ﺑﺎﺯﮔﺸﺖ ﺑﻪ ﺻﻔﺤﻪ ﻗﺒﻠﯽ
خرید پکیج
تعداد آیتم قابل مشاهده باقیمانده : 3 مورد
نسخه الکترونیک
medimedia.ir

Pathogenesis and prevention of aminoglycoside nephrotoxicity and ototoxicity

Pathogenesis and prevention of aminoglycoside nephrotoxicity and ototoxicity
Literature review current through: Jan 2024.
This topic last updated: Aug 16, 2023.

INTRODUCTION — The main concerns with the use of aminoglycoside antibiotics are nephrotoxicity and ototoxicity. This topic will review what is known about the pathogenesis of these complications and how the nephrotoxicity might be prevented. The manifestations of and risk factors for aminoglycoside nephrotoxicity are discussed separately. (See "Manifestations of and risk factors for aminoglycoside nephrotoxicity".)

NEPHROTOXICITY — Acute kidney injury (AKI) due to acute tubular necrosis is a relatively common complication of aminoglycoside therapy, with a rise in the serum creatinine concentration of more than 0.5 to 1 mg/dL (44 to 88 micromol/L) or a 50 percent increase in serum creatinine concentration from baseline occurring in 10 to 20 percent of adult patients [1,2]. A rise in serum creatinine occurs in 20 to 33 percent of pediatric patients, in whom the use of gentamicin is high, especially in neonates in the intensive care unit [3]. Aminoglycosides are freely filtered across the glomerulus; almost all of the drug is then excreted, with 5 to 10 percent of a parenteral intravenous dose being taken up and sequestered by the proximal tubule cells (PTCs), where the aminoglycoside can achieve concentrations vastly exceeding the concurrent serum concentration [4].

The intracellular accumulation of aminoglycosides is confined primarily to the S1 and S2 segments of the proximal tubule. However, following kidney ischemia, the S3 portion is also a site of intracellular aminoglycoside concentrations. AKI can develop even if serum drug levels are closely monitored [5]. Animal models suggest that damage to tubular collecting duct cells also occurs [6].

Dose frequency also may be important as multiple human studies suggest that giving a large dose of aminoglycoside once a day is as effective an antimicrobial regimen and less nephrotoxic than giving aminoglycosides in the conventional, divided-dose regimen [7-11] (see "Dosing and administration of parenteral aminoglycosides"). Aminoglycosides may also have a deleterious effect on the developing kidney in preterm and small for gestational age infants [12].

Renal transport of aminoglycosides

Proximal tubule cell transport and charge — Multiple amine groups on the aminoglycoside molecule confer a cationic charge at physiologic pH [13,14]. As a result, aminoglycoside molecules readily bind to anion phospholipids within the plasma membrane of the PTC in a saturable, electrostatic manner [13-16]. The relative affinity of an aminoglycoside for the PTC plasma membrane correlates with the nephrotoxicity observed in clinical practice [5,17-19]:

Neomycin, which has the highest affinity for the PTC binding site, has the greatest nephrotoxicity of the aminoglycosides.

Tobramycin and gentamicin have lower binding affinity, conferring less nephrotoxicity.

Amikacin has even less binding affinity and, likewise, has less nephrotoxic potential than tobramycin and gentamicin.

Streptomycin, which has the least affinity for the PTC binding site, has the least nephrotoxicity [5,17,18].

Aminoglycosides binding to the anionic phospholipids of the PTC may be transferred to the transmembrane protein megalin or bind directly to megalin by electrostatic interactions, and are subsequently endocytosed [13,14,20-35]. Megalin is a 600 kD cell surface scavenger receptor and has an affinity for positively charged proteins and other molecules like aminoglycosides [36]. Megalin binds to cubilin; both proteins are abundantly expressed in the renal proximal tubules and many other tissues, including glomerular podocytes and ciliary and inner ear epithelium [13,26,37].

Once megalin/cubilin-mediated endocytosis has occurred, endosomes containing the aminoglycosides are transported through the endocytic system, where they can either localize to lysosomes or be trafficked, in a retrograde fashion, first to the Golgi complex [33,38-41], then to the endoplasmic reticulum, and finally to the cytosol [42-45]. In the cytosol, aminoglycoside molecules accumulate in subcellular organelles such as the mitochondria and the nucleus, where they inhibit mitochondrial function and cause increased production of reactive oxygen species [44].

This pathway of the aminoglycosides to the Golgi complex and subcellular organelles is consistent with the disruption in both protein sorting and synthesis and mitochondrial function observed in aminoglycoside nephrotoxicity.

