Metalloproteinases of Social Determinants of Health: Comparison
Please note this is a comparison between Version 3 by Camila Xu and Version 2 by Raffaele Serra.

There are three main families of metalloproteinases (MPs) that are involved in human health and disease: (1) the “matrix metalloproteinase” (MMP) family, (2) the “a disintegrin and metalloprotease” (ADAM) family, and (3) the “a disintegrin and metalloproteinase with thrombospondin motifs” (ADAMTS) family. MPs are relevant to public health because of their role in several diseases and, most of all, their role as biomarkers that also impact the quality of life and the psychosocial dimension of affected patients. In this context, new pathways to precision health and precision medicine have been opened in the area of MPs. 

  • metalloproteinases
  • MMP
  • ADAM
  • ADAMTS
  • history
  • complexity

1. Introduction

There are three main families of metalloproteinases (MPs) that are involved in human health and disease: (1) the “matrix metalloproteinase” (MMP) family, (2) the “a disintegrin and metalloprotease” (ADAM) family, and (3) the “a disintegrin and metalloproteinase with thrombospondin motifs” (ADAMTS) family. All these families belong to the superfamily of zinc endopeptidases, which are called metzincins [1,2,3,4,5,6,7][1][2][3][4][5][6][7].
In 1962, MMP families began to be studied and Woessner published the first study on this topic demonstrating that a protein derived from a rat was able to digest collagen [8]. In particular, the mammalian uterus is one of the most favorable tissues for studying protein catabolism, especially collagen, under physiological conditions, and in fact, in this study, collagen disappeared in the postpartum period during uterus involution in a rate-limiting model, suggesting the role of specific enzyme activity. Later in the same year (1962), Gross et al. [9] showed the rapid collagenolytic activity of large tissue masses in an anuran tadpole (tail, gill, gut, skin) during natural and hormone-induced metamorphosis, and this suggested the need to identify specific enzymes able to remove particular structural components during normal growth and development. Later, in 1966, Nagai et al. purified from a tadpole the first member of the MMP family, MMP-1, initially called tadpole collagenase [10]. In that period, several MMPs and tissue inhibitors of metalloproteinases (TIMPs) were characterized [11] and, ultimately, in 1988, Birkedal-Hansen published the first review on the MMP family [12].
In 1987, the first member of the ADAM family, ADAM-1, initially known as fertilization protein PH-30 or fertilin-α, was identified; subsequently, in 1992, ADAMs were actually classified as a new family of MPs. In particular, ADAM-1 was found to act as a fusion peptide during sperm–egg fusion due to cell adhesion and protease properties; hence, it was evident that it has a role in sperm–egg interactions. Furthermore, ADAMs are also similar to the snake venom metalloproteinase family (SVM), also known as the snake venom disintegrins family [13,14,15][13][14][15].
In 1997, the ADAMTS1 gene, expressed in the cachexigenic colon 26 adenocarcinoma sublines, and the relative protein ADAMTS-1, the first member of the ADAMTS family, were characterized [16]. MPs are directly related to the homeostasis of the extracellular matrix (ECM), a biochemical center that includes collagen, elastin, and other proteins that are involved in providing structural and functional support to several tissues [7].

2. The Matrix Metalloproteinase (MMP) Family

At present, there are 28 MMP family members in vertebrates, of which at least 23 are present in humans, and they may be secreted as soluble enzymes or be bound to the cell membranes (the so-called membrane-type (MT) MMPs). Furthermore, MT-MMPs may be bound to the cell membranes by a COOH-terminal transmembrane domain or by a glycosylphosphatidyl-inositol (GPI) anchor [7]. Generally, MMPs have a signal peptide that serves to lead them to the endoplasmic reticulum (ER), a prodomain that serves to maintain them as inactive zymogens, a catalytic domain with three histidine residues bound to a zinc-binding site, and a proline-rich hinge region and a C-terminal hemopexin-like (HPX) domain (not present in some MMPs, such as MMP-7, MMP-23, and MMP-26) involved in substrate binding (Figure 1).
Figure 1. Schematic representation of MMPs. MMP: matrix metalloproteinase; MT-MMP: membrane type-matrix metalloproteinase; GMPI: glycosylphosphatidyl-inositol; Zn2+: Zinc ion.
While the MMP catalytic domains are similar among family members as they are highly conserved, the HPX domains of MMPs are particular to each MMP member [7,18,19,20,21][7][17][18][19][20]. Considering substrate specificity and homology, MMPs can be classified into six subgroups: collegenases, gelatinases, stromelysins, matrilysins, MT-MMPs, and ungrouped MMPs [7,11,22,23][7][11][21][22] (Table 1).
Table 1.
MMP family members.

References

  1. Ielapi, N.; Andreucci, M.; Licastro, N.; Faga, T.; Grande, R.; Buffone, G.; Mellace, S.; Sapienza, P.; Serra, R. Precision Medicine and Precision Nursing: The Era of Biomarkers and Precision Health. Int. J. Gen. Med. 2020, 13, 1705–1711.
