1
|
Menke AF, Heitplatz B, Van Marck V, Pavenstädt H, Jehn U. Hydroxychloroquine-Induced Renal Phospholipidosis: Case Report and Review of Differential Diagnoses. Case Rep Nephrol Dial 2024; 14:20-29. [PMID: 38370571 PMCID: PMC10871737 DOI: 10.1159/000536448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Accepted: 01/20/2024] [Indexed: 02/20/2024] Open
Abstract
Introduction Renal phospholipidosis describes the accumulation of phospholipids in the lysosomes of kidney cells, in particular podocytes. Originally, this was described primarily in the context of the lysosomal storage disorder Fabry disease. It is now known that a variety of drugs can lead to the accumulation of lysosomal phospholipids. Case Presentation We present the case of a 69-year-old female patient suffering chronic kidney disease and systemic lupus erythematosus who underwent a kidney biopsy because of a further increase in serum creatinine levels. There was no evidence of lupus nephritis, but electron microscopy showed zebra bodies as a morphological sign of phospholipidosis. This was most likely drug-induced after 25 years of continuous medication with hydroxychloroquine. A renal biopsy 2 years and 6 months earlier, when the renal function of the patient was distinctively better, showed no signs of renal phospholipidosis. Afterward, medication with hydroxychloroquine was discontinued, and renal function parameters remained stable in the 1-year course. Conclusion This case raises the question of how severely impaired renal function affects the risk of hydroxychloroquine-induced renal phospholipidosis and underlines that hydroxychloroquine should be administered with caution in patients with kidney insufficiency. Moreover, we provide a review of the causes of renal phospholipidosis, which have been described in the literature and give an overview of possible differential diagnoses in cases with histologically proven phospholipidosis in renal biopsies.
Collapse
Affiliation(s)
- Amélie Friederike Menke
- Division of General Internal Medicine, Nephrology and Rheumatology, Department of Medicine D, University Hospital of Münster, Münster, Germany
| | - Barbara Heitplatz
- Gerhard-Domagk-Institut of Pathology, University Hospital of Münster, Münster, Germany
| | - Veerle Van Marck
- Gerhard-Domagk-Institut of Pathology, University Hospital of Münster, Münster, Germany
| | - Hermann Pavenstädt
- Division of General Internal Medicine, Nephrology and Rheumatology, Department of Medicine D, University Hospital of Münster, Münster, Germany
| | - Ulrich Jehn
- Division of General Internal Medicine, Nephrology and Rheumatology, Department of Medicine D, University Hospital of Münster, Münster, Germany
| |
Collapse
|
2
|
Gould S, Templin MV. Off target toxicities and links with physicochemical properties of medicinal products, including antibiotics, oligonucleotides, lipid nanoparticles (with cationic and/or anionic charges). Data review suggests an emerging pattern. Toxicol Lett 2023; 384:14-29. [PMID: 37454775 DOI: 10.1016/j.toxlet.2023.07.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 07/10/2023] [Accepted: 07/12/2023] [Indexed: 07/18/2023]
Abstract
Toxicology is an essential part of any drug development plan. Circumnavigating the risk of failure because of a toxicity issue can be a challenge, and failure in late development is extremely costly. To identify potential risks, it requires more than just understanding the biological target. The toxicologist needs to consider a compound's structure, it's physicochemical properties (including the impact of the overall formulation), as well as the biological target (e.g., receptor interactions). Understanding the impact of the physicochemical properties can be used to predict potential toxicities in advance by incorporating key endpoints in early screening strategies and/or used to compare toxicity profiles across lead candidates. This review discussed the risks of off-target and/or non-specific toxicities that may be associated with the physicochemical properties of compounds, especially those carrying dominant positive or negative charges, including amphiphilic small molecules, peptides, oligonucleotides and lipids/liposomes/lipid nanoparticles. The latter of which are being seen more and more in drug development, including the recent Covid pandemic, where mRNA and lipid nanoparticle technology is playing more of a role in vaccine development. The translation between non-clinical and clinical data is also considered, questioning how a physicochemical driven toxicity may be more universal across species, which means that such toxicity may be reassuringly translatable between species and as such, this information may also be considered as a support to the 3 R's, particularly in the early screening stages of a drug development plan.