Prevention

Clinical strategies — Clinical strategies that may minimize the potential for nephrotoxicity include:

Selection of the least toxic aminoglycoside, when possible. Gentamicin is considered the most nephrotoxic, followed in decreasing order of nephrotoxicity by tobramycin, amikacin, netilmicin, and streptomycin [1].

Correcting hypokalemia and hypomagnesemia prior to administering an aminoglycoside.

Other effective clinical strategies include avoiding aminoglycosides in patients with reduced effective arterial volume (or optimizing volume status prior to giving aminoglycosides), adjusting the dose for kidney function, limiting the duration of therapy to 7 to 10 days, and minimizing concomitant nephrotoxic medications [46].

Pharmacokinetically monitoring aminoglycoside therapy, serum creatinine, and utilizing a once-daily dosing regimen in selected patients have also demonstrated benefit [7-9,46-49]. (See "Dosing and administration of parenteral aminoglycosides".)

Agents — Several agents have emerged in preclinical studies as potential compounds to prevent aminoglycoside nephrotoxicity. Despite their potential, none have been adopted clinically for the prevention of aminoglycoside nephrotoxicity [50].

One class of compounds, the anionic polyamino acids (PAAs), which include polyaspartic acid and polyglutamic acid, has been extensively evaluated [14,45,51-57]. Early studies showed that PAA interferes with the binding of the aminoglycoside to the PTC plasma membrane [55-57]. Subsequent work showed that PAA binds directly to aminoglycosides and can displace them from negatively charged lysosomes [58]. This led researchers to postulate that PAA affords kidney protection by directly binding to the aminoglycoside or by displacing it from the lysosome and thus preventing its trafficking through the PTC [58].

Other compounds evaluated for their potential role in preventing aminoglycoside nephrotoxicity include antioxidants. As examples, desferrioxamine, methimazole, vitamin E, vitamin C, and selenium have been effective in preventing gentamicin nephrotoxicity [45,59,60]. Superoxide dismutase, lipoic acid, dimethyl-sulfoxide (DMSO), N-acetylcysteine (NAC), melatonin, and the peroxisome proliferator-activated receptor gamma agonist pioglitazone have also demonstrated utility in preventing aminoglycoside nephrotoxicity in preclinical models [59,61-67]. Cilastatin competes with megalin for binding to gentamicin and may inhibit toxicity [68]. In an animal model, inhibiting megalin-mediated endocytosis with the reversible ligand Lrpap1, also known as receptor-associated protein (RAP), prevented both clathrin-mediated and clathrin-independent proximal tubule endocytosis and minimized gentamicin nephrotoxicity [69].

A gentamicin congener that retains its antimicrobial efficacy with a lower potential for aminoglycoside nephrotoxicity has been isolated. Commercially available gentamicin is not a homogeneous compound; rather, it is a mixture of C1, C1a, C2, and C2a congeners that differ in their nephrotoxic potential [70]. The C2 compound was the specific congener shown to be bactericidal with less nephrotoxicity [70]. Via immunofluorescent techniques, the C2 congener is transported intracellularly to the Golgi complex in reduced quantities. This is likely the reason for its lack of nephrotoxicity [70].

Independent of their antimicrobial activity, aminoglycosides are characterized by their ability to induce read-through of premature termination codons, making them potential therapies in the treatment of inherited diseases such as cystic fibrosis [71-76]. Research to develop designer aminoglycosides for this purpose has led to the discovery of aminoglycoside derivatives with less cellular toxicity [71-74]. However, such compounds also have decreased bactericidal activity and thus would not be effective antibiotics [71,74]. Ongoing research in this area may lead to the discovery of other aminoglycoside derivatives that maintain antibacterial efficacy while minimizing cellular toxicity.

OTOTOXICITY — Aminoglycosides are associated with cochlear and vestibular toxicity in a substantial proportion of patients receiving the drug for prolonged periods, leading to hearing loss and disequilibrium, respectively [77,78]. The relative frequency with which these occur is uncertain, and both may not be clinically evident in the same patient. In one series of 33 patients presenting to a neurology clinic with vestibular symptoms, formal audiology testing revealed that 10 of 27 tested patients did not have associated hearing loss greater than expected for age [79]. In a later report on 25 patients, most were found to have no residual hearing loss [77].

Gentamicin and tobramycin are primarily vestibulotoxic, whereas neomycin, kanamycin, and amikacin are more ototoxic [78]. The clinical findings and diagnosis of individuals with aminoglycoside vestibulotoxicity are presented separately. (See "Causes of vertigo", section on 'Aminoglycoside toxicity'.)

Pathogenesis — The pathogenesis of aminoglycoside-induced ototoxicity with hearing loss is less understood. Sustained or excessive peak serum aminoglycoside concentrations are thought to be a risk factor.

