全部 标题 作者
关键词 摘要

OALib Journal期刊
ISSN: 2333-9721
费用:99美元

查看量下载量

相关文章

更多...
Pathogens  2013 

The Significance of Matrix Metalloproteinases in Parasitic Infections Involving the Central Nervous System

DOI: 10.3390/pathogens2010105

Keywords: matrix metalloproteinases, tissue inhibitor of metalloproteinases, malaria, trypanosomosis, toxoplasmosis, neurocysticercosis, angiostrongyloidosis

Full-Text   Cite this paper   Add to My Lib

Abstract:

Matrix metalloproteinases (MMPs) represent a large family of over twenty different secreted or membrane-bound endopeptidases, involved in many physiological (embryogenesis, precursor or stem cell mobilization, tissue remodeling during wound healing, etc.), as well as pathological (inflammation, tumor progression and metastasis in cancer, vascular pathology, etc.) conditions. For a long time, MMPs were considered only for the ability to degrade extracellular matrix (ECM) molecules (e.g., collagen, laminin, fibronectin) and to release hidden epitopes from the ECM. In the last few years, it has been fully elucidated that these molecules have many other functions, mainly related to the immune response, in consideration of their effects on cytokines, hormones and chemokines. Among others, MMP-2 and MMP-9 are endopeptidases of the MMP family produced by neutrophils, macrophages and monocytes. When infection is associated with leukocyte influx into specific organs, immunopathology and collateral tissue damage may occur. In this review, the involvement of MMPs and, in particular, of gelatinases in both protozoan and helminth infections will be described. In cerebral malaria, for example, MMPs play a role in the pathogenesis of such diseases. Also, trypanosomosis and toxoplasmosis will be considered for protozoan infections, as well as neurocysticercosis and angiostrongyloidosis, as regards helminthiases. All these situations have in common the proteolytic action on the blood brain barrier, mediated by MMPs.