Collapse
|
3
|
Mally A, Jarzina S. Mapping Adverse Outcome Pathways for Kidney Injury as a Basis for the Development of Mechanism-Based Animal-Sparing Approaches to Assessment of Nephrotoxicity. FRONTIERS IN TOXICOLOGY 2022; 4:863643. [PMID: 35785263 PMCID: PMC9242087 DOI: 10.3389/ftox.2022.863643] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Accepted: 03/11/2022] [Indexed: 02/04/2023] Open
Abstract
In line with recent OECD activities on the use of AOPs in developing Integrated Approaches to Testing and Assessment (IATAs), it is expected that systematic mapping of AOPs leading to systemic toxicity may provide a mechanistic framework for the development and implementation of mechanism-based in vitro endpoints. These may form part of an integrated testing strategy to reduce the need for repeated dose toxicity studies. Focusing on kidney and in particular the proximal tubule epithelium as a key target site of chemical-induced injury, the overall aim of this work is to contribute to building a network of AOPs leading to nephrotoxicity. Current mechanistic understanding of kidney injury initiated by 1) inhibition of mitochondrial DNA polymerase γ (mtDNA Polγ), 2) receptor mediated endocytosis and lysosomal overload, and 3) covalent protein binding, which all present fairly well established, common mechanisms by which certain chemicals or drugs may cause nephrotoxicity, is presented and systematically captured in a formal description of AOPs in line with the OECD AOP development programme and in accordance with the harmonized terminology provided by the Collaborative Adverse Outcome Pathway Wiki. The relative level of confidence in the established AOPs is assessed based on evolved Bradford-Hill weight of evidence considerations of biological plausibility, essentiality and empirical support (temporal and dose-response concordance).
Collapse
|
4
|
Bejoy J, Qian ES, Woodard LE. Tissue Culture Models of AKI: From Tubule Cells to Human Kidney Organoids. J Am Soc Nephrol 2022; 33:487-501. [PMID: 35031569 PMCID: PMC8975068 DOI: 10.1681/asn.2021050693] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
AKI affects approximately 13.3 million people around the world each year, causing CKD and/or mortality. The mammalian kidney cannot generate new nephrons after postnatal renal damage and regenerative therapies for AKI are not available. Human kidney tissue culture systems can complement animal models of AKI and/or address some of their limitations. Donor-derived somatic cells, such as renal tubule epithelial cells or cell lines (RPTEC/hTERT, ciPTEC, HK-2, Nki-2, and CIHP-1), have been used for decades to permit drug toxicity screening and studies into potential AKI mechanisms. However, tubule cell lines do not fully recapitulate tubular epithelial cell properties in situ when grown under classic tissue culture conditions. Improving tissue culture models of AKI would increase our understanding of the mechanisms, leading to new therapeutics. Human pluripotent stem cells (hPSCs) can be differentiated into kidney organoids and various renal cell types. Injury to human kidney organoids results in renal cell-type crosstalk and upregulation of kidney injury biomarkers that are difficult to induce in primary tubule cell cultures. However, current protocols produce kidney organoids that are not mature and contain off-target cell types. Promising bioengineering techniques, such as bioprinting and "kidney-on-a-chip" methods, as applied to kidney nephrotoxicity modeling advantages and limitations are discussed. This review explores the mechanisms and detection of AKI in tissue culture, with an emphasis on bioengineered approaches such as human kidney organoid models.