One hypothesis is related to receptors for N-methyl-D-aspartate (NMDA), which are present at the synapse between cochlear hair cells and neural afferents [80]. Aminoglycosides can mimic the positive modulation of polyamines at these receptors, possibly producing excitotoxic damage. The observation that the administration of NMDA antagonists markedly attenuates hearing loss in animals is consistent with this hypothesis. Furthermore, there is a high correlation between in vitro activation of the receptor and relative cochlear toxicity in humans (gentamicin > tobramycin > amikacin > neomycin). In addition to these functional changes, aminoglycosides also may induce structural changes, such as loss of target innervation and degeneration of spiral ganglion neurons [81].

Aminoglycosides create reactive oxygen species that damage the inner ear. Support for this hypothesis includes the findings of aminoglycoside-induced reactive oxygen species in vitro [78], and experimental evidence in animals showing ototoxicity prevention with antioxidant agents [82,83].

There appears to be a genetic predisposition to the development of ototoxicity with aminoglycosides. Point mutations in the small mitochondrial (12S) ribosomal RNA gene (eg, a nucleotide 1555 A to G substitution) have been described in a number of families with inherited susceptibility to ototoxicity [50,78,84,85]. The mutant mitochondrial human ribosomal RNA binds aminoglycosides with high affinity; in comparison, the wild-type human ribosomal RNA does not bind aminoglycosides at all [86]. Aminoglycoside binding presumably exacerbates the inherent mitochondrial defect induced by the mutation, resulting in a reduction in the overall translation rate below the minimum level required for normal mitochondrial function and increasing the generation of reactive oxygen species [87]. The prevalence of point mutations among individuals of European ancestry is approximately 1 in 500 [88,89].

Prevention of ototoxicity — Traditionally, a number of the strategies used to help prevent the development of ototoxicity due to aminoglycosides are the same as those used to prevent nephrotoxicity. These strategies include once-daily dosing and careful monitoring of serum drug concentrations. However, ototoxicity has been reported even in those with target serum levels and once-daily dosing [7,79]. (See "Dosing and administration of parenteral aminoglycosides", section on 'Ototoxicity'.)

Another approach is to use audiometric testing among patients receiving aminoglycoside therapy. However, hearing loss may occur even after the termination of antibiotic therapy [79,90].

Genetic screening of patients prior to administering of aminoglycosides would be a valuable tool to aid in the prevention of ototoxicity [88,89]. A rapid screening test has been used in neonates to detect the m.1555A>G variant in a clinically relevant timeframe [91]. The patient and family history also remain valuable tools available to prevent aminoglycoside-induced ototoxicity.

Given the relative rarity of the 12S ribosomal point-mutations that confer susceptibility to aminoglycoside-induced hearing loss, it may not be economically feasible to perform general screening or for most laboratories to carry these diagnostic tests. Although aminoglycosides remain powerful tools in our antibiotic armamentarium, there are reasonable alternatives that provide similar spectrums of bacterial coverage. For patients that require gram-negative bacterial coverage and whose personal or family history suggests a risk for ototoxicity, considering these antibiotic alternatives may be a more prudent approach.

Based upon the observations that oxidative stress may cause ototoxicity, the efficacy of N-acetylcysteine (NAC), a thiol-containing antioxidant, was evaluated in hemodialysis patients receiving gentamicin [92]. In this study, 53 hemodialysis patients with catheter-induced bacteremia were randomly assigned to gentamicin plus NAC (600 mg twice daily) or gentamicin alone. Pure-tone audiograms were performed at baseline, one week, and at six weeks after gentamicin therapy was stopped. NAC was given until the first otologic examination, which was approximately one week after completion of antibiotic therapy. Compared with the control group, NAC therapy significantly lowered the incidence of audiologic toxicity (25 versus 60 percent). Protection primarily occurred in the high-frequency range, and NAC was not associated with any adverse effects.

Another study that included 60 patients on continuous ambulatory peritoneal dialysis (CAPD) suggested that oral administration of NAC prevented ototoxicity related to intraperitoneal amikacin and vancomycin and may have improved high-frequency function at four weeks [93].

We believe that further study in a larger number of patients and in other clinical settings is required before NAC is routinely used to prevent ototoxicity. However, given the safety of NAC, some experts suggest that NAC should be administered to all patients with end-stage kidney disease receiving an aminoglycoside [94,95].

Limited evidence in nondialysis patients suggests that aspirin may provide some protection against ototoxicity [96-98]. Whether this finding extends to dialysis patients without causing significant adverse effects is unclear.