  2. Costa, D.; Andreucci, M.; Ielapi, N.; Serraino, G.F.; Mastroroberto, P.; Bracale, U.M.; Serra, R. Molecular Determinants of Chronic Venous Disease: A Comprehensive Review. Int. J. Mol. Sci. 2023, 24, 1928.
  3. Serra, R.; Gallelli, L.; Butrico, L.; Buffone, G.; Caliò, F.G.; De Caridi, G.; Massara, M.; Barbetta, A.; Amato, B.; Labonia, M.; et al. From varices to venous ulceration: The story of chronic venous disease described by metalloproteinases. Int. Wound J. 2017, 14, 233–240.
  4. Costa, D.; Andreucci, M.; Ielapi, N.; Serraino, G.F.; Mastroroberto, P.; Bracale, U.M.; Serra, R. Vascular Biology of arterial aneurysms. Ann. Vasc. Surg. 2023; in press.
  5. Stöcker, W.; Bode, W. Structural features of a superfamily of zinc-endopeptidases: The metzincins. Curr. Opin. Struct. Biol. 1995, 5, 383–390.
  6. Wächter, J.; Shannon, M.J.; Beristain, A.G. Transcriptomic mapping of the metzincin landscape in human trophoblasts. Gene Expr. Patterns GEP 2022, 46, 119283.
  7. Laronha, H.; Caldeira, J. Structure and Function of Human Matrix Metalloproteinases. Cells 2020, 9, 1076.
  8. Woessner, J.F., Jr. Catabolism of collagen and non-collagen protein in the rat uterus during post-partum involution. Biochem. J. 1962, 83, 304–314.
  9. Gross, J.; Lapiere, C.M. Collagenolytic activity in amphibian tissues: A tissue culture assay. Proc. Natl. Acad. Sci. USA 1962, 48, 1014–1022.
  10. Nagai, Y.; Lapiere, C.M.; Gross, J. Tadpole collagenase. Preparation and purification. Biochemistry 1966, 5, 3123–3130.
  11. Iyer, R.P.; Patterson, N.L.; Fields, G.B.; Lindsey, M.L. The history of matrix metalloproteinases: Milestones, myths, and misperceptions. Am. J. Physiol. Heart Circ. Physiol. 2012, 303, H919–H930.
  12. Birkedal-Hansen, H. From tadpole collagenase to a family of matrix metalloproteinases. J. Oral Pathol. 1988, 17, 445–451.
  13. Zhong, S.; Khalil, R.A. A Disintegrin and Metalloproteinase (ADAM) and ADAM with thrombospondin motifs (ADAMTS) family in vascular biology and disease. Biochem. Pharmacol. 2019, 164, 188–204.
  14. Wong, G.E.; Zhu, X.; Prater, C.E.; Oh, E.; Evans, J.P. Analysis of fertilin alpha (ADAM1)-mediated sperm-egg cell adhesion during fertilization and identification of an adhesion-mediating sequence in the disintegrin-like domain. J. Biol. Chem. 2001, 276, 24937–24945.
  15. Giebeler, N.; Zigrino, P. A Disintegrin and Metalloprotease (ADAM): Historical Overview of Their Functions. Toxins 2016, 8, 122.
  16. Kuno, K.; Kanada, N.; Nakashima, E.; Fujiki, F.; Ichimura, F.; Matsushima, K. Molecular cloning of a gene encoding a new type of metalloproteinase-disintegrin family protein with thrombospondin motifs as an inflammation associated gene. J. Biol. Chem. 1997, 272, 556–562.
  17. Noël, A.; Gutiérrez-Fernández, A.; Sounni, N.E.; Behrendt, N.; Maquoi, E.; Lund, I.K.; Cal, S.; Hoyer-Hansen, G.; López-Otín, C. New and paradoxical roles of matrix metalloproteinases in the tumor microenvironment. Front. Pharmacol. 2012, 3, 140.
  18. Cabral-Pacheco, G.A.; Garza-Veloz, I.; Castruita-De la Rosa, C.; Ramirez-Acuña, J.M.; Perez-Romero, B.A.; Guerrero-Rodriguez, J.F.; Martinez-Avila, N.; Martinez-Fierro, M.L. The Roles of Matrix Metalloproteinases and Their Inhibitors in Human Diseases. Int. J. Mol. Sci. 2020, 21, 9739.
  19. Dufour, A.; Sampson, N.S.; Zucker, S.; Cao, J. Role of the hemopexin domain of matrix metalloproteinases in cell migration. J. Cell. Physiol. 2008, 217, 643–651.
  20. Alford, V.M.; Kamath, A.; Ren, X.; Kumar, K.; Gan, Q.; Awwa, M.; Tong, M.; Seeliger, M.A.; Cao, J.; Ojima, I.; et al. Targeting the Hemopexin-like Domain of Latent Matrix Metalloproteinase-9 (proMMP-9) with a Small Molecule Inhibitor Prevents the Formation of Focal Adhesion Junctions. ACS Chem. Biol. 2017, 12, 2788–2803.
  21. Shi, Y.; Ma, X.; Fang, G.; Tian, X.; Ge, C. Matrix metalloproteinase inhibitors (MMPIs) as attractive therapeutic targets: Recent progress and current challenges. NanoImpact 2021, 21, 100293.