Collapse
Affiliation(s)
- Julie Bejoy
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Eddie S. Qian
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Lauren E. Woodard
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee,Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville, Tennessee,Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee
| |
Collapse
|
5
|
Choung HYG, Jean-Gilles J, Goldman B. Myeloid bodies is not an uncommon ultrastructural finding. Ultrastruct Pathol 2022; 46:130-138. [PMID: 35100945 DOI: 10.1080/01913123.2021.2022054] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
The presence of myeloid bodies (MBs) is classically associated with Fabry disease (FD). However, MBs are also identified in patients without clinical evidence of FD. We attempt to further understand the clinicopathologic significance of incidental MBs in those without FD. Among the 4400 renal biopsies accessioned at the University of Rochester Medical Center from 2010 to 2021, we identified 32 cases showing MBs, 6 of which had FD. Medications were compared between a non-FG and a control-group of randomly selected cases without MBs (non-MBs). Both Fabry-group (FG) and non-Fabry-group (non-FG) were predominantly middle-aged (mean 48 years vs 56, respectively). Non-FG had slight female predominance (1:4), while all in FG were female. The majority of both non-FG and non-MBs cohort were on the same medications reported to cause phospholipidosis except sertraline and hydralazine (p = .04), which were more frequent in non-FG. Ultrastructurally, non-FG tended to show focal MBs in predominantly podocytes, while FG showed more extensive MBs in not only podocytes but also parietal, tubular, endothelial, and myocyte cells (p = .03). In addition, half of FG had another superimposed renal disease including kappa-light chain deposition disease, thin-basement membrane nephropathy, and lithium-related changes. MBs are encountered not only in FD but in other settings including CADs, toxins, and other inheritable diseases. Although secondary causes of MBs typically show less extensive involvement compared to FD, these features overlap. Given the challenges in diagnosing female carriers, the finding of MBs, though not specific to FD, may be the only clue that leads to further work-up and timely diagnosis, underscoring the importance of considering FD among other etiologies in differential diagnosis.
Collapse
Affiliation(s)
- Hae Yoon Grace Choung
- Department of Pathology and Laboratory Medicine, Division of Renal Pathology and Electron Microscopy, University of Rochester Medical Center, Rochester, NY, USA
| | - Jerome Jean-Gilles
- Department of Pathology and Laboratory Medicine, Division of Renal Pathology and Electron Microscopy, University of Rochester Medical Center, Rochester, NY, USA
| | - Bruce Goldman
- Department of Pathology and Laboratory Medicine, Division of Renal Pathology and Electron Microscopy, University of Rochester Medical Center, Rochester, NY, USA
| |
Collapse
|
6
|
Hinkovska-Galcheva V, Treadwell T, Shillingford JM, Lee A, Abe A, Tesmer JJG, Shayman JA. Inhibition of lysosomal phospholipase A2 predicts drug-induced phospholipidosis. J Lipid Res 2021; 62:100089. [PMID: 34087196 PMCID: PMC8243516 DOI: 10.1016/j.jlr.2021.100089] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 05/26/2021] [Accepted: 05/27/2021] [Indexed: 11/17/2022] Open
Abstract
Phospholipidosis, the excessive accumulation of phospholipids within lysosomes, is a pathological response observed following exposure to many drugs across multiple therapeutic groups. A clear mechanistic understanding of the causes and implications of this form of drug toxicity has remained elusive. We previously reported the discovery and characterization of a lysosome-specific phospholipase A2 (PLA2G15) and later reported that amiodarone, a known cause of drug-induced phospholipidosis, inhibits this enzyme. Here, we assayed a library of 163 drugs for inhibition of PLA2G15 to determine whether this phospholipase was the cellular target for therapeutics other than amiodarone that cause phospholipidosis. We observed that 144 compounds inhibited PLA2G15 activity. Thirty-six compounds not previously reported to cause phospholipidosis inhibited PLA2G15 with IC50 values less than 1 mM and were confirmed to cause phospholipidosis in an in vitro assay. Within this group, fosinopril was the most potent inhibitor (IC50 0.18 μM). Additional characterization of the inhibition of PLA2G15 by fosinopril was consistent with interference of PLA2G15 binding to liposomes. PLA2G15 inhibition was more accurate in predicting phospholipidosis compared with in silico models based on pKa and ClogP, measures of protonation, and transport-independent distribution in the lysosome, respectively. In summary, PLA2G15 is a primary target for cationic amphiphilic drugs that cause phospholipidosis, and PLA2G15 inhibition by cationic amphiphilic compounds provides a potentially robust screening platform for potential toxicity during drug development.