Less ototoxic forms of aminoglycosides may also be useful in preventing ototoxicity. An animal study of an aminoglycoside congener (N1MS) has demonstrated excellent activity against Escherichia coli, Klebsiella pneumoniae, and extended-spectrum B-lactamases while preserving hair cells and hearing relative to its parent compound [99]. Similar results were found with gentamicin C1a, a congener of commercial gentamicin, and apramycin, an aminoglycoside used in veterinarian medicine [100].

SOCIETY GUIDELINE LINKS — Links to society and government-sponsored guidelines from selected countries and regions around the world are provided separately. (See "Society guideline links: Chronic kidney disease in adults".)

SUMMARY AND RECOMMENDATIONS

Prevalence of nephrotoxicity – Acute kidney injury (AKI) due to acute tubular necrosis is a relatively common complication of aminoglycoside therapy, affecting 10 to 20 percent of patients. (See 'Nephrotoxicity' above.)

Varying degrees of nephrotoxicity – Since aminoglycoside molecules readily bind to anionic phospholipids and megalin within the plasma membrane of the proximal tubule cell (PTC) in a saturable, electrostatic manner, the relative affinity of an aminoglycoside for the PTC plasma membrane correlates with the nephrotoxicity observed in clinical practice. Neomycin, which has the highest affinity for the PTC binding site, has the greatest nephrotoxicity of the aminoglycosides. Streptomycin, which has the least affinity for the PTC binding site, has the least nephrotoxicity. (See 'Proximal tubule cell transport and charge' above.)

Prevention of nephrotoxicity

For prevention, clinical strategies are primarily utilized to minimize the potential for nephrotoxicity. These include selection of the least toxic aminoglycoside when possible, correcting hypokalemia and hypomagnesemia prior to administering an aminoglycoside, avoiding aminoglycosides in patients with reduced effective arterial volume, adjusting the dose for kidney function, limiting the duration of therapy to 7 to 10 days, and minimizing concomitant nephrotoxic medications. We suggest utilizing a once-daily dosing regimen in selected patients. (Grade 2B). (See 'Clinical strategies' above.)

Although several agents have emerged as potential compounds to prevent aminoglycoside nephrotoxicity, none have been adopted clinically. (See 'Agents' above.)

Ototoxicity – Aminoglycosides are associated with cochlear and vestibular toxicity in a substantial proportion of patients receiving the drug for prolonged periods, leading to hearing loss and disequilibrium, respectively. The pathogenesis of aminoglycoside-induced ototoxicity with hearing loss is less understood. (See 'Ototoxicity' above and 'Pathogenesis' above.)

Prevention of ototoxicity

The main strategies to prevent the development of ototoxicity due to aminoglycosides are once-daily dosing and careful monitoring of serum drug concentrations. However, some experts also administer N-acetylcysteine (NAC) to patients with end-stage kidney disease receiving an aminoglycoside. (See 'Prevention of ototoxicity' above.)

Rapid tests for mitochondrial RNA mutations that confer increased sensitivity to aminoglycoside ototoxicity have been developed and may play a role in the future.

ACKNOWLEDGMENT — The UpToDate editorial staff acknowledges Brian S Decker, MD, PharmD (deceased), who contributed to an earlier version of this topic review.

