We use cookies to improve your experience. By continuing to browse this site, you accept our cookie policy.×
Skip main navigation
Aging Health
Bioelectronics in Medicine
Biomarkers in Medicine
Breast Cancer Management
CNS Oncology
Colorectal Cancer
Concussion
Epigenomics
Future Cardiology
Future Medicine AI
Future Microbiology
Future Neurology
Future Oncology
Future Rare Diseases
Future Virology
Hepatic Oncology
HIV Therapy
Immunotherapy
International Journal of Endocrine Oncology
International Journal of Hematologic Oncology
Journal of 3D Printing in Medicine
Lung Cancer Management
Melanoma Management
Nanomedicine
Neurodegenerative Disease Management
Pain Management
Pediatric Health
Personalized Medicine
Pharmacogenomics
Regenerative Medicine
Research Articlecc iconby iconnc iconnd icon

Nanoparticle conversion to biofilms: in vitro demonstration using serum-derived mineralo-organic nanoparticles

    Tsui-Yin Wong

    Laboratory of Nanomaterials, Chang Gung University, Gueishan, Taoyuan 33302, Taiwan

    Center for Molecular & Clinical Immunology, Chang Gung University, Gueishan, Taoyuan 33302, Taiwan

    Authors contributed equally

    Search for more papers by this author

    ,
    Hsin-Hsin Peng

    Laboratory of Nanomaterials, Chang Gung University, Gueishan, Taoyuan 33302, Taiwan

    Center for Molecular & Clinical Immunology, Chang Gung University, Gueishan, Taoyuan 33302, Taiwan

    Authors contributed equally

    Search for more papers by this author

    ,
    Cheng-Yeu Wu

    Laboratory of Nanomaterials, Chang Gung University, Gueishan, Taoyuan 33302, Taiwan

    Center for Molecular & Clinical Immunology, Chang Gung University, Gueishan, Taoyuan 33302, Taiwan

    Research Center of Bacterial Pathogenesis, Chang Gung University, Gueishan, Taoyuan 33302, Taiwan

    ,
    Jan Martel

    Laboratory of Nanomaterials, Chang Gung University, Gueishan, Taoyuan 33302, Taiwan

    Center for Molecular & Clinical Immunology, Chang Gung University, Gueishan, Taoyuan 33302, Taiwan

    ,
    David M Ojcius

    Center for Molecular & Clinical Immunology, Chang Gung University, Gueishan, Taoyuan 33302, Taiwan

    Department of Biomedical Sciences, University of the Pacific, Arthur Dugoni School of Dentistry, San Francisco, CA 94103, USA

    ,
    Fu-Yung Hsu

    Department of Materials Engineering, Ming Chi University of Technology, Taishan, New Taipei City 24301, Taiwan

    &
    John D Young

    *Author for correspondence:

    E-mail Address: dingeyoung@hotmail.com

    Laboratory of Nanomaterials, Chang Gung University, Gueishan, Taoyuan 33302, Taiwan

    Center for Molecular & Clinical Immunology, Chang Gung University, Gueishan, Taoyuan 33302, Taiwan

    Laboratory of Cellular Physiology & Immunology, Rockefeller University, New York, NY 10021, USA

    Biochemical Engineering Research Center, Ming Chi University of Technology, Taishan, New Taipei City 24301, Taiwan

    Published Online:https://doi.org/10.2217/nnm.15.171

    Aims: Mineralo-organic nanoparticles (NPs) detected in biological fluids have been described as precursors of physiological and pathological calcifications in the body. Our main objective was to examine the early stages of mineral NP formation in body fluids. Materials & methods: A nanomaterial approach based on atomic force microscopy, dynamic light scattering, electron microscopy and spectroscopy was used. Results: The mineral particles, which contain the serum proteins albumin and fetuin-A, initially precipitate in the form of round amorphous NPs that gradually grow in size, aggregate and coalesce to form crystalline mineral films similar to the structures observed in calcified human arteries. Conclusion: Our study reveals the early stages of particle formation and provides a platform to analyze the role(s) of mineralo-organic NPs in human tissues.