  22. Jones, L.; Ghaneh, P.; Humphreys, M.; Neoptolemos, J.P. The matrix metalloproteinases and their inhibitors in the treatment of pancreatic cancer. Ann. N. Y. Acad. Sci. 1999, 880, 288–307.
  23. Leber, T.M.; Balkwill, F.R. Zymography: A single-step staining method for quantitation of proteolytic activity on substrate gels. Anal. Biochem. 1997, 249, 24–28.
  24. Lei, Z.; Jian, M.; Li, X.; Wei, J.; Meng, X.; Wang, Z. Biosensors and bioassays for determination of matrix metalloproteinases: State of the art and recent advances. J. Mater. Chem. B 2020, 8, 3261–3291.
  25. Hadler-Olsen, E.; Fadnes, B.; Sylte, I.; Uhlin-Hansen, L.; Winberg, J.O. Regulation of matrix metalloproteinase activity in health and disease. FEBS J. 2011, 278, 28–45.
  26. Zhu, P.; Chen, C.; Wu, D.; Chen, G.; Tan, R.; Ran, J. AGEs-induced MMP-9 activation mediated by Notch1 signaling is involved in impaired wound healing in diabetic rats. Diabetes Res. Clin. Pract. 2022, 186, 109831.
  27. Cook, L.; Sengelmann, M.; Winkler, B.; Nagl, C.; Koch, S.; Schlomann, U.; Slater, E.P.; Miller, M.A.; von Strandmann, E.P.; Dörsam, B.; et al. ADAM8-Dependent Extracellular Signaling in the Tumor Microenvironment Involves Regulated Release of Lipocalin 2 and MMP-9. Int. J. Mol. Sci. 2022, 23, 1976.
  28. Klein, T.; Bischoff, R. Physiology and pathophysiology of matrix metalloproteases. Amino Acids 2011, 41, 271–290.
  29. de Almeida, L.G.N.; Thode, H.; Eslambolchi, Y.; Chopra, S.; Young, D.; Gill, S.; Devel, L.; Dufour, A. Matrix Metalloproteinases: From Molecular Mechanisms to Physiology, Pathophysiology, and Pharmacology. Pharmacol. Rev. 2022, 74, 712–768.
  30. Vafadari, B.; Salamian, A.; Kaczmarek, L. MMP-9 in translation: From molecule to brain physiology, pathology, and therapy. J. Neurochem. 2016, 139 (Suppl. S2), 91–114.
  31. Braundmeier, A.G.; Fazleabas, A.T.; Nowak, R.A. Extracellular matrix metalloproteinase inducer expression in the baboon endometrium: Menstrual cycle and endometriosis. Reproduction 2010, 140, 911–920.
  32. Morris, S.A.; Korach, K.S.; Burns, K.A. Unique Sensitivity of Uterine Tissue and the Immune System for Endometriotic Lesion Formation. Front. Physiol. 2021, 12, 805784.
  33. Amar, S.; Smith, L.; Fields, G.B. Matrix metalloproteinase collagenolysis in health and disease. Biochim. Biophys. Acta Mol. Cell Res. 2017, 1864, 1940–1951.
  34. Kaczmarek, L.; Lapinska-Dzwonek, J.; Szymczak, S. Matrix metalloproteinases in the adult brain physiology: A link between c-Fos, AP-1 and remodeling of neuronal connections? EMBO J. 2002, 21, 6643–6648.
  35. Mittal, R.; Patel, A.P.; Debs, L.H.; Nguyen, D.; Patel, K.; Grati, M.; Mittal, J.; Yan, D.; Chapagain, P.; Liu, X.Z. Intricate Functions of Matrix Metalloproteinases in Physiological and Pathological Conditions. J. Cell. Physiol. 2016, 231, 2599–2621.
  36. Serra, R.; Buffone, G.; Falcone, D.; Molinari, V.; Scaramuzzino, M.; Gallelli, L.; de Franciscis, S. Chronic venous leg ulcers are associated with high levels of metalloproteinases-9 and neutrophil gelatinase-associated lipocalin. Wound Repair Regen. Off. Publ. Wound Heal. Soc. Eur. Tissue Repair Soc. 2013, 21, 395–401.
  37. Horecka, A.; Hordyjewska, A.; Biernacka, J.; Dąbrowski, W.; Zubilewicz, T.; Malec, A.; Musik, I.; Kurzepa, J. Intense remodeling of extracellular matrix within the varicose vein: The role of gelatinases and vascular endothelial growth factor. Ir. J. Med. Sci. 2021, 190, 255–259.
  38. Chen, Y.; Peng, W.; Raffetto, J.D.; Khalil, R.A. Matrix Metalloproteinases in Remodeling of Lower Extremity Veins and Chronic Venous Disease. Prog. Mol. Biol. Transl. Sci. 2017, 147, 267–299.
  39. Busceti, M.T.; Grande, R.; Amato, B.; Gasbarro, V.; Buffone, G.; Amato, M.; Gallelli, L.; Serra, R.; de Franciscis, S. Pulmonary embolism, metalloproteinsases and neutrophil gelatinase associated lipocalin. Acta Phlebol. 2013, 14, 115–121.
