4,125
Views
482
CrossRef citations to date
0
Altmetric
REVIEW ARTICLE

Matrix metalloproteinases: Contribution to pathogenesis, diagnosis and treatment of periodontal inflammation

, , , , , , , & show all
Pages 306-321 | Published online: 08 Jul 2009

Abstract

Matrix metalloproteinases (MMPs) form a family of enzymes that mediate multiple functions both in the tissue destruction and immune responses related to periodontal inflammation. The expression and activity of MMPs in non‐inflamed periodontium is low but is drastically enhanced to pathologically elevated levels due to the dental plaque and infection‐induced periodontal inflammation. Soft and hard tissue destruction during periodontitis and peri‐implantitis are thought to reflect a cascade of events involving bacterial virulence factors/enzymes, pro‐inflammatory cytokines, reactive oxygen species and MMPs. However, recent studies suggest that MMPs can also exert anti‐inflammatory effects in defence of the host by processing anti‐inflammatory cytokines and chemokines, as well as by regulating apoptotic and immune responses. MMP‐inhibitor (MMPI)‐drugs, such as doxycycline, can be used as adjunctive medication to augment both the scaling and root planing‐treatment of periodontitis locally and to reduce inflammation systematically. Furthermore, MMPs present in oral fluids (gingival crevicular fluid (GCF), peri‐implant sulcular fluid (PISF), mouth‐rinses and saliva) can be utilized to develop new non‐invasive, chair/bed‐side, point‐of‐care diagnostics for periodontitis and dental peri‐implantitis.

Abbreviations
AP‐1=

activating protein‐1

(CA)n=

microsatellite containing tandem repeats of deoxy (cytidine, adenine) sequences

ECM=

extracellular matrix

EDTA=

ethylenediamine tetraacetic acid

FDA=

Food and Drug Administration

IFMA=

time‐resolved immunofluorometric assay

LPS=

lipopolysaccharide

RUNT=

RUNT‐domain containing transcription factor

RUNX=

RUNX‐related protein containing transcription factor

TGF=

transforming growth factor

TPA=

tetradecanoylphorbol acetate

Introduction

Four decades ago, the first collagenolytic enzyme was discovered Citation1. This collagenase (MMP‐1) was responsible for the involution of the tadpole tail during amphibian morphogenesis Citation1. Today, it is recognized that the matrix metalloproteinases (MMPs) constitute a multigene family of over 25 structurally related but genetically distinct secreted or cell surface‐associated proteolytic enzymes that can process or degrade numerous extracellular, pericellular and non‐matrix substrates Citation2–8. In order to classify the MMP superfamily it is important to recognize their specific characteristics. Each MMP consists of a specific sequence of domains with several different motifs Citation2–8. This domain structure includes the signal peptide, the propeptide domain, the catalytic domain, and the C‐terminal hemopexin‐like domain, which are present in almost all MMPs Citation2–8. However, certain MMPs have additional domains, such as a transmembrane or a cytoplasmic domain Citation2–8. To date 25 members of the MMP family have been identified and characterized, including 22 found in the human genome. Structural similarities of the MMPs suggest that they have originated from a common ancestor by a series of gene duplication Citation2–8. MMP genes coding for MMP‐1, ‐3, ‐7, ‐8, ‐10, ‐12, ‐13, ‐20, ‐21 and ‐28 are located in the chromosome 11 at 11q21‐23, chromosome 10, and chromosome 17, respectively, and the others are located in chromosomes 1, 8, 12, 14, 16, 20 and 22 Citation2–6. Since MMPs can collaboratively degrade almost all tissue and structural components of connective tissue matrices and basement membranes, their activities are strictly controlled at different levels Citation2–8. In addition, as described in detail below, certain MMPs can act as metabolic regulators by processing various non‐matrix bioactive molecules to modulate various cellular, immunologic, intracellular, apoptotic and anti‐inflammatory responses Citation3–5.

Key messages

  • Matrix metalloproteinases (MMPs) are among the key mediators of irreversible tissue destruction in periodontitis and peri‐implantitis

  • MMPs can be inhibited by doxycycline in periodontitis with beneficial clinical outcome

  • MMPs can be diagnostically utilized in point‐of‐care bed/chair‐side tests

  • MMP inhibition by doxycycline seemingly also exert systemically beneficial anti‐inflammatory effects

Structural and functional features of matrix metalloproteinases (MMPs)

For an enzyme to be classified as an MMP Citation1–8, two conserved motifs are required, namely the prodomain and the catalytic domain. The prodomain usually consists of about 80 amino acids and contains a conserved cysteine residue which can bind to the zinc atom in the catalytic domain. The disruption of this bond, usually mediated as a result of cleavage(s) of the prodomain, is required for activation of the latent proenzyme to its catalytically competent form. Serine and cysteine proteinases of mammalian and microbial origins as well as other MMPs can proteolytically process the prodomain, which results directly form this limited proteolysis, or from a subsequent autocatalytic process which results in the lower molecular weight active form(s) Citation2–8. The importance of calcium ions for the optimal catalytic competence of MMPs is reflected by the inhibition of the enzyme activity by metal chelators such as EDTA and the tetracyclines Citation2–8.

In addition to the conserved pro‐ and catalytic domains, MMPs contain various other functional domains mediating substrate specificity, recognition and interaction with other proteins and molecules Citation2–8. All MMPs except MMPs ‐7, ‐23 and ‐26 have a proline‐rich hinge region and a hemopexin‐like C‐terminal domain Citation2–8. The hemopexin domain is involved in the binding of tissue inhibitors of metalloproteinases (TIMPs) and in the cleavage of the native triple helical collagen Citation2–8. Both ends of the hemopexin domain contain cysteine residues, which by formation of a disulfide bridge folds the domain into a four‐bladed propeller structure Citation4. The two gelatinases, MMP‐2 and ‐9, contain repeats of fibronectin type II modules that are responsible for the gelatin‐binding properties Citation8. Membrane‐type MMPs (MT‐MMPs) and MMP‐11, ‐21, ‐23 and ‐28 contain a furin‐cleavage site between the propeptide and catalytic domain, which is proteolytic activation site for the furin‐like enzymes Citation8. Four of the six membrane‐type MMPs (MT1‐, MT2‐, MT3‐ and MT5‐MMP) contain a single transmembrane and a single cytosolic domain, and MT4‐ and MT6‐MMPs are connected to the cell surface with a C‐terminal hydrophobic extension which acts as a glycosyl phosphatidyl inositol (GPI)‐anchoring signal Citation9. These domains direct the MT‐MMPs onto a cell surface Citation9,10.

Collagenases

MMP‐1 (collagenase‐1) is produced as two different forms, mainly as a 52 kDa less‐glycosylated protein but also as a 57 kDa glycosylated form. After removal of the propeptide, the active 42 and 48 kDa MMP‐1 species can digest fibrillar interstitial collagens at a single specific site, Gly775‐Ile776 in α1(I)‐chain and Gly775‐Leu776 in α2(I)‐chain producing the classical αA (3/4)‐ and αB (1/4)‐cleavage products Citation1,2,Citation6. Mutations in these collagenase‐cleavage sites can render the collagen fibrils resistant to MMP‐1 and other collagenases (MMP‐8, ‐13) Citation2,Citation6. MMP‐1 has a wide range of other substrates, including various structural extracellular matrix (ECM) components as well as soluble non‐matrix mediators and other MMPs Citation2, Citation6.

Collagenase‐2 (MMP‐8), also called neutrophil collagenase, has previously been thought to be expressed solely by leukocytes of the polymorphonuclear (PMN) lineage cell line Citation2,Citation6,Citation11. MMP‐8 is synthesized during the maturation of PMNs in the bone marrow; it then becomes glycosylated and is prestored in the subcellular specific granules, from where it can be released by selective degranulation in response to appropriate triggering stimuli Citation2,Citation6,Citation11–14. The neutrophil (PMN)‐type MMP‐8 is more glycosylated than other MMPs including the fibroblast‐type MMP‐8 Citation11,Citation15; these carbohydrate moieties of PMN‐type MMP‐8 are believed to act as targeting signals directing its subcellular location into intracellular specific granules of PMNs Citation2,Citation6,Citation11,Citation15 and may explain the sensitivity of PMN‐type MMP‐8 to activation and inactivation by reactive oxygen species Citation11,Citation14,Citation16. MMP‐8 can also be produced (de novo expressed) by articular chondrocytes Citation17,18, and during various inflammatory diseases such as bronchitis, asthma, periodontitis and arthritis by resident synovial and gingival fibroblasts, epithelial cells/keratinocytes, odontoblasts, oral cancer cells, monocyte/macrophages and plasma cells Citation15,Citation19–27. Like MMP‐1, MMP‐8 also has wide substrate specificity; and its activities overlap with the other collagenases (MMP‐1 and ‐13) Citation2,Citation6,Citation11,Citation28. Recently, MMP‐8 has been shown to exert unexpected anti‐inflammatory defensive and protective characteristics against the spread of experimental skin cancer and inflammatory lung diseases probably by processing anti‐inflammatory cytokines and chemokines as well as regulating inflammatory cell apoptosis and immune response Citation29–31.

The third member of the collagenase subfamily is collagenase‐3 (MMP‐13) which was originally cloned from a human breast cancer cDNA library Citation32. MMP‐13 seems to have a very restricted expression but broad substrate specificity and is a catalytically very efficient enzyme Citation6,Citation33–36. MMP‐13 is expressed during bone development as well as in fetal and gingival/oral wound healing Citation21,Citation27,Citation34–36. Enhanced MMP‐13 levels have been shown to be expressed in many pathological tissue destructive conditions like arthritis, periodontal disease, chronic ulcers, atherosclerosis and many types of malignant tumours Citation6,Citation21,Citation27,Citation32–38. MMP‐13 expression is also related to the invasive capacity of various malignant cell lines and tumours Citation6,Citation32,Citation35,Citation38. The ability of MMP‐13 to digest type II collagen ten times more efficiently than types I or III suggests that MMP‐13 also is involved in the pathophysiology of the arthritides Citation33,Citation36,37. In addition, MMP‐13 can also process chemokines and can be inhibited by tetracyclines and bisphosphonate alendronate Citation5,6,Citation34–36.

Gelatinases

The two gelatinases, 72 kDa gelatinase A (MMP‐2) and 92 kDa gelatinase B (MMP‐9) Citation2–8, have rather similar substrate specificity Citation8. Both progelatinases can be isolated as complexes with TIMPs; proMMP‐2 binds to TIMP‐2 and proMMP‐9 to TIMP‐1 Citation2–8. MMP‐2 has a characteristic activation mechanism on the cell surface involving MT‐MMPs and involving also TIMP‐2 Citation2–10, whereas human proMMP‐9 is sensitive to activation by a serine proteinase, human trypsin‐2 and oxidants Citation8,Citation11.