    Papers of special note have been highlighted as: •• of considerable interest

    References

    • 1 Abedin M, Tintut Y, Demer LL. Vascular calcification: mechanisms and clinical ramifications. Arterioscler. Thromb. Vasc. Biol. 24(7), 1161–1170 (2004).
    • 2 Alexopoulos N, Raggi P. Calcification in atherosclerosis. Nat. Rev. Cardiol. 6, 681–688 (2009).
    • 3 Giachelli CM. Ectopic calcification: gathering hard facts about soft tissue mineralization. Am. J. Pathol. 154(3), 671–675 (1999).
    • 4 Doherty TM, Asotra K, Fitzpatrick LA et al. Calcification in atherosclerosis: bone biology and chronic inflammation at the arterial crossroads. Proc. Natl Acad. Sci. USA 100(20), 11201–11206 (2003).
    • 5 Kapustin AN, Shanahan CM. Calcium regulation of vascular smooth muscle cell-derived matrix vesicles. Trends Cardiovasc. Med. 22(5), 133–137 (2012).
    • 6 Höhling HJ, Arnold S, Plate U, Stratmann U, Wiesmann HP. Analysis of a general principle of crystal nucleation, formation in the different hard tissues. Adv. Dent. Res. 11, 462–466 (1997).
    • 7 Robinson C. Self-oriented assembly of nano-apatite particles: a subunit mechanism for building biological mineral crystals. J. Dent. Res. 86(8), 677–679 (2007).•• Describes the initial observation of round calcium phosphate NPs as precursors of mineralization in developing enamel.
    • 8 Heiss A, Eckert T, Aretz A et al. Hierarchical role of fetuin-A and acidic serum proteins in the formation and stabilization of calcium phosphate particles. J. Biol. Chem. 283(21), 14815–14825 (2008).
    • 9 Bertazzo S, Gentleman E, Cloyd KL, Chester AH, Yacoub MH, Stevens MM. Nano-analytical electron microscopy reveals fundamental insights into human cardiovascular tissue calcification. Nat. Mater. 12(6), 576–583 (2013).
    • 10 Martel J, Young JD. Purported nanobacteria in human blood as calcium carbonate nanoparticles. Proc. Natl Acad. Sci. USA 105(14), 5549–5554 (2008).
    • 11 Young JD, Martel J, Young L, Wu CY, Young A, Young D. Putative nanobacteria represent physiological remnants and culture by-products of normal calcium homeostasis. PLoS ONE 4(2), e4417 (2009).
    • 12 Young JD, Martel J, Young D et al. Characterization of granulations of calcium and apatite in serum as pleomorphic mineralo-protein complexes and as precursors of putative nanobacteria. PLoS ONE 4(5), e5421 (2009).
    • 13 Wu CY, Martel J, Young D, Young JD. Fetuin-A/albumin-mineral complexes resembling serum calcium granules and putative nanobacteria. demonstration of a dual inhibition-seeding concept. PLoS ONE 4(11), e8058 (2009).
    • 14 Young JD, Martel J. The rise and fall of nanobacteria. Sci. Am. 302(1), 52–59 (2010).
    • 15 Martel J, Wu CY, Young JD. Critical evaluation of gamma-irradiated serum used as feeder in the culture and demonstration of putative nanobacteria and calcifying nanoparticles. PLoS ONE 5(4), e10343 (2010).
    • 16 Martel J, Young D, Young A et al. Comprehensive proteomic analysis of mineral nanoparticles derived from human body fluids and analyzed by liquid chromatography-tandem mass spectrometry. Anal. Biochem. 418, 111–125 (2011).
    • 17 Peng HH, Martel J, Lee YH, Ojcius DM, Young JD. Serum-derived nanoparticles: de novo generation and growth in vitro, and internalization by mammalian cells in culture. Nanomedicine (Lond.) 6(4), 643–658 (2011).
    • 18 Martel J, Young D, Peng HH, Wu CY, Young JD. Biomimetic properties of minerals and the search for life in the Martian meteorite ALH84001. Ann. Rev. Earth Planet. Sci. 40, 167–193 (2012).
    • 19 Peng HH, Wu CY, Young D et al. Physicochemical and biological properties of biomimetic mineralo-protein nanoparticles formed spontaneously in biological fluids. Small 9(13), 2297–2307 (2013).
    • 20 Wu CY, Young L, Young D, Martel J, Young JD. Bions: a family of biomimetic mineralo-organic complexes derived from biological fluids. PLoS ONE 8(9), e75501 (2013).
    • 21 Wu CY, Martel J, Cheng WC, He CC, Ojcius DM, Young JD. Membrane vesicles nucleate mineralo-organic nanoparticles and induce carbonate apatite precipitation in human body fluids. J. Biol. Chem. 288(42), 30571–30584 (2013).•• Shows that membrane vesicles found in blood may initiate the formation of mineralo-organic NPs and calcification in biological fluids.
    • 22 Martel J, Peng HH, Young D, Wu CY, Young JD. Of nanobacteria, nanoparticles, biofilms and their role in health and disease: facts, fancy, and future. Nanomedicine (Lond.) 9(4), 483–499 (2014).
    • 23 Wu CY, Young D, Martel J, Young JD. A story told by a single nanoparticle in the body fluid: demonstration of dissolution-reprecipitation of nanocrystals in a biological system. Nanomedicine (Lond.) 10(17), 2659–2676 (2015).