References

[1]  Visse, R.; Nagase, H. Matrix metalloproteinases and tissue inhibitors of metalloproteinases: structure, function, and biochemistry. Circ. Res. 2003, 92, 827–839, doi:10.1161/01.RES.0000070112.80711.3D.
[2]  Kumar, V.; Fausto, N.; Abbas, A. Robbins and Cotran: Pathologic Basis of Disease, 7th ed.; Elsevier: Philadelphia, PA, USA, 2004.
[3]  Ingber, D.E. Mechanical control of tissue morphogenesis during embryological development. Int. J. Dev. Biol. 2006, 50, 255–266, doi:10.1387/ijdb.052044di.
[4]  Maleski, M.; Hockfield, S. Glial cells assemble hyaluronan-based pericellular matrices in vitro. Glia 1997, 20, 193–202, doi:10.1002/(SICI)1098-1136(199707)20:3<193::AID-GLIA3>3.0.CO;2-9.
[5]  Rauch, U. Modeling an extracellular environment for axonal pathfinding and fasciculation in the central nervous system. Cell Tissue Res. 1997, 290, 349–356, doi:10.1007/s004410050940.
[6]  Fujita, M.; Spray, D.C.; Choi, H.; Saez, J.; Jefferson, D.M.; Hertzberg, E.; Rosenberg, L.C.; Reid, L.M. Extracellular matrixregulation ofcell-cell communicationand tissue-specific gene expression in primary liver cultures. Prog. Clin. Biol. Res. 1986, 226, 333–360.
[7]  Noguera, R.; Nieto, O.A.; Tadeo, I.; Fari?as, F.; Alvaro, T. Extracellular matrix, biotensegrity and tumor microenvironment. An update and overview. Histol. Histopathol. 2012, 27, 693–705.
[8]  Mosher, D.F.; Adams, J.B. Adhesion-modulating/matricellular ECM protein families: A structural, functional and evolutionary appraisal. Matrix Biol. 2012, 31, 155–161, doi:10.1016/j.matbio.2012.01.003.
[9]  Lin, C.Q.; Bissell, M.J. Multi-faceted regulation of cell differentiation by extracellular matrix. FASEB J. 1993, 7, 737–743.
[10]  Werb, Z.; Sympson, C.J.; Alexander, C.M.; Thomasset, N.; Lund, L.R.; MacAuley, A.; Ashkenas, J.; Bissell, M.J. Extracellular matrix remodeling and the regulation of epithelial-stromal interactions during differentiation and involution. Kidney Int. Suppl. 1996, 54, S68–S74.
[11]  Gentili, C.; Cancedda, R. Cartilage and bone extracellular matrix. Curr. Pharm. Des. 2009, 15, 1334–1348, doi:10.2174/138161209787846739.
[12]  Davis, G.E.; Donald, R.; Senger, D.R. Endothelial Extracellular Matrix: Biosynthesis, Remodeling, and Functions During Vascular Morphogenesis and Neovessel Stabilization. Circ. Res. 2005, 97, 1093–1107, doi:10.1161/01.RES.0000191547.64391.e3.
[13]  Plopper, G. The extracellular matrix and cell adhesion, in Cells; Lewin, B., Cassimeris, L., Lingappa, V., Plopper, G., Sudbury, M.A., Eds.; Jones and Bartlett: Burlington, MA, USA, 2007.
[14]  Wiese, S.; Karus, M.; Faissner, A. Astrocytes as a source for extracellular matrix molecules and cytokines. Front Pharmacol. 2012, 3, 120, doi:10.3389/fphar.2012.00120.
[15]  Nicholson, C.; Syková, E. Extracellular space structure revealed by diffusion analysis. Trends Neurosci. 1998, 21, 207–215, doi:10.1016/S0166-2236(98)01261-2.
[16]  Mott, J.D.; Werb, Z. Regulation of matrix biology by matrix metalloproteinases. Curr. Opin. Cell. Biol. 2004, 16, 558–564, doi:10.1016/j.ceb.2004.07.010.
[17]  Nagase, H.; Visse, R.; Murphy, G. Structure and function of matrix metalloproteinases and TIMPs. Cardiovascular Res. 2006, 69, 562–573, doi:10.1016/j.cardiores.2005.12.002.
[18]  Roycik, M.D.; Fang, X.; Sang, Q.X. A fresh prospect of extracellular matrix hydrolytic enzymes and their substrates. Curr. Pharm. Des. 2009, 15, 1295–1308, doi:10.2174/138161209787846676.
[19]  Hijova, E. Matrix metalloproteinases: their biological functions and clinical implication. Bratisl. Lek. Listy. 2005, 106, 127–132.
[20]  Ghajar, C.M.; George, S.C.; Putnam, A.J. Matrix metalloproteinase control of capillary morphogenesis. Crit. Rev. Eukaryot. Gene Expr. 2008, 18, 251–278, doi:10.1615/CritRevEukarGeneExpr.v18.i3.30.
[21]  Page-McCaw, A.; Ewald, A.J.; Werb, Z. Matrix metalloproteinases and the regulation of tissue remodelling. Nat. Rev. Mol. Cell Biol. 2007, 8, 221–233, doi:10.1038/nrm2125.
[22]  Ravi, A.; Pallavi, G.; Sitaraman, S.V. matrix metalloproteinases in inflammatory bowel disease: boon or a bane? Inflamm. Bowel Dis. 2007, 13, 97–107, doi:10.1002/ibd.20011.
[23]  Yong, V.W.; Power, C.; Forsyth, P.; Edwards, D.R. Metalloproteinases in biology and pathology of the nervous system. Nature Rev. 2001, 2, 502–511, doi:10.1038/35081571.
[24]  Dzwonek, J.; Rylski, M.; Kaczmarek, L. Matrix metalloproteinases and their endogenous inhibitors in neuronal physiology of the adult brain. FEBS Lett. 2004, 567, 129–135, doi:10.1016/j.febslet.2004.03.070.
