amino acids can be incorporated into the primary structure of proteins during translation, leading to the synthesis of selenoproteins, which play several critical roles in wound healing88-90 (Figure 3).

Figure 3

Role in the Ras/Raf/MEK/ERK pathway: activation of Raf through selenoprotein SelK. Selenium directly influences the Ras/Raf/MEK/ERK pathway through SelK. SelK interacts with the Raf-1 kinase to enhance its binding to Ras and thereby initiate Raf-1 activation. This, in turn, leads to the phosphorylation and activation of MEK and ERK. Furthermore, selenium supplementation has been shown to increase the expression of Ras and Raf-1 in certain cell types.91,92

Role in the PI3K/Akt pathway: activation of PI3K through selenoprotein TrxR. Similarly, selenium influences the PI3K pathway by being an essential cofactor for selenoprotein TrxR.93,94 TrxR, through its redox activity, affects the formation and stability of RTK dimers.94-96 RTKs are upstream regulators of PI3K. When activated by ligand binding, they undergo dimerization and autophosphorylation, leading to the recruitment of PI3K and initiation of the PI3K pathway.97 Additionally, it has been noted that selenium administration increases the expression and phosphorylation of Akt, thereby amplifying the signaling output of the PI3K pathway.97-99

Role in all pathways: modulating key enzymes for successful pathway activation. Selenium is a cofactor mandatory for the activation of GPx, a selenoprotein antioxidant enzyme that neutralizes free radicals to minimize oxidative stress.100 It has been observed that a strong messenger RNA expression of GPx1 during the inflammatory process aids wound healing. The neutralizing activity of selenium facilitates maintaining a favorable environment for VEGF expression, endothelial cell proliferation, and the development of capillary-like structures.100,101 Selenium has also been shown to inhibit PTPs, a class of signaling enzymes that dephosphorylate phosphotyrosine residues on proteins involved in signaling pathways, including EGFR, in order to deactivate them.102 By inhibiting PTP activity, selenium thereby prolongs the phosphorylation and activation of proteins in the Ras/Raf/MEK/ERK and PI3K pathways, allowing for sustained signaling to activate the pathways (and related genes) responsible for successful wound healing.103

Limitations

As with all review articles, this article is limited by the complete reliance on previously published research. However, the authors of the current article ensured the incorporation of a variety of primary sources (original research articles) and secondary sources (review articles) when synthesizing the text in this manuscript, with preference given to recently published and peer-reviewed articles.

Conclusion

Fish skin is a promising potential candidate to improve wound healing due to its uniquely high omega-3, collagen, and selenium content. These components have shown to boost the pathways that regulate the natural wound healing process (IL-10 signaling, Ras/Raf/MEK/ERK, and PI3K pathways), which in turn boosts the natural wound healing process. Notable cellular effects of omega-3, collagen, and selenium include the inhibition of pro-inflammatory components and creation of anti-inflammatory compounds involved in interleukin signaling. Additional notable effects include the upregulation of Ras and PI3K activation in the Ras/Raf/MEK/ERK and PI3K pathways, respectively, through altering lipid composition (via omega-3), binding to collagen receptors (via collagen), and modulating selenoproteins (via selenium). The mechanisms discussed herein emphasize the necessity to investigate the application of fish skin as a practical and commercial wound healing agent in clinical settings. Additional wound healing properties of fish skin, such as microbial protection of open wounds, should be studied in the future to ascertain their potential.

Acknowledgements

Affiliations: 1Department of Diet & Nutritional Sciences, Rashid Latif Khan University, Lahore, Punjab, Pakistan 2University Institute of Diet & Nutritional Sciences, The University of Lahore, Lahore, Punjab, Pakistan 3Institute of Food Sciences and Nutrition, University of Sargodha, Sargodha, Punjab, Pakistan

ORCID: Murtaza, 0000-0002-0893-7397

Disclosure: The authors disclose no financial or other conflicts of interest.

