Next Article in Journal
Association between Proton Pump Inhibitors and Hearing Impairment: A Nested Case-Control Study
Next Article in Special Issue
Histological Evidence of Wound Healing Improvement in Rats Treated with Oral Administration of Hydroalcoholic Extract of Vitis labrusca
Previous Article in Journal
Immunomodulatory Expression of Cathelicidins Peptides in Pulp Inflammation and Regeneration: An Update
Previous Article in Special Issue
Vitamin D Deficiency Reduces Vascular Reactivity of Coronary Arterioles in Male Rats
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Molecular Changes Underlying Genistein Treatment of Wound Healing: A Review

1
Department of Pharmacology, Faculty of Medicine, Pavol Jozef Šafárik University, 040 11 Košice, Slovakia
2
Department of Biomedical Research, East-Slovak Institute of Cardiovascular Diseases, Inc., 040 11 Košice, Slovakia
3
Department of Pharmacognosy and Botany, Faculty of Pharmacy, Comenius University, 832 32 Bratislava, Slovakia
4
Laboratory of Cell Interactions, Faculty of Medicine, Pavol Jozef Šafárik University, 040 11 Košice, Slovakia
5
Prague Burn Center, Third Faculty of Medicine, Charles University, 100 34 Prague, Czech Republic
*
Author to whom correspondence should be addressed.
Curr. Issues Mol. Biol. 2021, 43(1), 127-141; https://doi.org/10.3390/cimb43010011
Submission received: 5 May 2021 / Revised: 12 May 2021 / Accepted: 16 May 2021 / Published: 17 May 2021

Abstract

:
Estrogen deprivation is one of the major factors responsible for many age-related processes including poor wound healing in postmenopausal women. However, the reported side-effects of estrogen replacement therapy (ERT) have precluded broad clinical administration. Therefore, selective estrogen receptor modulators (SERMs) have been developed to overcome the detrimental side effects of ERT on breast and/or uterine tissues. The use of natural products isolated from plants (e.g., soy) may represent a promising source of biologically active compounds (e.g., genistein) as efficient alternatives to conventional treatment. Genistein as natural SERM has the unique ability to selectively act as agonist or antagonist in a tissue-specific manner, i.e., it improves skin repair and simultaneously exerts anti-cancer and chemopreventive properties. Hence, we present here a wound healing phases-based review of the most studied naturally occurring SERM.

1. Introduction

Improved healthcare in developed countries has led to an increase in the size of the elderly population. Therefore, almost one third of a woman’s life occurs in the post-menopausal period. Estrogen deprivation is one of the major factors responsible for many age-related processes including poor wound healing in postmenopausal women [1]. Wound healing is a dynamic event running in four basic phases, i.e., hemostasis, inflammation, proliferation, and maturation/remodeling, which overlap with each other in a consecutive manner restoring the disrupted integrity of tissues/organs. Immediately following blood clotting inflammatory/immune cells enter the injury site to eliminate pathogens and remove necrotic tissue [2]. Fibroblasts located in the granulation tissue excrete and remodel extracellular matrix (ECM) proteins, whereas differentiated myofibroblasts contract the wound [3]. In addition, endothelial cells create new vessels and provide nutritional support for the newly formed tissue [4]. To create a mechanical barrier between the outer and inner environment, keratinocytes migrate across the wound (over the granulation tissue) restoring the integrity of the organism [5].
Previously, it was shown that women receiving estrogen replacement therapy (ERT) have a lower incidence of chronic wounds [6,7]. Moreover, improved skin wound healing was also observed following topical application of estrogen [8]. In this clinical study ERT was shown to significantly reduce wound size, increase collagen deposition, and improve wound stiffness. Further beneficial effects of ERT in elderly women involve reduced risk of bone fracture, colorectal cancer, and incidence of menopausal symptoms [9,10]. However, reported side-effects of estradiol substitution have precluded a common clinical introduction of ERT. Accordingly, selective estrogen receptor modulators (SERMs) were studied to overcome the detrimental side effects of ERT on breast and/or uterine tissues [11,12]. A renewed strategy directed towards the use of natural products isolated from plants (e.g., soy) might represent a promising source of biologically active compounds (e.g., genistein) as efficient alternatives to conventional treatment. Hence, we present here a systematical, wound healing phases-based, review of the most studied naturally occurring SERM genistein.

2. Methods—Literature Search Strategy

The effects of genistein on wound healing were searched on PubMed using the term “genistein wound healing” for relevant English-written papers (Available online: https://pubmed.ncbi.nlm.nih.gov/?term=genistein+wound+healing&sort=date, accessed on 31 March 2021), the search identified 84 articles. The present paper reviews the most relevant studies and includes the appropriate references.

3. (Phyto)Estrogen Signaling in Wound Healing and Skin Aging

Menopause-induced estrogen deficiency is accompanied by changes in skin morphology and physiology involving decrease in thickness, collagen, and water content as well as loss of elasticity and increase in fragility [13,14,15]. Lower estrogen levels associated with the menopause also result in impaired wound healing. In particular, the negative effect of the menopause on wound healing results from fragile skin that tears and bruises easily and is thus susceptible to trauma [16].
Phytoestrogens are non-steroidal compounds of plant origin structurally and functionally similar to human estrogen [17]. Depending on endogenous estrogen levels and estrogen receptors (ERs) expression, phytoestrogens may act (tissue dependent) as agonists and/or antagonists, thus presenting a subclass of SERMs [18]. Genistein (4′,5,7-trihydroxyisoflavone) is a naturally occurring phytoestrogen (commonly found in soy products including soy beans except for soy sauce [19]) and the most studied natural SERM. The most prevalent form of genistein is the biologically active genistein-7β-glucoside, which on digestion or fermentation leads to release of the aglycone form [20]. Although well absorbed in the small intestine, genistein has low oral bioavailability due to low water-solubility, extensive metabolism, and high expression level of efflux transporters, especially breast cancer resistance protein (BCRP) [21,22,23]. Structurally genistein resembles 17β-estradiol, mostly due to the similar position and distance between the OH groups, crucial for the binding ability of both phytoestrogens and estrogens towards ERs. However, genistein has a higher binding affinity for ER-β than ER-α resulting in functional diversity between both molecules [24]. ER-β is predominantly expressed in the epidermis, dermal fibroblasts, blood vessels, and hair follicles and represents the main mediator of estrogen action in the skin [25,26]. Pharmacological effects of genistein are mediated through competitive agonizing/antagonizing of ERs. Apart from genomic estrogen signaling, several non-ER effects of genistein have been described including activation of G protein-coupled receptor 30 (GPR30) [27] and inhibition of protein-tyrosine kinases (PTK) [28,29], topoisomerase II [30], platelet-derived growth factor (PDGF) [31], and epidermal growth factor (EGF)-induced c-Fos expression, as well as inhibition of diacylglycerol synthesis [32] and angiogenesis [33,34]. Additionally, genistein modulates various features of cancer including cell cycle, apoptosis, angiogenesis, and metastasis [35]. Intriguingly, activity of genistein revealed a concentration dependent pattern where higher concentrations inhibited the activity of PTK associated with the EGF receptor, while lower concentrations triggered estrogenic activity [36,37].

