Lactoferrin in Aseptic and Septic Inflammation
Abstract
:1. Iron and Inflammation
2. Lactoferrin
3. Lactoferrin Against Aseptic Inflammation
3.1. Lactoferrin Against Anemia of Inflammation
3.2. Lactoferrin Against Inflammation-Related Preterm Delivery
3.3. Lactoferrin Against Inflammation in Alzheimer’s Disease
3.4. Lactoferrin Against Inflammation in Type 2 Diabetes
4. Lactoferrin Against Septic Inflammation
4.1. Lactoferrin Against Inflammation Related to Chlamydia trachomatis Infection
4.2. Lactoferrin Against Inflammation Related to Cystic Fibrosis Lung Infection
5. Lactoferrin against Aseptic and Septic Inflammatory Bowel Disease
6. Conflicting Data on Lactoferrin Anti-Inflammatory Activity in In Vitro versus In Vivo Models
7. Conclusions
Author Contributions
Funding
Acknowledgments
Conflicts of Interest
References
- Ma, Y.; Abbate, V.; Hider, R.C. Iron-sensitive fluorescent probes: Monitoring intracellular iron pools. Metallomics 2015, 7, 212–222. [Google Scholar] [CrossRef] [PubMed]
- Andrews, N.C. Disorders of iron metabolism. N. Engl. J. Med. 2000, 341, 1986–1995. [Google Scholar] [CrossRef] [PubMed]
- Valenti, P.; Antonini, G. Lactoferrin: An important host defence against microbial and viral attack. Cell. Mol. Life Sci. 2005, 62, 2576–2587. [Google Scholar] [CrossRef] [PubMed]
- Bonaccorsi di Patti, M.C.; Cutone, A.; Polticelli, F.; Rosa, L.; Lepanto, M.S.; Valenti, P.; Musci, G. The ferroportin-ceruloplasmin system and the mammalian iron homeostasis machine: Regulatory pathways and the role of lactoferrin. BioMetals 2018, 31, 399–414. [Google Scholar] [CrossRef]
- Paesano, R.; Natalizi, T.; Berlutti, F.; Valenti, P. Body iron delocalization: The serious drawback in iron disorders in both developing and developed countries. Pathog. Glob. Health 2012, 106, 200–216. [Google Scholar] [CrossRef] [PubMed]
- Ganz, T. Iron and infection. Int. J. Hematol. 2018, 107, 7–15. [Google Scholar] [CrossRef] [PubMed]
- Donovan, A.; Lima, C.A.; Pinkus, J.L.; Pinkus, G.S.; Zon, L.I.; Robine, S.; Andrews, N.C. The iron exporter ferroportin/Slc40a1 is essential for iron homeostasis. Cell Metab. 2005, 1, 191–200. [Google Scholar] [CrossRef] [PubMed]
- Brissot, P.; Troadec, M.B.; Loréal, O.; Brissot, E. Pathophysiology and classification of iron overload diseases; update 2018. Transfus. Clin. Biol. 2019, 26, 80–88. [Google Scholar] [CrossRef] [PubMed]
- Kono, S.; Yoshida, K.; Tomosugi, N.; Terada, T.; Hamaya, Y.; Kanaoka, S.; Miyajima, H. Biological effects of mutant ceruloplasmin on hepcidin-mediated internalization of ferroportin. Biochim. Biophys. Acta 2010, 1802, 968–975. [Google Scholar] [CrossRef] [PubMed]
- Persichini, T.; De Francesco, G.; Capone, C.; Cutone, A.; di Patti, M.C.; Colasanti, M.; Musci, G. Reactive oxygen species are involved in ferroportin degradation induced by ceruloplasmin mutant Arg701Trp. Neurochem. Int. 2012, 60, 360–364. [Google Scholar] [CrossRef]
- Krause, A.; Neitz, S.; Mägert, H.J.; Schulz, A.; Forssmann, W.G.; Schulz-Knappe, P.; Adermann, K. LEAP-1, a novel highly disulfide-bonded human peptide, exhibits antimicrobial activity. FEBS Lett. 2000, 1480, 147–150. [Google Scholar] [CrossRef]
- Park, C.H.; Valore, E.V.; Waring, A.J.; Ganz, T. Hepcidin, a urinary antimicrobial peptide synthesized in the liver. J. Biol. Chem. 2001, 276, 7806–7810. [Google Scholar] [CrossRef] [PubMed]
- Hunter, H.N.; Fulton, D.B.; Ganz, T.; Vogel, H.J. The solution structure of human hepcidin, a peptide hormone with antimicrobial activity that is involved in iron uptake and hereditary hemochromatosis. J. Biol. Chem. 2002, 277, 37597–37603. [Google Scholar] [CrossRef]
- Qiao, B.; Sugianto, P.; Fung, E.; Del-Castillo-Rueda, A.; Moran-Jimenez, M.J.; Ganz, T.; Nemeth, E. Hepcidin-induced endocytosis of ferroportin is dependent on ferroportin ubiquitination. Cell Metab. 2012, 15, 918–924. [Google Scholar] [CrossRef]
- Fisher, A.L.; Nemeth, E. Iron homeostasis during pregnancy. Am. J. Clin. Nutr. 2017, 106, 1567S–1574S. [Google Scholar] [CrossRef]
- Casu, C.; Nemeth, E.; Rivella, S. Hepcidin agonists as therapeutic tools. Blood 2018, 131, 1790–1794. [Google Scholar] [CrossRef] [PubMed]
- Nemeth, E.; Ganz, T. Anemia of inflammation. Hematol. Oncol. Clin. N. Am. 2014, 28, 671–681. [Google Scholar] [CrossRef] [PubMed]
- Coffey, R.; Ganz, T. Iron homeostasis: An anthropocentric perspective. J. Biol. Chem. 2017, 292, 12727–12734. [Google Scholar] [CrossRef] [PubMed]
- Cutone, A.; Frioni, A.; Berlutti, F.; Valenti, P.; Musci, G.; Bonaccorsi di Patti, M.C. Lactoferrin prevents LPS-induced decrease of the iron exporter ferroportin in human monocytes/macrophages. BioMetals 2014, 27, 807–813. [Google Scholar] [CrossRef]
- Cutone, A.; Rosa, L.; Lepanto, M.S.; Scotti, M.J.; Berlutti, F.; Bonaccorsi di Patti, M.C.; Musci, G.; Valenti, P. Lactoferrin efficiently counteracts the inflammation-induced changes of the iron homeostasis system in macrophages. Front. Immunol. 2017, 15. [Google Scholar] [CrossRef]
- Ludwiczek, S.; Aigner, E.; Theurl, I.; Weiss, G. Cytokine-mediated regulation of iron transport in human monocytic cells. Blood 2003, 101, 4148–4154. [Google Scholar] [CrossRef] [PubMed]
- Weinstein, D.A.; Roy, C.N.; Fleming, M.D.; Loda, M.F.; Wolfsdorf, J.I.; Andrews, N.C. Inappropriate expression of hepcidin is associated with iron refractory anemia: Implications for the anemia of chronic disease. Blood 2002, 100, 3776–3781. [Google Scholar] [CrossRef] [PubMed]
- Lepanto, M.S.; Rosa, L.; Cutone, A.; Conte, M.P.; Paesano, R.; Valenti, P. Efficacy of Lactoferrin Oral Administration in the Treatment of Anemia and Anemia of Inflammation in Pregnant and Non-pregnant Women: An Interventional Study. Front. Immunol. 2018, 9. [Google Scholar] [CrossRef]
- McDonald, B.; Pittman, K.; Menezes, G.B.; Hirota, S.A.; Slaba, I.; Waterhouse, C.C.; Beck, P.L.; Muruve, D.A.; Kubes, P. Intravascular danger signals guide neutrophils to sites of sterile inflammation. Science 2010, 330, 362–366. [Google Scholar] [CrossRef] [PubMed]
- Kruzel, M.L.; Zimecki, M.; Actor, J.K. Lactoferrin in a Context of Inflammation-Induced Pathology. Front. Immunol. 2017, 8, 1438. [Google Scholar] [CrossRef]
- Johansson, B. Isolation of an iron-containing red protein from human milk. Acta Chem. Scand. 1960, 14, 510–512. [Google Scholar] [CrossRef]
- Montreuil, J.; Tonnelat, J.; Mullet, S. Preparation and properties of lactosiderophilin (lactotransferrin) of human milk. Biochim. Biophys. Acta 1960, 45, 413–421. [Google Scholar] [CrossRef]
- Groves, M.L. The isolation of a red protein from milk. J. Am. Chem. Soc. 1960, 82, 3345–3350. [Google Scholar] [CrossRef]
- Baker, E.N. Structure and reactivity of transferrins. Adv. Inorg. Chem. 1994, 41, 389–463. [Google Scholar]
- Anderson, B.F.; Baker, H.M.; Norris, G.E.; Rumball, S.V.; Baker, E.N. Apolactoferrin structure demonstrates ligand-induced conformational change in transferrins. Nature 1990, 344, 784–787. [Google Scholar] [CrossRef]
