ALL Metrics
-
Views
-
Downloads
Get PDF
Get XML
Cite
Export
Track
Review

Molecular mediators involved in skin healing: a narrative review

[version 1; peer review: 1 approved with reservations]
PUBLISHED 27 Apr 2022
Author details Author details
OPEN PEER REVIEW
REVIEWER STATUS

This article is included in the Cell & Molecular Biology gateway.

Abstract

Background: The expression of biological mediators, such as growth factors and cytokines, after skin damage, and their balance, is important to guarantee proliferation, differentiation and migration of cells and extracellular matrix, as well as to the homeostasis during tissue remodeling. The present review means to clarify their functions over inflammation, proliferation and remodeling stages of skin regeneration.
Methods: Reviews, experimental studies and clinical trials included in this paper were search on PubMed database using the following terms: platelet-derived growth factor, interleukin 1, tumor necrosis factor alpha, keratinocyte growth factor, transforming growth factor beta, endothelial vascular growth factor, matrix metalloproteinase 9, tissue metalloproteinase inhibitor 1, interferon gamma, transcription nuclear factor kappa B, skin inflammation, skin cell proliferation, skin extracellular matrix and skin regeneration.
Results and Discussion: Several studies on the signaling mechanism of these mediators in normal and impaired healing have already been developed and have shown promising results. Currently, some of these mediators are already direct focuses of potential clinical therapies that address the treatment of acute and chronic skin wounds. These mediators, basically synthetized by platelets, macrophages, vascular endothelial cells, fibroblasts and keratinocytes, act on epidermis and dermis proliferation, hypertrophy and cell migration, resulting in formation of granulation tissue, reepithelization and extracellular matrix remodeling and angiogenesis. A better understanding of the action of these mediators can provide greater knowledge not only of their roles in the natural healing process but also in the presence of disorders that affect skin repair.
Conclusion: Future studies aimed to understand the mechanism of action of these mediators in the different types of cells involved in wound healing may lead to the discovery of new therapeutics to optimize the treatment of skin pathologies.

Keywords

olecular mediators, biomarkers, inflammation, extracellular matrix, cell proliferation, skin regeneration, skin healing, wounds.

Introduction

Wound healing involves three basic phases: inflammation, proliferation and remodeling.1 Immediately after an injury, platelets are activated and degranulate releasing chemokines and growth factors (e.g. platelet-derived growth factor – PDGF), which act forming a fibrin clot and promoting local.2,3

In the inflammatory phase neutrophils, monocytes and macrophages are recruited to remove cell’s debris and possible microorganisms.13

In the proliferative phase, from 72 hours up to 2-3 weeks, there is proliferation and migration of quiescent cells (fibroblasts, keratinocytes, endothelial cells) aiming at reepithelization.3,4

Cytokines (e.g. interleukin 1 – IL-1, tumor necrosis factor alpha – TNF-α) and growth factors (e.g. keratinocyte growth factor – KGF, transforming growth factor beta – TGF-β and vascular endothelial growth factor – VEGF) are also present in this phase.2,5,6

VEGF mediates angiogenesis, ensuring nutrition to the new formed tissue, and reestablishes the extracellular matrix (ECM),5 formed by proteoglycans, collagen III, elastin and laminin.3

In the remodeling phase, which can last for months or even years, there is a gradual degradation of extracellular matrix and type III collagen, formation of type I collagen and reorganization of these collagen fibers in the dermis.3 This event is controlled by matrix metalloproteinase (MMPs) and their inhibitors (TIMPs), balancing apoptosis and new cell differentiation.2,3,7

Biological mediators, such as growth factors and cytokines, are crucial to the process of wound healing. Improving our knowledge in the role of growth factors and cytokines involved in the regulation of inflammatory, proliferative and remodeling responses during skin healing may have a significant impact on wound therapy.

Generally, these mediators were able to induce keratinocytes and fibroblast proliferation, hypertrophy and migration, resulting in a regenerated skin with a new epidermis and dermis.