  40. Raffetto, J.D.; Ross, R.L.; Khalil, R.A. Matrix metalloproteinase 2-induced venous dilation via hyperpolarization and activation of K+ channels: Relevance to varicose vein formation. J. Vasc. Surg. 2007, 45, 373–380.
  41. Serraino, G.F.; Jiritano, F.; Costa, D.; Ielapi, N.; Battaglia, D.; Bracale, U.M.; Mastroroberto, P.; Andreucci, M.; Serra, R. Metalloproteinases in Cardiac Surgery: A Systematic Review. Biomolecules 2023, 13, 113.
  42. Luchian, I.; Goriuc, A.; Sandu, D.; Covasa, M. The Role of Matrix Metalloproteinases (MMP-8, MMP-9, MMP-13) in Periodontal and Peri-Implant Pathological Processes. Int. J. Mol. Sci. 2022, 23, 1806.
  43. Andreucci, M.; Provenzano, M.; Faga, T.; Michael, A.; Patella, G.; Mastroroberto, P.; Serraino, G.F.; Bracale, U.M.; Ielapi, N.; Serra, R. Aortic Aneurysms, Chronic Kidney Disease and Metalloproteinases. Biomolecules 2021, 11, 194.
  44. Provenzano, M.; Andreucci, M.; Garofalo, C.; Faga, T.; Michael, A.; Ielapi, N.; Grande, R.; Sapienza, P.; Franciscis, S.; Mastroroberto, P.; et al. The Association of Matrix Metalloproteinases with Chronic Kidney Disease and Peripheral Vascular Disease: A Light at the End of the Tunnel? Biomolecules 2020, 10, 154.
  45. Serra, R.; Grande, R.; Montemurro, R.; Butrico, L.; Caliò, F.G.; Mastrangelo, D.; Scarcello, E.; Gallelli, L.; Buffone, G.; de Franciscis, S. The role of matrix metalloproteinases and neutrophil gelatinase-associated lipocalin in central and peripheral arterial aneurysms. Surgery 2015, 157, 155–162.
  46. 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. Ann. Vasc. Surg. 2013, 27, 1185.e5–1185.e7.
  47. Mei, K.; Chen, Z.; Wang, Q.; Luo, Y.; Huang, Y.; Wang, B.; Gu, R. The role of intestinal immune cells and matrix metalloproteinases in inflammatory bowel disease. Front. Immunol. 2023, 13, 1067950.
  48. Chowkwale, M.; Lindsey, M.L.; Saucerman, J.J. Intercellular model predicts mechanisms of inflammation-fibrosis coupling after myocardial infarction. J. Physiol. 2022; online ahead of print.
  49. Wagner, J.; Kumar, Y.; Lautenbach, A.; von Kroge, P.; Wolter, S.; Mann, O.; Izbicki, J.; Gagliani, N.; Duprée, A. Fatty acid-binding protein-4 (FABP4) and matrix metalloproteinase-9 (MMP9) as predictive values for nonalcoholic steatohepatitis (NASH). Lipids Health Dis. 2023, 22, 1.
  50. Seegar, T.C.; Blacklow, S.C. Domain integration of ADAM family proteins: Emerging themes from structural studies. Exp. Biol. Med. 2019, 244, 1510–1519.
  51. Meyer-Schwesinger, C.; Seipold, L.; Saftig, P. Ectodomain shedding by ADAM proteases as a central regulator in kidney physiology and disease. Biochim. Biophys. Acta Mol. Cell Res. 2022, 1869, 119165.
  52. Lichtenthaler, S.F.; Lemberg, M.K.; Fluhrer, R. Proteolytic ectodomain shedding of membrane proteins in mammals-hardware, concepts, and recent developments. EMBO J. 2018, 37, e99456.
  53. Clark, P. Protease-mediated ectodomain shedding. Thorax 2014, 69, 682–684.
  54. Tatsumi, M.; Kishi, T.; Ishida, S.; Kawana, H.; Uwamizu, A.; Ono, Y.; Kawakami, K.; Aoki, J.; Inoue, A. Ectodomain shedding of EGFR ligands serves as an activation readout for TRP channels. PLoS ONE 2023, 18, e0280448.
  55. Ma, X.; Takahashi, Y.; Wu, W.; Liang, W.; Chen, J.; Chakraborty, D.; Li, Y.; Du, Y.; Benyajati, S.; Ma, J.X. ADAM17 mediates ectodomain shedding of the soluble VLDL receptor fragment in the retinal epithelium. J. Biol. Chem. 2021, 297, 101185.
  56. Brown, M.S.; Ye, J.; Rawson, R.B.; Goldstein, J.L. Regulated intramembrane proteolysis: A control mechanism conserved from bacteria to humans. Cell 2000, 100, 391–398.
  57. Butrico, L.; Barbetta, A.; Ciranni, S.; Mastroroberto, P.; Andreucci, M.; De Franciscis, S.; Serra, R. Role of metalloproteinases and their inhibitors in the development of abdominal aortic aneurysm: Current insights and systematic review of the literature. Chirurgia 2017, 30, 151–159.
  58. Kelwick, R.; Desanlis, I.; Wheeler, G.N.; Edwards, D.R. The ADAMTS (A Disintegrin and Metalloproteinase with Thrombospondin motifs) family. Genome Biol. 2015, 16, 113.
