Open access peer-reviewed chapter

Inorganic Polyphosphates Are Important for Cell Survival and Motility of Human Skin Keratinocytes and Play a Role in Wound Healing

Written By

Cynthia M. Simbulan-Rosenthal, Bonnie C. Carney, Anirudh Gaur, Manish Moghe, Elliott Crooke, Lauren T. Moffatt, Jeffrey W. Shupp and Dean S. Rosenthal

Submitted: 04 March 2019 Reviewed: 31 May 2019 Published: 28 June 2019

DOI: 10.5772/intechopen.87183

From the Edited Volume

Contemporary Topics about Phosphorus in Biology and Materials

Edited by David G. Churchill, Maja Dutour Sikirić, Božana Čolović and Helga Füredi Milhofer

Chapter metrics overview

942 Chapter Downloads

View Full Metrics

Abstract

Inorganic polyphosphate (polyP) is a simple ancient polymer of linear chains of orthophosphate residues linked by high energy phospho-anhydride bonds ubiquitously found in all organisms. Despite its structural simplicity, it plays diverse functional roles. polyP is involved in myriad of processes including serving as microbial phosphagens, buffer against alkalis, Ca2+ storage, metal-chelating agents, pathogen virulence, cell viability and proliferation, structural component and chemical chaperones, and in the microbial stress response. In mammalian cells, polyP has been implicated in blood coagulation, inflammation, bone differentiation, cell bioenergetics, signal transduction, Ca2+-signaling, neuronal excitability, as a protein-stabilizing scaffold, and in wound healing, among others. This chapter will discuss (1) polyP metabolism and roles of polyP in prokaryotic and eukaryotic cells, (2) the contribution of polyP to survival, cell proliferation, and motility involved in wound healing in human skin keratinocytes, (3) the use of polyP-containing platelet-rich plasma (PRP) to promote wound healing in acute and chronic wounds, including burns, and (4) the use of polyP-containing PRP in excisional wound models to promote faster healing. While polyP shows promise as a therapeutic agent to accelerate healing for acute and chronic wounds, the molecular mechanisms as a potent modulator of the wound healing process remain to be elucidated.

Keywords

  • inorganic polyphosphate
  • wound healing
  • keratinocytes
  • platelet-rich plasma

1. Introduction

PolyP is a simple prebiotic molecule that varies in chain length between three and several thousand inorganic phosphates linked by phosphoanhydride bonds (Figure 1A). It is continuously synthesized from ATP or GTP and degraded by cellular enzymes in bacteria [1, 2, 3, 4, 5] and eukaryotes, yet its pleiotropic functions remain to be clarified. In bacteria, but not mammalian cells, the enzymes that catalyze these activities have been identified [6, 7, 8, 9, 10]. polyP is synthesized following osmotic, oxidative, UVB, or other cellular stresses, and augments bacterial survival [11, 12, 13, 14, 15, 16]; four roles have been proposed: an energy source, chelation of metal ions, storage of phosphate and response to cellular stresses. Necessary for bacterial survival, polyP is implicated in essential biological processes in prokaryotes including stress response, motility, biofilm formation [17, 18, 19, 20, 21, 22], virulence, sporulation, and quorum-sensing [21, 22, 23]. Enzymes involved in polyP synthesis and degradation have been isolated and characterized in bacteria and other lower eukaryotes, and maintain tight control of polyP levels, as might be expected for a polymer controlling vital biological processes.

Figure 1.

(A) Schematic of linear structure of polyP, and its synthesis by polyphosphate kinase (PPK) and degradation by exopolyphosphatases (PPX1) or endopolyphosphatases (PPN). Constitutive (B and D) or Tet-inducible PPX1 (C and E) expression slows wound healing in cultured human keratinocytes (modified with permission from [33]).

1.1 PolyP in prokaryotes

Drawing from the cellular ATP pool, bacterial polyphosphate kinase (PPK1) catalyzes the reversible transfer of the terminal γ-phosphate of ATP to polyP [24], whereas PPK2, expressed in other prokaryotes, transfers the terminal inorganic phosphate (Pi) from polyP to GDP to form GTP [4, 25, 26]. In contrast, the exopolyphosphatase PPX1 hydrolyzes polyP into phosphate monomers, thus maintaining phosphate homeostasis [5]. PPK levels and activity are tightly regulated, maintaining steady-state polyP concentrations in the bacterial cytosol at low micromolar levels, even in mutant strains deficient in PPX [27]. PolyP synthesis is upregulated during nutrient deprivation [28, 29], or during osmotic [28], acidic pH [30], oxidative [31], or heat [32] stresses, potentially depleting cellular ATP pools by converting millimolar levels of ATP to long polyP chains (>1000 Pi) [31]. polyP levels are measured using enzyme-based assays that employ ppk to generate ATP from ADP, using luciferase as a reporter [33]. In another assay, cells are labeled with 32Pi; polyP is isolated and hydrolyzed with PPX1, and thin-layer chromatography or phosphoimage analysis is performed [17]. Toluidine blue binding assays are effective for different chain-lengths but are relatively insensitive. Recently, a rapid and simple method has been described [34, 35].31P-NMR Spectroscopy is an effective and accurate method for measuring polyP in intact cells [36]. Electron ionization mass spectrometry [37], cryoelectron tomography and spectroscopy imaging [38] have also been employed. Protein affinity labeling in vivo uses the affinity of a recombinant polyP-binding domain of E. coli PPX1 (PPXbd) [39], which we and other investigators have used to specifically inhibit the function of polyP.

Not surprisingly, ppk mutants are extremely sensitive to environmental stresses [13, 31, 32, 40, 41], and exhibit reduced motility, virulence, and biofilm production [42]. ppk gene expression is regulated by σ38, a transcriptional regulator for late stationary phase genes [43] and polyP, in turn, amplifies its own synthesis by inducing transcription of the gene encoding σ38 (RpoS) [29, 41, 42]. In response to oxidative or heat stress, polyP synthesis is regulated at a transcriptional and/or post-translational level, as PPK synthesis and levels are altered by antisense RNA that target ppk mRNA transcripts [44] as well as transient inactivation of PPX by stress-sensitive regulators that allow polyP levels to remain high until normal conditions are restored [45].

1.2 PolyP in eukaryotes

While polyP is found in eukaryotes from protists to mammalian cells [11], the mechanism of polyP synthesis remains largely unknown for most eukaryotic organisms [1, 45], except for S. cerevisiae, where the vacuolar transporter chaperone 4 (VTC4) synthesizes polyP from ATP and then transports the polymer into vacuoles [46, 47]. Vacuolar polyP maintains phosphate homeostasis by appropriating phosphate during growth in phosphate-rich conditions [46], and releases phosphate during the cell cycle to provide precursors for DNA replication [48].

There is no sequence or structural homology between the polyP-synthesizing enzymes PPK1, PPK2, or VTC4, and homologues have yet to be found in higher eukaryotes [1]. While phylogenetic analysis of the prokaryotic branch reveals no clear homologues of E. coli PPK in a large number of polyP-synthesizing species [49], a few enzymes have been identified that use polyP as phosphate donor in reactions that can be reversed in the presence of excess substrate in vitro [49, 50]. In the absence of a polyP-synthesizing enzyme, polyP may be synthesized by the mitochondrial proton-motive force [51], in a complex process involving intact mitochondrial membranes [52]. Decreased polyP production resulting from depolarization of the mitochondrial membrane [52, 53] suggests that this may be a spontaneous process that does not need catalysis. Alternatively, inositol phosphates have also been implicated in polyP metabolism, since polyP levels are diminished in cells lacking the enzyme that synthesizes highly phosphorylated inositols [54, 55, 56, 57]. Similar to polyP synthesis, polyP-degrading enzymes, such as yeast PPX1, have been found in lower eukaryotes, while mammalian polyP-specific degradation enzymes are mostly uncharacterized. However, h-prune, which regulates cell migration, also acts as a exopolyphosphatase for short-chain polyP in vitro [58, 59].

Subcellular fractionation, immunofluorescent staining, and biochemical quantification reveal that polyP is localized to the nucleus, cell membrane, cytoplasm, and intracellular organelles in mammalian cells. It is specifically enriched in nucleoli, acidocalcisomes (organelles rich in protons, calcium (Ca2+), and phosphorus), and mitochondria [11, 60, 61, 62, 63]. In the brain, astrocytes secrete polyP, which is taken up by neurons, indicating both intra- and extracellular localization [64, 65]. Similar to vesicular packaging of ATP, astrocytes release polyP via exocytosis from vesicular nucleotide transporter (VNUT)-containing vesicles [65]. A putative G protein-coupled receptor in D. discoideum mediates cell surface binding of extracellular polyP, which as a signaling molecule, elicits differential effects on cell-substratum adhesion and cytoskeletal F-actin levels [66].

