Skip to main content

Interaction of the AKT and β-catenin signalling pathways and the influence of photobiomodulation on cellular signalling proteins in diabetic wound healing

Abstract

The induction of a cells destiny is a tightly controlled process that is regulated through communication between the matrix and cell signalling proteins. Cell signalling activates distinctive subsections of target genes, and different signalling pathways may be used repeatedly in different settings. A range of different signalling pathways are activated during the wound healing process, and dysregulated cellular signalling may lead to reduced cell function and the development of chronic wounds. Diabetic wounds are chronic and are characterised by the inability of skin cells to act in response to reparative inducements. Serine/threonine kinase, protein kinase B or AKT (PKB/AKT), is a central connection in cell signalling induced by growth factors, cytokines and other cellular inducements, and is one of the critical pathways that regulate cellular proliferation, survival, and quiescence. AKT interacts with a variety of other pathway proteins including glycogen synthase kinase 3 beta (GSK3β) and β-catenin. Novel methodologies based on comprehensive knowledge of activated signalling pathways and their interaction during normal or chronic wound healing can facilitate quicker and efficient diabetic wound healing. In this review, we focus on interaction of the AKT and β-catenin signalling pathways and the influence of photobiomodulation on cellular signalling proteins in diabetic wound healing.

Background

Protein components that serve as cellular receptors, cytoplasmic couriers, and nuclear effectors constitute the cellular signalling pathway for signal transduction and gene expression. Cell receptor proteins are transmembrane, having an extracellular domain to which growth factors (GFs)/cytokines bind, and an intracellular enzyme linked domain. To induce and convey signals to the cytoplasmic couriers and nuclear effectors, cell receptor proteins interact with distinctive chemical messengers including ligands such as GF and cytokines. Ligands are released by several cell types to signal themselves or others. Ligand binding creates an intermediate form of the receptors cytosolic domain, and this change instigates a sequence of downstream biochemical reactions [1,2,3]. Receptors are classified into five groups: cell surface, enzyme-linked, internal, G-protein-coupled (GPC), and ion channel. When the released ligand binds to receptors on the same cell that released them, the altered cellular behavior is said to be autocrine. Ligands can also be released and passed directly across to neighboring cells to form gap junction cell activation, and when they are passed onto nearby cells, or travel through the bloodstream to distant cellular receptors, it is referred to as paracrine and endocrine cell activation, respectively [4, 5].

Cellular signalling is mediated by molecular alterations that switch on and off a given cellular response or function, and this is accomplished via several mechanisms including desensitising receptors to their ligands, degradation of signalling proteins, and restoration of normal concentration of the ligands [6]. Moreover, the presence of specific extracellular and transmembrane signalling competitors and activators influence cellular reaction. For instance, the Wnt/β-catenin signalling pathway is inhibited by extracellular factors including frizzled-related proteins (FRPs), cerberus, dickkopf proteins (Dkks), sclerostin domain-containing 1 (Wise/SOST), Wnt-inhibitory factor 1 (WIF-1), insulin-like GF binding protein 4 (IGFBP-4), and transmembrane factors including Wnt-activated inhibitory factor 1 (Waif1/5T4), and adenomatosis polyposis coli down-regulated 1 (APCD1). Literature shows that Norrin and R-spondin advances the Wnt/β-catenin pathway by increasing stabilisation and phosphorylation of β-catenin [7]. The complexity and specificity of cellular signalling can also be controlled by compartmentalised signalling from distinct centers such as organelle specific pools of mitogen-activated protein kinases (MAPKs) within the golgi apparatus that can rapidly switch a particular signalling pathway on or off [8]. Defects in cellular signalling or pathogenic distress can cause numerous cellular alterations through the modification of signalosomes leading to the pathways hyper- or hypo-sensitive condition, and continuous and uncontrolled signalling can be detrimental [9]. Several factors including genetic and epigenetic changes can cause uncontrollable activation of cellular signalling pathways, resulting in disadvantageous cellular outcomes including over proliferation and attenuated getaway mechanisms that under normal circumstances control their growth, migration, and survival. These alterations can lead to the development of cancer and its progression, and have also been shown to be involved in delayed healing in chronic diabetic wounds [10].

Diabetes mellitus (DM) is a chronic disease that arises when the pancreas is not able to produce insulin or when cells do not make good use of insulin, a phenomenon that leads to elevated blood glucose levels (hyperglycaemia). In 2021, an estimated 537 million adults aged between 20 and 79 years were living with diabetes, and this number is projected to increase to 783 million by 2045. In the same year, DM was responsible for roughly 6.7 million deaths [11]. The increasing global incidence of diabetes is associated with a high occurrence of chronic diabetic wounds, and this complication is characterised by disrupted molecular factors involved in normal wound healing [12]. According to Abbas and Boulton [13], almost 25% of diabetic patients develop chronic wounds of the foot, with recurrence rates of 70% after treatment, and out of these, 85% may require lower limb amputation. They further pointed out that diabetic patients with one amputated lower limb are at risk of developing a chronic wound on the remaining limb, with a 70% mortality rate within five years after the first amputation.

Chronic hyperglycaemia is linked to impairment and failure of various organs and tissues (Fig. 1), and is one of the major contributing factors for the development of wound chronicity [14]. When diabetic patients develop chronic wounds, they become at high risk of developing distressing complications such as infection and limb amputation. In DM, altered tissue repair is related to abnormalities including neuropathic, vascular, and immune function. Standard treatment methods that tightly control blood glucose levels and thorough wound care are essential. However, the prognosis for the healing of diabetic wounds is very poor [11]. Typically, uncontrolled DM, oxidative stress, and heightened cell apoptosis leads to the development of chronic non-healing wounds. Hyperglycaemia-induced protein glycation generates superoxide free radicals that lead to the formation of reactive products, lipid peroxidation, and damage to cellular molecules [15]. Diabetic wounds develop due to poor glycaemic control, peripheral vascular disease, underlying damage to the nervous system (neuropathy), and inadequate foot care. In the presence of neuropathy and peripheral vascular disease, the healing of bruises or injuries may be delayed, leading to the development of chronic diabetic wounds. Mostly, diabetic wounds of the foot develop in areas that encounter persistent pressure and trauma, where hardened skin breaks due to repeated pressure in the presence of motor neuropathy [16, 17]. Sensory neuropathy is the main cause for sensory loss to trauma, and the consequence of persistent trauma on a hardened skin is subcutaneous haemorrhage, which eventually erodes and becomes ulcerated [16].

Fig. 1
figure 1

Chronic hyperglycaemia and development of chronic diabetic wounds

Characteristically, the presence of hyperglycemia, hypertension, and hypercholesterolemia in DM are risk factors for several complications such as damage to the nervous system, renal system (nephropathy), and eyes (retinopathy), and development of cardiovascular and peripheral vascular diseases and stroke. It is suggested that the control of blood pressure, glucose, and cholesterol levels can reduce the risk of developing these complications [18]. The growing number of diabetic patients with chronic wounds has caused serious social and economic burdens worldwide, and routine therapeutic strategies have been challenging for the patient as well as the healthcare system, mostly typified with high costs and unsatisfactory results. In this regard, the development of innovative cost-effective therapeutic methods that can improve patient outcomes is gaining increased attention [19, 20].