[25]  Sedlacek, R.; Mauch, S.; Kolb, B.; Sch?tzlein, C.; Eibel, H.; Peter, H.H.; Schmitt, J.; Krawinkel, U. Matrix metalloproteinase MMP-19 (RASI-1) is expressed on the surface of activated peripheral blood mononuclear cells and is detected as an autoantigen in rheumatoid arthritis. Immunobiol. 1998, 198, 408–423, doi:10.1016/S0171-2985(98)80049-1.
[26]  Tetlow, L.C.; Adlam, D.J.; Woolley, D.E. Matrix metalloproteinase and proinflammatory cytokine production by chondrocytes of human osteoarthritic cartilage: associations with degenerative changes. Arthritis Rheum. 2001, 44, 585–594, doi:10.1002/1529-0131(200103)44:3<585::AID-ANR107>3.0.CO;2-C.
[27]  Yoshida, W.; Uzuki, M.; Nishida, J.; Shimamura, T.; Sawai, T. Examination of in vivo gelatinolytic activity in rheumatoid arthritis synovial tissue using newly developed in situ zymography and image analyzer. Clin. Exp. Rheumatol. 2009, 27, 587–593.
[28]  Creemers, E.E.; Cleutjens, J.P.; Smits, J.F.; Daemen, M.J. Matrix metalloproteinase inhibition after myocardial infarction: a new approach to prevent heart failure? Circ. Res. 2001, 89, 201–210, doi:10.1161/hh1501.094396.
[29]  Siefert, S.A.; Sarkar, R. Matrix metalloproteinases in vascular physiology and disease. Vascular. 2012, 20, 210–216, doi:10.1258/vasc.2011.201202.
[30]  Briasoulis, A.; Tousoulis, D.; Papageorgiou, N.; Kampoli, A.M.; Androulakis, E.; Antoniades, C.; Tsiamis, E.; Latsios, G.; Stefanadis, C. Novel therapeutic approaches targeting matrix metalloproteinases in cardiovascular disease. Curr. Top. Med. Chem. 2012, 12, 1214–1221, doi:10.2174/1568026611208011214.
[31]  Srivastava, P.K.; Dastidar, S.G.; Ray, A. Chronic obstructive pulmonary disease: role of matrix metalloproteases and future challenges of drug therapy. Expert. Opin. Investig. Drugs. 2007, 16, 1069–1078, doi:10.1517/13543784.16.7.1069.
[32]  Oikonomidi, S.; Kostikas, K.; Tsilioni, I.; Tanou, K.; Gourgoulianis, K.I.; Kiropoulos, T.S. Matrix metalloproteinases in respiratory diseases: from pathogenesis to potential clinical implications. Curr. Med. Chem. 2009, 16, 1214–1228, doi:10.2174/092986709787846587.
[33]  Mocchegiani, E.; Giacconi, R.; Costarelli, L. Metalloproteases/anti-metalloproteases imbalance in chronic obstructive pulmonary disease: genetic factors and treatment implications. Curr. Opin. Pulm. Med. 2011, 17, S11–S19, doi:10.1097/01.mcp.0000410743.98087.12.
[34]  Leppert, D.; Lindberg, R.L.; Kappos, L.; Leib, S.L. Matrix metalloproteinases: multifunctional effectors of inflammation in multiple sclerosis and bacterial meningitis. Brain Res. Rev. 2001, 36, 249–257, doi:10.1016/S0165-0173(01)00101-1.
[35]  Parks, W.C.; Wilson, C.L.; Lòpez-Boado, Y.S. Matrix metalloproteinases as modulators of inflammation and innate immunity. Nat. Rev. 2004, 4, 617–628.
[36]  Gharagozlian, S.; Svennevig, K.; Bangstad, H.J.; Winberg, J.O.; Kolset, S.O. Matrix metalloproteinases in subjects with type 1 diabetes. BMC Clin. Pathol. 2009, 16, 7, doi:10.1186/1472-6890-9-7.
[37]  Coussens, L.M.; Werb, Z. Matrix metalloproteinases and the development of cancer. Chem. Biol. 1996, 3, 895–904, doi:10.1016/S1074-5521(96)90178-7.
[38]  van Kempen, L.C.; Coussens, L.M. MMP-9 potentiates pulmonary metastasis formation. Cancer Cell. 2002, 2, 251–252, doi:10.1016/S1535-6108(02)00157-5.
[39]  Egeblad, M.; Werb, Z. New functions for the matrix metalloproteinases in cancer progression. Nat. Rev. Cancer. 2002, 2, 161–174, doi:10.1038/nrc745.
[40]  Zucker, S.; Vacirca, J. Role of matrix metalloproteinases (MMPs) in colorectal cancer. Cancer Metastasis Rev. 2004, 23, 101–117, doi:10.1023/A:1025867130437.
[41]  Affara, N.I.; Andreu, P.; Coussens, L.M. Delineating protease functions during cancer development. Methods Mol. Biol. 2009, 539, 1–32, doi:10.1007/978-1-60327-003-8_1.
[42]  Rooprai, H.K.; McCormick, D. Proteases and their inhibitors in human brain tumours: a review. Anticancer Res. 1997, 17, 4151–4162.
[43]  Avolio, C.; Ruggieri, M.; Giuliani, F.; Liuzzi, G.M.; Leante, R.; Riccio, P.; Livrea, P.; Trojano, M. Serum MMP-2 and MMP-9 are elevated in ifferent multiple sclerosis subtypes. J. Neuroimmunol. 2003, 136, 46–53, doi:10.1016/S0165-5728(03)00006-7.
[44]  Backstrom, J.R.; Miller, C.A.; Tokes, Z.A. Characterization of neutral proteinases from Alzheimer-affected and control brain specimens: identification of calcium-dependent metalloproteinases from the hippocampus. J. Neurochem. 1992, 58, 983–992, doi:10.1111/j.1471-4159.1992.tb09352.x.
[45]  Créange, A.; Sharshar, T.; Planchenault, T.; Christov, C.; Poron, F.; Raphae¨l, J.C.; Gherardi, R.K. Matrix metalloproteinase-9 is increased and correlates with severity in Guillain-Barre′ syndrome. Neurol. 1999, 53, 1683–1691, doi:10.1212/WNL.53.8.1683.