Thus, this review aims to address the role of some molecular mediators, such as: PDGF, IL-1, TNF-α, KGF, TGF-β, VEGF, MMP-9, TIMP-1, IFN-γ and NF-kB active in three phases of wound healing (Figure 1).

28539a28-3a1b-4889-af3d-1e964677150a_figure1.gif

Figure 1. Figure showing the histological aspects of skin in; A) homeostasis, and during wound healing phases: B) inflammation, C) proliferation, D) remodeling. Observe the cellular skin layers: E - Epidermis, D - Dermis, H - Hipodermis (subcutaneous tissue). In B) the fibrin clot is represented in red, in C) the eschar is in brown and in D) the matrix remodeling is in green. Some molecular markers are important for skin healing and regeneration, such as: PDGF, IL-1, TNF-α, KGF, TGF-β, VEGF, MMP-9, TIMP-1, IFN-γ and NF-kB, being synthetized by specific cells (macrophages, neutrophils, keratinocytes, fibroblasts and endothelial cells). Photo by the author.

Methods

Reviews, experimental studies and clinical trials included in this paper were searched on PUBMED database using the following terms: platelet-derived growth factor (PDGF), interleukin 1 (IL-1), tumor necrosis factor alpha (TNF-α), keratinocyte growth factor (KGF), transforming growth factor beta (TGF-β), endothelial vascular growth factor (VEGF), matrix metalloproteinase 9 (MMP-9), tissue metalloproteinase inhibitor 1 (TIMP-1), interferon gamma (IFN-γ), transcription nuclear factor kappa B (NF-kB), skin inflammation, skin cell proliferation, skin extracellular matrix and skin regeneration. A total of 121 references, from 1980 to 2020, were incorporated.

Results

Platelet-derived growth factor

The role of PDGF includes all stages of healing, from the mediation of inflammation, angiogenesis, proliferation of fibroblasts and formation of granulation tissue.2

PDGF family consists of five distinct homodimeric isoforms of glycoproteins (PDGF-AA, PDGF-BB, PDGF-CC, PDGF-DD and PDGF-AB), which: PDGF-AA is synthesized by epithelial, muscle and neural cells; PDGF-BB by endothelial cells and megakaryocytes; PDGF-CC by epithelial, endothelial cells and neurons and PDGF-DD by fibroblasts and vascular smooth muscle.8

The five isoforms are also synthesized by platelets, macrophages, vascular endothelial cells, fibroblasts and keratinocytes, and has a mitogenic effects over mesenchymal cells through proteolytic reactions mediated by receptors platelet-derived growth factor alpha and beta (PDGFR-α and PDGFR-β).810

In normal skin or chronic wounds, PDGF levels are almost absent.11 However, during healing, PDGF act on fibroblast proliferation and chemotaxis9,12 mediate inflammation, angiogenesis and form the granulation tissue.8,9,13

Interleukin 1

IL-1 is a pro-inflammatory cytokine found in two forms: interleukin 1 alpha (IL-1α) and interleukin 1 beta (IL-1β). Both act on keratinocytes proliferation and migration, angiogenesis control and MECs remodeling.1416 During the inflammatory phase, IL-1 is highly produced by keratinocytes, in the epidermis15,16; high levels of IL-1α and IL-1β are related to negative changes in the prognosis of skin inflammatory and proliferative events17 and to the synthesis and regulation of other inflammatory mediators.18

Antagonists receptors recombinant human (IL-1Ra) and mouse (IL-36Ra) inhibit the expression of high levels of IL-1 and other cytokines during intense skin inflammatory and fibrotic responses.19 Inhibition of IL-1 by IL-1Ra results in a decrease in pro-inflammatory cytokines (IL-1α, IL-1β, IL-12 and IFN-γ) and increase of IL-10, M2 macrophages, endothelial cells and granulation tissue, confirming their role over cell differentiation and proliferation.20

