Open access peer-reviewed chapter

Matrix Metalloproteinases (MMPs) and Diabetic Foot: Pathophysiological Findings and Recent Developments in Their Inhibitors of Natural as well as Synthetic Origin

Written By

Kirandeep Kaur, Atamjit Singh, Shivani Attri, Danish Malhotra, Aditi Verma, Neena Bedi and Preet Mohinder Singh Bedi

Submitted: 02 February 2020 Reviewed: 22 May 2020 Published: 19 November 2020

DOI: 10.5772/intechopen.92982

From the Edited Volume

The Eye and Foot in Diabetes

Edited by Jeffery Grigsby and Fethi Derbel

Chapter metrics overview

613 Chapter Downloads

View Full Metrics

Abstract

Management of diabetic foot remains a major challenge for healthcare system. Though wound healing is a multiphase process and involved multiple biomarkers that acts in stepwise manner, pathophysiology diabetic foot ulcers is still not much clear and need standardization. Matrix metalloproteinases (MMPs) are often linked with non-healing characteristic of diabetic foot ulcers. They play vital roles in various phases of healing process. Major functions are removal of damaged extracellular matrix in inflammatory phase, breakdown of capillary basement membrane prior to angiogenesis and facilitation in fibroblast migration during proliferation phase. For efficient healing, these enzymes are needed in certain amount only. Imbalance of these enzymes leads to excessive degradation which has been linked with the non-healing nature of diabetic ulcers. This chapter will shed light on the role of MMP’s in various phases of wound healing and the inhibitors of MMP’s from natural as well as synthetic origin. It would help researchers and physicians to the understand nature of diabetic foot more clearly and design of strategies for diabetic foot management.

Keywords

  • diabetic foot
  • matrix Metalloproteinases
  • MMP inhibitors
  • wound healing
  • inflammation

1. Introduction

Wound healing is a complex mechanism involves cascade of inter-related events, i.e., hemostasis, inflammation, proliferation and remodeling [1]. Various skin cells including epidermal, dermal, immune and endothelial cells are involved in initiating remodeling process. In wounded are, various signaling pathways and cellular mechanisms are observed to be active at same time which are responsible for ongoing the healing process. Moreover, various cellular events such as blood clotting, fibroplasia, re-epithelization and matrix deposition along with neovascularization are also involved in the process [1, 2, 3, 4, 5]. Skin is the largest organ of human body and responsible to control thermoregulation, fluid imbalance and protection of other internal organs against microbes [6]. In wounds, this barrier gets disrupted and become prone to the microbial infections. The bacterial burden invades the layers beneath epidermis and also the deeper tissues associated with extracellular matrix (ECM) which worsens the wound state [7]. The matrix metalloproteinases (MMPs) are the zinc dependent proteolytic enzymes which were firstly discovered in the tadpoles having function of collagen degradation [8]. Total 24 MMPs have been reported so far having different substrate specificities and functions [9, 10]. The MMPs have been reported to be involved in cellular interactions, cell- matrix interactions by altering the levels of cytokines, growth factors and various biological fragments hidden in ECM [11, 12, 13, 14, 15]. MMPs indirectly modulates the cellular behavior by altering the cell surface receptors, junctional proteins and various cellular processes such as cell death and inflammation [16, 17]. MMPs play important role during microbial infection of wounds, the disrupted fragments of ECM possess antimicrobial activity which makes the MMPs to be the major component involved in healing process of wounds [18]. However, the bacteria itself are able to produce proteolytic enzymes which leads to the accumulation of degraded matrix components [19, 20, 21]. At the same time in some cases MMPs have been proven to be suitable candidate for gearing up the wound healing process [22, 23] but on other hands, several investigations reported the deregulation of these enzymes to be responsible for worsening the healing process and conversion of acute wounds to chronic wounds. This book chapter will focus on the various implications of MMPs in the chronic wounds along with their inhibitors of natural as well as synthetic origin.

Advertisement

2. Chronic wounds and infections

The disruption of skin barrier leads to increases susceptibility of bacterial strains to invade the wounds. The interaction of various bacteria/microbes has specificity with different matrix components turns to bacterial colonization. The bacterial colonization increases the bacterial burden in damaged wound site [24, 25]. This microbial colonization is the onset to the journey of an acute wound towards chronic wound [26, 27]. The minute to higher quantities of bacterial population is found in each and every acute wound known as contamination [28]. The quantity and the severity of these bacterial strains vary from wound to wound. If these bacterial population contains some pathogenic strains then there is a high risk of contamination turning into infection [29, 30]. Bacteria have ability to form biofilm with the help of self-secreting extracellular polymeric substances [31]. Biofilms involves the different layers of bacteria stick with each other to form thick films. These biofilms hinder the proper functioning of immune system of host [32]. The biofilms make the bacteria hard to evade from bacterial bed and delays the healing process [33]. The most prevalent bacteria found in the chronic wounds are Staphylococcus aureus, Pseudomonas aeruginosa, Proteus mirabilis and Escherichia coli which prolongs the healing process [30, 34].

Advertisement

3. Chronic wounds and MMPs

The elevated level of proteolytic activity of MMPs is considered as the major factor responsible for impaired wound healing [35, 36]. The MMPs have capability to degrade ECM, non-ECM components, trans-membrane proteins, cell surface receptors and diminishes the function of cytokines and growth factors by decreasing their level [37, 38, 39, 40, 41, 42, 43]. The tissue inhibitors of MMPs (TIMPs) are found to be decreased in chronic wounds that make the situation more worsen [44, 45, 46, 47].

From the exudates of chronic wounds, it has been found that the proteolytic activity is surprisingly 116-fold higher than acute wounds. It marks the presence of MMPs in high levels [48, 49]. The plethora varies from wound to wound due to the specificity of different type of bacteria with ECM structural integrity [50, 51]. The Staphylococcus aureus has been found to increase the level of MMP-1 and -9 whereas Pseudomonas aeruginosa is known for raising the level of elastases [52, 53]. The bacterial pathogens not only target the host immune cells such as macrophages and neutrophils, but also attract the ECM matrix to release proteolytic enzymes which further mediates the release of MMPs. As in case of Pseudomonas aeruginosa the proteolytic enzyme thermolysin protease activates MMP-1, -8 and -9 [53]. Lipopolysaccharide derived serine proteinases activates pro-MMP-9 [54]. Different MMPs are upregulated specifically by various bacterial strains such as Corynebacterium striatum specifically gave rise to the level of MMP-2 and -9 and so as the other strains are specific for the modulation of MMPs [55, 56, 57].

Advertisement

4. MMPs and wounds

MMP family consists of 28 members, out of which 26 are expressed in humans and their homologs are found in birds, plants as well as algae also [58]. MMPs can be divided on the basis of similarity of protein fold- known an ‘Clans’ and on the basis of evolutionary relationships- called as ‘Families’. The MMP class consists of 8 clans and almost 40 families. There are basically, two ways of classifying the MMPs, which can be described as following:

  1. In accordance to the organization of the substrate specificity and homology:

    1. Collagenases (MMP-1, MMP-8, MMP-2)

    2. Gelatinases (MMP-2 and MMP-9)

    3. Stromelysins (MMP-3, MMP-10, MMP-12)

    4. Matrilysins (MMP-7, MMP-26)

    5. Membrane Type (MT) MMPs (MT-MMP-14, -15, -16, -17, -24, -25)

    6. Other MMPs (MMP-19, -20, -21, -22, -23, -27, -28)

  2. In accordance to the structure of the MMPs:

    1. Archetypal MMP (type-1 collagenases)

    2. Martilysins: lacks the hemopexin domain

    3. Gelatinases: Comprised of three type II fibronectin domains

    4. MT-MMPs: Localized at the surface of cell membrane

Table 1 represents the classification of the MMPs based upon their substrate and targets. This classification provides wide range of information including their distribution in human body [59, 60].