4. Inflammatory Phase and Oxidative Stress

Inflammation is a crucial process involved in wound healing. During the inflammatory phase the innate immune response is activated leading to the recruitment of immune cells to the injury site and removal of invading pathogens and dead cells/tissues [38]. The imbalance between pro-inflammatory and anti-inflammatory signaling has a crucial influence on the morphological and functional outcome of scarring [39]. In particular, prolonged/chronic inflammation may cause deregulation of keratinocyte and fibroblast differentiation/activation which may in turn lead to disturbance in the wound healing process. frequently resulting in excessive scar formation [40]. Intriguingly, despite inappropriately excessive inflammatory response associated with age-related delayed wound healing [41], the menopause is rather associated with reduced scarring and ECM deposition [13,42]. The effect of genistein on inflammatory phase of wound healing and oxidative stress is shown in Figure 1.
In particular, macrophages are of high importance in wound healing as they attenuate inflammation, clear cell debris, as well as coordinate tissue repair. Genistein can inhibit activation of macrophages exposed to inflammatory stimulus by inhibiting activation of transcription factor nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB), as well as signal transducer and activator of transcription 1 (STAT-1) [43]. In detail, transcription factor NF-κB controls the expression of inducible chemokines, cell adhesion molecules, vasoactive proteins, and anti-apoptotic proteins important for cellular stress response. The NF-κB signaling pathway is triggered by inflammatory activators such as UV light, cytokines, free radicals, oxidized LDL (ox-LDL,) and viral antigens [44]. One part of the innate immune response regulated by NF-κB is NOD-like receptor protein-3 (NLRP3) inflammasome. NLRP3 inflammasome is reported to have an important role in promoting the early stage of skin wound healing [45]. Genistein supplementation had a positive effect on NLRP3 expression which was reduced by diabetes in mice [46]. Apart from in vitro data, the accelerated wound closure rate observed in the early stages of healing was accompanied by decreased levels of inflammatory markers TNF-α and NF-κB in genistein treated mice [46,47]. Furthermore, an in vivo study showed that genistein treatment can shift macrophage phenotype from M1 to M2 in rats with experimental colitis [48]. The switch from pro-inflammatory M1 phenotype to inflammation-resolving M2 phenotype in macrophages is crucial for the transition from the inflammatory to the proliferation phase of wound healing [40]. Genistein was also able to inhibit inducible nitric oxide synthase (iNOS) activity and production of cytokines, promoting inflammation such as tumor necrosis factor alpha (TNF-α), interleukin (IL)-6 and IL-1β in lipopolysaccharide (LPS)-stimulated macrophages [48].
Furthermore, genistein prevented LPS-induced decrease in adenosine monophosphate-activated protein kinase (AMPK) phosphorylation [49]. Of note, a novel molecule synthesized using genistein, 7-difluoromethoxy-5,4′-dimethoxy-genistein (DFMG), was able to attenuate macrophage activation induced by co-culture with lysophosphatidylcholine (LPC)-injured human umbilical vein endothelial cells (HUVE-12) cells, via inhibition of NF-κB through the signaling pathway involving toll like receptor 4 (TLR4) and myeloid differentiation factor 88 (MyD88) [50]. Impact of gamma irradiation, a common food preserving procedure, was also studied in soy bean compounds including genistein [51]. The experiment revealed that the bioactivity of genistein through gamma-irradiation does not increase macrophage cytotoxicity. Moreover, gamma-irradiated genistein exerts anti-inflammatory action through inhibition of mitogen-activated protein kinase (MAPK) and NF-κB signaling pathways [52].
Apart from macrophages, genistein can ameliorate the immune response of granulocytes and T cells in vivo. In detail, the cell-mediated immune response in ovariectomized (OVX) mice was tested by monitoring the delayed-type hypersensitivity (DTH). Treatment of OVX mice with 8–80 mg/kg genistein decreased DTH by 46–67%. Furthermore, histopathological examination revealed reduced cell-infiltration and lower numbers of CD4+ and CD8+ T cells in lymph nodes [53]. A similar suppressive effect on DTH was observed in non-OVX mice fed with genistein (30 mg/kg) where the granulocyte dependent inflammatory response was also ameliorated [54].
Moreover, genistein affected secretion of cytokines, growth factors, and other signaling molecules produced by non-inflammatory cells with considerable impact on the inflammation associated with the healing process. For example, in human umbilical vein endothelial cells (HUVECs), genistein attenuated the expression of pro-inflammatory mediators NF-κB, E-selectin, P-selectin, monocyte chemoattractant protein-1 (MCP-1), IL-8, Vascular cell adhesion protein 1 (VCAM-1), and Intercellular adhesion molecule 1 (ICAM-1) triggered by ox-LDL [55]. Pretreatment of HUVECs with genistein prior to exposition to inflammatory stimulus lowered the induced expression of pro-inflammatory mediators, such as TNF-α, IL-1β, MCP-1, IL-8, and ICAM-1 on both mRNA and protein levels [56]. A similar anti-inflammatory effect was observed in keratinocytes stimulated with TNF-α where genistein treatment resulted in the inhibition of NF-κB nuclear translocation and expression of inflammatory cytokines (TNF-α, IL-6 and IL-23) [57].
Genistein treatment improved pathological scores of cutaneous skin lesions in mice and impaired expression of proinflammatory factors IL-1β, IL-6, TNF-α, C-C motif chemokine ligand 2 (CCL2), IL-17, and IL-23 induced by imiquimod [57]. Notably, the anti-inflammatory effect of genistein may also be related to its antioxidative properties. It was shown that treatment of human keratinocytes and fibroblasts with genistein could prevent skin aging via modulation of glutathione (GSH) content and reactive oxygen species (ROS) release, endothelial/inducible (e/i)NOS dependent NO release, matrix metalloproteinases (MMPs) expression, and mitochondria membrane potential through mechanisms involving p38 MAPK, Akt and extracellular signal-regulated kinases 1/2 (ERK1/2) as downstream signaling associated with ERs and GPR30 [58], as well as by increasing superoxide dismutase (SOD) activity and B-cell lymphoma 2 (Bcl-2) expression in endothelial cells [59,60]. Importantly, as was shown in an in vivo study where OVX mice were co-treated with ER antagonist, the ameliorative effect of genistein on the inflammatory stage of wound healing seems to be independent of ER-mediated signaling. However, genistein treatment affected the next step of the healing process through ER-signaling as accelerated re-epithelization induced by genistein treatment was partially reversed by ER antagonist (ICI 182,780), but had little impact on the anti-inflammatory effect of genistein [61].
As shown in a mice model, genistein decreases expression of Cu/Zn-SOD and Mn-SOD to a low level, which was shown to be sufficient for coping with oxidative stress in the early stages of wound healing [47]. Furthermore, genistein was shown to be able to modulate the antioxidant defense system impaired by diabetic condition in mice and restored its function in the early stages of wound healing [46]. Additionally, genistein reduced oxidative stress in diabetic mice through suppression of iNOS and forkhead box O transcription factor 1 (FoxO1) activity. This resulted not only in alleviation of delayed wound healing but also in improved angiogenesis [62]. Protection against oxidative stress in skin was also observed in male rats fed with an isoflavone mixture of genistein and daidzein (2 or 20 mg/kg). The isoflavone mixture-treated group showed significantly greater thickness of the skin epidermis as well as higher amounts of collagen and elastic fibers in the dermis. Skin homogenates of the treated group showed a decrease in catalase activity and inhibited lipid peroxide formation in a dose dependent manner [63].

5. Proliferation Phase

The proliferative phase of wound repair involves formation of the granulation tissue including fibroblast proliferation, ECM deposition, angiogenesis, and wound re-epithelization [64]. In particular, several menopause-associated fibroblast malfunctions were reported in elderly women. The underlying mechanisms include microRNA-7 up-regulation, impaired function of EGF receptor, hyaluronan synthase 2, resulting in Januse kinase (JAK)/STAT1 over-activation [65]. In this context estrogen down-regulates microRNA-7 expression which attenuates STAT1 and also induces a rapid re-organization of the cytoskeleton in dermal fibroblast via the non-genomic GPR30 axis [66,67]. The effect of genistein on proliferation phase of wound healing is shown in Figure 2.

5.1. Fibroblasts

Dermal fibroblasts can transiently express α-smooth muscle actin (SMA) to obtain a myofibroblast-like phenotype [68]. Myofibroblasts play crucial role in ECM synthesis and remodeling, thus precise regulation of fibroblast-to-myofibroblast differentiation, e.g., with the main driving source molecule TGF-β1, is essential for proper wound closure [69]. Importantly, absence of initial TGF-β1 stimulus resulted in increased myofibroblast apoptosis. Interestingly, if initial TGF-β1 stimulus is exchanged to IL-10, the motility of fibroblasts increased and myofibroblasts number decreased [70]. Intriguingly, the crosstalk between ER and TGF-β1 signaling was reported [71,72]. In detail, ER signaling suppresses TGF-β1-induced activation of Sma and MAD-related protein 3 (SMAD3), whereas TGF-β1 signaling stimulates ER-mediated transcriptional activation [73]. From this point of view the question of whether genistein treatment interacts with TGF-β1-induced fibroblast-to-myofibroblast differentiation should be explored in further research.
Genistein was shown to be able to influence collagen production in human dermal fibroblasts under oxidative stress conditions induced by t-BHP (t-butylhydroperoxide). Oxidative stress diminished collagen synthesis in fibroblasts and treatment of cells with genistein exerted a biphasic effect on collagen expression. Low concentration (1 µM) restored the decreased collagen expression whereas high concentration (100 µM) enhanced the inhibitory action of t-BHP. Protective action of the lower tested genistein concentration (1 µM) was mediated via modulation of the insulin-like growth factor 1 (IGF-1) receptor expression and ERK1/2 associated pathway [74].
In addition, genistein may also improve treatment of non-healing leg ulcers, typical of increased proteolytic activity and strong expression of MMPs, urokinase-type plasminogen activator (uPA), and extracellular MMP inducer [75]. As shown in human gingival fibroblasts, genistein was able to inhibit basal uPA activity. Moreover, genistein was also able to inhibit EGF-stimulated uPA production. In detail, genistein inhibited phosphorylation of the EGF receptor following stimulation inhibiting EGF-related activation of JNK and ERK1/2 [76].