Regarding, IL-1β Inhibition signaling pathway leads to a decrease in the pro-inflammatory M1 macrophages, an increase in the anti-inflammatory macrophages M2 and growth factors resulting in optimization of skin wounds healing.21

Tumor necrosis factor alpha

TNF-α is a pro-inflammatory cytokine synthesized by several cells (fibroblasts, keratinocytes, monocytes, macrophages, eosinophils and T cells), especially in the initial phase of healing,22 which acts on the regulation of immune, inflammatory and proliferative responses (fibroblasts and keratinocytes) through type 1 (TNFR1 - p55) and type 2 (TNFR2 - p75) receptors, which trigger internal signals in target cells, leading to NF-kB activation and these responses.23

Rapid skin healing occurred in the absence of the TNFR1-p55, with reduction of cytokines and inflammatory infiltrates, and increase of angiogenesis and collagen I24; suggesting that TNFR1-p55 can negatively affect repair by inducing leukocyte infiltration at the wound site and decreasing reepithelization.2325

Low levels of TNF-α stimulate the inflammatory process and synthesis of growth factors by macrophages, optimizing healing. However, at high levels, TNF-α impairs repair by increasing synthesis and activity of MMPs and decreasing synthesis of their inhibitors (TIMPs), which results in degradation of the ECM, inhibition of cell migration and collagen deposition and, consequent, delay in the process of healing, that is, chronification of the wound.14

The TNF-α deficiency leads to less granulation tissue formation and slower reepithelization.26

Keratinocyte growth factor

Keratinocyte growth factor is poorly synthesized in intact skin tissue but becomes highly active in fibroblasts and keratinocytes after epithelial injury. Both, KGF-1 and KGF-2, can modulate the proliferation and migration of keratinocytes, resulting in reepithelization and epidermis regeneration.27

KGF-1, highly synthesized by fibroblasts at the beginning of healing, binds to the fibroblast growth factor receptor 2 (FGFR2-IIIb) in keratinocytes, regulating migration, proliferation and differentiation.2832

KGF-2 has a mitogenic effect similar to KGF-1, but binding to the specific KGFR receptor.33,34 During wound healing, KGF also acts in the formation of granulation tissue in addition to protecting cells from toxicity induced by active oxygen species.35

Transforming growth factor beta

TGF-β family consists of the TGF-β1, TGF-β2 and TGF-β3 isoforms36,37 and is synthesized by platelets, macrophages, fibroblasts and keratinocytes, and constitute an important antiproliferative mediator to control the different stages of healing,14,37 by recruiting inflammatory cells, inhibiting proliferation and migration of endothelial cells and, consequently, angiogenesis, inhibiting fibroblasts and keratinocytes migration and ECM production (collagen and fibronectin), promoting remodeling and adequate repair of epidermis.3642

TGF-β1 is synthesized at high levels prior to reepithelialization and is related to cellular hypertrophy36,43; it can later inhibit the MMPs synthesis leading to an increase in collagen fibers.36 TGF-β3 is synthesized during the initial stage of healing and can improve repair, preventing fibroplasia.39,43

The main mechanism by which TGF-β inhibits and/or controls cell growing occurs through the interaction between both type 1 (TGF-βRI) and type 2 (TGF-βRII) receptors, promoting specific pathway signaling (TGF-β/SMAD), phosphorylation of R-Smads 2 or 3 and binding to Smad4, and, consequently, growth inhibition by regulation of target genes.42,44,45

Vascular endothelial growth factor

Neovascularization, mediated by the VEGF family, is an important event in the skin repair and can occur mediated by endothelial progenitors (EPCs).46,47

The VEGF family consists of six isoforms of pro-angiogenic glycoproteins: VEGF-A, VEGF-B, VEGF-C, VEGF-D, VEGF-E and placental growth factor (PIGF), VEGF-A121, VEGF-A165, VEGF-A189 and VEGF-A206.41,48,49