  59. Porter, S.; Clark, I.M.; Kevorkian, L.; Edwards, D.R. The ADAMTS metalloproteinases. Biochem. J. 2005, 386, 15–27.
  60. Santamaria, S.; de Groot, R. ADAMTS proteases in cardiovascular physiology and disease. Open Biol. 2020, 10, 200333.
  61. DeYoung, V.; Singh, K.; Kretz, C.A. Mechanisms of ADAMTS13 regulation. J. Thromb. Haemost. 2022, 20, 2722–2732.
  62. Grosse, G.M.; Leotescu, A.; Sieweke, J.T.; Schneppenheim, S.; Budde, U.; Ziegler, N.L.; Biber, S.; Gabriel, M.M.; Ernst, J.; Schuppner, R.; et al. ADAMTS-13 activity in stroke of known and unknown cause: Relation to vascular risk factor burden. Front. Neurol. 2023, 13, 1045478.
  63. Brew, K.; Nagase, H. The tissue inhibitors of metalloproteinases (TIMPs): An ancient family with structural and functional diversity. Biochim. Biophys. Acta 2010, 1803, 55–71.
  64. Bauer, E.A.; Stricklin, G.P.; Jeffrey, J.J.; Eisen, A.Z. Collagenase production by human skin fibroblasts. Biochem. Biophys. Res. Commun. 1975, 64, 232–240.
  65. Verstappen, J.; Von den Hoff, J.W. Tissue inhibitors of metalloproteinases (TIMPs): Their biological functions and involvement in oral disease. J. Dent. Res. 2006, 85, 1074–1084.
  66. Murphy, G. Tissue inhibitors of metalloproteinases. Genome Biol. 2011, 12, 233.
  67. Rose, K.W.J.; Taye, N.; Karoulias, S.Z.; Hubmacher, D. Regulation of ADAMTS Proteases. Front. Mol. Biosci. 2021, 8, 701959.
  68. Khanafer, K.; Ghosh, A.; Vafai, K. Correlation between MMP and TIMP levels and elastic moduli of ascending thoracic aortic aneurysms. Cardiovasc. Revascularization Med. Incl. Mol. Interv. 2019, 20, 324–327.
  69. Serra, R.; Gallelli, L.; Buffone, G.; Molinari, V.; Stillitano, D.M.; Palmieri, C.; de Franciscis, S. Doxycycline speeds up healing of chronic venous ulcers. Int. Wound J. 2015, 12, 179–184.
  70. Serra, R.; Gallelli, L.; Conti, A.; De Caridi, G.; Massara, M.; Spinelli, F.; Buffone, G.; Caliò, F.G.; Amato, B.; Ceglia, S.; et al. The effects of sulodexide on both clinical and molecular parameters in patients with mixed arterial and venous ulcers of lower limbs. Drug Des. Dev. Ther. 2014, 8, 519–527.
  71. Serra, R.; Grande, R.; Buffone, G.; Gallelli, L.; De Franciscis, S. The effects of minocycline on extracellular matrix in patients with chronic venous leg ulcers. Acta Phlebol. 2013, 14, 99–107.
  72. de Franciscis, S.; Gallelli, L.; Battaglia, L.; Molinari, V.; Montemurro, R.; Stillitano, D.M.; Buffone, G.; Serra, R. Cilostazol prevents foot ulcers in diabetic patients with peripheral vascular disease. Int. Wound J. 2015, 12, 250–253.
  73. Cione, E.; Piegari, E.; Gallelli, G.; Caroleo, M.C.; Lamirata, E.; Curcio, F.; Colosimo, F.; Cannataro, R.; Ielapi, N.; Colosimo, M.; et al. Expression of MMP-2, MMP-9, and NGAL in Tissue and Serum of Patients with Vascular Aneurysms and Their Modulation by Statin Treatment: A Pilot Study. Biomolecules 2020, 10, 359.
  74. Li, K.; Tay, F.R.; Yiu, C.K.Y. The past, present and future perspectives of matrix metalloproteinase inhibitors. Pharmacol. Ther. 2020, 207, 107465.
  75. Serra, R.; Ielapi, N.; Barbetta, A.; Andreucci, M.; de Franciscis, S. Novel biomarkers for cardiovascular risk. Biomark. Med. 2018, 12, 1015–1024.
  76. Galliera, E.; Tacchini, L.; Corsi Romanelli, M.M. Matrix metalloproteinases as biomarkers of disease: Updates and new insights. Clin. Chem. Lab. Med. 2015, 53, 349–355.
  77. Fonseca, F.L.; da Costa Aguiar Alves, B.; Azzalis, L.A.; Belardo, T.M. Matrix Metalloproteases as Biomarkers of Disease. Methods Mol. Biol. 2017, 1579, 299–311.
  78. Liu, C.H.; Di, Y.P. Matrix Metallopeptidase-Gene Signature Predicts Stage I Lung Adenocarcinoma Survival Outcomes. Int. J. Mol. Sci. 2023, 24, 2382.