[46]  Lim, G.P.; Backstrom, J.R.; Cullen, M.J.; Miller, C.A.; Atkinson, R.D.; Tokes, Z.A. Matrix metalloproteinases in the neocortex and spinal cord of amyotrophic lateral sclerosis patients. J. Neurochem. 1996, 67, 251–259.
[47]  Rosenberg, G.A. Matrix metalloproteinases in brain injury. J. Neurotrauma 1995, 12, 833–842, doi:10.1089/neu.1995.12.833.
[48]  Lukes, A.; Mun-Bryce, S.; Lukes, M.; Rosenberg, G.A. Extracellular matrix degradation by metalloproteinases and central nervous system diseases. Mol. Neurobiol. 1999, 19, 267–284, doi:10.1007/BF02821717.
[49]  Liuzzi, G.M.; Mastroianni, C.M.; Santacroce, M.P.; Fanelli, M.; D’Agostino, C.; Vullo, V.; Riccio, P. Increased activity of matrix metalloproteinases in the cerebrospinal fluid of patients with HIV-associated neurological disease. J. Neurovirol. 2000, 6, 156–163, doi:10.3109/13550280009013159.
[50]  Sterlicht, M.D.; Werb, Z. How matrix metalloproteinases regulate cell behaviour. Ann. Rev. Cell Dev. Biol. 2001, 17, 463–516, doi:10.1146/annurev.cellbio.17.1.463.
[51]  Welgus, H.G.; Campbell, E.J.; Cury, J.D.; Eisen, A.Z.; Senior, R.M.; Wilhelm, S.M.; Goldberg, G.I. Neutral metalloproteinases produced by human mononuclear phagocytes. Enzyme profile, regulation, and expression during cellular development. J. Clin. Invest. 1990, 86, 1496–1502, doi:10.1172/JCI114867.
[52]  Leppert, D.; Waubant, E.; Galardy, R.; Bunnett, N.W.; Hauser, S.L. T cell gelatinases mediate basement membrane transmigration in vitro. J. Immunol. 1995, 154, 4379–4389.
[53]  Masure, S.; Proost, P.; van Damme, J.; Opdenakker, G. Purification and identification of 91-kDa neutrophil gelatinase: release by the activating peptide interleukin-8. Eur. J. Biochem. 1991, 198, 391–398, doi:10.1111/j.1432-1033.1991.tb16027.x.
[54]  Wells, G.M.; Catlin, G.; Cossins, J.A.; Mangan, M.; Ward, G.A.; Miller, K.M.; Clements, J.M. Quantitation of matrix metalloproteinases in cultured rat astrocytes using the polymerase chain reaction with a multi-competitor cDNA standard. Glia 1996, 18, 332–340, doi:10.1002/(SICI)1098-1136(199612)18:4<332::AID-GLIA7>3.0.CO;2-Z.
[55]  Gottschall, P.E.; Yu, X. Cytokines regulate gelatinase A and B (matrix metalloproteinase 2 and 9) activity in cultured rat astrocytes. J. Neurochem. 1995, 64, 1513–1520, doi:10.1046/j.1471-4159.1995.64041513.x.
[56]  Okada, S.; Kita, H.; George, T.J.; Gleich, G.J.; Leiferman, K.M. Migration of eosinophils through basement membrane components in vitro: role of matrix metalloproteinase-9. Am. J. Respir. Cell. Mol. Biol. 1997, 17, 519–528.
[57]  Nielsen, B.S.; Timshel, S.; Kjeldsen, L.; Sehested, M.; Pyke, C.; Borregaard, N.; Dano, K. 92 kDa type IV collagenase (MMP-9) is expressed in neutrophils and macrophages but not in malignant epithelial cells in human colon cancer. Int. J. Cancer 1996, 65, 57–62, doi:10.1002/(SICI)1097-0215(19960103)65:1<57::AID-IJC10>3.0.CO;2-F.
[58]  Herron, G.S.; Werb, Z.; Dwyer, K.; Banda, M.J. Secretion of metalloproteinases by stimulated capillary endothelial cells. In Production of procollagenase and prostromelysin exceeds expression of proteolytic activity. J. Biol. Chem. 1986, 261, 2810–2813.
[59]  Birkedal-Hansen, H.; Moore, W.G.; Bodden, M.K.; Windsor, L.J.; Birkedal-Hansen, B.; DeCarlo, A.; Engler, J.A. Matrix metalloproteinases: a review. Crit. Rev. Oral Biol. Med. 1993, 4, 197–250.
[60]  Birkedal-Hansen, H.; Yamada, S.; Windsor, J.; Pollard, A.H.; Lyons, G.; Stetler-Stevenson, W.; Birkedal-Hansen, B. Matrix metalloproteinases. Curr. Protoc. Cell Biol. 2008, 10, 8.
[61]  Uría, J.A.; López-Otín, C. Matrilysin-2, a new matrix metalloproteinase expressed in human tumors and showing the minimal domain organization required for secretion, latency, and activity. Cancer Res. 2000, 60, 4745–4751.
[62]  Nagase, H.; Woessner, J.F., Jr. Matrix metalloproteinases. J. Biol. Chem. 1999, 274, 21491–21494, doi:10.1074/jbc.274.31.21491.
[63]  Overall, C.M. Molecular determinants of metalloproteinase substrate specificity: matrix metalloproteinase substrate binding domains, modules, and exosites. Mol. Biotechnol. 2002, 22, 51–86, doi:10.1385/MB:22:1:051.
[64]  Stamenkovic, I. Extracellular matrix remodelling: the role of matrix metalloproteinases. J. Pathol. 2003, 200, 448–464, doi:10.1002/path.1400.
[65]  Steffens, B.; Hakkinen, L.; Larjava, H. Proteolytic events of wound-healing-coordinated interactions among matrix metalloproteinanses (MMPs), integrins, and extracellular matrix molecules. Crit. Rev. Oral Biol. Med. 2001, 12, 373–398, doi:10.1177/10454411010120050201.