Interaction between AKT and β-catenin pathways in cell survival

The Phosphoinositide 3-kinase (PI3K)/AKT and wingless/integrated (Wnt)/β-catenin pathways are basic regulators of cell proliferation, differentiation, development, and apoptosis, and their relationship is so close such that they have become a distinctive therapeutic target [21]. AKT plays a critical function in cellular survival, and is activated when ligands bind to cell surface receptor tyrosine kinases (RTKs), resulting in the activation of the PI3K/AKT pathway (Fig. 2). PI3K/AKT signalling is regulated by the tumour suppressor gene phosphatase and tensin homologue (PTEN), representing both lipid and protein phosphatase that negatively regulates PI3K/AKT signalling via the dephosphorylated phosphatidylinositol-3,4,5-trisphosphate (PIP3) to phosphatidylinositol 4,5-bisphosphate (PIP2) [22]. The Wnt/β-catenin genes encode for a group of secreted GF proteins, and in humans 19 Wnt genes have been identified. Wnt proteins play a critical role in regulating cell proliferation, migration, and fate. Alternatively, β-catenin, a 781 amino acid protein, is encoded by catenin beta 1 (CTNNB1), and cytoplasmic β-catenin is continuously degraded by an adenomatous polyposis coli (APC)/GSK3β/Axin destruction complex that switches off the pathway which, in the presence and binding of Wnt, is inhibited, resulting in the Wnt/β-catenin pathway switching on (Fig. 3). When the Wnt/β catenin pathway is switched on and is activated, there is increased levels of cytoplasmic β-catenin that is translocated into the nucleus [23].

Fig. 2
figure 2

Phosphatidylinositol 3 kinase/Protein Kinase B (PI3K/AKT) signalling

Fig. 3
figure 3

The cellular Wnt/β-catenin pathway

Cytosolic build-up of β-catenin promotes the stability of nuclear p53, and reduced expression of caspases, c-myc, and regulation of B-cell lymphoma 2 (BCL-2) and cell apoptosis. In the nucleus, β-catenin acts as a transcriptional co-activator that interacts with T-cell factor/lymphoid enhancer-binding factor (TCF/LEF) to form a transcription regulator for genes related to cell proliferation, survival and cancer [24]. The PI3K/AKT and Wnt/β-catenin pathways are essential in the regulation of all the phases of the wound healing process. A wound showing no signs of healing for more than eight weeks becomes chronic, mostly due to DM and reduced cell signalling for cellular migration, proliferation, and survival [10]. Increased blood glucose concentration affects cellular signalling and inhibits cellular proliferation, migration, and angiogenesis, and ultimately wound healing. According to Zhang et al. [25], high glucose prevents the differentiation of stem cells via the reduced expression of cytosolic β-catenin, epidermal growth factor receptor (EGFR), and cyclin D1. Furthermore, they revealed a close interaction between Wnt/β-catenin and P13K/AKT signalling during inflammatory responses, cell proliferation and differentiation, and wound healing. In agreement to these suggestions, Wei et al. [14], demonstrated the involvement of P13K/AKT signalling proteins in angiogenesis and fibroblast activation during diabetic wound healing, and the PI3K/AKT facilitated inhibition of GSK3β affects the stability and nuclear translocation of β-catenin.

PI3K/AKT and Wnt/β-catenin signalling share receptors, including GSK3β, receptor frizzled (FZD), adaptor dishevelled (Dvl), dishevelled egl-10 and pleckstrin (DEP) domain-containing mTOR interacting protein (DEPTOR), and eukaryotic translation initiation factor 4E (eIF4E) that are engaged in both pathways. GSK3β is inhibited when both PI3K/AKT and Wnt/β-catenin signalling are activated via distinctive upstream activation [26]. Prossomariti and colleagues [27], clarified the detailed interaction of these pathways through the GSK3β FZD, Dvl, DEPTOR and eIF4E receptors. The existence of an inter-connectivity between the PI3K/AKT and Wnt/β-catenin pathways has been observed during cellular proliferation and survival. Recently, Fleming-de-Moraes et al. [28], observed an increased activation of these two pathways and enhanced cell migration and proliferation when both signalling pathways were simultaneously stimulated with IGF 1 and Wnt. However, these effects were reduced in the presence of a specific PI3K inhibitor, suggesting some dependency involving these two signalling pathways [29]. The EGF-induced stimulation of β-catenin is dependent on the activation of AKT, and EGFR activation causes the Wnt-free β-catenin activation. Cross communication between the PI3K/AKT and Wnt/β-catenin pathways is critical, and therapeutic options targeting major proteins for the PI3K/AKT and Wnt/β-catenin pathways may affect cellular function and wound healing [30].

Molecular defects in chronic diabetic wounds

The development of chronic diabetic wounds is mostly caused by diminished GF production, cellular migration, proliferation, angiogenic response, and collagen accumulation, and the unbalanced accumulation of extracellular matrix (ECM) components and matrix metalloproteinases (MMPs) [31, 32]. In addition, there are suggestions that the reduced activity of various signalling pathways including insulin, AMP-activated protein kinase (AMPK), and ligand-activated transcription factor pathways such as peroxisome proliferator-activated receptors (PPARs), may play a critical role in the development of chronic diabetic wounds [33, 34]. Stojadinovic et al. [35], alluded that delayed diabetic wound healing may be affected by overexpression of c-myc and nuclear localisation of β-catenin coupled with reduced and irregular localisation of EGFR, and initiation of glucocorticoid signalling. Recent reports indicate that variations in the signalling pathways such as the AKT/mammalian target of rapamycin; mTOR, (AKT/mTOR), results in the reduction of diabetic wound healing [36, 37].

The activation of the insulin pathways, including insulin receptor/Src Homology 2 (IR/SH2), SHC signalling adaptor protein/extracellular signal regulated kinases (SHC/ERKs), and IR/PI3K/AKT generally initiates wound healing [36, 38]. In this regard, Yu et al. [39], reported that topical application of insulin stimulates diabetic wound healing. In their study, insulin treated diabetic wounded rats showed an increase in activated AKT and GSK3β. GSK3β proteins are related to several signalling pathways including Wnt/β-catenin, nuclear factor kappa B (NF-κB), AKT, and nuclear factor erythroid 2 related factor 2 (Nrf2). Dysregulated GSK3β signalling is associated with the development of inflammatory syndromes, diabetic wounds, nephropathy, and retinopathy, and plays a critical role in angiogenesis and modification of the diabetic wound healing process [40]. Although the mechanisms for GSK3β regulation during PI3K/AKT and Wnt/β-catenin signal transduction is partly understood, it is well known that PI3K/AKT inhibits the activity of GSK3β kinase (Fig. 4), one of the key components of the β-catenin degradation and phosphorylation complex. Increased phosphorylation of β-catenin is central to its ubiquitination, degradation and reduced cellular activity [41]. To show the interaction and crosstalk between PI3K/AKT and Wnt/β-catenin pathways, Saraceno et al. [42] demonstrated that perinatal asphyxia (PA) affects the IGF receptor (IGFR)/PI3K/AKT/GSK3β/β-catenin signalling pathways in neuroprotection and neuronal plasticity. In agreement to these observations, Li et al. [43], demonstrated that the activation of human amniotic mesenchymal stem cells (hAMSC) is mediated by PI3K/AKT dependent GSK3β/β-catenin signalling to yield a favorable therapeutic benefit for skin wound healing. When Hoke et al. [44], analysed the expression of signalling protein ratios for PI3K and IGFR, mTOR and IGFR, p53 and IGFR, cyclooxygenase 2 (COX 2) and IGFR, BCL-2-antagonist/killer (BAK) and IGFR, and Caspase 9 and IGFR, they noted a considerable relationship of the values to the healing outcomes in chronic diabetic wounds. In these observations, AKT was seen to be a central regulator for most of the signalling pathways involved in the regulation of cellular migration, proliferation, differentiation, and wound healing [45].