Correspondence: Sarosh Malik, MS; The University of Lahore, 186 D Punjab Cooperative Housing Society Ghazi Road Lahore Cantt, Lahore, Punjab 54000 Pakistan; saroshmalik12@gmail.com

Manuscript Accepted: April 26, 2024

How Do I Cite This?

Basharat S, Malik S, Samad HUA, Murtaza MA. Diving deep into healing: the promising role of fish skin in wound recovery. Wounds. 2024;36(8):274-280. doi:10.25270/wnds/23134

References

1. Dąbrowska AK, Spano F, Derler S, Adlhart C, Spencer ND, Rossi RM. The relationship between skin function, barrier properties, and body-dependent factors. Skin Res Technol. 2017;24(2):165-174. doi:10.1111/srt.12424

2. Murray RZ, West ZE, Cowin AJ, Farrugia BL. Development and use of biomaterials as wound healing therapies. Burns Trauma. 2019;7. doi:10.1186/s41038-018-0139-7

3. Guo S, DiPietro LA. Factors affecting wound healing. J Dental Res. 2010;89(3):219-229. doi:10.1177/0022034509359125

4. Reinke JM, Sorg H. Wound repair and regeneration. Eur Surg Res. 2012;49(1):35-43. doi:10.1159/000339613

5. Eming SA, Martin P, Tomic-Canic M. Wound repair and regeneration: mechanisms, signaling, and translation. Sci Transl Med. 2014;6(265):265sr6. doi:10.1126/scitranslmed.3009337

6. Wang Z, Zheng L, Lian C, Qi Y, Li W, Wang S. Human umbilical cord-derived mesenchymal stem cells relieve hind limb ischemia by promoting angiogenesis in mice. Stem Cells Develop. 2019;28(20):1384-1397. doi:10.1089/scd.2019.0115

7. Bielefeld KA, Amini-Nik S, Alman BA. Cutaneous wound healing: recruiting developmental pathways for regeneration. Cell Mol Life Sci. 2012;70(12):2059-2081. doi:10.1007/s00018-012-1152-9

8. Olivera S, Tomic-Canic M. Human ex vivo wound healing model. Wound Repair Regen. 2013;25(6):1017-1026. doi:10.1111/wrr.12600

9. Matei AM, Caruntu C, Tampa M, et al. Applications of nanosized-lipid-based drug delivery systems in wound care. Appl Sci. 2021;11(11):4915. doi:10.3390/app11114915

10. Herrmann M, Diederichs S, Melnik S, et al. Extracellular vesicles in musculoskeletal pathologies and regeneration. Front Bioeng Biotechnol. 2021;8:624096. doi:10.3389/fbioe.2020.624096

11. Koh TJ, DiPietro LA. Inflammation and wound healing: the role of the macrophage. Exp Rev Mol Med. 2011;13:e23. doi:10.1017/s1462399411001943

12. Gurtner GC, Werner S, Barrandon Y, Longaker MT. Wound repair and regeneration. Nature. 2008;453(7193):314-321. doi:10.1038/nature07039

13. Park CH, Abramson ZR, Taba M, et al. Three-dimensional micro-computed tomographic imaging of alveolar bone in experimental bone loss or repair. J Periodontol. 2007;78(2):273-281. doi:10.1902/jop.2007.060252

14. Zhang M, Zhao X. Alginate hydrogel dressings for advanced wound management. Int J Biol Macromol. 2020;162:1414-1428. doi:10.1016/j.ijbiomac.2020.07.311

15. Almadani YH, Vorstenbosch J, Davison PG, Murphy AM. Wound healing: a comprehensive review. Semin Plast Surg. 2021;35(3):141-144. doi:10.1055/s-0041-1731791

16. Gabryšová L, Alvarez-Martinez M, Luisier R, et al. c-Maf controls immune responses by regulating disease-specific gene networks and repressing IL-2 in CD4+ T cells. Nature Immunology. 2018;19(5):497-507. doi:10.1038/s41590-018-0083-5. Erratum in: Nat Immunol. 2019;20(3):374. doi:10.1038/s41590-019-0331-3