VEGF-A is the main pro-angiogenic growth factor related to wound healing,41,46,49 being expressed mainly by endothelial cells and platelets, but also by fibroblasts, keratinocytes and macrophages,50,51 once their specific tyrosine kinase 1 receptor Flt-1 (VEGFR-1) and Flk/KDR-1 (VEGFR-2) are also present in these cells.46,52,53

According to Ref. 54 an increase in angiogenesis can be mediated by VEGF-A synthesized by monocytes and macrophages in chronic venous wounds. Such a process is possible because of the interactions between endothelial and inflammatory cells.

VEGF-A increases vascular permeability in the early stages of skin repair51 and stimulates quiescent endothelial cells to interact with adjacent cells in full state of proliferation and migration, promoting vascular growth.46,55 The balance in VEGF-A levels is also important to promote normal or delayed dermis’ healing, re-epithelialization and wound contraction.46,56,57

VEGFR-2 receptor is also quite important to control angiogenesis, since binding to VEGF activates the signaling pathways of protein kinase B, resulting in inhibition of apoptosis and induction of cell proliferation.58

Matrix metalloproteinase 9

Different types of matrix metalloproteinases (MMPs), such as collagenases, gelatinases, stromelysins, are present in several tissues performing specific functions regulated by different metalloproteinase inhibitors.59,60

MMP-9 or gelatinase B, one of the 25 existing isoforms, is synthesized by basal cells and keratinocytes60,61 and has an important role in the skin healing,62 regulating the acute and chronic inflammation63,64 through the activation and control of cytokines and chemokines,65,66 controlling the release of growth factors,59 angiogenesis67 and migration/proliferation of keratinocytes68; regulating communication cells-ECM; promoting ECM reorganization and, consequently, tissue reepithelization.63,64,66

The deficiency of MMP-9 in rat skin results in an inflammatory process, with a high deposit of IL-1α and disorganization of basement membrane and ECM64; as well as the absence of MMP-9 results in decrease of angiogenesis and delay in reepithelization, with scarcity of keratinocyte migration at the edges of the wound.68,69

MMP-9 is inhibited by tissue metalloproteinase inhibitor-1 (TIMP-1) and the balance between them promotes adequate skin repair63; high levels of MMP-9 and decrease in TIMP-1 result in slow healing in wounds of diabetic rats, for example.69

Tissue inhibitor of metalloproteinase-1

TIMPs (tissue inhibitor of metalloproteinase), TIMP-1, TIMP-2, TIMP-3 and TIMP-4, regulate several cellular mechanisms, depending or not on MMPs.7072

In wound healing, TIMP-1 plays an important role in inflammation by modulating the expression of cytokines and growth factors (PDGF and TGF-β1); regulating cell differentiation, angiogenesis and apoptosis.7176 It also acts in the remodeling of the ECM, stimulating synthesis and degradation of protein.72,73

TIMP-1 increases proliferation of fibroblasts,74 protects quiescent endothelial cells75 and epithelial cells76 against TNF-α induced apoptosis through activation of the phosphatidylinositol-3-kinase (PI3K)/Akt signaling pathways and Akt mitogen-activated kinase (ERK/MAPK), respectively.

Together with Glycosylphosphatidylinositol (TIMP-1-GPI), TIMP-1 stimulates migration and proliferation of epidermal keratinocytes, reducing dermal myofibroblasts and the secretion of TGF-β, improving skin repair.76

Interferon gamma

IFN-γ is a pro-inflammatory cytokines (Type I: IFN-α and IFN-β; Type II: IFN-γ) produced by several cells, mainly by T-lymphocytes (Th1) and natural killer cells in response to viral infections and events involving inflammatory and mitogenic activities.77,78 IFN-γ responds to type 1 (IFNGR1) and type 2 (IFNGR2) receptors79 and acts by signaling the JAK1/2 STAT pathway and tyrosine kinase 2 (TYK2).80