  79. Sun, W.P.; Du, X.; Chen, J.J. Biomarkers for Predicting the Occurrence and Progression of Atrial Fibrillation: Soluble Suppression of Tumorigenicity 2 Protein and Tissue Inhibitor of Matrix Metalloproteinase-1. Int. J. Clin. Pract. 2022, 2022, 6926510.
  80. Stojanovic, S.K.; Stamenkovic, B.N.; Cvetkovic, J.M.; Zivkovic, V.G.; Apostolovic, M.R.A. Matrix Metalloproteinase-9 Level in Synovial Fluid-Association with Joint Destruction in Early Rheumatoid Arthritis. Medicina 2023, 59, 167.
  81. Brusa, S.; Terracciano, D.; Bruzzese, D.; Fiorenza, M.; Stanziola, L.; Pinchera, B.; Valente, V.; Gentile, I.; Cittadini, A.; Mormile, I.; et al. Circulating tissue inhibitor of metalloproteinases 1 (TIMP-1) at COVID-19 onset predicts severity status. Front. Med. 2022, 9, 1034288.
  82. Kicman, A.; Niczyporuk, M.; Kulesza, M.; Motyka, J.; Ławicki, S. Utility of Matrix Metalloproteinases in the Diagnosis, Monitoring and Prognosis of Ovarian Cancer Patients. Cancer Manag. Res. 2022, 14, 3359–3382.
  83. Noh, J.W.; Jang, J.H.; Yoon, H.S.; Kim, K.B.; Heo, M.H.; Jang, H.E.; Kim, Y.J.; Lee, Y. Evaluation of Salivary Biomarkers of Periodontal Disease Based on Smoking Status: A Systematic Review. Int. J. Environ. Res. Public Health 2022, 19, 14619.
  84. Serra, R.; Gallelli, L.; Grande, R.; Amato, B.; De Caridi, G.; Sammarco, G.; Ferrari, F.; Butrico, L.; Gallo, G.; Rizzuto, A.; et al. Hemorrhoids and matrix metalloproteinases: A multicenter study on the predictive role of biomarkers. Surgery 2016, 159, 487–494.
  85. Tsiknia, A.A.; Sundermann, E.E.; Reas, E.T.; Edland, S.D.; Brewer, J.B.; Galasko, D.; Banks, S.J.; Alzheimer’s Disease Neuroimaging Initiative. Sex differences in Alzheimer’s disease: Plasma MMP-9 and markers of disease severity. Alzheimer’s Res. Ther. 2022, 14, 160.
  86. Marks, J. Dossier: Le groupe des Dix, des précurseurs de l’interdisciplinarité–Biology and complexity: Edgar Morin and Henri Atlan. Nat. Sci. Sociétés 2019, 27, 159–168.
  87. Waldrop, M.M. Complexity: The Emerging Science at the Edge of Order and Chaos; Simon and Schuster: New York, NY, USA, 1993.
  88. Coveney, P.V. Self-Organization and Complexity: A New Age for Theory, Computation and Experiment. Philos. Trans. Math. Phys. Eng. Sci. 2003, 361, 1057–1079.
  89. Morin, E. Introduction à la Pensée Complexe; Editors du Seuil: Paris, France, 2005.
  90. Serra, R. Matrix Metalloproteinases in Health and Disease. Biomolecules 2020, 10, 1138.
  91. Morin, E. Restricted complexity, general complexity. In Worldviews, Science and Us: Philosophy and Complexity; Gershenson, C., Aerts, D., Edmonds, B., Eds.; World Scientific: Singapore, 2007; pp. 5–29.
  92. Heath-Carpentier, A. The Challenge of Complexity: Essays by Edgar Morin; Liverpool University Press: Liverpool, UK, 2022.
  93. Costa, D. Diversity and Health: Two Sides of the Same Coin. Ital. Sociol. Rev. 2023, 13, 69–90.
  94. Costa, D.; Ielapi, N.; Caprino, F.; Giannotta, N.; Sisinni, A.; Abramo, A.; Ssempijja, L.; Andreucci, M.; Bracale, U.M.; Serra, R. Social Aspects of Diabetic Foot: A Scoping Review. Soc. Sci. 2022, 11, 149.
  95. Kalra, S.; Baruah, M.P.; Sahay, R. Salutogenesis in Type 2 Diabetes Care: A Biopsychosocial Perspective. Indian J. Endocrinol. Metab. 2018, 22, 169–172.
  96. Soon, K.; Acton, C. Pain-induced stress: A barrier to wound healing. Wounds UK 2006, 2, 92–101.
  97. Alexander, S.J. Time to get serious about assessing- and managing-psychosocial issues associated with chronic wounds. Curr. Opin. Support. Palliat. Care 2013, 7, 95–100.
  98. Fu, K.; Zheng, X.; Chen, Y.; Wu, L.; Yang, Z.; Chen, X.; Song, W. Role of matrix metalloproteinases in diabetic foot ulcers: Potential therapeutic targets. Front. Pharmacol. 2022, 13, 1050630.
  99. Hariono, M.; Yuliani, S.H.; Istyastono, E.P.; Riswanto, F.D.O.; Adhipandito, C.F. Matrix metalloproteinase 9 (MMP9) in wound healing of diabetic foot ulcer: Molecular target and structure-based drug design. Vasc. Med. 2018, 22, 1–13.