    • 24 Kajander EO, Kuronen I, Akerman K, Pelttari A, Ciftcioglu N. Nanobacteria from blood, the smallest culturable autonomously replicating agent on Earth. Proc. Soc. Photo Opt. Instrum. Eng. 3111, 420–428 (1997).
    • 25 Kajander EO, Ciftcioglu N. Nanobacteria: an alternative mechanism for pathogenic intra- and extracellular calcification and stone formation. Proc. Natl Acad. Sci. USA 95(14), 8274–8279 (1998).
    • 26 Kajander EO, Ciftcioglu N, Miller-Hjelle MA, Hjelle JT. Nanobacteria: controversial pathogens in nephrolithiasis and polycystic kidney disease. Curr. Opin. Nephrol. Hypertens. 10(3), 445–452 (2001).
    • 27 Ciftcioglu N, Mckay DS, Mathew G, Kajander EO. Nanobacteria: fact or fiction? Characteristics, detection, and medical importance of novel self-replicating, calcifying nanoparticles. J. Investig. Med. 54(7), 385–394 (2006).
    • 28 Gilman H, Hukins DW. Seeded growth of hydroxyapatite in the presence of dissolved albumin at constant composition. J. Inorg. Biochem. 55(1), 31–39 (1994).
    • 29 Garnett J, Dieppe P. The effects of serum and human albumin on calcium hydroxyapatite crystal growth. Biochem. J. 266(3), 863–868 (1990).
    • 30 Schinke T, Amendt C, Trindl A, Poschke O, Muller-Esterl W, Jahnen-Dechent W. The serum protein alpha2-HS glycoprotein/fetuin inhibits apatite formation in vitro and in mineralizing calvaria cells: a possible role in mineralization and calcium homeostasis. J. Biol. Chem. 271(34), 20789–20796 (1996).
    • 31 Jahnen-Dechent W, Heiss A, Schäfer C, Ketteler M. Fetuin-A regulation of calcified matrix metabolism. Circulation 108, 1494–1509 (2011).
    • 32 Wong TY, Wu CY, Martel J et al. Detection and characterization of mineralo-organic nanoparticles in human kidneys. Sci. Rep. (2015) (In Press).
    • 33 Posner AS. Crystal chemistry of bone mineral. Physiol. Rev. 49, 760–792 (1969).
    • 34 Jahnen-Dechent W. Lot's wife's problem revisited: how we prevent pathological calcification. In. Biomineralization. Bauerlein E (Ed.). Wiley, Weinheim, Germany, 243–267 (2005).
    • 35 Jahnen-Dechent W, Schäfer C, Heiss A, Grotzinger J. Systemic inhibition of spontaneous calcification by the serum protein alpha 2-HS glycoprotein/fetuin. Z. Kardiol. 90, 47–56 (2001).
    • 36 Schäfer C, Heiss A, Schwarz A et al. The serum protein alpha 2-Heremans-Schmid glycoprotein/fetuin-A is a systemically acting inhibitor of ectopic calcification. J. Clin. Invest. 112(3), 357–366 (2003).•• Seminal study that identified the role of fetuin-A as a systemic calcification inhibitor in mice.
    • 37 Herrmann M, Schäfer C, Heiss A et al. Clearance of fetuin-A – containing calciprotein particles is mediated by scavenger receptor-A. Circ. Res. 111(5), 575–584 (2012).
    • 38 Addadi L, Raz S, Weiner S. Taking advantage of disorder: amorphous calcium carbonate and its roles in biomineralization. Adv. Mater. 15(12), 959–970 (2003).
    • 39 Gower LB. Biomimetic model systems for investigating the amorphous precursor pathway and its role in biomineralization. Chem. Rev. 108, 4551–4627 (2008).•• Review describing the critical role of amorphous mineral as a precursor phase in various biomineralization processes.
    • 40 Beniash E, Metzler RA, Lam RSK, Gilbert PUPA. Transient amorphous calcium phosphate in forming enamel. J. Struct. Biol. 166, 133–143 (2009).
    • 41 Reich W. Die Bione. Zur Entstehung des Vegetativen Lebens. Sexpol-Verlag, Oslo, Norway (1938).
    • 42 Amos FF, Dai L, Kumar R, Khan SR, Gower LB. Mechanism of formation of concentrically laminated spherules. implication to Randall's plaque and stone formation. Urol. Res. 37, 11–17 (2009).
    • 43 Evan AP, Coe FL, Lingeman JE et al. Mechanism of formation of human calcium oxalate renal stones on Randall's plaque. Anat. Rec. (Hoboken) 290(10), 1315–1323 (2007).
    • 44 Ryall RL. The future of stone research: rummagings in the attic, Randall's plaque, nanobacteria, and lessons from phylogeny. Urol. Res. 36, 77–97 (2008).
    • 45 Bassett DC, Grover LM, Müller FA, McKee MD, Barralet JE. Serum protein controlled nanoparticle synthesis. Adv. Funct. Mat. 21(15), 2968–2977 (2011).
    • 46 Chen XR, Bai J, Yuan SJ et al. Calcium phosphate nanoparticles are associated with inorganic phosphate-induced osteogenic differentiation of rat bone marrow stromal cells. Chem. Biol. Interact. 5(238), 111–117 (2015).
    • 47 He G, Gajjeraman S, Schultz D et al. Spatially and temporally controlled biomineralization is facilitated by interaction between self-assembled dentin matrix protein 1 and calcium phosphate nuclei in solution. Biochemistry 44(49), 16140–16148 (2005).
    • 48 Schwartz MK, Hunter LW, Huebner M, Lieske JC, Miller VM. Characterization of biofilm formed by human-derived nanoparticles. Nanomedicine (Lond.) 4(8), 931–941 (2009).