Nanodiagnosis and Nanotreatment of CVDs: Comparison
Please note this is a comparison between Version 2 by Vivi Li and Version 1 by Muhammad Nadeem Zafar.

Cardiovascular diseases (CVDs) are the world’s leading cause of mortality and represent a large contributor to the costs of medical care. Although tremendous progress has been made for the diagnosis of CVDs, there is an important need for more effective early diagnosis and the design of novel diagnostic methods. The diagnosis of CVDs generally relies on signs and symptoms depending on molecular imaging (MI) or on CVD-associated biomarkers. For early-stage CVDs, however, the reliability, specificity, and accuracy of the analysis is still problematic. Because of their unique chemical and physical properties, nanomaterial systems have been recognized as potential candidates to enhance the functional use of diagnostic instruments. Nanomaterials such as gold nanoparticles, carbon nanotubes, quantum dots, lipids, and polymeric nanoparticles represent novel sources to target CVDs. The special properties of nanomaterials including surface energy and topographies actively enhance the cellular response within CVDs. The availability of newly advanced techniques in nanomaterial science opens new avenues for the targeting of CVDs. 

  • nanotechnology
  • diagnosis
  • treatment
  • cardiovascular diseases
Please wait, diff process is still running!

References

  1. WHO. Cardiovascular Diseases (CVDs). 2017. Available online: (accessed on 4 July 2018).
  2. Flora, G.D.; Nayak, M.K. A brief review of cardiovascular diseases, associated risk factors and current treatment regimes. Curr. Pharm. Des. 2019, 25, 4063–4084.
  3. Zamani, P.; Fereydouni, N.; Butler, A.E.; Navashenaq, J.G.; Sahebkar, A. The therapeutic and diagnostic role of exosomes in cardiovascular diseases. Trends Cardiovasc. Med. 2019, 29, 313–323.
  4. Braunwald, E. Cardiomyopathies: An overview. Circ. Res. 2017, 121, 711–721.
  5. Yusuf, S.; Joseph, P.; Rangarajan, S.; Islam, S.; Mente, A.; Hystad, P.; Brauer, M.; Kutty, V.R.; Gupta, R.; Wielgosz, A. Modifiable risk factors, cardiovascular disease, and mortality in 155 722 individuals from 21 high-income, middle-income, and low-income countries (PURE): A prospective cohort study. Lancet 2020, 395, 795–808.
  6. Chiba, A.; Watanabe-Takano, H.; Miyazaki, T.; Mochizuki, N. Cardiomyokines from the heart. Cell. Mol. Life Sci. 2018, 75, 1349–1362.
  7. Putzu, A.; de Carvalho, C.M.P.D.; de Almeida, J.P.; Belletti, A.; Cassina, T.; Landoni, G.; Hajjar, L.A. Perioperative statin therapy in cardiac and non-cardiac surgery: A systematic review and meta-analysis of randomized controlled trials. Ann. Intensive Care 2018, 8, 95.
  8. Diaconu, C.C.; Marcu, D.R.; Bratu, O.G.; Stanescu, A.M.A.; Gheorghe, G.; Hlescu, A.A.; Mischianu, D.L.; Manea, M. Beta-blockers in cardiovascular therapy: A review. J. Mind Med. Sci. 2019, 6, 216–223.
  9. Zhang, P.; Zhu, L.; Cai, J.; Lei, F.; Qin, J.-J.; Xie, J.; Liu, Y.-M.; Zhao, Y.-C.; Huang, X.; Lin, L. Association of inpatient use of angiotensin converting enzyme inhibitors and angiotensin II receptor blockers with mortality among patients with hypertension hospitalized with COVID-19. Circ. Res. 2020, 126, 1671–1681.
  10. Ambrosy, A.P.; Mentz, R.J.; Fiuzat, M.; Cleland, J.G.; Greene, S.J.; O’Connor, C.M.; Teerlink, J.R.; Zannad, F.; Solomon, S.D. The role of angiotensin receptor–neprilysin inhibitors in cardiovascular disease-existing evidence, knowledge gaps, and future directions. Eur. J. Heart Fail. 2018, 20, 963–972.