Collapse
Affiliation(s)
- Vania Hinkovska-Galcheva
- Department of Internal Medicine, University of Michigan Medical School, University of Michigan, Ann Arbor, MI, USA
| | - Taylour Treadwell
- Department of Internal Medicine, University of Michigan Medical School, University of Michigan, Ann Arbor, MI, USA
| | - Jonathan M Shillingford
- Department of Internal Medicine, University of Michigan Medical School, University of Michigan, Ann Arbor, MI, USA
| | - Angela Lee
- Department of Internal Medicine, University of Michigan Medical School, University of Michigan, Ann Arbor, MI, USA
| | - Akira Abe
- Department of Internal Medicine, University of Michigan Medical School, University of Michigan, Ann Arbor, MI, USA
| | - John J G Tesmer
- Departments of Biological Sciences and Medicinal Chemistry and Pharmacology, Purdue University, West Lafayette, IN, USA
| | - James A Shayman
- Department of Internal Medicine, University of Michigan Medical School, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
7
|
Dabrowski M, Bukowy-Bieryllo Z, Jackson CL, Zietkiewicz E. Properties of Non-Aminoglycoside Compounds Used to Stimulate Translational Readthrough of PTC Mutations in Primary Ciliary Dyskinesia. Int J Mol Sci 2021; 22:ijms22094923. [PMID: 34066907 PMCID: PMC8125088 DOI: 10.3390/ijms22094923] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Revised: 04/28/2021] [Accepted: 05/03/2021] [Indexed: 12/30/2022] Open
Abstract
Primary ciliary dyskinesia (PCD) is a rare disease with autosomal recessive inheritance, caused mostly by bi-allelic gene mutations that impair motile cilia structure and function. Currently, there are no causal treatments for PCD. In many disease models, translational readthrough of premature termination codons (PTC-readthrough) induced by aminoglycosides has been proposed as an effective way of restoring functional protein expression and reducing disease symptoms. However, variable outcomes of pre-clinical trials and toxicity associated with long-term use of aminoglycosides prompt the search for other compounds that might overcome these problems. Because a high proportion of PCD-causing variants are nonsense mutations, readthrough therapies are an attractive option. We tested a group of chemical compounds with known PTC-readthrough potential (ataluren, azithromycin, tylosin, amlexanox, and the experimental compound TC007), collectively referred to as non-aminoglycosides (NAGs). We investigated their PTC-readthrough efficiency in six PTC mutations found in Polish PCD patients, in the context of cell and cilia health, and in comparison to the previously tested aminoglycosides. The NAGs did not compromise the viability of the primary nasal respiratory epithelial cells, and the ciliary beat frequency was retained, similar to what was observed for gentamicin. In HEK293 cells transfected with six PTC-containing inserts, the tested compounds stimulated PTC-readthrough but with lower efficiency than aminoglycosides. The study allowed us to select compounds with minimal negative impact on cell viability and function but still the potential to induce PTC-readthrough.