[66]  Shapiro, S.D.; Kobayashi, D.K.; Ley, T.J. Cloning and characterization of a unique elastolytic metalloproteinase produced by human alveolar macrophages. J. Biol. Chem. 1993, 268, 23824–23829.
[67]  Salmela, M.T.; Karjalainen-Lindsberg, M.L.; Puolakkainen, P.; Saarialho-Kere, U. Upregulation and differential expression of matrilysin (MMP-7) and metalloelastase (MMP-12) and their inhibitors TIMP-1 and TIMP-3 in Barrett's oesophageal adenocarcinoma. Br. J. Cancer 2001, 85, 383–392, doi:10.1054/bjoc.2001.1929.
[68]  Wilson, C.L.; Matrisian, L.M. Matrilysin: an epithelial matrix metalloproteinase with potentially novel functions. Int. J. Biochem. Cell Biol. 1996, 28, 123–136, doi:10.1016/1357-2725(95)00121-2.
[69]  Park, H.I.; Ni, J.; Gerkema, F.E.; Liu, D.; Belozerow, V.; Sang, Q.X.A. Identification and characterization of human endometase (matrix metalloproteinase-26) from endometrial tumor. J. Biol. Chem. 2000, 27, 20540–20544.
[70]  Galewskaa, Z.; Romanowicza, L.; Jaworskib, S.; Bańkowskia, E. Matrix metalloproteinases, MMP-7 and MMP-26, in plasma and serum of control and preeclamptic umbilical cord blood. Eur. J. Obstetrics Gynecol. Repr. Biol. 2010, 150, 152–156, doi:10.1016/j.ejogrb.2010.03.007.
[71]  Massova, I.; Kotra, L.P.; Fridman, R.; Mobashery, S. Matrix metalloproteinases: structures, evolution, and diversification. FASEB J. 1998, 12, 1075–1095.
[72]  Das, S.; Mandal, M.; Chakraborti, T.; Mandal, A.; Chakraborti, S. Structure and evolutionary aspects of matrix metalloproteinases: a brief overview. Mol. Cell Biochem. 2003, 253, 31–40, doi:10.1023/A:1026093016148.
[73]  Ii, M.; Yamamoto, H.; Adachi, Y.; Maruyama, Y.; Shinomura, Y. Role of matrix metalloproteinase-7 (matrilysin) in human cancer invasion, apoptosis, growth, and angiogenesis. Exp. Biol. Med. (Maywood) 2006, 231, 20–27.
[74]  Jones, C.B.; Sane, D,C.; Herrington, D.M. Matrix metalloproteinases: a review of their structure and role in acute coronary syndrome. Cardiovasc. Res. 2003, 59, 812–823, doi:10.1016/S0008-6363(03)00516-9.
[75]  Fillmore, H.L.; VanMeter, T.E.; Broaddus, W.C. Membrane-type matrix metalloproteinases (MT-MMPs): expression and function during glioma invasion. J. Neurooncol. 2001, 53, 187–202, doi:10.1023/A:1012213604731.
[76]  Zucker, S.; Pei, D.; Cao, J.; Lopez-Otin, C. Membrane type-matrix metalloproteinases (MT-MMP). Curr. Top. Dev. Biol. 2003, 54, 1–74, doi:10.1016/S0070-2153(03)54004-2.
[77]  Stracke, O.J.; Fosang, J.A.; Last, K.; Mercuri, A.F.; Pendas, M.A.; Llano, E.; Perris, R.; Di Cesare, E.P. Matrix metalloproteinases 19 and 20 cleave aggrecan and cartilage oligomeric matrix protein (COMP). FEBS Lett. 2000, 478, 52–56, doi:10.1016/S0014-5793(00)01819-6.
[78]  Lohi, J.; Wilson, C.L.; Roby, J.D.; Parks, W.C. Epilysin, a novel human matrix metalloproteinase (MMP-28) expressed in testis and keratinocytes and in response to injury. J. Biol. Chem. 2001, 276, 10134–10144.
[79]  Fini, M.E.; Cook, J.R.; Mohan, R. Proteolytic mechanisms in corneal ulceration and repair. Arch. Dermatol. Res. 1998, 290, S12–S23, doi:10.1007/PL00007449.
[80]  van den Berg, W.B. The role of cytokines and growth factors in cartilage destruction in osteoarthritis and rheumatoid arthritis. J. Rheumatol. 1999, 58, 136–141.
[81]  Cawston, T.E.; Wilson, A.J. Understanding the role of tissue degrading enzymes and their inhibitors in development and disease. Best Pract. Res. Clin. Rheumatol. 2006, 20, 983–1002, doi:10.1016/j.berh.2006.06.007.
[82]  Yan, C.; Boyd, D.D. Regulation of matrix metalloproteinase gene expression. J. Cell. Physiol. 2007, 211, 19–26, doi:10.1002/jcp.20948.
[83]  Clark, I.M.; Swingler, T.E.; Sampieri, C.L.; Edwards, D.R. The regulation of matrix metalloproteinases and their inhibitors. Int. J. Biochem. Cell. Biol. 2008, 40, 1362–1378, doi:10.1016/j.biocel.2007.12.006.
[84]  Springman, E.B.; Angleton, E.L.; Birkedal-Hansen, H.; Van Wart, H.E. Multiple modes of activation of latent human fibroblast collagenase: evidence for the role of a Cys73 active-site zinc complex in latency and a “cysteine switch” mechanism for activation. Proc. Natl. Acad. Sci. USA 1990, 87, 364–368.
[85]  Van Wart, H.E. Birkedal-Hansen, H. The cysteine switch: a principle of regulation of metalloproteinase activity with potential applicability to the entire matrix metalloproteinase gene family. Proc. Natl. Acad. Sci. USA 1990, 87, 5578–5582, doi:10.1073/pnas.87.14.5578.
[86]  Kotra, L.P.; Zhang, L.; Fridman, R.; Orlando, R.; Mobashery, S. N-Glycosylation pattern of the zymogenic form of human matrix metalloproteinase-9. Bioorg. Chem. 2002, 30, 356–370, doi:10.1016/S0045-2068(02)00501-1.