17. Rutz S, Ouyang W. Regulation of interleukin-10 and interleukin-22 expression in T helper cells. Curr Opin Immunol. 2011;23(5):605-612. doi:10.1016/j.coi.2011.07.018

18. Jarnicki AG, Lysaght J, Todryk S, Mills KH. Suppression of antitumor immunity by IL-10 and TGF-beta-producing T cells infiltrating the growing tumor: influence of tumor environment on the induction of CD4+ and CD8+ regulatory T cells. J Immunol. 2006;177(2):896-904. doi:10.4049/jimmunol.177.2.896

19. Landén NX, Li D, Ståhle M. Transition from inflammation to proliferation: a critical step during wound healing. Cell Mol Life Sci. 2016;73(20):3861-3885. doi:10.1007/s00018-016-2268-0

20. Geraghty NJ, Watson D, Adhikary SR, Sluyter R. P2X7 receptor in skin biology and diseases. World J Dermatol. 2016;5(2):72. doi:10.5314/wjd.v5.i2.72

21. Negut I, Dorcioman G, Grumezescu V. Scaffolds for wound healing applications. Polymers (Basel). 2020;12(9):2010. doi:10.3390/polym12092010

22. Zhang Y, Wang X, Whang Y, Li X, Zhang X, Zhang X. Chitosan/epidermal growth factor loaded electrospun nanofivers for wound healing. Int J Biol Macromol. 2018;120:28-33. doi:10.1016/j.ijbiomac.2018.07.001

23. Li X, Wang X, Chen D, Chen Y, Wei L, Li X. Platelet rich plasma combined with epidermal growth factor promotes wound healing in diabetic rats by modulating the expression of inflammatory cytokines Int J Molec Med. 2018;24:8449-8458. doi:10.12659/msm.912596

24. Kim HS, Sun X, Lee JH, Kim HW, Fu X, Leong KW. Advanced drug delivery systems and artificial skin grafts for skin wound healing. Adv Drug Deliv Rev. 2019;146:209-239. doi:10.1016/j.addr.2018.12.014

25. Widgerow AD, King K, Tocco-Tussardi I, et al. The burn wound exudate—an under-utilized resource. Burns. 2015;41(1):11-17. doi:10.1016/j.burns.2014.06.002

26. Chen L, Tredget EE, Wu PY, Wu Y. Paracrine factors of mesenchymal stem cells recruit macrophages and endothelial lineage cells and enhance wound healing. PLoS One. 2008;3(4):e1886. doi:10.1371/journal.pone.0001886. Erratum in: PLoS One. 2024;19(4):e0302417

27. Ren S, Chen J, Duscher D, et al. Microvesicles from human adipose stem cells promote wound healing by optimizing cellular functions via AKT and ERK signaling pathways. Stem Cell Res Ther. 2019;10(1):47. doi:10.1186/s13287-019-1152-x

28. Grassian AR, Schafer ZT, Brugge JS. ErbB2 stabilizes epidermal growth factor receptor (EGFR) expression via Erk and Sprouty2 in extracellular matrix-detached cells. J Biol Chem. 2011;286(1):79-90. doi:10.1074/jbc.m110.169821

29. Harrisingh MC, Perez-Nadales E, Parkinson DB, Malcolm DS, Mudge AW, Lloyd AC. The Ras/Raf/ERK signalling pathway drives Schwann cell dedifferentiation. EMBO J. 2004;23(15):3061-3071. doi:10.1038/sj.emboj.7600309

30. Boudra R, Ramsey MR. 1433 Role of BNC1 in keratinocytes proliferation and migration: a critical regulator of wound healing? J Investigat Dermatol. 2018;138(5):S243. doi:10.1016/j.jid.2018.03.1451

31. Ghilardi SJ, O’Reilly BM, Sgro AE. Intracellular signaling dynamics and their role in coordinating tissue repair. Wiley Interdiscip Rev Syst Biol Med. 2020;12(3):e1479. doi:10.1002/wsbm.1479