Eukaryotic polyP levels are in the 20–100 μM range (expressed as Pi concentration), with chain lengths ranging from 50 to 800 Pi residues (rat tissues) [67], averaging ~80 Pi residues in human platelets [68] to 200 Pi residues in yeast [67], compared to bacterial polyP, which can range up to thousands of Pi units long [53]; however, up to 130 mm medium-sized polyP chains are stored in dense granules in thrombocytes and mast cells [62, 68, 69]. Brain tissue exhibits among the highest polyP levels (~100 µm), which drop with age and neurodegenerative disease [11, 70, 71, 72], consistent with the role of polyP in stabilizing protein unfolding intermediates as amyloid-like precursors [32]. High levels of polyphosphate are also found in osteoblast matrix vesicles, the initial sites of bone mineral formation [73]. PolyP concentrations and chain lengths are dynamic, and depend on growth conditions of cells; for example in Plasmodia, polyP has an average chain length of 100 Pi, which is degraded to 10 Pi during sporulation [67].

Studies on the myriad roles of polyP in higher eukaryotes have recently gained momentum. PolyP is directly or indirectly involved in diverse cell processes, including control of cell bioenergetics, signal transduction, activation of the mitochondrial permeability transition pore (mPTP), Ca2+-signaling [74, 75], and maintenance of the mitochondrial membrane potential [74]. Associated with mPTP [74] and voltage-gated channels, polyP regulates neuronal excitability [76] and astroglial signaling [64]. About 39% of intracellular polyP pools in astrocytes are in mitochondria [77], playing a role in bioenergetics [52, 77] and Ca2+-handling [74, 78, 79]. As a signaling molecule, polyP released from astrocytes can mediate the physiological response to brain hypoxia [65].

In addition to its role as a gliotransmitter in the autonomic nervous system [64], the polymer also interacts with a variety of proteins, such as mammalian target of rapamycin (mTOR), fibroblast growth factor (FGF)-2, TRPM8, integrin β1, and glycosomal and ribosomal proteins and enzymes, consequently modulating cell survival and cell growth [80, 81, 82, 83, 84, 85, 86]. A fascinating finding of several recent studies is that polyP can covalently and non-enzymatically modify a small number of specific proteins in yeast [81] and humans containing lysine residues located in poly-acidic, serine, and lysine-rich (PASK) motifs, some of which are involved in ribosome biogenesis [87].

PolyP is involved in mTOR signaling, cell proliferation, and apoptosis [74, 80], and stimulates the mTOR pathway [80] at concentrations normally found in mammalian cells (0.15–1.5 mM); [11], suggesting a role for the polymer in mammalian cell proliferation. By promoting release of translation initiation factor eIF4E, mTOR stimulates initiation of translation, particularly proteins involved in cell growth and proliferation. PolyP also enhances the mitogenic activity of FGF-2 by promoting its binding to cell surface receptors [83]. PolyP appears to regulate apoptosis by inducing activation of caspase-3 in human plasma cells [88]. This polyanion also chelates metals, such as manganese and cadmium, blocking metal-induced cell damage [89, 90].

Other roles for polyP in mammalian cells include coagulation via activation of blood clotting factor XII [69], inflammation [91] as well as Ca2+ chelation for bone mineralization and osteogenic differentiation [92]. The polymer also contributes to pro-inflammatory responses upon release from mast cells [62]. Finally, serving as a stabilizing scaffold for protein-folding intermediates, polyP was recently shown to work as a protein-like chaperone protecting cells against stress-induced protein aggregation [93].

Advertisement

2. The contribution of polyP to cell survival, proliferation, and motility involved in wound healing in skin keratinocytes

Inorganic polyP shows promise in different phases of wound healing, including hemostasis and re-epithelialization, as polyP is a normal component of different cells that play a role in this process, including platelets, dermal fibroblasts, and keratinocytes. The use of polyP as a therapeutic for acute and chronic wounding has begun to garner interest, in part because of experiments elucidating its role in wound healing [33], as well as in hemostasis [69, 94, 95, 96, 97, 98, 99]. We have recently shown a role for polyP in the response to UV survival, cell motility, and wound healing [33]. Addition of exogenous polyP increased the rate of wound healing in standard scratch wound assays in vitro [33].

Whereas candidates for mammalian polyP metabolism have been shown to exhibit additional enzymatic activities [59], more specific polyphosphatases have been identified in lower eukaryotes including yeast, trypanosomes, and Dictyostelium. We therefore used exopolyphosphatase derived from S. cerevisiae (ScPPX1) to target intracellular polyP in human skin keratinocytes, an obvious choice for UV resistance, motility, and wound healing. The functions of polyP in the response of keratinocytes to UVB or wounding was studied by expressing ScPPX1, which selectively breaks down endogenous inorganic polyP, and not phosphoproteins, DNA, RNA, or nucleotide mono-, di-, or triphosphates [6]. Cells depleted of intracellular polyP by ScPPX1 expression exhibited increased sensitivity to UVB via enhanced apoptosis, and impaired wound healing [33]. Human keratinocytes stably expressing constitutive HisPPX1 or tetracycline (Tet)-inducible HisPPX1 were used to deplete cells of endogenous polyP, and study its role in wound healing assays performed on confluent monolayers, mimicking cell re-epithelialization during wound healing in vivo. RT-PCR and immunoblot analysis confirmed PPX1 expression in stable HisPPX1-expressing cells or in the presence of Tet (Figure 1B and C). Scratch gaps demonstrate marked attenuation of wound healing following constitutive HisPPX1 or Tet-induced expression (Figure 1D and E).

Since keratinocyte proliferation and migration are crucial to re-epithelialization during wound healing, the contribution of polyP to cell growth and motility involved in wound healing was next determined. Vector control cells exhibited significantly higher rates of cell growth as well as BrdU incorporation into newly synthesized DNA in cells at the wound edge, compared with polyP-depleted HisPPX1-expressing cells (Figure 2). Further, real time monitoring and measurement of cell motility performed in an xCelligence impedance-based system revealed significant decreases in cell motility in polyP-depleted keratinocytes (Figure 3). These results demonstrate that polyP depletion by either constitutive or inducible expression of PPX1 retards the rate of wound healing in human skin keratinocytes, by decreasing cell proliferation and motility. To determine if the loss of endogenous polyP can be supplemented with exogenous extracellular polyP, ScPPX1-expressing cells were grown in the presence of different concentrations of polyP, or with polyP-rich platelet lysate (next section). Exogenously added polyP was found to accelerate wound healing in human keratinocytes in polyP dose-response experiments on confluent monolayers of keratinocytes subjected to scratch wound healing assays (Figure 4). Interestingly, both intracellular and extracellular polyP dose-dependently increased the rate of wound healing in vitro.

Figure 2.

Constitutive PPX1 expression decreases growth rate of keratinocytes. Viable cell counts were performed over 11 days, and growth curves plotted for HisPPX1-expressing cells compared to vector cells (top). HisPPX1 and vector controls were subjected to scratch assays and proliferation was measured by in situ BrdU incorporation in cells at the wound edge (middle and bottom; modified with permission from [33]).

Figure 3.

Real-time monitoring and measurement of cell motility was performed in an xCelligence impedance-based system. Total number of cells attached to the bottom chamber were measured every 15 min over 24 hours, and are shown as technical duplicates, with assays repeated twice (left). Percentage of cell migration at the 24-hour time-point based on 100,000 cells plated at the top chamber (right; modified with permission from [33]).

Figure 4.

PolyP dose-dependently accelerates wound healing in cultured human keratinocyte, using polyP at 1 μM (left) or 10 μM (right; modified with permission from [33]).

Advertisement

3. The use of polyP-containing PRP to promote wound healing in acute and chronic wounds, including burns

Wound healing is a highly coordinated process involving biochemical and physiological interplay of keratinocytes and fibroblasts to restore skin integrity. Platelets are components of blood responsible for blood clotting and wound healing. Although the importance of platelets in wound healing has been extensively studied, the bioactive substance playing a major role in skin re-epithelialization during wound healing is still unclear. PRP has been shown to support the survival and proliferation of human keratinocytes [100], and is currently used as therapeutic for both acute and chronic wounds (for review see [101]). Discovery of the release of growth factors triggered an interest in using PRP for wound healing, and platelet lysates have been examined as a replacement for using fetal bovine serum in cell cultures, which may contain contaminants such as prions, or elicit an unwanted immune response in patients. Platelet-rich lysates derived from platelets are a by-product of blood preparation, and are thus inexpensive. Most studies focused on lysates for mesenchymal stromal cell culture for cell therapy, in which platelets are activated by thrombin and CaCl2, or by freeze-thaw.

In recent years, PRP has gained traction in many different specialties including in dermatology where it is used to treat acne [102], scarring [103], and alopecia [104, 105], in regenerative medicine where it is used to treat acute and chronic injuries to bone and cartilage [106, 107], in orthopedics and sports medicine where it is used to treat rotator cuff tears, osteoarthritis of the knee, hamstring injuries, and Achilles tendinopathy [108, 109, 110], in dentistry where it is used during tooth extractions, periodontal surgery, and dental implant surgery [111, 112], and more recently in wound healing to promote enhanced healing [113, 114, 115, 116, 117, 118]. PRP is an autologous blood product generated from multiple rounds of centrifugation that serve to concentrate the number of platelets in plasma. PRP contains high concentrations of growth factors such as platelet-derived growth factor (PDGF), epidermal growth factor (EGF), and transforming growth factor beta (TGFβ) compared to plasma and whole blood [119, 120]. It also contains higher levels of pro- and anti-inflammatory cytokines that promote enhanced healing. Lastly, PRP is known to contain inorganic polyP which is continually synthesized from ATP or GTP, and is degraded by cellular enzymes in bacteria and eukaryotes.