Fig. 4
figure 4

The interaction between Wnt/β-catenin and P13K/AKT signalling

Management of chronic diabetic wounds

The management of chronic diabetic wounds involves the use of different agents that affect molecular targets by, among others, up- or down-regulating the expression of GFs, MMP, collagen synthesis and degradation, and activation of angiogenesis. GFs are critical for tissue restoration, and due to their diversity in action they have become an attractive treatment agent for stimulating tissue restoration in chronic diabetic wounds. GFs facilitate the movement of cells into the wound, and increase their proliferation and deposition of ECM [46, 47]. Typically, the cellular spatial and temporary release of GFs in response to injury is to make best use of regenerative benefits and reduce side effects to hasten wound healing [12]. There are several families of GFs that are expressed by different cell types involved with wound healing including, but not limited to, epidermal growth factor (EGF), vascular-endothelial growth factor (VEGF), fibroblast growth factor (FGF), platelet-derived growth factor (PDGF), Wnt, and transforming growth factor (TGF). They stimulate a cascade of cellular signalling pathways, including the PI3K/AKT and β-catenin pathways for different cellular functions that advance wound healing [48, 49]. However, MMP mediated ECM and GF/cytokine mortification in chronic diabetic wounds reduce their bioavailability for effective wound healing. Hence, techniques that may deliver stimulators and inhibitors of GFs, angiogenesis, MMP, ECM deposition, gene transcription, stem cell infiltration, and cell rejuvenation in diabetic wound healing are desirable [48, 50].

EGFR promotes the proliferation and movement of cells including keratinocytes and fibroblasts, improves collagen production and ECM synthesis, and its role in normal and uncontrolled wound healing has been expansively investigated [51]. The activation of EGFR has been identified as a mechanism through which GFs control normal and uncontrolled cellular function, and this has led to the development of EGFR inhibitors for cancer treatment and topical GF application to enhance cutaneous wound healing. Therapies targeting EGF/EGFR have shown to be beneficial for the management of both cancer and cutaneous wounds [52]. Wei et al. [53], elucidated that recombinant human EGF (rhEGF) promote epithelial and fibroblast cell proliferation and migration in vitro, and that vacuum sealing drainage (VSD) in combination with rhEGF effectively stimulate wound healing compared to other treatment methods in vivo. However, the chronic wound environment and increased MMPs readily degrades exogenous GFs [54, 55]. The cellular release of VEGF in response to tissue injury promotes angiogenesis and the healing of chronic diabetic wounds [56]. The release of VEGF is induced in the presence of hypoxia, and advances reepithelialisation and increased blood vessel formation during the wound healing process. The use of VEGF treatments is inhibited mainly due to vasopermeability, and in clinical trials, recombinant VEGF has shown minor clinical benefits requiring further studies on their effect in chronic wound healing. Several synthetically derived GFs for the treatment of chronic wounds are available, and comes in many forms including injections, gels, solutions, ointments, and creams. However, the efficacy of these treatment techniques for chronic diabetic wounds is not satisfactory [57, 58]. Usually, the treatment of diabetic wounds involves different techniques and agents that affect molecular targets. For example, dressings ranging from silk, cotton, or linen gauze pads and sponges, to transparent absorbent acrylic pads, hydrocolloids and hydrofibers, or topical cadexomer iodine, honey, hypochlorous acid, sterile saline, and superoxidised therapeutic techniques among others, have been developed [59]. However, due to short term benefits, and inadequate evidence of efficacy and cost-effectiveness of the available treatment methods for chronic diabetic wounds, several other novel treatment methods including photobiomodulation (PBM), formally known as low level light/laser therapy (LLLT), have been suggested [60, 61].

Photobiomodulation therapy (PBMT) in chronic diabetic wounds

Photobiomodulation therapy (PBMT) is the use of red and near infra-red light over wounds, lesions, or injuries to improve tissue and wound healing, and lessen inflammation and pain. It has been defined as an effective and low-priced commercially obtainable treatment method that can easily be used at home or in hospitals with an excellent safety profile [62, 63]. PBM was first pioneered in 1983 by Endre Mester, and involves nonthermal and low powered noninvasive lasers or light emitting diodes (LEDs). At a cellular level, PBM works by influencing different biological processes involved in tissue repair, regeneration, and wound healing. It has a biphasic dose reaction relationship, promoting wound healing at lower doses, while harmful or no effect is observed at elevated levels. It is an effectual treatment method used as an adjuvant for both acute and chronic wounds, skin restoration, musculoskeletal disorders, pain management and hair growth [64]. PBM increases the cell membrane potential and the performance of the sodium–potassium pump (Na + /K±ATPase), resulting in increased adenosine triphosphate (ATP) production (Fig. 5). The resultant increase in ATP is useful for the normalisation of cell activity to promote tissue restoration [65].

Fig. 5
figure 5

Mechanism of photobiomodulation (PBM) therapy

PBM is frequently used at wavelengths ranging between 600 and 1100 nm and fluences between 1 and 4J/cm2 with power output between 10 and 90 mW [64,65,66]. Literature suggests that PBM regulates the effects of high blood glucose on endothelial cells, reduces the concentration of TNF-α, augments proliferation of fibroblast cells, and quickens tissue repair in diabetic chronic wounds [67]. Lenifa et al. [68] and Santos et al. [69], reported accelerated wound healing in PBM treated chronic wounds, mainly via the release of cytokines and GFs, interleukin 1 alpha (IL-1α) and interleukin 8 (IL-8), fibroblast cell proliferation, migration and differentiation, and wound restoration (Fig. 6). Furthermore, PBM has been demonstrated to increase the expression of PDGF and TGF beta (TGF-β), and escalate the production of collagen and ECM to promote wound healing [70]. A photochemical response to PBM starts when the photon energy is absorbed by photoreceptors, including cytochrome c oxidase (CCO) on the electron transport chain, resulting in the increased production of ATP, reactive oxygen species (ROS), and nitric oxide (NO) in the cell [71]. Khan et al. [72], suggested that the PBM mediated activation of endogenous TGF-β1 plays a vital role in wound healing. During the acute wound healing process, cell adhesion, proliferation, migration and delineation is mediated by the TGF-β1 dependent signalling pathways including PI3K/AKT and Wnt/β-catenin, forming an interdependence web of signal transduction to achieve effective wound healing [73, 74]. Liang et al. [75], noted that the prosurvival effect of PBM on amyloid β peptide (Aβ) induced apoptosis was attained through the AKT/GSK3β/β-catenin pathway and the inhibition of GSK3β that resulted in the cytosolic accumulation of β-catenin and nuclear translocation. PBM initiates a massive prospective for precise therapy in different types of cutaneous diseases, and the most effective use of PBM necessitates the identification of its mechanisms when light interacts with tissue including the dosage [76, 77]. Wavelength, fluence and frequency of irradiation is critical for cellular mechanisms of PBM in wound healing. Nevertheless, the influence of PBM may differ according to the type of cells, and in this regard, there is still controversy for its use [77,78,79]. According to Tran et al. [80], it has been reported that LED-light irradiation with wavelengths between 630 and 940 nm has no harmful effects on humans. However, blue LED light with wavelengths of 400–500 nm may be cytotoxic and cause injury to the skin, eyes, and some other human tissues. This danger is popularly known as a ‘‘blue light hazard’’. The clinical demonstration of PBM in different medical disciplines is summarized in Table 1.