92 kDa MMP‐9 can be expressed by various cell lines such as keratinocytes, osteoclasts, eosinophils, neutrophils and macrophages Citation2–11. Increased MMP‐9 production and activation has been detected in many kinds of inflammatory and malignant diseases Citation8 such as periodontitis, peri‐implantitis and pericoronitis as well as various malignant tumours Citation8,Citation39–43. MMP‐9 expression and activation can be induced by many inflammatory cytokines such as tumour necrosis factor‐α (TNF‐α), interleukin‐1 (IL‐1), interferon‐γ (IFN‐ γ), IL‐2, but also decreased by IL‐10 and activation IL‐4 Citation8,Citation42,43. Correspondingly, its production and activation can be controlled with the ‘biological’ drugs exemplified by the etanercept‐type TNF‐blockers Citation42.

Stromelysins

The stromelysin subfamily consists of stromelysin‐1 (MMP‐3) and stromelysin‐2 (MMP‐10) Citation2–6. Their main substrates are basement membrane components such as laminins and type IV collagen but not interstitial collagens Citation2–6. Stromelysins can also cleave various ECM components and activate other proMMPs Citation2–6. MMP‐3 appears to have many roles; for example, it can induce tissue remodelling and destruction, apoptosis or promote differentiation Citation2–6. MMP‐3 has also been shown to be involved in the gingival tissue and crevicular fluid cooperative MMP‐activation cascades in periodontitis Citation28,Citation43,44.

Membrane‐type MMPs

Membrane‐type MMPs (MT‐MMPs) differ from the other MMPs in that they are bound to the cell surface Citation9,10,Citation45. Six different MT‐MMPs have been identified and characterized, MT1‐, MT2‐, MT3‐ and MT5‐MMPs have a transmembrane and a C‐terminal cytosolic domain Citation2–5,Citation9,10,Citation45,46, whereas MT4‐MMP and MT6‐MMP are bound to the cell surface with a C‐terminal hydrophopic extension that acts as a glycosyl phosphatidyl inositol (GPI) anchor Citation2–5,Citation9,10.

MT‐MMPs can modify the pericellular environment by degrading various ECM components Citation9,10,Citation45. MT‐MMPs can digest native type I–III collagens Citation45, and can activate other MMPs like proMMP‐2, proMMP‐8 and proMMP‐13 Citation45–47. MT1‐MMP expression is regulated by various cytoskeleton‐ECM interactions (45–59). MT1‐MMP plays also an important role in the angiogenesis and in tumour growth and spread Citation45. MT5‐MMP has been detected in brain and MT6‐MMP in PMNs Citation48,49, and increased expression of the MT‐MMPs has been found in many malignant tumours, and in inflammatory conditions such as arthritis and periodontitis Citation49–54. Soluble or shedded form(s) of MT1‐MMP (MMP‐14) and MT6‐MMP (MMP‐25) have been detected in culture media of human renal mesangial cells, breast cancer cells, gingival fibroblasts, periodontitis‐affected human gingival crevicular fluid (GCF), inflamed bronchoalveolar lavage fluid and tear fluid Citation47,Citation54–58.

Tissue inhibitors of matrix metalloproteinases (TIMPs)

TIMPs participate in the regulation of the ECM metabolism Citation2–5,Citation35,Citation59–61. The TIMP family consists of four members (TIMPs 1–4). TIMPs share some important structural features and an ability to inhibit MMPs, but exert also specific or selective distinct biochemical characteristics and expression profiles Citation2–5,Citation35,Citation59–61.

TIMPs are in general variably glycosylated proteins with molecular masses of 21–34 kDa, their nucleotide sequence homology is close to 25%. TIMPs contain 12 conserved cysteine residues capable of pairing to 6 disulphide bridges, of which 3 are located in the N‐terminal and 3 in the C‐terminal domain Citation2–5,Citation35,Citation59–61. The conserved N‐terminal domain of TIMPs is usually responsible for their biological activities and MMP binding Citation2–5,Citation61. Disulfide bonds in the N‐terminal domain are required for the MMP inhibitory activity Citation2–5,Citation61. The C‐terminal domains participate in the complex formation with the proMMPs Citation2–5,Citation35,Citation59–61. TIMPs 1–4 can inhibit all MMPs but TIMP‐1 is a relatively ineffective inhibitor of MMP‐19 and membrane‐type MMPs. TIMPs ‐1, ‐2 and ‐4 are secreted extracellular proteins, whereas TIMP‐3 is bound to the extracellular matrix Citation2. TIMPs ‐1, ‐2 and ‐3 are expressed by several cell types, and the TIMP‐4 expression has been found in various tissues including brain, heart, ovary and skeletal muscle Citation2–5,Citation35,Citation59–61. The primary function of TIMPs is regarded to be MMP inhibition, but TIMPs can exert also other functions, which may include MMP transportation and stabilization, MMP focalization to the cell surface via MT‐MMP binding, inhibition of angiogenesis, promotion of bone‐resorbing activity, growth factor‐like activity, and pro‐ and anti‐apoptotic functions Citation2,Citation62,63. All these natural functions of TIMPs cannot be obtained with the synthetic MMP‐inhibitors Citation59–63.

Regulation of MMP activity and expression

MMP activity is regulated at multiple levels, including transcription, secretion/degranulation activation of the zymogen, inhibition of activity, extracellular inhibitors, localization either inside or outside the cell and internalization as well as by clearance Citation2–8,Citation11–16,Citation28,Citation35,Citation59,Citation64.

Activity and expression of most MMPs are maintained at undetectable low or quiescent levels in intact normal tissues Citation2–8,Citation35,Citation59,Citation64. The expression of MMP genes is regulated by transcription factors, co‐activators and co‐repressor proteins Citation2–8, Citation59. Transcriptional activation can be stimulated by a variety of pro‐inflammatory cytokines, hormones and growth factors, such as TNF‐α, IL‐1, IL‐6, epidermal growth factor, platelet‐derived growth factor and fibroblast growth factor Citation2–8,Citation28,Citation34,35,Citation57,Citation59,Citation61,Citation64–69. For example, CD40‐ligand can induce MMP‐1 expression, insulin MMP‐12 expression, hypoxia the expression of MMP‐2, MT1‐MMP and MMP‐13, and hyperoxia the expression of MMP‐8 and ‐9 Citation2–8,Citation34,35,Citation59,Citation61,Citation64–69. Activation of latent proMMPs can occur intracellularly, at the cell surface by MT‐MMPs and in the extracellular space by other proteases Citation2–8,Citation45–47. Plasmin and trypsins are regarded to be important serine proteinase activators of MMPs in vivoCitation2–8,Citation11–16,Citation70–72. Cysteine proteinases can activate proMMPs released from gingival fibroblasts and proforms of serine proteinases Citation57. Bacterial and candidal proteinases as well as reactive oxygen species can activate proMMPs Citation11,Citation14,Citation16,Citation59,Citation61,Citation73–75. Activated MMPs can further participate in the processing and activation of other MMPs in mutual activation cascades Citation2–8,Citation45–47. Human trypsin‐2 is one of the potential candidates to be the initial activating serine proteinase of proMMP‐activation cascade Citation70–72. Furthermore, human trypsin‐2 can also directly degrade native interstitial type I and II collagens Citation71,72. In most cases, proMMP activation involves the direct removal of the prodomain, resulting in about 10 kDa lower molecular weight active forms Citation2,Citation6,Citation8,Citation70–73, but recent studies have also shown that certain MMPs can be active while in full‐size or even complex forms evidently due to the interaction(s) with substrates, ligands, inhibitors and oxidants Citation16,Citation39,Citation59,Citation76,77.

MMPs in periodontal inflammation

Bacterial dental plaque is currently considered to be the major risk factor for the development of periodontal disease Citation28,Citation64,Citation73,Citation75,Citation78. Periodontal destruction is due to uncontrolled inflammatory response to the bacterial insult consisting of a group of Gram‐negative anaerobic periodontopathogenic bacteria organized as a biofilm Citation28,Citation64,Citation73,Citation75,Citation78. The mechanisms involved in the initiation and progression of periodontal diseases are not clearly understood Citation28,Citation64,Citation75. The toxins, enzymes and metabolites of potent periodontopathogenic bacteria present in the dental plaque are considered to be the initiating factors of the host immune response which is primarily responsible for the tissue destruction. It is the host cells which produce and release proinflammatory mediators (cytokines and chemokines) as well as reactive oxygen species, prostaglandins and various proteinases Citation11–13,Citation28,Citation57,Citation75,Citation79. Lipopolysaccharides (LPS) derived from bacterial membrane have the capacity to activate host epithelial cells to express and release proinflammatory cytokines such as interleukin‐1, interleukin‐8, tumour necrosis factor‐α, prostaglandins and proteases Citation11–15,Citation28, Citation57, Citation64, Citation75. LPS represents one of the many microbe‐derived ligands for the toll‐like receptors, which are involved in the microbe‐induced inflammatory responses in periodontitis and other diseases Citation64,Citation75. Chemoattractant signals are initiated and transmigrating recruited leukocytes and monocyte/macrophages to amplify inflammation at the local site Citation28,Citation64. The next stage of circulus vitiosus of periodontal inflammation follows; resident periodontal ligament and other cells in the periodontium, such as gingival fibroblasts, monocyte/macrophages, gingival sulcular epithelial cells/oral keratinocytes, osteoblasts/osteoclasts and endothelial cells, are activated in response to various pro‐inflammatory stimuli to express and secrete pro‐inflammatory cytokines, cysteine proteinases and MMPs Citation12,13,Citation15,Citation28,Citation57,Citation64,Citation74,75,Citation78.

The involvement of cytokines in MMP transcription

Three important regulatory mechanisms can control the mode and extent of MMP expression in the inflamed periodontium: regulation of transcription levels and secretion; activation of the proenzyme in the extracellular milieu and on the cell surfaces; and, finally, regulation/inhibition of MMP activity by their endogenous inhibitors (TIMPs) Citation2–8,Citation11–13,Citation28,Citation35,Citation64,Citation80. In addition, MMPs are degraded and targets of clearance Citation2–8,Citation28,Citation59,Citation61,Citation64,Citation80,81. MMP genes are expressed when required in a physiological extracellular matrix remodelling or in the pathological tissue destruction Citation2–8,Citation28,Citation59,Citation64,Citation80,81. Furthermore, certain inductive MMPs, such as MMP‐8 and ‐9, have recently been shown to protect against the spread of experimental lung inflammation and skin cancer Citation28–31,Citation59,Citation79. MMP‐1 and ‐2 are known to be constitutively expressed at low levels in the periodontium Citation2–8,Citation28,Citation64,65. However, the expression of inductive MMPs can be up‐ or down‐regulated by pro‐ and anti‐inflammatory cytokines, extracellular matrix proteins, bacterial virulence factors and enzymes, cell stress, cell shape changes, cellular contacts and communication, and by phorbol esters Citation2–8,Citation28,Citation35,Citation59,Citation64,65,Citation80,81. Furthermore, MMP expression can be induced or repressed by MMPs' own substrate(s) either in native intact and/or cleaved/processed forms with various outcomes Citation4–6,Citation59,Citation64,Citation80,81. For example, type I collagen can act as a ligand for the discoidin‐domain containing receptor‐like tyrosine kinases, which can up‐regulate MMP‐1 expression when activated by intact type I collagen and which become inactivated if the type I collagen ligand is degraded by collagenases Citation59, Citation80–83.

