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

Delivering drugs to tubular cells and organelles: the application of nanodrugs in acute kidney injury

    Zhi Li‡

    Department of Nephrology, The Third Xiangya Hospital, Central South University, Changsha, 410013, China

    The Critical Kidney Disease Research Center of Central South University, Changsha, 410013, China

    ‡Authors contributed equally

    Search for more papers by this author

    ,
    Xiao Fan‡

    Department of Nephrology, The Third Xiangya Hospital, Central South University, Changsha, 410013, China

    The Critical Kidney Disease Research Center of Central South University, Changsha, 410013, China

    ‡Authors contributed equally

    Search for more papers by this author

    ,
    Jialong Fan

    College of Biology, Hunan University, Changsha, 410082, China

    ,
    Wei Zhang

    Department of Nephrology, The Third Xiangya Hospital, Central South University, Changsha, 410013, China

    The Critical Kidney Disease Research Center of Central South University, Changsha, 410013, China

    ,
    Jun Liu

    Department of Nephrology, The Third Xiangya Hospital, Central South University, Changsha, 410013, China

    The Critical Kidney Disease Research Center of Central South University, Changsha, 410013, China

    ,
    Bin Liu

    *Author for correspondence:

    E-mail Address: binliu2001@hotmail.com

    College of Biology, Hunan University, Changsha, 410082, China

    Department of Physiology & Pathophysiology, NHC Key Laboratory of Metabolic Cardiovascular Diseases Research, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, 750004, China

    &
    Hao Zhang

    **Author for correspondence:

    E-mail Address: zhanghaoliaoqing@163.com

    Department of Nephrology, The Third Xiangya Hospital, Central South University, Changsha, 410013, China

    The Critical Kidney Disease Research Center of Central South University, Changsha, 410013, China

    Published Online:https://doi.org/10.2217/nnm-2023-0200

    Acute kidney injury (AKI) is a common clinical syndrome with limited treatment options and high mortality rates. Proximal tubular epithelial cells (PTECs) play a key role in AKI progression. Subcellular dysfunctions, including mitochondrial, nuclear, endoplasmic reticulum and lysosomal dysfunctions, are extensively studied in PTECs. These studies have led to the development of potential therapeutic drugs. However, clinical development of those drugs faces challenges such as low solubility, short circulation time and severe systemic side effects. Nanotechnology provides a promising solution by improving drug properties through nanocrystallization and enabling targeted delivery to specific sites. This review summarizes advancements and limitations of nanoparticle-based drug-delivery systems in targeting PTECs and subcellular organelles, particularly mitochondria, for AKI treatment.