32. Phimnuan P, Dirand Z, Tissot M, et al. Beneficial effects of a blended fibroin/aloe gel extract film on the biomolecular mechanism(s) via the MAPK/ERK pathway relating to diabetic wound healing. ACS Omega. 2023;8(7):6813-6824. doi:10.1021/acsomega.2c07507

33. Makieva S, Giacomini E, Ottolina J, Sanchez A, Papaleo E, Viganò P. Inside the endometrial cell signaling subway: mind the gap(s). Int J Mol Sci. 2018;19(9):2477. doi:10.3390/ijms19092477

34. Ras Raf MEK ERK signaling pathway - overview, regulation and role in pathology [Internet]. JJ Medicine; 2017 March 10 [cited 2023 June 01]. Video: 6:09 min. https://www.youtube.com/watch?v=i1f2RbogiDw

35. Zhang W, Bai X, Zhao B, et al. Cell-free therapy based on adipose tissue stem cell-derived exosomes promotes wound healing via the PI3K/Akt signaling pathway. Exp Cell Res. 2018;370(2):333-342. doi:10.1016/j.yexcr.2018.06.035

36. Zhang J, Liu X, Li H, et al. Exosomes/tricalcium phosphate combination scaffolds can enhance bone regeneration by activating the PI3K/Akt signaling pathway. Stem Cell Res Ther. 2016;7(1). doi:10.1186/s13287-016-0391-3

37. Li G, Li YY, Sun JE, Lin W hua, Zhou RX. ILK–PI3K/AKT pathway participates in cutaneous wound contraction by regulating fibroblast migration and differentiation to myofibroblast. Lab Invest. 2016;96(7):741-751. doi:10.1038/labinvest.2016.48

38. Zhang YM, Zhang ZQ, Liu YY, et al. Requirement of Gαi1/3-Gab1 signaling complex for keratinocyte growth factor-induced PI3K-AKT-mTORC1 activation. J Invest Dermatol. 2015;135(1):181-191. doi:10.1038/jid.2014.326

39. Yuan X, Han L, Fu P, et al. Cinnamaldehyde accelerates wound healing by promoting angiogenesis via up-regulation of PI3K and MAPK signaling pathways. Lab Invest. 2018;98(6):783-798. doi:10.1038/s41374-018-0025-8

40. Xing W, Guo W, Zou CH, et al. Acemannan accelerates cell proliferation and skin wound healing through AKT/mTOR signaling pathway. J Dermatol Sci. 2015;79(2):101-109. doi:10.1016/j.jdermsci.2015.03.016

41. Shi H, Cheng Y, Ye J, et al. bFGF promotes the migration of human dermal fibroblasts under diabetic conditions through reactive oxygen species production via the PI3K/Akt-Rac1-JNK pathways. Int J Biol Sci. 2015;11(7):845-859. doi:10.7150/ijbs.11921

42. Wang L, Sun H, Fu S, Chao S, Hou H, Liu X. The role of PI3K/AKT/ENOS pathway in spinal cord injury and the recovery of motor function. Int J Clin Exp Med. 2020;13(7):4960-4965. doi:10.23736/s0031-0808.20.03974-9

43. Fan S, Qi D, Yu Q, et al. Intermedin alleviates the inflammatory response and stabilizes the endothelial barrier in LPS-induced ARDS through the PI3K/Akt/eNOS signaling pathway. Int Immunopharmacol. 2020;88:106951. doi:10.1016/j.intimp.2020.106951

44. Su YC, Lee WC, Wang CC, Yeh SA, Chen WH, Chen PJ. Targeting PI3K/AKT/mTOR signaling pathway as a radiosensitization in head and neck squamous cell carcinomas. Int J Mol Sci. 2022;23(24):15749. doi:10.3390/ijms232415749

45. Kirsner RS, Margolis DJ, Baldursson BT, et al. Fish skin grafts compared to human amnion/chorion membrane allografts: a double-blind, prospective, randomized clinical trial of acute wound healing. Wound Repair Regen. 2019;28(1):75-80. doi:10.1111/wrr.12761

46. Micheal S, Winters C, Khan M. Acellular fish skin graft use for diabetic lower extremity wound healing: a retrospective study of 58 ulcerations and a literature review. Wounds. 2019;31(10):262-268.