  11. Rodriguez-Araujo, G.; Krentz, A.J. Utility of invasive and non-invasive cardiovascular research. In Translational Research Methods in Diabetes, Obesity, and Nonalcoholic Fatty Liver Disease: A Focus on Early Phase Clinical Drug Development; Springer: Cham, Switzerland, 2019; pp. 275–308.
  12. Shi, C.; Xie, H.; Ma, Y.; Yang, Z.; Zhang, J. Nanoscale technologies in highly sensitive diagnosis of cardiovascular diseases. Front. Bioeng. Biotechnol. 2020, 8, 531.
  13. Morris, S.A.; Slesnick, T.C. Magnetic resonance imaging. In Visual Guide to Neonatal Cardiology; John Wiley & Sons Ltd.: Hoboken, NJ, USA, 2018; pp. 104–108.
  14. Alsharqi, M.; Woodward, W.; Mumith, J.; Markham, D.; Upton, R.; Leeson, P. Artificial intelligence and echocardiography. Echo Res. Pract. 2018, 5, R115–R125.
  15. Picard, F.; Sayah, N.; Spagnoli, V.; Adjedj, J.; Varenne, O. Vasospastic angina: A literature review of current evidence. Arch. Cardiovasc. Dis. 2019, 112, 44–55.
  16. Schoepf, U.J. CT of the Heart; Humana Press: Totowa, NJ, USA, 2019.
  17. Oikonomou, E.K. Molecular Imaging to Guide Precision Diagnosis and Prevention of Cancer Therapeutics-Related Cardiac Dysfunction; Taylor & Francis: Abingdon, UK, 2020.
  18. Clerico, A.; Lippi, G. The state-of-the-art of “high-sensitivity” immunoassay for measuring cardiac troponin I and T. J. Lab. Precis. Med. 2018, 3, 53.
  19. Chandarana, M.; Curtis, A.; Hoskins, C. The use of nanotechnology in cardiovascular disease. Appl. Nanosci. 2018, 8, 1607–1619.
  20. Raber, I.; McCarthy, C.P.; Vaduganathan, M.; Bhatt, D.L.; Wood, D.A.; Cleland, J.G.; Blumenthal, R.S.; McEvoy, J.W. The rise and fall of aspirin in the primary prevention of cardiovascular disease. Lancet 2019, 393, 2155–2167.
  21. Univers, J.; Long, C.; Tonks, S.A.; Freeman, M.B. Systemic hypersensitivity reaction to endovascular stainless steel stent. J. Vasc. Surg. 2018, 67, 615–617.
  22. Bejarano, J.; Navarro-Marquez, M.; Morales-Zavala, F.; Morales, J.O.; Garcia-Carvajal, I.; Araya-Fuentes, E.; Flores, Y.; Verdejo, H.E.; Castro, P.F.; Lavandero, S. Nanoparticles for diagnosis and therapy of atherosclerosis and myocardial infarction: Evolution toward prospective theranostic approaches. Theranostics 2018, 8, 4710.
  23. Tian, L.; Lu, L.; Feng, J.; Melancon, M.P. Radiopaque nano and polymeric materials for atherosclerosis imaging, embolization and other catheterization procedures. Acta Pharm. Sin. B 2018, 8, 360–370.
  24. Richardson, J.J.; Caruso, F. Nanomedicine toward 2040. Nano Lett. 2020, 20, 1481–1482.
  25. Barani, M.; Torkzadeh-Mahani, M.; Mirzaei, M.; Nematollahi, M.H. Comprehensive evaluation of gene expression in negative and positive trigger-based targeting niosomes in HEK-293 cell line. Iran. J. Pharm. Res. IJPR 2020, 19, 166.
  26. Bilal, M.; Barani, M.; Sabir, F.; Rahdar, A.; Kyzas, G.Z. Nanomaterials for the treatment and diagnosis of Alzheimer’s disease: An overview. NanoImpact 2020, 20, 100251.
  27. Mombini, S.; Mohammadnejad, J.; Bakhshandeh, B.; Narmani, A.; Nourmohammadi, J.; Vahdat, S.; Zirak, S. Chitosan-PVA-CNT nanofibers as electrically conductive scaffolds for cardiovascular tissue engineering. Int. J. Biol. Macromol. 2019, 140, 278–287.