Many MMP, including MMP‐1, ‐3, ‐7, ‐9, ‐10, ‐12 and ‐13 gene promoters, comprise activating protein‐1 (AP‐1) consensus elements, such as AP‐1 binding site, polyoma virus enhancer activator or PEA1, tetradecanoylphorbol acetate (TPA)‐responsive element or TPE, one of which is located at about ‐70. The AP‐1 binding site is activated upon interaction with AP‐1 consisting of certain activated and complexed transcription factors such as the products of immediate early genes Fos and Jun. Other important MMP promoters include polyoma enhancer activator 3 site, which is not found in MMP‐2, binding Ets erythroblastosis 26, transformation‐specific protein first discovered in the E26 avian erythroblastosis virus, nuclear factor Kappa B binding sites binding NF Kappa B, and RUNX‐2 binding site, only found in MMP‐13 promoter, and binding RUNT domain factor‐2 Citation2–8,Citation65–69,Citation80,81,Citation84. These MMP promoters can be up‐regulated by various proinflammatory cytokines, growth factors, the extracellular matrix metalloproteinase inducer (EMMPRIN) and bacterial virulence factors Citation2–8,Citation28,Citation64–69,Citation80,81,Citation84. Resident cells, i.e. gingival fibroblasts, epithelial cells/keratinocytes, osteoblasts/osteoclasts, periodontal ligament cells, in the periodontium together with recruited inflammatory cells, i.e., neutrophils, monocytes/macrophages and plasma cells, can thus be both stimulated or down‐regulated by various cytokines Citation2–8,Citation20,Citation28,Citation43,44,Citation57,Citation64–69,Citation80,81,Citation84. For example, transforming growth factor (TGF)‐β can suppress the MMP‐1, ‐3 and ‐8 gene transcription but can also induce the MMP‐13 gene expression Citation2–8,Citation20,Citation28,Citation34–36,Citation65–69,Citation80,81,Citation84.

IL‐1β and TNF‐ α can up‐regulate MMP‐3, ‐8 and ‐9 gene and protein expression by gingival fibroblasts, and IL‐1β and TNF‐α exert the same effect on osteoblast MMP‐13 Citation28,Citation43,44,Citation57,Citation64,Citation65–69. On the other hand IL‐6 does not exert effects on keratinocyte MMP expression, but TGF‐1β can repress the transcription of the most MMP genes by keratinocytes but can up‐regulate MMP‐2 Citation35,36,Citation65,Citation68,69. Furthermore, IL‐8 and bacterial virulence factors have been shown to be essential to recruit and trigger extravasating neutrophil influx to the sites of periodontal inflammation and can further promote neutrophil degranulation to release large amounts of oxidatively activated presynthetized and prepacked both MMP‐8 and ‐9 Citation11–16,Citation64,Citation85,86; this can occur with or without de novo induction of MMP‐8 and ‐9 mRNA expression Citation22,23.

Recent studies have revealed exocites, i.e. MMP substrate binding, processing and cleavage sites locating outside the catalytic domain and active site of MMPs; the exocites can cleave non‐collagenous/non‐matrix MMP substrates including certain pro‐ and anti‐inflammatory cytokines and chemokines Citation2–8,Citation59,Citation64,Citation80. Exocites thus broaden the substrate and tissue targeting profile of MMPs Citation2–8,Citation59,Citation64,Citation80. The exocite processings and cleavages have been found to result usually in small‐size molecules or peptides Citation59,Citation64,Citation80, the effects of which may in fact be clearly more potent than the full‐size parent or native molecule Citation3,Citation5,Citation59. In this regard, IL‐8 expressed by periodontitis‐affected gingival sulcular epithelial cells can provoke the release of MMP‐8 and ‐9 by recruited neutrophils and is also a potential target of the released MMPs Citation59,Citation64,Citation80,Citation85,86. Thus, proteolysis of various non‐matrix matrix substrates such as cytokines, chemokines, receptors, adhesion molecules, complement components and serpins is currently receiving as much attention as the traditional roles of MMP‐8 and ‐9 in the degradation of the extracellular matrix and basement membrane during pathogenesis of both inflammatory and malignant diseases Citation2–8,Citation11,Citation28–31,Citation59,Citation64,Citation79,80,Citation85,86. Therefore, the levels and involvement of MMPs can no longer be regarded as crucial solely because of tissue destructive activity, but must also be considered crucial for the protective anti‐inflammatory immune processes Citation2–8,Citation28,Citation59,Citation64.

Matrix metalloproteinase promoter region polymorphisms associated with periodontitis and peri‐implantitis

MMP gene expression levels can be affected by the genetic variation(s), and promoter region polymorphisms can thus be involved in the development and progression of various malignant Citation87–91 and also inflammatory diseases such as periodontal disease, dental peri‐implantitis, osteoporosis, coronary heart disease and multiple sclerosis Citation92–109.

An insertion or deletion of guanine at position ‐1607 in the human MMP‐1 promoter region generates two different alleles: one with single guanine C (G) and the other with two guanines (2G). The 2G allele together with adjacent adenosine generates a core binding site, 5′‐GGA ‐3′, for Ets transcription factors located immediately adjacent to the AP‐1 site causing a 37‐fold increase in the transcriptioned activity of MMP‐1 Citation87. It has been shown that the 2G allele significantly increases the transcription of MMP‐1 relative to the 1G allele Citation87. The 2G allele displays enhanced MMP‐1 transcription in both tumour cells and normal fibroblasts, and the presence of this allele has been associated with the enhanced development of ovarian cancer and metastatic melanomas Citation87–92. The presence of the 2G allele has recently been associated with severe chronic periodontitis and early dental implant failures (peri‐implantitis) Citation93–95. However, in Japanese and Czech populations no association between the corresponding MMP‐1 polymorphism and periodontitis could be found, suggesting that the genetic variation in different races may explain only part of the various outcomes Citation96,97.

Two functional genetic polymorphisms have been detected in the MMP‐9 promoter Citation98,99, namely a SNP (single nucleotide polymorphism) at position ‐1562 and a d(CA)n microsatellite repeat at position ‐90. The SNP is a C‐to‐T substitution that increases the transcriptional activity of MMP‐9 Citation98,99. The C‐1562T polymorphism has been found to be associated with the coronary atherosclerosis Citation99. The (CA)n is a multi‐allelic microsatellite polymorphism, where the common form is the (CA)14. The (CA)14‐repeat has only 50% of the transcriptional activity of the MMP‐9 promoter when compared to the (CA)21‐repeat allele Citation89. An association between the (CA)n repeats and abdominal aortic aneurysm and intracranial aneurysm has been found, but there have also been variant results Citation100–103. No associations have been found between MMP‐9 promoter polymorphisms with the susceptibility or severity of periodontal diseases or peri‐implantitis Citation104.

A 5A/6A polymorphism has been found in the MMP‐3 (stromelysin‐1) promoter. This SNP has been found to be associated with atherosclerosis Citation105,106. The frequency of the 5A allele is clearly enhanced in the affected individuals relative to the control subjects, and the risk of acute myocardial infarction in individuals carrying one or two copies of the 5A allele has been estimated to be 2.25‐fold Citation107,108. Itagaki et al. Citation96 did not find support for the hypothesis that MMP‐1 or MMP‐3 promoter polymorphisms could influence the susceptibility to or progression of periodontitis. Holla et al. Citation109 found that polymorphisms in the MMP‐2 promoter did not contribute significantly to the susceptibility to periodontitis, and de Souza et al. Citation104 found that polymorphisms in the TIMP‐2 promoter did not allow any conclusions regarding enhanced susceptibility to or severity of periodontitis.

Taken together, there are potential reasons why MMP‐1, ‐2, ‐3 and ‐9 promoter polymorphisms do not show significant influence on the susceptibility to periodontitis or dental peri‐implantitis Citation93–98,Citation105,Citation108,109. In the case the cells carrying the genetic variations of MMPs would pathologically excessively up‐regulate their MMP expressions leading to MMP over‐production, the enhanced prevalence of systemic collagenolytic tissue destructive disorders such cancers, arthritides, skin diseases, ulcers, eye and lung diseases in addition to local periodontitis could be expected Citation2–8,Citation28,Citation64,Citation110,111. However, periodontitis patients are usually systemically rather healthy although enhanced periodontal tissue destruction involving locally up‐regulated and activated gingival tissue and GCF MMPs has been associated to certain systemic diseases such as unbalanced diabetes and cardiovascular diseases Citation112,113.

Recent MMP single gene knock‐out studies have evidenced that the affected mice do not show any particular diseases Citation114,115. Since the MMP members share the most common extracellular matrix and non‐matrix substrates, and eventually can compensate for lost functions, it can be concluded that single gene polymorphism(s) of a certain individual MMP may be not enough to exert significant effects on periodontal disease. Overall, the MMP transcription at the sites of periodontal inflammation is regulated and affected by the combined action of multiple local factors including bacteria, microbial virulence factors, enzymes and metabolites, host's adhesion and immune molecules, hormones, cytokines and growth factors etc. Citation11–13,Citation28,Citation64,65. Thus, in addition to genetic factors there are distinct and multiple local molecular factors involved in the periodontal soft and hard tissue destruction; shifting from bacterial infection into host cell behaviour, including various cytokine‐, serine proteinase‐/serpin‐, MMP‐ and TIMP‐cascades Citation28,Citation33,Citation65. Noteworthy, certain MMPs such as MMP‐8 and ‐9 have recently been found to exert, in addition to their surrogate tissue destructive properties, also unexpected protective and defensive anti‐inflammatory properties Citation28–31,Citation59,Citation64,Citation79.

Characteristics and diagnostic utilization of MMPs present in the diseased periodontal tissues, gingival crevicular fluid (GCF), and pharmacological inhibition of MMPs in periodontitis

GCF has been found to contain large amounts of serum proteins, inflammatory mediators, host tissue and cell degradation products as well as microbial metabolites and enzymes Citation7,Citation28,Citation64. Especially proteinases such as MMPs, as well as neutrophil elastase and cathepsin G, are thought to play a central role in the regulation of periodontal tissue turnover in health and disease. These enzymes in GCF or in peri‐implant sulcular fluid (PISF) are considered to reflect periodontal and dental peri‐implant health and disease Citation7,Citation28,Citation64,Citation116,117. The collection and analysis of GCF and PISF samples could provide a useful non‐invasive means to assess and monitor the pathophysiological status of the periodontium and dental peri‐implant tissues in a site‐specific manner Citation28,Citation64,Citation116,117. Thus, high expectations have been placed on GCF and PISF enzymes in the search for molecular indicators or markers to guide clinicians towards early chair‐side and point‐of‐care detection and monitoring of periodontal and dental peri‐implant health and disease Citation7,Citation28,Citation64,Citation116,117.