  1. Humes HD. Aminoglycoside nephrotoxicity. Kidney Int 1988; 33:900.
  2. Moore RD, Smith CR, Lipsky JJ, et al. Risk factors for nephrotoxicity in patients treated with aminoglycosides. Ann Intern Med 1984; 100:352.
  3. McWilliam SJ, Antoine DJ, Smyth RL, Pirmohamed M. Aminoglycoside-induced nephrotoxicity in children. Pediatr Nephrol 2017; 32:2015.
  4. Galløe AM, Graudal N, Christensen HR, Kampmann JP. Aminoglycosides: single or multiple daily dosing? A meta-analysis on efficacy and safety. Eur J Clin Pharmacol 1995; 48:39.
  5. Smith CR, Lipsky JJ, Laskin OL, et al. Double-blind comparison of the nephrotoxicity and auditory toxicity of gentamicin and tobramycin. N Engl J Med 1980; 302:1106.
  6. Huang H, Jin WW, Huang M, et al. Gentamicin-Induced Acute Kidney Injury in an Animal Model Involves Programmed Necrosis of the Collecting Duct. J Am Soc Nephrol 2020; 31:2097.
  7. Munckhof WJ, Grayson ML, Turnidge JD. A meta-analysis of studies on the safety and efficacy of aminoglycosides given either once daily or as divided doses. J Antimicrob Chemother 1996; 37:645.
  8. Hatala R, Dinh T, Cook DJ. Once-daily aminoglycoside dosing in immunocompetent adults: a meta-analysis. Ann Intern Med 1996; 124:717.
  9. Ferriols-Lisart R, Alós-Almiñana M. Effectiveness and safety of once-daily aminoglycosides: a meta-analysis. Am J Health Syst Pharm 1996; 53:1141.
  10. Barza M, Ioannidis JP, Cappelleri JC, Lau J. Single or multiple daily doses of aminoglycosides: a meta-analysis. BMJ 1996; 312:338.
  11. Prins JM, Büller HR, Kuijper EJ, et al. Once versus thrice daily gentamicin in patients with serious infections. Lancet 1993; 341:335.
  12. Samiee-Zafarghandy S, van den Anker JN. Nephrotoxic effects of the aminoglycosides on the developing kidney. J Ped Neonat Individ Med 2013; 2:1.
  13. Nagai J, Takano M. Molecular aspects of renal handling of aminoglycosides and strategies for preventing the nephrotoxicity. Drug Metab Pharmacokinet 2004; 19:159.
  14. Sastrasinh M, Knauss TC, Weinberg JM, Humes HD. Identification of the aminoglycoside binding site in rat renal brush border membranes. J Pharmacol Exp Ther 1982; 222:350.
  15. Giuliano RA, Verpooten GA, Verbist L, et al. In vivo uptake kinetics of aminoglycosides in the kidney cortex of rats. J Pharmacol Exp Ther 1986; 236:470.
  16. Mingeot-Leclercq MP, Tulkens PM. Aminoglycosides: nephrotoxicity. Antimicrob Agents Chemother 1999; 43:1003.
  17. Smith CR, Baughman KL, Edwards CQ, et al. Controlled comparison of amikacin and gentamicin. N Engl J Med 1977; 296:349.
  18. Lerner AM, Reyes MP, Cone LA, et al. Randomised, controlled trial of the comparative efficacy, auditory toxicity, and nephrotoxicity of tobramycin and netilmicin. Lancet 1983; 1:1123.
  19. Williams PD, Bennett DB, Gleason CR, Hottendorf GH. Correlation between renal membrane binding and nephrotoxicity of aminoglycosides. Antimicrob Agents Chemother 1987; 31:570.
  20. Moestrup SK, Cui S, Vorum H, et al. Evidence that epithelial glycoprotein 330/megalin mediates uptake of polybasic drugs. J Clin Invest 1995; 96:1404.
  21. Hammond TG, Majewski RR, Kaysen JH, et al. Gentamicin inhibits rat renal cortical homotypic endosomal fusion: role of megalin. Am J Physiol 1997; 272:F117.
  22. Christensen EI, Birn H. Megalin and cubilin: multifunctional endocytic receptors. Nat Rev Mol Cell Biol 2002; 3:256.
  23. Christensen EI, Birn H. Megalin and cubilin: synergistic endocytic receptors in renal proximal tubule. Am J Physiol Renal Physiol 2001; 280:F562.
  24. Christensen EI, Willnow TE. Essential role of megalin in renal proximal tubule for vitamin homeostasis. J Am Soc Nephrol 1999; 10:2224.
  25. Kerjaschki D, Farquhar MG. The pathogenic antigen of Heymann nephritis is a membrane glycoprotein of the renal proximal tubule brush border. Proc Natl Acad Sci U S A 1982; 79:5557.
  26. Lundgren S, Carling T, Hjälm G, et al. Tissue distribution of human gp330/megalin, a putative Ca(2+)-sensing protein. J Histochem Cytochem 1997; 45:383.
  27. Schmitz C, Hilpert J, Jacobsen C, et al. Megalin deficiency offers protection from renal aminoglycoside accumulation. J Biol Chem 2002; 277:618.