  28. Mukhtar, M.; Ali, H.; Ahmed, N.; Munir, R.; Talib, S.; Khan, A.S.; Ambrus, R. Drug delivery to macrophages: A review of nano-therapeutics targeted approach for inflammatory disorders and cancer. Expert Opin. Drug Deliv. 2020, 17, 1239–1257.
  29. Sevostyanov, M.; Baikin, A.; Sergienko, K.; Shatova, L.; Kirsankin, A.; Baymler, I.; Shkirin, A.; Gudkov, S. Biodegradable stent coatings on the basis of PLGA polymers of different molecular mass, sustaining a steady release of the thrombolityc enzyme streptokinase. React. Funct. Polym. 2020, 150, 104550.
  30. Sun, J.; Sun, K.; Bai, K.; Chen, S.; Wang, F.; Zhao, F.; Hong, N.; Hu, H. A novel braided biodegradable stent for use in congenital heart disease: Short-term results in porcine iliac artery. J. Biomed. Mater. Res. Part A 2019, 107, 1667–1677.
  31. Omid, S.O.; Goudarzi, Z.; Kangarshahi, L.M.; Mokhtarzade, A.; Bahrami, F. Self-expanding stents based on shape memory alloys and shape memory polymers. J. Compos. Compd. 2020, 2, 92–98.
  32. Maleki, B.; Alinezhad, H.; Atharifar, H.; Tayebee, R.; Mofrad, A.V. One-pot synthesis of polyhydroquinolines catalyzed by ZnCl2 supported on nano Fe3O4@ SiO2. Org. Prep. Proced. Int. 2019, 51, 301–309.
  33. Cervadoro, A.; Palomba, R.; Vergaro, G.; Cecchi, R.; Menichetti, L.; Decuzzi, P.; Emdin, M.; Luin, S. Targeting inflammation with nanosized drug delivery platforms in cardiovascular diseases: Immune cell modulation in atherosclerosis. Front. Bioeng. Biotechnol. 2018, 6, 177.
  34. Sandoval-Yañez, C.; Castro Rodriguez, C. Dendrimers: Amazing platforms for bioactive molecule delivery systems. Materials 2020, 13, 570.
  35. Dizaj, S.M.; Rad, A.A.; Safaei, N.; Salatin, S.; Ahmadian, E.; Sharifi, S.; Vahed, S.Z.; Lotfipour, F.; Shahi, S. The application of nanomaterials in cardiovascular diseases: A review on drugs and devices. J. Pharm. Pharm. Sci. 2019, 22, 501–515.
  36. Thompson, L.C.; Sheehan, N.L.; Walters, D.M.; Lust, R.M.; Brown, J.M.; Wingard, C.J. Airway exposure to modified multi-walled carbon nanotubes perturbs cardiovascular adenosinergic signaling in mice. Cardiovasc. Toxicol. 2019, 19, 168–177.
  37. Giménez, V.M.M.; Fuentes, L.B.; Kassuha, D.E.; Manucha, W. Current Drug Nano-targeting Strategies for Improvement in the diagnosis and treatment of prevalent pathologies such as cardiovascular and renal diseases. Curr. Drug Targets 2019, 20, 1496–1504.
  38. Tu, Y.; Sun, Y.; Fan, Y.; Cheng, Z.; Yu, B. Multimodality molecular imaging of cardiovascular disease based on nanoprobes. Cell. Physiol. Biochem. 2018, 48, 1401–1415.
  39. Li, T.; Liang, W.; Xiao, X.; Qian, Y. Nanotechnology, an alternative with promising prospects and advantages for the treatment of cardiovascular diseases. Int. J. Nanomed. 2018, 13, 7349.
  40. Deng, Y.; Zhang, X.; Shen, H.; He, Q.; Wu, Z.; Liao, W.; Yuan, M. Application of the nano-drug delivery system in treatment of cardiovascular diseases. Front. Bioeng. Biotechnol. 2020, 7, 489.
  41. Su, M.; Dai, Q.; Chen, C.; Zeng, Y.; Chu, C.; Liu, G. Nano-medicine for thrombosis: A precise diagnosis and treatment strategy. Nano-Micro Lett. 2020, 12, 96.