Collapse
Affiliation(s)
- Maciej Dabrowski
- Institute of Human Genetics, Polish Academy of Sciences, 60-479 Poznan, Poland; (M.D.); (Z.B.-B.)
| | - Zuzanna Bukowy-Bieryllo
- Institute of Human Genetics, Polish Academy of Sciences, 60-479 Poznan, Poland; (M.D.); (Z.B.-B.)
| | - Claire L. Jackson
- Primary Ciliary Dyskinesia Centre, NIHR Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, Southampton SO16 6YD, UK;
- School of Clinical and Experimental Sciences, University of Southampton Faculty of Medicine, Southampton SO16 6YD, UK
| | - Ewa Zietkiewicz
- Institute of Human Genetics, Polish Academy of Sciences, 60-479 Poznan, Poland; (M.D.); (Z.B.-B.)
- Correspondence:
| |
Collapse
|
8
|
Vervaet BA, Nast CC, Schreurs G, Jayasumana C, Herath C, Kojc N, De Broe ME. Chronic interstitial nephritis in agricultural communities: a new perspective on etiology, diagnosis and mechanism. Nephrol Ther 2021; 17S:S45-S50. [PMID: 33910697 DOI: 10.1016/j.nephro.2020.02.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Accepted: 02/12/2020] [Indexed: 01/26/2023]
Abstract
Chronic interstitial nephritis in agricultural communities is a devastating kidney disease with a globally increasing prevalence. Its cause is unknown. Two predominant etiologies are hypothesised: recurrent episodes of dehydration and exposure to environmental toxins, such as agrochemicals and metals. In this review, we summarise arguments on: 1) why heat stress/dehydration is an unlikely cause of this disease and 2) why chronic interstitial nephritis in agricultural communities is to be considered a toxin-induced nephropathy. Mechanistically, we provide arguments for a putative role of pesticides on the one hand, and the calcineurin pathway on the other hand, both of which require further investigation. Finally, we summarise several important perspectives for research on chronic interstitial nephritis in agricultural communities.
Collapse
Affiliation(s)
- Benjamin A Vervaet
- Laboratory of Pathophysiology, University of Antwerp, Universiteitsplein 1, 2610 Antwerp, Belgium
| | | | - Gerd Schreurs
- Laboratory of Pathophysiology, University of Antwerp, Universiteitsplein 1, 2610 Antwerp, Belgium
| | - Channa Jayasumana
- Faculty of Medicine, Rajatrata University of Sri Lanka, Anuradhapura, Sri Lanka
| | - Chula Herath
- Sri Jayewardenepura General Hospital, Colombo, Sri Lanka
| | - Nika Kojc
- Department of Pathology, University of Ljubljana, Ljubljana, Slovenia
| | - Marc E De Broe
- Laboratory of Pathophysiology, University of Antwerp, Universiteitsplein 1, 2610 Antwerp, Belgium.
| |
Collapse
|
9
|
Green DM, Wang M, Krasin M, Srivastava D, Onder S, Jay DW, Ness KK, Greene W, Lanctot JQ, Shelton KC, Zhu L, Mulrooney DA, Ehrhardt MJ, Davidoff AM, Robison LL, Hudson MM. Kidney Function after Treatment for Childhood Cancer: A Report from the St. Jude Lifetime Cohort Study. J Am Soc Nephrol 2021; 32:983-993. [PMID: 33653686 PMCID: PMC8017532 DOI: 10.1681/asn.2020060849] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Accepted: 12/23/2020] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND Survivors of childhood cancer may be at increased risk for treatment-related kidney dysfunction. Although associations with acute kidney toxicity are well described, evidence informing late kidney sequelae is less robust. METHODS To define the prevalence of and risk factors for impaired kidney function among adult survivors of childhood cancer who had been diagnosed ≥10 years earlier, we evaluated kidney function (eGFR and proteinuria). We abstracted information from medical records about exposure to chemotherapeutic agents, surgery, and radiation treatment and evaluated the latter as the percentage of the total kidney volume treated with ≥5 Gy (V5), ≥10 Gy (V10), ≥15 Gy (V15), and ≥20 Gy (V20). We also used multivariable logistic regression models to assess demographic and clinical factors associated with impaired kidney function and Elastic Net to perform model selection for outcomes of kidney function. RESULTS Of the 2753 survivors, 51.3% were men, and 82.5% were non-Hispanic White. Median age at diagnosis was 7.3 years (interquartile range [IQR], 3.3-13.2), and mean age was 31.4 years (IQR, 25.8-37.8) at evaluation. Time from diagnosis was 23.2 years (IQR, 17.6-29.7). Approximately 2.1% had stages 3-5 CKD. Older age at evaluation; grade ≥2 hypertension; increasing cumulative dose of ifosfamide, cisplatin, or carboplatin; treatment ever with a calcineurin inhibitor; and volume of kidney irradiated to ≥5 or ≥10 Gy increased the odds for stages 3-5 CKD. Nephrectomy was significantly associated with stages 3-5 CKD in models for V15 or V20. CONCLUSIONS We found that 2.1% of our cohort of childhood cancer survivors had stages 3-5 CKD. These data may inform screening guidelines and new protocol development.