47. Fiakos G, Kuang Z, Lo E. Improved skin regeneration with acellular fish skin grafts. Engineer Regen. 2020;1:95-101. doi:10.1016/j.engreg.2020.09.002

48. Stone R, Saathoff EC, Larson DA, et al. Accelerated wound closure of deep partial thickness burns with acellular fish skin graft. Int J Mol Sci. 2021;22(4):1590. doi:10.3390/ijms22041590

49. Liu Y, Ramakrishnan VV, Dave D. Lipid class and fatty acid composition of oil extracted from Atlantic salmon by-products under different optimization parameters of enzymatic hydrolysis. Biocatalysis Agricultur Biotechnol. 2020;30:101866. doi:10.1016/j.bcab.2020.101866

50. Calder PC. Omega-3 fatty acids and inflammatory processes: from molecules to man. Biochem Socy Trans. 2017;45(5):1105-1115. doi:10.1042/bst20160474

51. Ishak WMW, Katas H, Yuen NP, Abdullah MA, Zulfakar MH. Topical application of omega-3-, omega-6-, and omega-9-rich oil emulsions for cutaneous wound healing in rats. Drug Deliv Transl Res. 2018;9(2):418-433. doi:10.1007/s13346-018-0522-8

52. Seth NE, Chopra DI, Lev-Toh H. Fish skin grafts with omega-3 for treatment of chronic wounds: exploring the role of omega-3 fatty acids in wound healing and a review of clinical healing outcomes. Surg Technol Int. 2022;46(10):40:38-46.

53. McCusker MM, Grant-Kels JM. Healing fats of the skin: the structural and immunologic roles of the omega-6 and omega-3 fatty acids. Clin Dermatol. 2010;28(4):440-451. doi:10.1016/j.clindermatol.2010.03.020

54. Balić A, Vlašić D, Žužul K, Marinović B, Bukvić Mokos Z. Omega-3 versus omega-6 polyunsaturated fatty acids in the prevention and treatment of inflammatory skin diseases. Int J Mol Sci. 2020;21(3):741. doi:10.3390/ijms21030741

55. Schunck WH, Konkel A, Fischer R, Weylandt KH. Therapeutic potential of omega-3 fatty acid-derived epoxyeicosanoids in cardiovascular and inflammatory diseases. Pharmacol Therapeut. 2018;183:177-204. doi:10.1016/j.pharmthera.2017.10.016

56. Kwon Y. Immuno-resolving ability of resolvins, protectins, and maresins derived from omega-3 fatty acids in metabolic syndrome. Mol Nutr Food Res. 2019;64(4):1900824. doi:10.1002/mnfr.201900824

57. Prasad P, Anjali P, Sreedhar RV. Plant-based stearidonic acid as sustainable source of omega-3 fatty acid with functional outcomes on human health. Crit Rev Food Sci Nutr. 2020;61(10):1725-1737. doi:10.1080/10408398.2020.1765137

58. Minokawa Y, Sawada Y, Nakamura M. The influences of omega-3 polyunsaturated fatty acids on the development of skin cancers. Diagnostics (Basel). 2021;11(11):2149. doi:10.3390/diagnostics11112149

59. Woodrow T, Chant T, Chant H. Treatment of diabetic foot wounds with acellular fish skin graft rich in omega-3: a prospective evaluation. J Wound Care. 2019;28(2):76-80. doi:10.12968/jowc.2019.28.2.76