  42. Solaimuthu, A.; Vijayan, A.N.; Murali, P.; Korrapati, P.S. Nano-biosensors and their relevance in tissue engineering. Curr. Opin. Biomed. Eng. 2020, 13, 84–93.
  43. Lenz, T.; Nicol, P.; Castellanos, M.I.; Engel, L.-C.; Lahmann, A.L.; Alexiou, C.; Joner, M. Small dimension—big impact! Nanoparticle-enhanced non-invasive and intravascular molecular imaging of atherosclerosis in vivo. Molecules 2020, 25, 1029.
  44. Christodoulides, N.; McRae, M.P.; Simmons, G.W.; Modak, S.S.; McDevitt, J.T. Sensors that learn: The evolution from taste fingerprints to patterns of early disease detection. Micromachines 2019, 10, 251.
  45. Li, T.; Feng, Z.-Q.; Qu, M.; Yan, K.; Yuan, T.; Gao, B.; Wang, T.; Dong, W.; Zheng, J. Core/shell piezoelectric nanofibers with spatial self-orientated β-phase nanocrystals for real-time micropressure monitoring of cardiovascular walls. ACS Nano 2019, 13, 10062–10073.
  46. Jiang, W.; Rutherford, D.; Vuong, T.; Liu, H. Nanomaterials for treating cardiovascular diseases: A review. Bioact. Mater. 2017, 2, 185–198.
  47. Barani, M.; Bilal, M.; Rahdar, A.; Arshad, R.; Kumar, A.; Hamishekar, H.; Kyzas, G.Z. Nanodiagnosis and nanotreatment of colorectal cancer: An overview. J. Nanopart. Res. 2021, 23, 18.
  48. Barani, M.; Bilal, M.; Sabir, F.; Rahdar, A.; Kyzas, G.Z. Nanotechnology in ovarian cancer: Diagnosis and treatment. Life Sci. 2020, 266, 118914.
  49. Barani, M.; Mukhtar, M.; Rahdar, A.; Sargazi, G.; Thysiadou, A.; Kyzas, G.Z. Progress in the application of nanoparticles and graphene as drug carriers and on the diagnosis of brain infections. Molecules 2021, 26, 186.
  50. Barani, M.; Mukhtar, M.; Rahdar, A.; Sargazi, S.; Pandey, S.; Kang, M. Recent advances in nanotechnology-based diagnosis and treatments of human osteosarcoma. Biosensors 2021, 11, 55.
  51. Ghazy, E.; Kumar, A.; Barani, M.; Kaur, I.; Rahdar, A.; Behl, T. Scrutinizing the therapeutic and diagnostic potential of nanotechnology in thyroid cancer: Edifying drug targeting by nano-oncotherapeutics. J. Drug Deliv. Sci. Technol. 2020, 61, 102221.
  52. Hasanein, P.; Rahdar, A.; Barani, M.; Baino, F.; Yari, S. Oil-in-water microemulsion encapsulation of antagonist drugs prevents renal ischemia-reperfusion injury in rats. Appl. Sci. 2021, 11, 1264.
  53. Mukhtar, M.; Bilal, M.; Rahdar, A.; Barani, M.; Arshad, R.; Behl, T.; Brisc, C.; Banica, F.; Bungau, S. Nanomaterials for diagnosis and treatment of brain cancer: Recent updates. Chemosensors 2020, 8, 117.
  54. Qindeel, M.; Barani, M.; Rahdar, A.; Arshad, R.; Cucchiarini, M. Nanomaterials for the diagnosis and treatment of urinary tract infections. Nanomaterials 2021, 11, 546.
  55. Rahdar, A.; Hajinezhad, M.R.; Sargazi, S.; Bilal, M.; Barani, M.; Karimi, P.; Kyzas, G.Z. Biochemical effects of deferasirox and deferasirox-loaded nanomicellesin iron-intoxicated rats. Life Sci. 2021, 270, 119146.
  56. Rahdar, A.; Sargazi, S.; Barani, M.; Shahraki, S.; Sabir, F.; Aboudzadeh, M.A. Lignin-stabilized doxorubicin microemulsions: Synthesis, physical characterization, and in vitro assessments. Polymers 2021, 13, 641.
  57. Sabir, F.; Barani, M.; Rahdar, A.; Bilal, M.; Nadeem, M. How to face skin cancer with nanomaterials: A review. Biointerface Res. Appl. Chem. 2021, 11, 11931–11955.