Fig. 6
figure 6

The therapeutic mechanism of photobiomodulation (PBM) in chronic diabetic wounds

Table 1 Clinical application of PBM different medical disciplines [81,82,83,84,85,86,87,88,89,90,91,92,93,94]

While PBM has been extensively studied, clarifying the fundamental means of its action may advance the understanding of its application, therapeutic benefits, and side effects [95]. In wound healing, PBM at different wavelengths and fluences have been used, resulting in variable data [96]. Variation in PBM protocols in wound management require continued controlled research to get recommendations regarding the desirable parameters [97]. Oliveira et al. [98], noted that a wavelength between 400–800 nm is the most efficient light spectrum in chronic diabetic wounds. The basic mechanism of action and the biological effect of PBM is not clear, arousing interest for research. It is evident that there is a close connection between PBM activated mitochondrial retrograde cellular signalling and molecular processes, including the stimulation and control of cytosolic kinases and downstream cascades for cellular activities required for suitable wound healing [99].

Conclusions

The management of chronic wounds is of great clinical and public concern, and the causes of delayed and impaired healing in diabetic patients are essential for research and development of novel treatment techniques. The phasic release of GFs/cytokines and the activation of their specific pathways during the wound healing process progresses the healing of acute wounds, and dysregulation of this process contributes to the development of wound chronicity [100,101,102,103,104]. However, the direct interaction of different signalling pathways during wound restoration processes remains uncertain. Therapeutic approaches including single/dual GF, cytokine stimulators and inhibitors, proteinase inhibitors and stem cell therapy are considered for diabetic chronic wounds. There is an interaction between the action of PI3K/AKT and Wnt/β-catenin pathways in cellular proliferation and survival [19]. Treatment methods that enhance the cellular release of cytokines and GFs can augment wound healing and the skin regeneration process. The consideration of the PBM-stimulated biological effects at cellular and molecular levels is key to the advancement of PBMT in the treatment of diabetic wounds [105]. In our effort to understand the molecular effects of PBM on diabetic wounds, we are, in our present work, exploring the relationship and activation of the GSK3β and β-catenin pathways in diabetic wounded cells after PBM in vitro.

Availability of data and materials

Data sharing is not applicable to this article as no datasets were generated or analysed during the current study.

Abbreviations

AKT:

Protein kinase B

AMPK:

AMP-activated protein kinase

APCD1:

Adenomatosis polyposis coli down-regulated 1

ATP:

Adenosine triphosphates

BAK:

BCL-2-antagonist/killer

BAK:

Bcl-2 homologous antagonist killer

BCL-2:

B cell lymphoma 2

CCO:

Cytochrome c oxidase

COX 2:

Cyclooxygenase 2

CTNNB1:

Catenin beta 1

Dkks:

Cerberus, dickkopf proteins

DM:

Diabetes mellitus

ECM:

Extracellular matrix

EGF:

Epidermal growth factor

EGFR:

Epidermal growth factor receptor

ERKs:

Extracellular signal regulated kinases

ERK-Sp1:

ERK-specificity protein 1

FGF:

Fibroblast growth factor

FRPs:

Frizzled-related proteins

GF:

Growth factor

GPC:

G-Protein-coupled

GSK3β :

Glycogen synthase kinase 3 beta

hAMSC:

Human amniotic mesenchymal stem cell

IGFBP-4:

Insulin-like growth factor binding protein 4

IGFR:

IGF receptor

IL:

Interleukin

LEDs:

Light emitting diodes

LEF:

Lymphoid enhancer-binding factor

LLLT:

Low level light therapy

MAPK:

Mitogen-activated protein kinase

MMPs:

Matrix metalloproteinase

mTOR:

Mammalian target of rapamycin

mTOR1:

Mammalian target of rapamycin complex 1

mTOR2:

Mammalian target of rapamycin complex 2

NF-Κb:

Nuclear factor kappa-light-chain-enhancer of activated B cell

NO:

Nitric oxide

Nrf2:

Nuclear factor erythroid 2 related factor 2

PA:

Perinatal asphyxia

PBM:

Photobiomodulation

PBMT:

Photobiomodulation therapy

PDGF:

Platelet-derived growth factor

PI3K:

Phosphatidylinositol 3 kinase

PI3P3 :

Phosphatidylinositol-3,4,5-trisphosphate

PIP2 :

Phosphatidylinositol 4,5-bisphosphate

PPARs:

Peroxisome proliferator-activated receptors

PTEN:

Phosphatase and tensin

rhEGF:

Recombinant human EGF

ROS:

Reactive oxygen species

RTKs:

Receptor tyrosine kinases

SH2:

Src homology 2

TCF:

T cell factor

TGF:

Transforming growth factor

TGF‐β:

TGF beta

TNF-α:

Tumour necrosis factor-alpha

VEGF:

Vascular endothelial growth factor

VEGF:

Vascular-endothelial growth factor

Waif1:

Wnt-activated inhibitory factor 1

WIF-1:

Wnt-inhibitory factor 1

Wise/SOST:

Sclerostin domain-containing

Wnt:

Wingless/integrated

Α :

Alpha

Β :

Beta

References

  1. Bondos SE, Dunker AK, Uversky VN. Intrinsically disordered proteins play diverse roles in cell signaling. Cell Commun Signal. 2022;20:20. https://doi.org/10.1186/s12964-022-00821-7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Bhatnagar S, Kumar A. Therapeutic targeting of signaling pathways in muscular dystrophy. J Mol Med. 2010;88:155–66. https://doi.org/10.1007/s00109-009-0550-4.

    Article  CAS  PubMed  Google Scholar 

  3. Espinosa-Sánchez A, Suárez-Martínez E, Sánchez-Díaz L, Carner A. Therapeutic targeting of signaling pathways related to cancer stemness. Front Oncol. 2020;10:1533. https://doi.org/10.3389/fonc.2020.01533.

    Article  PubMed  PubMed Central  Google Scholar 

  4. Miller EJ, Lappin SL. Physiology, cellular receptor. 2022. In: StatPearls [Internet]. Treasure Island (FL): StatPearls Publishing; 2023 Jan. https://www.ncbi.nlm.nih.gov/books/NBK554403/. Accessed 28 Apr 2022.

  5. Jones B. The therapeutic potential of GLP-1 receptor biased agonism. Br J Pharmacol. 2022;179:492–510. https://doi.org/10.1111/bph.15497.

    Article  CAS  PubMed  Google Scholar 

  6. Tabibzadeh A, Tameshkel FS, Moradi Y, Soltani S, Moradi-Lakeh M, Ashrafi GH, et al. Signal transduction pathway mutations in gastrointestinal (GI) cancers: a systematic review and meta-analysis. Sci Rep. 2020;10:18713. https://doi.org/10.1038/s41598-020-73770-1.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Chiarini F, Paganelli F, Martelli AM, Evangelisti C. The role played by Wnt/β-catenin signaling pathway in acute lymphoblastic leukemia. Int J Mol Sci. 2020;21:1098. https://doi.org/10.3390/ijms21031098.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Liu Y, Liu Y, Deng J, Li W, Nie X. Fibroblast growth factor in diabetic foot ulcer: Progress and therapeutic prospects. Front Endocrinol. 2021;12:744868. https://doi.org/10.3389/fendo.2021.744868.

    Article  Google Scholar 

  9. Jones JI, Nguyen TT, Peng Z, Chang M. Targeting MMP-9 in diabetic foot ulcers. J Pharm. 2019;12:79. https://doi.org/10.3390/ph12020079.

    Article  CAS  Google Scholar 

  10. Zhao XF. G protein-coupled receptors function as cell membrane receptors for the steroid hormone 20-hydroxyecdysone. Cell Commun Signal. 2020;18:146. https://doi.org/10.1186/s12964-020-00620-y.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. International diabetes federation (IDF) school of diabetes. https://www.idfdiabeteschool.org/what-is-diabetes. Accessed 15 Dec 2021.