  100. Jones, J.I.; Nguyen, T.T.; Peng, Z.; Chang, M. Targeting MMP-9 in Diabetic Foot Ulcers. Pharmaceuticals 2019, 12, 79.
  101. Chuang, S.Y.; Yang, S.H.; Chen, T.Y.; Pang, J.H. Cilostazol inhibits matrix invasion and modulates the gene expressions of MMP-9 and TIMP-1 in PMA-differentiated THP-1 cells. Eur. J. Pharmacol. 2011, 670, 419–426.
  102. Li, H.; Sheng, Z.; Khan, S.; Zhang, R.; Liu, Y.; Zhang, Y.; Yong, V.W.; Xue, M. Matrix Metalloproteinase-9 as an Important Contributor to the Pathophysiology of Depression. Front. Neurol. 2022, 13, 861843.
  103. Cukor, D. Introduction: Psychosocial Issues in Kidney Disease. Semin. Nephrol. 2021, 41, 485–486.
  104. Cukor, D.; Cohen, S.D.; Kimmel, P.L. Psychosocial Aspects of Chronic Kidney Disease: Exploring the Impact of CKD, Dialysis, and Transplantation on Patients; Academic Press: Cambridge, MA, USA, 2021.
  105. Bayne, D.F.; Shune, S.E. A Biopsychosocial Model of Mealtime Management in Persons with Dementia, an Asset-Based Approach to Patient-Centered Care. Geriatrics 2022, 7, 112.
  106. Ismail, Z.; Smith, E.E.; Geda, Y.; Sultzer, D.; Brodaty, H.; Smith, G.; Agüera-Ortiz, L.; Sweet, R.; Miller, D.; Lyketsos, C.G.; et al. Neuropsychiatric symptoms as early manifestations of emergent dementia: Provisional diagnostic criteria for mild behavioral impairment. Alzheimer’s Dement. 2015, 12, 195–202.
  107. Edmondson, D.; Newman, J.; Whang, W.; Davidson, K. Emotional triggers in myocardial infarction: Do they matter? Eur. Heart J. 2013, 34, 300–306.
  108. Mittleman, M.; Mostofsky, E. Physical, psychological and chemical triggers of acute cardiovascular events. Circulation 2011, 124, 346–354.
  109. Cilli, E.; Ranieri, J.; Guerra, F.; Ferri, C.; Di Giacomo, D. Cardiovascular disease, self-care and emotional regulation processes in adult patients: Balancing unmet needs and quality of life. BioPsychoSocial Med. 2022, 16, 20.
  110. Monami, M.; Marchionni, N. Psychological disorders and cardiovascular diseases. G. Ital. Cardiol. 2007, 8, 335–348.
  111. WHO. Adherence to Long-Term Therapies: Evidence for Action; WHO: Geneva, Switzerland, 2003.
  112. Thomas, H.N.; Thurston, R.C. A biopsychosocial approach to women’s sexual function and dysfunction at midlife: A narrative review. Maturitas 2016, 87, 49–60.
  113. Ventegodt, S. Sex and the quality of life in Denmark. Arch. Sex Behav. 1998, 27, 295–307.
  114. Mercer, C.H.; Fenton, K.A.; Johnson, A.M.; Wellings, K.; Macdowall, W.; McManus, S.; Nanchahal, K.; Erens, B. Sexual function problems and help seeking behaviour in Britain: National probability sample survey. BMJ Clin. Res. Ed. 2003, 327, 426–427.
  115. Newby, A.C. Metalloproteinase expression in monocytes and macrophages and its relationship to atherosclerotic plaque instability. Arterioscler. Thromb. Vasc. Biol. 2008, 28, 2108–2114.
  116. Zhang, N.; Liu, C.; Jin, L.; Zhang, R.; Wang, T.; Wang, Q.; Chen, J.; Yang, F.; Siebert, H.C.; Zheng, X. Ketogenic Diet Elicits Antitumor Properties through Inducing Oxidative Stress, Inhibiting MMP-9 Expression, and Rebalancing M1/M2 Tumor-Associated Macrophage Phenotype in a Mouse Model of Colon Cancer. J. Agric. Food Chem. 2020, 68, 11182–11196.
  117. Seraphim, P.M.; Leal, E.C.; Moura, J.; Gonçalves, P.; Gonçalves, J.P.; Carvalho, E. Lack of lymphocytes impairs macrophage polarization and angiogenesis in diabetic wound healing. Life Sci. 2020, 254, 117813.
  118. Chen, B.; Hong, H.; Sun, Y.; Chen, C.; Wu, C.; Xu, G.; Bao, G.; Cui, Z. Role of macrophage polarization in osteoarthritis (Review). Exp. Ther. Med. 2022, 24, 757.
  119. Serra, R.; Jiritano, F.; Bracale, U.M.; Ielapi, N.; Licastro, N.; Provenzano, M.; Andreucci, M.; Rizzuto, A.; Mastroroberto, P.; Serraino, G.F. Novel biomarkers in cardiovascular surgery. Biomark. Med. 2021, 15, 307–318.