The role of polyP secreted by platelets and present in PRP on cell proliferation and wound healing was investigated in human HaCaT keratinocytes co-transduced with either ScPPX1 or vector control, along with DsRed or GFP, respectively, as fluorescent markers in order to visualize and track cells that have reduced or normal levels of polyP. Cells stably expressing fluorescent-tagged DsRed-PPX1 or GFP-empty vector were incubated with platelet lysate (4%) supplemented with or without exogenous pure polyP (1 μM). In both vector-GFP control and polyP-depleted PPX1-DsRed cells treated with polyP, platelet lysate, or platelet lysate + polyP, cell growth curves revealed a significant increase in cell proliferation compared to untreated controls (data to be published elsewhere). PolyP quantification in platelet lysates using a micromolar polyP assay kit showed that a 4% platelet lysate contains ~8 μM polyP, which was within the range used for exogenously added polyP. This assay measures increase in fluorescence intensity (emission 550 nm, excitation 415 nm) of a PPD dye upon binding to polyP.

Cell migration/scratch assays were performed on PPX1-DsRed or vector-GFP control keratinocytes to assess the effects on wound healing and cell motility. Fluorescent pictures were taken at 10 min intervals for 36 hours using an EVOS FL time-lapse imaging system, and gap closure was quantified by Image J. In both GFP-vector cells and PPX1-expressing cells, the rate of wound closure in the scratch assays were significantly increased when cells were incubated either with platelet lysate alone, polyP alone, or both (data to be published elsewhere). These results together indicate that exogenous polyP, delivered either purified or from platelet-enriched plasma, can accelerate wound healing.

To assess whether the increased rate of wound healing is attributable to polyP in platelet lysates, specific polyP inhibitors (polyP-binding protein PPXbd or UHRA-9, a kind gift from Dr. James Morrissey) were utilized in wound healing assays. PPXbd, a recombinant polyP-binding domain of E. coli exopolyphosphatase, binds to platelet-derived polyP and blocks FXI activation, thrombin and fibrin generation, and consequently, inhibiting polyP procoagulant activity [99]. Interestingly, the enhanced rates of wound healing in vector control or polyP-depleted ScPPX-expressing cells induced by supplementation with exogenous extracellular polyP from pure polyP or in platelet lysates, was completely reversed by addition of the polyP inhibitors PPXbd or UHRA-9 (data to be published elsewhere). PolyP secreted by platelets and present in platelet lysate or PRP may therefore play an essential role in re-epithelialization during wound healing.

3.1 Chronic wounds

The acceleration of wound healing is of paramount importance in the setting of acute and chronic wounds, as well as burn wounds. Open chronic wounds are a significant cause of additional morbidity in patient populations that already have a plethora of comorbidities [121]. Significant improvements in complete healing were reported in chronic wounds treated with PRP compared to no topical treatments in a 2011 systematic review and meta-analysis on the use of PRP in acute and chronic wounds [101]. Another review of PubMed and Cochrane databases found significant benefit of PRP for diabetic chronic wounds, specifically in wounds unresponsive to standard of care treatment options [113]. A third systematic review of nine randomized controlled clinical trials (RCT) suggested that well-designed high-powered RCTs are needed to demonstrate increased wound healing with PRP treatment [122].

Treatment of 56 patients with diabetic foot ulcers with twice weekly applications of PRP resulted in complete healing in 86% of patients in the treated groups vs. only 68% in the control group [123]. Animal models using exosomes derived from PRP for full thickness skin wounds in a diabetic rat model also showed increased healing, as well as increased fibroblast proliferation and migration [124]. Platelet-rich fibrin also improved diabetic animal skin wound healing [125]. Overall, while there is no consensus on this treatment modality in chronic wounds, it is becoming widely used, and many trials seek to understand its potential beneficial effects. Improvements in open wound area have been shown in a number of animal and clinical studies.

3.2 Acute wounds

Meta-analysis of rodent and non-rodent studies using a systematic review conducted under preferred reported items for systematic review of interventions (PRISMA) guidelines indicated that the treatment of wounds with PRP resulted in reduction of open wound area [126]. In addition to its role in wound healing, PRP reduced complications such as wound infection, exudate (mass of cells and fluid that seeps out of a wound), drainage, and hematoma formation [101]. PRP and PRP with keratinocyte and fibroblast cells were shown to increase re-epithelialization at 7–14 days post-injury in mouse models, compared to non-treated controls [127]. In full thickness porcine wounds treated with the secreted proteins of PRP, wound re-epithelialization and collagen deposition were significantly increased in treated animals vs. saline controls [128]. Thus, PRP may improve wound healing in acute surgical wounds by secreting growth factors that support local microenvironments that promotes healing [129]. PRP’s effectiveness has also been shown in bone grafting, cartilage regeneration, and non-cutaneous surgical procedures. The impact of PRP on normal and damaged (derived from chronic ulcers or irradiated) fibroblasts have been described [130]. In addition, despite the lack of reproducibility of platelet concentrations due to differences in manufacturer-specific protocols for PRP preparation and differences in treatment methodologies, PRP has been shown to affect fibroblast proliferation and migration in a number of in vitro studies. As with chronic wounds, it is unclear why some studies, but not others, show a beneficial effect of PRP treatment.

3.3 Burns

PRP has been used as a topical treatment to accelerate wound healing in burn wounds, however, like in chronic and acute wounds, its use is still debated due to conflicting results [114]. Some papers recommend its use [115, 117, 118, 131, 132, 133, 134, 135], while others have shown non-significant changes in outcomes after treatment with PRP, and advise caution in using it in a wide-spread manner [114, 136, 137, 138]. A review of PRP for burns concluded that PRP may be useful in regeneration of dermal structures, increasing graft-take, and increasing re-epithelialization, but recommended further research on characterization of the mechanisms by which PRP can improve burn wound healing, donor site healing, and scar outcomes [139].

The use of side-by-side treatment of a split thickness skin graft (STSG) donor site with standard treatment or with PRP showed complete re-epithelialization in the PRP-treated side at day 11 vs. day 13 for the control. Histological samples taken from these healing wounds, and by H and E staining revealed increased epidermal thickness in PRP-treated wounds, as well as a significant increase in the number of blood vessels. After platelet concentrate in conjunction with STSG was used for deep burns, monitoring of viscoelastic properties of the resultant scars over 12 months revealed that the skin’s return to normal viscoelastic properties was accelerated in burns treated with PRP compared to controls [133]. Compared to historic institutional standard of care controls, treatment of deep partial thickness (DPT) burns using PRP applied with the autograft during skin grafting, pain scores, inflammation, pruritis (itchiness), cosmesis of the scar, and perfusion all showed improved outcomes [135].

Animal models of burn injury in rats treated with topical PRP or control showed that PRP treatment resulted in increased hydroxyproline, decreased inflammatory cells infiltration, but no difference in fibroblast collagen production or angiogenesis [134]. In a rat animal model of DPT burns, PRP was effective in increasing % wound closure, but showed little effectiveness in the full thickness injury group. PRP treatment resulted in increased neo-epidermal thickness at day 21, as well as decreases in CD31, 68, and 163, TGFβ1, MMP2, and MPO+ cells indicating an increased resolution of inflammation [118]. PRP injection in burn wound scars in a rat animal model of burn injury also showed pain-associated markers to be decreased with treatment [132]. While positive healing was observed in most models, in a recent study using a swine model of burn, tangential excision, and grafting with or without PRP, PRP showed similar effect on re-epithelialization and scarring in full thickness wounds compared to control wounds [136].

In a randomized clinical trial from 2018, 27 patients with DPT burns that did not get autografted were treated with lyophilized PRP powder, and showed a significant increased percent wound closure at 3 weeks in the treated group compared to control. Additionally, the infection rate in the PRP group was 26%, while 33% of control patients had postoperative infections [115]. PRP’s concentrated secretion of growth factors may include basic fibroblast growth factor, epidermal growth factor, platelet-derived growth factor (PDGF), insulin-like growth factor, transforming growth factor β (TGFβ1), and vascular endothelial growth factor (VEGF) as probable mechanisms by which it can accelerate healing [139]. However, quantification of growth factors TGFβ1, PDGF-AA, and VEGF in a cohort of five burn patients compared to five healthy volunteers showed comparable levels of growth factors in the PRP from burn patients and health volunteers. Thus, there may be an additional factor in PRP that is possibly altered in burn patients (or patients with other pathologies such as diabetes or other conditions that would lead them to have surgical procedures yielding acute wounds) that may contribute to its success in treating some wounds and failure in others. Due to the effect burns have on the pathophysiology of blood coagulopathy [140, 141, 142] and capillary endotheliopathy [143, 144] after injury, it is reasonable to assume that platelets from burn patients may have differing levels of polyP. Associating this data with what is known about polyP, its ubiquitous presence in all prokaryotic and eukaryotic organisms, and its role in the response to cellular stress, it was hypothesized that polyP may contribute to wound healing.