60. Lullove E, Liden B, McEneaney P, et al. Evaluating the effect of omega-3–rich fish skin in the treatment of chronic, nonresponsive diabetic foot ulcers: penultimate analysis of a multicenter, prospective, randomized controlled trial. Wounds. 2022;34(4):e34-e36. doi:10.25270/wnds/2022.e34e36

61. Abbasnezhad S, Biazar E, Aavani F, Kamalvand M, Heidari Keshel S, Pourjabbar B. Chemical modification of acellular fish skin as a promising biological scaffold by carbodiimide cross-linker for wound healing. Int Wound J. 2022;20(5):1566-1577. doi:10.1111/iwj.14012

62. Dorweiler B, Trinh TT, Dünschede F, et al. The marine Omega3 wound matrix for treatment of complicated wounds. Gefässchirurgie. 2018;23(S2):46-55. doi:10.1007/s00772-018-0428-2

63. Severing A, Rembe J, Füllerer M, Stürmer EK. Impact of the chronic wound microenvironment and marine omega-3 fatty acids on skin cell regeneration processes. Experiment Dermatol. 2021;31(5):725-735. doi:10.1111/exd.14506

64. Trinh TT, Dünschede F, Vahl CF, Dorweiler B. Marine Omega3 wound matrix for the treatment of complicated wounds. Phlebologie. 2016;45(02):93-98. doi:10.12687/phleb2305-2-2016

65. Torrissen M, Ytteborg E, Svensen H, et al. Investigation of the functions of n-3 very-long-chain PUFAs in skin using in vivo Atlantic salmon and in vitro human and fish skin models. Br J Nutr. 2023:1-17. doi:10.1017/s0007114523001150

66. Jana S, Datta P, Das H, Ghosh PR, Kundu B, Nandi SK. Engineering vascularizing electrospun dermal grafts by integrating fish collagen and ion-doped bioactive glass. ACS Biomater Sci Engin. 2022;8(2):734-752. doi:10.1021/acsbiomaterials.1c01098

67. Blanco M, Vázquez J, Pérez-Martín R, Sotelo C. Hydrolysates of fish skin collagen: an opportunity for valorizing fish industry byproducts. Marine Drugs. 2017;15(5):131. doi:10.3390/md15050131

68. Sotelo CG, Comesaña MB, Ariza PR, Pérez-Martín RI. Characterization of collagen from different discarded fish species of the west coast of the Iberian Peninsula. J Aquat Food Prod Technol. 2015;25(3):388-399. doi:10.1080/10498850.2013.865283

69. Wang L, Wang Q, Qian J, et al. Bioavailability bioavailable forms of collagen after oral administration to rats. J Agric Food Chem. 2015;63(14):3752-3756. doi:10.1021/jf5057502

70. Alves APNN, Lima Júnior EM, Piccolo NS, et al. Study of tensiometric properties, microbiological and collagen content in nile tilapia skin submitted to different sterilization methods. Cell Tissue Bank. 2018;19(3):373-382. doi:10.1007/s10561-017-9681-y

71. Subhan F, Kang HY, Lim Y, et al. Fish scale collagen peptides protect against CoCl2/TNF-α-induced cytotoxicity and inflammation via inhibition of ROS, MAPK, and NF-κB pathways in HaCaT cells. Oxid Med Cell Longev. 2017;2017:9703609. doi:10.1155/2017/9703609

72. Chen Y, Jin H, Yang F, et al. Physicochemical, antioxidant properties of giant croaker (Nibea japonica) swim bladders collagen and wound healing evaluation. Int J Biol Macromol. 2019;138:483-491. doi:10.1016/j.ijbiomac.2019.07.111

73. Shang Y, Yao S, Qiao X, et al. Evaluations of marine collagen peptides from tilapia skin on experimental oral ulcer model of mice. Mater Today Commun. 2021;26:101893. doi:10.1016/j.mtcomm.2020.101893