- Moore, S.A.; Anderson, B.F.; Groom, C.R.; Haridas, M.; Baker, E.N. Three-dimensional structure of diferric bovine lactoferrin at 2.8 A resolution. J. Mol. Biol. 1997, 274, 222–236. [Google Scholar] [CrossRef]
- Baker, E.N.; Rumball, S.V.; Anderson, B.F. Transferrins: Insights into structure and function from studies on lactoferrin. Trends Biochem. Sci. 1987, 12, 350–353. [Google Scholar] [CrossRef]
- Bruns, C.M.; Nowalk, A.J.; Arvai, A.S.; McTigue, M.A.; Vaughan, K.G.; Mietzner, T.A.; McRee, D.E. Structure of Haemophilus influenzae Fe(+3)-binding protein reveals convergent evolution within a superfamily. Nat. Struct. Biol. 1997, 4, 919–924. [Google Scholar] [CrossRef]
- Gerstein, M.; Anderson, B.F.; Norris, G.E.; Baker, E.N.; Lesk, A.M.; Chothia, C. Domain closure in lactoferrin. Two hinges produce a see-saw motion between alternative close-packed interfaces. J. Mol. Biol. 1993, 234, 357–372. [Google Scholar] [CrossRef]
- Castillo, E.; Pérez, M.D.; Franco, I.; Calvo, M.; Sánchez, L. Kinetic and thermodynamic parameters for heat denaturation of human recombinant lactoferrin from rice. Biochem. Cell Biol. 2012, 90, 389–396. [Google Scholar] [CrossRef]
- Rosa, L.; Cutone, A.; Lepanto, M.S.; Scotti, M.J.; Conte, M.P.; Paesano, R.; Valenti, P. Physico-chemical properties influence the functions and efficacy of commercial bovine lactoferrins. BioMetals 2018, 31, 301–312. [Google Scholar] [CrossRef]
- Baker, H.M.; Baker, E.N. A structural perspective on lactoferrin function. Biochem. Cell Biol. 2012, 90, 320–328. [Google Scholar] [CrossRef]
- Berlutti, F.; Pantanella, F.; Natalizi, T.; Frioni, A.; Paesano, R.; Polimeni, A.; Valenti, P. Antiviral properties of lactoferrin: A natural immunity molecule. Molecules 2011, 16, 6992–7018. [Google Scholar] [CrossRef]
- Jiang, R.; Lopez, V.; Kelleher, S.L.; Lönnerdal, B. Apo- and holo-lactoferrin are both internalized by lactoferrin receptor via clathrin-mediated endocytosis but differentially affect ERK-signaling and cell proliferation in Caco-2 cells. J. Cell Physiol. 2011, 226, 3022–3031. [Google Scholar] [CrossRef]
- Bellamy, W.; Takase, M.; Wakabayashi, H.; Kavase, K.; Tomita, M. Antibacterial spectrum of lactoferricin B, a potent bactericide peptide derived from the N-terminal region of bovine lactoferrin. J. Appl. Bacteriol. 1992, 73, 472–479. [Google Scholar] [CrossRef]
- Van der Kraan, M.I.; Groenink, J.; Nazmi, K.; Veerman, E.C.; Bolscher, J.G. Nieuw Amerongen AV. Lactoferrampin: A novel antimicrobial peptide in the N1-domain of bovine lactoferrin. Peptides 2004, 25, 177–183. [Google Scholar] [CrossRef] [PubMed]
- Karav, S.; German, J.B.; Rouquié, C.; Le Parc, A.; Barile, D. Studying Lactoferrin N-Glycosylation. Int. J. Mol. Sci. 2017, 18, 870. [Google Scholar] [CrossRef] [PubMed]
- Baker, E.N.; Baker, H.M. A structural framework for understanding the multifunctional character of lactoferrin. Biochimie 2009, 91, 3–10. [Google Scholar] [CrossRef]
- Haridas, M.; Anderson, B.F.; Baker, E.N. Structure of human diferric lactoferrin refined at 2.2 A resolution. Acta Crystallogr. D Biol. Crystallogr. 1995, 51, 629–646. [Google Scholar] [CrossRef]
- Van Veen, H.A.; Geerts, M.E.; van Berkel, P.H.; Nuijens, J.H. The role of N-linked glycosylation in the protection of human and bovine lactoferrin against tryptic proteolysis. Eur. J. Biochem. 2004, 271, 678–684. [Google Scholar] [CrossRef]
- Barboza, M.; Pinzon, J.; Wickramasinghe, S.; Froehlich, J.W.; Moeller, I.; Smilowitz, J.T.; Ruhaak, L.R.; Huang, J.; Lönnerdal, B.; German, J.B. Glycosylation of human milk lactoferrin exhibits dynamic changes during early lactation enhancing its role in pathogenic bacteria-host interactions. Mol. Cell Proteom. 2012, 11, M111.015248. [Google Scholar] [CrossRef] [PubMed]
- Masson, P.L.; Heremans, J.F.; Dive, C. An iron-binding protein common to many external secretions. Clin. Chim. Acta 1966, 14, 735–739. [Google Scholar] [CrossRef]
- Masson, P.L.; Heremans, J.F.; Schonne, E. Lactoferrin, an iron-binding protein in neutrophilic leukocytes. J. Exp. Med. 1969, 130, 643–658. [Google Scholar] [CrossRef]
- Alexander, D.B.; Iigo, M.; Yamauchi, K.; Suzui, M.; Tsuda, H. Lactoferrin: An alternative view of its role in human biological fluids. Biochem. Cell Biol. 2012, 90, 279–306. [Google Scholar] [CrossRef]
- Rosa, L.; Cutone, A.; Lepanto, M.S.; Paesano, R.; Valenti, P. Lactoferrin: A natural glycoprotein involved in iron and inflammatory homeostasis. Int. J. Mol. Sci. 2017, 18, 1985. [Google Scholar] [CrossRef]
- Rossi, P.; Giansanti, F.; Boffi, A.; Ajello, M.; Valenti, P.; Chiancone, E.; Antonini, G. Ca2+ binding to bovine lactoferrin enhances protein stability and influences the release of bacterial lipopolysaccharide. Biochem. Cell Biol. 2002, 80, 41–48. [Google Scholar] [CrossRef]
- Legrand, D. Lactoferrin, a key molecule in immune and inflammatory processes. Biochem. Cell Biol. 2012, 90, 252–268. [Google Scholar] [CrossRef]
- Ward, P.P.; Paz, E.; Conneely, O.M. Multifunctional roles of lactoferrin: A critical overview. Cell. Mol. Life Sci. 2005, 62, 2540–2548. [Google Scholar] [CrossRef] [PubMed]
- Lang, J.; Yang, N.; Deng, J.; Liu, K.; Yang, P.; Zhang, G.; Jiang, C. Inhibition of SARS pseudovirus cell entry by lactoferrin binding to heparan sulfate proteoglycans. PLoS ONE 2011, 6, e23710. [Google Scholar] [CrossRef] [PubMed]
- Albar, A.H.; Almehdar, H.A.; Uversky, V.N.; Redwan, E.M. Structural heterogeneity and multifunctionality of lactoferrin. Curr. Protein Pept. Sci. 2014, 15, 778–797. [Google Scholar] [CrossRef] [PubMed]
- Legrand, D.; Vigié, K.; Said, E.A.; Elass, E.; Masson, M.; Slomianny, M.C.; Carpentier, M.; Briand, J.P.; Mazurier, J.; Hovanessian, A.G. Surface nucleolin participates in both the binding and endocytosis of lactoferrin in target cells. Eur. J. Biochem. 2004, 271, 303–317. [Google Scholar] [CrossRef] [PubMed]
- Suzuki, Y.A.; Lopez, V.; Lönnerdal, B. Mammalian lactoferrin receptors: Structure and function. Cell. Mol. Life Sci. 2005, 62, 2560–2575. [Google Scholar] [CrossRef]
- Suzuki, Y.A.; Wong, H.; Ashida, K.Y.; Schryvers, A.B.; Lönnerdal, B. The N1 domain of human lactoferrin is required for internalization by Caco-2 cells and targeting to the nucleus. Biochemistry 2008, 47, 10915–10920. [Google Scholar] [CrossRef] [PubMed]
- Wrackmeyer, U.; Hansen, G.H.; Seya, T.; Danielsen, E.M. Intelectin: A novel lipid raft-associated protein in the enterocyte brush border. Biochemistry 2006, 45, 9188–9197. [Google Scholar] [CrossRef]
- Mancinelli, R.; Olivero, F.; Carpino, G.; Overi, D.; Rosa, L.; Lepanto, M.S.; Cutone, A.; Franchitto, A.; Alpini, G.; Onori, P.; et al. Role of lactoferrin and its receptors on biliary epithelium. Biometals 2018, 31, 369–379. [Google Scholar] [CrossRef] [PubMed]
- Ashida, K.; Sasaki, H.; Suzuki, Y.A.; Lönnerdal, B. Cellular internalization of lactoferrin in intestinal epithelial cells. Biometals 2004, 17, 311–315. [Google Scholar] [CrossRef] [PubMed]