It has been reported that the predominant MMPs in periodontitis‐affected GCF and peri‐implantitis‐affected PISF are PMN‐derived MMP‐8 and ‐9 Citation7,Citation116–123. However, the resident cells (sulcular epithelial cells/keratinocytes, gingival and periodontal ligament fibroblasts, endothelial cells, monocyte/macrophages, plasma cells, osteoblasts/osteoclasts etc.) in the periodontium likely also (albeit to a smaller extent) contribute MMP‐8 and ‐9 during periodontitis and peri‐implantitis Citation11–14,Citation21–24,Citation26–28,Citation40–44, Citation64. Diseased GCF and PISF contain elevated levels of MMP‐3, MT1‐MMP (MMP‐14), MT6‐MMP (MMP‐25), MMP‐7, and MMP‐26 in active forms relative to periodontally healthy GCF and PISF Citation21,Citation39–44,Citation54,Citation120–122; whereas MMP‐1 and ‐2 have been detected only in low levels in diseased GCF and PISF Citation7,Citation28,Citation64,65. The pathologically elevated and activated MMPs can form and act in co‐operative cascades in the periodontitis‐affected gingival tissue and GCF Citation28,Citation42–44. Furthermore, markedly enhanced expression of MMP‐13 (collagenase‐3) has been demonstrated in the gingival tissue sections from chronic periodontitis patients suggesting MMP‐13 expression is important in the proliferation and intrusion of the activated epithelium into the periodontal connective tissues and in the progression of attachment loss and deepening of periodontal pockets Citation21,Citation27,28,Citation64.

Thus, the predominant MMPs present in the inflamed gingival tissue, gingival crevicular fluid (GCF), saliva/mouth‐rinse‐samples as well as dental peri‐implant sulcular fluid (PISF) are PMN‐derived inductive MMP‐8 and ‐9 and epithelial or bone cell‐derived MMP‐13, rather than constitutively expressed MMP‐1 and MMP‐2 Citation7,Citation27,28,Citation64,65,Citation116–123. Levels and degree of activation of PMN‐derived MMP‐8 and ‐9 and the bone‐ or epithelial cell‐derived MMP‐13 have been shown to increase with increasing periodontal disease severity and activity and to decrease following periodontal treatment (scaling and root planing) Citation7,Citation28,Citation64,Citation116–123. Disturbances between MMP and TIMP ratios have been implicated in the ethiopathogenesis of periodontitis Citation7,Citation28,Citation44,Citation64. Taken together, the breakdown of gingival collagen fibres in periodontal soft and hard tissues (alveolar bone resorption) by the cooperative action and cascade of both MMPs and cytokines contributes the detectable clinical signs and outcome of periodontitis and peri‐implantitis including enhanced pocket formation, attachment loss, bone resorption, gingival recessions, increased tooth/dental implant mobility and finally tooth/dental implant loss Citation7,Citation28,Citation64. In this regard a systemic pharmaceutical capable of down‐regulating the pathologically elevated levels of MMPs could be very useful as an adjunctive treatment/medication in periodontitis or dental peri‐implantitis Citation7,Citation120,Citation123,124.

A number of MMP inhibitors have been synthesized, but the major drawbacks of these molecules are the timing of the medication and adverse side effects Citation125. The fact that only a limited number of compounds have reached advanced clinical trials in spite of extensive efforts by almost all major pharmaceutical companies indicates that the development of MMP inhibitors is very demanding and challenging Citation125,126. In this regard, the ideal selectivity profile of MMP inhibitors and the target MMPs for various malignant and inflammatory disease states are unclear Citation28,Citation59,Citation64,Citation123,Citation125,126. Musculoskeletal syndrome has been reported among the most common side effect in clinical studies using MMP inhibitors, and events such as joint pain, stiffness, oedema, skin discoloration, and reduced skin mobility have been observed Citation59,Citation125. In arthritis and periodontitis the excessive inhibition of MMPs surprisingly exacerbates rather than alleviates the disease Citation59,Citation125,Citation127. In this regard, Bjornsson et al. Citation127 recently described the effects of a potent and efficient broad‐spectrum hydroxamic peptide‐based MMP‐inhibitor, Batimastat, on periodontal destruction in a rat model. Surprisingly, instead of reducing the severity of experimental periodontitis, Batimastat actually enhanced the periodontal tissue destruction Citation127. What has not been sufficiently appreciated is that a minimal, but not pathologically excessive, level of MMPs seems to exert protective and anti‐inflammatory functions Citation29–31,Citation79. Thus, we have proposed that ‘leaky’, less efficient MMP‐inhibitors, such as the tetracycline‐based MMP‐inhibitors, may be safe and effective because they only reduce the pathologically excessive MMPs but do not reduce MMP levels/activities below those required for physiological or anti‐inflammatory functions Citation7,Citation123,124,Citation128, whereas the potent and efficient synthetic MMP‐inhibitors, such as Batimastat, excessively reduce these MMPs below the physiologic or protective basal levels eventually resulting in clinically significant and harmful side effects Citation59,Citation125,Citation127. Some effective and selective in vitro MMP inhibitors, such as cyclic peptides produced in phage display libraries, are still in preclinical testing Citation126. Tetracyclines, especially doxycycline, which, in addition to their antimicrobial activity, can act as MMP inhibitors, an effect which is independent of their antimicrobial activity Citation7,Citation120,Citation123,124. Doxycycline inhibits MMPs at many levels: 1) inhibits the catalytic activity, 2) suppresses MMP gene expression, 3) prevents the proteolytic and oxidative activation of MMPs and 4) prevents the oxidative and MMP‐dependent inactivation of serpins such as α1‐antitrypsin Citation7,Citation15,Citation120,Citation123,124. Presently Periostat® (doxycycline hyclate, CollaGenex Pharmaceuticals Inc.) is the only collagenase inhibitor approved by the Food and Drug Administration (FDA) and is clinically employed for the adjunctive treatment of periodontitis Citation123,124,Citation129. This ‘low‐dose’ doxycycline formulation, used at 20 mg b.i.d., has been shown to have lost its antibiotic/anti‐bacterial activity Citation7,Citation123,124,Citation129. A low‐dose or sub‐antimicrobial dose of doxycycline medication (LDD or SDD) does not promote musculoskeletal syndrome side effects, is capable of inhibiting the pathologically elevated GCF and gingival tissue collagenase activity without causing their complete inhibition, and does not generate the development of microbial resistance to antibiotics Citation7,Citation123,124,Citation128,129.

Studies have demonstrated that low‐dose doxycycline (LDD) used as a systemic adjunctive medication to scaling and root planing (SRP) for periodontitis patients significantly improves the clinical outcome and attachment and reduces pocket depth, slowing the progression of periodontitis when compared with SRP alone Citation7,Citation123,124,Citation129. Furthermore, extensive literature has shown that systemic diseases such as diabetes and cardiovascular diseases share links between biomarkers of systemic inflammation (C‐reactive protein [CRP] IL‐6, TNF‐α and MMP‐9) and periodontitis Citation112,113,Citation130,131. Severe periodontitis patients have significantly elevated serum CRP levels relative to patients with moderate or mild periodontitis Citation130,131. Moreover, Golub et al. Citation132 have proposed a ‘two‐hit’ model of periodontitis wherein the second ‘hit’, systemic inflammation, co‐induces periodontal tissue destruction in concert with the first ‘hit’, the microbial products such as LPS. Cardiology research groups have repeatedly in large‐scale clinical studies demonstrated that plasma CRP and other biomarkers (i.e., IL‐6, TNF‐α and MMP‐9) can be utilized as predictors of future cardiovascular diseases including heart attacks Citation133,134. In this regard, our group has reported that low‐dose doxycycline medication, but not placebo, can significantly (but not completely) Citation128 reduces the plasma levels of these systemic pro‐inflammatory biomarkers including also MMPs in patients with acute coronary syndromes (ACS) Citation135,136 (). A prospective, randomized double‐blind pilot study of 6‐month of LDD—or placebo—treatment was performed to reduce systemic inflammation and prevent conditions promoting plaque rupture Citation135,136. LDD medication was shown to cause greater reductive effect on plasma CRP values (but no effect of placebo) of ACS patients when baseline plasma CRP values are ⩾5 µg/mL (high‐sensitivity CRP), and little or no effect of LDD medication was observed when baseline CRP values are ⩽5 µg/mL (). Furthermore, Western immunoblotting and zymographic analysis Citation22,23,Citation70,Citation135,136 of plasma samples revealed that LDD medication decreased MMP‐8 and ‐9 levels by 30% and 50%, respectively, after 6‐month therapy, whereas placebo (6 months) had no statistically significant effect (). In addition, the major species of MMP‐8 present in human plasma is the PMN‐type MMP‐8 isoform (15, Sorsa T, Golub LM, Brown DL, Konttinen YT, et al., unpublished data).

Figure 1. Effects of 6‐month regimen of low‐dose doxycycline (LDD; B = before, A = after) or placebo on plasma levels of pro‐inflammatory biomarkers in patients (n = 30) with acute coronary syndromes (ACS). (a): MMP‐8; (b): MMP‐9; (c): C‐reactive protein (CRP). MMP‐8 and ‐9 values represent densitometric scanning units and CRP represent µg/mL, and are expressed as means + SDs. (MMP = matrix metalloproteinases.)

Figure 1. Effects of 6‐month regimen of low‐dose doxycycline (LDD; B = before, A = after) or placebo on plasma levels of pro‐inflammatory biomarkers in patients (n = 30) with acute coronary syndromes (ACS). (a): MMP‐8; (b): MMP‐9; (c): C‐reactive protein (CRP). MMP‐8 and ‐9 values represent densitometric scanning units and CRP represent µg/mL, and are expressed as means + SDs. (MMP = matrix metalloproteinases.)