5.2. Angiogeneis

Angiogenesis is the formation of new blood capillaries from existing vasculature through sprouting. This process is crucial in physiological as well as pathological conditions e.g., wound repair, inflammation, invasion, and tumor metastasis [77]. In the process of wound repair, angiogenesis is essential for the delivery of oxygen and nutrients to the newly developed tissue. Angiogenesis is comprised of consecutive actions including migration and proliferation of endothelial cells orchestrated by growth factors, oxygen levels, and proteases [2]. Recently, it was revealed that genistein can impact the growth and budding of endothelial cells, the formation of new capillaries, and some of the signaling pathways connected to angiogenesis [78] also via alteration of vascular endothelial growth factor (VEGF). VEGF is one of the most important regulators of angiogenesis, of which inhibition leads to significant decrease in blood vessel formation [79]. Importantly, the effect of genistein on VEGF expression is dose and cell type dependent [33,80,81,82,83].
In HUVECs, genistein decreased both basal and hypoxia stimulated VEGF and VEGF receptor (VEGFR) expressions on both protein [83] and mRNA [84] levels. In addition, genistein was able to inhibit VEGF-induced HUVEC proliferation at 10 µM concentration [83]. In detail, genistein treatment prevents HUVEC activation by VEGF through activation of MAPK and inhibition of PTK signaling, resulting in decreased production and activity of MMP-2 and MMP-9 as well as decreased activation of c-Jun N-terminal kinase (JNK) and p38 [33]. Furthermore, cell pre-treatment with genistein blocks the VEGF/bFGF induced MMP-1 and uPA expression and activation of MMP-2 through modulation of their inhibitor expression TIMP-1, TIMP-2, plasminogen activator inhibitor 1 (PAI-1), endostatin, angiostatin, and thrombospondin [85,86]. Of note, MMPs support angiogenesis by degrading the components of ECM, promoting endothelial cell invasion and sprouting [87].
However, genistein treatment of HUVECs showed a bivalent dose-dependent effect on in vitro endothelial tube formation. At low concentration (0.001–1 µM), genistein was able to stimulate tube-like structure sprouting, while in contrast at higher tested concentration (25–100 µM) genistein hindered angiogenesis [88]. Interestingly, treatment with 1 µM genistein averted the loss of tubule network impaired by high glucose concentration, whereas lower genistein concentration (0.1 µM) did not manifest such an effect [62].
Furthermore, genistein modulated angiogenesis by alteration of proangiogenic cytokines and protease expressions. In low doses (0.01–50 µM), genistein was shown to exert a positive effect on the secretion of bFGF, EGF, angiogenin, angiopoietin-2, MMP-9, and uPA receptor in HUVECs [88]. Interestingly, co-treatment of endothelial cells with VEGF and genistein at low concentrations (100 nM) showed a synergic effect on the up-regulation of six angiogenesis promoting genes—MMP14, VEGF-A, CTGF, C-X-C motif chemokine 5 (CXCL5), IL-6 and integrin β3 (ITGB3)—as well as three angiogenesis inhibiting gene expressions—Collagen Type XVIII Alpha 1 Chain (COL18A1), Tissue inhibitor of metalloproteinases (TIMP)-2, and TIMP-3 [59]. Therefore, we also used Western blot to confirm changes in gene expressions on the protein level. Our experiment revealed that out of nine dysregulated genes on the mRNA level, six (VEGF, CTGF, CXCL5, Integrin β3, TIMP2 and COL18A; TIMP3 not performed) were deregulated also on the protein level (Figure 3).
Of note, comparison of wound biopsies from OVX and sham operated rats showed a decrease in TGF-β1, VEGF, MMP-2, MMP-9, TIMP-1, and TIMP-2 levels after ovariectomy. Treatment of OVX rats with 1 mg/kg of genistein aglycone significantly restored expression of these proteins whilst lower doses of genistein were shown to increase collagen layer thickness and skin breaking strength [89]. Furthermore, genistein was shown to be able to restore VEGF and TGF-1β, while tissue transglutaminase 2 (TG2) expression decreased due to estrogen deficiency. Intriguingly, genistein (at lower tested concentrations) was able to exert a greater effect than 17-α-ethinyl oestradiol [90].
It was also demonstrated that genistein can impact angiogenesis by affecting migration and adhesion of endothelial cells. Impairment of cell–cell adhesion was observed after treatment of HUVECs with 10 µM concentration of genistein. Limited adhesion was accompanied by down-regulation of mRNA and the protein expression of cell adhesion related genes including VE-cadherin, gap junction protein alpha 1 (connexin 43), integrin alpha V, and multimerin [91]. Furthermore, genistein treatment affected expression of additional proteins linked to adhesion and pro-inflammatory proteins—monocyte chemoattractant protein 1 (MCP-1) and ICAM-1 in brain microvascular endothelial cells (BMECs) [92]. Interestingly, genistein (0.1–5 µM) was shown to inhibit TNF-α-induced endothelial inflammation by decreasing production of adhesion molecules and chemokines such as sICAM-1, sVCAM-1, sE-selectin, MCP-1, and IL-8 [93]. Additionally, genistein also known as a non-selective tyrosine kinase inhibitor is able to decrease migration of endothelial cells by disrupting FAK/paxillin signaling [94].

5.3. Epidermis Regeneration

Wound re-epithelization is a crucial process in rebuilding the mechanical barrier between the outer and inner environments. Estrogen also imparts a potent mitogenic effect on keratinocytes, promoting in vitro and in vivo migration [95], which might be affected by the estrogen-mediated ER-β interaction with keratinocytes. ER-β led to an increase in cell proliferation and keratin-19 expression, as well as a decrease in galectin-1 expression. Fittingly, in rat wounds treated with the ER-β agonist, epidermal regeneration was accelerated. In the present study, we provide information on the nuclear ER-β through which selective estrogen receptor agonists affect the expression pattern of selected markers, thus modulating keratinocyte proliferation (increased Ki67 expression) and differentiation (increased keratin 19 expression) [96]. In addition, we demonstrated that the pharmacological activation of ER-α and -β has a different impact on wound healing [96,97].
Genistein among other phytoestrogen-containing extracts modulated the differentiation pattern of keratinocytes (HaCaT) and estrogen positive breast cancer cells (MCF-7). Interestingly, phytoestrogen treatment was shown to increase the expression of luminal marker keratin-8 in MCF-7, but not in HaCaT cells, whereas the expression of other keratins (i.e., 14 and 19) remained rather stable in both used cell lines [98]. Furthermore, estrogen accelerated skin wound healing by promoting keratinocyte proliferation via non-genomic ERK/Akt signaling [99]. Genistein was shown to stimulate hyaluronic acid production in keratinocytes culture in vitro and in vivo [100].

6. Maturation and Remodeling Phase

It is well known that abnormalities in wound repair range from non-healing wounds to excessive fibrosis and scarring. Excessive deposition of ECM components and collagens during the maturation/remodeling phase of wound healing may lead to development of hypertrophic scars or keloids. In comparison to hypertrophic scars, which do not grow beyond the boundaries of the wound site, keloids often grow beyond the original extent of the wound. However, both hypertrophic scars and keloids may cause physiological and psychological discomfort [101]. Hypertrophic scar, a more common form of excessive scarring, is often a result of surgical procedures, trauma, radiation, or burn injuries. Although, the exact mechanism of keloid and hypertrophic scar development is not known, various cytokines have been shown to be implicated including IL-6, IL-8, IL-10, and TGF-β as well as various growth factors including for example PDGF [101]. The effect of genistein on maturation phase of wound healing is shown in Figure 2.
In hypertrophic scars, genistein at high concentration (100 µM) was shown to be capable of arresting fibroblast proliferation and suppressing collagen production. On the contrary, normal skin fibroblasts were not affected, which indicates action specificity. In detail, the anti-proliferative effect of genistein was mediated through inhibition of Rat sarcoma (Ras), Rapidly accelerated fibrosarcoma (Raf), ERK, and p38 proteins involved in the MAPK/ERK signaling pathway [28]. Furthermore, in rat cardiac fibroblasts, genistein was shown to inhibit proliferation through GPR30 signaling by suppression of the cell cycle proteins, cyclin B1, and cyclin-dependent kinase 1(CDK1) [27].
Abnormal scarring can also result in keloid formation, characterized by imbalanced ECM synthesis and degradation. One of the features observed in keloids is abnormal activity of transcription factor AP-1 (activator protein 1) connected with disturbed expression of its subunits (c-Fos, c-Jun). In cooperation with other transcription factors, AP-1 induces expression of MMPs and regulates expression of growth factors and cytokines [102]. The expression of C-JUN was higher in keratinocytes treated with both tested concentrations of genistein (37 and 370 µM). On the other hand, in fibroblasts, genistein regulated C-JUN expression in a dose-dependent manner. In normal dermal fibroblasts a lower tested concentration of genistein resulted in higher C-JUN expression whereas in keloid fibroblasts C-JUN expression was more increased at the higher tested concentration of genistein. Genistein at higher tested concentration inhibited C-FOS expression in normal fibroblasts, but the expression of C-FOS was stimulated in keloid fibroblasts in a concentration dependent manner. Of note, genistein, at 370 and 37 µM, inhibited FOS-B expression in keratinocytes [103].
When compared to normal fibroblasts, keloid fibroblasts express higher amounts of connective tissue growth factor (CTGF) on mRNA and protein levels. Genistein was shown to be able to decrease CTGF expression on both levels in a dose dependent manner. Moreover, genistein was also capable of attenuating TGFβ1, β2, and β3 gene expression. Interestingly, the cytoprotective effect of genistein was revealed due to Bcl-2 gene expression stimulation [104]. Furthermore, it was also described in HMVEC-d cells, that the genistein induced Bcl-2 production is mediated rather via ER-β [60].