- Liao, Y.; Lopez, V.; Shafizadeh, T.B.; Halsted, C.H.; Lönnerdal, B. Cloning of a pig homologue of the human lactoferrin receptor: Expression and localization during intestinal maturation in piglets. Comp. Biochem. Physiol. A Mol. Integr. Physiol. 2007, 148, 584–590. [Google Scholar] [CrossRef]
- Legrand, D. Overview of Lactoferrin as a Natural Immune Modulator. J. Pediatr. 2016, 173. [Google Scholar] [CrossRef]
- Losfeld, M.E.; Khoury, D.E.; Mariot, P.; Carpentier, M.; Krust, B.; Briand, J.P.; Mazurier, J.; Hovanessian, A.G.; Legrand, D. The cell surface expressed nucleolin is a glycoprotein that triggers calcium entry into mammalian cells. Exp. Cell Res. 2009, 315, 357–369. [Google Scholar] [CrossRef] [PubMed]
- Liao, Y.; Jiang, R.; Lönnerdal, B. Biochemical and molecular impacts of lactoferrin on small intestinal growth and development during early life. Biochem. Cell Biol. 2012, 90, 476–484. [Google Scholar] [CrossRef] [PubMed]
- Kitagawa, H.; Yoshizawa, Y.; Yokoyama, T.; Takeuchi, T.; Talukder, M.J.; Shimizu, H.; Ando, K.; Harada, E. Persorption of bovine lactoferrin from the intestinal lumen into the systemic circulation via the portal vein and the mesenteric lymphatics in growing pigs. J. Vet. Med. Sci. 2003, 65, 567–572. [Google Scholar] [CrossRef]
- Fischer, R.; Debbabi, H.; Blais, A.; Dubarry, M.; Rautureau, M.; Boyaka, P.N.; Tome, D. Uptake of ingested bovine lactoferrin and its accumulation in adult mouse tissues. Int. Immunopharmacol. 2007, 7, 1387–1393. [Google Scholar] [CrossRef] [PubMed]
- Pisetsky, D. Cell death in the pathogenesis of immune-mediated diseases: The role of HMGB1 and DAMP-PAMP complexes. Swiss Med. Wkly. 2011, 141, w13256. [Google Scholar] [CrossRef] [PubMed]
- Shin, K.; Wakabayashi, H.; Yamauchi, K.; Yaeshima, T.; Iwatsuki, K. Recombinant human intelectin binds bovine lactoferrin and its peptides. Biol. Pharm. Bull. 2008, 31, 1605–1608. [Google Scholar] [CrossRef]
- Chung, E.Y.; Kim, S.; Ma, X.J. Regulation of cytokine production during phagocytosis of apoptotic cells. Cell Res. 2006, 16, 154–161. [Google Scholar] [CrossRef] [PubMed]
- Matzinger, P. Tolerance, danger, and the extended family. Annu. Rev. Immunol. 1994, 12, 991–1045. [Google Scholar] [CrossRef]
- Valenti, P.; Greco, R.; Pitari, G.; Rossi, P.; Ajello, M.; Melino, G.; Antonini, G. Apoptosis of Caco-2 intestinal cells invaded by Listeria monocytogenes: Protective effect of lactoferrin. Exp. Cell Res. 1999, 250, 197–202. [Google Scholar] [CrossRef]
- Blais, A.; Fan, C.; Voisin, T.; Aattouri, N.; Dubarry, M.; Blachier, F.; Tomé, D. Effects of lactoferrin on intestinal epithelial cell growth and differentiation: An in vivo and in vitro study. Biometals 2014, 27, 857–874. [Google Scholar] [CrossRef]
- Wessling-Resnick, M. Iron homeostasis and the inflammatory response. Annu. Rev. Nutr. 2010, 30, 105–122. [Google Scholar] [CrossRef]
- Anker, S.D.; Comin, C.J.; Filippatos, G.; Willenheimer, R.; Dickstein, K.; Drexler, H.; Lüscher, T.F.; Bart, B.; Banasiak, W.; Niegowska, J.; et al. Ferric carboxymaltose in patients with heart failure and iron deficiency. N. Engl. J. Med. 2009, 361, 2436–2448. [Google Scholar] [CrossRef] [PubMed]
- Tim Goodnough, L.; Comin-Colet, J.; Leal-Noval, S.; Ozawa, S.; Takere, J.; Henry, D.; Javidroozi, M.; Hohmuth, B.; Bisbe, E.; Gross, I.; et al. Management of anemia in patients with congestive heart failure. Am. J. Hematol. 2017, 92, 88–93. [Google Scholar] [CrossRef] [PubMed]
- Weiss, G.; Ganz, T.; Goodnough, L.T. Anemia of inflammation. Blood 2019, 133, 40–50. [Google Scholar] [CrossRef]
- Frazer, D.M.; Anderson, G.J. The orchestration of body iron intake: How and where do enterocytes receive their cues? Blood Cells Mol. Dis. 2003, 30, 288–297. [Google Scholar] [CrossRef]
- Pettersson, T.; Rosenlöf, K.; Friman, C.; Mickos, A.; Teppo, A.M.; Fyhrquist, F. Successful treatment of the anemia of rheumatoid arthritis with subcutaneously administered recombinant human erythropoietin. Slower response in patients with more severe inflammation. Scand. J. Rheumatol. 1993, 22, 188–193. [Google Scholar] [CrossRef] [PubMed]
- Kato, Y.; Takagi, C.; Tanaka, J.; Masaki, Y.; Furuya, H. Effect of daily subcutaneous administration of recombinant erythropoietin on chronic anemia in rheumatoid arthritis. Intern. Med. 1994, 33, 193–197. [Google Scholar] [CrossRef] [PubMed]
- Peeters, H.R.; Jongen-Lavrencic, M.; Vreugdenhil, G.; Swaak, A.J. Effect of recombinant human erythropoietin on anemia and disease activity in patients with rheumatoid arthritis and anemia of chronic disease: A randomized placebo controlled double blind 52 weeks clinical trial. Ann. Rheum. Dis. 1996, 55, 739–744. [Google Scholar] [CrossRef]
- Paesano, R.; Pacifici, E.; Benedetti, S.; Berlutti, F.; Frioni, A.; Polimeni, A.; Valenti, P. Safety and efficacy of lactoferrin versus ferrous sulphate in curing iron deficiency and iron deficiency anaemia in hereditary thrombophilia pregnant women: An interventional study. Biometals 2014, 27, 999–1006. [Google Scholar] [CrossRef]
- Paesano, R.; Torcia, F.; Berlutti, F.; Pacifici, E.; Ebano, V.; Moscarini, M.; Valenti, P. Oral administration of lactoferrin increases hemoglobin and total serum iron in pregnant women. Biochem. Cell Biol. 2006, 84, 377–380. [Google Scholar] [CrossRef]
- Rosendaal, F.R. Venous thrombosis: Prevalence and interaction of risk factors. Haemostasis 1999, 1, 1–9. [Google Scholar] [CrossRef]
- Khan, S.; Dickerman, J.D. Hereditary thrombophilias. Thromb. J. 2006, 4. [Google Scholar] [CrossRef] [PubMed]
- Szecsi, P.B.; Jørgensen, M.; Klajnbard, A.; Andersen, M.R.; Colov, N.P.; Stender, S. Haemostatic reference intervals in pregnancy. Thromb. Haemost. 2010, 103, 718–727. [Google Scholar] [CrossRef]
- Simcox, L.E.; Ormesher, L.; Tower, C.; Greer, I.A. Thrombophilia and Pregnancy Complications. Int. J. Mol. Sci. 2015, 16, 28418–28428. [Google Scholar] [CrossRef] [PubMed]
- Poredos, P.; Jezovnik, M.K. The role of inflammation in venous thromboembolism and the link between arterial and venous thrombosis. Int. Angiol. 2007, 26, 306–311. [Google Scholar]
- Corna, G.; Campana, L.; Pignatti, E.; Castiglioni, A.; Tagliafico, E.; Bosurgi, L.; Campanella, A.; Brunelli, S.; Manfredi, A.A.; Apostoli, P.; et al. Polarization dictates iron handling by inflammatory and alternatively activated macrophages. Haematologica 2010, 95, 1814–1822. [Google Scholar] [CrossRef]
- Recalcati, S.; Locati, M.; Marini, A.; Santambrogio, P.; Zaninotto, F.; De Pizzoli, M.; Zammataro, L.; Girelli, D.; Cairo, G. Differential regulation of iron homeostasis during human macrophage polarized activation. Eur. J. Immunol. 2010, 40, 824–835. [Google Scholar] [CrossRef]
- Behnia, F.; Sheller, S.; Menon, R. Mechanistic Differences Leading to Infectious and Sterile Inflammation. Am. J. Reprod. Immunol. 2016, 75, 505–518. [Google Scholar] [CrossRef] [PubMed]
- Nadeau-Vallée, M.; Obari, D.; Palacios, J.; Brien, M.È.; Duval, C.; Chemtob, S.; Girard, S. Sterile inflammation and pregnancy complications: A review. Reproduction 2016, 152. [Google Scholar] [CrossRef]