    Graphical abstract

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

    References

    • 1. Abebe A, Kumela K, Belay M, Kebede B, Wobie Y. Mortality and predictors of acute kidney injury in adults: a hospital-based prospective observational study. Sci. Rep. 11(1), 15672 (2021).
    • 2. Kellum JA, Romagnani P, Ashuntantang G, Ronco C, Zarbock A, Anders HJ. Acute kidney injury. Nat. Rev. Dis. Primers 7(1), 52 (2021).
    • 3. Ronco C, Bellomo R, Kellum JA. Acute kidney injury. Lancet 394(10212), 1949–1964 (2019).
    • 4. Hukriede NA, Soranno DE, Sander V et al. Experimental models of acute kidney injury for translational research. Nat. Rev. Nephrol. 18(5), 277–293 (2022).
    • 5. Kuwabara S, Goggins E, Okusa MD. The pathophysiology of sepsis-associated AKI. Clin. J. Am. Soc. Nephrol. 17(7), 1050–1069 (2022).
    • 6. Park SJ, Li C, Chen YM. Endoplasmic reticulum calcium homeostasis in kidney disease: pathogenesis and therapeutic targets. Am. J. Pathol. 191(2), 256–265 (2021).
    • 7. Lankadeva YR, May CN, Bellomo R, Evans RG. Role of perioperative hypotension in postoperative acute kidney injury: a narrative review. Br. J. Anaesth. 128(6), 931–948 (2022).
    • 8. Perazella MA, Rosner MH. Drug-induced acute kidney injury. Clin. J. Am. Soc. Nephrol. 17(8), 1220–1233 (2022).
    • 9. Liu BC, Tang TT, Lv LL, Lan HY. Renal tubule injury: a driving force toward chronic kidney disease. Kidney Int. 93(3), 568–579 (2018).
    • 10. Li Z, Liu Z, Luo M et al. The pathological role of damaged organelles in renal tubular epithelial cells in the progression of acute kidney injury. Cell Death Discov. 8(1), 239 (2022).
    • 11. Jourde-Chiche N, Fakhouri F, Dou L et al. Endothelium structure and function in kidney health and disease. Nat. Rev. Nephrol. 15(2), 87–108 (2019).
    • 12. Molema G, Zijlstra JG, van Meurs M, Kamps J. Renal microvascular endothelial cell responses in sepsis-induced acute kidney injury. Nat. Rev. Nephrol. 18(2), 95–112 (2022).
    • 13. Zhang K, Li R, Chen X et al. Renal endothelial cell-targeted extracellular vesicles protect the kidney from ischemic injury. Adv. Sci. (Weinh.) 10(3), e2204626 (2023).
    • 14. Wang S, Wang Y, Lai X et al. Minimalist nanocomplex with dual regulation of endothelial function and inflammation for targeted therapy of inflammatory vascular diseases. ACS Nano 17(3), 2761–2781 (2023).
    • 15. Oroojalian F, Charbgoo F, Hashemi M et al. Recent advances in nanotechnology-based drug delivery systems for the kidney. J. Control. Rel. 321, 442–462 (2020).
    • 16. Geo HN, Murugan DD, Chik Z et al. Renal nano-drug delivery for acute kidney Injury: current status and future perspectives. J. Control. Rel. 343, 237–254 (2022).
    • 17. Kamaly N, He JC, Ausiello DA, Farokhzad OC. Nanomedicines for renal disease: current status and future applications. Nat. Rev. Nephrol. 12(12), 738–753 (2016).
    • 18. Kamada H, Tsutsumi Y, Sato-Kamada K et al. Synthesis of a poly(vinylpyrrolidone-co-dimethyl maleic anhydride) co-polymer and its application for renal drug targeting. Nat. Biotechnol. 21(4), 399–404 (2003).
    • 19. Wei H, Jiang D, Yu B et al. Nanostructured polyvinylpyrrolidone–curcumin conjugates allowed for kidney-targeted treatment of cisplatin induced acute kidney injury. Bioact. Mater. 19, 282–291 (2023).
    • 20. Zhu Z, Liu X, Li P et al. Renal clearable quantum dot–drug conjugates modulate labile iron species and scavenge free radicals for attenuating chemotherapeutic drug-induced acute kidney injury. ACS Appl. Mater. Interfaces 15(18), 21854–21865 (2023).