74. Xiao Z, Liang P, Chen J, et al. A peptide YGDEY from tilapia gelatin hydrolysates inhibits UVB-mediated skin photoaging by regulating MMP -1 and MMP -9 expression in HaCaT cells. Photochem Photobiol. 2019;95(6):1424-1432. doi:10.1111/php.13135

75. Gaspar-Pintiliescu A, Stanciuc AM, Craciunescu O. Natural composite dressings based on collagen, gelatin and plant bioactive compounds for wound healing: a review. Int J Biol Macromol. 2019;138:854-865. doi:10.1016/j.ijbiomac.2019.07.155

76. Zhang Z, Zhu H, Zheng Y, et al. The effects and mechanism of collagen peptide and elastin peptide on skin aging induced by D-galactose combined with ultraviolet radiation. J Photochem Photobiol B. 2020;210:111964. doi:10.1016/j.jphotobiol.2020.111964

77. Kumar LV, Shakila RJ, Jeyasekaran G. In vitro anti-cancer, anti-diabetic, anti-inflammation and wound healing properties of collagen peptides derived from unicorn leatherjacket (Aluterus monoceros) at different hydrolysis. Turkish J Fish Aquat Sci. 2019;4(19):551-60.

78. MacLeod AS, Mansbridge JN. The innate immune system in acute and chronic wounds. Adv Wound Care (New Rochelle). 2016;5(2):65-78. doi:10.1089/wound.2014.0608

79. Alves AL, Costa-Gouveia J, Vieira de Castro J, et al. Study of the immunologic response of marine-derived collagen and gelatin extracts for tissue engineering applications. Acta Biomater. 2022;141:123-131. doi:10.1016/j.actbio.2022.01.009

80. Kalekar LA, Cohen JN, Prevel N, et al. Regulatory T cells in skin are uniquely poised to suppress profibrotic immune responses. Sci Immunol. 2019;4(39):eaaw2910. doi:10.1126/sciimmunol.aaw2910

81. Zeltz C, Gullberg D. The integrin–collagen connection – a glue for tissue repair? J Cell Sci. 2016;129(6):1284-1284. doi:10.1242/jcs.188672

82. Huang H, Wright S, Zhang J, Brekken RA. Getting a grip on adhesion: Cadherin switching and collagen signaling. Biochim Biophys Acta Mol Cell Res. 2019;1866(11):118472. doi:10.1016/j.bbamcr.2019.04.002

83. Orgel JPRO, Madhurapantula RS. A structural prospective for collagen receptors such as DDR and their binding of the collagen fibril. Biochim Biophys Acta Mol Cell Res. 2019;1866(11):118478. doi:10.1016/j.bbamcr.2019.04.008

84. Elango J, Hou C, Bao B, Wang S, Maté Sánchez de Val JE, Wenhui W. The molecular interaction of collagen with cell receptors for biological function. Polymers. 2022;14(5):876. doi:10.3390/polym14050876

85. Woltersdorf C, Bonk M, Leitinger B, et al. The binding capacity of α1β1-, α2β1- and α10β1-integrins depends on non-collagenous surface macromolecules rather than the collagens in cartilage fibrils. Matrix Biol. 2017;63:91-105. doi:10.1016/j.matbio.2017.02.001

86. Borza CM, Bolas G, Zhang X, et al. The collagen receptor discoidin domain receptor 1b enhances integrin β1-mediated cell migration by interacting with talin and promoting Rac1 activation. Front Cell Dev Biol. 2022;10:836797. doi:10.3389/fcell.2022.836797

87. Boraschi-Diaz I, Wang J, Mort JS, Komarova SV. Collagen type I as a ligand for receptor-mediated signaling. Front Phys. 2017;5. doi:10.3389/fphy.2017.00012

88. Hariharan S, Dharmaraj S. Selenium and selenoproteins: it’s role in regulation of inflammation. Inflammopharmacology. 2020;28(3):667-695. doi:10.1007/s10787-020-00690-x

89. Mehrabi T, Mesgar AS, Mohammadi Z. Bioactive glasses: a promising therapeutic ion release strategy for enhancing wound healing. ACS Biomater Sci Engin. 2020;6(10):5399-5430. doi:10.1021/acsbiomaterials.0c00528