- Hurst, S.M.; Wilkinson, T.S.; McLoughlin, R.M.; Jones, S.; Horiuchi, S.; Yamamoto, N.; Rose-John, S.; Fuller, G.M.; Topley, N.; Jones, S.A. Il-6 and its soluble receptor orchestrate a temporal switch in the pattern of leukocyte recruitment seen during acute inflammation. Immunity 2001, 14, 705–714. [Google Scholar] [CrossRef]
- Saito, S.; Nakashima, A.; Shima, T.; Ito, M. Th1/Th2/Th17 and regulatory T-cell paradigm in pregnancy. Am. J. Reprod. Immunol. 2010, 63, 601–610. [Google Scholar] [CrossRef] [PubMed]
- Scholl, T.O. Iron status during pregnancy: Setting the stage for mother and infant. Am. J. Clin. Nutr. 2005, 81, 1218S–1222S. [Google Scholar] [CrossRef]
- Paesano, R.; Berlutti, F.; Pietropaoli, M.; Goolsbee, W.; Pacifici, E.; Valenti, P. Lactoferrin efficacy versus ferrous sulfate in curing iron disorders in pregnant and non-pregnant women. Int. J. Immunopathol. Pharmacol. 2010, 23, 577–587. [Google Scholar] [CrossRef]
- Wenstrom, K.D.; Andrews, W.W.; Tamura, T.; DuBard, M.B.; Johnston, K.E.; Hemstreet, G.P. Elevated amniotic fluid interleukin-6 levels at genetic amniocentesis predict subsequent pregnancy loss. Am. J. Obstet. Gynecol. 1996, 175, 830–833. [Google Scholar] [CrossRef]
- Bogavac, M.; Brkic, S.; Simin, N.; Celic, D. Mid-pregnancy interleukin levels in serum and amniotic fluid as predictors of preterm delivery. J. Matern. Fetal Neonatal Med. 2012. [Google Scholar] [CrossRef] [PubMed]
- Xiao, J.P.; Yin, Y.X.; Gao, Y.F.; Lau, S.; Shen, F.; Zhao, M.; Chen, Q. The increased maternal serum levels of IL-6 are associated with the severity and onset of preeclampsia. Cytokine 2012, 60, 856–860. [Google Scholar] [CrossRef]
- Locci, M.; Nazzaro, G.; Merenda, A.; Pisaturo, M.L.; Laviscio, P.; Poppiti, R.; Miranda, M.; Stile, A.; De Placido, G. Atosiban vs ritodrine used prophylactically with cerclage in ICSI pregnancies to prevent pre-term birth in women identified as being at high risk on the basis of transvaginal ultrasound scan. J. Obstet. Gynaecol. 2006, 26, 396–401. [Google Scholar] [CrossRef]
- Berghella, V.; Rafael, T.J.; Szychowski, J.M.; Rust, O.A.; Owen, J. Cerclage for short cervix on ultrasonography in women with singleton gestations and previous pre-term birth: A meta-analysis. Obstet. Gynecol. 2011, 117, 663–671. [Google Scholar] [CrossRef]
- Locci, M.; Nazzaro, G.; Miranda, M.; Salzano, E.; Montagnani, S.; Castaldo, C.; De Placido, G. Vaginal lactoferrin in asymptomatic patients at low risk for pre-term labour for shortened cervix: Cervical length and interleukin-6 changes. J. Obstet. Gynaecol. 2013, 33, 144–148. [Google Scholar] [CrossRef]
- Mathews, T.J.; MacDorman, M.F. Infant mortality statistics from the 2007 period linked birth/infant death data set. Natl. Vital. Stat. Rep. 2011, 59, 1–30. [Google Scholar]
- Christiaens, I.; Zaragoza, D.B.; Guilbert, L.; Robertson, S.A.; Mitchell, B.F.; Olson, D.M. Inflammatory processes in preterm and term parturition. J. Reprod. Immunol. 2008, 79, 50–57. [Google Scholar] [CrossRef] [PubMed]
- Estrada-Gutierrez, G.; Gomez-Lopez, N.; Zaga-Clavellina, V.; Giono-Cerezo, S.; Espejel-Nuñez, A.; Gonzalez-Jimenez, M.A.; Espino y Sosa, S.; Olson, D.M.; Vadillo-Ortega, F. Interaction between pathogenic bacteria and intrauterine leukocytes triggers alternative molecular signaling cascades leading to labor in women. Infect. Immun. 2010, 78, 4792–4799. [Google Scholar] [CrossRef] [PubMed]
- Genc, M.R.; Ford, C.E. The clinical use of inflammatory markers during pregnancy. Curr. Opin. Obstet. Gynecol. 2010, 22, 116–121. [Google Scholar] [CrossRef]
- Mitchell, M.D.; Dudley, D.J.; Edwin, S.S.; Schiller, S.L. Interleukin-6 stimulates prostaglandin production by human amnion and decidual cells. Eur. J. Pharmacol. 1991, 192, 189–191. [Google Scholar] [CrossRef]
- Lyon, D.; Cheng, C.Y.; Howland, L.; Rattican, D.; Jallo, N.; Pickler, R.; Brown, L.; McGrath, J. Integrated review of cytokines in maternal, cord, and newborn blood: Part I--associations with preterm birth. Biol. Res. Nurs. 2010, 11, 371–376. [Google Scholar] [CrossRef] [PubMed]
- Gotsch, F.; Gotsch, F.; Romero, R.; Erez, O.; Vaisbuch, E.; Kusanovic, J.P.; Mazaki-Tovi, S.; Kim, S.K.; Hassan, S.; Yeo, L. The preterm parturition syndrome and its implications for understanding the biology, risk assessment, diagnosis, treatment and prevention of preterm birth. J. Matern. Fetal Neonatal Med. 2009, 2, 5–23. [Google Scholar] [CrossRef]
- Vesce, F.; Giugliano, E.; Bignardi, S.; Cagnazzo, E.; Colamussi, C.; Marci, R.; Valente, N.; Seraceni, S.; Maritati, M.; Contini, C. Vaginal lactoferrin administration before genetic amniocentesis decreases amniotic interleukin-6 levels. Gynecol. Obstet. Invest. 2014, 77, 245–249. [Google Scholar] [CrossRef] [PubMed]
- Trentini, A.; Maritati, M.; Cervellati, C.; Manfrinato, M.C.; Gonelli, A.; Volta, C.A.; Vesce, F.; Greco, P.; Dallocchio, F.; Bellini, T.; et al. Vaginal Lactoferrin Modulates PGE2, MMP-9, MMP-2, and TIMP-1 Amniotic Fluid Concentrations. Mediators Inflamm. 2016, 2016. [Google Scholar] [CrossRef] [PubMed]
- Maritati, M.; Comar, M.; Zanotta, N.; Seraceni, S.; Trentini, A.; Corazza, F.; Vesce, F.; Contini, C. Influence of vaginal lactoferrin administration on amniotic fluid cytokines and its role against inflammatory complications of pregnancy. J. Inflamm. (Lond.) 2017, 14. [Google Scholar] [CrossRef]
- Cheng, X.S.; Zhao, K.P.; Jiang, X.; Du, L.L.; Li, X.H.; Ma, Z.W.; Yao, J.; Luo, Y.; Duan, D.X.; Wang, J.Z.; Zhou, X.W. Nmnat2 attenuates Tau phosphorylation through activation of PP2A. J. Alzheimers Dis. 2013, 36, 185–195. [Google Scholar] [CrossRef]
- Ikonomovic, M.D.; Klunk, W.E.; Abrahamson, E.E.; Wuu, J.; Mathis, C.A.; Scheff, S.W.; Mufson, E.J.; DeKosky, S.T. Precuneus amyloid burden is associated with reduced cholinergic activity in Alzheimer disease. Neurology 2011, 77, 39–47. [Google Scholar] [CrossRef]
- Colvez, A.; Joel, M.E.; Ponton-Sanchez, A.; Royer, A.C. Health status and work burden of Alzheimer patients’ informal caregivers: Comparisons of five different care programs in the European Union. Health Policy 2002, 60, 219–233. [Google Scholar] [CrossRef]
- Fuller, S.; Steele, M.; Münch, G. Activated astroglia during chronic inflammation in Alzheimer’s disease—Do they neglect their neurosupportive roles? Mutat. Res. 2010, 690, 40–49. [Google Scholar] [CrossRef]
- Cunningham, C.; Wilcockson, D.C.; Campion, S.; Lunnon, K.; Perry, V.H. Central and systemic endotoxin challenges exacerbate the local inflammatory response and increase neuronal death during chronic neurodegeneration. J. Neurosci. 2005, 25, 9275–9284. [Google Scholar] [CrossRef]
- Dal Prà, I.; Chiarini, A.; Gui, L.; Chakravarthy, B.; Pacchiana, R.; Gardenal, E.; Whitfield, J.F.; Armato, U. Do astrocytes collaborate with neurons in spreading the “infectious” aβ and Tau drivers of Alzheimer’s disease? Neuroscientist. 2015, 2, 9–29. [Google Scholar] [CrossRef]