    • 21. Duan R, Li Y, Zhang R et al. Reversing acute kidney injury through coordinated interplay of anti-inflammation and iron supplementation. Adv. Mater. 35(28), e2301283 (2023). •• Fe3O4 is an excellent nanocarrier, but its use during acute kidney injury (AKI) may exacerbate iron death in renal tubular epithelial cells. This study discovered for the first time that nicotinamide mononucleotide can inhibit inflammation, eliminate the local microenvironment that can induce iron death, and utilize Fe3O4 to improve anemia and promote AKI recovery.
    • 22. Li Y, Wang G, Wang T et al. PEGylated gambogic acid nanoparticles enable efficient renal-targeted treatment of acute kidney injury. Nano Lett. 23(12), 5641–5656 (2023).
    • 23. Alidori S, Akhavein N, Thorek DL et al. Targeted fibrillar nanocarbon RNAi treatment of acute kidney injury. Sci. Transl. Med. 8(331), 331ra339 (2016).
    • 24. Yan R, Cui W, Ma W, Li J, Liu Z, Lin Y. Typhaneoside–tetrahedral framework nucleic acids system: mitochondrial recovery and antioxidation for acute kidney injury treatment. ACS Nano 17(9), 8767–8781 (2023).
    • 25. Choi HS, Mathew AP, Uthaman S et al. Inflammation-sensing catalase-mimicking nanozymes alleviate acute kidney injury via reversing local oxidative stress. J. Nanobiotechnol. 20(1), 205 (2022).
    • 26. Wang S, Chen Y, Han S et al. Selenium nanoparticles alleviate ischemia reperfusion injury-induced acute kidney injury by modulating GPx-1/NLRP3/Caspase-1 pathway. Theranostics 12(8), 3882–3895 (2022).
    • 27. Jiang D, Ge Z, Im HJ et al. DNA origami nanostructures can exhibit preferential renal uptake and alleviate acute kidney injury. Nat. Biomed. Eng. 2(11), 865–877 (2018).
    • 28. Williams RM, Shah J, Mercer E et al. Kidney-targeted redox scavenger therapy prevents cisplatin-induced acute kidney injury. Front. Pharmacol. 12, 790913 (2021).
    • 29. Han SJ, Williams RM, D’Agati V, Jaimes EA, Heller DA, Lee HT. Selective nanoparticle-mediated targeting of renal tubular Toll-like receptor 9 attenuates ischemic acute kidney injury. Kidney Int. 98(1), 76–87 (2020).
    • 30. Vallorz EL, Janda J, Mansour HM, Schnellmann RG. Kidney targeting of formoterol containing polymeric nanoparticles improves recovery from ischemia reperfusion-induced acute kidney injury in mice. Kidney Int. 102(5), 1073–1089 (2022).
    • 31. Deng X, Zeng T, Li J et al. Kidney-targeted triptolide-encapsulated mesoscale nanoparticles for high-efficiency treatment of kidney injury. Biomater. Sci. 7(12), 5312–5323 (2019).
    • 32. Nurunnabi M, Khatun Z, Huh KM et al. In vivo biodistribution and toxicology of carboxylated graphene quantum dots. ACS Nano 7(8), 6858–6867 (2013).
    • 33. Wang H, Yu D, Fang J et al. Phenol-like group functionalized graphene quantum dots structurally mimicking natural antioxidants for highly efficient acute kidney injury treatment. Chem. Sci. 11(47), 12721–12730 (2020).
    • 34. Yu H, Lin T, Chen W et al. Size and temporal-dependent efficacy of oltipraz-loaded PLGA nanoparticles for treatment of acute kidney injury and fibrosis. Biomaterials 219, 119368 (2019). • After inducing ischemia–reperfusion injury (IRI) damage on one side of a mouse’s kidney while keeping the other side healthy, different sizes of PLGA-DSPE-PEG nanoparticles were injected. Nanoparticles with a size of 100 nm accumulated in the IRI-side renal tubular epithelial cells but not in the healthy-side renal tubular epithelial cells.
    • 35. Leeuwis JW, Nguyen TQ, Dendooven A, Kok RJ, Goldschmeding R. Targeting podocyte-associated diseases. Adv. Drug Deliv. Rev. 62(14), 1325–1336 (2010).
    • 36. Williams RM, Shah J, Ng BD et al. Mesoscale nanoparticles selectively target the renal proximal tubule epithelium. Nano Lett. 15(4), 2358–2364 (2015). •• It was serendipitously discovered that PLGA-PEG nanoparticles of medium size accumulated in renal tubular epithelial cells, not via the conventional brush-border absorption but by entry from the basolateral side of the renal tubules. PEG modification was indispensable for this accumulation pathway.