7. Discussion and Conclusions

Better understanding of the biological processes following genistein treatment are inevitable for potential/novel phytoestrogen-based pharmacological manipulations of wound repair. The present data summarize the effects of genistein (Table 1) as natural SERM that has the unique ability to selectively act as agonist or antagonist in a tissue-specific manner. Therefore, genistein improves skin repair [47,90] and simultaneously exerts anti-cancer [105] and chemopreventive properties [106]. As a result several clinical trials combining genistein and/or its analogues with conventional chemotherapeutics have been conducted and point to the safety and efficiency of this drug [107,108]. Nevertheless, further research, including the discovery of the exact underlying mechanisms and optimal wound-type specific management protocol, needs to be performed so that these findings may be applied in practice. Therefore, translation of the reviewed phytoestrogen and estrogen-like compound-mediated signaling to a clinical setting remains a challenge for further research.

Author Contributions

Conceptualization, M.Č. and P.G.; literature search, M.Č., V.L. and P.M.; writing—original draft preparation, M.Č., V.L. and P.G.; supervision, P.G.; funding acquisition, P.G. All authors have read and agreed to the published version of the manuscript.

Funding

Data shown in this review were obtained with the support of the Slovak Research and Development Agency under contract No. APVV-16-0207.

Informed Consent Statement

Not applicable.

Data Availability Statement

Adult human dermal microvascular endothelial cells (HMVEC-d) were obtained from Lonza (Lonza Walkersville, Inc., Walkersville, MD, USA). Detailed description of used methods can be provided by authors on request.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

AMPKAdenosine monophosphate-activated protein kinase
AP-1Activator protein 1
BCRPBreast cancer resistance protein
bFGFBasic fibroblast growth factor
Bcl-2B-cell lymphoma 2
BMECsBrain microvascular endothelial cells
CCLC-C motif chemokine
CDCluster of differentiation
COL18A1Collagen Type XVIII Alpha 1 Chain
CTGFConnective tissue growth factor
CXCLC-X-C motif chemokine ligand
ECMExtracellular matrix
EGFEpidermal growth factor
eNOSEndothelial nitric oxide synthase
EREstrogen receptor
ERKSignal-regulated kinase
ERTEstrogen replacement therapy
FoxO1Orkhead box O transcription factor 1
GPR30G protein-coupled receptor 30
GSHGlutathione
GTGranulation tissue
HaCaTHuman keratinocyte cell line
HMVEC-dHuman dermal microvascular vein endothelial cells
HUVECHuman umbilical vein endothelial cell
ICAM-1Intercellular adhesion molecule 1
IGFInsulin growth factor
ILInterleukin
iNOSInducible nitric oxide synthase
ITGB3Integrin β3
JAKJanuse kinase
LPCLysophosphatidylcholine
LPSLipopolysaccharide
MAPKMitogen-activated protein kinase
MCF-7Michigan Cancer Foundation-7
MCP-1Monocyte chemoattractant protein-1
MMPMatrix metalloproteinase
MyD88Myeloid differentiation factor 88
NF-κBNuclear factor kappa-light-chain-enhancer of activated B cells
NLRP3NOD-like receptor protein-3
OVXOvariectomized
ox-LDLOxidized low density lipoprotein
PDGFPlatelet-derived growth factor
PTKProtein tyrosine kinase
ROSReactive oxygen species
SMASmooth muscle actin
SMADSma and MAD-related protein 3
SERMSelective estrogen receptor modulator
STAT-1Signal transducer and activator of transcription 1
TIMPTissue inhibitor of matrix metalloproteinase
TG2Transglutaminase 2
TGFTransforming growth factor
TNFTumor necrosis factor
t-BHPt-Butylhydroperoxide
uPAUrokinase-type plasminogen activator
VEGFVascular endothelial growth factor
VEGFRVascular endothelial growth factor receptor