A major issue, when considering the use of low‐dose doxycycline (LDD) MMP‐inhibitory medication therapy, is the duration of the therapy and detection or monitoring the link between the clinical and biological outcomes Citation123,124,Citation129. The classical periodontal diagnosis determines mainly the previous periodontal tissue destruction, i.e. the disease outcome rather than disease activity Citation137. Evaluation of periodontal disease activity before significant destruction, and measurement of successful treatment or disease arrest, would allow the treatment to be directed to the right patient, possibly even to the site of high disease activity, at the right moment Citation116,117,Citation137. There is a need for a convenient chair‐side, point‐of‐care test for diagnosis and monitoring of periodontal diseases Citation28,Citation64,Citation137. MMP‐8 in GCF is a potential candidate for such a test Citation27,28,Citation44,Citation64,Citation116–120. We have recently developed monoclonal antibodies for MMP‐8 Citation15 to be utilized in a chair‐side dip‐stick test for MMP‐8 that allowed the development of a novel sensitive, specific, rapid and practical immunological chair‐side point‐of‐care dip‐stick test for MMP‐8 in GCF and peri‐implant sulcular fluid (PISF) Citation15,Citation116,117,Citation120–123,Citation138. This test resembles pregnancy home test kits Citation116,117,Citation138, can conveniently be performed by a dentist without specific equipment, and measures the GCF MMP‐8 levels in 5 minutes Citation138 (). Clinicians prefer such rapid (<10 min from start to finish) chair‐side point‐of‐care tests Citation28,Citation64,Citation116,117,Citation137,138. The test can differentiate healthy and gingivitis sites from periodontitis sites Citation138. The test stick results () were found to be in good agreement with the quantitative MMP‐8 immunofluorometric assay (IFMA) analysis from GCF collected from periodontally healthy individuals and from gingivitis and periodontitis patients when all positive test results (+ to +++) were analysed (). The agreement was very good when the Kappa‐statistics (κ = 0 0.81) were calculated, and the chair‐side GCF MMP‐8 test provided a sensitivity of 0.83 and specificity of 0.96 Citation138. The elevated test‐stick (++ to +++) results and MMP‐8 IFMA analysis from GCF during the maintenance phase of periodontal treatment (SRP) preceded the clinical attachment loss (). The dip‐stick technology () allows the use of one or more MMPs, TIMPs and tissue degradation products Citation28,Citation64,Citation116–123,Citation138 in a combination test. GCF MMP‐8 level testing is a useful tool to monitor the beneficial effects of adjunctive sub‐antimicrobial doxycycline medication for periodontitis patients Citation139–141. This rapid point‐of‐care test developed for periodontitis is a useful tool also for monitoring of peri‐implantitis Citation116,117,Citation121,122. It can be adjusted to monitor the follow‐up of orthodontic tooth movement Citation142,143. Evidently the principle of this chair‐side point‐of‐care MMP‐8 test could be adapted for the arthritic, eye and lung diseases Citation22,23,Citation144–149. In addition to GCF Citation138–141, the effect of the MMP inhibitory medication can be followed by utilizing both MMP activity assays and immunoassays from salivary, oral rinse, tear fluid and serum/plasma samples Citation135,136,Citation138,Citation144–149 ().

Figure 2. Test positive result of the MMP‐8 specific chair‐side dip‐stick is confirmed with the appearance of the blue line in the catching zone (left, indicated by arrows). (MMP = matrix metalloproteinases.)

Figure 2. Test positive result of the MMP‐8 specific chair‐side dip‐stick is confirmed with the appearance of the blue line in the catching zone (left, indicated by arrows). (MMP = matrix metalloproteinases.)

Figure 3. Scattergram of periodontitis patients' sites (n = 133) at baseline; correlation of MMP‐8 concentration (assessed by IFMA, µg/L) and MMP‐8 specific chair‐side dipstick test results (0 = negative; 1 = +; 2 = ++; 3 = +++). (IFMA = time‐resolved immunofluorometric assay; MMP = matrix metalloproteinases.)

Figure 3. Scattergram of periodontitis patients' sites (n = 133) at baseline; correlation of MMP‐8 concentration (assessed by IFMA, µg/L) and MMP‐8 specific chair‐side dipstick test results (0 = negative; 1 = +; 2 = ++; 3 = +++). (IFMA = time‐resolved immunofluorometric assay; MMP = matrix metalloproteinases.)

Figure 4. MMP‐8 concentration in GCF (µg/L, bars) collected from 5–8 distinct sites from periodontitis patients (n = 8) and assessed by IFMA and MMP‐8 specific chair‐side dipstick test (−; +; ++ and +++) precede loss of attachment (AL, mm) during maintenance phase of periodontal treatment (scaling and root planing). MMP‐8 concentrations and AL are represented as means. (GCF = gingival crevicular fluid; IFMA = time‐resolved immunofluorometric assay; MMP = matrix metalloproteinases.)

Figure 4. MMP‐8 concentration in GCF (µg/L, bars) collected from 5–8 distinct sites from periodontitis patients (n = 8) and assessed by IFMA and MMP‐8 specific chair‐side dipstick test (−; +; ++ and +++) precede loss of attachment (AL, mm) during maintenance phase of periodontal treatment (scaling and root planing). MMP‐8 concentrations and AL are represented as means. (GCF = gingival crevicular fluid; IFMA = time‐resolved immunofluorometric assay; MMP = matrix metalloproteinases.)

Conclusions

Overall, the excessive levels of PMN‐derived MMP‐8 and ‐9 are more associated with the progression, course and treatment of periodontitis and can contribute to the disease ethiopathogenesis in different ways, through cleavage(s) of fibrillar collagens, basement membrane and other matrix macromolecules, inactivation of proteinase inhibitors (α1‐antitrypsin and α2‐macroglobulin) and non‐matrix substrates including chemokines, such as CXC chemokine, a neutrophil chemoattract Citation3,Citation5,Citation28,Citation59,Citation64. However, lower levels of these MMPs can exert anti‐inflammatory protection by regulating the inflammatory cell and apoptotic responses Citation30,31,Citation79. In this respect, recently unexpected inhibitory effects of MMP‐8 in experimental skin cancer progression have been reported Citation29. MMP‐8 gene deletion in mice increased tumour development after carcinogen treatment, and genetic manipulation of metastasic cells to overproduce MMP‐8 resulted in decreased metastasis dissemination Citation29,Citation150. In summary, these observations suggest that MMPs associated with periodontal inflammation seem to act in cascades Citation28,Citation43,44,Citation64 and to exert multiple functions regulating both the surrogate tissue destruction and protective immune functions Citation28–31,Citation64,Citation79,Citation151, and in oral fluids such as GCF and PISF these MMPs can be utilized as diagnostic tools Citation116,117,Citation138. Synthetic MMP‐inhibitors such as doxycycline and CMT‐3 (6‐demethyl‐6‐deoxy‐4‐dedimethyl‐amino‐tetracycline—which has been chemically stripped—free of this antibiotic activity) can be used to support both the periodontal treatment (scaling and root planing) Citation7,Citation120,Citation123,124,Citation129,Citation139–141 and reduce systemic inflammation Citation135,136.

Acknowledgements

The research in our laboratories has been supported grants from the Academy of Finland and the HUCH‐EVO (TYH 5306, TYH 6104 and TI 020Y0002) Foundation.