  12. Patel S, Srivastava S, Singh MR, Singh D. Mechanistic insight into diabetic wounds: Pathogenesis, molecular targets and treatment strategies to pace wound healing. Biomed Pharmacother. 2019;112:108615. https://doi.org/10.1016/j.biopha.2019.108615.

    Article  CAS  PubMed  Google Scholar 

  13. Abbas ZG, Boulton AJM. Diabetic foot ulcer disease in African continent: ‘From clinical care to implementation’ – Review of diabetic foot in last 60 years—1960 to 2020. Diabetes Res Clin Pract. 2022;183:109155. https://doi.org/10.1016/j.diabres.2021.109155.

    Article  PubMed  Google Scholar 

  14. Wei P, Zhong C, Yang X, Shu F, Xiao S, Gong T, et al. Exosomes derived from human amniotic epithelial cells accelerate diabetic wound healing via PI3K-AKT-mTOR-mediated promotion in angiogenesis and fibroblast function. Burns Trauma. 2020. https://doi.org/10.1093/burnst/tkaa020.

    Article  PubMed  PubMed Central  Google Scholar 

  15. Deng L, Du C, Song P, Chen T, Rui S, Armstrong DG, et al. The role of oxidative stress and antioxidants in diabetic wound healing. Oxid Med Cell Longev. 2021. https://doi.org/10.1155/2021/8852759.

    Article  PubMed  PubMed Central  Google Scholar 

  16. Tottoli EM, Dorati R, Genta I, Chiesa E, Pisani S, Conti B. Skin wound healing process and new emerging technol-ogies for skin wound care and regeneration. J Pharm. 2020;12:735. https://doi.org/10.3390/pharmaceutics12080735.

    Article  CAS  Google Scholar 

  17. Oliver TI, Mutluoglu M. Diabetic foot ulcer. [Updated 2021 Aug 19]. In: StatPearls [Internet]. Treasure Island (FL): StatPearls Publishing; 2021 Jan-. Available from: https://www.ncbi.nlm.nih.gov/books/NBK537328/. Accessed 22 Dec 2021.

  18. Jordan H, Kowalewski C, Zimek Z, Fiedor P, Kaminski A, Oldak T, et al. Chronic diabetic wounds and their treatment with skin substitutes. Cells. 2021;10:655. https://doi.org/10.3390/cells10030655.

    Article  CAS  Google Scholar 

  19. Guo YJ, Pan WW, Liu SB, Shen ZF, Xu Y, Hu LL. ERK/MAPK signalling pathway and tumorigenesis (Review). Spandidos publications. 2020;19:1997–2007. https://doi.org/10.3892/etm.2020.8454.

    Article  CAS  Google Scholar 

  20. Nair A, Chauhan P, Saha B, Kubatzky KF. Conceptual evolution of cell signalling. Int J Mol Sci. 2019;20:3292. https://doi.org/10.3390/ijms20133292.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Langhammer TS, Roolf C, Krohn S, Kretzschmar C, Huebner R, Rolfs A, et al. PI3K/AKT signaling interacts with Wnt/β-catenin signaling but does not induce an accumulation of β-catenin in the nucleus of acute lymphoblastic leukemia cell lines. Blood. 2013;122:4886. https://doi.org/10.1182/blood.V122.21.4886.4886.

    Article  Google Scholar 

  22. Kalimuthu S, Se-Kwon K. Cell survival and apoptosis signalling as therapeutic target for cancer: marine bioactive compounds. Int J Mol Sci. 2013;14:2334–54. https://doi.org/10.3390/ijms14022334.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Jung YS, Park J. Wnt signaling in cancer: therapeutic targeting of Wnt signaling beyond β-catenin and the destruction complex. Exp Mol Med. 2020;52:183–91. https://doi.org/10.1038/s12276-020-0380-6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Trejo-Solis C, Escamilla-Ramirez A, Jimenez-Farfan D, Castillo-Rodriguez RA, Flores-Najera A, et al. Crosstalk of the Wnt/β-Catenin Signalling pathway in the induction of apoptosis on cancer cells. Pharmaceuticals. 2021;14:871. https://doi.org/10.3390/ph14090871.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Zhang H, Nie X, Shi X, Zhao J, Chen Y, Yao Q, et al. Regulatory mechanisms of the Wnt/β-catenin pathway in diabetic cutaneous ulcers. Front Pharmacol. 2018;9:1114. https://doi.org/10.3389/fphar.2018.01114.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Kaidanovich-Beilin O, Woodgett JR. GSK3: functional insights from cell biology and animal models. Front Mol Neurosci. 2011;4:40. https://doi.org/10.3389/fnmol.2011.00040.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Prossomariti A, Piazzi G, Alquati C, Ricciardiello L. Are Wnt/β-catenin and PI3K/AKT/mTORC1 distinct pathways in colorectal cancer? Cell Mol Gastroenterol Hepatol. 2020;10:491–506. https://doi.org/10.1016/j.jcmgh.2020.04.007.

    Article  PubMed  PubMed Central  Google Scholar 

  28. Fleming-de-Moraes CD, Rocha MR, Tessmann JW, Martins de Araujo W, Morgado-Diaz JA. Crosstalk between PI3K/AKT and Wnt/β-catenin pathways promote colorectal cancer progression regardless of mutational status. Cancer Biol Ther. 2022;23(1):1–13. https://doi.org/10.1080/15384047.2022.2108690.

  29. Dong J, Xu X, Zhang Q, Yuan Z, Tan B. The PI3K/AKT pathway promotes fracture healing through its crosstalk with Wnt/β-catenin. Exp Cell Res. 2020;394:112137. https://doi.org/10.1016/j.yexcr.2020.112137.

    Article  CAS  PubMed  Google Scholar 

  30. Spampinato SF, Caruso GI, De Pasquale R, Sortino MA, Merlo S. The treatment of impaired wound healing in diabetes: looking among old drugs. J Pharm. 2020;13:60. https://doi.org/10.3390/ph13040060.

    Article  CAS  Google Scholar 

  31. Armstrong DG, Boulton AJM, Bus SA. Diabetic foot ulcers and their recurrence. N Engl J Med. 2017;376:2367–75. https://doi.org/10.1056/NEJMra1615439.

    Article  PubMed  Google Scholar 

  32. Chu E, Mychasiuk R, Hibbs ML, Semple BD. Dysregulated phosphoinositide 3-kinase signaling in microglia: shaping chronic neuroinflammation. J Neuroinflammation. 2021;18:276. https://doi.org/10.1186/s12974-021-02325-6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Liu Q, Chen L, Hu L, Guo Y, Shen X. Small molecules from natural sources, targeting signaling pathways in diabetes. Biochim Biophys Acta. 2010;1799:854–65. https://doi.org/10.1016/j.bbagrm.2010.06.004.

    Article  CAS  PubMed  Google Scholar 

  34. Bauters C, Lamblin N, Mc Fadden EP, Belle EV, Millaire A, de Groote P. Influence of diabetes mellitus on heart failure risk and outcome. Cardiovasc Diabetol. 2003. https://doi.org/10.1186/1475-2840-2-1.

    Article  PubMed  PubMed Central  Google Scholar 

  35. Stojadinovic O, Brem H, Vouthounis C, Lee B, Fallon J, Stallcup M, et al. Molecular pathogenesis of chronic wounds: the role of beta-catenin and c-myc in the inhibition of epithelialization and wound healing. Am J Pathol. 2005;167:59–69. https://doi.org/10.1016/s0002-9440(10)62953-7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Huang H, Cui W, Qiu W, Zhu M, Zhao R, Zeng D, et al. Impaired wound healing results from the dysfunction of the AKT/mTOR pathway in diabetic rats. J Dermatol Sci. 2015;79:241–51. https://doi.org/10.1016/j.jdermsci.2015.06.002.