Advertisement

4. PolyP and polyP-containing platelet rich plasma accelerates re-epithelialization in vitro and in vivo

A HaCaT keratinocyte polyP-depleted cell line and vector control was used in growth curves and scratch assays to evaluate polyP, platelet lysate, or combined treatment to accelerate wound healing in vitro. PolyP-containing PRP was also evaluated as a treatment in a splinted model of excisional wounding in vivo. Exogenous polyP was also spiked into PRP to assess its role. Treatment with the polyP-containing treatments increased cell growth and attenuated open wound area in vitro (p < 0.001). Addition of a polyP inhibitor abrogated these effects (p < 0.0001). PRP-treated wounds re-epithelialized faster compared to untreated wounds when analyzed at Days 3 and 5 (n = 6 wounds, p < 0.05). Re-epithelialization was further enhanced by exogenous polyP addition to PRP as evidenced by elongated epithelial tongues (Figure 5) in the low and high dose PRP + polyP treatment groups compared to PRP alone (n = 8 wounds, p < 0.05; data to be published elsewhere). Due to its autologous nature, PRP serves as a safe and efficacious option for accelerating wound healing, and may be enhanced by the exogenous addition of polyP.

Figure 5.

Untreated or treated wounds were excised on day 5 and fixed in formalin, paraffin embedded, sectioned, and stained with H&E. Sections were imaged, and composited to create an image with areas of normal skin on both sides with the epithelial tongue (orange arrow) protruding from each side of normal skin. The epithelial gap is demarcated by the black arrow where no epithelium is present. Scale bar = 200 μm. Representative histology of untreated (A), PRP only treated (B), PRP + 10 μM polyP-treated wounds are shown. Epithelial tongue length was measured with Image J and quantified.

4.1 PRP-treated wounds heal faster than controls in vivo

PRP was generated based on a previously published protocol [145]. Briefly, whole blood was collected, and centrifuged to create platelet-poor plasma (PPP) and a pellet of platelets. PPP was then removed and the platelets were resuspended and activated with thrombin and calcium chloride (CaCl2) to form a “biobandage”-like gel. Whole blood, packed RBCS, and PRP were stained with Wright and Giemsa stains to confirm PRP platelet concentration. A murine model of full thickness excisional wound healing was used where 6 mm punch biopsies were created on dorsal flanks of C57BL/6 mice [146, 147]. Wounds were splinted to encourage healing by re-epithelialization as opposed to contracture. They were subsequently treated with PRP, or no treatment was applied. Tegaderm dressing was applied and kept in place for 3 days. Pictures were taken at days 3, 4, 5, 6, or 7. By day 7, wounds were mostly closed.

In a second experiment, wounded mice were divided into four treatment groups: untreated, PRP only, PRP + 10 μM polyP (low dose) and PRP + 100 μM polyP (high dose). Doses of polyP were calculated by examining historical data from the literature on platelet levels, as well as polyP concentrations in platelets. The mean platelet count for C57BL/6 mice is 9.85 ± 1.40 × 1011 platelets/L, however, because there are no reliable reports of polyP concentrations per platelet in mice, human platelet counts and polyP concentrations were extrapolated for this experiment [148]. Human platelet concentrations range from 1.5 to 4.5 × 1011 platelets/L [149]. It is also known that platelets contain 0.74 ± 0.08 μmol polyP/1 × 1011 platelets [150]. Therefore, whole blood should contain between 1 and 3 μM polyP, and PRP, which contains at least 3-fold higher levels of platelets compared to whole blood, should contain 3–9 μM of polyP. We added ~10 μM in the low dose group, and a 10× concentration compared to the low dose group for the high dose group (100 μM). Lyophilized polyP was reconstituted in dH2O to 1 M. PolyP took up 10% of the total treatment volume of PRP.

Treatments were applied on Day 0, and Tegaderm™ dressings stayed in place through day 3. On day 3 and 4, dressings were removed, and pictures were taken. On day 5, dressings were removed, pictures were taken, and wounds were excised with underlying fascia and were sewn into histological cassettes to retain wound orientation. These samples were then paraffin embedded and H&E stained. Sections were imaged, and epithelial tongue length was measured using Image J. Epithelial tongues were defined as new epithelium if there was no uninjured dermis underneath the epithelium.

Compared to whole blood, PRP contained a higher concentration of platelets (data to be published elsewhere). The splinted wound model was used to shift the healing towards re-epithelialization instead of the normal contraction observed in mice. PRP application was easily applied as a “bio-bandage” gel-like liquid (data to be published elsewhere). The 6 mm punch biopsies allowed for the creation of similar wound size between animal groups at day 0 (0.33 cm2 ± 0.13 vs. 0.39 ± 0.15 cm2, p = n.s.; data to be published elsewhere). At days 3 (0.09 ± 0.06 vs. 0.23 ± 0.13) and 5 (0.12 ± 0.07 vs. 0.25 ± 0.12) PRP-treated wounds had significantly smaller open wound areas compared to control animals (n = 6, p < 0.05). At days 6 and 7, this difference leveled off.

4.2 Exogenous spiking of PRP with polyP further accelerates healing in vivo

To further investigate the potential role of polyP in PRP, wounds were treated with PRP or with PRP with low or high dose-spiked polyP. Untreated wounds were largely open by day 5, while PRP treated wounds were smaller and contained newly formed epithelium (data to be published elsewhere). Spiking with low or high dose polyP further stimulated epithelialization, and wounds were smaller with increasing doses. By histomorphometric analysis, epithelial tongues can be seen by H&E staining. In untreated wounds, these tongues are small and shallow. PRP treatment results in a more proliferative epithelium that is thicker and longer than untreated samples. Spiking with low and high dose polyP creates longer epithelial tongues). Epithelial tongue measurement by Image J shows a significant decrease in tongue length in untreated and PRP only treated vs. PRP + high dose polyP (737.38 ± 121.21 and 925.55 ± 214.17 vs. 1186.91 ± 255.06 μM, n = 8, p < 0.0001, p < 0.05). PRP + high dose polyP-treated wounds also had significantly longer epithelial tongues compared to PRP + low dose polyP (n = 8, p < 0.05). PRP contains polyP at a concentration near 5 μM. The exogenous addition of polyP to the PRP promoted keratinocyte growth and proliferation, as is evidence by the increased epithelial tongue length with increasing doses of polyP administration.

Advertisement

5. Conclusion

PolyP plays key roles in essential biological processes in bacteria, and its increasing importance in eukaryotes is becoming apparent, including its participation in blood coagulation and wound healing. Recent advances in measurement and localization of polyP, along with our growing understanding of polyP metabolism and its interaction with specific proteins allows us to begin to analyze mechanisms responsible for cell-specific roles of polyP. Our ability to regulate polyP in eukaryotic cells opens possibilities for therapeutic intervention. Future work related to wound healing should be aimed at investigating the specific roles of intra- and extracellular polyP in keratinocytes, as well as the potential importance of polyP in other skin cells, including dermal fibroblasts, as these cells make up the majority of the skin. As polyP is also secreted by activated platelets and is important for normal blood clotting, the application of polyP-containing PRP as a biologic dressing may positively contribute to wound healing. We have completed two clinical trials using platelet-rich plasma for wound healing, a phenomenon that may be explained by the presence of polyP. PolyP levels and chain lengths should also be quantified in healthy and pathologic conditions in order to assess appropriate levels when treating acute or chronic wounds in the future.

Advertisement

Acknowledgments

Authors are grateful for the technical assistance of Sixian Song in the scratch gap assays, and Dr. James Morrissey (University of Michigan Medical School) for the generous gift of polyP, and its inhibitors PPXbd and UHRA-9, used in their studies. This work was partially supported by the NIH STTR grant 1R41ES026908 (to DSR), and the office of the Dean of Research, Georgetown University School of Medicine.

Advertisement

Conflict of interest

The authors have no conflicts of interest to declare.