References

  • Gross J., Lapiere C. M. Collagenolytic activity in amphibian tissues: a tissue culture assay. Proc Natl Acad Sci 1962; 48: 1014–22
  • Visse R., Nagase H. Matrix metalloproteinases and tissue inhibitors of metalloproteinases: structure, function, and biochemistry. Circ Res 2003; 92: 827–39
  • Egeblad M., Werb Z. New functions for the matrix metalloproteinases in cancer progression. Nat Rev Cancer 2002; b: 161–74
  • Murphy G., Knäuper V. Relating matrix metalloproteinase structure to function: why the ‘hemopexin’ domain?. Matrix Biol 1997; 15: 511–8
  • McCawley L. J., Matrisian L. M. Matrix metalloproteinases: they're not just for matrix anymore!. Curr Opin Cell Biol 2001; 13: 534–40
  • Ala‐aho R., Kähäri V. ‐M. Collagenases in cancer. Biochimie 2005; 87: 273–86
  • Golub L. M., Wolff M., Roberts S., Lee H. M., Leung M., Payonk G. S. Treating periodontal diseases by blocking tissue‐destructive enzymes. J Am Dent Assoc 1994; 125: 163–9, discussion 169–71
  • Björklund M., Koivunen E. Gelatinase‐mediated migration and invasion of cancer cells. Biochim Biophys Acta 2005; 25: 37–69
  • Itoh Y., Kajita M., Kinoh H., Mori H., Okada A., Seiki M. Membrane type 4 matrix metallloproteinase (MT4‐MMP, MMP‐17) is a glycosyl‐phosphatidyl inositol‐anchored proteinase. J Biol Chem 1999; 274: 34260–6
  • Kojima S., Itoh Y., Matsumoto S., Masuho Y., Seiki M. Membrane‐type 6 matrix metalloproteinase is the second glycosyl‐phosphatidyl inositol‐anchored MMP. FEBS Lett 2000; 480: 142–6
  • Weiss S. J. Tissue destruction by neutrophils. N Eng J Med 1989; 320: 365–76
  • Ding Y., Uitto V. J., Haapasalo M., Lounatmaa K., Konttinen Y. T., Salo T., et al. Membrane components of Treponema denticola trigger proteinase release from human polymorphonuclear leukocytes. J Dent Res 1996; 75: 1986–93
  • Ding Y., Haapasalo M., Kerosuo E., Lounatmaa K., Kotiranta A., Sorsa T. Release and activation of human neutrophil matrix metallo‐ and serine proteinases during phagocytosis of Fusobacterium nucleatum, Porphyromonas gingivalis and Treponema denticola. J Clin Periodontol 1997; 24: 237–48
  • Weiss S. J., Peppin G., Ortiz X., Ragsdale C., Test S. T. Oxidative autoactivation of latent collagenase by human neutrophils. Science 1985; 227: 747–9
  • Hanemaaijer R., Sorsa T., Konttinen Y. T., Ding Y., Sutinen M., Visser H., et al. Matrix metalloproteinase‐8 is expressed in rheumatoid synovial fibroblasts and endothelial cells. Regulation by tumor necrosis factor‐alpha and doxycycline. J Biol Chem 1997; 272: 31504–9
  • Saari H., Suomalainen K., Lindy O., Konttinen Y. T., Sorsa T. Activation of latent human neutrophil collagenase by reactive oxygen species and serine proteases. Biochem Biophys Res Commun 1990; 171: 979–87
  • Cole A. A., Kuettner K. E. MMP‐8 (neutrophil collagenase) mRNA and aggrecanase cleavage products are present in normal and osteoarthritic human articular cartilage. Acta Orthop Scand 1995; 98–102
  • Cole A. A., Chubinskaya S., Schumacher B., Huch K., Szabo G., Yao J., et al. Chondrocyte matrix metalloproteinase‐8. Human articular chondrocytes express neutrophil collagenase. J Biol Chem 1996; 271: 11023–6
  • Chubinskaya S., Huch K., Mikecz K., Cs‐Szabo G., Hasty K. A., Kuettner K. E., et al. Chondrocyte matrix metalloproteinase‐8: up‐regulation of neutrophil collagenase by interleukin‐1 beta in human cartilage from knee and ankle joints. Lab Invest 1996; 74: 232–40
  • Palosaari H., Wahlgren J., Larmas M., Ronka H., Sorsa T., Salo T., et al. The expression of MMP‐8 in human odontoblasts and dental pulp cells is down‐regulated by TGF‐beta1. J Dent Res 2000; 79: 77–84
  • Tervahartiala T., Pirilä E., Ceponis A., Maisi P., Salo T., Tuter G., et al. The in vivo expression of the collagenolytic matrix metalloproteinases (MMP‐2, ‐8, ‐13, and ‐14) and matrilysin (MMP‐7) in adult and localized juvenile periodontitis. J Dent Res 2000; 79: 1969–77
  • Prikk K., Maisi P., Pirilä E., Reintam M. A., Salo T., Sorsa T., et al. Airway obstruction correlates with collagenase‐2 (MMP‐8) expression and activation in bronchial asthma. Lab Invest 2002; 82: 1535–45
  • Prikk K., Maisi P., Pirila E., Sepper R., Salo T., Wahlgren J., et al. In vivo collagenase‐2 (MMP‐8) expression by human bronchial epithelial cells and monocytes/macrophages in bronchiectasis. J Pathol 2001; 194: 232–8
  • Wahlgren J., Maisi P., Sorsa T., Sutinen M., Tervahartiala T., Pirila E., et al. Expression and induction of collagenases (MMP‐8 and ‐13) in plasma cells associated with bone‐destructive lesions. J Pathol 2001; 194: 217–24
  • Moilanen M., Pirilä E., Grenman R., Sorsa T., Salo T. Expression and regulation of collagenase‐2 (MMP‐8) in head and neck squamous cell carcinomas. J Pathol 2002; 197: 72–81
  • Bachmeier B. E., Nerlich A. G., Boukamp P., Lichtinghagen R., Tschesche H., Fritz H., et al. Human keratinocyte cell lines differ in the expression of the collagenolytic matrix metalloproteinases‐1,‐8, and ‐13 and of TIMP‐1. Biol Chem 2000; 38: 509–16
  • Kiili M., Cox S. W., Chen H. Y., Wahlgren J., Maisi P., Eley B. M., et al. Collagenase‐2 (MMP‐8) and collagenase‐3 (MMP‐13) in adult periodontitis: molecular forms and levels in gingival crevicular fluid and immunolocalisation in gingival tissue. J Clin Periodontol 2002; 29: 224–32
  • Sorsa T., Tjäderhane L., Salo T. Matrix metalloproteinases (MMPs) in oral diseases. Oral Dis 2004; 10: 311–8
  • Balbin M., Fueyo A., Tester A. M., Pendas A. M., Pitiot A. S., Astudillo A., et al. Loss of collagenase‐2 confers increased skin tumor susceptibility to male mice. Nat Genet 2003; 35: 252–7
  • Owen C. A., Hu Z., Lopez‐Otin C., Shapiro S. D. Membrane‐bound matrix metalloproteinase‐8 on activated polymorphonuclear cells is a potent, tissue inhibitor of metalloproteinase‐resistant collagenase and serpinase. J Immunol 2004; 172: 7791–803
  • Gueders M. M., Balbin M., Rocks N., Foidart J. M., Gosset P., Louis R., et al. Matrix metalloproteinase‐8 deficiency promotes granulocytic allergen‐induced airway inflammation. J Immunol 2005; 175: 2589–97
  • Freije J. M., Diez‐Itza I., Balbin M., Sanchez L. M., Blasco R., Tolivia J., et al. Molecular cloning and expression of collagenase‐3, a novel human matrix metalloproteinase produced by breast carcinomas. J Biol Chem 1994; 269: 16766–73
  • Knäuper V., Lopez‐Otin C., Smith B., Knight G., Murphy G. Biochemical characterization of human collagenase‐3. J Biol Chem 1996; 271: 1544–50
  • Johansson N., Kähäri V. M. Matrix metalloproteinases in squamous cell carcinoma. Histol Histopathol 2000; 15: 225–37
  • Kähäri V. M., Saarialho‐Kere U. Matrix metalloproteinases and their inhibitors in tumour growth and invasion. Ann Med 1999; 31: 34–45
  • Konttinen Y. T., Salo T., Hanemaaijer R., Valleala H., Sorsa T., Sutinen M., et al. Collagenase‐3 (MMP‐13) and its activators in rheumatoid arthritis: localization in the pannus‐hard tissue junction and inhibition by alendronate. Matrix Biol 1999; 18: 401–12
  • Lindy O., Konttinen Y. T., Sorsa T., Ding Y., Santavirta S., Ceponis A., et al. Matrix metalloproteinase 13 (collagenase 3) in human rheumatoid synovium. Arthritis Rheum 1997; 40: 1391–9
  • Ylipalosaari M., Thomas G. J., Nystrom M., Salhimi S., Marshall J. F., Huotari V., et al. Alpha v beta 6 integrin down‐regulates the MMP‐13 expression in oral squamous cell carcinoma cells. Exp Cell Res 2005; 309: 273–83
  • Westerlund U., Ingman T., Lukinmaa P. L., Salo T., Kjeldsen L., Borregaard N., et al. Human neutrophil gelatinase and associated lipocalin in adult and localized juvenile periodontitis. J Dent Res 1996; 75: 1553–63
  • Ma J., Kitti U., Hanemaaijer R., Teronen O. P., Sorsa T. A., Natah S., et al. Gelatinase B is associated with peri‐implant bone loss. Clin Oral Implants Res 2003; 14: 709–13
  • Makela M., Salo T., Uitto V. J., Larjava H. Matrix metalloproteinases (MMP‐2 and MMP‐9) of the oral cavity: cellular origin and relationship to periodontal status. J Dent Res 1994; 73: 1397–406
  • Beklen A., Laine M., Venta I., Hyrkäs T., Konttinen Y. T. Role of TNF‐{alpha} and its receptors in pericoronitis. J Dent Res 2005; 84: 1178–82
  • Beklen A., Tuter G., Sorsa T., Hanemaaijer R., Virtanen I., Tervahartiala T., et al. Gingival tissue and crevicular fluid co‐operation in adult periodontitis. J Dent Res 2006; 85: 59–63
  • Ingman T., Tervahartiala T., Ding Y., Tschesche H., Haerian A., Kinane D. F., et al. Matrix metalloproteinases and their inhibitors in gingival crevicular fluid and saliva of periodontitis patients. J Clin Periodontol 1996; 23: 1127–32
  • Seiki M. Membrane‐type matrix metalloproteinases. APMIS 1999; 107: 137–43
  • Knauper V., Will H., Lopez‐Otin C., Smith B., Atkinson S. J., Stanton H., et al. Cellular mechanisms for human procollagenase‐3 (MMP‐13) activation. Evidence that MT1‐MMP (MMP‐14) and gelatinase a (MMP‐2) are able to generate active enzyme. J Biol Chem 1996; 271: 17124–31
  • Holopainen J. M., Moilanen J. A., Sorsa T., Kivela‐Rajamaki M., Tervahartiala T., Vesaluoma M. H., et al. Activation of matrix metalloproteinase‐8 by membrane type 1‐MMP and their expression in human tears after photorefractive keratectomy. Invest Ophthalmol Vis Sci 2003; 44: 2550–6
  • Sekine‐Aizawa Y., Hama E., Watanabe K., Tsubuki S., Kanai‐Azuma M., Kanai Y., et al. Matrix metalloproteinase (MMP) system in brain: identification and characterization of brain‐specific MMP highly expressed in cerebellum. Eur J Neurosci 2001; 13: 935–48
  • Matsuda A., Itoh Y., Koshikawa N., Akizawa T., Yana I., Seiki M. Clusterin, an abundant serum factor, is a possible negative regulator of MT6‐MMP/MMP‐25 produced by neutrophils. J Biol Chem 2003; 278: 36350–7
  • Drew A. F., Blick T. J., Lafleur M. A., Tim E. L., Robbie M. J., Rice G. E., et al. Correlation of tumor‐ and stromal‐derived MT1‐MMP expression with progression of human ovarian tumors in SCID mice. Gynecol Oncol 2004; 95: 437–48
  • Toft‐Hansen H., Nuttall R. K., Edwards D. R., Owens T. Key metalloproteinases are expressed by specific cell types in experimental autoimmune encephalomyelitis. J Immunol 2004; 173: 5209–18
  • Nie J., Pei D. Rapid inactivation of alpha‐1‐proteinase inhibitor by neutrophil specific leukolysin/membrane‐type matrix metalloproteinase 6. Exp Cell Res 2004; 296: 145–50
  • Nie J., Pei D. Direct activation of pro‐matrix metalloproteinase‐2 by leukolysin/membrane‐type 6 matrix metalloproteinase/matrix metalloproteinase 25 at the asn(109)‐Tyr bond. Cancer Res 2003; 63: 6758–62