[87]  Yang, Z.; Strickland, D.K.; Bornstein, P. Extracellular MMP-2 levels are regulated by the low-density lipoprotein-related scavenger receptor and thrombospondin 2. J. Biol. Chem. 2001, 276, 8403–8408.
[88]  Murphy, G. Tissue inhibitors of metalloproteinases. Murphy Genome Biol. 2011, 12, 1–7.
[89]  Brew, K.; Dinakarpandian, D.; Nagase, H. Tissue inhibitors of metalloproteinases: evolution, structure and function. Biochim. Biophys. Acta. 2000, 1477, 267–283, doi:10.1016/S0167-4838(99)00279-4.
[90]  Bode, W.; Maskos, K. Structural basis of the matrix metalloproteinases and their physiological inhibitors, the tissue inhibitors of metalloproteinases. Biol. Chem. 2003, 384, 863–872.
[91]  Gomez, D.E.; Alonso, D.F.; Yoshiji, H.; Thorgeirsson, U.P. Tissue inhibitors of metalloproteinases: structure, regulation and biological functions. Eur. J. Cell. Biol. 1997, 7, 111–122.
[92]  Jiang, Y.; Goldberg, I.D.; Shi, Y.E. Complex roles of tissue inhibitors of metalloproteinases in cancer. Oncogene 2002, 21, 2245–2252, doi:10.1038/sj.onc.1205291.
[93]  Guedez, L.; Stetler-Stevenson, W.G.; Wolff, L.; Wang, J.; Fukushima, P.; Mansoor, A.; Stetler-Stevenson, M. In vitro suppression of programmed cell death of B cells by tissue inhibitor of metalloproteinases-1. J. Clin. Invest. 1998, 102, 2002–2010, doi:10.1172/JCI2881.
[94]  Fata, J.E.; Leco, K.J.; Voura, E.B.; Yu, H.Y.; Waterhouse, P.; Murphy, G.; Moorehead, R.A.; Khokha, R. Accelerated apoptosis in the Timp-3-deficient mammary gland. J. Clin. Invest. 2001, 108, 831–841.
[95]  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, doi:10.1111/j.1742-4658.2010.07920.x.
[96]  Geurts, N.; Opdenakker, G.; Van den Steen, P.E. Matrix metalloproteinases as therapeutic targets in protozoan parasitic infections. Pharmacol. Ther. 2012, 133, 257–279, doi:10.1016/j.pharmthera.2011.11.008.
[97]  Snow, R.W.; Guerra, C.A.; Noor, A.M.; Myint, H.Y.; Hay, S.I. The global distribution of clinical episodes of Plasmodium falciparum malaria. Nature 2005, 434, 214–217, doi:10.1038/nature03342.
[98]  Kappe, S.H.; Vaughan, A.M.; Boddey, J.A.; Cowman, A.F. Thatwas then but this is now: malaria research in the time of an eradication agenda. Science 2010, 328, 862–866, doi:10.1126/science.1184785.
[99]  van der Heyde, H.C.; Nolan, J.; Combes, V.; Gramaglia, I.; Grau, G.E. A unified hypothesis for the genesis of cerebral malaria: sequestration, inflammation and hemostasis leading to microcirculatory dysfunction. Trends Parasitol. 2006, 22, 503–508, doi:10.1016/j.pt.2006.09.002.
[100]  Miller, L.H.; Dror, I.; Baruch, K.M.; Ogobara, K.D. The pathogenic basis of malaria. Nature 2002, 415, 673–679, doi:10.1038/415673a.
[101]  Armah, H.; Dodoo, A.K.; Wiredu, E.K.; Stiles, J.K.; Adjei, A.A.; Gyasi, R.K.; Tettey, Y. High-level cerebellar expression of cytokines and adhesion molecules in fatal, paediatric, cerebral malaria. Ann. Trop. Med. Parasitol. 2005, 99, 629–647, doi:10.1179/136485905X51508.
[102]  Shikani, H.J.; Freeman, B.D.; Lisanti, M.P.; Weiss, L.M.; Tanowitz, H.B.; Desruisseaux, M.S. Cerebral malaria:We Have Come a Long Way. Am. J. Pathol. 2012, 181, 1484–1492, doi:10.1016/j.ajpath.2012.08.010.
[103]  Abbott, N.J.; R?nnb?ck, L.; Hansson, E. Astrocyte-endothelial interactions at the blood-brain barrier. Nat. Rev. Neurosci. 2006, 7, 41–53, doi:10.1038/nrn1824.
[104]  Yong, V.W. Metalloproteinases: mediators of pathology and regeneration in the CNS. Nat. Rev. Neurosci. 2005, 6, 931–944, doi:10.1038/nrn1807.
[105]  Rosenberg, G.A.; Kornfeld, M.; Estrada, E.; Kelley, R.O.; Liotta, L.A.; Stetler-Stevenson, W.G. TIMP-2 reduces proteolytic opening of blood-brain barrier by type IV collagenase. Brain Res. 1992, 576, 203–207, doi:10.1016/0006-8993(92)90681-X.
[106]  Wilson, E.H.; Weninger, W.; Hunter, C.A. Trafficking of immune cells in the central nervous system. J. Clin. Invest. 2010, 120, 1368–1379, doi:10.1172/JCI41911.
[107]  Griffiths, M.J.; Shafi, M.J.; Popper, S.J.; Hemingway, C.A.; Kortok, M.M.; Wathen, A.; Rockett, K.A.; Mott, R.; Levin, M.; Newton, C.R.; Marsh, K.; Relman, D.A.; Kwiatkowski, D.P. Genomewide analysis of the host response to malaria in Kenyan children. J. Infect. Dis. 2005, 191, 1599–1611.
[108]  Dietmann, A.; Helbok, R.; Lackner, P.; Issifou, S.; Lell, B.; Matsiegui, P.B.; Reindl, M.; Schmutzhard, E.; Kremsner, P.G. Matrix metalloproteinases and their tissue inhibitors (TIMPs) in Plasmodium falciparum malaria: serum levels of TIMP-1 are associated with disease severity. J. Infect. Dis. 2008, 197, 1614–1620, doi:10.1086/587943.