This glycoprotein has anti-proliferative and anti-fibrotic effects, inhibiting fibroblast proliferation, collagen, cytokines IL-4 and TGF-β synthesis77,81; acts in the activation of macrophages and control the collagenase synthesis by these cells81; regulates the immune activity of stem cells of different tissues.82

The presence of IFN-γ, at high levels, can delay or impair skin repair,80,8385 by decreasing angiogenesis, synthesis, deposition, organization and stability of collagen in ECM, and controlling cell proliferation.86,87

However, reduction or absence of IFN-γ leds to early angiogenesis, by increasing VEGF and decreasing cytokine CXCL10 levels, attenuating inflammation, proliferating keratinocytes and fibroblasts, increasing expression of FGF-7, IGF and EGF, improving reepithelization, collagen deposition and synthesis of TGF-β1, accelerating, consequently, healing and wound contraction.82,88,89

Transcription nuclear factor kappa B

NF-kB family (p50/p105, p65/RelA, c-Rel/RelB and p52/p100) regulates genes involved in a variety of cellular processes, including skin healing, by activating and controlling the inflammatory response, the proliferative and migratory cellular activities, the expression of MMPs, the release and activity of growth factors, TGF-β and collagen, resulting in reepithelization9092 and ECM remodeling.93

The NF-kB can be activated through several cellular stimuli, such as the phosphorylation of the IkB kinase complex (IKK), decreasing the secretion of cytokines (TNF-α, IL-I and IL-6) in human dermal fibroblasts.93

However, the inhibition of the NF-kB signaling pathway by the IkB kinase inhibitor (IKK) complex resulted in marked tissue inflammation due to high expression of cytokines. In keratinocytes, the inhibition of NF-kB leads to skin inflammation, through TNF-α high expression, and epidermal hyperplasia.94

Discussion

Role of mediators in wound healing and re-epithelialization

Growth factors and cytokines are essential mediators to initiate the skin healing process, which starts by an inflammatory phase, followed by epidermis and dermis proliferation and remodeling.16

During the inflammatory phase keratinocytes, in the epidermis,15,16 produce IL-1. Like a positive-feedback, IL-1α and IL-1β act in the proliferation, differentiation and migration of keratinocytes,1416 resulting in reepithelization.9092

Antagonists receptors IL-1Ra and IL-36Ra, generally act by inhibiting IL-1 in the presence of intense inflammation and/or cutaneous fibrosis,19 accelerating the inflammatory phase and consequently closing the wound.95 IL-1Ra accelerated reepithelialization,96 leading to earlier skin healing in rats.95,97 IL-1 also plays an important role in this phase of healing activating neutrophils and macrophages infiltration.95

As well described by the literature, PDGFs activate some inflammatory mediators,9,12 induce8,9,13 and control98 the granulation tissue formation.

Associated to KGF-1, PDGF-BB mediate the proliferation of keratinocytes from mesenchymal cells derived from adipose tissue during healing, inducing faster reepithelialization, resulting in an epidermis similar to that found in normal skin.99

PDGF-BB is also important for fibroblasts growing and collagen deposition,100 suggesting its effectiveness in repairing acute and chronic wounds,98 promoting ECM remodeling.94 In addition, PDGF-BB acts in the oxidative balance, decreasing (3rd day) and increasing (7th day) the levels of nitric oxide,101 controlling the rate of wound contraction.102

The synthesis of KGF by fibroblasts is essential for modulating the proliferation and differentiation of keratinocytes. These cells interfere with the fibroblast response by modulating genes linked to ECM, changing phenotypes, synthesis of IL-1 and MMPs during healing. The balance between keratinocyte synthesis and apoptosis mediated by KGF is fundamental for the prevention of cutaneous fibrosis.103

Kerotinocytes and fibroblasts also activate TGF-β family,3642 VEGF and EPCs47,48,98 improving formation of granulation tissue.103