    • 37. Aird WC. Phenotypic heterogeneity of the endothelium: II. Representative vascular beds. Circ. Res. 100(2), 174–190 (2007).
    • 38. Brenner BM, Troy JL, Daugharty TM. On the mechanism of inhibition in fluid reabsorption by the renal proximal tubule of the volume-expanded rat. J. Clin. Invest. 50(8), 1596–1602 (1971).
    • 39. Williams RM, Shah J, Tian HS et al. Selective nanoparticle targeting of the renal tubules. Hypertension 71(1), 87–94 (2018).
    • 40. Vallorz EL, Blohm-Mangone K, Schnellmann RG, Mansour HM. Formoterol PLGA-PEG nanoparticles induce mitochondrial biogenesis in renal proximal tubules. AAPS J. 23(4), 88 (2021).
    • 41. Otsuka H, Nagasaki Y, Kataoka K. PEGylated nanoparticles for biological and pharmaceutical applications. Adv. Drug Deliv. Rev. 55(3), 403–419 (2003).
    • 42. Zhang F, Liu MR, Wan HT. Discussion about several potential drawbacks of PEGylated therapeutic proteins. Biol. Pharm. Bull. 37(3), 335–339 (2014).
    • 43. Christensen EI, Birn H, Storm T, Weyer K, Nielsen R. Endocytic receptors in the renal proximal tubule. Physiology 27(4), 223–236 (2012).
    • 44. Qin S, Wu B, Gong T, Zhang ZR, Fu Y. Targeted delivery via albumin corona nanocomplex to renal tubules to alleviate acute kidney injury. J. Control. Rel. 349, 401–412 (2022).
    • 45. Pang M, Duan S, Zhao M et al. Co-delivery of celastrol and lutein with pH sensitive nano micelles for treating acute kidney injury. Toxicol. Appl. Pharmacol. 450, 116155 (2022).
    • 46. Matsuura S, Katsumi H, Suzuki H et al. L-serine-modified polyamidoamine dendrimer as a highly potent renal targeting drug carrier. Proc. Natl Acad. Sci. USA 115(41), 10511–10516 (2018).
    • 47. Knight SF, Kundu K, Joseph G et al. Folate receptor-targeted antioxidant therapy ameliorates renal ischemia–reperfusion injury. J. Am. Soc. Nephrol. 23(5), 793–800 (2012).
    • 48. Huang C, Zeng T, Li J et al. Folate receptor-mediated renal-targeting nanoplatform for the specific delivery of triptolide to treat renal ischemia/reperfusion injury. ACS Biomater. Sci. Eng. 5(6), 2877–2886 (2019).
    • 49. Du B, Zhao M, Wang Y et al. Folic acid-targeted pluronic F127 micelles improve oxidative stress and inhibit fibrosis for increasing AKI efficacy. Eur. J. Pharmacol. 930, 175131 (2022).
    • 50. Hu JB, Li SJ, Kang XQ et al. CD44-targeted hyaluronic acid–curcumin prodrug protects renal tubular epithelial cell survival from oxidative stress damage. Carbohydr. Polym. 193, 268–280 (2018).
    • 51. Huang ZW, Shi Y, Zhai YY et al. Hyaluronic acid coated bilirubin nanoparticles attenuate ischemia reperfusion-induced acute kidney injury. J. Control. Rel. 334, 275–289 (2021).
    • 52. Walkon LL, Strubbe-Rivera JO, Bazil JN. Calcium overload and mitochondrial metabolism. Biomolecules 12(12), 1891 (2022).
    • 53. Liu D, Shu G, Jin F et al. ROS-responsive chitosan-SS31 prodrug for AKI therapy via rapid distribution in the kidney and long-term retention in the renal tubule. Sci. Adv. 6(41), eabb7422 (2020). • Inspired by phospholipid serine as the substrate of KIM-1, chitosan modified with L-serine and loaded with SS31 formed nanoparticles for AKI treatment. In this system, L-serine binds to the highly expressed KIM-1 on damaged renal tubular epithelial cells, enabling targeted delivery. The reactive oxygen species environment within the cells triggers the release of SS31, which itself targets the mitochondrial inner membrane and clears mitochondrial reactive oxygen species.
    • 54. Li J, Duan Q, Wei X, Wu J, Yang Q. Kidney-targeted nanoparticles loaded with the natural antioxidant rosmarinic acid for acute kidney injury treatment. Small 18(48), e2204388 (2022).
    • 55. Tang TT, Wang B, Li ZL et al. KIM-1 targeted extracellular vesicles: a new therapeutic platform for RNAi to treat AKI. J. Am. Soc. Nephrol. 32(10), 2467–2483 (2021).