S. no.TypeClassSubstrates and targetsDistribution
1.MMP-1CollagenasesCollagen (I, II, III, VII, VIII, X), gelatin, aggrecan, nidogen, perlecan, proteoglycan link protein, serpins, tenascin C, Versican, casein, α1-antichymotrypsin, α1-antitrypsin, α1-proteinase inhibitor, IGF-BP-3 and -5, IL-1β, L-selectin, ovostatin, PAR-1, pro-TNF-α and SDF-1.Endothelium, SMCs, fibroblasts, platelets, macrophages and varicose veins (interstitial/fibroblast collagenase).
2.MMP-2GelatinasesCollagen (I, II, III, IV, V, VII, X, XI), gelatin, aggrecan, elastin, fibronectin, laminin, nidogen, proteoglycan link protein, versican, active MMP-9 and MMP-13, FGF-R1, IGFBP-3 and -5, IL-1β, pro-TNF-α and TGF-β.Endothelium, VSM, Adventitia, platelets, leukocytes, aortic aneurysm and varicose veins.
3.MMP-3StromelysinsCollagen (II, III, IV, IX, X, XI), gelatin, aggrecan, decorin, elastin, fibronectin, laminin, nidogen, perlecan, proteoglycan, proteoglycan link protein, versican, casein, α1-antichymotrypsin, α1-proteinase inhibitor, antithrombin III, E-cadherin, fibrinogen, IGF-BP-3, L-selectin, ovostatin, pro-HB-EGF, pro-IL-1β, proMMP-1, -8, and -9, pro-TNF-α and SDF-1Endothelium, intima, VSM, platelets, coronary artery disease, hypertension, varicose veins, synovial fibroblasts and tumor invasion.
4.MMP-7MatrilysinsCollagen (IV, X), gelatin, aggrecan, elastin, enactin, fibronectin, laminin, proteoglycan link protein, casein, β4 integrin, decorin, defensin, E-cadherin, Fas ligand, plasminogen, proMMP-2, -7, and -8, pro-TNF-α, syndecan and transferrin.Endothelium, intima, VSM, uterus and varicose veins (PUMP).
5.MMP-8CollagenasesCollagen (I, II, III, V, VII, VIII, X), gelatin, aggrecan, elastin, fibronectin, laminin, Nidogen, α2-Antiplasmin and proMMP-8.Macrophages and neutrophils (PMNL or neutrophil collagenase).
6.MMP-9GelatinasesCollagen (IV, V, VII, X, XIV), gelatin, aggrecan, elastin, fibronectin, laminin, nidogen, proteoglycan link protein, versican, CXCL5, IL-1β, IL2-R, plasminogen, pro-TNF-α, SDF-1 and TGF-β.Endothelium, VSM, adventitia, micro vessels, macrophages, aortic aneurysm and varicose veins.
7.MMP-10StromelysinsCollagen (III, IV, V), gelatin, aggrecan, elastin, fibronectin, laminin, nidogen, Casein, proMMP-1, -8, and -10.Atherosclerosis, uterus, preeclampsia, arthritis and carcinoma cells.
8.MMP-11StromelysinsAggrecan, fibronectin, laminin, α1-Antitrypsin, α1-proteinase inhibitor and IGF-BP-1.Brain, uterus and angiogenesis.
9.MMP-12Other enzymesCollagen IV, gelatin, elastin, fibronectin, laminin, casein and plasminogen.SMCs, fibroblasts, macrophages and great saphenous vein.
10.MMP-13CollagenasesCollagen (I, II, III, IV), gelatin, aggrecan, fibronectin, laminin, perlecan, tenascin, casein, PAR-1, plasminogen activator 2, proMMP-9 and-13, and SDF-1.SMCs, macrophages, varicose veins, pre-eclampsia and breast cancer.
11.MMP-14MT-MMPCollagen (I, II, III), gelatin, aggrecan, elastin, fibrin, fibronectin, laminin, nidogen, perlecan, proteoglycan, tenascin, vitronectin, αvβ3 integrin, CD44, proMMP-2 and -13, pro-TNF-α, SDF-1, α1-proteinase inhibitor and tissue transglutaminase.VSM, fibroblasts, platelets, brain, uterus and angiogenesis.
12.MMP-15MT-MMPCollagen I, gelatin, aggrecan, fibronectin, laminin, nidogen, perlecan, tenascin, vitronectin, proMMP-2 and -13, and tissue transglutaminaseFibroblasts, leukocytes and pre-eclampsia
13.MMP-16MT-MMPCollagen I, Aggrecan, fibronectin, laminin, perlecan, vitronectin, Casein, proMMP-2 and -13Leukocytes and angiogenesis.
14.MMP-17MT-MMPGelatin and fibrinBrain and breast cancer.
15.MMP-18CollagenasesCollagen (I, II, III), gelatin and α1-AntitrypsinXenopus (amphibian, Xenopus collagenase), heart, lung and colon.
16.MMP-19Other enzymesCollagen (I, IV), gelatin, aggrecan, fibronectin, laminin, nidogen, tenascin and casein.Liver
17.MMP-20Other enzymesCollagen (V), aggrecan, cartilage oligomeric protein and amelogeninTooth enamel
18.MMP-21Other enzymesα1-AntitrypsinFibroblasts, macrophages and placenta
19.MMP-22Other enzymesGelatinChicken fibroblasts.
20.MMP-23Other enzymesGelatinOvary, testis, prostate and Other (type II) MT-MMP.
21.MMP-24MT-MMPGelatin, Chondroitin sulphate, dermatinsulfate, fibrin, fibronectin, N-cadherin and proMMP-2 and -13Leukocytes, lung, pancreas, kidney, brain, astrocytoma and glioblastoma.
22.MMP-25MT-MMPCollagen IV, gelatin, fibrin, fibronectin, proMMP-2 and α1-proteinase inhibitorLeukocytes (leukolysin), anaplastic astrocytomas and glioblastomas.
23.MMP-26MatrilysinsCollagen IV, gelatin, fibrinogen, fibronectin, vitronectin, casein, β1-proteinase inhibitor, fibrin, fibronectin and proMMP-2.Breast cancer and endometrial tumors.
24.MMP-27Other enzymesHeart, leukocytes, macrophages, kidney, endometrium, menstruation, bone, osteoarthritis and breast cancer.
25.MMP-28Other enzymesCaseinSkin and keratinocytes.

Table 1.

Distribution of MMPs in human body with their substrates and targets.

4.1 Collagenases

Collagenases are the enzymes known for their cleavage action on the bunch of extracellular components, i.e., Collagen, Aggrecan, Versican, Perlecan, etc., which are responsible for ECM accumulation. Collagenases activity has been found to be higher in chronic wounds with positively alleviated levels of MMP-1 and MMP-8 whereas the TIMP get downregulated. MMP-1 is known as collagenase-1. After the tissue rupturing, the integral proteins when coordinated with the keratinocytes alleviate the level of MMP-1. Furthermore, the MMP-1 degrades the ECM components and increases the turnover of proliferating cells at the other end of keratinocytes [61]. In the proliferation phase of wound healing the MMP-1 level is found to be high whereas the TIMPs are lower in the initial phases. On reaching the final phase of wound repair, i.e., the remodeling/re-epithelization the situation gets vice n versa. Some laminin isoforms of keratinocytes also regulate the MMPs level in various phases of wound repair [62]. In recent past several investigations report the dysregulation of MMP-1 in chronic wounds, i.e., even high level of MMP-1 is found in remodeling phase leads to damaged diabetic foot [63, 64]. The MMP/TIMP ratio is a crucial factor for repairing wounds [65]. The dermal ulcers also known as lipodermosclerosis are enriched with MMP-1 and MMP-2 associated with downregulation of TIMP-2 [66, 67]. Some immune cells stimulate the production of MMPs, i.e., collagenases and gelatinases [57, 68]. Among which the neutrophils derived MMP-8 (Collagenase-2) has been found to play an important role in pathophysiology of wounds. The upregulation of MMP-8 is majorly responsible for non-healing of wounds, i.e., for the state of chronic wounds [40, 69, 70]. On the contrary, MMP-8 has stronger affinity towards collagen-1 hence provide tensile strength to the wound tissues in the re-epithelization phase. Even in some reports MMP-8 is found to act as pro-enzyme in wound repair [71, 72]. Stromal cells derived MMP-13, collagenase-3 is reported to be highly expressed in wound site where as absence in the epidermis indicates its pivotal role in the formation of granulation tissue and extracellular matrix [63].

4.2 Gelatinases

Gelatinases, i.e., gelatinase A (MMP-2) and gelatinase B (MMP-9) have the broader specificity towards the substrates therefore leads to enhanced depletion of ECM components and retard the process of angiogenesis [6, 73]. The alleviated level of these MMPs has been found in the exudates of chronic wounds [46]. However, they possess broad specificity but an excellent substrate specificity exists between both the gelatinases. MMP-9 erodes the pro healing and other growth factors that leads to delayed in healing process however positively influence the inflammatory phase. The upregulation of MMP-9 degrades the specific biomarkers of wound healing, i.e., the vascular endothelial growth factor and dermatopontin and makes them non-functional. However MMP-2 stimulates the deterioration of laminin 332, enhance the keratinocytes migration and promotes the healing process [74, 75, 76]. The inflammatory cytokines (Interleukins; IL1-α, IL1-β, IL-2, IL-17, C reactive protein, Insulin like Growth Factors-1, Transforming Growth Factor-α) stimulates the release of protein called Neutrophil gelatinase associated lipocalin (NGAL). This NGAL activates the MMP-9 and makes the NGAL-MMP-9 complex which is considered as the underlying cause of slow healing in diabetic wounds. Diabetic wounds have been reported to be enriched with MMP-9, MMP-9-NGAL complex, NGAL and neutrophil. However, the situation gets opposite when given the insulin treatment [77, 78, 79, 80].

4.3 Stromelysins and other MMPs

Stromelysin-1 and -2, i.e., MMP-3 and -10, respectively. MMP-3 along with collagenase-1 is found in distal end and stimulates the keratinocytes proliferation whereas MMP-10 is present in the starting edge of keratinocytes [60, 81]. MMP-3 regulates the migration of fibroblasts to the wound site resulting in wound contraction. On other hand, MMP-10 is responsible for the keratinocyte cell death and slow down the healing process. MMP-3 is a major activator of MMP-9 hence also contributing to inflammatory phase [82]. Other MMPs such as MMP-12, -7 and -14 are activated by stromal macrophages. MMP-7 interact with cyndecans and integrins to promote the skin regeneration in remodeling phase [83]. MMP-14 is majorly present in the fibroblasts on the wound bed. The level of MMP-12 gets naturally increased during inflammatory phase. These MMPs not only contribute in the cellular signaling pathways but also triggers the stimulation of other MMPs [22, 84, 85, 86].

Advertisement

5. MMPs in wound healing

5.1 Hemostasis

Followed by the tissue injury, the blood clotting and platelet aggregation is the former step in wound healing. The extrinsic and intrinsic system regulates the accumulation of platelets at wound site by means of coagulation factors and thrombocytes respectively [87]. The cytokines and other associated growth factors trigger the constriction of vessels which fills the voids in the wound area and lead to clot formation. The former step is followed by the vasodilation where the thrombocytes and fibroblasts like fibronectin, vitronectin and thrombospondin leads to form the provisional scaffold like wound matrix which allows the migration of keratinocytes, endothelial cells and leukocytes [88]. These platelets and leukocytes stimulate cytokines and growth factors which further assists the inflammatory process. The interleukins IL-1α, β, IL-6 and TNF-α are engaged in this process. Furthermore, the collagen synthesis is mediated by FGF-b, IGF, TGF-β and angiogenesis which get activated by FGF-B, VEGF subunit A, TGF-β and HIF-1 [89, 90]. Hemostasis is the initial phase in wound healing process and MMPs does not have any significant interference in this phase.