TGF-β1 contributes to epidermis and dermis thickness,104 acting as an antiproliferative mediator in the healing phases, inhibiting migratory events and controlling cellular hypertrophy, usually caused by myofibroblast clusters. The suppression of myofibroblast synthesis and consequent inhibition of smooth muscle α-actin and collagen deposition is important in the control of hypertrophic healing.105

VEGF promotes angiogenesis, increases rate of wound contraction and, consequently, early skin healing.106 Low levels of VEGF and PDGF were found both in healing and in the intact skin of mice.107 Both VEGFR-1 and VEGFR-2 receptors58 are targets for VEGF binding, which induces cell proliferation.108

During healing, the persistent presence of pro-inflammatory M1 macrophages in the granulation tissue impairs the repair, however this event can be regulated by the signaling of the VEGFR-1 receptor, which acts on the balance of the levels of pro-inflammatory M1 and anti-inflammatory macrophages M2 to promote angiogenesis.108

The activation of VEGFR-2 increases expression of IL-10, which reduces macrophages and, consequently, inflammation and differentiation of myofibroblasts in cutaneous wounds, as well as increases density of angiogenesis, improving the quality of repair.109

Other pro-inflammatory cytokines synthesis, such as TNF-α22 and NF-kB23 were activated by IL-1 and fibroblasts. TNF-α acts on the inflammatory process and on the proliferative and remodeling events of ECM during healing.14,26 Its imbalance interferes in tissue repair, since at low levels TNF-α positively modulates inflammation, inducing proliferation of keratinocytes110 and reepithelization14; at high levels TNF-α inhibits cell migration, increases MMPs synthesis and ECM degradation.14 Persistent inflammation, with M1 macrophage and neutrophil infiltration, TNF-α and TIMP-1 were observed in chronic skin wounds.111113

NF-kB also acts on inflammation, proliferation and cell migration during skin repair, through the mediation of growth factors, collagen synthesis and MMPs, promoting re-epithelialization91 and tissue remodeling.93

Keratinocytes also contribute to the regulation of TIMPs/MMPs activities, modulating ECM by inactivating MMPs, increasing TIMP-1.114 Myofibroblasts also contribute to the regulation of TIMPs/MMPs during skin repair.115

MMP-9 is involved in the inflammatory,65,66 proliferative67,68 and remodeling phases during skin healing.63,64,66 Therefore, the balance between MMP-9 synthesis and degradation by TIMP-1 is essential for adequate repair.63 Both high levels of MMP-9 and very low levels impair the migratory activity of keratinocytes and consequently the reepithelialization and wound remodeling of epidermis.116

Exogenous MMP-9 decreases collagen IV, delaying wound healing in rats, suggesting that MMP-9 interferes with the composition of basement membrane proteins, which prevent keratinocyte migration, accession and restructuring of the epidermis.117

Just like the MMP-9, TIMP-1 also acts during the healing phases by modulating events related to ECM inflammation, proliferation, migration and remodeling7376; and the balance in their expression levels contributes to the prevention of fibrosis and to a better skin repair.114

An increase in TIMP-1 and a decrease in MMP-9,115,118 improved angiogenesis, wound contraction115 and reduced inflammation.118 TIMP-1-GPI blocked the secretion of MMPs by altering the association of MMPs with the cell surface, improving the proliferation of dermal fibroblasts and reducing the expression of fibrotic genes, suggesting that this complex may help control fibrosis during cutaneous healing.119

As the proliferative phase is established, IFN-γ regulates the neutrophilic inflammatory responses120 and the immune activity of skin stem cells,82 initiating the remodeling phase,8183,8588 and the anti-proliferative and anti-fibrotic events,78,83 as inhibits the proliferation of fibroblasts and, consequently, the synthesis of collagen,120 cytokines and TGF-β.77,81 In complete absence of IFN-γ, collagen deposition and wound resistance to traction were reduced.121

Conclusion

The complete healing of a wound is essential to restore the structure and function and aesthetics of the injured tissue. Biological mediators play an important role in the healing process. A better understanding of the role of these mediators can improve knowledge not only about physiological healing, but also about tissue healing and regeneration in the presence of skin and/or systemic disorders or pathologies.