References

  1. 1. Rao NN, Gomez-Garcia MR, Kornberg A. Inorganic polyphosphate: Essential for growth and survival. Annual Review of Biochemistry. 2009;78:605-647. DOI: 10.1146/annurev.biochem.77.083007.093039
  2. 2. Brown MR, Kornberg A. The long and short of it: Polyphosphate PPK and bacterial survival. Trends in Biochemical Sciences. 2008;33(6):284-290. DOI: 10.1016/j.tibs.2008.04.005
  3. 3. Gomez-Garcia MR, Kornberg A. Formation of an actin-like filament concurrent with the enzymatic synthesis of inorganic polyphosphate. Proceedings of the National Academy of Sciences of the United States of America. 2004;101(45):15876-15880. DOI: 10.1073/pnas.0406923101
  4. 4. Zhang H, Ishige K, Kornberg A. A polyphosphate kinase (PPK2) widely conserved in bacteria. Proceedings of the National Academy of Sciences of the United States of America. 2002;99(26):16678-16683. DOI: 10.1073/pnas.262655199
  5. 5. Akiyama M, Crooke E, Kornberg A. An exopolyphosphatase of Escherichia coli. The enzyme and its ppx gene in a polyphosphate operon. The Journal of Biological Chemistry. 1993;268(1):633-639
  6. 6. Wurst H, Kornberg A. A soluble exopolyphosphatase of Saccharomyces cerevisiae. Purification and characterization. The Journal of Biological Chemistry. 1994;269(15):10996-11001
  7. 7. Wurst H, Shiba T, Kornberg A. The gene for a major exopolyphosphatase of Saccharomyces cerevisiae. Journal of Bacteriology. 1995;177(4):898-906
  8. 8. Lichko LP, Kulakovskaya TV, Kulaev IS. Inorganic polyphosphate and exopolyphosphatase in the nuclei of Saccharomyces cerevisiae: Dependence on the growth phase and inactivation of the PPX1 and PPN1 genes. Yeast. 2006;23(10):735-740. DOI: 10.1002/yea.1391
  9. 9. Sethuraman A, Rao NN, Kornberg A. The endopolyphosphatase gene: Essential in Saccharomyces cerevisiae. Proceedings of the National Academy of Sciences of the United States of America. 2001;98(15):8542-8547. DOI: 10.1073/pnas.151269398
  10. 10. Luginbuehl E et al. The exopolyphosphatase TbrPPX1 of Trypanosoma brucei. BMC Microbiology. 2011;11:4. DOI: 10.1186/1471-2180-11-4
  11. 11. Kumble KD, Kornberg A. Inorganic polyphosphate in mammalian cells and tissues. The Journal of Biological Chemistry. 1995;270(11):5818-5822
  12. 12. Rao NN, Kornberg A. Inorganic polyphosphate regulates responses of Escherichia coli to nutritional stringencies, environmental stresses and survival in the stationary phase. Progress in Molecular and Subcellular Biology. 1999;23:183-195
  13. 13. Crooke E et al. Genetically altered levels of inorganic polyphosphate in Escherichia coli. The Journal of Biological Chemistry. 1994;269(9):6290-6295
  14. 14. Alcantara C et al. Accumulation of polyphosphate in Lactobacillus spp. and its involvement in stress resistance. Applied and Environmental Microbiology. 2014;80(5):1650-1659. DOI: 10.1128/AEM.03997-13
  15. 15. Nikel PI et al. Accumulation of inorganic polyphosphate enables stress endurance and catalytic vigour in Pseudomonas putida KT2440. Microbial Cell Factories. 2013;12:50. DOI: 10.1186/1475-2859-12-50
  16. 16. Singh R et al. Polyphosphate deficiency in Mycobacterium tuberculosis is associated with enhanced drug susceptibility and impaired growth in Guinea pigs. Journal of Bacteriology. 2013;195(12):2839-2851. DOI: 10.1128/JB.00038-13
  17. 17. Rao NN, Liu S, Kornberg A. Inorganic polyphosphate in Escherichia coli: The phosphate regulon and the stringent response. Journal of Bacteriology. 1998;180(8):2186-2193
  18. 18. Rao NN, Kornberg A. Inorganic polyphosphate supports resistance and survival of stationary-phase Escherichia coli. Journal of Bacteriology. 1996;178(5):1394-1400
  19. 19. Rashid MH, Kornberg A. Inorganic polyphosphate is needed for swimming, swarming, and twitching motilities of Pseudomonas aeruginosa. Proceedings of the National Academy of Sciences of the United States of America. 2000;97(9):4885-4890. DOI: 10.1073/pnas.060030097
  20. 20. Rashid MH, Rao NN, Kornberg A. Inorganic polyphosphate is required for motility of bacterial pathogens. Journal of Bacteriology. 2000;182(1):225-227
  21. 21. Shi X, Rao NN, Kornberg A. Inorganic polyphosphate in Bacillus cereus: Motility, biofilm formation, and sporulation. Proceedings of the National Academy of Sciences of the United States of America. 2004;101(49):17061-17065. DOI: 10.1073/pnas.0407787101
  22. 22. Rashid MH et al. Polyphosphate kinase is essential for biofilm development, quorum sensing, and virulence of Pseudomonas aeruginosa. Proceedings of the National Academy of Sciences of the United States of America. 2000;97(17):9636-9641. DOI: 10.1073/pnas.170283397
  23. 23. Kim KS et al. Inorganic polyphosphate is essential for long-term survival and virulence factors in Shigella and Salmonella spp. Proceedings of the National Academy of Sciences of the United States of America. 2002;99(11):7675-7680. DOI: 10.1073/pnas.112210499
  24. 24. Ahn K, Kornberg A. Polyphosphate kinase from Escherichia coli. Purification and demonstration of a phosphoenzyme intermediate. The Journal of Biological Chemistry. 1990;265(20):11734-11739
  25. 25. Ishige K, Zhang H, Kornberg A. Polyphosphate kinase (PPK2), a potent, polyphosphate-driven generator of GTP. Proceedings of the National Academy of Sciences of the United States of America. 2002;99(26):16684-16688. DOI: 10.1073/pnas.262655299
  26. 26. Nocek B et al. Polyphosphate-dependent synthesis of ATP and ADP by the family-2 polyphosphate kinases in bacteria. Proceedings of the National Academy of Sciences of the United States of America. 2008;105(46):17730-17735. DOI: 10.1073/pnas.0807563105
  27. 27. Rudat AK et al. Mutations in Escherichia coli polyphosphate kinase that lead to dramatically increased in vivo polyphosphate levels. Journal of Bacteriology. 2018;200(6):pii: e00697-17. DOI: 10.1128/JB.00697-17
  28. 28. Ault-Riche D et al. Novel assay reveals multiple pathways regulating stress-induced accumulations of inorganic polyphosphate in Escherichia coli. Journal of Bacteriology. 1998;180(7):1841-1847
  29. 29. Shiba T et al. Inorganic polyphosphate and the induction of rpoS expression. Proceedings of the National Academy of Sciences of the United States of America. 1997;94(21):11210-11215
  30. 30. Mullan A, Quinn JP, McGrath JW. Enhanced phosphate uptake and polyphosphate accumulation in Burkholderia cepacia grown under low pH conditions. Microbial Ecology. 2002;44(1):69-77. DOI: 10.1007/s00248-002-3004-x
  31. 31. Gray MJ et al. Polyphosphate is a primordial chaperone. Molecular Cell. 2014;53(5):689-699. DOI: 10.1016/j.molcel.2014.01.012
  32. 32. Yoo NG et al. Polyphosphate stabilizes protein unfolding intermediates as soluble amyloid-like oligomers. Journal of Molecular Biology. 2018;430(21):4195-4208. DOI: 10.1016/j.jmb.2018.08.016
  33. 33. Simbulan-Rosenthal CM et al. Inorganic polyphosphates are important for cell survival and motility of human skin keratinocytes. Experimental Dermatology. 2015;24(8):636-639. DOI: 10.1111/exd.12729
  34. 34. Werner TP, Amrhein N, Freimoser FM. Novel method for the quantification of inorganic polyphosphate (iPoP) in Saccharomyces cerevisiae shows dependence of iPoP content on the growth phase. Archives of Microbiology. 2005;184(2):129-136. DOI: 10.1007/s00203-005-0031-2
  35. 35. Freimoser FM et al. Systematic screening of polyphosphate (poly P) levels in yeast mutant cells reveals strong interdependence with primary metabolism. Genome Biology. 2006;7(11):R109. DOI: 10.1186/gb-2006-7-11-r109
  36. 36. Zakrzewska J, Zizic M, Zivic M. The effect of anoxia on PolyP content of Phycomyces blakesleeanus mycelium studied by 31P NMR spectroscopy. Annals of the New York Academy of Sciences. 2005;1048:482-486. DOI: 10.1196/annals.1342.073
  37. 37. Choi BK, Hercules DM, Houalla M. Characterization of polyphosphates by electrospray mass spectrometry. Analytical Chemistry. 2000;72(20):5087-5091
  38. 38. Comolli LR, Kundmann M, Downing KH. Characterization of intact subcellular bodies in whole bacteria by cryo-electron tomography and spectroscopic imaging. Journal of Microscopy. 2006;223(Pt 1):40-52. DOI: 10.1111/j.1365-2818.2006.01597.x