  28. Cui S, Verroust PJ, Moestrup SK, Christensen EI. Megalin/gp330 mediates uptake of albumin in renal proximal tubule. Am J Physiol 1996; 271:F900.
  29. Nagai J, Tanaka H, Nakanishi N, et al. Role of megalin in renal handling of aminoglycosides. Am J Physiol Renal Physiol 2001; 281:F337.
  30. Christensen EI, Birn H, Verroust P, Moestrup SK. Membrane receptors for endocytosis in the renal proximal tubule. Int Rev Cytol 1998; 180:237.
  31. Bennett WM. Mechanisms of aminoglycoside nephrotoxicity. Clin Exp Pharmacol Physiol 1989; 16:1.
  32. Olbricht CJ, Fink M, Gutjahr E. Alterations in lysosomal enzymes of the proximal tubule in gentamicin nephrotoxicity. Kidney Int 1991; 39:639.
  33. Ford DM, Dahl RH, Lamp CA, Molitoris BA. Apically and basolaterally internalized aminoglycosides colocalize in LLC-PK1 lysosomes and alter cell function. Am J Physiol 1994; 266:C52.
  34. Takano M, Ohishi Y, Okuda M, et al. Transport of gentamicin and fluid-phase endocytosis markers in the LLC-PK1 kidney epithelial cell line. J Pharmacol Exp Ther 1994; 268:669.
  35. Hori R, Okuda M, Ohishi Y, et al. Surface binding and intracellular uptake of gentamicin in the cultured kidney epithelial cell line (LLC-PK1). J Pharmacol Exp Ther 1992; 261:1200.
  36. Dagil R, O'Shea C, Nykjær A, et al. Gentamicin binds to the megalin receptor as a competitive inhibitor using the common ligand binding motif of complement type repeats: insight from the nmr structure of the 10th complement type repeat domain alone and in complex with gentamicin. J Biol Chem 2013; 288:4424.
  37. Spiegel DM, Shanley PF, Molitoris BA. Mild ischemia predisposes the S3 segment to gentamicin toxicity. Kidney Int 1990; 38:459.
  38. Wedeen RP, Batuman V, Cheeks C, et al. Transport of gentamicin in rat proximal tubule. Lab Invest 1983; 48:212.
  39. Silverblatt FJ, Kuehn C. Autoradiography of gentamicin uptake by the rat proximal tubule cell. Kidney Int 1979; 15:335.
  40. Servais H, Van Der Smissen P, Thirion G, et al. Gentamicin-induced apoptosis in LLC-PK1 cells: involvement of lysosomes and mitochondria. Toxicol Appl Pharmacol 2005; 206:321.
  41. Sandoval R, Leiser J, Molitoris BA. Aminoglycoside antibiotics traffic to the Golgi complex in LLC-PK1 cells. J Am Soc Nephrol 1998; 9:167.
  42. Sandvig K, Ryd M, Garred O, et al. Retrograde transport from the Golgi complex to the ER of both Shiga toxin and the nontoxic Shiga B-fragment is regulated by butyric acid and cAMP. J Cell Biol 1994; 126:53.
  43. Lord JM, Roberts LM. Toxin entry: retrograde transport through the secretory pathway. J Cell Biol 1998; 140:733.
  44. Sandoval RM, Molitoris BA. Gentamicin traffics retrograde through the secretory pathway and is released in the cytosol via the endoplasmic reticulum. Am J Physiol Renal Physiol 2004; 286:F617.
  45. Sandvig K, Garred O, van Helvoort A, et al. Importance of glycolipid synthesis for butyric acid-induced sensitization to shiga toxin and intracellular sorting of toxin in A431 cells. Mol Biol Cell 1996; 7:1391.
  46. Guo X, Nzerue C. How to prevent, recognize, and treat drug-induced nephrotoxicity. Cleve Clin J Med 2002; 69:289.
  47. Baciewicz AM, Sokos DR, Cowan RI. Aminoglycoside-associated nephrotoxicity in the elderly. Ann Pharmacother 2003; 37:182.
  48. Bailey TC, Little JR, Littenberg B, et al. A meta-analysis of extended-interval dosing versus multiple daily dosing of aminoglycosides. Clin Infect Dis 1997; 24:786.
  49. Bacopoulou F, Markantonis SL, Pavlou E, Adamidou M. A study of once-daily amikacin with low peak target concentrations in intensive care unit patients: pharmacokinetics and associated outcomes. J Crit Care 2003; 18:107.
  50. Fu X, Wan P, Li P, et al. Mechanism and Prevention of Ototoxicity Induced by Aminoglycosides. Front Cell Neurosci 2021; 15:692762.
  51. Ali BH. Agents ameliorating or augmenting experimental gentamicin nephrotoxicity: some recent research. Food Chem Toxicol 2003; 41:1447.
  52. Beauchamp D, Laurent G, Maldague P, et al. Protection against gentamicin-induced early renal alterations (phospholipidosis and increased DNA synthesis) by coadministration of poly-L-aspartic acid. J Pharmacol Exp Ther 1990; 255:858.