    Article  CAS  PubMed  Google Scholar 

  37. Guo W, Qiu W, Ao X, Li W, He X, Ao L, et al. Low-concentration DMSO accelerates skin wound healing by AKT/mTOR-mediated cell proliferation and migration in diabetic mice. Br J Pharmacol. 2020;177:3327–41. https://doi.org/10.1111/bph.15052.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Lima MH, Caricilli AM, de Abreu L, Araújo EP, Pelegrinelli FF, Thirone AC, et al. Topical insulin accelerates wound healing in diabetes by enhancing the AKT and ERK pathways: a double-blind placebo-controlled clinical trial. PLoS ONE. 2012;7:e36974. https://doi.org/10.1371/journal.pone.0036974.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Yu T, Gao M, Yang P, Pei Q, Liu D, Wang D, et al. Topical insulin accelerates cutaneous wound healing in insulin-resistant diabetic rats. Am J Transl Res. 2017;9:4682–93.

    CAS  PubMed  PubMed Central  Google Scholar 

  40. Wang X, Li W, Lu S, Ma Z. Modulation of the wound healing through noncoding RNA interplay and GSK-3β/NF-κB signaling interaction. Int J Genomics. 2021. https://doi.org/10.1155/2021/9709290.

    Article  PubMed  PubMed Central  Google Scholar 

  41. Wu D, Pan W. GSK3: a multifaceted kinase in Wnt signaling. Trends Biochem Sci. 2010;35:161–8. https://doi.org/10.1016/j.tibs.2009.10.002.

    Article  CAS  PubMed  Google Scholar 

  42. Saraceno GE, Bellini MJ, Garcia-Segura LM, Capani F. Estradiol Activates PI3K/AKT/GSK3 pathway under chronic neurodegenerative conditions triggered by perinatal asphyxia. Front Pharmacol. 2018;9:335. https://doi.org/10.3389/fphar.2018.00335.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Li JY, Ren KK, Zhang WJ, Xiao L, Wu HY, Liu QY, et al. Human amniotic mesenchymal stem cells and their paracrine factors promote wound healing by inhibiting heat stress-induced skin cell apoptosis and enhancing their proliferation through activating PI3K/AKT signaling pathway. Stem Cell Res Ther. 2019;10:247. https://doi.org/10.1186/s13287-019-1366-y.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Hoke GD, Ramos C, Hoke NN, Crossland MC, Shawler LG, Boykin JV. Atypical diabetic foot ulcer keratinocyte protein signaling correlates with impaired wound healing. J Diabetes Res. 2016. https://doi.org/10.1155/2016/1586927.

    Article  PubMed  PubMed Central  Google Scholar 

  45. Xu H, Zhu J, Hu C, Song H, Li Y. Inhibition of microRNA-181a may suppress proliferation and invasion and promote apoptosis of cervical cancer cells through the PTEN/AKT/FOXO1 pathway. J Physiol Biochem. 2016;72:721–32. https://doi.org/10.1007/s13105-016-0511-7.

    Article  CAS  PubMed  Google Scholar 

  46. Jere S, Houreld N, Abrahamse H. Role of the PI3K/AKT (mTOR and GSK3β) signalling pathway and photobiomodulation in diabetic wound healing. Cytokine Growth Factor Rev. 2019;50:52–9. https://doi.org/10.1016/j.cytogfr.2019.03.001.

    Article  CAS  PubMed  Google Scholar 

  47. Ren X, Zhao M, Lash B, Martino MM, Julier Z. Growth factor engineering strategies for regenerative medicine applications. Front Bioeng Biotechnol. 2020;7:469. https://doi.org/10.3389/fbioe.2019.00469.

    Article  PubMed  PubMed Central  Google Scholar 

  48. Jimi S, Jaguparov A, Nurkesh A, Sultankulov B, Saparov A. Sequential delivery of cryogel released growth factors and cytokines accelerates wound healing and improves tissue regeneration. Front Bioeng Biotechnol. 2020;8:345. https://doi.org/10.3389/fbioe.2020.00345.

    Article  PubMed  PubMed Central  Google Scholar 

  49. Vaidyanathan L. Growth factors in wound healing—a review. Biomed Pharmacol J. 2021. https://doi.org/10.13005/bpj/2249.

    Article  Google Scholar 

  50. Park JW, Hwang SR, Yoon IS. Advanced growth factor delivery systems in wound management and skin regeneration. Molecules. 2017;22:1259. https://doi.org/10.3390/molecules22081259.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Berry-Kilgour C, Cabral J, Wise L. Advancements in the delivery of growth factors and cytokines for the treatment of cutaneous wound indications. Adv Wound Caref. 2021;10:596–622. https://doi.org/10.1089/wound.2020.1183.

    Article  Google Scholar 

  52. Cheng Y, Li Y, Huang S, Yu F, Bei Y, Zhang Y, et al. Hybrid freeze-dried dressings composed of epidermal growth factor and recombinant human-like collagen enhance cutaneous wound healing in rats. Front Bioeng Biotechnol. 2020;8:742. https://doi.org/10.3389/fbioe.2020.00742.

    Article  PubMed  PubMed Central  Google Scholar 

  53. Wei S, Wang W, Li L, Meng HY, Feng CZ, Dong YY, et al. Recombinant human epidermal growth factor combined with vacuum sealing drainage for wound healing in Bama pigs. Military Med Res. 2021;8:18. https://doi.org/10.1186/s40779-021-00308-5.

    Article  CAS  Google Scholar 

  54. Bodnar RJ. Epidermal growth factor and epidermal growth factor receptor: the yin and yang in the treatment of cutaneous wounds and cancer. Adv Wound Caref. 2013;2:24–9. https://doi.org/10.1089/wound.2011.0326.

    Article  Google Scholar 

  55. Ferrer-Tasies L, Santana H, Cabrera-Puig I, González-Mira E, Ballell-Hosa L, Castellar-Álvarez C, et al. Recombinant human epidermal growth factor/quatsome nanoconjugates: A robust topical delivery system for complex wound healing. Adv Therap. 2021;4:2000260. https://doi.org/10.1002/adtp.202000260.

    Article  CAS  Google Scholar 

  56. White MJV, Briquez PS, White DAV, Hubbell JA. VEGF-A, PDGF-BB and HB-EGF engineered for promiscuous super affinity to the extracellular matrix improve wound healing in a model of type 1 diabetes. Regen Med. 2021;6:76. https://doi.org/10.1038/s41536-021-00189-1.

    Article  CAS  Google Scholar 

  57. Yamakawa S, Hayashida K. Advances in surgical applications of growth factors for wound healing. Burn Trauma. 2019;7:10. https://doi.org/10.1186/s41038-019-0148-1.

    Article  Google Scholar 

  58. Veith AP, Henderson K, Spencer A, Sligar AD, Baker AB. Therapeutic strategies for enhancing angiogenesis in wound healing. Adv Drug Deliv Rev. 2019;146:97–125. https://doi.org/10.1016/j.addr.2018.09.010.

    Article  CAS  PubMed  Google Scholar 

  59. Ongarora BG. Recent technological advances in the management of chronic wounds: a literature review. Health Sci Rep. 2022;5:e641. https://doi.org/10.1002/hsr2.641.

    Article  PubMed  PubMed Central  Google Scholar 

  60. Alexiadou K, Doupis J. Management of diabetic foot ulcers. Diabetes Ther. 2012;3:4. https://doi.org/10.1007/s13300-012-0004-9.