5.2 Proliferation and re-epithelization

The proliferation phase includes the granulation tissue to cover wound area by the strong network of vessels. Platelets are shifted to the injury site, to form the clot. Besides this, the platelets have another important function to stimulate the movement of neutrophils and macrophages to the wound site triggered by the release of platelets derived growth factors [91]. This factor is also engaged in mediating the collagenases the fibroblastic cells especially MMP-8 which have major role in tissue damage. MMP-8 are also released by neutrophils during the wound infection and assists the wound debridement and rearrangement of damaged collagen-I [92]. The synthesis of another MMPs such as MMP-1,2,3,9 are also driven by the platelets. MMP-1 and 2 has important function to control the adhesion of platelets and conglomeration [44, 88]. Moreover, MMP-9 filters the different collagen types and regulates the release of inflammatory cytokines such as IGN-γ and TGF-β. The collagen and fibroblast synthesis which in turn form the collective tissue network is also regulated by MMP-9. The new capillary formation at the wound site is also associated with movement of fibroblasts within the fibrin network which promotes angiogenesis and leads to neovascularization and re-epithelization [88]. The process of re-epithelization is also get started by the signaling pathways regulated by the endothelial and non-endothelial cells which involve various cytokines such as EGF, KGF, IGF-1 and NGF [90]. The basic component of the endothelial cells known as laminin exists in various isoforms. Among which the laminin isoform-5 have pivotal role in induction of keratinocyte migration and MMP-9 activation. Cell movement is a major role of MMP-9 hence plays an important role in re-epithelization process [93]. Another MMPs such as MMP-14 and MMP-2 breaks laminin isoform 5 and release a factor which when interacts with the epidermal growth factor (EGF) turns up the movement of cells [94, 95]. One more factor FGF-2 released by macrophages when interacts with the heparin sulphate enhances the growth of endothelial and fibroblast cells. The vascular endothelial growth factor (VEGF) released by macrophages activates the cell migration and proliferation of keratinocytes and endothelial cells which include MMP-1, 2, 9 and 13 hence play a major role in wound healing [96, 97].

5.3 Matrix formation and remodeling

The final stage of wound healing is remodeling phase. It involves the upregulation of collagen turnover but decline in proliferation of fibroblast [98]. Moreover, the keratinocytes reach fibrin clot by crossing granulation tissue matrix [99]. The collagen-I replacement with collagen type III indicates the maturation of wound [100, 101]. However, in the early phase of remodeling phase fibronectin and fibroblasts are get displaced by collagen type I and III and proteoglycans which in turn enhances the tensile strength and integrity of wound matrix [102]. The level of myofibroblast and blood vessels get increased while reaching the end of this phase and high density of these two leads to the closure of wound [63, 103].

5.4 Proteolysis in wound repair

Many processes in wound healing such as keratinocyte migration, angiogenesis and re-epithelization are generally followed by the extra cellular matrix (ECM) degradation [104]. The MMPs are majorly involved in this proteolytic degradation. MMP-19 and 28 are present in keratinocytes of basal stratum and superbasals [105]. Moreover, MMP-19 is also found in the hair follicles, endothelial cells, arteries and veins [106]. MMP-1 expression is found to be upregulated in dermis part of the wounds where basal membrane is destroyed and promotes re-epithelization process and triggers the binding of keratinocytes with type-1 collagen [65]. Collagen type I is known to upregulate the level of MMP-1 whereas collagen type III and other basement proteins do not promote the MMP-1 synthesis. MMP-1 activates α1β2 integrin to synthesize collagen type-1 [16]. The MMP1- α1β2 complex enhances the migration of keratinocytes therefore boost up the re-epithelization process [107]. During the process of basement membrane formation followed by re-epithelization, MMP-1 expression gets knockdown by the cellular junctions of basal membrane proteins [16, 108]. Moreover, MMP-13 which is mainly present in the dermis along with MMP-1 regulates the fibroblast proliferation mediated by matrix shrinkage and matrix stiffness [109, 110]. MMP-8 stored in cellular granules are secreted when get activated by macrophages [111]. The overexpression of MMP-8 is found in the damaged wounds. MMP-13 downregulation is balanced by MMP-8 which slow down the healing process by improper infiltration of neutrophils, improper re-epithelization and constant inflammatory syndrome [111, 112]. As given in the classification section the stromelysins such as MMP-3 and MMP-10 are present in the epidermal cells i.e. proliferating keratinocytes. These MMPs especially MMP-3 has major role in disruption of fibrin containing provisional matrix and formation of new basal matrix after remodeling [113]. This process is majorly carried out by cytokines and other growth factors such as FGF-b and HB-EGF [114]. Furthermore, the MMP-9 has also an important role in final phase that shaping the epidermal layer at the end during wound repair. In addition, MMP-2, -9, -19 and MT1-MMP are stored and released by the endothelial cells [115]. Among which MMP-2 and -9 have pivotal role in degradation of mature blood vessels and sprouting/growth of new blood vessels by activating angiogenesis related growth factors and cytokines [86, 89, 116, 117]. MT1-MMP possess proteolytic activity against mature collagen and fibrin by crossing the thick network of fibrin proteins in stroma of damaged tissues [118]. Whereas, MMP-19 is involved in growth process of endothelial cells, epithelial cells, fibroblast cells and small vasculature within macrophages [119].

The wounds that persist more than 4–6 weeks are generally recognized as chronic wounds [120]. Wounds such as venous leg ulcers [72, 121], diabetic foot ulcers [122, 123] and that caused due to pressure [66] are considered as chronic or delayed wounds. Some wounds which appears to be acute at initial stages but may turns to chronic one while reaching the final phase of healing are also categorized under chronic wounds. Main examples of these types of wounds are surgical wounds and traumatic wounds. These chronic wounds are specifically characterized by the altered levels on MMPs.

Abnormal structural integrity of fibrin network, increased tendon rigidity and altered volume and level of biochemical substances indicates the delayed and chronic wounds [124]. Proteolytic activity of MMPs has major impact on healing process of chronic wounds. Besides this MMP-3 and MMP-13 along with MMP-9 are actively found in the normal as well as diabetic foot ulcers. Where MMP-3 and MMP-9 have been upregulated, MMP-3 has been found to be knockdown in chronic wounds. The overexpression of MMP-13 and MMP-9 is associated with high glucose concentration at wound site [125]. The imbalance between MMP and TIMP level is a major cause of hyperglycemia, hyperlipidemia and hypertension during the condition of chronic wounds/diabetic ulcers [126, 127]. In the state of chronic wounds, the migration of inflammatory cells is followed by imbalanced fibroblast clotting which lead to secrete the ECM proteins. Meanwhile, MMPs have been found to increase the fibroblast proliferation and collagen degradation via TGF-β1 signaling [127, 128]. Higher production of gelatinases has been observed in the diabetic wounds. Conclusively MMPs in this state are associated with degradation of ECM components but at the same time are also responsible for the recovery of traumatic wounds by regenerating the capillary and blood vessels at the respective site [129].

Advertisement

6. Levels of MMPs in diabetic wounds

In the state of diabetic wounds, the glucose level is significantly higher [130]. Elevated levels of MMPs have been found in these wounds because of oxidative stress and end products of glycation which may lead to diabetic peripheral arterial disease [61, 131]. Degradation of ECM due to MMPs especially MMP-1, -2 and -9 turn these diabetic wounds to get more worse [132]. The mismatch between the extent of degradation and repairing of ECM is a critical factor to cause delay in wound healing process i.e. chronic wounds. Therefore, it necessitates the ECM components to be in controlled condition for boosting up the healing process [131]. Any other disease condition in diabetic ulcer may worsen the healing process due to imbalanced availability of cytokines and other growth factors needed for the healing of wounds [133, 134]. In each phase of diabetic wound repair, i.e., hemostasis, inflammation, proliferation and remodeling there has been altered expression of MMPs [135]. Epithelial remodeling is associated with raised levels of MMP-1, -8, -9 and downregulation of TIMPs. The fibronectin degradation is majorly carried out by MMP-9 which leads to cell migration and proliferation [136]. MMP-1, -8 and -9 have been reported to be upregulated in the venous wounds due to absence of TIMP [73, 81]. Patients with metabolic syndrome have been found to be overly expressed with MMP-2 and -9 in their serum sample. The mutations in the gene expression of MMP-9 can also be a cause for delayed healing. Increased expression of MMP-9, TNF-α and other growth factors in diabetic foot ulcers has been found and concluded that they could be linked with slow-to-heal ulcers in diabetics and therefore a target for new therapeutic management [137].

Advertisement

7. Therapeutical targeting of MMPs

7.1 Synthetic approaches

MMPs has pivotal role in diabetic wounds hence they are the major target for the researchers. MMPs possess high resemblance in their structural morphology therefore it is difficult to target specifically one MMP at the time especially when they fall under same category such as gelatinase-A and gelatinase-B [138, 139]. Moreover, MMP-8 has been found to boost up healing in diabetic wounds in absence of MMP-9 and vice versa which necessitates the specific inhibition of MMP-9 without having any interaction with MMP-8. There are many broad spectrum MMP inhibitors (MMPI) which have been already investigated for the purpose but we need more selective therapeutics over the existing one [140]. Many small structural molecules have been discovered yet to target the same. In an investigation, it has been reported that the racemic mixture, i.e., (R, S)-ND-336 possess 55-fold more activity than the R or S isomer alone to target MMP-9 specifically than MMP-8 [74]. Moreover, based upon the Ki values the R isomer has been reported to be 10-fold more potent than the S isomer for selective inhibition of MMP-9 [141]. As it has been known that the synthetic molecule (R, S)-ND-336 falls under thiirane class, its structural ring gets unlatched and produce thiolate which gets interacted with the zinc ion within MMP-9 and inhibit its function. Reversal of the given process is very slow therefore it shows long lasting retention time. R-ND-336 has been investigated to be more effective than FDA approved drug becaplermin for the respective purpose [141]. Enhanced specificity for inhibition of MMPs can be obtained by using antibody approach. In the recent past, GS-5745 is an antibody being investigated for specific inhibition of MMP-9. It has dual mechanism to act on i.e. by interacting and hindering the active site of MMP-9 and another one is to cleave the MMP-3 zymogen which is involved in activation of MMP-9 [142, 143, 144, 145]. Moreover, another two antibodies being investigated under the clinical trials are SSDS-3 and REGA-3G12 have been observed to possess selective inhibition against MMP-9 [146, 147]. Furthermore, the above antibodies have been more explored for the cancer targets hence there are many future possibilities to explore the wound healing potential of the above candidates [148]. Wound dressings are being commonly used for the purpose of healing and controlling the exudate secretion [149]. Many MMP inhibitors have been used to incorporate into these wound dressings. But these inhibitors are generally found to be non-specific, i.e., the broad-spectrum inhibitors. Most commonly used MMP-inhibitors in wound healing are bisphosphonates [150]. Another hypothesis involves the use of atelocollagen type I to be used along with 4-vinyl-benzyl chloride to specifically inhibit the MMPs present in wound exudates [151]. In addition, another clinical candidate GM-6001 exploited as wound dressing has broad spectrum activity but found to be less effecting in healing diabetic wounds than the therapeutics having specific inhibitors against MMPs [152]. RNA is a basic nucleotide to synthesize gene encoding MMPs [153]. So, the therapies being approached to inhibit RNA which in turn inhibit the MMPs at gene level have the high potential to heal the diabetic wounds than other therapeutics [154]. But the major obstacle is to deliver the siRNA to the target site [155, 156, 157]. Therefore, to overcome this, the star shaped cationic polymer such as cyclodextrins have been reported to be used for the purpose as they have very low toxicity [158]. Furthermore, β-CD-(D3)7/MMP-9-siRNA has been found in an investigation to inhibit MMP-9 in the diabetic wounds where MMPs level is quite high due to TIMP knockdown which in turn promotes the healing process. These siRNAs are supposed to be taken by the fibroblasts on wound site [159, 160]. But when the siRNA was used alone in a further study it has been found to cause liver and kidney toxicity [161]. In addition, the miRNA-139 and miRNA-335 has been reported to possess an excellent potential in healing the diabetic wounds via inhibiting MMP-9 [162].