  53. Kishore BK, Lambricht P, Laurent G, et al. Mechanism of protection afforded by polyaspartic acid against gentamicin-induced phospholipidosis. II. Comparative in vitro and in vivo studies with poly-L-aspartic, poly-L-glutamic and poly-D-glutamic acids. J Pharmacol Exp Ther 1990; 255:875.
  54. Ramsammy LS, Josepovitz C, Lane BP, Kaloyanides GJ. Polyaspartic acid protects against gentamicin nephrotoxicity in the rat. J Pharmacol Exp Ther 1989; 250:149.
  55. Williams PD, Hottendorf GH, Bennett DB. Inhibition of renal membrane binding and nephrotoxicity of aminoglycosides. J Pharmacol Exp Ther 1986; 237:919.
  56. Kishore BK, Ibrahim S, Lambricht P, et al. Comparative assessment of poly-L-aspartic and poly-L-glutamic acids as protectants against gentamicin-induced renal lysosomal phospholipidosis, phospholipiduria and cell proliferation in rats. J Pharmacol Exp Ther 1992; 262:424.
  57. Josepovitz C, Pastoriza-Munoz E, Timmerman D, et al. Inhibition of gentamicin uptake in rat renal cortex in vivo by aminoglycosides and organic polycations. J Pharmacol Exp Ther 1982; 223:314.
  58. Kishore BK, Kállay Z, Lambricht P, et al. Mechanism of protection afforded by polyaspartic acid against gentamicin-induced phospholipidosis. I. Polyaspartic acid binds gentamicin and displaces it from negatively charged phospholipid layers in vitro. J Pharmacol Exp Ther 1990; 255:867.
  59. Ali BH. Gentamicin nephrotoxicity in humans and animals: some recent research. Gen Pharmacol 1995; 26:1477.
  60. Ben Ismail TH, Ali BH, Bashir AA. Influence of iron, deferoxamine and ascorbic acid on gentamicin-induced nephrotoxicity in rats. Gen Pharmacol 1994; 25:1249.
  61. Ali BH, Bashir AK. Effect of superoxide dismutase treatment on gentamicin nephrotoxicity in rats. Gen Pharmacol 1996; 27:349.
  62. Ali BH, Mousa HM. Effect of dimethyl sulfoxide on gentamicin-induced nephrotoxicity in rats. Hum Exp Toxicol 2001; 20:199.
  63. Sandhya P, Mohandass S, Varalakshmi P. Role of DL alpha-lipoic acid in gentamicin induced nephrotoxicity. Mol Cell Biochem 1995; 145:11.
  64. Mazzon E, Britti D, De Sarro A, et al. Effect of N-acetylcysteine on gentamicin-mediated nephropathy in rats. Eur J Pharmacol 2001; 424:75.
  65. Reiter RJ, Tan DX, Sainz RM, et al. Melatonin: reducing the toxicity and increasing the efficacy of drugs. J Pharm Pharmacol 2002; 54:1299.
  66. Ozbek E, Turkoz Y, Sahna E, et al. Melatonin administration prevents the nephrotoxicity induced by gentamicin. BJU Int 2000; 85:742.
  67. Medić B, Stojanović M, Rovčanin B, et al. Pioglitazone attenuates kidney injury in an experimental model of gentamicin-induced nephrotoxicity in rats. Sci Rep 2019; 9:13689.
  68. Hori Y, Aoki N, Kuwahara S, et al. Megalin Blockade with Cilastatin Suppresses Drug-Induced Nephrotoxicity. J Am Soc Nephrol 2017; 28:1783.
  69. Wagner MC, Sandoval RM, Yadav SPS, et al. Lrpap1 (RAP) Inhibits Proximal Tubule Clathrin Mediated and Clathrin Independent Endocytosis, Ameliorating Renal Aminoglycoside Nephrotoxicity. Kidney360 2023; 4:591.
  70. Sandoval RM, Reilly JP, Running W, et al. A non-nephrotoxic gentamicin congener that retains antimicrobial efficacy. J Am Soc Nephrol 2006; 17:2697.
  71. Nudelman I, Glikin D, Smolkin B, et al. Repairing faulty genes by aminoglycosides: development of new derivatives of geneticin (G418) with enhanced suppression of diseases-causing nonsense mutations. Bioorg Med Chem 2010; 18:3735.
  72. Wang D, Belakhov V, Kandasamy J, et al. The designer aminoglycoside NB84 significantly reduces glycosaminoglycan accumulation associated with MPS I-H in the Idua-W392X mouse. Mol Genet Metab 2012; 105:116.
  73. Rowe SM, Sloane P, Tang LP, et al. Suppression of CFTR premature termination codons and rescue of CFTR protein and function by the synthetic aminoglycoside NB54. J Mol Med (Berl) 2011; 89:1149.
  74. Nudelman I, Rebibo-Sabbah A, Cherniavsky M, et al. Development of novel aminoglycoside (NB54) with reduced toxicity and enhanced suppression of disease-causing premature stop mutations. J Med Chem 2009; 52:2836.