Collapse
Affiliation(s)
- Daniel M. Green
- Department of Epidemiology and Cancer Control, St. Jude Children’s Research Hospital, Memphis, Tennessee
- Department of Oncology, St. Jude Children’s Research Hospital, Memphis, Tennessee
| | - Mingjuan Wang
- Biostatistics, St. Jude Children’s Research Hospital, Memphis, Tennessee
| | - Matthew Krasin
- Department of Radiation Oncology, St. Jude Children’s Research Hospital, Memphis, Tennessee
| | | | - Songul Onder
- Division of Nephrology, Department of Medicine, University of Tennessee Health Sciences Center, Memphis, Tennessee
- Division of Nephrology, Department of Pediatrics, LeBonheur Children’s Hospital, Memphis, Tennessee
| | - Dennis W. Jay
- Department of Pathology, St. Jude Children’s Research Hospital, Memphis, Tennessee
| | - Kirsten K. Ness
- Department of Epidemiology and Cancer Control, St. Jude Children’s Research Hospital, Memphis, Tennessee
| | - William Greene
- Department of Pharmaceutical Sciences, St. Jude Children’s Research Hospital, Memphis, Tennessee
| | - Jennifer Q. Lanctot
- Department of Epidemiology and Cancer Control, St. Jude Children’s Research Hospital, Memphis, Tennessee
| | - Kyla C. Shelton
- Department of Epidemiology and Cancer Control, St. Jude Children’s Research Hospital, Memphis, Tennessee
| | - Liang Zhu
- Biostatistics, St. Jude Children’s Research Hospital, Memphis, Tennessee
| | - Daniel A. Mulrooney
- Department of Epidemiology and Cancer Control, St. Jude Children’s Research Hospital, Memphis, Tennessee
- Department of Oncology, St. Jude Children’s Research Hospital, Memphis, Tennessee
- Department of Pediatrics, University of Tennessee Health Sciences Center, Memphis, Tennessee
| | - Matthew J. Ehrhardt
- Department of Epidemiology and Cancer Control, St. Jude Children’s Research Hospital, Memphis, Tennessee
- Department of Oncology, St. Jude Children’s Research Hospital, Memphis, Tennessee
| | - Andrew M. Davidoff
- Department of Pediatrics, University of Tennessee Health Sciences Center, Memphis, Tennessee
- Department of Surgery, St. Jude Children’s Research Hospital, Memphis, Tennessee
| | - Leslie L. Robison
- Department of Epidemiology and Cancer Control, St. Jude Children’s Research Hospital, Memphis, Tennessee
| | - Melissa M. Hudson
- Department of Epidemiology and Cancer Control, St. Jude Children’s Research Hospital, Memphis, Tennessee
- Department of Oncology, St. Jude Children’s Research Hospital, Memphis, Tennessee
- Department of Pediatrics, University of Tennessee Health Sciences Center, Memphis, Tennessee
| |
Collapse
|
10
|
Vervaet BA, Nast CC, Schreurs G, De Broe ME. The authors reply. Kidney Int 2020; 97:1058-1059. [PMID: 32331581 DOI: 10.1016/j.kint.2020.01.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Accepted: 01/23/2020] [Indexed: 10/24/2022]
Affiliation(s)
| | - Cynthia C Nast
- Department of Pathology, Cedars Sinai Medical Center, Los Angeles, California, USA
| | - Gerd Schreurs
- Laboratory of Pathophysiology, University of Antwerp, Antwerp, Belgium
| | - Marc E De Broe
- Laboratory of Pathophysiology, University of Antwerp, Antwerp, Belgium
| |
Collapse
|
11
|