7.2 MMPs inhibitors of natural origin

Natural products have huge resource of biologically active molecules and provided large number of biologically active compounds to clinical practice for treatment of wide range of diseases and disorders. Considering capability of natural products in drug development, wide range of researchers across the globe has screened numerous constituents from natural sources for MMPs modulation activity. Major bioactive constituents (Figure 1) from natural sources with MMP modulation potential has been discussed below.

Figure 1.

Various MMP inhibitors of natural origin.

Withaferin A (3-azido Withaferin A) a naturally derived steroidal lactone from plant Withania somnifera, exhibits various pharmacological activities. In vitro studies revealed that withaferin A inhibit MMP-2 by up regulating the expression of pro-apoptotic protein par-4. Stimulation of par-4 by withaferin A enhances apoptosis by extrinsic pathway of apoptosis by activating FADD-caspase-8-caspase-3 in dose dependent manner [163]. Cantharidin is a natural compound obtained from Mylabris phalerata and showed remarkable inhibition of cell migration and invasiveness by suppressing MMP-2 and MMP-9 through affecting their upstream markers such as NF-ĸB, c-Jun and AP-1 in A375.S2 cells based wound healing and matrigel chamber invasion assays. Cantharidin exerts anti-cell migration and anti-invasiveness property by suppressing MMP-2 and MMP-9 via modulating their upstream markers such as NF-ĸB, c-Jun and AP-1. Cantharidin also down regulate the expression of NF-ĸB, p65 and proteins involved in PI3K-Akt (PI3K, ERK1/2, Rock1 and FAK) and MAPK (p38, ERK and JNK) signaling cascade [164]. Celastrol is another phytoconstituent of Tripterygium wilfordii having anti-proliferative activity. Celastrol inhibit metastasis and invasiveness of ovarian cancer cells (SKOV3 and OVCAR-3) by knockdown the expression of NF-κB/MMP-9. NF-κB modulates IκBα degradation, p65 translocation blocking and MMP-9 suppression [165]. Gallic acid has been proved to inhibit cell metastasis and invasiveness in PC-3 cells by degrading expressions of MMP-2 and MMP-9 otherwise they degrade the ECM and delayed wound healing. Gallic acid has been also observed to activate the TIMP-1 which is natural regulator of MMP-2 [166]. Ginsenoside Rd is obtained from Panax ginseng leaf which increases proliferation, migration and shows protective effect in human dermal fibroblast (HDFs) and keratinocyte progenitor cells which enhances healing of skin wound. Being a steroidal moiety it easily permeate to cell membrane and enhance healing of skin both in laser burn and excision wound by increasing expression of CREB, cAMP along with reducing expression of MMP-1 [167]. Lycorine a natural compound widely distributed in plant family Amaryllidaceae. Lycorine shows antimigratory effect in HepG2 cells by reducing the expression of MMP-2 and MMP-9. Lycorine increase polymerized F-actin by blocking the normal turnover of the actin cytoskeleton and a loss of depolymerized G-actin. The activation of ROCK1 in cells pre-treated with lycorine shows decrease in expression of cofilin, cyclinA, cyclin B1, cdc2, MMP-9 and MMP-2 which shows lycorine inhibiting cell proliferation and migration in HepG2 cells via inhibition of ROCK1/cofilin-induced actin dynamics [168]. Naringin is a natural flavonoid present in citrus fruits. Treatment of naringin reduce the expression of p-ERK and p-JNK which are molecular markers involved in MAPK signaling pathway in turn reduce the expression of MMP-2 and MMP-9 [169]. Pinosylvin is a natural compound present in Pinus species. Pinosylvin downregulate the expressions of MMP-1, MMP-2 and MMP-9 in human fibrosarcoma HT1080 cells. Quercetin is another well-known inhibitor of MMP-2 and MMP-9 [170]. Platycodin D from Platycodon grandiflorum exhibit anti-invasive and antimetastatic activity in human breast cancer cells (MDA-MB-231). It inhibit cell invasion by down regulating the Mrna expression of MMP-9 [171]. Formononetin is a natural compound found in Astragalus membranaceus, Trifolium pratense, Glycyrrhiza glabra and Pueraria lobata. It shows inhibitory effect on the breast cancer cells progression, migration and invasiveness by suppressing the effect of MMP-2 and MMP-9 along with upregulating the expression of matrix metalloproteinase inhibitors such as TIMP-1 and TIMP-2 [172]. Zeylenone is natural oxide with anticancer activity. In the present study, Zey is reported to have anti-cancer activity against prostate cancer. Zeylenone has been reported to suppress the expression of MMP-2, MMP-9 and upregulate the expression of TIMP-1 and collagen-1 in DU145 cells [173]. Curcumin is a natural compound extracted from plant Curcuma longa (Zingiberaceae). Curcumin possesses anti-oxidant, anti-inflammatory and anti-cancer activity and along with wound healing property. The mechanism of curcumin behind wound healing is the lowering the expression of TNF-α and increased proportion of α-SMA and collagen in fibroblast. Matrix metalloproteinase especially MMP-9 helpful in tissue migration and remodeling and someway helpful in normal wound healing. Curcumin treated cells negatively regulate the expression of MMP-9 and reduced the expression of TNF-α which positively modulate MMP-9. Curcumin also regulate the expression of NF-κB induce by TNF-α. Thus curcumin enhances wound healing activity by downregulating expression of MMP-9 and increase value of collagen by regulating expression of NF-κB induce by TNF-α in fibroblasts [174]. Shikonin is a natural component presents in Lithospermum erythrorhizon which exhibit decent wound healing property. Shikonin has been proved for its inhibitory effect on migration and invasiveness of U87 and U251cells by inhibiting the expression of MMP-2 and MMP-9 [175, 176].

Advertisement

8. Conclusion

From the ancient past wound healing has known to be a complicated topic as it involves many complex and unclear mechanisms. Moreover, wound healing process in diabetes like state get delay and more worsen. In the recent past various MMPs have been found to play a key role in the healing of diabetic foot. Structurally, it mainly has zinc on its active site. Furthermore, it is categorized in various types based upon the different substrate it cleaves, i.e., collagenases, gelatinases, stromelysins and various other MMPs. Also, the modulation of expression of various MMPs significantly alters the healing process. In addition, TGF-β has been reported to be the signaling pathway for MMPs to act upon for healing of chronic wounds. Moreover, different phases of wound repair involve alteration in level of various MMPs. Among various MMPs, MMP-9 has been widely discussed and investigated enzyme in the recent past and has also been considered as major culprit in altering the healing rate. The overexpression of various MMPs extends the time of healing or may devastate the condition. Therefore, various MMP inhibitors either of natural or synthetic origin have been explored for the purpose. Most of these candidates are under clinical trial and has proven to be very selective and effective for healing chronic wounds. Besides the wound healing MMPs possess therapeutic effectiveness for various other diseases. In future, there are various possibilities to explore and unlash various mechanisms of MMPs for chronic wound healing.

Advertisement

Acknowledgments

Authors are grateful to the University Grants Commission for providing NFOBC to Atamjit Singh. The authors are also thankful to Guru Nanak Dev University, Amritsar for providing various facilities to carry out the work.

Advertisement

Conflict of interest

The authors confirm that this chapter content has no conflicts of interest.