- Mohamed, W.A.; Salama, R.M.; Schaalan, M.F. A pilot study on the effect of lactoferrin on Alzheimer’s disease pathological sequelae: Impact of the p-Akt/PTEN pathway. Biomed. Pharmacother. 2019, 111, 714–723. [Google Scholar] [CrossRef]
- Zhang, B.; Gaiteri, C.; Bodea, L.G.; Wang, Z.; Mcelwee, J.; Podtelezhnikov, A.A.; Zhang, C.; Xie, T.; Tran, L.; Dobrin, R.; et al. Integrated systems approach identifies genetic nodes and networks in late-onset Alzheimer’s disease. Cell 2013, 153, 707–720. [Google Scholar] [CrossRef]
- Du, L.; Zhao, Z.; Cui, A.; Zhu, Y.; Zhang, L.; Liu, J.; Shi, S.; Fu, C.; Han, X.; Gao, W.; et al. Increased iron deposition on brain quantitative susceptibility mapping correlates with decreased cognitive function in Alzheimer’s disease. ACS Chem. Neurosci. 2018, 9, 1849–1857. [Google Scholar] [CrossRef]
- Liu, J.L.; Fan, Y.G.; Yang, Z.S.; Wang, Z.Y.; Guo, C. Iron and Alzheimer’s Disease: From Pathogenesis to Therapeutic Implications. Front. Neurosci. 2018, 12. [Google Scholar] [CrossRef]
- Gerlach, M.; Ben-Shachar, D.; Riederer, P.; Youdim, M.B. Altered brain metabolism of iron as a cause of neurodegenerative diseases? J. Neurochem. 1994, 63, 793–807. [Google Scholar] [CrossRef] [PubMed]
- Agrawal, M.; Saraf, S.; Saraf, S.; Antimisiaris, S.G.; Hamano, N.; Li, S.D.; Chougule, M.; Shoyele, S.A.; Gupta, U.; Ajazuddin; Alexander, A. Recent advancements in the field of nanotechnology for the delivery of anti-Alzheimer drug in the brain region. Expert Opin. Drug Deliv. 2018, 15, 589–617. [Google Scholar] [CrossRef] [PubMed]
- Meng, Q.; Wang, A.; Hua, H.; Jiang, Y.; Wang, Y.; Mu, H.; Wu, Z.; Sun, K. Intranasal delivery of Huperzine A to the brain using lactoferrin-conjugated N-trimethylated chitosan surface-modified PLGA nanoparticles for treatment of Alzheimer’s disease. Int. J. Nanomed. 2018, 13, 705–718. [Google Scholar] [CrossRef]
- Crapper McLachlan, D.R.; Dalton, A.J.; Kruck, T.P.; Bell, M.Y.; Smith, W.L.; Kalow, W.; Andrews, D.F. Intramuscular desferrioxamine in patients with Alzheimer’s disease. Lancet 1991, 337, 1304–1308. [Google Scholar] [CrossRef]
- Guo, C.; Wang, P.; Zhong, M.L.; Wang, T.; Huang, X.S.; Li, J.Y.; Wang, Z.Y. Deferoxamine inhibits iron induced hippocampal tau phosphorylation in the Alzheimer transgenic mouse brain. Neurochem. Int. 2013, 62, 165–172. [Google Scholar] [CrossRef]
- Guo, C.; Wang, T.; Zheng, W.; Shan, Z.Y.; Teng, W.P.; Wang, Z.Y. Intranasal deferoxamine reverses iron-induced memory deficits and inhibits amyloidogenic APP processing in a transgenic mouse model of Alzheimer’s disease. Neurobiol. Aging 2013, 34, 562–575. [Google Scholar] [CrossRef]
- May, P.M.; Bulman, R.A. The present status of chelating agents in medicine. Prog. Med. Chem. 1983, 20, 225–336. [Google Scholar]
- Cuajungco, M.P.; Faget, K.Y.; Huang, X.; Tanzi, R.E.; Bush, A.I. Metal chelation as a potential therapy for Alzheimer’s disease. Ann. N. Y. Acad. Sci. 2000, 920, 292–304. [Google Scholar] [CrossRef]
- Brown, R.D.; Rickard, K.A.; Kronenberg, H. Lactoferrin in the myeloproliferative disorders: A search for granulopoietic regulator defects. Br. J. Haematol. 1985, 59, 617–626. [Google Scholar] [CrossRef]
- Valverde, F.; Lopez-Mascaraque, L.; De Carlos, J.A. Distribution and morphology of Alz-50-immunoreactive cells in the developing visual cortex of kittens. J. Neurocytol. 1990, 19, 662–671. [Google Scholar] [CrossRef]
- Kawamata, T.; Tooyama, I.; Yamada, T.; Walker, D.G.; Mcgeer, P.L. Lactotransferrin immunocytochemistry in Alzheimer and normal human brain. Am. J. Pathol. 1993, 142, 1574–1585. [Google Scholar]
- Leveugle, B.; Spik, G.; Perl, D.P.; Bouras, C.; Fillit, H.M.; Hof, P.R. The iron-binding protein lactotransferrin is present in pathologic lesions in a variety of neurodegenerative disorders: A comparative immunohistochemical analysis. Brain Res. 1994, 650, 20–31. [Google Scholar] [CrossRef]
- Qian, Z.M.; Wang, Q. Expression of iron transport proteins and excessive iron accumulation in the brain in neurodegenerative disorders. Brain Res. Brain Res. 1998, 27, 257–267. [Google Scholar] [CrossRef]
- Arnold, R.R.; Cole, M.F.; McGhee, J.R. A bacteriocidal effect for human lactoferrin. Science 1977, 197, 263–265. [Google Scholar] [CrossRef]
- Fillebeen, C.; Descamps, L.; Dehouck, M.P.; Fenart, L.; Benaissa, M.; Spik, G.; Cecchelli, R.; Pierce, A. Receptor-mediated transcytosis of lactoferrin through the blood-brain barrier. J. Biol. Chem. 1999, 274, 7011–7017. [Google Scholar] [CrossRef]
- Guo, C.; Yang, Z.H.; Zhang, S.; Chai, R.; Xue, H.; Zhang, Y.H.; Li, J.Y.; Wang, Z.Y. Intranasal Lactoferrin Enhances α-Secretase-Dependent Amyloid Precursor Protein Processing via the ERK1/2-CREB and HIF-1α Pathways in an Alzheimer’s Disease Mouse Model. Neuropsychopharmacology 2017, 42, 2504–2515. [Google Scholar] [CrossRef] [PubMed]
- Hotamisligil, G.S.; Peraldi, P.; Budavari, A.; Ellis, R.; White, M.F.; Spiegelman, B.M. IRS-1-mediated inhibition of insulin receptor tyrosine kinase activity in TNF-alpha- and obesity-induced insulin resistance. Science 1996, 271, 665–668. [Google Scholar] [CrossRef]
- Yin, M.J.; Yamamoto, Y.; Gaynor, R.B. The anti-inflammatory agents aspirin and salicylate inhibit the activity of I(kappa)B kinase-beta. Nature 1998, 396, 77–80. [Google Scholar] [CrossRef] [PubMed]
- Aguirre, V.; Werner, E.D.; Giraud, J.; Lee, Y.H.; Shoelson, S.E.; White, M.F. Phosphorylation of Ser307 in insulin receptor substrate-1 blocks interactions with the insulin receptor and inhibits insulin action. J. Biol. Chem. 2002, 277, 1531–1537. [Google Scholar] [CrossRef]
- Paz, K.; Hemi, R.; LeRoith, D.; Karasik, A.; Elhanany, E.; Kanety, H.; Zick, Y. A molecular basis for insulin resistance. Elevated serine/threonine phosphorylation of IRS-1 and IRS-2 inhibits their binding to the juxtamembrane region of the insulin receptor and impairs their ability to undergo insulin-induced tyrosine phosphorylation. J. Biol. Chem. 1997, 272, 29911–29918. [Google Scholar] [CrossRef]
- Mandrup-Poulsen, T. The role of interleukin-1 in the pathogenesis of IDDM. Diabetologia 1996, 39, 1005–1029. [Google Scholar] [CrossRef]
- Feve, B.; Bastard, J.P. The role of interleukins in insulin resistance and type 2 diabetes mellitus. Nat. Rev. Endocrinol. 2009, 5, 305–311. [Google Scholar] [CrossRef] [PubMed]
- Mohanty, P.; Hamouda, W.; Garg, R.; Aljada, A.; Ghanim, H.; Dandona, P. Glucose challenge stimulates reactive oxygen species (ROS) generation by leucocytes. J. Clin. Endocrinol. Metab. 2000, 85, 2970–2973. [Google Scholar] [CrossRef]
- Dhindsa, S.; Tripathy, D.; Mohanty, P.; Ghanim, H.; Syed, T.; Aljada, A.; Dandona, P. Differential effects of glucose and alcohol on reactive oxygen species generation and intranuclear nuclear factor-kappaB in mono- nuclear cells. Metabolism 2004, 53, 330–334. [Google Scholar] [CrossRef]
- Larsen, C.M.; Faulenbach, M.; Vaag, A.; Ehses, J.A.; Donath, M.Y.; Mandrup-Poulsen, T. Sustained effects of interleukin-1 receptor antagonist treatment in type 2 diabetes. Diabetes Care 2009, 32, 1663–1668. [Google Scholar] [CrossRef]