Downes KJ, Hayes M, Fitzgerald JC, Pais GM, Liu J, Zane NR, Goldstein SL, Scheetz MH, Zuppa AF. Mechanisms of antimicrobial-induced nephrotoxicity in children. J Antimicrob Chemother 2020; 75:1-13. [PMID: 31369087 PMCID: PMC6910165 DOI: 10.1093/jac/dkz325] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Drug-induced nephrotoxicity is responsible for 20% to 60% of cases of acute kidney injury in hospitalized patients and is associated with increased morbidity and mortality in both children and adults. Antimicrobials are one of the most common classes of medications prescribed globally and also among the most common causes of nephrotoxicity. A broad range of antimicrobial agents have been associated with nephrotoxicity, but the features of kidney injury vary based on the agent, its mechanism of injury and the site of toxicity within the kidney. Distinguishing nephrotoxicity caused by an antimicrobial agent from other potential inciting factors is important to facilitate both early recognition of drug toxicity and prompt cessation of an offending drug, as well as to avoid unnecessary discontinuation of an innocuous therapy. This review will detail the different types of antimicrobial-induced nephrotoxicity: acute tubular necrosis, acute interstitial nephritis and obstructive nephropathy. It will also describe the mechanism of injury caused by specific antimicrobial agents and classes (vancomycin, aminoglycosides, polymyxins, antivirals, amphotericin B), highlight the toxicodynamics of these drugs and provide guidance on administration or monitoring practices that can mitigate toxicity, when known. Particular attention will be paid to paediatric patients, when applicable, in whom nephrotoxin exposure is an often-underappreciated cause of kidney injury.
Collapse
Affiliation(s)
- Kevin J Downes
- Department of Pediatrics, The University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Division of Infectious Diseases, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Center for Clinical Pharmacology, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Center for Pediatric Clinical Effectiveness, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Molly Hayes
- Antimicrobial Stewardship Program, Center for Healthcare Quality & Analytics, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Julie C Fitzgerald
- Division of Critical Care Medicine, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Anesthesiology and Critical Care, The University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Gwendolyn M Pais
- Department of Pharmacy Practice, Chicago College of Pharmacy, Midwestern University, Downers Grove, IL, USA
- Pharmacometrics Center of Excellence, Chicago College of Pharmacy, Midwestern University, Downers Grove, IL, USA
| | - Jiajun Liu
- Department of Pharmacy Practice, Chicago College of Pharmacy, Midwestern University, Downers Grove, IL, USA
- Pharmacometrics Center of Excellence, Chicago College of Pharmacy, Midwestern University, Downers Grove, IL, USA
- Department of Pharmacy, Northwestern Memorial Hospital, Chicago, IL, USA
| | - Nicole R Zane
- Center for Clinical Pharmacology, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Stuart L Goldstein
- Department of Pediatrics, University of Cincinnati, Cincinnati, OH, USA
- Center for Acute Care Nephrology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| | - Marc H Scheetz
- Department of Pharmacy Practice, Chicago College of Pharmacy, Midwestern University, Downers Grove, IL, USA
- Pharmacometrics Center of Excellence, Chicago College of Pharmacy, Midwestern University, Downers Grove, IL, USA
- Department of Pharmacy, Northwestern Memorial Hospital, Chicago, IL, USA
- Department of Pharmacology, College of Graduate Studies, Midwestern University, Downers Grove, IL, USA
| | - Athena F Zuppa