References

  1. 1. Diegelmann RF. From the selected works of Robert F. Diegelmann Ph. D. Frontiers in Bioscience. 2004;9:283-289
  2. 2. Shaw TJ, Martin P. Wound repair at a glance. Journal of Cell Science. 2009;122(18):3209-3213
  3. 3. Yamaguchi Y, Yoshikawa K. Cutaneous wound healing: An update. The Journal of Dermatology. 2001;28(10):521-534
  4. 4. Martin P. Wound healing--aiming for perfect skin regeneration. Science. 1997;276(5309):75-81
  5. 5. Schultz GS, Davidson JM, Kirsner RS, Bornstein P, Herman IM. Dynamic reciprocity in the wound microenvironment. Wound Repair and Regeneration. 2011;19(2):134-148
  6. 6. Cooper S. The biology of the skin. Journal of the Royal Society of Medicine. 2002;95:109
  7. 7. Yager DR, Zhang LY, Liang HX, Diegelmann RF, Cohen IK. Elevated matrix metalloproteinase levels and activity compared to surgical wound fluids. Journal of Investigative Dermatology. 1996;107(5):743-748
  8. 8. Gross J, Lapiere CM. Collagenolytic activity in amphibian tissues: A tissue culture assay. Proceedings of the National Academy of Sciences of the United States of America. 1962;48(6):1014
  9. 9. Puente XS, Sánchez LM, Overall CM, López-Otín C. Human and mouse proteases: A comparative genomic approach. Nature Reviews. Genetics. 2003;4(7):544-558
  10. 10. Fanjul-Fernández M, Folgueras AR, Cabrera S, López-Otín C. Matrix metalloproteinases: Evolution, gene regulation and functional analysis in mouse models. Biochimica et Biophysica Acta (BBA)-Molecular Cell Research. 2010;1803(1):3-19
  11. 11. Levi E, Fridman R, Miao HQ , Ma YS, Yayon A, Vlodavsky I. Matrix metalloproteinase 2 releases active soluble ectodomain of fibroblast growth factor receptor 1. Proceedings of the National Academy of Sciences. 1996;93(14):7069-7074
  12. 12. Preece G, Murphy G, Ager A. Metalloproteinase-mediated regulation of L-selectin levels on leucocytes. Journal of Biological Chemistry. 1996;271(20):11634-11640
  13. 13. Suzuki M, Raab G, Moses MA, Fernandez CA, Klagsbrun M. Matrix metalloproteinase-3 releases active heparin-binding EGF-like growth factor by cleavage at a specific juxtamembrane site. Journal of Biological Chemistry. 1997;272(50):31730-31737
  14. 14. Rundhaug JE. Matrix metalloproteinases and angiogenesis. Journal of Cellular and Molecular Medicine. 2005;9(2):267-285
  15. 15. Solomonov I, Zehorai E, Talmi-Frank D, Wolf SG, Shainskaya A, Zhuravlev A, et al. Distinct biological events generated by ECM proteolysis by two homologous collagenases. Proceedings of the National Academy of Sciences. 2016;113(39):10884-10889
  16. 16. Sternlicht MD, Werb Z. How matrix metalloproteinases regulate cell behavior. Annual Review of Cell and Developmental Biology. 2001;17(1):463-516
  17. 17. Chen Q , Jin M, Yang F, Zhu J, Xiao Q , Zhang L. Matrix metalloproteinases: Inflammatory regulators of cell behaviors in vascular formation and remodeling. Mediators of Inflammation. 2013;2013:928315
  18. 18. Brennan EP, Reing J, Chew D, Myers-Irvin JM, Young EJ, Badylak SF. Antibacterial activity within degradation products of biological scaffolds composed of extracellular matrix. Tissue Engineering. 2006;12(10):2949-2955
  19. 19. Suleman L. Extracellular bacterial proteases in chronic wounds: A potential therapeutic target? Advances in Wound Care. 2016;5(10):455-463
  20. 20. Maeda H, Molla A. Pathogenic potentials of bacterial proteases. Clinica Chimica Acta. 1989;185(3):357-367
  21. 21. Wong W, Wijeyewickrema LC, Kennan RM, Reeve SB, Steer DL, Reboul C, et al. S1 pocket of a bacterially derived subtilisin-like protease underpins effective tissue destruction. Journal of Biological Chemistry. 2011;286(49):42180-42187
  22. 22. Ravanti L, Kähäri VM. Matrix metalloproteinases in wound repair. International Journal of Molecular Medicine. 2000;6(4):391-798
  23. 23. Steffensen B, Häkkinen L, Larjava H. Proteolytic events of wound-healing—Coordinated interactions among matrix metalloproteinases (MMPs), integrins, and extracellular matrix molecules. Critical Reviews in Oral Biology and Medicine. 2001;12(5):373-398
  24. 24. Lähteenmäki K, Virkola R, Sarén A, Emödy L, Korhonen TK. Expression of plasminogen activator Pla of Yersinia pestis enhances bacterial attachment to the mammalian extracellular matrix. Infection and Immunity. 1998;66(12):5755-5762
  25. 25. Chauhan N, Wrobel A, Skurnik M, Leo JC. Yersinia adhesins: An arsenal for infection. PROTEOMICS–Clinical Applications. 2016;10(9-10):949-963
  26. 26. Stevens DL, Bisno AL, Chambers HF, Dellinger EP, Goldstein EJ, Gorbach SL, et al. Practice guidelines for the diagnosis and management of skin and soft tissue infections: 2014 update by the Infectious Diseases Society of America. Clinical Infectious Diseases. 2014;59(2):e10-e52
  27. 27. McMenemy K. Skin and soft tissue infections. Physician Assistant Clinics. 2017;2(2):165-176
  28. 28. Edwards R, Harding KG. Bacteria and wound healing. Current Opinion in Infectious Diseases. 2004;17(2):91-96
  29. 29. Cooper R, Lawrence JC. The isolation and identification of bacteria from wounds. Journal of Wound Care. 1996;5(7):335-340
  30. 30. Bessa LJ, Fazii P, Di Giulio M, Cellini L. Bacterial isolates from infected wounds and their antibiotic susceptibility pattern: Some remarks about wound infection. International Wound Journal. 2015;12(1):47-52
  31. 31. Flemming HC, Wingender J. The biofilm matrix. Nature Reviews Microbiology. 2010;8(9):623-633
  32. 32. Metcalf DG, Bowler PG. Biofilm delays wound healing: A review of the evidence. Burns & Trauma. 2013;1(1):5-12
  33. 33. Attinger C, Wolcott R. Clinically addressing biofilm in chronic wounds. Advances in Wound Care. 2012;1(3):127-132
  34. 34. Dowd SE, Sun Y, Secor PR, Rhoads DD, Wolcott BM, James GA, et al. Survey of bacterial diversity in chronic wounds using pyrosequencing, DGGE, and full ribosome shotgun sequencing. BMC Microbiology. 2008;8(1):43
  35. 35. Rayment EA, Upton Z. Finding the culprit: A review of the influences of proteases on the chronic wound environment. The International Journal of Lower Extremity Wounds. 2009;8(1):19-27
  36. 36. Trengove NJ, Stacey MC, Macauley S, Bennett N, Gibson J, Burslem F, et al. Analysis of the acute and chronic wound environments: The role of proteases and their inhibitors. Wound Repair and Regeneration. 1999;7(6):442-452
  37. 37. Shapiro SD. Matrix metalloproteinase degradation of extracellular matrix: biological consequences. Current Opinion in Cell Biology. 1998;10(5):602-608
  38. 38. Yue J, Zhang K, Chen J. Role of integrins in regulating proteases to mediate extracellular matrix remodeling. Cancer Microenvironment. 2012;5(3):275-283
  39. 39. Bullen EC, Longaker MT, Updike DL, Benton R, Ladin D, Hou Z, et al. Tissue inhibitor of metalloproteinases-1 is decreased and activated gelatinases are increased in chronic wounds. Journal of Investigative Dermatology. 1995;104(2):236-240
  40. 40. Schultz GS, Wysocki A. Interactions between extracellular matrix and growth factors in wound healing. Wound Repair and Regeneration. 2009;17(2):153-162
  41. 41. Krishnaswamy VR, Manikandan M, Munirajan AK, Vijayaraghavan D, Korrapati PS. Expression and integrity of dermatopontin in chronic cutaneous wounds: A crucial factor in impaired wound healing. Cell and Tissue Research. 2014;358(3):833-841
  42. 42. Mast BA, Schultz GS. Interactions of cytokines, growth factors, and proteases in acute and chronic wounds. Wound Repair and Regeneration. 1996;4(4):411-420
  43. 43. Kryczka J, Stasiak M, Dziki L, Mik M, Dziki A, Cierniewski CS. Matrix metalloproteinase-2 cleavage of the β1 integrin ectodomain facilitates colon cancer cell motility. Journal of Biological Chemistry. 2012;287(43):36556-36566
  44. 44. Armstrong DG, Jude EB. The role of matrix metalloproteinases in wound healing. Journal of the American Podiatric Medical Association. 2002;92(1):12-18
  45. 45. Yanhan R, Guosheng G, Min Y, Driver VR. Role of matrix metalloproteinases in chronic wound healing: Diagnostic and therapeutic implications. Chinese Medical Journal. 2014;127(8):1572-1581
  46. 46. Lobmann R, Ambrosch A, Schultz G, Waldmann K, Schiweck S, Lehnert H. Expression of matrix-metalloproteinases and their inhibitors in the wounds of diabetic and non-diabetic patients. Diabetologia. 2002;45(7):1011-1016
  47. 47. Robson MC. The role of growth factors in the healing of chronic wounds. Wound Repair and Regeneration. 1997;5(1):12-17
  48. 48. Gaffney J, Solomonov I, Zehorai E, Sagi I. Multilevel regulation of matrix metalloproteinases in tissue homeostasis indicates their molecular specificity in vivo. Matrix Biology. 2015;44:191-199
  49. 49. Tarnuzzer RW, Schultz GS. Biochemical analysis of acute and chronic wound environments. Wound Repair and Regeneration. 1996;4(3):321-325
  50. 50. Santala A, Saarinen J, Kovanen P, Kuusela P. Activation of interstitial collagenase, MMP-1, by Staphylococcus aureus cells having surface-bound plasmin: A novel role of plasminogen receptors of bacteria. FEBS Letters. 1999;461(3):153-156
  51. 51. Calander AM, Starckx S, Opdenakker G, Bergin P, Quiding-Järbrink M, Tarkowski A. Matrix metalloproteinase-9 (gelatinase B) deficiency leads to increased severity of Staphylococcus aureus-triggered septic arthritis. Microbes and Infection. 2006;8(6):1434-1439