- van Asseldonk, E.J.; Stienstra, R.; Koenen, T.B.; Joosten, L.A.; Netea, M.G.; Tack, C.J. Treatment with anakinra improves disposition index but not insulin sensitivity in nondiabetic subjects with the metabolic syndrome: A randomized, double-blind, placebo-controlled study. J. Clin. Endocrinol. Metab. 2011, 96, 2119–2126. [Google Scholar] [CrossRef]
- Rissanen, A.; Howard, C.P.; Botha, J.; Thuren, T.; Global Investigators. Effect of anti-IL-1β antibody (canakinumab) on insulin secretion rates in impaired glucose tolerance or type 2 diabetes: Results of a randomized, placebo-controlled trial. Diabetes Obes. Metab. 2012, 14, 1088–1096. [Google Scholar] [CrossRef]
- Hensen, J.; Howard, C.P.; Walter, V.; Thuren, T. Impact of interleukin-1β antibody (canakinumab) on glycaemic indicators in patients with type 2 diabetes mellitus: Results of secondary endpoints from a randomized, placebo-controlled trial. Diabetes Metab. 2013, 39, 524–531. [Google Scholar] [CrossRef] [PubMed]
- Goldfine, A.B.; Fonseca, V.; Jablonski, K.A.; Pyle, L.; Staten, M.A.; Shoelson, S.E. The effects of salsalate on glycemic control in patients with type 2 diabetes: A randomized trial. Ann. Intern. Med. 2010, 152, 346–357. [Google Scholar] [CrossRef]
- Goldfine, A.B.; Fonseca, V.; Jablonski, K.A.; Chen, Y.D.; Tipton, L.; Staten, M.A.; Shoelson, S.E.; Targeting inflammation Using Salsalate in Type 2 Diabetes Study Team. Salicylate (salsalate) in patients with type 2 diabetes: A randomized trial. Ann. Intern.Med. 2013, 159, 1–12. [Google Scholar] [CrossRef] [PubMed]
- Mohamed, W.A.; Schaalan, M.F. Antidiabetic efficacy of lactoferrin in type 2 diabetic pediatrics; controlling impact on PPAR-γ, SIRT-1, and TLR4 downstream signaling pathway. Diabetol. Metab. Syndr. 2018, 10. [Google Scholar] [CrossRef]
- Ley, R.E.; Peterson, D.A.; Gordon, J.I. Ecological and evolutionary forces shaping microbial diversity in the human intestine. Cell 2006, 124, 837–848. [Google Scholar] [CrossRef]
- O’Hara, A.M.; Shanahan, F. The gut flora as a forgotten organ. EMBO Rep. 2006, 7, 688–693. [Google Scholar] [CrossRef]
- Sorini, C.; Cardoso, R.F.; Gagliani, N.; Villablanca, E.J. Commensal Bacteria-Specific CD4+ T Cell Responses in Health and Disease. Front. Immunol. 2018, 9. [Google Scholar] [CrossRef]
- Sekirov, I.; Finlay, B.B. The role of the intestinal microbiota in enteric infection. J. Physiol. 2009, 587, 4159–4167. [Google Scholar] [CrossRef] [PubMed]
- Kamada, N.; Chen, G.Y.; Inohara, N.; Núñez, G. Control of pathogens and pathobionts by the gut microbiota. Nat. Immunol. 2013, 14, 685–690. [Google Scholar] [CrossRef]
- Dethlefsen, L.; McFall-Ngai, M.; Relman, D.A. An ecological and evolutionary perspective on human-microbe mutualism and disease. Nature 2007, 449, 811–818. [Google Scholar] [CrossRef]
- Tomlin, H.; Piccinini, A.M. A complex interplay between the extracellular matrix and the innate immune response to microbial pathogens. Immunology 2018, 155, 186–201. [Google Scholar] [CrossRef]
- Gras, S.; Van Rhijn, I.; Shahine, A.; Le Nours, J. Molecular recognition of microbial lipid-based antigens by T cells. Cell. Mol. Life Sci. 2018, 75, 1623–1639. [Google Scholar] [CrossRef]
- Adlerova, L.; Bartoskova, A.; Faldyna, M. Lactoferrin: A review. Veterinarni Medicina 2008, 457–468. [Google Scholar] [CrossRef]
- Drago-Serrano, M.E.; Campos-Rodríguez, R.; Carrero, J.C.; de la Garza, M. Lactoferrin: Balancing Ups and Downs of Inflammation Due to Microbial Infections. Int. J. Mol. Sci. 2017, 18, 501. [Google Scholar] [CrossRef] [PubMed]
- Manzoni, P.; Rinaldi, M.; Cattani, S.; Pugni, L.; Romeo, M.G.; Messner, H.; Stolfi, I.; Decembrino, L.; Laforgia, N.; Vagnarelli, F.; et al. Bovine lactoferrin supplementation for prevention of late-onset sepsis in very low-birth-weight neonates: A randomized trial. JAMA 2009, 7, 1421–1428. [Google Scholar] [CrossRef] [PubMed]
- Turin, C.G.; Zea-Vera, A.; Pezo, A.; Cruz, K.; Zegarra, J.; Bellomo, S.; Cam, L.; Llanos, R.; Castañeda, A.; Tucto, L.; et al. Lactoferrin for prevention of neonatal sepsis. BioMetals 2014, 27, 1007–1016. [Google Scholar] [CrossRef] [PubMed]
- Pammi, M.; Suresh, G. Enteral lactoferrin supplementation for prevention of sepsis and necrotizing enterocolitis in preterm infants. Cochrane Database Syst. Rev. 2017, 6. [Google Scholar] [CrossRef]
- Telang, S. Lactoferrin: A Critical Player in Neonatal Host Defense. Nutrients 2018, 10, 1228. [Google Scholar] [CrossRef]
- Larsen, B.; Monif, G.R. Understanding the bacterial flora of the female genital tract. Clin. Infect. Dis. 2001, 32, 69–77. [Google Scholar] [CrossRef] [PubMed]
- Oakley, B.B.; Fiedler, T.L.; Marrazzo, J.M.; Fredricks, D.N. Diversity of human vaginal bacterial communities and associations with clinically defined bacterial vaginosis. Appl. Environ. Microbiol. 2008, 74, 4898–4909. [Google Scholar] [CrossRef]
- Russo, R.; Edu, A.; De Seta, F. Study on the effects of an oral lactobacilli and lactoferrin complex in women with intermediate vaginal microbiota. Arch. Gynecol. Obstet. 2018, 298, 139–145. [Google Scholar] [CrossRef]
- Anderson, M.R.; Klink, K.; Cohrssen, A. Evaluation of vaginal complaints. JAMA 2004, 291, 1368–1379. [Google Scholar] [CrossRef]
- Farage, M.A.; Miller, K.W.; Ledger, W.J. Determining the cause of vulvovaginal symptoms. Obstet. Gynecol. Surv. 2008, 63, 445–464. [Google Scholar] [CrossRef]
- Hainer, B.L.; Gibson, M.V. Vaginitis: Diagnosis and Treatment. Am. Fam. Physician. 2011, 83, 807–815. [Google Scholar]
- Hedges, S.R.; Barrientes, F.; Desmond, R.A.; Schwebke, J.R. Local and systemic cytokine levels in relation to changes in vaginal flora. J. Infect. Dis. 2006, 193, 556–562. [Google Scholar] [CrossRef]
- Beigi, R.H.; Yudin, M.H.; Cosentino, L.; Meyn, L.A.; Hillier, S.L. Cytokines, pregnancy, and bacterial vaginosis: Comparison of levels of cervical cytokines in pregnant and non-pregnant women with bacterial vaginosis. J. Infect. Dis. 2007, 196, 1355–1360. [Google Scholar] [CrossRef] [PubMed]
- Rampersauda, R.; Randis, T.M.; Ratner, A.J. Microbiota of the upper and lower genital tract. Semin. Fetal Neonatal Med. 2012, 17, 51–57. [Google Scholar] [CrossRef]
- Valenti, P.; Rosa, L.; Capobianco, D.; Lepanto, M.S.; Schiavi, E.; Cutone, A.; Paesano, R.; Mastromarino, P. Role of Lactobacilli and Lactoferrin in the Mucosal Cervicovaginal Defense. Front. Immunol. 2018, 9, 376. [Google Scholar] [CrossRef]
- Peipert, J.F.; Montagno, A.B.; Cooper, A.S.; Sung, C.J. Bacterial vaginosis as a risk factor for upper genital tract infection. Am. J. Obstet. Gynecol. 1997, 177, 1184–1187. [Google Scholar] [CrossRef]
- Brotman, R.M.; Klebanoff, M.A.; Nansel, T.R.; Yu, K.F.; Andrews, W.W.; Zhang, J.; Schwebke, J.R. Bacterial vaginosis assessed by gram stain and diminished colonization resistance to incident gonococcal, chlamydial, and trichomonal genital infection. J. Infect. Dis. 2010, 202, 1907–1915. [Google Scholar] [CrossRef]