  58. Pala, R.; Pattnaik, S.; Busi, S.; Nauli, S.M. Nanomaterials as novel cardiovascular theranostics. Pharmaceutics 2021, 13, 348.
  59. Nicolosi, D.; Scalia, M.; Nicolosi, V.M.; Pignatello, R. Encapsulation in fusogenic liposomes broadens the spectrum of action of vancomycin against Gram-negative bacteria. Int. J. Antimicrob. Agents 2010, 35, 553–558.
  60. Darwitan, A.; Wong, Y.S.; Nguyen, L.T.; Czarny, B.; Vincent, A.; Nedumaran, A.M.; Tan, Y.F.; Muktabar, A.; Tang, J.K.; Ng, K.W. Liposomal nanotherapy for treatment of atherosclerosis. Adv. Healthc. Mater. 2020, 9, 2000465.
  61. Matoba, T.; Koga, J.-I.; Nakano, K.; Egashira, K.; Tsutsui, H. Nanoparticle-mediated drug delivery system for atherosclerotic cardiovascular disease. J. Cardiol. 2017, 70, 206–211.
  62. Dasa, S.S.K.; Suzuki, R.; Gutknecht, M.; Brinton, L.T.; Tian, Y.; Michaelsson, E.; Lindfors, L.; Klibanov, A.L.; French, B.A.; Kelly, K.A. Development of target-specific liposomes for delivering small molecule drugs after reperfused myocardial infarction. J. Control. Release 2015, 220, 556–567.
  63. Lestini, B.J.; Sagnella, S.M.; Xu, Z.; Shive, M.S.; Richter, N.J.; Jayaseharan, J.; Case, A.J.; Kottke-Marchant, K.; Anderson, J.M.; Marchant, R.E. Surface modification of liposomes for selective cell targeting in cardiovascular drug delivery. J. Control. Release 2002, 78, 235–247.
  64. Van der Valk, F.M.; van Wijk, D.F.; Lobatto, M.E.; Verberne, H.J.; Storm, G.; Willems, M.C.; Legemate, D.A.; Nederveen, A.J.; Calcagno, C.; Mani, V. Prednisolone-containing liposomes accumulate in human atherosclerotic macrophages upon intravenous administration. Nanomed. Nanotechnol. Biol. Med. 2015, 11, 1039–1046.
  65. Laing, S.T.; Moody, M.R.; Kim, H.; Smulevitz, B.; Huang, S.-L.; Holland, C.K.; McPherson, D.D.; Klegerman, M.E. Thrombolytic efficacy of tissue plasminogen activator-loaded echogenic liposomes in a rabbit thrombus model. Thromb. Res. 2012, 130, 629–635.
  66. Zhong, H.; Deng, Y.; Wang, X.; Yang, B. Multivesicular liposome formulation for the sustained delivery of breviscapine. Int. J. Pharm. 2005, 301, 15–24.
  67. Huang, G.; Zhou, Z.; Srinivasan, R.; Penn, M.S.; Kottke-Marchant, K.; Marchant, R.E.; Gupta, A.S. Affinity manipulation of surface-conjugated RGD peptide to modulate binding of liposomes to activated platelets. Biomaterials 2008, 29, 1676–1685.
  68. Januzzi, J.L., Jr.; Maisel, A.S.; Silver, M.; Xue, Y.; DeFilippi, C. Natriuretic peptide testing for predicting adverse events following heart failure hospitalization. Congest. Heart Fail. 2012, 18, S9–S13.
  69. Al Meslmani, B.M.; Mahmoud, G.F.; Bakowsky, U. Development of expanded polytetrafluoroethylene cardiovascular graft platform based on immobilization of poly lactic-co-glycolic acid nanoparticles using a wet chemical modification technique. Int. J. Pharm. 2017, 529, 238–244.
  70. Ahadian, S.; Huyer, L.D.; Estili, M.; Yee, B.; Smith, N.; Xu, Z.; Sun, Y.; Radisic, M. Moldable elastomeric polyester-carbon nanotube scaffolds for cardiac tissue engineering. Acta Biomater. 2017, 52, 81–91.