    • 56. Yan J, Wang Y, Zhang J, Liu X, Yu L, He Z. Rapidly blocking the calcium overload/ROS production feedback loop to alleviate acute kidney injury via microenvironment-responsive BAPTA-AM/BAC co-delivery nanosystem. Small 19(17), e2206936 (2023).
    • 57. Huang Z, Chun C, Li X. Kidney targeting peptide-modified biomimetic nanoplatforms for treatment of acute kidney injury. J. Control. Rel. 358, 368–381 (2023). •• The principle of ‘homologous targeting’ was first extended to areas beyond tumor-targeted delivery. Renal tubular epithelial cell membrane-coated nanoparticles exhibited better targeting effects than uncoated nanoparticles. Their targeting ability was further enhanced by modification with the renal-targeting peptide KCSAVPLC.
    • 58. Liu Z, Liu X, Yang Q, Yu L, Chang Y, Qu M. Neutrophil membrane-enveloped nanoparticles for the amelioration of renal ischemia–reperfusion injury in mice. Acta Biomater. 104, 158–166 (2020).
    • 59. Tang TT, Wang B, Wu M et al. Extracellular vesicle-encapsulated IL-10 as novel nanotherapeutics against ischemic AKI. Sci. Adv. 6(33), eaaz0748 (2020).
    • 60. Gatti S, Bruno S, Deregibus MC et al. Microvesicles derived from human adult mesenchymal stem cells protect against ischaemia–reperfusion-induced acute and chronic kidney injury. Nephrol. Dial. Transplant. 26(5), 1474–1483 (2011).
    • 61. Zhao M, Liu S, Wang C et al. Mesenchymal stem cell-derived extracellular vesicles attenuate mitochondrial damage and inflammation by stabilizing mitochondrial DNA. ACS Nano 15(1), 1519–1538 (2021).
    • 62. Cao H, Cheng Y, Gao H et al. In vivo tracking of mesenchymal stem cell-derived extracellular vesicles improving mitochondrial function in renal ischemia–reperfusion injury. ACS Nano 14(4), 4014–4026 (2020).
    • 63. Yuan ZX, Zhang ZR, Zhu D et al. Specific renal uptake of randomly 50% N-acetylated low molecular weight chitosan. Mol. Pharm. 6(1), 305–314 (2009).
    • 64. Wang DW, Li SJ, Tan XY et al. Engineering of stepwise-targeting chitosan oligosaccharide conjugate for the treatment of acute kidney injury. Carbohydr. Polym. 256, 117556 (.2021).
    • 65. Kaunitz JD, Cummins VP, Mishler D, Nagami GT. Inhibition of gentamicin uptake into cultured mouse proximal tubule epithelial cells by L-lysine. J. Clin. Pharmacol. 33(1), 63–69 (1993).
    • 66. Wischnjow A, Sarko D, Janzer M et al. Renal targeting: peptide-based drug delivery to proximal tubule cells. Bioconjug. Chem. 27(4), 1050–1057 (2016).
    • 67. Wang J, Poon C, Chin D et al. Design and in vivo characterization of kidney-targeting multimodal micelles for renal drug delivery. Nano Res. 11(10), 5584–5595 (2018).
    • 68. Schreiber A, Theilig F, Schweda F, Hocherl K. Acute endotoxemia in mice induces downregulation of megalin and cubilin in the kidney. Kidney Int. 82(1), 53–59 (2012).
    • 69. Wu X, Tang S, Dai Q et al. Vitamin D–vitamin D receptor alleviates oxidative stress in ischemic acute kidney injury via upregulating glutathione peroxidase 3. FASEB J. 37(2), e22738 (2023).
    • 70. Wang S, Huang S, Liu X, He Y, Liu Y. Paricalcitol ameliorates acute kidney injury in mice by suppressing oxidative stress and inflammation via Nrf2/HO-1 signaling. Int. J. Mol. Sci. 24(2), 969 (2023).
    • 71. Du J, Jiang S, Hu Z et al. Vitamin D receptor activation protects against lipopolysaccharide-induced acute kidney injury through suppression of tubular cell apoptosis. Am. J. Physiol. Renal Physiol. 316(5), F1068–F1077 (2019).
    • 72. Siegelman MH, Degrendele HC, Estess P. Activation and interaction of CD44 and hyaluronan in immunological systems. J. Leukoc. Biol. 66(2), 315–321 (1999).
    • 73. Lewington AJ, Padanilam BJ, Martin DR, Hammerman MR. Expression of CD44 in kidney after acute ischemic injury in rats. Am. J. Physiol. Regul. Integr. Comp. Physiol. 278(1), R247–254 (2000).