  52. 52. Schmidtchen A, Holst E, Tapper H, Björck L. Elastase-producing Pseudomonas aeruginosa degrade plasma proteins and extracellular products of human skin and fibroblasts, and inhibit fibroblast growth. Microbial Pathogenesis. 2003;34(1):47-55
  53. 53. Okamoto T, Akaike T, Suga M, Tanase S, Horie H, Miyajima S, et al. Activation of human matrix metalloproteinases by various bacterial proteinases. Journal of Biological Chemistry. 1997;272(9):6059-6066
  54. 54. Min D, Moore AG, Bain MA, Breit SN, Lyons JG. Activation of macrophage promatrix metalloproteinase-9 by lipopolysaccharide-associated proteinases. The Journal of Immunology. 2002;168(5):2449-2455
  55. 55. DeCarlo AA Jr, Windsor LJ, Bodden MK, Harber GJ, Birkedal-Hansen B, Birkedal-Hansen H. Activation and novel processing of matrix metalloproteinases by a thiol-proteinase from the oral anaerobe Porphyromonas gingivalis. Journal of Dental Research. 1997;76(6):1260-1270
  56. 56. Oggioni MR, Memmi G, Maggi T, Chiavolini D, Iannelli F, Pozzi G. Pneumococcal zinc metalloproteinase ZmpC cleaves human matrix metalloproteinase 9 and is a virulence factor in experimental pneumonia. Molecular Microbiology. 2003;49(3):795-805
  57. 57. Serra R, Grande R, Buffone G, Molinari V, Perri P, Perri A, et al. Extracellular matrix assessment of infected chronic venous leg ulcers: Role of metalloproteinases and inflammatory cytokines. International Wound Journal. 2016;13(1):53-58
  58. 58. Wang X, Khalil RA. Matrix metalloproteinases, vascular remodeling, and vascular disease. In: Advances in Pharmacology. 2018;81:241-330
  59. 59. Li W, Saji S, Sato F, Noda M, Toi M. Potential clinical applications of matrix metalloproteinase inhibitors and their future prospects. The International Journal of Biological Markers. 2013;28(2):117-130
  60. 60. Gueders MM, Foidart JM, Noel A, Cataldo DD. Matrix metalloproteinases (MMPs) and tissue inhibitors of MMPs in the respiratory tract: Potential implications in asthma and other lung diseases. European Journal of Pharmacology. 2006;533(1-3):133-144
  61. 61. Martins VL, Caley M, O’Toole EA. Matrix metalloproteinases and epidermal wound repair. Cell and Tissue Research. 2013;351(2):255-268
  62. 62. Parks WC. Matrix metalloproteinases in repair. Wound Repair and Regeneration. 1999;7(6):423-432
  63. 63. Saarialho-Kere UK. Patterns of matrix metalloproteinase and TIMP expression in chronic ulcers. Archives of Dermatological Research. 1998;290(1):S47-S54
  64. 64. McCawley LJ, Matrisian LM. Matrix metalloproteinases: They’re not just for matrix anymore! Current Opinion in Cell Biology. 2001;13(5):534-540
  65. 65. Sudbeck BD, Pilcher BK, Welgus HG, Parks WC. Induction and repression of collagenase-1 by keratinocytes is controlled by distinct components of different extracellular matrix compartments. Journal of Biological Chemistry. 1997;272(35):22103-22110
  66. 66. Barone EJ, Yager DR, Pozez AL, Olutoye OO, Crossland MC, Diegelmann RF, et al. Interleukin-1alpha and collagenase activity are elevated in chronic wounds. Plastic and Reconstructive Surgery. 1998;102(4):1023-1027
  67. 67. Muller M, Trocme C, Lardy B, Morel F, Halimi S, Benhamou PY. Matrix metalloproteinases and diabetic foot ulcers: The ratio of MMP-1 to TIMP-1 is a predictor of wound healing. Diabetic Medicine. 2008;25(4):419-426
  68. 68. Herouy YA, Nockowski PI, Schöpf ER, Norgauer JO. Lipodermatosclerosis and the significance of proteolytic remodeling in the pathogenesis of venous ulceration. International Journal of Molecular Medicine. 1999;3(5):511-516
  69. 69. Herouy Y, Pornschlegel G, Stetter C, Grenz H, Schöpf E, Norgauer J, et al. Lipodermatosclerosis is characterized by elevated expression and activation of matrix metalloproteinases: Implications for venous ulcer formation. Journal of Investigative Dermatology. 1998;111(5):822-827
  70. 70. Percival SL, Finnegan S, Donelli G, Vuotto C, Rimmer S, Lipsky BA. Antiseptics for treating infected wounds: Efficacy on biofilms and effect of pH. Critical Reviews in Microbiology. 2016;42(2):293-309
  71. 71. Harris IR, Yee KC, Walters CE, Cunliffe WJ, Kearney JN, Wood EJ, et al. Cytokine and protease levels in healing and non-healing chronic venous leg ulcers. Experimental Dermatology. 1995;4(6):342-349
  72. 72. Nwomeh BC, Liang HX, Cohen IK, Yager DR. MMP-8 is the predominant collagenase in healing wounds and nonhealing ulcers. Journal of Surgical Research. 1999;81(2):189-195
  73. 73. Amato B, Coretti G, Compagna R, Amato M, Buffone G, Gigliotti D, et al. Role of matrix metalloproteinases in non-healing venous ulcers. International Wound Journal. 2015;12(6):641-645
  74. 74. Gao M, Nguyen TT, Suckow MA, Wolter WR, Gooyit M, Mobashery S, et al. Acceleration of diabetic wound healing using a novel protease–anti-protease combination therapy. Proceedings of the National Academy of Sciences. 2015;112(49):15226-15231
  75. 75. Nagase H, Woessner JF. Matrix metalloproteinases. Journal of Biological Chemistry. 1999;274(31):21491-21494
  76. 76. Vu TH, Werb Z. Matrix metalloproteinases: Effectors of development and normal physiology. Genes & Development. 2000;14(17):2123-2133
  77. 77. Lauer G, Sollberg S, Cole M, Krieg T, Eming SA, Flamme I, et al. Expression and proteolysis of vascular endothelial growth factor is increased in chronic wounds. Journal of Investigative Dermatology. 2000;115(1):12-18
  78. 78. Krishnaswamy VR, Balaguru UM, Chatterjee S, Korrapati PS. Dermatopontin augments angiogenesis and modulates the expression of transforming growth factor beta 1 and integrin alpha 3 beta 1 in endothelial cells. European Journal of Cell Biology. 2017;96(3):266-275
  79. 79. Serra R, Gallelli L, Grande R, Amato B, De Caridi G, Sammarco G, et al. Hemorrhoids and matrix metalloproteinases: A multicenter study on the predictive role of biomarkers. Surgery. 2016;159(2):487-494
  80. 80. de Franciscis S, Mastroroberto P, Gallelli L, Buffone G, Montemurro R, Serra R. Increased plasma levels of metalloproteinase-9 and neutrophil gelatinase-associated lipocalin in a rare case of multiple artery aneurysm. Annals of Vascular Surgery. 2013;27(8):1185-e5
  81. 81. Serra R, Buffone G, Falcone D, Molinari V, Scaramuzzino M, Gallelli L, et al. Chronic venous leg ulcers are associated with high levels of metalloproteinases-9 and neutrophil gelatinase-associated lipocalin. Wound Repair and Regeneration. 2013;21(3):395-401
  82. 82. Abdollahi M, Ng TS, Rezaeizadeh A, Aamidor S, Twigg SM, Min D, et al. Insulin treatment prevents wounding associated changes in tissue and circulating neutrophil MMP-9 and NGAL in diabetic rats. PLoS One. 2017;12(2)
  83. 83. Krampert M, Bloch W, Sasaki T, Bugnon P, Rulicke T, Wolf E, et al. Activities of the matrix metalloproteinase stromelysin-2 (MMP-10) in matrix degradation and keratinocyte organization in wounded skin. Molecular Biology of the Cell. 2004;15(12):5242-5254
  84. 84. Bullard KM, Lund L, Mudgett JS, Mellin TN, Hunt TK, Murphy B, et al. Impaired wound contraction in stromelysin-1–deficient mice. Annals of Surgery. 1999;230(2):260
  85. 85. Chen P, Abacherli LE, Nadler ST, Wang Y, Li Q , Parks WC. MMP7 shedding of syndecan-1 facilitates re-epithelialization by affecting α2β1 integrin activation. PLoS One. 2009;4(8)
  86. 86. Mirastschijski U, Impola U, Jahkola T, Karlsmark T, Ågren MS, Saarialho-Kere U. Ectopic localization of matrix metalloproteinase-9 in chronic cutaneous wounds. Human Pathology. 2002;33(3):355-364
  87. 87. Heng MC. Wound healing in adult skin: Aiming for perfect regeneration. International Journal of Dermatology. 2011;50(9):1058-1066
  88. 88. Robson MC, Steed DL, Franz MG. Wound healing: Biologic features and approaches to maximize healing trajectories. Current Problems in Surgery. 2001;2(38):72-140
  89. 89. Woo YC, Park SS, Subieta AR, Brennan TJ. Changes in tissue pH and temperature after incision indicate acidosis may contribute to postoperative pain. Anesthesiology: The Journal of the American Society of Anesthesiologists. 2004;101(2):468-475
  90. 90. Werner S, Grose R. Regulation of wound healing by growth factors and cytokines. Physiological Reviews. 2003;83(3):835-870
  91. 91. Heldin CH, Westermark B. Mechanism of action and in vivo role of platelet-derived growth factor. Physiological Reviews. 1999;79(4):1283-1316
  92. 92. Park JH, Jeong YJ, Park KK, Cho HJ, Chung IK, Min KS, et al. Melittin suppresses PMA-induced tumor cell invasion by inhibiting NF-κB and AP-1-dependent MMP-9 expression. Molecules and Cells. 2010;29(2):209-215
  93. 93. Momota Y, Suzuki N, Kasuya Y, Kobayashi T, Mizoguchi M, Yokoyama F, et al. Laminin alpha3 LG4 module induces keratinocyte migration involvement of matrix metalloproteinase-9. Journal of Receptor and Signal Transduction Research. 2005;25(1):1-17
  94. 94. Giannelli G, Falk-Marzillier J, Schiraldi O, Stetler-Stevenson WG, Quaranta V. Induction of cell migration by matrix metalloprotease-2 cleavage of laminin-5. Science. 1997;277(5323):225-228
  95. 95. Koshikawa N, Giannelli G, Cirulli V, Miyazaki K, Quaranta V. Role of cell surface metalloprotease MT1-MMP in epithelial cell migration over laminin-5. The Journal of Cell Biology. 2000;148(3):615-624
  96. 96. Mäkelä M, Larjava H, Pirilä E, Maisi P, Salo T, Sorsa T, et al. Matrix metalloproteinase 2 (gelatinase A) is related to migration of keratinocytes. Experimental Cell Research. 1999;251(1):67-78