[109]  Mun-Bryce, S.; Rosenberg, G.A. Gelatinase B modulates selective opening of the blood-brain barrier during inflammation. Am. J. Physiol. 1998, 274, R1203–R1211.
[110]  Muroski, M.E.; Roycik, M.D.; Newcomer, R.G.; Van den Steen, P.E.; Opdenakker, G.; Monroe, H.R.; Sahab, Z.J.; Sang, Q.X. Matrix metalloproteinase-9/gelatinase B is a putative therapeutic target of chronic obstructive pulmonary disease and multiple sclerosis. Curr. Pharm. Biotechnol. 2008, 9, 34–46, doi:10.2174/138920108783497631.
[111]  Prato, M.; D'Alessandro, S.; Van den Steen, P.E.; Opdenakker, G.; Arese, P.; Taramelli, D.; Basilico, M. Natural haemozoin modulates matrix metalloproteinases and induces morphological changes in human microvascular endothelium. Cell Microbiol. 2011, 13, 1275–1285, doi:10.1111/j.1462-5822.2011.01620.x.
[112]  Deininger, M.H.; Winkler, S.; Kremsner, P.G.; Meyermann, R.; Schluesener, H.J. Angiogenic proteins in brains of patients who died with cerebral malaria. J. Neuroimmunol. 2003, 142, 101–111, doi:10.1016/S0165-5728(03)00250-9.
[113]  Deininger, M.H.; Kremsner, P.G.; Meyermann, R.; Schluesener, H. Macrophages/microglial cells in patients with cerebral malaria. Eur. Cytokine Netw. 2002, 13, 173–185.
[114]  Kristensson, K.; Nyg?rd, M.; Bertini, G.; Bentivoglio, M. African trypanosome infections of the nervous system: parasite entry and effects on sleep and synaptic functions. Prog. Neurobiol. 2010, 91, 152–171, doi:10.1016/j.pneurobio.2009.12.001.
[115]  Matthews, K.R.; Gull, K. Cycles within cycles: the interplay between differentiation and cell division in Trypanosoma brucei. Parasitol. Today. 1994, 10, 473–476, doi:10.1016/0169-4758(94)90159-7.
[116]  Enanga, B.; Burchmore, R. J.; Stewart, M.L.; Barrett, M.P. Sleeping sickness and the brain. Cell. Mol. Life Sci. 2002, 59, 845–858, doi:10.1007/s00018-002-8472-0.
[117]  Grab, D. J.; Kennedy, P.G. Traversal of human and animal trypanosomes across the blood-brain barrier. J. Neurovirol. 2008, 14, 344–351, doi:10.1080/13550280802282934.
[118]  Hainard, A.; Tiberti, N.; Robin, X.; Ngoyi, D.M.; Matovu, E.; Enyaru, J.C.; Müller, M.; Turck, N.; Ndung'u, J.M.; Lejon, V.; Sanchez, J.C. Matrix metalloproteinase-9 and intercellular adhesion molecule 1 are powerful staging markers for human African trypanosomiasis. Trop. Med. Int. Health. 2011, 16, 119–126, doi:10.1111/j.1365-3156.2010.02642.x.
[119]  Agrawal, S.; Anderson, P.; Durbeej, M.; van Rooijen, N.; Ivars, F.; Opdenakker, G.; Sorokin, L.M. Dystroglycan is selectively cleaved at the parenchymal basement membrane at sites of leukocyte extravasation in experimental autoimmune encephalomyelitis. J. Exp. Med. 2006, 203, 1007–1019, doi:10.1084/jem.20051342.
[120]  Masocha, W.; Rottenberg, M.E.; Kristensson, K. Minocycline impedes African trypanosome invasion of the brain in a murine model. Antimicrob. Agents Chemother. 2006, 50, 1798–1804, doi:10.1128/AAC.50.5.1798-1804.2006.
[121]  Darcy, F.; Santoro, F. Toxoplasmosis. In Parasitic Infections and the Immune System; Kierszenbaum, F., Ed.; Academic Press: Waltham, MA, USA, 1994; pp. 163–201.
[122]  Wong, S.Y.; Remington, J.S. Biology of Toxoplasma gondii. AIDS 1993, 7, 299–316, doi:10.1097/00002030-199303000-00001.
[123]  Gazzinelli, R.; Xu, Y.; Hieny, S.; Cheever, A.; Sher, A. Simultaneous depletion of CD4+ and CD8+ T lymphocytes is required to reactivate chronic infection with Toxoplasma gondii. J. Immunol. 1992, 149, 175–180.
[124]  Strack, A.; Asensio, V.C.; Campbell, I.L.; Schluter, D.; Deckert, M. Chemokines are differentially expressed by astrocytes, microglia and inflammatory leukocytes in Toxoplasma encephalitis and critically regulated by interferon-gamma. Acta Neuropathol. 2002, 103, 458–468, doi:10.1007/s00401-001-0491-7.
[125]  Clark, R.T.; Nance, J.P.; Noor, S.; Wilson, E.H. T cell production of matrix metalloproteases and inhibition of parasite clearance by TIMP-1 during chronic toxoplasma infection in the brain. ASN Neurol. 2010, 3, 1–12.
[126]  García, H.H.; Gonzalez, A.E.; Evans, C.A.; Gilman, R.H. Cysticercosis Working Group in Peru. Taenia solium cysticercosis. Lancet 2003, 362, 547–556.
[127]  Sciutto, E.; Fragoso, G.; Fleury, A.; Laclette, J.P.; Sotelo, J.; Aluja, A.; Vargas, L.; Larralde, C. Taenia solium disease in humans and pigs:an ancient parasitosis disease rooted in developing countries and emerging as a major health problem of global dimensions. Microbes Infect. 2000, 2, 1875–1890, doi:10.1016/S1286-4579(00)01336-8.
[128]  Alvarez, J.I.; Teale, J.M. Multiple expression of matrix metalloproteinases in murine neurocysticercosis: implications for leukocyte migration through multiple central nervous system barriers. Brain Res. 2008, 1214, 145–158, doi:10.1016/j.brainres.2008.03.036.