References

  1. Hall, G.; Phillips, T.J. Estrogen and skin: The effects of estrogen, menopause, and hormone replacement therapy on the skin. J. Am. Acad. Derm. 2005, 53, 555–568. [Google Scholar] [CrossRef]
  2. Coma, M.; Frohlichova, L.; Urban, L.; Zajicek, R.; Urban, T.; Szabo, P.; Novak, S.; Fetissov, V.; Dvorankova, B.; Smetana, K., Jr.; et al. Molecular changes underlying hypertrophic scarring following burns involve specific deregulations at all wound healing stages (inflammation, proliferation and maturation). Int. J. Mol. Sci. 2021, 22, 897. [Google Scholar] [CrossRef]
  3. Pakshir, P.; Noskovicova, N.; Lodyga, M.; Son, D.O.; Schuster, R.; Goodwin, A.; Karvonen, H.; Hinz, B. The myofibroblast at a glance. J. Cell Sci. 2020, 133. [Google Scholar] [CrossRef] [PubMed]
  4. Guerra, A.; Belinha, J.; Jorge, R.N. Modelling skin wound healing angiogenesis: A review. J. Theor. Biol. 2018, 459, 1–17. [Google Scholar] [CrossRef] [PubMed]
  5. Muse, M.E.; Crane, J.S. Physiology, epithelialization. In Statpearls; StatPearls Publishing: Treasure Island, FL, USA, 2021. [Google Scholar]
  6. Kovacs, E.J. Aging, traumatic injury, and estrogen treatment. Exp. Gerontol. 2005, 40, 549–555. [Google Scholar] [CrossRef] [PubMed]
  7. Margolis, D.J.; Knauss, J.; Bilker, W. Hormone replacement therapy and prevention of pressure ulcers and venous leg ulcers. Lancet 2002, 359, 675–677. [Google Scholar] [CrossRef]
  8. Ashcroft, G.S.; Greenwell-Wild, T.; Horan, M.A.; Wahl, S.M.; Ferguson, M.W. Topical estrogen accelerates cutaneous wound healing in aged humans associated with an altered inflammatory response. Am. J. Pathol. 1999, 155, 1137–1146. [Google Scholar] [CrossRef] [Green Version]
  9. Warren, M.P. A comparative review of the risks and benefits of hormone replacement therapy regimens. Am. J. Obstet. Gynecol. 2004, 190, 1141–1167. [Google Scholar] [CrossRef] [PubMed]
  10. Pataky, M.W.; Young, W.F.; Nair, K.S. Hormonal and metabolic changes of aging and the influence of lifestyle modifications. Mayo Clin. Proc. 2021, 96, 788–814. [Google Scholar] [CrossRef]
  11. Kelly, P.M.; Keely, N.O.; Bright, S.A.; Yassin, B.; Ana, G.; Fayne, D.; Zisterer, D.M.; Meegan, M.J. Novel selective estrogen receptor ligand conjugates incorporating endoxifen-combretastatin and cyclofenil-combretastatin hybrid scaffolds: Synthesis and biochemical evaluation. Molecules 2017, 22, 1440. [Google Scholar] [CrossRef]
  12. Cassidy, A. Potential risks and benefits of phytoestrogen-rich diets. Int. J. Vitam. Nutr. Res. 2003, 73, 120–126. [Google Scholar] [CrossRef] [PubMed]
  13. Affinito, P.; Palomba, S.; Sorrentino, C.; Di Carlo, C.; Bifulco, G.; Arienzo, M.P.; Nappi, C. Effects of postmenopausal hypoestrogenism on skin collagen. Maturitas 1999, 33, 239–247. [Google Scholar] [CrossRef]
  14. Brincat, M.; Moniz, C.J.; Studd, J.W.; Darby, A.; Magos, A.; Emburey, G.; Versi, E. Long-term effects of the menopause and sex hormones on skin thickness. Br. J. Obstet. Gynaecol. 1985, 92, 256–259. [Google Scholar] [CrossRef] [PubMed]
  15. Calleja-Agius, J.; Brincat, M. The effect of menopause on the skin and other connective tissues. Gynecol. Endocrinol. 2012, 28, 273–277. [Google Scholar] [CrossRef] [PubMed]
  16. Brincat, M.P.; Baron, Y.M.; Galea, R. Estrogens and the skin. Climacteric 2005, 8, 110–123. [Google Scholar] [CrossRef] [PubMed]
  17. Ososki, A.L.; Kennelly, E.J. Phytoestrogens: A review of the present state of research. Phytother Res. 2003, 17, 845–869. [Google Scholar] [CrossRef]
  18. Brzezinski, A.; Debi, A. Phytoestrogens: The “natural” selective estrogen receptor modulators? Eur. J. Obstet. Gynecol. Reprod. Biol. 1999, 85, 47–51. [Google Scholar] [CrossRef]
  19. Chadha, R.; Bhalla, Y.; Jain, A.; Chadha, K.; Karan, M. Dietary soy isoflavone: A mechanistic insight. Nat. Prod. Commun. 2017, 12, 627–634. [Google Scholar] [CrossRef] [Green Version]
  20. Fukutake, M.; Takahashi, M.; Ishida, K.; Kawamura, H.; Sugimura, T.; Wakabayashi, K. Quantification of genistein and genistin in soybeans and soybean products. Food Chem. Toxicol. 1996, 34, 457–461. [Google Scholar] [CrossRef]
  21. Yang, Z.; Zhu, W.; Gao, S.; Yin, T.; Jiang, W.; Hu, M. Breast cancer resistance protein (abcg2) determines distribution of genistein phase ii metabolites: Reevaluation of the roles of abcg2 in the disposition of genistein. Drug Metab. Dispos. 2012, 40, 1883–1893. [Google Scholar] [CrossRef] [Green Version]
  22. Motlekar, N.; Khan, M.A.; Youan, B.B.C. Preparation and characterization of genistein containing poly(ethylene glycol) microparticles. J. Appl. Polym. Sci. 2006, 101, 2070–2078. [Google Scholar] [CrossRef]
  23. Kwon, S.H.; Kang, M.J.; Huh, J.S.; Ha, K.W.; Lee, J.R.; Lee, S.K.; Lee, B.S.; Han, I.H.; Lee, M.S.; Lee, M.W.; et al. Comparison of oral bioavailability of genistein and genistin in rats. Int. J. Pharm. 2007, 337, 148–154. [Google Scholar] [CrossRef]
  24. Kuiper, G.G.; Lemmen, J.G.; Carlsson, B.; Corton, J.C.; Safe, S.H.; van der Saag, P.T.; van der Burg, B.; Gustafsson, J.A. Interaction of estrogenic chemicals and phytoestrogens with estrogen receptor beta. Endocrinology 1998, 139, 4252–4263. [Google Scholar] [CrossRef]
  25. Thornton, M.J.; Taylor, A.H.; Mulligan, K.; Al-Azzawi, F.; Lyon, C.C.; O’Driscoll, J.; Messenger, A.G. The distribution of estrogen receptor beta is distinct to that of estrogen receptor alpha and the androgen receptor in human skin and the pilosebaceous unit. J. Investig. Derm. Symp. Proc. 2003, 8, 100–103. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Campbell, L.; Emmerson, E.; Davies, F.; Gilliver, S.C.; Krust, A.; Chambon, P.; Ashcroft, G.S.; Hardman, M.J. Estrogen promotes cutaneous wound healing via estrogen receptor beta independent of its antiinflammatory activities. J. Exp. Med. 2010, 207, 1825–1833. [Google Scholar] [CrossRef] [Green Version]
  27. Wang, H.; Zhao, Z.; Lin, M.; Groban, L. Activation of gpr30 inhibits cardiac fibroblast proliferation. Mol. Cell. Biochem. 2015, 405, 135–148. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  28. Cao, C.; Li, S.; Dai, X.; Chen, Y.; Feng, Z.; Zhao, Y.; Wu, J. Genistein inhibits proliferation and functions of hypertrophic scar fibroblasts. Burn. J. Int. Soc. Burn. Inj. 2009, 35, 89–97. [Google Scholar] [CrossRef] [PubMed]
  29. Nakashima, S.; Koike, T.; Nozawa, Y. Genistein, a protein tyrosine kinase inhibitor, inhibits thromboxane a2-mediated human platelet responses. Mol. Pharmacol. 1991, 39, 475–480. [Google Scholar]
  30. Mizushina, Y.; Shiomi, K.; Kuriyama, I.; Takahashi, Y.; Yoshida, H. Inhibitory effects of a major soy isoflavone, genistein, on human DNA topoisomerase ii activity and cancer cell proliferation. Int. J. Oncol. 2013, 43, 1117–1124. [Google Scholar] [CrossRef] [Green Version]
  31. Little, P.J.; Getachew, R.; Rezaei, H.B.; Sanchez-Guerrero, E.; Khachigian, L.M.; Wang, H.; Liao, S.; Zheng, W.; Ballinger, M.L.; Osman, N. Genistein inhibits pdgf-stimulated proteoglycan synthesis in vascular smooth muscle without blocking pdgfbeta receptor phosphorylation. Arch. Biochem. Biophys. 2012, 525, 25–31. [Google Scholar] [CrossRef]
  32. Liu, X.J.; Yang, L.; Mao, Y.Q.; Wang, Q.; Huang, M.H.; Wang, Y.P.; Wu, H.B. Effects of the tyrosine protein kinase inhibitor genistein on the proliferation, activation of cultured rat hepatic stellate cells. World J. Gastroenterol. 2002, 8, 739–745. [Google Scholar] [CrossRef] [PubMed]
  33. Yu, X.; Zhu, J.; Mi, M.; Chen, W.; Pan, Q.; Wei, M. Anti-angiogenic genistein inhibits vegf-induced endothelial cell activation by decreasing ptk activity and mapk activation. Med. Oncol. 2012, 29, 349–357. [Google Scholar] [CrossRef]