  97. 97. McCawley LJ, O’Brien P, Hudson LG. Epidermal growth factor (EGF)-and scatter factor/hepatocyte growth factor (SF/HGF)-mediated keratinocyte migration is coincident with induction of matrix metalloproteinase (MMP)-9. Journal of Cellular Physiology. 1998;176(2):255-265
  98. 98. da Cunha CB, Klumpers DD, Li WA, Koshy ST, Weaver JC, Chaudhuri O, et al. Influence of the stiffness of three-dimensional alginate/collagen-I interpenetrating networks on fibroblast biology. Biomaterials. 2014;35(32):8927-8936
  99. 99. Jacinto A, Martinez-Arias A, Martin P. Mechanisms of epithelial fusion and repair. Nature Cell Biology. 2001;3(5):E117-E123
  100. 100. Hinz B. Formation and function of the myofibroblast during tissue repair. Journal of Investigative Dermatology. 2007;127(3):526-537
  101. 101. Wysocki AB, Staiano-Coico L, Grinneill F. Wound fluid from chronic leg ulcers contains elevated levels of metalloproteinases MMP-2 and MMP-9. Journal of Investigative Dermatology. 1993;101(1):64-68
  102. 102. Takeo M, Lee W, Ito M. Wound healing and skin regeneration. Cold Spring Harbor Perspectives in Medicine. 2015;5(1):a023267
  103. 103. Rosińczuk J, Taradaj J, Dymarek R, Sopel M. Mechanoregulation of wound healing and skin homeostasis. BioMed Research International. 2016;2016:3943481
  104. 104. Arpino V, Brock M, Gill SE. The role of TIMPs in regulation of extracellular matrix proteolysis. Matrix Biology. 2015;44:247-254
  105. 105. Sadowski T, Dietrich S, Müller M, Havlickova B, Schunck M, Proksch E, et al. Matrix metalloproteinase-19 expression in normal and diseased skin: Dysregulation by epidermal proliferation. Journal of Investigative Dermatology. 2003;121(5):989-996
  106. 106. Inoue M, Kratz G, Haegerstrand A, Ståhle-Bäckdahl M. Collagenase expression is rapidly induced in wound-edge keratinocytes after acute injury in human skin, persists during healing, and stops at re-epithelialization. Journal of Investigative Dermatology. 1995;104(4):479-483
  107. 107. Pilcher BK, Dumin JA, Sudbeck BD, Krane SM, Welgus HG, Parks WC. The activity of collagenase-1 is required for keratinocyte migration on a type I collagen matrix. The Journal of Cell Biology. 1997;137(6):1445-1457
  108. 108. Di Colandrea T, Chada KK, Wang L, Wille J, D’Armiento J. Epidermal expression of collagenase delays wound-healing in transgenic mice. Journal of Investigative Dermatology. 1998;111(6):1029-1033
  109. 109. Ravanti L, Toriseva M, Penttinen R, Crombleholme T, Foschi M, Han J, et al. Expression of human collagenase-3 (MMP-13) by fetal skin fibroblasts is induced by transforming growth factor β via p38 mitogen-activated protein kinase. The FASEB Journal. 2001;15(6):1098-1100
  110. 110. Toriseva MJ, Ala-aho R, Karvinen J, Baker AH, Marjomäki VS, Heino J, et al. Collagenase-3 (MMP-13) enhances remodeling of three-dimensional collagen and promotes survival of human skin fibroblasts. Journal of Investigative Dermatology. 2007;127(1):49-59
  111. 111. Holmbeck K, Bianco P, Caterina J, Yamada S, Kromer M, Kuznetsov SA, et al. MT1-MMP-deficient mice develop dwarfism, osteopenia, arthritis, and connective tissue disease due to inadequate collagen turnover. Cell. 1999;99(1):81-92
  112. 112. Nissinen L, Kähäri VM. Matrix metalloproteinases in inflammation. Biochimica et Biophysica Acta (BBA) - General Subjects. 2014;1840(8):2571-2580
  113. 113. Rechardt O, Vaalamo M, Höök-Nikanne J, Saarialho-Kere U, Elomaa O, Pääkkönen K, et al. Stromelysin-2 is upregulated during normal wound repair and is induced by cytokines. Journal of Investigative Dermatology. 2000;115(5):778-787
  114. 114. Visse R, Nagase H. Matrix metalloproteinases and tissue inhibitors of metalloproteinases: Structure, function, and biochemistry. Circulation Research. 2003;92(8):827-839
  115. 115. Chen W, Fu X, Ge S, Sun T, Sheng Z. Differential expression of matrix metalloproteinases and tissue-derived inhibitors of metalloproteinase in fetal and adult skins. International Journal of Biochemistry & Cell Biology. 2007;39(5):997-1005
  116. 116. Mohan R, Chintala SK, Jung JC, Villar WV, McCabe F, Russo LA, et al. Matrix metalloproteinase gelatinase B (MMP-9) coordinates and effects epithelial regeneration. The Journal of Biological Chemistry. 2002;277(3):2065-2072
  117. 117. Bergers G, Brekken R, McMahon G, Th V, Itoh T, Tamaki K, et al. Matrix metalloproteinase-9 triggers the angiogenic switch during carcinogenesis. Nature Cell Biology. 2000;2(10):737-744
  118. 118. Chun TH, Sabeh F, Ota I, Murphy H, McDonagh KT, Holmbeck K, et al. MT1-MMP-dependent neovessel formation within the confines of the three-dimensional extracellular matrix. The Journal of Cell Biology. 2004;167(4):757-767
  119. 119. Impola U, Toriseva M, Suomela S, Jeskanen L, Hieta N, Jahkola T, et al. Matrix metalloproteinase-19 is expressed by proliferating epithelium but disappears with neoplastic dedifferentiation. International Journal of Cancer. 2003;103(6):709-716
  120. 120. Lazaro JL, Izzo V, Meaume S, Davies AH, Lobmann R, Uccioli L. Elevated levels of matrix metalloproteinases and chronic wound healing: An updated review of clinical evidence. Journal of Wound Care. 2016;25(5):277-287
  121. 121. Rayment EA, Upton Z, Shooter GK. Increased matrix metalloproteinase-9 (MMP-9) activity observed in chronic wound fluid is related to the clinical severity of the ulcer. The British Journal of Dermatology. 2008;158(5):951-961
  122. 122. Lobmann R, Ambrosch A, Schultz G, Waldmann K, Schiweck S, Lehnert H. Expression of matrix-metalloproteinases and their inhibitors in the wounds of diabetic and nondiabetic patients. Diabetologia. 2002;45(7):1011-1016
  123. 123. Krisp C, Jacobsen F, McKay MJ, Molloy MP, Steinstraesser L, Wolters DA. Proteome analysis reveals antiangiogenic environments in chronic wounds of diabetes mellitus type 2 patients. Proteomics. 2013;13(17):2670-2681
  124. 124. de Oliveira RR, Lemos A, de Castro Silveira PV, da Silva RJ, de Moraes SR. Alterations of tendons in patients with diabetes mellitus: A systematic review. Diabetic Medicine: A Journal of the British Diabetic Association. 2011;28(8):886-895
  125. 125. Izzo V, Meloni M, Vainieri E, Giurato L, Ruotolo V, Uccioli L. High matrix metalloproteinase levels are associated with dermal graft failure in diabetic foot ulcers. The International Journal of Lower Extremity Wounds. 2014;13(3):191-196
  126. 126. Kadoglou NP, Daskalopoulou SS, Perrea D, Liapis CD. Matrix metalloproteinases and diabetic vascular complications. Angiology. 2005;56(2):173-189
  127. 127. Ban CR, Twigg SM. Fibrosis in diabetes complications: Pathogenic mechanisms and circulating and urinary markers. Vascular Health and Risk Management. 2008;4(3):575-596
  128. 128. Mendias CL, Gumucio JP, Davis ME, Bromley CW, Davis CS, Brooks SV. Transforming growth factor-beta induces skeletal muscle atrophy and fibrosis through the induction of atrogin-1 and scleraxis. Muscle & Nerve. 2012;45(1):55-59
  129. 129. Dinh T, Tecilazich F, Kafanas A, Doupis J, Gnardellis C, Leal E, et al. Mechanisms involved in the development and healing of diabetic foot ulceration. Diabetes. 2012;61(11):2937-2947
  130. 130. Lu J, Guo JH, Xl T, Zhang C, Zhao M, Zhang QW, et al. Tiron inhibits UVB-induced AP-1 binding sites transcriptional activation on MMP-1 and MMP-3 promoters by MAPK signaling pathway in human dermal fibroblasts. PLoS One. 2016;11(8):e0159998
  131. 131. Chung AW, Hsiang YN, Matzke LA, McManus BM, van Breemen C, Okon EB. Reduced expression of vascular endothelial growth factor paralleled with the increased angiostatin expression resulting from the upregulated activities of matrixmetalloproteinase-2 and -9 in human type 2 diabetic arterial vasculature. Circulation Research. 2006;99(2):140-148
  132. 132. Death AK, Fisher EJ, McGrath KC, Yue DK. High glucose alters matrix metalloproteinase expression in two key vascular cells: Potential impact on atherosclerosis in diabetes. Atherosclerosis. 2003;168(2):263-269
  133. 133. Yadav SS, Singh MK, Dwivedi P, Mandal RK, Usman K, Khattri S, et al. Significance of impaired serum gelatinases activities in metabolic syndrome. Toxicology International. 2014;21(1):107-111
  134. 134. Li Z, Guo S, Yao F, Zhang Y, Li T. Increased ratio of serum matrix metalloproteinase-9 against TIMP-1 predicts poor wound healing in diabetic foot ulcers. Journal of Diabetes and its Complications. 2013;27(4):380-382
  135. 135. Menghini R, Uccioli L, Vainieri E, Pecchioli C, Casagrande V, Stoehr R, et al. Expression of tissue inhibitor of metalloprotease 3 is reduced in ischemic but not neuropathic ulcers from patients with type 2 diabetes mellitus. Acta Diabetologica. 2013;50(6):907-910
  136. 136. Lopez-Lopez N, Gonzalez-Curiel I, Trevino-Santa Cruz MB, Rivas-Santiago B, TrujilloPaez V, Enciso-Moreno JA, et al. Expression and vitamin D-mediated regulation of matrix metalloproteinases (MMPs) and tissue inhibitors of metalloproteinases (TIMPs) in healthy skin and in diabetic foot ulcers. Archives of Dermatological Research. 2014;306(9):809-821
  137. 137. Burrow JW, Koch JA, Chuang HH, Zhong W, Dean DD, Sylvia VL. Nitric oxide donors selectively reduce the expression of matrix metalloproteinases-8 and -9 by human diabetic skin fibroblasts. The Journal of Surgical Research. 2007;140(1):90-98