  • Emingil G., Kuula H., Sorsa T., Atilla G. Gingival crevicular fluid matrix metalloproteinase‐25 and ‐26 levels in periodontal disease. J Periodontol 2006; 77: 664–71
  • Kazes I., Delarue F., Hagege J., Bouzhir‐Sima L., Rondeau E., Sraer J. D., et al. Soluble latent membrane‐type 1 matrix metalloprotease secreted by human mesangial cells is activated by urokinase. Kidney Int 1998; 54: 1976–84
  • Li H., Bauzon D. E., Xu X., Tschesche H., Cao J., Sang Q. A. Immunological characterization of cell‐surface and soluble forms of membrane type 1 matrix metalloproteinase in human breast cancer cells and in fibroblasts. Mol Carcinog 1998; 22: 84–94
  • Cox S. W., Eley B. M., Kiili M., Asikainen A., Tervahartiala T., Sorsa T. Collagen degradation by interleukin‐1β‐stimulated gingival fibroblasts is accompanied by release and activation of multiple matrix metalloproteinases and cysteine proteinases. Oral Dis 2006; 12: 34–40
  • Maisi P., Prikk K., Sepper R., Pirilä E., Salo T., Hietanen J., et al. Soluble membrane‐type 1 matrix metalloproteinase (MT1‐MMP) and gelatinase A (MMP‐2) in induced sputum and bronchoalveolar lavage fluid of human bronchial asthma and bronchiectasis. APMIS 2002; 110: 771–82
  • Folgueras A. R., Pendas A. M., Sanchez L. M., Lopez‐Otin C. Matrix metalloproteinases in cancer: from new functions to improved inhibition strategies. Int J Dev Biol 2004; 48: 411–24
  • Baker A. H., Zaltsman A. B., George S. J., Newby A. C. Divergent effects of tissue inhibitor of metalloproteinase‐1, ‐2, or ‐3 overexpression on rat vascular smooth muscle cell invasion, proliferation, and death in vitro. TIMP‐3 promotes apoptosis. J Clin Invest 1998; 101: 1478–87
  • Woessner J. F., Jr. Matrix metalloproteinases and their inhibitors in connective tissue remodeling. FASEB J 1991; 5: 2145–54
  • Shibutani T., Yamashita K., Aoki T., Iwayama Y., Nishikawa T., Hayakawa T. Tissue inhibitors of metalloproteinases (TIMP‐1 and TIMP‐2) stimulate osteoclastic bone resorption. J Bone Miner Metab 1999; 17: 245–51
  • Sobue T., Hakeda Y., Kobayashi Y., Hayakawa H., Yamashita K., Aoki T., et al. Tissue inhibitor of metalloproteinases 1 and 2 directly stimulate the bone‐resorbing activity of isolated mature osteoclasts. J Bone Miner Res 2001; 16: 2205–14
  • Uitto V. J., Overall C. M., McCulloch C. Proteolytic host cell enzymes in gingival crevice fluid. Periodontol 2000 2003; 31: 77–104
  • Birkedal‐Hansen H. Role of matrix metalloproteinases in human periodontal diseases. J Periodontol 1993; 64: 474–84
  • Julovi S. M., Yasuda T., Shimizu M., Hiramitsu T., Nakamura T. Inhibition of interleukin‐1beta‐stimulated production of matrix metalloproteinases by hyaluronan via CD44 in human articular cartilage. Arthritis Rheum 2004; 50: 516–25
  • Liacini A., Sylvester J., Li W. Q., Huang W., Dehnade F., Ahmad M., et al. Induction of matrix metalloproteinase‐13 gene expression by TNF‐alpha is mediated by MAP kinases, AP‐1, and NF‐kappaB transcription factors in articular chondrocytes. Exp Cell Res 2003; 288: 208–17
  • Damiens C., Fortun Y., Charrier C., Heymann D., Padrines M. Modulation by soluble factors of gelatinase activities released by osteoblastic cells. Cytokine 2000; 12: 1727–31
  • Johnson L. L., Dyer R., Hupe D. J. Matrix metalloproteinases. Curr Opin Chem Biol 1998; 2: 466–71
  • Sorsa T., Salo T., Koivunen E., Tyynela J., Konttinen Y. T., Bergmann U., et al. Activation of type IV procollagenases by human tumor‐associated trypsin‐2. J Biol Chem 1997; 272: 21067–74
  • Moilanen M., Sorsa T., Stenman M., Nyberg P., Lindy O., Vesterinen J., et al. Tumor‐associated trypsinogen‐2 (trypsinogen‐2) activates procollagenases (MMP‐1, ‐8, ‐13) and stromelysin‐1 (MMP‐3) and degrades type I collagen. Biochemistry 2003; 42: 5414–20
  • Stenman M., Ainola M., Valmu L., Bjartell A., Ma G., Stenman U. H., et al. Trypsin‐2 degrades human type II collagen and is expressed and activated in mesenchymally transformed rheumatoid arthritis synovitis tissue. Am J Pathol 2005; 167: 1119–24
  • Sorsa T., Ingman T., Suomalainen K., Haapasalo M., Konttinen Y. T., Lindy O., et al. Identification of proteases from periodontopathogenic bacteria as activators of latent human neutrophil and fibroblast‐type interstitial collagenases. Infect Immun 1992; 60: 4491–5
  • Okamoto T., Akuta T., Tamura F., van Der Vliet A., Akaike T. Molecular mechanism for activation and regulation of matrix metalloproteinases during bacterial infections and respiratory inflammation. Biol Chem 2004; 385: 997–1006
  • Potempa J., Banbula A., Travis J. Role of bacterial proteinases in matrix destruction and modulation of host responses. Periodontol 2000 2000; 24: 153–92
  • Bannikov G. A., Karelina T. V., Collier I. E., Marmer B. L., Goldberg G. I. Substrate binding of gelatinase B induces its enzymatic activity in the presence of intact propeptide. J Biol Chem 2002; 277: 16022–7
  • Fedarko N. S., Jain A., Karadag A., Fisher L. W. Three small integrin binding ligand N‐linked glycoproteins (SIBLINGs) bind and activate specific matrix metalloproteinases. FASEB J 2004; 18: 734–6
  • Sorsa T., Ding Y. L., Ingman T., Salo T., Westerlund U., Haapasalo M., et al. Cellular source, activation and inhibition of dental plaque collagenase. J Clin Periodontol 1995; 22: 709–17
  • McMillan S. J., Kearley J., Campbell J. D., Zhu X. W., Larbi K. Y., Shipley J. M., et al. Matrix metalloproteinase‐9 deficiency results in enhanced allergen‐induced airway inflammation. J Immunol 2004; 172: 2586–94
  • Sternlicht M. D., Werb Z. How matrix metalloproteinases regulate cell behavior. Annu Rev Cell Dev Biol 2001; 17: 463–516
  • Fini M. E., Cook J. R., Mohan R., Brinckerhoff C. E. Regulation of matrix metalloproteinase gene expression. Matrix metalloproteinases, W. C Parks, R. P Mecham. Academic Press, California 1998; 299–356
  • Shrivastava A., Radziejewski C., Campbell E., Zhu X. W., Larbi K. Y., Shipley J. M., et al. An orphan receptor tyrosine kinase family whose members serve as nonintegrin collagen receptors. Mol Cell 1997; 1: 25–34
  • Vogel W., Gish G. D., Alves F., Pawson T. The discoidin domain receptor tyrosine kinases are activated by collagen. Mol Cell 1997; 1: 13–23
  • Uria J. A., Jimenez M. G., Balbin M., Freije J. M., Lopez‐Otin C. Differential effects of transforming growth factor‐beta on the expression of collagenase‐1 and collagenase‐3 in human fibroblasts. J Biol Chem 1998; 273: 9769–77
  • Opdenakker G., Van den Steen P. E., Dubois B., Nelissen I., Van Coillie E., Masure S., et al. Gelatinase B functions as regulator and effector in leukocyte biology. J Leukoc Biol 2001; 69: 851–9
  • Peppin G. J., Weiss S. J. Activation of the endogenous metalloproteinase, gelatinase, by triggered human neutrophils. Proc Natl Acad Sci U S A 1986; 83: 4322–6
  • Rutter J. L., Mitchell T. I., Buttice G., Meyers J., Gusella J. F., Ozelius L. J., et al. A single nucleotide polymorphism in the matrix metalloproteinase‐1 promoter creates an Ets binding site and augments transcription. Cancer Res 1998; 58: 5321–5
  • Noll W. W., Belloni D. R., Rutter J. L., Storm C. A., Schned A. R., Titus‐Ernstoff L., et al. Loss of heterozygosity on chromosome 11q22‐23 in melanoma is associated with retention of the insertion polymorphism in the matrix metalloproteinase‐1 promoter. Am J Pathol 2001; 158: 691–7
  • Shimajiri S., Arima N., Tanimoto A., Murata Y., Hamada T., Wang K. Y., et al. Shortened microsatellite d(CA)21 sequence down‐regulates promoter activity of matrix metalloproteinase 9 gene. FEBS Lett 1999; 455: 70–4
  • Przybylowska K., Zielinska J., Zadrozny M., Krawczyk T., Kulig A., Wozniak P., et al. An association between the matrix metalloproteinase 1 promoter gene polymorphism and lymph node metastasis in breast cancer. J Exp Clin Cancer Res 2004; 23: 121–5
  • Kanamori Y., Matsushima M., Minaguchi T., Kobayashi K., Sagae S., Kudo R., et al. Correlation between expression of the matrix metalloproteinase‐1 gene in ovarian cancers and an insertion/deletion polymorphism in its promoter region. Cancer Res 1999; 59: 4225–7
  • Thiry‐Blaise L. M., Taquet A. N., Reginster J. Y., Nusgens B., Franchimont P., Lapiere C. M. Investigation of the relationship between osteoporosis and the collagenase gene by means of polymorphism of the 5'upstream region of this gene. Calcif Tissue Int 1995; 56: 88–91
  • de Souza A. P., Trevilatto P. C., Scarel‐Caminaga R. M., Brito R. B., Line S. R. MMP‐1 promoter polymorphism: association with chronic periodontitis severity in a Brazilian population. J Clin Periodontol 2003; 30: 154–8
  • Santos M. C. L., Campos M. I. G., de Souza A. P., Trevilatto P. C., Line S. R. Analysis of MMP‐1 and MMP‐9 promoter polymorphism in early osseointegrated implant failure. Int J Maxillofac Implants 2004; 19: 38–43
  • Cao Z., Li C., Jin L., Corbet E. F. Association of matrix metalloproteinase‐1 promoter polymorphism with generalized aggressive periodontitis in a Chinese population. J Periodontal Res 2005; 40: 427–31
  • Itagaki M., Kubota T., Tai H., Shimada Y., Morozumi T., Yamazaki K. Matrix metalloproteinase‐1 and ‐3 gene promoter polymorphisms in Japanese patients with periodontitis. J Clin Periodontol 2004; 31: 764–9
  • Holla L. I., Jurajda M., Fassmann A., Dvorakova N., Znojil V., Vacha J. Genetic variations in the matrix metalloproteinase‐1 promoter and risk of susceptibility and/or severity of chronic periodontitis in the Czech population. J Clin Periodontol 2004; 31: 685–90
  • Zhang B., Ye S., Herrmann S. M., Eriksson P., de Maat M., Evans A., et al. Functional polymorphism in the regulatory region of gelatinase B gene in relation to severity of coronary atherosclerosis. Circulation 1999; 99: 1788–94
  • Wang J., Warzecha D., Wilcken D., Wang X. L. Polymorphism in the gelatinase B gene and the severity of coronary arterial stenosis. Clin Sci (Lond) 2001; 101: 87–92
  • Fiotti N., Zivadinov R., Altamura N., Nasuelli D., Bratina A., Tommasi M. A., et al. MMP‐9 microsatellite polymorphism and multiple sclerosis. J Neuroimmunol 2004; 152: 147–53
  • Wagner S., Kluge B., Koziol J. A., Grau A. J., Grond‐Ginsbach C. MMP‐9 polymorphisms are not associated with spontaneous cervical artery dissection. Stroke 2004; 35: e62–4
  • Nelissen I., Vandenbroeck K., Fiten P., Hillert J., Olsson T., Marrosu M. G., et al. Polymorphism analysis suggests that the gelatinase B gene is not a susceptibility factor for multiple sclerosis. J Neuroimmunol 2000; 105: 58–63
  • Peters D. G., Kassam A., St Jean P. L., Yonas H., Ferrell R. E. Functional polymorphism in the matrix metalloproteinase‐9 promoter as a potential risk factor for intracranial aneurysm. Stroke 1999; 30: 2612–6