[129]  Verma, A.; Prasad, K.N.; Nyati, K.K.; Singh, S.K.; Singh, A.K.; Paliwal, V.K.; Gupta, R.K. Association of MMP-2 and MMP-9 with clinical outcome of neurocysticercosis. Parasitol. 2011, 138, 1423–1428, doi:10.1017/S0031182011001259.
[130]  Heuser, K.; Hoddevik, E.H.; Taub?ll, E.; Gjerstad, L.; Indahl, U.; Kaczmarek, L.; Berg, P.R.; Lien, S.; Nagelhus, E.A.; Ottersen, O.P. Temporal lobe epilepsy and matrix metalloproteinase 9: a tempting relation but negative genetic association. Seizure 2010, 19, 335–338, doi:10.1016/j.seizure.2010.05.003.
[131]  Yin, P.; Yang, L.; Zhou, H.Y.; Sun, R.P. Matrix metalloproteinase-9 may be a potential therapeutic target in epilepsy. Med. Hypotheses 2011, 76, 184–186, doi:10.1016/j.mehy.2010.09.013.
[132]  Tsai, H.C.; Chung, L.Y.; Chen, E.R.; Liu, Y.C.; Lee, S.S.J.; Chen, Y.S.; Sy, C.L.; Wann, S.R.; Yen, C.M. Association of Matrix Metalloproteinase-9 and Tissue Inhibitors of Metalloproteinase-4 in Cerebrospinal Fluid with Blood-Brain Barrier Dysfunction in Patients with Eosinophilic Meningitis Caused by Angiostrongylus cantonensis. Am. J. Trop. Med. Hyg. 2008, 78, 20–27.
[133]  Wang, Q.P.; Wu, Z.D.; Wei, J.; Owen, R.L.; Lun, Z.R. Human Angiostrongylus cantonensis: an update. Eur. J. Clin. Microbiol. Infect. Dis. 2012, 31, 389–395, doi:10.1007/s10096-011-1328-5.
[134]  Nishimura, K.; Hung, T. Current views on geographic distribution and modes of infection of neurohelminthic diseases. J. Neurol Sci. 1997, 145, 5–14, doi:10.1016/S0022-510X(96)00293-6.
[135]  Hsu, W.Y.; Chen, J.Y.; Chien, C.T.; Chi, C.S.; Han, N.T. Eosinophilic meningitis caused by Angiostrongylus cantonensis. Pediatr. Infect. Dis. J. 1990, 9, 443–445, doi:10.1097/00006454-199006000-00016.
[136]  Sasaki, O.; Sugaya, H.; Ishida, K.; Yoshimura, K. Ablation of eosinophils with anti-IL-5 antibody enhances the survival of intracranial worms of Angiostrongylus cantonensis in the mouse. Parasite Immunol. 1993, 15, 349–354, doi:10.1111/j.1365-3024.1993.tb00619.x.
[137]  Sugaya, H.; Yoshimura, K. T-cell-dependent eosinophilia in the cerebrospinal fluid of the mouse infected with Angiostrongylus cantonensis. Parasite Immunol. 1998, 10, 127–138, doi:10.1111/j.1365-3024.1988.tb00209.x.
[138]  Lai, S.C.; Jiang, S.T.; Chen, K.M.; Lee, H.H. Matrix metalloproteinases activity demonstrated in the infective stage of the nematodes, Angiostrongylus cantonensis. Parasitol. Res. 2005, 97, 466–471.
[139]  Lee, H.H.; Chou, H.L.; Chen, K.M.; Lai, S.C. Association of matrix metalloproteinase-9 in eosinophilic meningitis of BALB/c mice caused by Angiostrongylus cantonensis. Parasitol. Res. 2004, 94, 321–328, doi:10.1007/s00436-004-1196-3.
[140]  Tseng, Y.K.; Tu, W.C.; Lee, H.H.; Chen, K.M.; Chou, H.L.; Lai, S.C. Ultrastructural localization of matrix metalloproteinase-9 in eosinophils from the cerebrospinal fluid of mice with eosinophilic meningitis caused by Angiostrongylus cantonensis. Ann. Trop. Med. Parasitol. 2004, 98, 831–841, doi:10.1179/000349804X3199.
[141]  Lai, S.C.; Twu, J.J.; Jiang, S.T.; Hsu, J.D.; Chen, K.M.; Chiaing, H.C.; Wang, C.J.; Tseng, C.K.; Shyu, L.Y.; Lee, H.H. Induction of matrix metalloproteinase-9 in murine eosinophilic meningitis caused by Angiostrongylus cantonensis. Ann. Trop. Med. Parasitol. 2004, 98, 715–724, doi:10.1179/000349804225021479.
[142]  Chen, K.M.; Lee, H.H.; Lu, K.H.; Tseng, Y.K.; Hsu, L.S.; Chou, H.L.; Lai, S.C. Association of matrix metalloproteinase-9 and Purkinje cell degeneration in mouse cerebellum caused by Angiostrongylus cantonensis. Int. J. Parasitol. 2004, 34, 1147–1156, doi:10.1016/j.ijpara.2004.07.004.
[143]  Chen, K.M.; Lee, H.H.; Chou, H.L.; Liu, J.Y.; Tsai, B.; Lai, S.C. Upregulation of MMP-9/TIMP-1 enzymatic system in eosinophilic meningitis caused by Angiostrongylus cantonensis. Int. J. Exp. Pathol. 2005, 86, 81–89, doi:10.1111/j.0959-9673.2005.00413.x.
[144]  Chen, K.M.; Liu, J.Y.; Lai, S.C.; Hsu, L.S.; Lee, H.H. Association of plasminogen activators and matrix metalloproteinase-9 proteolytic cascade with blood-CNS barrier damage of angiostrongyliasis. Int. J. Exp. Path. 2006, 87, 113–119, doi:10.1111/j.0959-9673.2006.00459.x.
[145]  Wei, P.C.; Tsai, C.H.; Chiu, P.S.; Lai, S.C. Matrix metalloproteinase-12 leads to elastin degradation in BALB/c mice with eosinophilic meningitis caused by Angiostrongylus cantonensis. Int. J. Parasitol. 2011, 41, 1175–1183, doi:10.1016/j.ijpara.2011.07.002.

Full-Text

comments powered by Disqus

Contact Us

service@oalib.com

QQ:3279437679

WhatsApp +8615387084133

WeChat 1538708413