  34. Fotsis, T.; Pepper, M.; Adlercreutz, H.; Fleischmann, G.; Hase, T.; Montesano, R.; Schweigerer, L. Genistein, a dietary-derived inhibitor of in vitro angiogenesis. Proc. Natl. Acad. Sci. USA 1993, 90, 2690–2694. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Tuli, H.S.; Tuorkey, M.J.; Thakral, F.; Sak, K.; Kumar, M.; Sharma, A.K.; Sharma, U.; Jain, A.; Aggarwal, V.; Bishayee, A. Molecular mechanisms of action of genistein in cancer: Recent advances. Front. Pharmacol. 2019, 10, 1336. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Akiyama, T.; Ishida, J.; Nakagawa, S.; Ogawara, H.; Watanabe, S.; Itoh, N.; Shibuya, M.; Fukami, Y. Genistein, a specific inhibitor of tyrosine-specific protein kinases. J. Biol. Chem. 1987, 262, 5592–5595. [Google Scholar] [CrossRef]
  37. Stahl, S.; Chun, T.Y.; Gray, W.G. Phytoestrogens act as estrogen agonists in an estrogen-responsive pituitary cell line. Toxicol. Appl. Pharmacol. 1998, 152, 41–48. [Google Scholar] [CrossRef]
  38. Ellis, S.; Lin, E.J.; Tartar, D. Immunology of wound healing. Curr. Dermatol. Rep. 2018, 7, 350–358. [Google Scholar] [CrossRef] [Green Version]
  39. Wilgus, T.A. Inflammation as an orchestrator of cutaneous scar formation: A review of the literature. Plast. Aesthet Res. 2020, 7. [Google Scholar] [CrossRef]
  40. Landen, N.X.; Li, D.Q.; Stahle, M. Transition from inflammation to proliferation: A critical step during wound healing. Cell. Mol. Life Sci. 2016, 73, 3861–3885. [Google Scholar] [CrossRef] [Green Version]
  41. Ashcroft, G.S.; Mills, S.J.; Lei, K.; Gibbons, L.; Jeong, M.J.; Taniguchi, M.; Burow, M.; Horan, M.A.; Wahl, S.M.; Nakayama, T. Estrogen modulates cutaneous wound healing by downregulating macrophage migration inhibitory factor. J. Clin. Investig. 2003, 111, 1309–1318. [Google Scholar] [CrossRef] [Green Version]
  42. Archer, D.F. Postmenopausal skin and estrogen. Gynecol. Endocrinol. 2012, 28, 2–6. [Google Scholar] [CrossRef]
  43. Hamalainen, M.; Nieminen, R.; Vuorela, P.; Heinonen, M.; Moilanen, E. Anti-inflammatory effects of flavonoids: Genistein, kaempferol, quercetin, and daidzein inhibit stat-1 and nf-kappab activations, whereas flavone, isorhamnetin, naringenin, and pelargonidin inhibit only nf-kappab activation along with their inhibitory effect on inos expression and no production in activated macrophages. Mediat. Inflamm. 2007, 2007, 45673. [Google Scholar]
  44. Brasier, A.R. The nf-kappab regulatory network. Cardiovasc. Toxicol. 2006, 6, 111–130. [Google Scholar] [CrossRef] [PubMed]
  45. Weinheimer-Haus, E.M.; Mirza, R.E.; Koh, T.J. Nod-like receptor protein-3 inflammasome plays an important role during early stages of wound healing. PLoS ONE 2015, 10, e0119106. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Eo, H.; Lee, H.J.; Lim, Y. Ameliorative effect of dietary genistein on diabetes induced hyper-inflammation and oxidative stress during early stage of wound healing in alloxan induced diabetic mice. Biochem. Biophys. Res. Commun. 2016, 478, 1021–1027. [Google Scholar] [CrossRef] [PubMed]
  47. Park, E.; Lee, S.M.; Jung, I.K.; Lim, Y.; Kim, J.H. Effects of genistein on early-stage cutaneous wound healing. Biochem. Biophys. Res. Commun. 2011, 410, 514–519. [Google Scholar] [CrossRef] [PubMed]
  48. Abron, J.D.; Singh, N.P.; Price, R.L.; Nagarkatti, M.; Nagarkatti, P.S.; Singh, U.P. Genistein induces macrophage polarization and systemic cytokine to ameliorate experimental colitis. PLoS ONE 2018, 13, e0199631. [Google Scholar] [CrossRef] [PubMed]
  49. Ji, G.; Zhang, Y.; Yang, Q.; Cheng, S.; Hao, J.; Zhao, X.; Jiang, Z. Genistein suppresses lps-induced inflammatory response through inhibiting nf-kappab following amp kinase activation in raw 264.7 macrophages. PLoS ONE 2012, 7, e53101. [Google Scholar] [CrossRef] [PubMed]
  50. Cong, L.; Yang, S.; Zhang, Y.; Cao, J.; Fu, X. Dfmg attenuates the activation of macrophages induced by coculture with lpcinjured huve12 cells via the tlr4/myd88/nfkappab signaling pathway. Int. J. Mol. Med. 2018, 41, 2619–2628. [Google Scholar]
  51. Zhang, Z.S.; Xie, Q.F.; Che, L.M. Effects of gamma irradiation on aflatoxin b1 levels in soybean and on the properties of soybean and soybean oil. Appl. Radiat. Isot. 2018, 139, 224–230. [Google Scholar] [CrossRef]
  52. Byun, E.B.; Sung, N.Y.; Yang, M.S.; Lee, B.S.; Song, D.S.; Park, J.N.; Kim, J.H.; Jang, B.S.; Choi, D.S.; Park, S.H.; et al. Anti-inflammatory effect of gamma-irradiated genistein through inhibition of nf-kappab and mapk signaling pathway in lipopolysaccharide-induced macrophages. Food Chem. Toxicol. 2014, 74, 255–264. [Google Scholar] [CrossRef] [PubMed]
  53. Yellayi, S.; Zakroczymski, M.A.; Selvaraj, V.; Valli, V.E.; Ghanta, V.; Helferich, W.G.; Cooke, P.S. The phytoestrogen genistein suppresses cell-mediated immunity in mice. J. Endocrinol. 2003, 176, 267–274. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Verdrengh, M.; Jonsson, I.M.; Holmdahl, R.; Tarkowski, A. Genistein as an anti-inflammatory agent. Inflamm. Res. 2003, 52, 341–346. [Google Scholar] [CrossRef]
  55. Zhang, H.; Zhao, Z.; Pang, X.; Yang, J.; Yu, H.; Zhang, Y.; Zhou, H.; Zhao, J. Genistein protects against ox-ldl-induced inflammation through microrna-155/socs1-mediated repression of nf-kb signaling pathway in huvecs. Inflammation 2017, 40, 1450–1459. [Google Scholar] [CrossRef] [PubMed]
  56. Lee, Y.W.; Lee, W.H. Protective effects of genistein on proinflammatory pathways in human brain microvascular endothelial cells. J. Nutr. Biochem. 2008, 19, 819–825. [Google Scholar] [CrossRef] [PubMed]
  57. Wang, A.; Wei, J.; Lu, C.; Chen, H.; Zhong, X.; Lu, Y.; Li, L.; Huang, H.; Dai, Z.; Han, L. Genistein suppresses psoriasis-related inflammation through a stat3-nf-kappab-dependent mechanism in keratinocytes. Int. Immunopharmacol. 2019, 69, 270–278. [Google Scholar] [CrossRef]
  58. Savoia, P.; Raina, G.; Camillo, L.; Farruggio, S.; Mary, D.; Veronese, F.; Graziola, F.; Zavattaro, E.; Tiberio, R.; Grossini, E. Anti-oxidative effects of 17 beta-estradiol and genistein in human skin fibroblasts and keratinocytes. J. Dermatol. Sci. 2018, 92, 62–77. [Google Scholar] [CrossRef] [Green Version]
  59. Faber, L.; Kovac, I.; Mitrengova, P.; Novotny, M.; Varinska, L.; Vasilenko, T.; Kello, M.; Coma, M.; Kuruc, T.; Petrova, K.; et al. Genistein improves skin flap viability in rats: A preliminary in vivo and in vitro investigation. Molecules 2018, 23, 1637. [Google Scholar] [CrossRef] [Green Version]
  60. Lachova, V.; Mitrengova, P.; Melegova, N.; Smetana, K., Jr.; Gal, P. Genistein induces bcl-2 expression in human dermal microvascular endothelial cells: A short report. Folia Biol. 2020, 66, 142–147. [Google Scholar]
  61. Emmerson, E.; Campbell, L.; Ashcroft, G.S.; Hardman, M.J. The phytoestrogen genistein promotes wound healing by multiple independent mechanisms. Mol. Cell. Endocrinol. 2010, 321, 184–193. [Google Scholar] [CrossRef]
  62. Tie, L.; An, Y.; Han, J.; Xiao, Y.; Xiaokaiti, Y.; Fan, S.; Liu, S.; Chen, A.F.; Li, X. Genistein accelerates refractory wound healing by suppressing superoxide and foxo1/inos pathway in type 1 diabetes. J. Nutr. Biochem. 2013, 24, 88–96. [Google Scholar] [CrossRef] [PubMed]
  63. Duchnik, E.; Kruk, J.; Baranowska-Bosiacka, I.; Pilutin, A.; Maleszka, R.; Marchlewicz, M. Effects of the soy isoflavones, genistein and daidzein, on male rats’ skin. Postepy Dermatol. Alergol. 2019, 36, 760–766. [Google Scholar] [CrossRef] [PubMed]
  64. Gal, P.; Varinska, L.; Faber, L.; Novak, S.; Szabo, P.; Mitrengova, P.; Mirossay, A.; Mucaji, P.; Smetana, K. How signaling molecules regulate tumor microenvironment: Parallels to wound repair. Molecules 2017, 22, 1818. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Midgley, A.C.; Bowen, T.; Phillips, A.O.; Steadman, R. Microrna-7 inhibition rescues age-associated loss of epidermal growth factor receptor and hyaluronan-dependent differentiation in fibroblasts. Aging Cell 2014, 13, 235–244. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Carnesecchi, J.; Malbouyres, M.; de Mets, R.; Balland, M.; Beauchef, G.; Vie, K.; Chamot, C.; Lionnet, C.; Ruggiero, F.; Vanacker, J.M. Estrogens induce rapid cytoskeleton re-organization in human dermal fibroblasts via the non-classical receptor gpr30. PLoS ONE 2015, 10, e0120672. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  67. Tsui, K.H.; Wang, P.H.; Chen, C.K.; Chen, Y.J.; Chiou, S.H.; Sung, Y.J.; Li, H.Y. Non-classical estrogen receptors action on human dermal fibroblasts. Taiwan. J. Obstet. Gynecol. 2011, 50, 474–478. [Google Scholar] [CrossRef] [Green Version]