  • de Souza A. P., Trevilatto P. C., Scarel‐Caminaga R. M., de Brito R. B., Jr., Barros S. P., Line S. R. Analysis of the MMP‐9 (C‐1562 T) and TIMP‐2 (G‐418C) gene promoter polymorphisms in patients with chronic periodontitis. J Clin Periodontol 2005; 32: 207–11
  • Ye S., Watts G. F., Mandalia S., Humphries S. E., Henney A. M. Preliminary report: genetic variation in the human stromelysin promoter is associated with progression of coronary atherosclerosis. Br Heart J 1995; 73: 209–15
  • de Maat M. P., Jukema J. W., Ye S., Zwinderman A. H., Moghaddam P. H., Beekman M., et al. Effect of the stromelysin‐1 promoter on efficacy of pravastatin in coronary atherosclerosis and restenosis. Am J Cardiol 1999; 83: 852–6
  • Terashima M., Akita H., Kanazawa K., Inoue N., Yamada S., Ito K., et al. Stromelysin promoter 5A/6A polymorphism is associated with acute myocardial infarction. Circulation 1999; 99: 2717–9
  • Ye S. Polymorphism in matrix metalloproteinase gene promoters: implication in regulation of gene expression and susceptibility of various diseases. Matrix Biol 2000; 19: 623–9
  • Holla L. I., Fassmann A., Vasku A., Goldbergova M., Beranek M., Znojil V., et al. Genetic variations in the human gelatinase A (matrix metalloproteinase‐2) promoter are not associated with susceptibility to, and severity of, chronic periodontitis. J Periodontol 2005; 76: 1056–60
  • Kinane D. F., Shiba H., Hart T. C. The genetic basis of periodontitis. Periodontol 2000 2005; 39: 91–117
  • Loos B. G., John R. P., Laine M. L. Identification of genetic risk factors for periodontitis and possible mechanisms of action. J Clin Periodontol 2005; 32: 159–79
  • Ryan M. E., Carnu O., Kamer A. The influence of diabetes on periodontal tissues. J Amer Dent Assoc 2003; 134: 395–405
  • Meurman J. H., Janket S. J., Qvarnstrom M., Nuutinen P. Dental infections and serum inflammatory markers in patients with and without severe heart disease. Oral Surg Oral Med Oral Pathol Oral Radiol Endod 2003; 96: 695–700
  • Zhou Z., Apte S. S., Soininen R., Cao R., Baaklini G. Y., Rauser R. W., et al. Impaired endochondral ossification and angiogenesis in mice deficient in membrane‐type matrix metalloproteinase I. Proc Natl Acad Sci U S A 2000; 97: 4052–7
  • Milner J. M., Cawston T. E. Matrix metalloproteinase knockout studies and the potential use of matrix metalloproteinase inhibitors in the rheumatic diseases. Curr Drug Targets Inflamm Allergy 2005; 4: 363–75
  • Sorsa T., Tervahartiala T., Stenman M., Suomalainen K., Mäntylä P. Chair‐side diagnostic point‐of‐care MMP tools in periodontitis and peri‐implantitis. Nordic Dentistry. Quintessence Int, Schou L, Copenhagen 2004; 79–95
  • Sorsa T., Mantyla P., Ronka H., Kallio P., Kallis G. B., Lundqvist C., et al. Scientific basis of a matrix metalloproteinase‐8 specific chair‐side test for monitoring periodontal and peri‐implant health and disease. Ann N Y Acad Sci 1999; 878: 130–40
  • Sorsa T., Uitto V. J., Suomalainen K., Vauhkonen M., Lindy S. Comparison of interstitial collagenases from human gingiva, sulcular fluid and polymorphonuclear leukocytes. J Periodontal Res 1988; 23: 386–93
  • Sorsa T., Suomalainen K., Uitto V. J. The role of gingival crevicular fluid and salivary interstitial collagenases in human periodontal diseases. Arch Oral Biol 1990; 35: 193S–196S
  • Sorsa T., Ding Y., Salo T., Lauhio A., Teronen O., Ingman T., et al. Effects of tetracyclines on neutrophil, gingival, and salivary collagenases. A functional and western‐blot assessment with special reference to their cellular sources in periodontal diseases. Ann N Y Acad Sci 1994; 732: 112–31
  • Kivelä‐Rajamäki M. J., Teronen O. P., Maisi P., Husa V., Tervahartiala T. I., Pirila E. M., et al. Laminin‐5 gamma2‐chain and collagenase‐2 (MMP‐8) in human peri‐implant sulcular fluid. Clin Oral Implants Res 2003; 14: 158–65
  • Kivelä‐Rajamäki M., Maisi P., Srinivas R., Tervahartiala T., Teronen O., Husa V., et al. Levels and molecular forms of MMP‐7 (matrilysin‐1) and MMP‐8 (collagenase‐2) in diseased human peri‐implant sulcular fluid. J Periodontal Res 2003; 38: 583–90
  • Golub L. M., Lee H. M., Ryan M. E., Giannobile W. V., Payne J., Sorsa T. Tetracyclines inhibit connective tissue breakdown by multiple non‐antimicrobial mechanisms. Adv Dent Res 1998; 12: 12–26
  • Preshaw P. M., Hefti A. F., Jepsen S., Etienne D., Walker C., Bradshaw M. H. Subantimicrobial dose doxycycline as adjunctive treatment for periodontitis. A review. J Clin Periodontol 2004; 31: 697–707
  • Coussens L. M., Fingleton B., Matrisian L. M. Matrix metalloproteinase inhibitors and cancer: trials and tribulations. Science 2002; 295: 2387–92
  • Koivunen E., Arap W., Valtanen H., Rainisalo A., Medina O. P., Heikkila P., et al. Tumor targeting with a selective gelatinase inhibitor. Nat Biotechnol 1999; 17: 768–74
  • Bjornsson M. J., Havemose‐Poulsen A., Stoltze K., Holmstrup P. Influence of the matrix metalloproteinase inhibitor batimastat (BB‐94) on periodontal bone destruction in Sprague‐Dawley rats. J Periodontal Res 2004; 39: 269–74
  • Sorsa T., Golub L. M. Is the excessive inhibition of matrix metalloproteinases (MMPs) by potent synthetic MMP inhibitors (MMPIs) desirable in periodontitis and other inflammatory diseases? That is: ‘Leaky’ MMPIs vs excessively efficient drugs. Oral Dis 2005; 11: 408–9
  • Ashley R. A. Clinical trials of a matrix metalloproteinase inhibitor in human periodontal disease. SDD Clinical Research Team. Ann N Y Acad Sci 1999; 878: 335–46
  • Craig R. G., Yip J. K., So M. K., Boylan R. J., Socransky S. S., Haffajee A. D. Relationship of destructive periodontal disease to the acute‐phase response. J Periodontol 2003; 74: 1007–16
  • D'Aiuto F., Parkar M., Andreou G., Suvan J., Brett P. M., Ready D., et al. Periodontitis and systemic inflammation: control of the local infection is associated with a reduction in serum inflammatory markers. J Dent Res 2004; 83: 156–60
  • Golub L. M., Payne J. B., Reinhardt R. A., Nieman G. Can systemic diseases co‐induce (not just exacerbate) periodontitis. A hypothetical ‘two‐hit’ model. J Dent Res 2006; 85: 102–05
  • Ridker P. M., Rifai N., Rose L., Buring J. E., Cook N. R. Comparison of C‐reactive protein and low‐density lipoprotein cholesterol levels in the prediction of first cardiovascular events. N Engl J Med 2002; 347: 1557–65
  • Blankenberg S., Rupprecht H. J., Poirier O., Bickel C., Smieja M., Hafner G., et al. AtheroGene Investigators. Plasma concentrations and genetic variation of matrix metalloproteinase 9 and prognosis of patients with cardiovascular disease. Circulation 2003; 107: 1579–85
  • Brown D. L., Desai K. K., Vakili B. A., Nouneh C., Lee H. M., Golub L. M. Clinical and biochemical results of the metalloproteinase inhibition with subantimicrobial doses of doxycycline to prevent acute coronary syndromes (MIDAS) pilot trial. Arterioscler Thromb Vasc Biol 2004; 24: 733–8
  • Brown D. L., Golub L. M., Lee H. M., Desai K., Sorsa T., Greenwald R. Effect of sub‐antimicrobial doxycycline on biomarkers in patients with acute coronary syndromes. J Dent Res 2003; 82, abstract 1443
  • Eley B. M., Cox S. W. Advances in periodontal diagnosis. 8. Commercial diagnostic kits based on GCF proteolytic and hydrolytic enzyme levels. Br Dent J 1998; 184b: 373–6
  • Mantyla P., Stenman M., Kinane D. F., Tikanoja S., Luoto H., Salo T., et al. Gingival crevicular fluid collagenase‐2 (MMP‐8) test stick for chair‐side monitoring of periodontitis. J Periodontal Res 2003; 38: 436–9
  • Golub L. M., Lee H. M., Greenwald R. A., Ryan M. E., Sorsa T., Salo T., et al. A matrix metalloproteinase inhibitor reduces bone‐type collagen degradation fragments and specific collagenases in gingival crevicular fluid during adult periodontitis. Inflamm Res 1997; 46: 310–9
  • Emingil G., Atilla G., Sorsa T., Luoto H., Kirilmaz L., Baylas H. The effect of adjunctive low‐dose doxycycline therapy on clinical parameters and gingival crevicular fluid matrix metalloproteinase‐8 levels in chronic periodontitis. J Periodontol 2004; 75: 106–15
  • Ryan M. E., Ramamurthy S., Golub L. M. Matrix metalloproteinases and their inhibition in periodontal treatment. Curr Opin Periodontol 1996; 3: 85–96
  • Apajalahti S., Sorsa T., Railavo S., Ingman T. The in vivo levels of matrix metalloproteinase‐1 and ‐8 in gingival crevicular fluid during initial orthodontic tooth movement. J Dent Res 2003; 82: 1018–22
  • Ingman T., Apajalahti S., Mantyla P., Savolainen P., Sorsa T. Matrix metalloproteinase‐1 and ‐8 in gingival crevicular fluid during orthodontic tooth movement: a pilot study during 1 month of follow‐up after fixed appliance activation. Eur J Orthod 2005; 27b: 202–7
  • Lauhio A., Salo T., Ding Y., Konttinen Y. T., Nordstrom D., Tschesche H., et al. In vivo inhibition of human neutrophil collagenase (MMP‐8) activity during long‐term combination therapy of doxycycline and non‐steroidal anti‐inflammatory drugs (NSAID) in acute reactive arthritis. Clin Exp Immunol 1994; 98: 21–8
  • Lauhio A., Salo T., Tjaderhane L., Lahdevirta J., Golub L. M., Sorsa T. Tetracyclines in treatment of rheumatoid arthritis. Lancet 1995; 346: 645–6
  • Fingleton B., Menon R., Carter K. J., Overstreet P. D., Hachey D. L., Matrisian L. M., et al. Proteinase activity in human and murine saliva as a biomarker for proteinase inhibitor efficacy. Clin Cancer Res 2004; 10: 7865–74
  • Nordstrom D., Lindy O., Lauhio A., Sorsa T., Santavirta S., Konttinen Y. T. Anti‐collagenolytic mechanism of action of doxycycline treatment in rheumatoid arthritis. Rheumatol Int 1998; 17: 175–80
  • Valleala H., Teronen O., Friman C., Sorsa T., Solovieva S., Konttinen Y. T. Inhibition of collagenase by a bisphosphonate‐group drug in patients with rheumatoid arthritis. J Rheumatol 2000; 27: 1570–2
  • Määttä M., Kari O., Tervahartiala T., Peltonen S., Kari M., Saari M., et al. Tear fluid levels of MMP‐8 are elevated in ocular rosacea‐modulation by oral doxycycline. Graefes Arch Clin Exp Ophtalmol 2006; 13: 1–6
  • Agarwal D., Goodison S., Nicholson B., Tarin D., Urquidi V. Expression of matrix metalloproteinase 8 (MMP‐8) and tyrosinase‐related protein‐1 (TYRP‐1) correlates with the absence of metastasis in an isogenic human breast cancer model. Differentiation 2003; 71: 114–25
  • Elkington P. T., O'Kane C. M., Friedland J. S. The paradox of matrix metalloproteinases in infectious disease. Clin Exp Immunol 2005; 142: 12–20

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.