    • 74. Fu Z, Fan Q, Zhou Y, Zhao Y, He Z. Elimination of intracellular calcium overload by BAPTA-AM-loaded liposomes: a promising therapeutic agent for acute liver failure. ACS Appl. Mater. Interfaces 11(43), 39574–39585 (2019).
    • 75. Wang Y, Pu M, Yan J et al. 1,2-Bis(2-aminophenoxy)ethane-N,N,N′,N′-tetraacetic acid acetoxymethyl ester loaded reactive oxygen species responsive hyaluronic acid–bilirubin nanoparticles for acute kidney injury therapy via alleviating calcium overload mediated endoplasmic reticulum stress. ACS Nano 17(1), 472–491 (2023).
    • 76. Cichy J, Pure E. The liberation of CD44. J. Cell Biol. 161(5), 839–843 (2003).
    • 77. Ichimura T, Bonventre JV, Bailly V et al. Kidney injury molecule-1 (KIM-1), a putative epithelial cell adhesion molecule containing a novel immunoglobulin domain, is up-regulated in renal cells after injury. J. Biol. Chem. 273(7), 4135–4142 (1998).
    • 78. Arai S, Kitada K, Yamazaki T et al. Apoptosis inhibitor of macrophage protein enhances intraluminal debris clearance and ameliorates acute kidney injury in mice. Nat. Med. 22(2), 183–193 (2016).
    • 79. Han WK, Bailly V, Abichandani R, Thadhani R, Bonventre JV. Kidney injury molecule-1 (KIM-1): a novel biomarker for human renal proximal tubule injury. Kidney Int. 62(1), 237–244 (2002).
    • 80. Ichimura T, Asseldonk EJPV, Humphreys BD, Gunaratnam L, Duffield JS, Bonventre JV. Kidney injury molecule-1 is a phosphatidylserine receptor that confers a phagocytic phenotype on epithelial cells. J. Clin. Invest. 118(5), 1657–1668 (2008).
    • 81. Freeman GJ, Casasnovas JM, Umetsu DT, Dekruyff RH. TIM genes: a family of cell surface phosphatidylserine receptors that regulate innate and adaptive immunity. Immunol. Rev. 235, 172–189 (2010).
    • 82. Kong L, Fan D, Zhou L, Wei S. The influence of modified molecular (D/L-serine) chirality on the theragnostics of PAMAM-based nanomedicine for acute kidney injury. J. Mater. Chem. B 9(43), 9023–9030 (2021).
    • 83. Luk BT, Zhang L. Cell membrane-camouflaged nanoparticles for drug delivery. J. Control. Rel. 220(Pt B), 600–607 (2015).
    • 84. Chen L, Hong W, Ren W, Xu T, Qian Z, He Z. Recent progress in targeted delivery vectors based on biomimetic nanoparticles. Signal Transduct. Target. Ther. 6(1), 225 (2021).
    • 85. Jin K, Luo Z, Zhang B, Pang Z. Biomimetic nanoparticles for inflammation targeting. Acta Pharm. Sin. B 8(1), 23–33 (2018).
    • 86. Deng J, Kohda Y, Chiao H et al. Interleukin-10 inhibits ischemic and cisplatin-induced acute renal injury. Kidney Int. 60(6), 2118–2128 (2001).
    • 87. Ouyang W, O’Garra A. IL-10 family cytokines IL-10 and IL-22: from basic science to clinical translation. Immunity 50(4), 871–891 (2019).
    • 88. Karp JM, Teol GSL. Mesenchymal stem cell homing: the devil is in the details. Cell Stem Cell 4(3), 206–216 (2009).
    • 89. Bruno S, Grange C, Deregibus MC et al. Mesenchymal stem cell-derived microvesicles protect against acute tubular injury. J. Am. Soc. Nephrol. 20(5), 1053–1067 (2009).
    • 90. Cheng YQ, Yue YX, Cao HM et al. Coassembly of hypoxia-sensitive macrocyclic amphiphiles and extracellular vesicles for targeted kidney injury imaging and therapy. J. Nanobiotechnol. 19(1), 451 (2021).
    • 91. Li Y, Wang J, Wientjes MG, Au JL. Delivery of nanomedicines to extracellular and intracellular compartments of a solid tumor. Adv. Drug Deliv. Rev. 64(1), 29–39 (2012).
    • 92. Tang C, Cai J, Yin XM, Weinberg JM, Venkatachalam MA, Dong Z. Mitochondrial quality control in kidney injury and repair. Nat. Rev. Nephrol. 17(5), 299–318 (2021).
    • 93. Liu Z, Li Y, Li C, Yu L, Chang Y, Qu M. Delivery of coenzyme Q10 with mitochondria-targeted nanocarrier attenuates renal ischemia–reperfusion injury in mice. Mater. Sci. Eng. C Mater. Biol. Appl. 131, 112536 (2021).