  138. 138. Overall CM, Kleifeld O. Towards third generation matrix metalloproteinase inhibitors for cancer therapy. British Journal of Cancer. 2006;94(7):941-946
  139. 139. Amar S, Minond D, Fields GB. Clinical implications of compounds designed to inhibit ECM-modifying metalloproteinases. Proteomics. 2017;17(23-24):1600389
  140. 140. Gooyit M, Peng Z, Wolter WR, Pi H, Ding D, Hesek D, et al. A chemical biological strategy to facilitate diabetic wound healing. ACS Chemical Biology. 2014;9(1):105-110
  141. 141. Nguyen TT, Ding D, Wolter WR, Pérez RL, Champion MM, Mahasenan KV, et al. Validation of matrix metalloproteinase-9 (MMP-9) as a novel target for treatment of diabetic foot ulcers in humans and discovery of a potent and selective small-molecule MMP-9 inhibitor that accelerates healing. Journal of Medicinal Chemistry. 2018;61(19):8825-8837
  142. 142. Appleby TC, Greenstein AE, Hung M, Liclican A, Velasquez M, Villaseñor AG, et al. Biochemical characterization and structure determination of a potent, selective antibody inhibitor of human MMP9. Journal of Biological Chemistry. 2017;292(16):6810-6820
  143. 143. Bendell JC, Starodub A, Shah MA, Sharma S, Wainberg ZA, Thai DL. Phase I study of GS-5745 alone and in combination with chemotherapy in patients with advanced solid tumors. Journal of Clinical Oncology. 2015;33(Suppl 15):4030-4030
  144. 144. Shah MA, Starodub A, Sharma S, Berlin J, Patel M, Wainberg ZA, et al. Andecaliximab/GS-5745 alone and combined with mFOLFOX6 in advanced gastric and gastroesophageal junction adenocarcinoma: Results from a phase I study. Clinical Cancer Research. 2018;24(16):3829-3837
  145. 145. Sela-Passwell N, Kikkeri R, Dym O, Rozenberg H, Margalit R, Arad-Yellin R, et al. Antibodies targeting the catalytic zinc complex of activated matrix metalloproteinases show therapeutic potential. Nature Medicine. 2012;18(1):143
  146. 146. Hu J, Van den Steen PE, Houde M, Ilenchuk TT, Opdenakker G. Inhibitors of gelatinase B/matrix metalloproteinase-9 activity: Comparison of a peptidomimetic and polyhistidine with single-chain derivatives of a neutralizing monoclonal antibody. Biochemical Pharmacology. 2004;67(5):1001-1009
  147. 147. Paemen L, Martens E, Masure S, Opdenakker G. Monoclonal antibodies specific for natural human neutrophil gelatinase B used for affinity purification, quantitation by two-site ELISA and inhibition of enzymatic activity. European Journal of Biochemistry. 1995;234(3):759-765
  148. 148. Hariono M, Yuliani SH, Istyastono EP, Riswanto FD, Adhipandito CF. Matrix metalloproteinase 9 (MMP9) in wound healing of diabetic foot ulcer: Molecular target and structure-based drug design. Wound Medicine. 2018;22:1-3
  149. 149. Vowden K, Vowden P. Wound dressings: Principles and practice. Surgery. 2014;32:462-467
  150. 150. Rayment EA, Dargaville TR, Shooter GK, George GA, Upton Z. Attenuation of protease activity in chronic wound fluid with bisphosphonate-functionalised hydrogels. Biomaterials. 2008;29(12):1785-1795
  151. 151. Tronci G, Yin J, Holmes RA, Liang H, Russell SJ, Wood DJ. Protease-sensitive atelocollagen hydrogels promote healing in a diabetic wound model. Journal of Materials Chemistry B. 2016;4(45):7249-7258
  152. 152. Ågren MS, Mirastschijski U, Karlsmark T, Saarialho-Kere UK. Topical synthetic inhibitor of matrix metalloproteinases delays epidermal regeneration of human wounds. Experimental Dermatology. 2001;10(5):337-348
  153. 153. Jeong EH, Kim H, Jang B, Cho H, Ryu J, Kim B, et al. Technological development of structural DNA/RNA-based RNAi systems and their applications. Advanced Drug Delivery Reviews. 2016;104:29-43
  154. 154. Wang J, Lu Z, Wientjes MG, Au JL. Delivery of siRNA therapeutics: Barriers and carriers. The AAPS Journal. 2010;12(4):492-503
  155. 155. Georgiou TK, Vamvakaki M, Patrickios CS, Yamasaki EN, Phylactou LA. Nanoscopic cationic methacrylate star homopolymers: Synthesis by group transfer polymerization, characterization and evaluation as transfection reagents. Biomacromolecules. 2004;5(6):2221-2229
  156. 156. Srinivasachari S, Fichter KM, Reineke TM. Polycationic β-cyclodextrin “click clusters”: Monodisperse and versatile scaffolds for nucleic acid delivery. Journal of the American Chemical Society. 2008;130(14):4618-4627
  157. 157. Xu FJ, Zhang ZX, Ping Y, Li J, Kang ET, Neoh KG. Star-shaped cationic polymers by atom transfer radical polymerization from β-cyclodextrin cores for nonviral gene delivery. Biomacromolecules. 2009;10(2):285-293
  158. 158. Cryan SA, Holohan A, Donohue R, Darcy R, O’Driscoll CM. Cell transfection with polycationic cyclodextrin vectors. European Journal of Pharmaceutical Sciences. 2004;21(5):625-633
  159. 159. Yang C, Zhu P, Yan L, Chen L, Meng R, Lao G. Dynamic changes in matrix metalloproteinase 9 and tissue inhibitor of metalloproteinase 1 levels during wound healing in diabetic rats. Journal of the American Podiatric Medical Association. 2009;99(6):489-496
  160. 160. Li N, Luo HC, Yang C, Deng JJ, Ren M, Xie XY, et al. Cationic star-shaped polymer as an siRNA carrier for reducing MMP-9 expression in skin fibroblast cells and promoting wound healing in diabetic rats. International Journal of Nanomedicine. 2014;9:3377
  161. 161. Li N, Luo HC, Ren M, Zhang LM, Wang W, Pan CL, et al. Efficiency and safety of β-CD-(D3) 7 as siRNA carrier for decreasing matrix metalloproteinase-9 expression and improving wound healing in diabetic rats. ACS Applied Materials & Interfaces. 2017;9(20):17417-17426
  162. 162. Wang W, Yang C, Yi Wang X, Yan Zhou L, Juan Lao G, Liu D, et al. MicroRNA-129 and-335 promote diabetic wound healing by inhibiting Sp1-mediated MMP-9 expression. Diabetes. 2018;67(8):1627-1638
  163. 163. Rah B, Amin H, Yousuf K, Khan S, Jamwal G, Mukherjee D, et al. A novel MMP-2 inhibitor 3-azidowithaferin A (3-azidoWA) abrogates cancer cell invasion and angiogenesis by modulating extracellular Par-4. PLoS One. 2012;7(9):e44039
  164. 164. Ji BC, Hsiao YP, Tsai CH, Chang SJ, Hsu SC, Liu HC, et al. Cantharidin impairs cell migration and invasion of A375. S2 human melanoma cells by suppressing MMP-2 and-9 through PI3K/NF-κB signaling pathways. Anticancer Research. 2015;35(2):729-738
  165. 165. Wang Z, Zhai Z, Du X. Celastrol inhibits migration and invasion through blocking the NF-κB pathway in ovarian cancer cells. Experimental and Therapeutic Medicine. 2017;14(1):819-824
  166. 166. Liu KC, Huang AC, Wu PP, Lin HY, Chueh FS, Yang JS, et al. Gallic acid suppresses the migration and invasion of PC-3 human prostate cancer cells via inhibition of matrix metalloproteinase-2 and-9 signaling pathways. Oncology Reports. 2011;26(1):177-184
  167. 167. Kim WK, Song SY, Oh WK, Kaewsuwan S, Tran TL, Kim WS, et al. Wound-healing effect of ginsenoside Rd from leaves of Panax ginseng via cyclic AMP-dependent protein kinase pathway. European Journal of Pharmacology. 2013;702(1-3):285-293
  168. 168. Liu W, Zhang Q , Tang Q , Hu C, Huang J, Liu Y, et al. Lycorine inhibits cell proliferation and migration by inhibiting ROCK1/cofilin-induced actin dynamics in HepG2 hepatoblastoma cells. Oncology Reports. 2018;40(4):2298-2306
  169. 169. Aroui S, Aouey B, Chtourou Y, Meunier AC, Fetoui H, Kenani A. Naringin suppresses cell metastasis and the expression of matrix metalloproteinases (MMP-2 and MMP-9) via the inhibition of ERK-P38-JNK signaling pathway in human glioblastoma. Chemico-Biological Interactions. 2016;244:195-203
  170. 170. Park EJ, Park HJ, Chung HJ, Shin Y, Min HY, Hong JY, et al. Antimetastatic activity of pinosylvin, a natural stilbenoid, is associated with the suppression of matrix metalloproteinases. The Journal of Nutritional Biochemistry. 2012;23(8):946-952
  171. 171. Chun J, Kim YS. Platycodin D inhibits migration, invasion, and growth of MDA-MB-231 human breast cancer cells via suppression of EGFR-mediated Akt and MAPK pathways. Chemico-Biological Interactions. 2013;205(3):212-221
  172. 172. Zhou R, Xu L, Ye M, Liao M, Du H, Chen H. Formononetin inhibits migration and invasion of MDA-MB-231 and 4T1 breast cancer cells by suppressing MMP-2 and MMP-9 through PI3K/AKT signaling pathways. Hormone and Metabolic Research. 2014;46(11):753-760
  173. 173. Zeng S, Zhu B, Zeng J, Wu W, Jiang C. Zeylenone represses the progress of human prostate cancer by downregulating the Wnt/β-catenin pathway. Molecular Medicine Reports. 2018;18(6):5572-5578
  174. 174. Yen YH, Pu CM, Liu CW, Chen YC, Chen YC, Liang CJ, et al. Curcumin accelerates cutaneous wound healing via multiple biological actions: The involvement of TNF-α, MMP-9, α-SMA, and collagen. International Wound Journal. 2018;15(4):605-617
  175. 175. Deng B, Qiu B. Shikonin inhibits invasiveness of osteosarcoma through MMP13 suppression. Tumor Biology. 2015;36(12):9311-9317
  176. 176. Zhang FY, Hu Y, Que ZY, Wang P, Liu YH, Wang ZH, et al. Shikonin inhibits the migration and invasion of human glioblastoma cells by targeting phosphorylated β-catenin and phosphorylated PI3K/Akt: A potential mechanism for the anti-glioma efficacy of a traditional Chinese herbal medicine. International Journal of Molecular Sciences. 2015;16(10):23823-23848

Written By

Kirandeep Kaur, Atamjit Singh, Shivani Attri, Danish Malhotra, Aditi Verma, Neena Bedi and Preet Mohinder Singh Bedi

Submitted: 02 February 2020 Reviewed: 22 May 2020 Published: 19 November 2020