    Article  PubMed  PubMed Central  Google Scholar 

  61. Chanque-Fossuo CN, Ho D, Dahle SE, Koo E, Isseroff RR, Jagdeo J. Low-level light therapy for treatment of diabetic foot ulcer: a review of clinical experiences. J Drugs Dermatol. 2016;15:843–8.

    Google Scholar 

  62. Crovetti G, Martinelli G, Issi M, Barone M, Guizzardi M, Campanati B, et al. Platelet gel for healing cutaneous chronic wounds. Transfus Apher Sci. 2004;30:145–51. https://doi.org/10.1016/j.transci.2004.01.004.

    Article  PubMed  Google Scholar 

  63. Raghuram AC, Yu RP, Lo AY, Sung CJ, Bircan M, Thompson HJ, et al. Role of stem cell therapies in treating chronic wounds: a systematic review. World J Stem Cells. 2020;12:659–75. https://doi.org/10.4252/wjsc.v12.i7.659.

    Article  PubMed  PubMed Central  Google Scholar 

  64. Bensadoun RJ, Epstein JB, Nair RG, Barasch A, Raber-Durlacher JE, Migliorati C, et al. World association for laser therapy (WALT). Safety and efficacy of photobiomodulation therapy in oncology: a systematic review. Cancer Med. 2020;22:8279–300. https://doi.org/10.1002/cam4.3582.

    Article  Google Scholar 

  65. Tam SY, Tam VCW, Ramkumar S, Khaw ML, Law HKW, Lee SWY. Review on the cellular mechanisms of low-level laser therapy use in oncology. Front Oncol. 2020;10:1255. https://doi.org/10.3389/fonc.2020.01255.

    Article  PubMed  PubMed Central  Google Scholar 

  66. Chittoria RK, Kumar SH. Low-level laser therapy (LLLT) in wound healing. In: Shiffman, M., Low, M. (eds) chronic wounds, wound dressings and wound healing. Recent clinical techniques, results, and research in wounds. Springer, Cham. 2018;6. https://doi.org/10.1007/15695_2017_89.

  67. Huang J, Chen J, Xiong S, Huang J, Liu Z. The effect of low-level laser therapy on diabetic foot ulcers: A meta-analysis of randomised controlled trials. Int Wound J. 2021;18:763–76. https://doi.org/10.1111/iwj.13577.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Lenifa Priyadarshini MJ, Kishore Babu EP, Imran TA. Effect of low level laser therapy on diabetic foot ulcers: a randomized control trial. Int Surg J. 2018;5:1008–15. https://doi.org/10.18203/2349-2902.isj20180821.

    Article  Google Scholar 

  69. Santos CM, dos Rocha RB, da Hazime FA, Cardoso VS. A systematic review and meta-analysis of the effects of low-level laser therapy in the treatment of diabetic foot ulcers. Int J Low Extrem Wounds. 2021;20:198–207. https://doi.org/10.1177/1534734620914439.

    Article  CAS  PubMed  Google Scholar 

  70. Bavaresco T, Pires AUB, Moraes VM, Osmarin VM, Silveira DT, Lucena AF. Low-level laser therapy for treatment of venous ulcers evaluated with the nursing outcome classification: study protocol for a randomized controlled trial. Trials. 2018;19:372. https://doi.org/10.1186/s13063-018-2729-x.

    Article  PubMed  PubMed Central  Google Scholar 

  71. Chen Q, Yang J, Yin H, Li Y, Qiu H, Gu Y, et al. Optimization of photo-biomodulation therapy for wound healing of diabetic foot ulcers in vitro and in vivo. Biomed Opt Express. 2022;13:2450–66. https://doi.org/10.1364/BOE.451135.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Khan I, Rahman SU, Tang E, Engel K, Hall B, Kulkarni AB, et al. Accelerated burn wound healing with photobiomodulation therapy involves activation of endogenous latent TGF-β1. Sci Rep. 2021;11:13371. https://doi.org/10.1038/s41598-021-92650-w.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Jere SW, Houreld NN, Abrahamse H. Effect of photobiomodulation on cellular migration and survival in diabetic and hypoxic diabetic wounded fibroblast cells. Lasers Med Sci. 2021;36:365–74. https://doi.org/10.1007/s10103-020-03041-y.

    Article  PubMed  Google Scholar 

  74. Mariam TG, Alemayehu A, Tesfaye E, Mequannt W, Temesgen K, Yetwale F, et al. Prevalence of diabetic foot ulcer and associated factors among adult diabetic patients who attend the diabetic follow-up clinic at the University of Gondar referral hospital, northwest Ethiopia, 2016 Institutional based cross-sectional study. J Diabetes Res. 2017. https://doi.org/10.1155/2017/2879249.

    Article  PubMed  PubMed Central  Google Scholar 

  75. Liang J, Liu L, Xing D. Photobiomodulation by low-power laser irradiation attenuates Aβ-induced cell apoptosis through the Akt/GSK3β/β-catenin pathway. Free Radic Biol Med. 2012;53(7):1459–67. https://doi.org/10.1016/j.freeradbiomed.2012.08.003.

    Article  CAS  PubMed  Google Scholar 

  76. Otterço AN, Andrade AL, Brassolatti P, Pinto KNZ, Araújo HSS, Parizotto NA. Photobiomodulation mechanisms in the kinetics of the wound healing process in rats. J Photochem Photobiol B Biol. 2018;183:22–9. https://doi.org/10.1016/j.jphotobiol.2018.04.010.

    Article  CAS  Google Scholar 

  77. Evans D, Houreld N, Abrahamse H. Low level laser therapy (LLLT) as an effective therapeutic modality for delayed wound healing. Ann N Y Acad Sci. 2005;1056:486–93. https://doi.org/10.1196/annals.1352.040.

    Article  Google Scholar 

  78. Mosca RC, Santos SN, Nogueira GEC, Pereira DL, Costa FC, Pereira JX, et al. The efficacy of photobiomodulation therapy in improving tissue resilience and healing of radiation skin damage. Photonics. 2022;9:10. https://doi.org/10.3390/photonics9010010.

    Article  Google Scholar 

  79. Dima R, TieppoFrancio V, Towery C, Davani S. Review of literature on low-level laser therapy benefits for nonpharmacological pain control in chronic pain and osteoarthritis. Altern Ther Health Med. 2018;24:8–10.

    PubMed  Google Scholar 

  80. Tran VV, Chae M, Moon JY, Lee YC. Light emitting diodes technology-based photobiomodulation therapy (PBMT) for dermatology and aesthetics: Recent applications, challenges, and perspectives. Opt Laser Technol. 2021;135:106698. https://doi.org/10.1016/j.optlastec.2020.106698.

    Article  CAS  Google Scholar 

  81. Yu S, Lan CCE, Yu HS. Mechanisms of repigmentation induced by photobiomodulation therapy in vitiligo. Exp Dermatol. 2019;28:10–4. https://doi.org/10.1111/exd.13823.

    Article  CAS  PubMed  Google Scholar 

  82. Gianfaldoni S, Tchernev G, Wollina U, Fioranelli M, Roccia MG, Gianfaldoni R, et al. An Overview of laser in dermatology: the past, the present and … the future (?). Open Access Maced J Med Sci. 2017;5:526–30. https://doi.org/10.3889/oamjms.2017.130.

    Article  PubMed  PubMed Central  Google Scholar 

  83. Husain Z, Alster T. The role of lasers and intense pulsed light technology in dermatology. Clin Cosmet Investig Dermatol. 2016;9:29–40. https://doi.org/10.2147/CCID.S69106.