- Sawada, M.; Otsuki, K.; Mitsukawa, K.; Yakuwa, K.; Nagatsuka, M.; Okai, T. Cervical inflammatory cytokines and other markers in the cervical mucus of pregnant women with lower genital tract infection. Int. J. Gynaecol. Obstet. 2006, 92, 117–121. [Google Scholar] [CrossRef]
- Grab, D.J.; Lonsdale-Eccles, J.D.; Oli, M.W.; Corbeil, L.B. Lactoferrin-binding proteins of Tritrichomonas foetus. J. Parasitol. 2001, 87, 1064–1070. [Google Scholar] [CrossRef]
- Chu, H.; Slepenkin, A.; Elofsson, M.; Keyser, P.; de la Maza, L.M.; Peterson, E.M. Candidate vaginal microbicides with activity against Chlamydia trachomatis and Neisseria gonorrhoeae. Int. J. Antimicrob. Agents 2010, 36, 145–150. [Google Scholar] [CrossRef] [PubMed]
- Spear, G.T.; Kendrick, S.R.; Chen, H.Y.; Thomas, T.T.; Bahk, M.; Balderas, R.; Ghosh, S.; Weinberg, A.; Landay, A.L. Multiplex immunoassay of lower genital tract mucosal fluid from women attending an urban STD clinic shows broadly increased IL1ß and lactoferrin. PLoS ONE 2011, 6, e19560. [Google Scholar] [CrossRef]
- Sessa, R.; Di Pietro, M.; Filardo, S.; Bressan, A.; Rosa, L.; Cutone, A.; Frioni, A.; Berlutti, F.; Paesano, R.; Valenti, P. Effect of bovine lactoferrin on Chlamydia trachomatis infection and inflammation. Biochem. Cell Biol. 2017, 95, 34–40. [Google Scholar] [CrossRef]
- Sessa, R.; Di Pietro, M.; Filardo, S.; Bressan, A.; Mastromarino, P.; Biasucci, A.V.; Rosa, L.; Cutone, A.; Berlutti, F.; Paesano, R.; et al. Lactobacilli-lactoferrin interplay in Chlamydia trachomatis infection. Pathog. Dis. 2017, 75. [Google Scholar] [CrossRef] [PubMed]
- Liao, H.; Liu, S.; Wang, H.; Su, H.; Liu, Z. Enhanced antifungal activity of bovine lactoferrin-producing probiotic Lactobacillus casei in the murine model of vulvovaginal candidiasis. BMC Microbiol. 2019, 19, 7. [Google Scholar] [CrossRef] [PubMed]
- Raulston, J.E. Response of Chlamydia trachomatis serovar E to iron restriction vitro and evidence for iron-regulated chlamydial proteins. Infect. Immun. 1997, 65, 4539–4547. [Google Scholar]
- Wu, H.F.; Monroe, D.M.; Church, F.C. Characterization of the glycosaminoglycan-binding region of lactoferrin. Arch. Biochem. Biophys. 1995, 317, 85–92. [Google Scholar] [CrossRef]
- Stallmann, S.; Hegemann, J.H. The Chlamydia trachomatis Ctad1 invasin exploits the human integrin 1 receptor for host cell entry. Cell. Microbiol. 2015, 18, 761–775. [Google Scholar] [CrossRef] [PubMed]
- Lipuma, J.J. The changing microbial epidemiology in cystic fibrosis. Clin. Microbiol. Rev. 2010, 299–323. [Google Scholar] [CrossRef]
- Salsgiver, E.L.; Fink, A.K.; Knapp, E.A.; Lipuma, J.J.; Olivier, K.N.; Marshall, B.C.; Saiman, L. Changing epidemiology of the respiratory bacteriology of patients with cystic fibrosis. Chest 2016, 390–400. [Google Scholar] [CrossRef]
- Hogardt, M.; Heesemann, J. Microevolution of Pseudomonas aeruginosa to a chronic pathogen of the cystic fibrosis lung. Curr. Top. Microbiol. Immunol. 2013, 358, 91–118. [Google Scholar] [CrossRef]
- Cockx, M.; Gouwy, M.; Van Damme, J.; Struyf, S. Chemoattractants and cytokines in primary ciliary dyskinesia and cystic fibrosis: Key players in chronic respiratory diseases. Cell Mol. Immunol. 2018, 312–323. [Google Scholar] [CrossRef]
- Dakin, C.J.; Numa, A.H.; Wang, H.; Morton, J.R.; Vertzyas, C.C.; Henry, R.L. Inflammation, infection, and pulmonary function in infants and young children with cystic fibrosis. Am. J. Respir. Crit. Care Med. 2002, 904–910. [Google Scholar] [CrossRef]
- Verhaeghe, C.; Remouchamps, C.; Hennuy, B.; Vanderplasschen, A.; Chariot, A.; Tabruyn, S.P.; Oury, C.; Bours, V. Role of IKK and ERK pathways in intrinsic inflammation of cystic fibrosis airways. Biochem. Pharmacol. 2007, 73, 1982–1994. [Google Scholar] [CrossRef]
- Bragonzi, A.; Horati, H.; Kerrigan, L.; Lorè, N.I.; Scholte, B.J.; Weldon, S. Inflammation and host-pathogen interaction: Cause and consequence in cystic fibrosis lung disease. J. Cyst. Fibros. 2018, 17, S40–S45. [Google Scholar] [CrossRef] [PubMed]
- Frioni, A.; Conte, M.P.; Cutone, A.; Longhi, C.; Musci, G.; di Patti, M.C.; Natalizi, T.; Marazzato, M.; Lepanto, M.S.; Puddu, P.; et al. Lactoferrin differently modulates the inflammatory response in epithelial models mimicking human inflammatory and infectious diseases. BioMetals 2014, 27, 843–856. [Google Scholar] [CrossRef]
- Valenti, P.; Catizone, A.; Pantanella, F.; Frioni, A.; Natalizi, T.; Tendini, M.; Berlutti, F. Lactoferrin decreases inflammatory response by cystic fibrosis bronchial cells invaded with Burkholderia cenocepacia iron-modulated biofilm. Int. J. Immunopathol. Pharmacol. 2011, 24, 1057–1068. [Google Scholar] [CrossRef]
- Valenti, P.; Frioni, A.; Rossi, A.; Ranucci, S.; De Fino, I.; Cutone, A.; Rosa, L.; Bragonzi, A.; Berlutti, F. Aerosolized bovine lactoferrin reduces neutrophils and pro-inflammatory cytokines in mouse models of Pseudomonas aeruginosa lung infections. Biochem. Cell Biol. 2017, 95, 41–47. [Google Scholar] [CrossRef]
- Lieschke, G.J.; Grail, D.; Hodgson, G.; Metcalf, D.; Stanley, E.; Cheers, C.; Fowler, K.J.; Basu, S.; Zhan, Y.F.; Dunn, A.R. Mice lacking granulocyte colony-stimulating factor have chronic neutropenia, granulocyte and macrophage progenitor cell deficiency, and impaired neutrophil mobilization. Blood 1994, 84, 1737–1746. [Google Scholar]
- Natori, T.; Sata, M.; Washida, M.; Hirata, Y.; Nagai, R.; Makuuchi, M. G-CSF stimulates angiogenesis and promotes tumor growth: Potential contribution of bone marrow-derived endothelial progenitor cells. Biochem. Biophys. Res. Commun. 2002, 297, 1058–1061. [Google Scholar] [CrossRef]
- Christensen, A.D.; Haase, C.; Cook, A.D.; Hamilton, J.A. Granulocyte colony-stimulating factor (G-CSF) plays an important role in immune complex-mediated arthritis. Eur. J. Immunol. 2016, 46, 1235–1245. [Google Scholar] [CrossRef] [PubMed]
- Zhao, X.; Xu, X.X.; Liu, Y.; Xi, E.Z.; An, J.J.; Tabys, D.; Liu, N. The In Vitro Protective Role of Bovine Lactoferrin on Intestinal Epithelial Barrier. Molecules 2019, 24, 148. [Google Scholar] [CrossRef] [PubMed]
- Lönnerdal, B. Infant formula and infant nutrition: Bioactive proteins of human milk and implications for composition of infant formulas. Am. J. Clin. Nutr. 2014, 99, 712S–717S. [Google Scholar] [CrossRef] [PubMed]
- Samadi, N.; Klems, M.; Untersmayr, E. The role of gastrointestinal permeability in food allergy. Ann. Allergy Asthma Im. 2018, 121, 168–173. [Google Scholar] [CrossRef]
- Arnott, I.D.; Kingstone, K.; Ghosh, S. Abnormal intestinal permeability predicts relapse in inactive Crohn disease. Scand. J. Gastroenterol. 2000, 35, 1163–1169. [Google Scholar]
- Weber, C.R. Dynamic properties of the tight junction barrier. Ann. N. Y. Acad. Sci. 2012, 1257, 77–84. [Google Scholar] [CrossRef] [PubMed]
- Reinhold, A.K.; Rittner, H.L. Barrier function in the peripheral and central nervous system—A review. Eur. J. Physiol. 2017, 469, 123–134. [Google Scholar] [CrossRef] [PubMed]