  71. Wennink, J.W.; Liu, Y.; Mäkinen, P.I.; Setaro, F.; de la Escosura, A.; Bourajjaj, M.; Lappalainen, J.P.; Holappa, L.P.; van den Dikkenberg, J.B.; Al Fartousi, M. Macrophage selective photodynamic therapy by meta-tetra (hydroxyphenyl) chlorin loaded polymeric micelles: A possible treatment for cardiovascular diseases. Eur. J. Pharm. Sci. 2017, 107, 112–125.
  72. Cormode, D.P.; Naha, P.C.; Fayad, Z.A. Nanoparticle contrast agents for computed tomography: A focus on micelles. Contrast Media Mol. Imaging 2014, 9, 37–52.
  73. Mosser, D.M.; Edwards, J.P. Exploring the full spectrum of macrophage activation. Nat. Rev. Immunol. 2008, 8, 958–969.
  74. Nakashiro, S.; Matoba, T.; Umezu, R.; Koga, J.-i.; Tokutome, M.; Katsuki, S.; Nakano, K.; Sunagawa, K.; Egashira, K. Pioglitazone-incorporated nanoparticles prevent plaque destabilization and rupture by regulating monocyte/macrophage differentiation in ApoE−/− mice. Arterioscler. Thromb. Vasc. Biol. 2016, 36, 491–500.
  75. Yoo, S.P.; Pineda, F.; Barrett, J.C.; Poon, C.; Tirrell, M.; Chung, E.J. Gadolinium-functionalized peptide amphiphile micelles for multimodal imaging of atherosclerotic lesions. ACS Omega 2016, 1, 996–1003.
  76. Kirana, C.; Rogers, P.F.; Bennett, L.E.; Abeywardena, M.Y.; Patten, G.S. Naturally derived micelles for rapid in vitro screening of potential cholesterol-lowering bioactives. J. Agric. Food Chem. 2005, 53, 4623–4627.
  77. Wang, S.; Wang, S.; Yu, X.; Zeng, J.; Li, H.; Yang, R.; Chen, W.; Dong, J. Magnetic nanoparticles functionalized with immobilized apolipoprotein antibodies for direct detection of non-high density lipoprotein cholesterol in human serum. Chem. Eng. J. 2020, 385, 123465.
  78. A Sharma, P.; Maheshwari, R.; Tekade, M.; Kumar Tekade, R. Nanomaterial based approaches for the diagnosis and therapy of cardiovascular diseases. Curr. Pharm. Des. 2015, 21, 4465–4478.
  79. Kievit, F.M.; Veiseh, O.; Fang, C.; Bhattarai, N.; Lee, D.; Ellenbogen, R.G.; Zhang, M. Chlorotoxin labeled magnetic nanovectors for targeted gene delivery to glioma. ACS Nano 2010, 4, 4587–4594.
  80. Ito, A.; Shinkai, M.; Honda, H.; Kobayashi, T. Medical application of functionalized magnetic nanoparticles. J. Biosci. Bioeng. 2005, 100, 1–11.
  81. Mouli, S.K.; Tyler, P.; McDevitt, J.L.; Eifler, A.C.; Guo, Y.; Nicolai, J.; Lewandowski, R.J.; Li, W.; Procissi, D.; Ryu, R.K. Image-guided local delivery strategies enhance therapeutic nanoparticle uptake in solid tumors. ACS Nano 2013, 7, 7724–7733.
  82. Chen, C.-C.; Lin, Y.-P.; Wang, C.-W.; Tzeng, H.-C.; Wu, C.-H.; Chen, Y.-C.; Chen, C.-P.; Chen, L.-C.; Wu, Y.-C. DNA− gold nanorod conjugates for remote control of localized gene expression by near infrared irradiation. J. Am. Chem. Soc. 2006, 128, 3709–3715.
  83. Chithrani, B.D.; Chan, W.C. Elucidating the mechanism of cellular uptake and removal of protein-coated gold nanoparticles of different sizes and shapes. Nano Lett. 2007, 7, 1542–1550.
  84. Ghann, W.E.; Aras, O.; Fleiter, T.; Daniel, M.-C. Syntheses and characterization of lisinopril-coated gold nanoparticles as highly stable targeted CT contrast agents in cardiovascular diseases. Langmuir 2012, 28, 10398–10408.