  39. 39. Saito K et al. Direct labeling of polyphosphate at the ultrastructural level in Saccharomyces cerevisiae by using the affinity of the polyphosphate binding domain of Escherichia coli exopolyphosphatase. Applied and Environmental Microbiology. 2005;71(10):5692-5701. DOI: 10.1128/AEM.71.10.5692-5701.2005
  40. 40. Groitl B et al. Pseudomonas aeruginosa defense systems against microbicidal oxidants. Molecular Microbiology. 2017;106(3):335-350. DOI: 10.1111/mmi.13768
  41. 41. Dahl JU et al. The anti-inflammatory drug mesalamine targets bacterial polyphosphate accumulation. Nature Microbiology. 2017;2:16267. DOI: 10.1038/nmicrobiol.2016.267
  42. 42. Fraley CD et al. A polyphosphate kinase 1 (ppk1) mutant of Pseudomonas aeruginosa exhibits multiple ultrastructural and functional defects. Proceedings of the National Academy of Sciences of the United States of America. 2007;104(9):3526-3531. DOI: 10.1073/pnas.0609733104
  43. 43. Maciag A et al. In vitro transcription profiling of the sigmaS subunit of bacterial RNA polymerase: Re-definition of the sigmaS regulon and identification of sigmaS-specific promoter sequence elements. Nucleic Acids Research. 2011;39(13):5338-5355. DOI: 10.1093/nar/gkr129
  44. 44. Silby MW, Nicoll JS, Levy SB. Regulation of polyphosphate kinase production by antisense RNA in Pseudomonas fluorescens Pf0-1. Applied and Environmental Microbiology. 2012;78(12):4533-4537. DOI: 10.1128/AEM.07836-11
  45. 45. Kuroda A, Kornberg A. Polyphosphate kinase as a nucleoside diphosphate kinase in Escherichia coli and Pseudomonas aeruginosa. Proceedings of the National Academy of Sciences of the United States of America. 1997;94(2):439-442
  46. 46. Ogawa N, DeRisi J, Brown PO. New components of a system for phosphate accumulation and polyphosphate metabolism in Saccharomyces cerevisiae revealed by genomic expression analysis. Molecular Biology of the Cell. 2000;11(12):4309-4321. DOI: 10.1091/mbc.11.12.4309
  47. 47. Hothorn M et al. Catalytic core of a membrane-associated eukaryotic polyphosphate polymerase. Science. 2009;324(5926):513-516. DOI: 10.1126/science.1168120
  48. 48. Bru S et al. Polyphosphate is involved in cell cycle progression and genomic stability in Saccharomyces cerevisiae. Molecular Microbiology. 2016;101(3):367-380. DOI: 10.1111/mmi.13396
  49. 49. Wang L et al. Distribution patterns of polyphosphate metabolism pathway and its relationships with bacterial durability and virulence. Frontiers in Microbiology. 2018;9:782. DOI: 10.3389/fmicb.2018.00782
  50. 50. Nahalka J, Patoprsty V. Enzymatic synthesis of sialylation substrates powered by a novel polyphosphate kinase (PPK3). Organic & Biomolecular Chemistry. 2009;7(9):1778-1780. DOI: 10.1039/b822549b
  51. 51. Pestov NA, Kulakovskaya TV, Kulaev IS. Inorganic polyphosphate in mitochondria of Saccharomyces cerevisiae at phosphate limitation and phosphate excess. FEMS Yeast Research. 2004;4(6):643-648. DOI: 10.1016/j.femsyr.2003.12.008
  52. 52. Pavlov E et al. Inorganic polyphosphate and energy metabolism in mammalian cells. The Journal of Biological Chemistry. 2010;285(13):9420-9428. DOI: 10.1074/jbc.M109.013011
  53. 53. Kornberg A, Rao NN, Ault-Riche D. Inorganic polyphosphate: A molecule of many functions. Annual Review of Biochemistry. 1999;68:89-125. DOI: 10.1146/annurev.biochem.68.1.89
  54. 54. Lonetti A et al. Identification of an evolutionarily conserved family of inorganic polyphosphate endopolyphosphatases. The Journal of Biological Chemistry. 2011;286(37):31966-31974. DOI: 10.1074/jbc.M111.266320
  55. 55. Cordeiro CD, Saiardi A, Docampo R. The inositol pyrophosphate synthesis pathway in Trypanosoma brucei is linked to polyphosphate synthesis in acidocalcisomes. Molecular Microbiology. 2017;106(2):319-333. DOI: 10.1111/mmi.13766
  56. 56. Ghosh S et al. Inositol hexakisphosphate kinase 1 maintains hemostasis in mice by regulating platelet polyphosphate levels. Blood. 2013;122(8):1478-1486. DOI: 10.1182/blood-2013-01-481549
  57. 57. Hou Q et al. Inhibition of IP6K1 suppresses neutrophil-mediated pulmonary damage in bacterial pneumonia. Science Translational Medicine. 2018;10(435):pii: eaal4045. DOI: 10.1126/scitranslmed.aal4045
  58. 58. Carotenuto M et al. H-prune through GSK-3beta interaction sustains canonical WNT/beta-catenin signaling enhancing cancer progression in NSCLC. Oncotarget. 2014;5(14):5736-5749. DOI: 10.18632/oncotarget.2169
  59. 59. Tammenkoski M et al. Human metastasis regulator protein H-prune is a short-chain exopolyphosphatase. Biochemistry. 2008;47(36):9707-9713. DOI: 10.1021/bi8010847
  60. 60. Aschar-Sobbi R et al. High sensitivity, quantitative measurements of polyphosphate using a new DAPI-based approach. Journal of Fluorescence. 2008;18(5):859-866. DOI: 10.1007/s10895-008-0315-4
  61. 61. Jimenez-Nunez MD et al. Myeloma cells contain high levels of inorganic polyphosphate which is associated with nucleolar transcription. Haematologica. 2012;97(8):1264-1271. DOI: 10.3324/haematol.2011.051409
  62. 62. Moreno-Sanchez D et al. Polyphosphate is a novel pro-inflammatory regulator of mast cells and is located in acidocalcisomes. The Journal of Biological Chemistry. 2012;287(34):28435-28444. DOI: 10.1074/jbc.M112.385823
  63. 63. Angelova PR et al. In situ investigation of mammalian inorganic polyphosphate localization using novel selective fluorescent probes JC-D7 and JC-D8. ACS Chemical Biology. 2014;9(9):2101-2110. DOI: 10.1021/cb5000696
  64. 64. Holmstrom KM et al. Signalling properties of inorganic polyphosphate in the mammalian brain. Nature Communications. 2013;4:1362. DOI: 10.1038/ncomms2364
  65. 65. Angelova PR et al. Signal transduction in astrocytes: Localization and release of inorganic polyphosphate. Glia. 2018;66(10):2126-2136. DOI: 10.1002/glia.23466
  66. 66. Suess PM, Tang Y, Gomer RH. The putative G protein-coupled receptor GrlD mediates extracellular polyphosphate sensing in Dictyostelium discoideum. Molecular Biology of the Cell. 2019;30(9):1118-1128. DOI: 10.1091/mbc.E18-10-0686
  67. 67. Chen KY. Study of polyphosphate metabolism in intact cells by 31-P nuclear magnetic resonance spectroscopy. Progress in Molecular and Subcellular Biology. 1999;23:253-273
  68. 68. Ruiz FA et al. Human platelet dense granules contain polyphosphate and are similar to acidocalcisomes of bacteria and unicellular eukaryotes. The Journal of Biological Chemistry. 2004;279(43):44250-44257. DOI: 10.1074/jbc.M406261200
  69. 69. Muller F et al. Platelet polyphosphates are proinflammatory and procoagulant mediators in vivo. Cell. 2009;139(6):1143-1156. DOI: 10.1016/j.cell.2009.11.001
  70. 70. Gabel NW, Thomas V. Evidence for the occurrence and distribution of inorganic polyphosphates in vertebrate tissues. Journal of Neurochemistry. 1971;18(7):1229-1242
  71. 71. Cremers CM et al. Polyphosphate: A conserved modifier of amyloidogenic processes. Molecular Cell. 2016;63(5):768-780. DOI: 10.1016/j.molcel.2016.07.016
  72. 72. Lorenz B et al. Changes in metabolism of inorganic polyphosphate in rat tissues and human cells during development and apoptosis. Biochimica et Biophysica Acta. 1997;1335(1-2):51-60. DOI: 10.1016/S0304-4165(96)00121-3
  73. 73. Li L et al. Long-chain polyphosphate in osteoblast matrix vesicles: Enrichment and inhibition of mineralization. Biochimica et Biophysica Acta: General Subjects. 2019;1863(1):199-209. DOI: 10.1016/j.bbagen.2018.10.003
  74. 74. Abramov AY et al. Targeted polyphosphatase expression alters mitochondrial metabolism and inhibits calcium-dependent cell death. Proceedings of the National Academy of Sciences of the United States of America. 2007;104(46):18091-18096. DOI: 10.1073/pnas.0708959104
  75. 75. Seidlmayer LK et al. Inorganic polyphosphate is a potent activator of the mitochondrial permeability transition pore in cardiac myocytes. The Journal of General Physiology. 2012;139(5):321-331. DOI: 10.1085/jgp.201210788