- Scaldaferri, F.; Pizzoferrato, M.; Gerardi, V.; Lopetuso, L.; Gasbarrini, A. The gut barrier: New acquisitions and therapeutic approaches. J. Clin. Gastroenterol. 2012, 46, S12–S17. [Google Scholar] [CrossRef]
- Lee, S.H. Intestinal permeability regulation by tight junction: Implication on inflammatory bowel diseases. Intest. Res. 2015, 13, 11–18. [Google Scholar] [CrossRef]
- Caccaro, R.; D’Incá, R.; Sturniolo, G.C. Clinical utility of calprotectin and lactoferrin as markers of inflammation in patients with inflammatory bowel disease. Expert Rev. Clin. Immunol. 2010, 6, 551–558. [Google Scholar] [CrossRef] [PubMed]
- Davidson, L.A.; Lönnerdal, B. Persistence of human milk proteins in the breast-fed infant. Acta Paediatr. Scand. 1987, 76, 733–740. [Google Scholar] [CrossRef]
- Dai, J.; Liu, W.Z.; Zhao, Y.P.; Hu, Y.B.; Ge, Z.Z. Relationship between fecal lactoferrin and inflammatory bowel disease. Scand. J. Gastroenterol. 2007, 42, 1440–1444. [Google Scholar] [CrossRef]
- Pfefferkorn, M.D.; Boone, J.H.; Nguyen, J.T.; Juliar, B.E.; Davis, M.A.; Parker, K.K. Utility of fecal lactoferrin in identifying Crohn disease activity in children. J. Pediatr. Gastroenterol. Nutr. 2010, 51, 425–428. [Google Scholar] [CrossRef] [PubMed]
- Karrasch, T.; Jobin, C. NF-kappaB and the intestine: Friend or foe? Inflamm. Bowel Dis. 2008, 14, 114–124. [Google Scholar] [CrossRef]
- Bain, C.C.; Mowat, A.M. Macrophages in intestinal homeostasis and inflammation. Immunol. Rev. 2014, 260, 102–117. [Google Scholar] [CrossRef] [PubMed]
- Togawa, J.; Nagase, H.; Tanaka, K.; Inamori, M.; Nakajima, A.; Ueno, N.; Saito, T.; Sekihara, H. Oral administration of lactoferrin reduces colitis in rats via modulation of the immune system and correction of cytokine imbalance. J. Gastroenterol. Hepatol. 2002, 17, 1291–1298. [Google Scholar] [CrossRef] [PubMed]
- Glasser, A.L.; Boudeau, J.; Barnich, N.; Perruchot, M.H.; Colombel, J.F.; Darfeuille-Michaud, A. Adherent invasive Escherichia coli strains from patients with Crohn’s disease survive and replicate within macrophages without inducing host cell death. Infect. Immun. 2001, 69, 5529–5537. [Google Scholar] [CrossRef]
- Darfeuille-Michaud, A.; Boudeau, J.; Bulois, P.; Neut, C.; Glasser, A.; Barnich, N.; Colombel, J.F. Hight prevalence of adherent-invasive Escherichia coli associated with ileal mucosa in Crohn’s disease. Gastroenterology 2004, 127, 412–421. [Google Scholar] [CrossRef] [PubMed]
- Sasaki, M.; Sitaraman, S.V.; Babbin, B.A.; Gerner-Smidt, P.; Ribot, E.M.; Garrett, N.; Alpern, J.A.; Akyildiz, A.; Theiss, A.L.; Nusrat, A.; et al. Invasive Escherichia coli are a feature of Crohn’s disease. Lab. Investig. 2007, 87, 1042–1105. [Google Scholar] [CrossRef]
- Rolhion, N.; Darfeuille-Michaud, A. Adherent-Invasive Escherichia coli in Inflammatory Bowel Disease. Inflamm. Bowel Dis. 2007, 13, 1277–1283. [Google Scholar] [CrossRef] [PubMed]
- Martinez-Medina, M.; Aldeguer, X.; Lopez-Siles, M.; Gonzalez-Huix, F.; Lopez-Oliu, C.; Dahbi, G.; Blanco, J.E.; Blanco, J.; Garcia-Gil, L.J.; Darfeuille-Michaud, A. Molecular diversity of Escherichia coli in the human gut: New ecological evidence supporting the role of adherent-invasive E. coli (AIEC) in Crohn’s disease. Inflamm. Bowel Dis. 2009, 15, 872–882. [Google Scholar] [CrossRef]
- Chassaing, B.; Etienne-Mesmin, L.; Bonnet, R.; Darfeuille-Michaud, A. Bile salts induce long polar fimbriae expression favouring Crohn’s disease-associated adherent-invasive Escherichia coli interaction with Peyer’s patches. Environ. Microbiol. 2013, 15, 355–371. [Google Scholar] [CrossRef] [PubMed]
- Iebba, V.; Conte, M.P.; Lepanto, M.S.; Di Nardo, G.; Santangelo, F.; Aloi, M.; Totino, V.; Checchi, M.P.; Longhi, C.; Cucchiara, S.; et al. Microevolution in fimH gene of mucosa-associated Escherichia coli strains isolated from pediatric patients with inflammatory bowel disease. Infect. Immun. 2012, 80, 1408–1417. [Google Scholar] [CrossRef] [PubMed]
- Conte, M.P.; Longhi, C.; Marazzato, M.; Conte, A.L.; Aleandri, M.; Lepanto, M.S.; Zagaglia, C.; Nicoletti, M.; Aloi, M.; Totino, V.; et al. Adherent-invasive Escherichia coli (AIEC) in pediatric Crohn’s disease patients: Phenotypic and genetic pathogenic features. BMC Res. Notes 2014, 7, 748. [Google Scholar] [CrossRef]
- Curran, C.S.; Demick, K.P.; Mansfield, J.M. Lactoferrin activates macrophages via TLR4-dependent and -independent signaling pathways. Cell. Immunol. 2006, 242, 23–30. [Google Scholar] [CrossRef] [PubMed]
- Puddu, P.; Carollo, M.G.; Belardelli, F.; Valenti, P.; Gessani, S. Role of endogenous interferon and LPS in the immunomodulatory effects of bovine lactoferrin in murine peritoneal macrophages. J. Leukoc. Biol. 2007, 82, 347–353. [Google Scholar] [CrossRef] [PubMed]
- Puddu, P.; Latorre, D.; Carollo, M.; Catizone, A.; Ricci, G.; Valenti, P.; Gessani, S. Bovine lactoferrin counteracts Toll-like receptor mediated activation signals in antigen presenting cells. PLoS ONE 2011, 6, e22504. [Google Scholar] [CrossRef] [PubMed]
- Berlutti, F.; Schippa, S.; Morea, C.; Sarli, S.; Perfetto, B.; Donnarumma, G.; Valenti, P. Lactoferrin downregulates pro-inflammatory cytokines upexpressed in intestinal epithelial cells infected with invasive or noninvasive Escherichia coli strains. Biochem. Cell Biol. 2006, 84, 351–357. [Google Scholar] [CrossRef]
- Cario, E.; Podolsky, D.K. Differential alteration in intestinal epithelial cell expression of toll-like receptor 3 (TLR3) and TLR4 in inflammatory bowel disease. Infect. Immun. 2000, 68, 7010–7017. [Google Scholar] [CrossRef]
- Vora, P.; Youdim, A.; Thomas, L.S.; Fukata, M.; Tesfay, S.Y.; Lukasek, K.; Michelsen, K.S.; Wada, A.; Hirayama, T.; Arditi, M.; et al. Beta-defensin-2 expression is regulated by TLR signaling in intestinal epithelial cells. J. Immunol. 2004, 173, 5398–5405. [Google Scholar] [CrossRef] [PubMed]
- Komatsu, A.; Satoh, T.; Wakabayashi, H.; Ikeda, F. Effects of bovine lactoferrin to oral Candida albicans and Candida glabrata isolates recovered from the saliva in elderly people. Odontology 2013, 103, 50–55. [Google Scholar] [CrossRef] [PubMed]
© 2019 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (http://creativecommons.org/licenses/by/4.0/).
Share and Cite
Lepanto, M.S.; Rosa, L.; Paesano, R.; Valenti, P.; Cutone, A. Lactoferrin in Aseptic and Septic Inflammation. Molecules 2019, 24, 1323. https://doi.org/10.3390/molecules24071323
Lepanto MS, Rosa L, Paesano R, Valenti P, Cutone A. Lactoferrin in Aseptic and Septic Inflammation. Molecules. 2019; 24(7):1323. https://doi.org/10.3390/molecules24071323
Chicago/Turabian StyleLepanto, Maria Stefania, Luigi Rosa, Rosalba Paesano, Piera Valenti, and Antimo Cutone. 2019. "Lactoferrin in Aseptic and Septic Inflammation" Molecules 24, no. 7: 1323. https://doi.org/10.3390/molecules24071323
APA StyleLepanto, M. S., Rosa, L., Paesano, R., Valenti, P., & Cutone, A. (2019). Lactoferrin in Aseptic and Septic Inflammation. Molecules, 24(7), 1323. https://doi.org/10.3390/molecules24071323