    • 94. Yu H, Jin F, Liu D et al. ROS-responsive nano-drug delivery system combining mitochondria-targeting ceria nanoparticles with atorvastatin for acute kidney injury. Theranostics 10(5), 2342–2357 (2020).
    • 95. Qin S, Liu C, Chen Y et al. Cobaltosic oxide–polyethylene glycol–triphenylphosphine nanoparticles ameliorate the acute-to-chronic kidney disease transition by inducing BNIP3-mediated mitophagy. Kidney Int. 103(5), 903–916 (2023).
    • 96. Liu D, Jin F, Shu G et al. Enhanced efficiency of mitochondria-targeted peptide SS-31 for acute kidney injury by pH-responsive and AKI-kidney targeted nanopolyplexes. Biomaterials 211, 57–67 (2019).
    • 97. Huang Q, Yang Y, Zhao T et al. Passively-targeted mitochondrial tungsten-based nanodots for efficient acute kidney injury treatment. Bioact. Mater. 21, 381–393 (2023).
    • 98. Smith RA, Porteous CM, Coulter CV, Murphy MP. Selective targeting of an antioxidant to mitochondria. Eur. J. Biochem. 263(3), 709–716 (1999).
    • 99. Murphy MP, Smith RA. Targeting antioxidants to mitochondria by conjugation to lipophilic cations. Ann. Rev. Pharmacol. Toxicol. 47, 629–656 (2007).
    • 100. Reily C, Mitchell T, Chacko BK, Benavides GA, Murphy MP, Darley-Usmar VM. Mitochondrially targeted compounds and their impact on cellular bioenergetics. Redox Biol. 1(1), 86–93 (2013).
    • 101. Battogtokh G, Choi YS, Kang DS et al. Mitochondria-targeting drug conjugates for cytotoxic, anti-oxidizing and sensing purposes: current strategies and future perspectives. Acta Pharm. Sin. B 8(6), 862–880 (2018).
    • 102. Zhao K, Zhao GM, Wu D et al. Cell-permeable peptide antioxidants targeted to inner mitochondrial membrane inhibit mitochondrial swelling, oxidative cell death, and reperfusion injury. J. Biol. Chem. 279(33), 34682–34690 (2004).
    • 103. Birk AV, Liu S, Soong Y et al. The mitochondrial-targeted compound SS-31 re-energizes ischemic mitochondria by interacting with cardiolipin. J. Am. Soc. Nephrol. 24(8), 1250–1261 (2013).
    • 104. Szeto HH. First-in-class cardiolipin-protective compound as a therapeutic agent to restore mitochondrial bioenergetics. Br. J. Pharmacol. 171(8), 2029–2050 (2014).
    • 105. Kumar S. Cellular and molecular pathways of renal repair after acute kidney injury. Kidney Int. 93(1), 27–40 (2018).
    • 106. Ruggiero A, Villa CH, Bander E et al. Paradoxical glomerular filtration of carbon nanotubes. Proc. Natl Acad. Sci. USA 107(27), 12369–12374 (2010).
    • 107. Ma X, Gong N, Zhong L, Sun J, Liang XJ. Future of nanotherapeutics: targeting the cellular sub-organelles. Biomaterials 97, 10–21 (2016).
    • 108. Ledford H. ‘Astonishing’ molecular syringe ferries proteins into human cells. Nature 616(7955), 18–19 (2023).
    • 109. Kreitz J, Friedrich MJ, Guru A et al. Programmable protein delivery with a bacterial contractile injection system. Nature 616(7956), 357–364 (2023). •• F Zhang’s team developed the Photorhabdus virulence cassette (PVC) bacterial injection system. Editing of the tail fiber of PVC enables it to recognize specific cells, thus achieving targeted delivery. The efficiency of the PVC delivery system is nearly 100%.
    • 110. Hulse M, Rosner MH. Drugs in development for acute kidney injury. Drugs 79(8), 811–821 (2019).
    • 111. Fouad AA, Al-Mulhim AS, Jresat I. Cannabidiol treatment ameliorates ischemia/reperfusion renal injury in rats. Life Sci. 91(7–8), 284–292 (2012).
    • 112. Kanlaya R, Thongboonkerd V. Protective effects of epigallocatechin-3-gallate from green tea in various kidney diseases. Adv. Nutr. 10(1), 112–121 (2019).
    • 113. Li HD, Meng XM, Huang C, Zhang L, Lv XW, Li J. Application of herbal traditional Chinese medicine in the treatment of acute kidney injury. Front. Pharmacol. 10, 376 (2019).