  120. Serraino, G.F.; Jiritano, F.; Costa, D.; Ielapi, N.; Napolitano, D.; Mastroroberto, P.; Bracale, U.M.; Andreucci, M.; Serra, R. Metalloproteinases and Hypertrophic Cardiomyopathy: A Systematic Review. Biomolecules 2023, 13, 665.
  121. Hunt, M.A.; Birmingham, T.B.; Skarakis-Doyle, E.; Vandervoort, A.A. Towards a biopsychosocial framework of osteoarthritis of the knee. Disabil. Rehabil. 2008, 30, 54–61.
  122. Ali, S.A.; Lee, K.; MacDermid, J.C. Applying the International Classification of Functioning, Disability and Health to understand osteoarthritis management in urban and rural community-dwelling seniors. Osteoarthr. Cartil. Open 2021, 3, 100132.
  123. Chen, Y.W.; Camp, P.G.; Coxson, H.O.; Road, J.D.; Guenette, J.A.; Hunt, M.A.; Reid, W.D. A Comparison of Pain, Fatigue, Dyspnea and their Impact on Quality of Life in Pulmonary Rehabilitation Participants with Chronic Obstructive Pulmonary Disease. COPD 2018, 15, 65–72.
  124. Novy, D.M.; Aigner, C.J. The biopsychosocial model in cancer pain. Curr. Opin. Support. Palliat. Care 2014, 8, 117–123.
  125. Syrjala, K.L.; Chapko, M.E. Evidence for a biopsychosocial model of cancer treatment-related pain. Pain 1995, 61, 69–79.
  126. Badura, A.S.; Grohmann, J.M. Psychological issues in pain perception and treatment in the elderly. Ann. Long-Term Care 2002, 10, 29–34.
  127. Masselin-Dubois, A.; Attal, N.; Fletcher, D.; Jayr, C.; Albi, A.; Fermanian, J.; Bouhassira, D.; Baudic, S. Are psychological predictors of chronic postsurgical pain dependent on the surgical model? A comparison of total knee arthroplasty and breast surgery for cancer. J. Pain 2013, 14, 854–864.
  128. Costa, D.; Ielapi, N.; Bevacqua, E.; Ciranni, S.; Cristodoro, L.; Torcia, G.; Serra, R. Social Determinants of Health and Vascular Diseases: A Systematic Review and Call for Action. Soc. Sci. 2023, 12, 214.
  129. Shafi, B.H.; Bøttcher, M.; Ejupi, A.; Jensen, G.; Osler, M.; Lange, T.; Prescott, E. Socioeconomic disparity in cardiovascular disease: Possible biological pathways based on a proteomic approach. Atherosclerosis 2022, 352, 62–68.
  130. Carey, G.; Malbon, E.; Carey, N.; Joyce, A.; Crammond, B.; Carey, A. Systems science and systems thinking for public health: A systematic review of the field. BMJ Open 2015, 5, e009002.
  131. Cabral, M.D.F.C.T.; Viana, A.L.; Gontijo, D.T. Use of the complexity paradigm in the field of health: Scope review. Esc. Anna Nery 2020, 24, e20190235.
  132. de Franciscis, S.; Fregola, S.; Gallo, A.; Argirò, G.; Barbetta, A.; Buffone, G.; Caliò, F.G.; De Caridi, G.; Amato, B.; Serra, R. PredyCLU: A prediction system for chronic leg ulcers based on fuzzy logic; part I—Exploring the venous side. Int. Wound J. 2016, 13, 1349–1353.
  133. Serra, R.; Bracale, U.M.; Barbetta, A.; Ielapi, N.; Licastro, N.; Gallo, A.; Fregola, S.; Turchino, D.; Gasbarro, V.; Mastroroberto, P.; et al. PredyCLU: A prediction system for chronic leg ulcers based on fuzzy logic; part II-Exploring the arterial side. Int. Wound J. 2020, 17, 987–991.
  134. Garvin, P.; Jonasson, L.; Nilsson, L.; Falk, M.; Kristenson, M. Plasma Matrix Metalloproteinase-9 Levels Predict First-Time Coronary Heart Disease: An 8-Year Follow-Up of a Community-Based Middle Aged Population. PLoS ONE 2015, 10, e0138290.
  135. Lundberg, A.K.; Jönsson, S.; Stenmark, J.; Kristenson, M.; Jonasson, L. Stress-induced release of matrix metalloproteinase-9 in patients with coronary artery disease: The possible influence of cortisol. Psychoneuroendocrinology 2016, 73, 117–124.
  136. Powell-Wiley, T.M.; Baumer, Y.; Baah, F.O.; Baez, A.S.; Farmer, N.; Mahlobo, C.T.; Pita, M.A.; Potharaju, K.A.; Tamura, K.; Wallen, G.R. Social determinants of cardiovascular disease. Circ. Res. 2022, 130, 782–799.
  137. Kasthurirathne, S.N.; Vest, J.R.; Menachemi, N.; Halverson, P.K.; Grannis, S.J. Assessing the capacity of social determinants of health data to augment predictive models identifying patients in need of wraparound social services. J. Am. Med. Inform. Assoc. 2018, 25, 47–53.
More
Video Production Service