  76. 76. Stotz SC et al. Inorganic polyphosphate regulates neuronal excitability through modulation of voltage-gated channels. Molecular Brain. 2014;7(1):42. DOI: 10.1186/1756-6606-7-42
  77. 77. Angelova PR et al. Role of inorganic polyphosphate in mammalian cells: From signal transduction and mitochondrial metabolism to cell death. Biochemical Society Transactions. 2016;44(1):40-45. DOI: 10.1042/BST20150223
  78. 78. Baev AY, Negoda A, Abramov AY. Modulation of mitochondrial ion transport by inorganic polyphosphate: Essential role in mitochondrial permeability transition pore. Journal of Bioenergetics and Biomembranes. 2017;49(1):49-55. DOI: 10.1007/s10863-016-9650-3
  79. 79. Elustondo PA et al. Mitochondrial permeability transition pore induction is linked to formation of the complex of ATPase C-subunit, polyhydroxybutyrate and inorganic polyphosphate. Cell Death Discovery. 2016;2:16070. DOI: 10.1038/cddiscovery.2016.70
  80. 80. Wang L et al. Inorganic polyphosphate stimulates mammalian TOR, a kinase involved in the proliferation of mammary cancer cells. Proceedings of the National Academy of Sciences of the United States of America. 2003;100(20):11249-11254. DOI: 10.1073/pnas.1534805100
  81. 81. Azevedo C, Livermore T, Saiardi A. Protein polyphosphorylation of lysine residues by inorganic polyphosphate. Molecular Cell. 2015;58(1):71-82. DOI: 10.1016/j.molcel.2015.02.010
  82. 82. Zakharian E et al. Inorganic polyphosphate modulates TRPM8 channels. PLoS ONE. 2009;4(4):e5404. DOI: 10.1371/journal.pone.0005404
  83. 83. Shiba T et al. Modulation of mitogenic activity of fibroblast growth factors by inorganic polyphosphate. The Journal of Biological Chemistry. 2003;278(29):26788-26792. DOI: 10.1074/jbc.M303468200
  84. 84. Segawa S et al. Probiotic-derived polyphosphate enhances the epithelial barrier function and maintains intestinal homeostasis through integrin-p38 MAPK pathway. PLoS ONE. 2011;6(8):e23278. DOI: 10.1371/journal.pone.0023278
  85. 85. Negreiros RS et al. Inorganic polyphosphate interacts with nucleolar and glycosomal proteins in trypanosomatids. Molecular Microbiology. 2018;110(6):973-994. DOI: 10.1111/mmi.14131
  86. 86. Azevedo C et al. Screening a protein Array with synthetic biotinylated inorganic polyphosphate to define the human PolyP-ome. ACS Chemical Biology. 2018;13(8):1958-1963. DOI: 10.1021/acschembio.8b00357
  87. 87. Bentley-DeSousa A et al. A screen for candidate targets of lysine polyphosphorylation uncovers a conserved network implicated in ribosome biogenesis. Cell Reports. 2018;22(13):3427-3439. DOI: 10.1016/j.celrep.2018.02.104
  88. 88. Hernandez-Ruiz L et al. Inorganic polyphosphate and specific induction of apoptosis in human plasma cells. Haematologica. 2006;91(9):1180-1186
  89. 89. Trilisenko L, Kulakovskaya E, Kulakovskaya T. The cadmium tolerance in Saccharomyces cerevisiae depends on inorganic polyphosphate. Journal of Basic Microbiology. 2017;57(11):982-986. DOI: 10.1002/jobm.201700257
  90. 90. Andreeva N et al. Adaptation of Saccharomyces cerevisiae to toxic manganese concentration triggers changes in inorganic polyphosphates. FEMS Yeast Research. 2013;13(5):463-470. DOI: 10.1111/1567-1364.12049
  91. 91. Dinarvand P et al. Polyphosphate amplifies proinflammatory responses of nuclear proteins through interaction with receptor for advanced glycation end products and P2Y1 purinergic receptor. Blood. 2014;123(6):935-945. DOI: 10.1182/blood-2013-09-529602
  92. 92. Tsutsumi K et al. Morphogenetic study on the maturation of osteoblastic cell as induced by inorganic polyphosphate. PLoS ONE. 2014;9(2):e86834. DOI: 10.1371/journal.pone.0086834
  93. 93. Xie L, Jakob U. Inorganic polyphosphate, a multifunctional polyanionic protein scaffold. The Journal of Biological Chemistry. 2019;294(6):2180-2190. DOI: 10.1074/jbc.REV118.002808
  94. 94. Choi SH, Smith SA, Morrissey JH. Polyphosphate is a cofactor for the activation of factor XI by thrombin. Blood. 2011;118(26):6963-6970. DOI: 10.1182/blood-2011-07-368811
  95. 95. Smith SA et al. Inhibition of polyphosphate as a novel strategy for preventing thrombosis and inflammation. Blood. 2012;120(26):5103-5110. DOI: 10.1182/blood-2012-07-444935
  96. 96. Smith SA et al. Polyphosphate exerts differential effects on blood clotting, depending on polymer size. Blood. 2010;116(20):4353-4359. DOI: 10.1182/blood-2010-01-266791
  97. 97. Smith SA, Morrissey JH. Polyphosphate: A new player in the field of hemostasis. Current Opinion in Hematology. 2014;21(5):388-394. DOI: 10.1097/MOH.0000000000000069
  98. 98. Szymusiak M et al. Colloidal confinement of polyphosphate on gold nanoparticles robustly activates the contact pathway of blood coagulation. Bioconjugate Chemistry. 2016;27(1):102-109. DOI: 10.1021/acs.bioconjchem.5b00524
  99. 99. Zhu S et al. FXIa and platelet polyphosphate as therapeutic targets during human blood clotting on collagen/tissue factor surfaces under flow. Blood. 2015;126(12):1494-1502. DOI: 10.1182/blood-2015-04-641472
  100. 100. Baik SY et al. Effects of platelet lysate preparations on the proliferation of HaCaT cells. Annals of Laboratory Medicine. 2014;34(1):43-50. DOI: 10.3343/alm.2014.34.1.43
  101. 101. Carter MJ, Fylling CP, Parnell LK. Use of platelet rich plasma gel on wound healing: A systematic review and meta-analysis. Eplasty. 2011;11:e38
  102. 102. Porwal S, Chahar YS, Singh PK. A comparative study of combined dermaroller and platelet-rich plasma versus dermaroller alone in acne scars and assessment of quality of life before and after treatment. Indian Journal of Dermatology. 2018;63(5):403-408. DOI: 10.4103/ijd.IJD_118_17
  103. 103. Alser OH, Goutos I. The evidence behind the use of platelet-rich plasma (PRP) in scar management: A literature review. Scars, Burns & Healing. 2018;4:2059513118808773. DOI: 10.1177/2059513118808773
  104. 104. Garg S, Manchanda S. Platelet-rich plasma-an ‘Elixir’ for treatment of alopecia: Personal experience on 117 patients with review of literature. Stem Cell Investigation. 2017;4:64. DOI: 10.21037/sci.2017.06.07
  105. 105. Alves R, Grimalt R. Double-blind, placebo-controlled pilot study on the use of platelet-rich plasma in women with female androgenetic alopecia. Dermatologic Surgery. 2018;44(1):132-133. DOI: 10.1097/DSS.0000000000001197
  106. 106. Liou JJ et al. Effect of platelet-rich plasma on chondrogenic differentiation of adipose- and bone marrow-derived mesenchymal stem cells. Tissue Engineering. Part A. 2018;24(19-20):1432-1443. DOI: 10.1089/ten.tea.2018.0065
  107. 107. Xie X, Zhang C, Tuan RS. Biology of platelet-rich plasma and its clinical application in cartilage repair. Arthritis Research & Therapy. 2014;16(1):204. DOI: 10.1186/ar4493
  108. 108. Chiavaras MM et al. IMpact of Platelet Rich plasma OVer alternative therapies in patients with lateral epicondylitis (IMPROVE): Protocol for a multicenter randomized controlled study: A multicenter, randomized trial comparing autologous platelet-rich plasma, autologous whole blood, dry needle tendon fenestration, and physical therapy exercises alone on pain and quality of life in patients with lateral epicondylitis. Academic Radiology. 2014;21(9):1144-1155. DOI: 10.1016/j.acra.2014.05.003
  109. 109. Hussain N, Johal H, Bhandari M. An evidence-based evaluation on the use of platelet rich plasma in orthopedics: A review of the literature. SICOT J. 2017;3:57. DOI: 10.1051/sicotj/2017036
  110. 110. Bansal H et al. Intra-articular injection in the knee of adipose derived stromal cells (stromal vascular fraction) and platelet rich plasma for osteoarthritis. Journal of Translational Medicine. 2017;15(1):141. DOI: 10.1186/s12967-017-1242-4
  111. 111. Huang Y et al. Platelet-rich plasma for regeneration of neural feedback pathways around dental implants: A concise review and outlook on future possibilities. International Journal of Oral Science. 2017;9(1):1-9. DOI: 10.1038/ijos.2017.1
  112. 112. Agrawal AA. Evolution, current status and advances in application of platelet concentrate in periodontics and implantology. World Journal of Clinical Cases. 2017;5(5):159-171. DOI: 10.12998/wjcc.v5.i5.159