  85. Almutairi, A.; Rossin, R.; Shokeen, M.; Hagooly, A.; Ananth, A.; Capoccia, B.; Guillaudeu, S.; Abendschein, D.; Anderson, C.J.; Welch, M.J. Biodegradable dendritic positron-emitting nanoprobes for the noninvasive imaging of angiogenesis. Proc. Natl. Acad. Sci. USA 2009, 106, 685–690.
  86. Lu, Y.; Sun, B.; Li, C.; Schoenfisch, M.H. Structurally diverse nitric oxide-releasing poly(propylene imine) dendrimers. Chem. Mater. 2011, 23, 4227–4233.
  87. Tomalia, D.A. Starburstr̀ dendrimers—Nanoscopic supermolecules according to dendritic rules and principles. In Macromolecular Symposia; Hüthig & Wepf Verlag: Basel, Switzerland; pp. 243–255.
  88. Venkatesh, S.; Li, M.; Saito, T.; Tong, M.; Rashed, E.; Mareedu, S.; Zhai, P.; Bárcena, C.; López-Otín, C.; Yehia, G.; et al. Mitochondrial LonP1 protects cardiomyocytes from ischemia/reperfusion injury in vivo. J. Mol. Cell. Cardiol. 2019, 128, 38–50.
  89. Taite, L.J.; West, J.L. Poly (ethylene glycol)-lysine dendrimers for targeted delivery of nitric oxide. J. Biomater. Sci. Polym. Ed. 2006, 17, 1159–1172.
  90. Bhadra, D.; Bhadra, S.; Jain, N.K. Pegylated lysine based copolymeric dendritic micelles for solubilization and delivery of artemether. J. Pharm. Pharm. Sci. Publ. Can. Soc. Pharm. Sci. Soc. Can. Des Sci. Pharm. 2005, 8, 467–482.
  91. Shao, W.; Arghya, P.; Yiyong, M.; Rodes, L.; Prakash, S. Carbon nanotubes for use in medicine: Potentials and limitations. Synth. Appl. Carbon Nanotub. Compos. 2013, 13, 285–311.
  92. Chen, Z.; Zhang, X.; Yang, R.; Zhu, Z.; Chen, Y.; Tan, W. Single-walled carbon nanotubes as optical materials for biosensing. Nanoscale 2011, 3, 1949–1956.
  93. Strus, M.C.; Chiaramonti, A.N.; Kim, Y.L.; Jung, Y.J.; Keller, R.R. Accelerated reliability testing of highly aligned single-walled carbon nanotube networks subjected to DC electrical stressing. Nanotechnology 2011, 22, 265713.
  94. Garibaldi, S.; Brunelli, C.; Bavastrello, V.; Ghigliotti, G.; Nicolini, C. Carbon nanotube biocompatibility with cardiac muscle cells. Nanotechnology 2005, 17, 391.
  95. Jayagopal, A.; Russ, P.K.; Haselton, F.R. Surface engineering of quantum dots for in vivo vascular imaging. Bioconjugate Chem. 2007, 18, 1424–1433.
  96. Larson, D.R.; Zipfel, W.R.; Williams, R.M.; Clark, S.W.; Bruchez, M.P.; Wise, F.W.; Webb, W.W. Water-soluble quantum dots for multiphoton fluorescence imaging in vivo. Science 2003, 300, 1434–1436.
  97. Ferrara, D.E.; Glaus, C.; Taylor, W.R. Targeting vascular epitopes using quantum dots. In Nanoparticles in Biomedical Imaging; Springer: Berlin/Heidelberg, Germany, 2008; pp. 443–461.
  98. Yan, M.; Zhang, Y.; Xu, K.; Fu, T.; Qin, H.; Zheng, X. An in vitro study of vascular endothelial toxicity of CdTe quantum dots. Toxicology 2011, 282, 94–103.
  99. Aizik, G.; Waiskopf, N.; Agbaria, M.; Ben-David-Naim, M.; Levi-Kalisman, Y.; Shahar, A.; Banin, U.; Golomb, G. Liposomes of quantum dots configured for passive and active delivery to tumor tissue. Nano Lett. 2019, 19, 5844–5852.
More
Video Production Service