  75. Richardson R, Smart M, Tracey-White D, et al. Mechanism and evidence of nonsense suppression therapy for genetic eye disorders. Exp Eye Res 2017; 155:24.
  76. Baradaran-Heravi A, Niesser J, Balgi AD, et al. Gentamicin B1 is a minor gentamicin component with major nonsense mutation suppression activity. Proc Natl Acad Sci U S A 2017; 114:3479.
  77. Dobie RA, Black FO, Pezsnecker SC, Stallings VL. Hearing loss in patients with vestibulotoxic reactions to gentamicin therapy. Arch Otolaryngol Head Neck Surg 2006; 132:253.
  78. Huth ME, Ricci AJ, Cheng AG. Mechanisms of aminoglycoside ototoxicity and targets of hair cell protection. Int J Otolaryngol 2011; 2011:937861.
  79. Black FO, Pesznecker S, Stallings V. Permanent gentamicin vestibulotoxicity. Otol Neurotol 2004; 25:559.
  80. Basile AS, Huang JM, Xie C, et al. N-methyl-D-aspartate antagonists limit aminoglycoside antibiotic-induced hearing loss. Nat Med 1996; 2:1338.
  81. Ernfors P, Duan ML, ElShamy WM, Canlon B. Protection of auditory neurons from aminoglycoside toxicity by neurotrophin-3. Nat Med 1996; 2:463.
  82. Wu WJ, Sha SH, Schacht J. Recent advances in understanding aminoglycoside ototoxicity and its prevention. Audiol Neurootol 2002; 7:171.
  83. Sinswat P, Wu WJ, Sha SH, Schacht J. Protection from ototoxicity of intraperitoneal gentamicin in guinea pig. Kidney Int 2000; 58:2525.
  84. Prezant TR, Agapian JV, Bohlman MC, et al. Mitochondrial ribosomal RNA mutation associated with both antibiotic-induced and non-syndromic deafness. Nat Genet 1993; 4:289.
  85. Casano RA, Johnson DF, Bykhovskaya Y, et al. Inherited susceptibility to aminoglycoside ototoxicity: genetic heterogeneity and clinical implications. Am J Otolaryngol 1999; 20:151.
  86. Hamasaki K, Rando RR. Specific binding of aminoglycosides to a human rRNA construct based on a DNA polymorphism which causes aminoglycoside-induced deafness. Biochemistry 1997; 36:12323.
  87. Guan MX, Fischel-Ghodsian N, Attardi G. A biochemical basis for the inherited susceptibility to aminoglycoside ototoxicity. Hum Mol Genet 2000; 9:1787.
  88. Bitner-Glindzicz M, Pembrey M, Duncan A, et al. Prevalence of mitochondrial 1555A-->G mutation in European children. N Engl J Med 2009; 360:640.
  89. Vandebona H, Mitchell P, Manwaring N, et al. Prevalence of mitochondrial 1555A-->G mutation in adults of European descent. N Engl J Med 2009; 360:642.
  90. Aran JM. Current perspectives on inner ear toxicity. Otolaryngol Head Neck Surg 1995; 112:133.
  91. McDermott JH, Mahaveer A, James RA, et al. Rapid Point-of-Care Genotyping to Avoid Aminoglycoside-Induced Ototoxicity in Neonatal Intensive Care. JAMA Pediatr 2022; 176:486.
  92. Feldman L, Efrati S, Eviatar E, et al. Gentamicin-induced ototoxicity in hemodialysis patients is ameliorated by N-acetylcysteine. Kidney Int 2007; 72:359.
  93. Tokgoz B, Ucar C, Kocyigit I, et al. Protective effect of N-acetylcysteine from drug-induced ototoxicity in uraemic patients with CAPD peritonitis. Nephrol Dial Transplant 2011; 26:4073.
  94. Feldman L, Sherman RA, Weissgarten J. N-acetylcysteine use for amelioration of aminoglycoside-induced ototoxicity in dialysis patients. Semin Dial 2012; 25:491.
  95. Li PK, Chow KM, Cho Y, et al. ISPD peritonitis guideline recommendations: 2022 update on prevention and treatment. Perit Dial Int 2022; 42:110.
  96. Chen Y, Huang WG, Zha DJ, et al. Aspirin attenuates gentamicin ototoxicity: from the laboratory to the clinic. Hear Res 2007; 226:178.
  97. Sha SH, Qiu JH, Schacht J. Aspirin to prevent gentamicin-induced hearing loss. N Engl J Med 2006; 354:1856.
  98. Rybak LP, Ramkumar V. Ototoxicity. Kidney Int 2007; 72:931.
  99. Huth ME, Han KH, Sotoudeh K, et al. Designer aminoglycosides prevent cochlear hair cell loss and hearing loss. J Clin Invest 2015; 125:583.
  100. Ishikawa M, García-Mateo N, Čusak A, et al. Lower ototoxicity and absence of hidden hearing loss point to gentamicin C1a and apramycin as promising antibiotics for clinical use. Sci Rep 2019; 9:2410.
Topic 7242 Version 25.0

References

آیا می خواهید مدیلیب را به صفحه اصلی خود اضافه کنید؟