  68. Hinz, B. The role of myofibroblasts in wound healing. Curr. Res. Transl. Med. 2016, 64, 171–177. [Google Scholar] [CrossRef]
  69. Desmouliere, A.; Geinoz, A.; Gabbiani, F.; Gabbiani, G. Transforming growth factor-beta 1 induces alpha-smooth muscle actin expression in granulation tissue myofibroblasts and in quiescent and growing cultured fibroblasts. J. Cell Biol. 1993, 122, 103–111. [Google Scholar] [CrossRef] [Green Version]
  70. Sapudom, J.; Wu, X.; Chkolnikov, M.; Ansorge, M.; Anderegg, U.; Pompe, T. Fibroblast fate regulation by time dependent tgf-beta1 and il-10 stimulation in biomimetic 3d matrices. Biomater. Sci. 2017, 5, 1858–1867. [Google Scholar] [CrossRef]
  71. Band, A.M.; Laiho, M. Crosstalk of tgf-beta and estrogen receptor signaling in breast cancer. J. Mammary Gland. Biol. Neoplasia 2011, 16, 109–115. [Google Scholar] [CrossRef]
  72. Stoica, A.; Saceda, M.; Fakhro, A.; Solomon, H.B.; Fenster, B.D.; Martin, M.B. The role of transforming growth factor-beta in the regulation of estrogen receptor expression in the mcf-7 breast cancer cell line. Endocrinology 1997, 138, 1498–1505. [Google Scholar] [CrossRef]
  73. Matsuda, T.; Yamamoto, T.; Muraguchi, A.; Saatcioglu, F. Cross-talk between transforming growth factor-beta and estrogen receptor signaling through smad3. J. Biol. Chem. 2001, 276, 42908–42914. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Sienkiewicz, P.; Surazynski, A.; Palka, J.; Miltyk, W. Nutritional concentration of genistein protects human dermal fibroblasts from oxidative stress-induced collagen biosynthesis inhibition through igf-i receptor-mediated signaling. Acta Pol. Pharm. 2008, 65, 203–211. [Google Scholar] [PubMed]
  75. Raffetto, J.D.; Ligi, D.; Maniscalco, R.; Khalil, R.A.; Mannello, F. Why venous leg ulcers have difficulty healing: Overview on pathophysiology, clinical consequences, and treatment. J. Clin. Med. 2020, 10, 29. [Google Scholar] [CrossRef] [PubMed]
  76. Smith, P.C.; Santibanez, J.F.; Morales, J.P.; Martinez, J. Epidermal growth factor stimulates urokinase-type plasminogen activator expression in human gingival fibroblasts. Possible modulation by genistein and curcumin. J. Periodontal Res. 2004, 39, 380–387. [Google Scholar] [CrossRef]
  77. Carmeliet, P.; Jain, R.K. Angiogenesis in cancer and other diseases. Nature 2000, 407, 249–257. [Google Scholar] [CrossRef]
  78. Mukund, V.; Mukund, D.; Sharma, V.; Mannarapu, M.; Alam, A. Genistein: Its role in metabolic diseases and cancer. Crit. Rev. Oncol. Hematol. 2017, 119, 13–22. [Google Scholar] [CrossRef] [PubMed]
  79. Varinska, L.; Gal, P.; Mojzisova, G.; Mirossay, L.; Mojzis, J. Soy and breast cancer: Focus on angiogenesis. Int. J. Mol. Sci. 2015, 16, 11728–11749. [Google Scholar] [CrossRef] [Green Version]
  80. Li, Y.Q.; Xing, X.H.; Wang, H.; Weng, X.L.; Yu, S.B.; Dong, G.Y. Dose-dependent effects of genistein on bone homeostasis in rats’ mandibular subchondral bone. Acta Pharmacol. Sin. 2012, 33, 66–74. [Google Scholar] [CrossRef]
  81. Si, H.; Yu, J.; Jiang, H.; Lum, H.; Liu, D. Phytoestrogen genistein up-regulates endothelial nitric oxide synthase expression via activation of camp response element-binding protein in human aortic endothelial cells. Endocrinology 2012, 153, 3190–3198. [Google Scholar] [CrossRef] [Green Version]
  82. Yan, G.R.; Zou, F.Y.; Dang, B.L.; Zhang, Y.; Yu, G.; Liu, X.; He, Q.Y. Genistein-induced mitotic arrest of gastric cancer cells by downregulating kif20a, a proteomics study. Proteomics 2012, 12, 2391–2399. [Google Scholar] [CrossRef] [PubMed]
  83. Guo, Y.; Wang, S.; Hoot, D.R.; Clinton, S.K. Suppression of vegf-mediated autocrine and paracrine interactions between prostate cancer cells and vascular endothelial cells by soy isoflavones. J. Nutr. Biochem. 2007, 18, 408–417. [Google Scholar] [CrossRef] [PubMed]
  84. Ambra, R.; Rimbach, G.; de Pascual Teresa, S.; Fuchs, D.; Wenzel, U.; Daniel, H.; Virgili, F. Genistein affects the expression of genes involved in blood pressure regulation and angiogenesis in primary human endothelial cells. Nutr. Metab. Cardiovasc. Dis. NMCD 2006, 16, 35–43. [Google Scholar] [CrossRef]
  85. Kim, M.H. Flavonoids inhibit vegf/bfgf-induced angiogenesis in vitro by inhibiting the matrix-degrading proteases. J. Cell. Biochem. 2003, 89, 529–538. [Google Scholar] [CrossRef] [PubMed]
  86. Su, S.J.; Yeh, T.M.; Chuang, W.J.; Ho, C.L.; Chang, K.L.; Cheng, H.L.; Liu, H.S.; Cheng, H.L.; Hsu, P.Y.; Chow, N.H. The novel targets for anti-angiogenesis of genistein on human cancer cells. Biochem. Pharmacol. 2005, 69, 307–318. [Google Scholar] [CrossRef] [PubMed]
  87. Lu, P.; Takai, K.; Weaver, V.M.; Werb, Z. Extracellular matrix degradation and remodeling in development and disease. Cold Spring Harb. Perspect. Biol. 2011, 3, a005058. [Google Scholar] [CrossRef]
  88. Berndt, S.; Issa, M.E.; Carpentier, G.; Cuendet, M. A bivalent role of genistein in sprouting angiogenesis. Planta Med. 2018, 84, 653–661. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  89. Polito, F.; Marini, H.; Bitto, A.; Irrera, N.; Vaccaro, M.; Adamo, E.B.; Micali, A.; Squadrito, F.; Minutoli, L.; Altavilla, D. Genistein aglycone, a soy-derived isoflavone, improves skin changes induced by ovariectomy in rats. Br. J. Pharmacol. 2012, 165, 994–1005. [Google Scholar] [CrossRef] [Green Version]
  90. Marini, H.; Polito, F.; Altavilla, D.; Irrera, N.; Minutoli, L.; Calo, M.; Adamo, E.B.; Vaccaro, M.; Squadrito, F.; Bitto, A. Genistein aglycone improves skin repair in an incisional model of wound healing: A comparison with raloxifene and oestradiol in ovariectomized rats. Br. J. Pharmacol. 2010, 160, 1185–1194. [Google Scholar] [CrossRef] [Green Version]
  91. Piao, M.; Mori, D.; Satoh, T.; Sugita, Y.; Tokunaga, O. Inhibition of endothelial cell proliferation, in vitro angiogenesis, and the down-regulation of cell adhesion-related genes by genistein. Combined with a cdna microarray analysis. Endothel. J. Endothel. Cell Res. 2006, 13, 249–266. [Google Scholar] [CrossRef]
  92. Lu, H.; Shi, J.X.; Zhang, D.M.; Chen, H.L.; Qi, M.; Yin, H.X. Genistein, a soybean isoflavone, reduces the production of pro-inflammatory and adhesion molecules induced by hemolysate in brain microvascular endothelial cells. Acta Neurol. Belg. 2009, 109, 32–37. [Google Scholar] [PubMed]
  93. Jia, Z.; Babu, P.V.; Si, H.; Nallasamy, P.; Zhu, H.; Zhen, W.; Misra, H.P.; Li, Y.; Liu, D. Genistein inhibits tnf-alpha-induced endothelial inflammation through the protein kinase pathway a and improves vascular inflammation in c57bl/6 mice. Int. J. Cardiol. 2013, 168, 2637–2645. [Google Scholar] [CrossRef] [Green Version]
  94. Montiel, M.; de la Blanca, E.P.; Jimenez, E. Angiotensin ii induces focal adhesion kinase/paxillin phosphorylation and cell migration in human umbilical vein endothelial cells. Biochem. Biophys. Res. Commun. 2005, 327, 971–978. [Google Scholar] [CrossRef] [PubMed]