90. Gao X, Zhang Z, Li Y, et al. Selenium deficiency facilitates inflammation following S aureus infection by regulating TLR2-related pathways in the mouse mammary gland. Biol Trace Element Res. 2016;172(2):449-457. doi:10.1007/s12011-015-0614-y

91. Zhang L, Xia H, Xia K, et al. Selenium regulation of the immune function of dendritic cells in mice through the ERK, Akt and RhoA/ROCK pathways. Biol Trace Element Res. 2020;199(9):3360-3370. doi:10.1007/s12011-020-02449-5

92. Li T, Sun Y, Wang J, Zhang C, Sun Y. Promoted skin wound healing by tail-amputated eisenia foetida proteins via the Ras/Raf/MEK/ERK signaling pathway. ACS Omega. 2023;8(15):13935-13943. doi:10.1021/acsomega.3c00317

93. Kipp AP. Selenium-dependent glutathione peroxidases during tumor development. Adv Cancer Res. 2017;136:109-138. doi:10.1016/bs.acr.2017.07.004

94. Park J, Lee JH, Yoon BS, et al. Additive effect of bFGF and selenium on expansion and paracrine action of human amniotic fluid-derived mesenchymal stem cells. Stem Cell Res Ther. 2018;9(1):293. doi:10.1186/s13287-018-1058-z

95. Jere SW, Houreld NN, Abrahamse H. Role of the PI3K/AKT (mTOR and GSK3β) signalling pathway and photobiomodulation in diabetic wound healing. Cytokine Growth Factor Rev. 2019;50:52-59. doi:10.1016/j.cytogfr.2019.03.001

96. Yao X, EI-Samahy MA, Fan L, et al. In vitro influence of selenium on the proliferation of and steroidogenesis in goat luteinized granulosa cells. Theriogenology. 2018;114:70-80. doi:10.1016/j.theriogenology.2018.03.014

97. Wang J, Lian S, He X, et al. Selenium deficiency induces splenic growth retardation by deactivating the IGF-1R/PI3K/Akt/mTOR pathway. Metallomics. 2018;10(11):1570-1575. doi:10.1039/c8mt00183a

98. Genchi G, Lauria G, Catalano A, Sinicropi MS, Carocci A. Biological activity of selenium and its impact on human health. Int J Mol Sci. 2023;24(3):2633. doi:10.3390/ijms24032633

99. Ramya S, Shanmugasundaram T, Balagurunathan R. Biomedical potential of actinobacterially synthesized selenium nanoparticles with special reference to anti-biofilm, anti-oxidant, wound healing, cytotoxic and anti-viral activities. J Trace Elements Med Biol. 2015;32:30-39. doi:10.1016/j.jtemb.2015.05.005

100. Hendren J, Kasnyik K, Williams C, Short S. Loss of glutathione peroxidase 1 attenuates colitis and is critical in activating epithelial regenerative responses. Inflamm Bowel Dis. 2021;27(Suppl 1):S27-S27. doi:10.1093/ibd/izaa347.060

101. Handy DE, Loscalzo J. The role of glutathione peroxidase-1 in health and disease. Free Radic Biol Med. 2022;188:146-161. doi:10.1016/j.freeradbiomed.2022.06.004

102. Muller AS, Most E, Pallauf J. Effects of a supranutritional dose of selenate compared with selenite on insulin sensitivity in type II diabetic dbdb mice. J Anim Physiol Anim Nutr. 2005;89(3-6):94-104. doi:10.1111/j.1439-0396.2005.00559.x

103. Møller NP, Iversen LF, Andersen HS, McCormack JG. Protein tyrosine phosphatases (PTPs) as drug targets: inhibitors of PTP-1B for the treatment of diabetes. Curr Opin Drug Discov Devel. 2000;3(5):527-540.

Advertisement

Advertisement

Advertisement