    Article  PubMed  PubMed Central  Google Scholar 

  84. Bodnar PM, Peshko AO, Prystupiuk OM, Voronko AA, Kyriienko DV, Mykhal’chyshyn HP, et al. Laser therapy in diabetes mellitus. Lik Sprava. 1999;6:125–8.

    Google Scholar 

  85. Lin L, Li J, Lin J, Tang S, Li Y. Effectiveness and safety of low-level laser therapy in diabetic peripheral neuropathy: a protocol for a systematic review and meta-analysis. Syst Rev. 2021;10:96. https://doi.org/10.1186/s13643-021-01656-y.

    Article  PubMed  PubMed Central  Google Scholar 

  86. https://www.nhs.uk/conditions/diabetic-retinopathy/treatment/. Accessed 16 Aug 2023.

  87. Kalinina OV, Alekseeva NV, Burtsev EM. Infrared laser therapy in distal diabetic polyneuropathy. Zh Nevrol Psikhiatr Im S S Korsakova. 1998;98:23–5.

    CAS  PubMed  Google Scholar 

  88. Sousa RG, Batista KN. Laser therapy in wound healing associated with diabetes mellitus—review. An Bras Dermatol. 2016;91:489–93. https://doi.org/10.1590/abd1806-4841.20163778.

    Article  PubMed  PubMed Central  Google Scholar 

  89. Verma SK, Maheshwari S, Singh RK, Chaudhari PK. Laser in dentistry: an innovative tool in modern dental practice. Natl J Maxillofac Surg. 2012;3:124–32. https://doi.org/10.4103/0975-5950.111342.

    Article  PubMed  PubMed Central  Google Scholar 

  90. Kathuria V, Dhillon JK, Kalra G. Low level laser therapy: a panacea for oral maladies. Laser Ther. 2015;24:215–23. https://doi.org/10.5978/islsm.15-RA-01.

    Article  PubMed  PubMed Central  Google Scholar 

  91. Cronshaw M, Parker S, Anagnostaki E, Mylona V, Lynch E, Grootveld M. Photobiomodulation dose parameters in dentistry: a systematic review and meta-analysis. Dent J (Basel). 2020;8:114. https://doi.org/10.3390/dj8040114.

    Article  PubMed  Google Scholar 

  92. Hashmi JT, Huang YY, Osmani BZ, Sharma SK, Naeser MA, Hamblin MR. Role of low-level laser therapy in neurorehabilitation. PM&R. 2010;2:292–305. https://doi.org/10.1016/j.pmrj.2010.10.013.

    Article  Google Scholar 

  93. Thunshelle C, Hamblin MR. Transcranial low-level laser (Light) therapy for brain injury. Photomed Laser Surg. 2016;34:587–98. https://doi.org/10.1089/pho.2015.4051.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. De la Torre JC. Treating cognitive impairment with transcranial low level laser therapy. J Photochem Photobiol B Biol. 2017;168:149–55. https://doi.org/10.1016/j.jphotobiol.2017.02.008.

    Article  CAS  Google Scholar 

  95. Egger A, Resnik SR, Aickara D, Maranda E, Kaiser M, Wikramanayake TC, et al. Examining the safety and efficacy of low-level laser therapy for male and female pattern hair loss: a review of the literature. Skin Appendage Disord. 2020;6:259–67. https://doi.org/10.1159/000509001.

    Article  PubMed  PubMed Central  Google Scholar 

  96. Pereira FLC, Ferreira MVL, da Silva MP, Rossi FM, Alves MP, Alves BLP. Use of a high-power laser for wound healing: a case report. J Lasers Med Sci. 2020;11:112–4. https://doi.org/10.15171/jlms.2020.19.

    Article  PubMed  PubMed Central  Google Scholar 

  97. Arjmand B, Khodadost M, Jahani Sherafat S, Rezaei Tavirani M, Ahmadi N, Hamzeloo Moghadam M, et al. Low-level laser therapy: potential and complications. J Lasers Med Sci. 2021;12:42. https://doi.org/10.34172/jlms.2021.42.

    Article  Google Scholar 

  98. Oliveira AC, Souza CJS, Braga JAC, Campos HLM, Silva TS, Maciel TS. Parameters used in electrophototherapy for wound healing: a scope review. Res Soc Dev. 2021;10:e45910616003.

    Article  Google Scholar 

  99. Ebrahimi P, Hadilou M, Naserneysari F, Dolatabadi A, Tarzemany R, Vahed N, et al. Effect of photobiomodulation in secondary intention gingival wound healing—a systematic review and meta-analysis. BMC Oral Health. 2021;21:258. https://doi.org/10.1186/s12903-021-01611-2.

    Article  PubMed  PubMed Central  Google Scholar 

  100. Wu S, Xing D. Intracellular signaling cascades following light irradiation. Laser Photonics Rev. 2014;8:115–30. https://doi.org/10.1002/lpor.201300015.

    Article  CAS  Google Scholar 

  101. Astuti SD, Sulistyo A, Setiawatie EM, Khasanah M, Purnobasuki H, Arifianto D, et al. An in-vivo study of photobiomodulation using 403 nm and 649 nm diode lasers for molar tooth extraction wound healing in wistar rats. Odontology. 2022;110:240–53. https://doi.org/10.1007/s10266-021-00653-w.

    Article  CAS  PubMed  Google Scholar 

  102. Hoversten KP, Kiemele LJ, Stolp AM, Takahashi PY, Verdoorn BP. Prevention, diagnosis, and management of chronic wounds in older adults. Mayo Clin Proc. 2020;95:2021–34. https://doi.org/10.1016/j.mayocp.2019.10.014.

    Article  PubMed  Google Scholar 

  103. Andrews KL, Derby KM, Jacobson TM, Sievers BA, Kiemele LJ. 24—Prevention and management of chronic wounds, editor(s): David X. Cifu, Braddom's physical medicine and rehabilitation (Sixth Edition), Elsevier. 2021;469–84.e4. doi:https://doi.org/10.1016/B978-0-323-62539-5.00024-2.

  104. Bowers S, Franco E. Chronic wounds: evaluation and management. Am Fam Physician. 2020;101:159–66.

    PubMed  Google Scholar 

  105. Jones RE, Foster DS, Longaker MT. Management of chronic wounds—2018. JAMA. 2018;320:1481–2. https://doi.org/10.1001/jama.2018.12426.

    Article  PubMed  Google Scholar 

Download references

Acknowledgements

Not applicable.

Funding

This work was supported by the South African Research Chairs Initiative of the Department of Science and Technology and National Research Foundation of South Africa (Grant No 98337), as well as grants received from the University of Johannesburg (URC), the African Laser Centre (ALC), the National Research Foundation (NRF, Grant No 129327), and the Council for Scientific and Industrial Research (CSIR)-National Laser Centre (NLC) Laser Rental Pool Programme.

Author information

Authors and Affiliations

Authors

Contributions

SWJ created, prepared and wrote the initial draft of the article. HA contributed in the supervision and mentorship of the lead author. NNH is the postgraduate supervisor of lead author. She played an oversight and supervisory responsibility, including mentorship, planning and execution of the research. She was involved in critical revision of drafts of the manuscript for intellectual content. All authors read and approved the final manuscript.

Corresponding author

Correspondence to Sandy Winfield Jere.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare that there is no conflict of interest regarding the publication of this paper.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Jere, S.W., Abrahamse, H. & Houreld, N.N. Interaction of the AKT and β-catenin signalling pathways and the influence of photobiomodulation on cellular signalling proteins in diabetic wound healing. J Biomed Sci 30, 81 (2023). https://doi.org/10.1186/s12929-023-00974-8

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12929-023-00974-8

Keywords