  95. Mukai, K.; Urai, T.; Asano, K.; Nakajima, Y.; Nakatani, T. Evaluation of effects of topical estradiol benzoate application on cutaneous wound healing in ovariectomized female mice. PLoS ONE 2016, 11, e0163560. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. Perzelova, V.; Sabol, F.; Vasilenko, T.; Novotny, M.; Kovac, I.; Slezak, M.; Durkac, J.; Holly, M.; Pilatova, M.; Szabo, P.; et al. Pharmacological activation of estrogen receptors-alpha and -beta differentially modulates keratinocyte differentiation with functional impact on wound healing. Int. J. Mol. Med. 2016, 37, 21–28. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  97. Novotny, M.; Vasilenko, T.; Varinska, L.; Smetana, K., Jr.; Szabo, P.; Sarissky, M.; Dvorankova, B.; Mojzis, J.; Bobrov, N.; Toporcerova, S.; et al. Er-alpha agonist induces conversion of fibroblasts into myofibroblasts, while er-beta agonist increases ecm production and wound tensile strength of healing skin wounds in ovariectomised rats. Exp. Dermatol. 2011, 20, 703–708. [Google Scholar] [CrossRef] [PubMed]
  98. Susanikova, I.; Puchl’ova, M.; Lachova, V.; Svajdlenka, E.; Mucaji, P.; Smetana, K., Jr.; Gal, P. Genistein and selected phytoestrogen-containing extracts differently modulate antioxidant properties and cell differentiation: An in vitro study in nih-3t3, hacat and mcf-7 cells. Folia Biol. 2019, 65, 24–35. [Google Scholar]
  99. Zhou, T.; Yang, Z.; Chen, Y.; Chen, Y.; Huang, Z.; You, B.; Peng, Y.; Chen, J. Estrogen accelerates cutaneous wound healing by promoting proliferation of epidermal keratinocytes via erk/akt signaling pathway. Cell. Physiol. Biochem. Int. J. Exp. Cell. Physiol. Biochem. Pharmacol. 2016, 38, 959–968. [Google Scholar] [CrossRef]
  100. Miyazaki, K.; Hanamizu, T.; Iizuka, R.; Chiba, K. Genistein and daidzein stimulate hyaluronic acid production in transformed human keratinocyte culture and hairless mouse skin. Ski. Pharmacol. Appl. Ski. Physiol. 2002, 15, 175–183. [Google Scholar] [CrossRef]
  101. Berman, B.; Maderal, A.; Raphael, B. Keloids and hypertrophic scars: Pathophysiology, classification, and treatment. Dermatol. Surg. 2017, 43, S3–S18. [Google Scholar] [CrossRef]
  102. Yates, S.; Rayner, T.E. Transcription factor activation in response to cutaneous injury: Role of ap-1 in reepithelialization. Wound Repair Regen. 2002, 10, 5–15. [Google Scholar] [CrossRef]
  103. Jurzak, M.; Adamczyk, K. Influence of genistein on c-jun, c-fos and fos-b of ap-1 subunits expression in skin keratinocytes, fibroblasts and keloid fibroblasts cultured in vitro. Acta Pol. Pharm. 2013, 70, 205–213. [Google Scholar] [PubMed]
  104. Jurzak, M.; Adamczyk, K.; Antonczak, P.; Garncarczyk, A.; Kusmierz, D.; Latocha, M. Evaluation of genistein ability to modulate ctgf mrna/protein expression, genes expression of tgfbeta isoforms and expression of selected genes regulating cell cycle in keloid fibroblasts in vitro. Acta Pol. Pharm. 2014, 71, 972–986. [Google Scholar] [PubMed]
  105. Kim, G.Y.; Suh, J.; Jang, J.H.; Kim, D.H.; Park, O.J.; Park, S.K.; Surh, Y.J. Genistein inhibits proliferation of brca1 mutated breast cancer cells: The gpr30-akt axis as a potential target. J. Cancer Prev. 2019, 24, 197–207. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  106. Sun, L.; Zhao, W.; Li, J.; Tse, L.A.; Xing, X.; Lin, S.; Zhao, J.; Ren, Z.; Zhang, C.X.; Liu, X. Dietary flavonoid intake and risk of esophageal squamous cell carcinoma: A population-based case-control study. Nutrition 2021, 89, 111235. [Google Scholar] [CrossRef]
  107. Pavese, J.M.; Krishna, S.N.; Bergan, R.C. Genistein inhibits human prostate cancer cell detachment, invasion, and metastasis. Am. J. Clin. Nutr. 2014, 100, 431S–436S. [Google Scholar] [CrossRef] [PubMed]
  108. Xiong, P.; Wang, R.; Zhang, X.; DeLa Torre, E.; Leon, F.; Zhang, Q.; Zheng, S.; Wang, G.; Chen, Q.H. Design, synthesis, and evaluation of genistein analogues as anti-cancer agents. Anticancer. Agents Med. Chem. 2015, 15, 1197–1203. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Figure 1. Cell-specific effects of genistein on wound healing during the inflammatory phase.
Figure 1. Cell-specific effects of genistein on wound healing during the inflammatory phase.
Cimb 43 00011 g001
Figure 2. Cell-specific effects of genistein on wound healing during the proliferation and maturation phases.
Figure 2. Cell-specific effects of genistein on wound healing during the proliferation and maturation phases.
Cimb 43 00011 g002
Figure 3. Western blot analysis of selected proteins performed on human microvascular endothelial cells (HMVEC-d) co-treated with VEGF (25 ng/mL) and increasing concentrations of genistein (1–1000 nM) after 48 h of incubation (MMP14—matrix metalloproteinase 14; VEGF-A—vascular endothelial growth factor A; CTGF—connective tissue growth factor; CXCL5—C-X-C motif chemokine 5; IL-6—interleukin 6; ITGB3—integrin β3; COL18A1—Collagen Type XVIII Alpha 1 Chain; TIMP-2—Tissue inhibitor of metalloproteinases 2).
Figure 3. Western blot analysis of selected proteins performed on human microvascular endothelial cells (HMVEC-d) co-treated with VEGF (25 ng/mL) and increasing concentrations of genistein (1–1000 nM) after 48 h of incubation (MMP14—matrix metalloproteinase 14; VEGF-A—vascular endothelial growth factor A; CTGF—connective tissue growth factor; CXCL5—C-X-C motif chemokine 5; IL-6—interleukin 6; ITGB3—integrin β3; COL18A1—Collagen Type XVIII Alpha 1 Chain; TIMP-2—Tissue inhibitor of metalloproteinases 2).
Cimb 43 00011 g003
Table 1. Genistein activity on wound healing.
Table 1. Genistein activity on wound healing.
Phase of Wound HealingTarget CellEffectReference
InflammationMacrophage↓ activation of NF-κB, STAT-1, iNOS, AMPK[43,48,49]
↑ expression of NLRP3[45,46]
↓ expression of TNF-α, NF-κB, Il6, IL-1β[46,47,48]
M1 → M2 shift promotion[48]
T cell,↓ infiltration[53]
Granulocyte↓ infiltration[54]
HUVEC↓ expression of NF-κB, TNF-α, E-selectin, P-selectin, MCP-1, IL-1β, IL-8, VCAM-1, ICAM-1[55,56]
HDMEC↑ activity of SOD[59,60]
↑ expression of Bcl-2[59,60]
Fibroblast↑ levels of GSH[58]
↓ expression of MMP-1, MMP-9[58]
↓ release of NO, ROS[58]
Keratinocyte↓ expression of TNF-α, IL-6, IL-23, MMP-1, MMP-9[57,58]
↑ levels of GSH[58]
↓ release of NO, ROS[58]
ProliferationFibroblast↑/↓ (low/high c) production of collagen[74]
↓ activity and production of uPA[76]
↓ activation of JNK, ERK1/2[76]
HUVEC↓ proliferation[83]
↓ expression of VEGF, VEGFR[83,84]
↑ signaling of MAPK[33]
↓ signaling of PTK[33]
↓ activation of c-Jun, JNK, p38[33,103]
↓ production/activation of MMP-2, MMP-9[33,84]
↓ VEFG/bFGF-induced expression of MMP-1, uPA and activation of MMP-2[85,86]
↑/↓ (low/high dose) stimulation of sprouting[62]
↑ (low dose) secretion of bFGF, EGF, angiogenin, angiopoietin-2, MMP-9 and uPA receptor[88]
↓ cell-cell adhesion, migration[91,94]
↓ expression of VE-cadherin, connexin 43, integrin αV, multimerin[91]
↓ TNF-α induced expression of MCP-1, IL-8, sICAM-1, sVCAM-1, E-selectin[93]
↓ signaling of FAK/paxillin[94]
↑ restores expression of VEGF, TGF-β1, TG2 in estrogen deficiency[90]
BMEC↓ expression of MCP-1, ICAM-1[92]
Keratinocyte↑ proliferation[99]
↑ signaling of ERk/Akt[99]
↑ production of hyaluronic acid[100]
↓ expression of FOS-B[103]
↑ expression of Bcl-2[60,104]
MaturationFibroblast↓ proliferation/collagen production in hypertrophic scars[28]
↓ signaling of MAPK/ERK in hypertrophic scars[28]
↑ expression of C-JUN, C-FOS in keloid fibroblasts[103]
↓ expression of CTGF, TGF-β(1, 2, 3) in keloid fibroblasts[104]
↑ increase; ↓ decrease.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Čoma, M.; Lachová, V.; Mitrengová, P.; Gál, P. Molecular Changes Underlying Genistein Treatment of Wound Healing: A Review. Curr. Issues Mol. Biol. 2021, 43, 127-141. https://doi.org/10.3390/cimb43010011

AMA Style

Čoma M, Lachová V, Mitrengová P, Gál P. Molecular Changes Underlying Genistein Treatment of Wound Healing: A Review. Current Issues in Molecular Biology. 2021; 43(1):127-141. https://doi.org/10.3390/cimb43010011

Chicago/Turabian Style

Čoma, Matúš, Veronika Lachová, Petra Mitrengová, and Peter Gál. 2021. "Molecular Changes Underlying Genistein Treatment of Wound Healing: A Review" Current Issues in Molecular Biology 43, no. 1: 127-141. https://doi.org/10.3390/cimb43010011

Article Metrics

Back to TopTop