Generally, these mediators were able to induce fibroblast and keratinocytes proliferation, hypertrophy and migration, resulting in a regenerated skin with a new epidermis and dermis. Future studies aimed to understand the mechanism of action of these mediators in the different types of cells involved in wound healing may lead to the discovery of new therapeutics to optimize the treatment of skin pathologies.

Data availability

No data are associated with this article.

Comments on this article Comments (0)

Version 1
VERSION 1 PUBLISHED 27 Apr 2022
Comment
Author details Author details
Competing interests
Grant information
Copyright
Download
 
Export To
metrics
Views Downloads
F1000Research - -
PubMed Central
Data from PMC are received and updated monthly.
- -
Citations
CITE
how to cite this article
dos Santos Leite C, César Pires O, Henrique Avi P et al. Molecular mediators involved in skin healing: a narrative review [version 1; peer review: 1 approved with reservations] F1000Research 2022, 11:465 (https://doi.org/10.12688/f1000research.111159.1)
NOTE: it is important to ensure the information in square brackets after the title is included in all citations of this article.
track
receive updates on this article
Track an article to receive email alerts on any updates to this article.

Open Peer Review

Current Reviewer Status: ?
Key to Reviewer Statuses VIEW
ApprovedThe paper is scientifically sound in its current form and only minor, if any, improvements are suggested
Approved with reservations A number of small changes, sometimes more significant revisions are required to address specific details and improve the papers academic merit.
Not approvedFundamental flaws in the paper seriously undermine the findings and conclusions
Version 1
VERSION 1
PUBLISHED 27 Apr 2022
Views
12
Cite
Reviewer Report 06 Oct 2022
David Leavesley, Wound Care Innovation for the Tropics (WCIT) Programme, Skin Research Institute of Singapore, Agency for Science, Technology and Research, Singapore, Singapore;  Innovations in Food and Chemical Safety (IFCS) Programme, Agency for Science, Technology and Research, Singapore, Singapore;  School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, NSW, Australia 
Approved with Reservations
VIEWS 12
The manuscript by dos Santos Leite et al, presents a review of the “expression of biological mediators, such as growth factors and cytokines, after skin damage, and their balance… to guarantee proliferation, differentiation and migration of cells and extracellular matrix, ... Continue reading
CITE
CITE
HOW TO CITE THIS REPORT
Leavesley D. Reviewer Report For: Molecular mediators involved in skin healing: a narrative review [version 1; peer review: 1 approved with reservations]. F1000Research 2022, 11:465 (https://doi.org/10.5256/f1000research.122851.r149095)
NOTE: it is important to ensure the information in square brackets after the title is included in all citations of this article.

Comments on this article Comments (0)

Version 1
VERSION 1 PUBLISHED 27 Apr 2022
Comment
Alongside their report, reviewers assign a status to the article:
Approved - the paper is scientifically sound in its current form and only minor, if any, improvements are suggested
Approved with reservations - A number of small changes, sometimes more significant revisions are required to address specific details and improve the papers academic merit.
Not approved - fundamental flaws in the paper seriously undermine the findings and conclusions
Sign In
If you've forgotten your password, please enter your email address below and we'll send you instructions on how to reset your password.

The email address should be the one you originally registered with F1000.

Email address not valid, please try again

You registered with F1000 via Google, so we cannot reset your password.

To sign in, please click here.

If you still need help with your Google account password, please click here.

You registered with F1000 via Facebook, so we cannot reset your password.

To sign in, please click here.

If you still need help with your Facebook account password, please click here.

Code not correct, please try again
Email us for further assistance.
Server error, please try again.