  113. 113. Picard F et al. The growing evidence for the use of platelet-rich plasma on diabetic chronic wounds: A review and a proposal for a new standard care. Wound Repair and Regeneration. 2015;23(5):638-643. DOI: 10.1111/wrr.12317
  114. 114. Picard F et al. Should we use platelet-rich plasma as an adjunct therapy to treat “acute wounds,” “burns,” and “laser therapies”: A review and a proposal of a quality criteria checklist for further studies. Wound Repair and Regeneration. 2015;23(2):163-170. DOI: 10.1111/wrr.12266
  115. 115. Yeung CY et al. Efficacy of lyophilised platelet-rich plasma powder on healing rate in patients with deep second degree burn injury: A prospective double-blind randomized clinical trial. Annals of Plastic Surgery. 2018;80(2S Suppl 1):S66-S69. DOI: 10.1097/SAP.0000000000001328
  116. 116. Hara T et al. Platelet-rich plasma stimulates human dermal fibroblast proliferation via a Ras-dependent extracellular signal-regulated kinase 1/2 pathway. Journal of Artificial Organs. 2016;19(4):372-377. DOI: 10.1007/s10047-016-0913-x
  117. 117. Kakudo N et al. Platelet-rich plasma promotes epithelialization and angiogenesis in a splitthickness skin graft donor site. Medical Molecular Morphology. 2011;44(4):233-236. DOI: 10.1007/s00795-010-0532-1
  118. 118. Venter NG et al. Use of platelet-rich plasma in deep second- and third-degree burns. Burns. 2016;42(4):807-814. DOI: 10.1016/j.burns.2016.01.002
  119. 119. Alves R, Grimalt R. A review of platelet-rich plasma: History, biology, mechanism of action, and classification. Skin Appendage Disorders. 2018;4(1):18-24. DOI: 10.1159/000477353
  120. 120. Arnoczky SP, Sheibani-Rad S. The basic science of platelet-rich plasma (PRP): What clinicians need to know. Sports Medicine and Arthroscopy Review. 2013;21(4):180-185. DOI: 10.1097/JSA.0b013e3182999712
  121. 121. Martinengo L et al. Prevalence of chronic wounds in the general population: Systematic review and meta-analysis of observational studies. Annals of Epidemiology. 2019;29:8-15. DOI: 10.1016/j.annepidem.2018.10.005
  122. 122. Brick N. Autologous platelet-rich plasma for treating chronic wounds. The American Journal of Nursing. 2013;113(8):54. DOI: 10.1097/01.NAJ.0000432965.18634.85
  123. 123. Ahmed M et al. Platelet-rich plasma for the treatment of clean diabetic foot ulcers. Annals of Vascular Surgery. 2017;38:206-211. DOI: 10.1016/j.avsg.2016.04.023
  124. 124. Guo SC et al. Exosomes derived from platelet-rich plasma promote the re-epithelization of chronic cutaneous wounds via activation of YAP in a diabetic rat model. Theranostics. 2017;7(1):81-96. DOI: 10.7150/thno.16803
  125. 125. Ding Y et al. Platelet-rich fibrin accelerates skin wound healing in diabetic mice. Annals of Plastic Surgery. 2017;79(3):e15-e19. DOI: 10.1097/SAP.0000000000001091
  126. 126. Tambella AM et al. Platelet-rich plasma to treat experimentally-induced skin wounds in animals: A systematic review and meta-analysis. PLoS ONE. 2018;13(1):e0191093. DOI: 10.1371/journal.pone.0191093
  127. 127. Law JX et al. Platelet-rich plasma with keratinocytes and fibroblasts enhance healing of full-thickness wounds. Journal of Tissue Viability. 2017;26(3):208-215. DOI: 10.1016/j.jtv.2017.05.003
  128. 128. Long DW et al. Controlled delivery of platelet-derived proteins enhances porcine wound healing. Journal of Controlled Release. 2017;253:73-81. DOI: 10.1016/j.jconrel.2017.03.021
  129. 129. Fernandez-Moure JS et al. Platelet-rich plasma: A biomimetic approach to enhancement of surgical wound healing. The Journal of Surgical Research. 2017;207:33-44. DOI: 10.1016/j.jss.2016.08.063
  130. 130. Devereaux J et al. Effects of platelet-rich plasma and platelet-poor plasma on human dermal fibroblasts. Maturitas. 2018;117:34-44. DOI: 10.1016/j.maturitas.2018.09.001
  131. 131. Maciel FB et al. Scanning electron microscopy and microbiological evaluation of equine burn wound repair after platelet-rich plasma gel treatment. Burns. 2012;38(7):1058-1065. DOI: 10.1016/j.burns.2012.02.029
  132. 132. Huang SH et al. Platelet-rich plasma injection in burn scar areas alleviates neuropathic scar pain. International Journal of Medical Sciences. 2018;15(3):238-247. DOI: 10.7150/ijms.22563
  133. 133. Klosova H et al. Objective evaluation of the effect of autologous platelet concentrate on post-operative scarring in deep burns. Burns. 2013;39(6):1263-1276. DOI: 10.1016/j.burns.2013.01.020
  134. 134. Ozcelik U et al. Effect of topical platelet-rich plasma on burn healing after partial-thickness burn injury. Medical Science Monitor. 2016;22:1903-1909
  135. 135. Prochazka V et al. Addition of platelet concentrate to dermo-epidermal skin graft in deep burn trauma reduces scarring and need for revision surgeries. Biomedical Papers of the Medical Faculty of the University Palacky, Olomouc, Czech Republic. 2014;158(2):242-258. DOI: 10.5507/bp.2013.070
  136. 136. Singer AJ et al. The effects of platelet rich plasma on healing of full thickness burns in swine. Burns. 2018;44(6):1543-1550. DOI: 10.1016/j.burns.2018.04.021
  137. 137. Marck RE, Middelkoop E, Breederveld RS. Considerations on the use of platelet-rich plasma, specifically for burn treatment. Journal of Burn Care & Research. 2014;35(3):219-227. DOI: 10.1097/BCR.0b013e31829b334e
  138. 138. Marck RE et al. The application of platelet-rich plasma in the treatment of deep dermal burns: A randomized, double-blind, intra-patient controlled study. Wound Repair and Regeneration. 2016;24(4):712-720. DOI: 10.1111/wrr.12443
  139. 139. Pallua N, Wolter T, Markowicz M. Platelet-rich plasma in burns. Burns. 2010;36(1):4-8. DOI: 10.1016/j.burns.2009.05.002
  140. 140. Tejiram S et al. In-depth analysis of clotting dynamics in burn patients. The Journal of Surgical Research. 2016;202(2):341-351. DOI: 10.1016/j.jss.2016.01.006
  141. 141. Shupp JW et al. Analysis of factor XIa, factor IXa and tissue factor activity in burn patients. Burns. 2018;44(2):436-444. DOI: 10.1016/j.burns.2017.08.003
  142. 142. Glas GJ, Levi M, Schultz MJ. Coagulopathy and its management in patients with severe burns. Journal of Thrombosis and Haemostasis. 2016;14(5):865-874. DOI: 10.1111/jth.13283
  143. 143. Luker JN et al. Shedding of the endothelial glycocalyx is quantitatively proportional to burn injury severity. Annals of Burns and Fire Disasters. 2018;31(1):17-22
  144. 144. Vigiola Cruz M et al. Plasma ameliorates endothelial dysfunction in burn injury. The Journal of Surgical Research. 2019;233:459-466. DOI: 10.1016/j.jss.2018.08.027
  145. 145. Messora MR, Nagata MJH, Furlaneto FAC, Dornelles RCM, Bomfim SRM, Deliberador TM, et al. A standardized research protocol for plateletrich plasma (PRP) preparation in rats. Revista Brasileira De Saude Ocupacional. 2011;8(3):299-304
  146. 146. Galiano RD et al. Quantitative and reproducible murine model of excisional wound healing. Wound Repair and Regeneration. 2004;12(4):485-492. DOI: 10.1111/j.1067-1927.2004.12404.x
  147. 147. Wang X et al. The mouse excisional wound splinting model, including applications for stem cell transplantation. Nature Protocols. 2013;8(2):302-309. DOI: 10.1038/nprot.2013.002
  148. 148. Barrios M et al. Comparative hemostatic parameters in BALB/c, C57BL/6 and C3H/He mice. Thrombosis Research. 2009;124(3):338-343. DOI: 10.1016/j.thromres.2008.11.001
  149. 149. Giacomini A et al. Platelet count and parameters determined by the Bayer ADVIA 120 in reference subjects and patients. Clinical and Laboratory Haematology. 2001;23(3):181-186
  150. 150. Morrissey JH, Choi SH, Smith SA. Polyphosphate: An ancient molecule that links platelets, coagulation, and inflammation. Blood. 2012;119(25):5972-5979. DOI: 10.1182/blood-2012-03-306605

Written By

Cynthia M. Simbulan-Rosenthal, Bonnie C. Carney, Anirudh Gaur, Manish Moghe, Elliott Crooke, Lauren T. Moffatt, Jeffrey W. Shupp and Dean S. Rosenthal

Submitted: 04 March 2019 Reviewed: 31 May 2019 Published: 28 June 2019