- Center for Clinical Pharmacology, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Division of Critical Care Medicine, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Anesthesiology and Critical Care, The University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| |
Collapse
|
12
|
Vervaet BA, Nast CC, Jayasumana C, Schreurs G, Roels F, Herath C, Kojc N, Samaee V, Rodrigo S, Gowrishankar S, Mousson C, Dassanayake R, Orantes CM, Vuiblet V, Rigothier C, D'Haese PC, De Broe ME. Chronic interstitial nephritis in agricultural communities is a toxin-induced proximal tubular nephropathy. Kidney Int 2019; 97:350-369. [PMID: 31892415 DOI: 10.1016/j.kint.2019.11.009] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Revised: 09/09/2019] [Accepted: 10/10/2019] [Indexed: 12/16/2022]
Abstract
Almost 30 years after the detection of chronic interstitial nephritis in agricultural communities (CINAC) its etiology remains unknown. To help define this we examined 34 renal biopsies from Sri Lanka, El Salvador, India and France of patients with chronic kidney disease 2-3 and diagnosed with CINAC by light and electron microscopy. In addition to known histopathology, we identified a unique constellation of proximal tubular cell findings including large dysmorphic lysosomes with a light-medium electron-dense matrix containing dispersed dark electron-dense non-membrane bound "aggregates". These aggregates associated with varying degrees of cellular/tubular atrophy, apparent cell fragment shedding and no-weak proximal tubular cell proliferative capacity. Identical lysosomal lesions, identifiable by electron microscopy, were observed in 9% of renal transplant implantation biopsies, but were more prevalent in six month (50%) and 12 month (67%) protocol biopsies and in indication biopsies (76%) of calcineurin inhibitor treated transplant patients. The phenotype was also found associated with nephrotoxic drugs (lomustine, clomiphene, lithium, cocaine) and in some patients with light chain tubulopathy, all conditions that can be directly or indirectly linked to calcineurin pathway inhibition or modulation. One hundred biopsies of normal kidneys, drug/toxin induced nephropathies, and overt proteinuric patients of different etiologies to some extent could demonstrate the light microscopic proximal tubular cell changes, but rarely the electron microscopic lysosomal features. Rats treated with the calcineurin inhibitor cyclosporine for four weeks developed similar proximal tubular cell lysosomal alterations, which were absent in a dehydration group. Overall, the finding of an identical proximal tubular cell (lysosomal) lesion in CINAC and calcineurin inhibitor nephrotoxicity in different geographic regions suggests a common paradigm where CINAC patients undergo a tubulotoxic mechanism similar to calcineurin inhibitor nephrotoxicity.
Collapse
Affiliation(s)
| | - Cynthia C Nast
- Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Channa Jayasumana
- Faculty of Medicine, Rajatrata University of Sri Lanka, Anuradhapura, Sri Lanka
| | - Gerd Schreurs
- Laboratory of Pathophysiology, University Antwerp, Antwerp, Belgium
| | - Frank Roels
- Department of Pathology, Ghent University, Gent, Belgium
| | - Chula Herath
- Department of Nephrology, Sri Jayewardenepura General Hospital, Colombo, Sri Lanka
| | - Nika Kojc
- Institute of Pathology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Vahid Samaee
- Electron Microscopy for Materials Science (EMAT), University of Antwerp, Antwerp, Belgium
| | - Sonali Rodrigo
- Department of Nephrology, Sri Jayewardenepura General Hospital, Colombo, Sri Lanka
| | | | - Christiane Mousson
- Department of Nephrology, Centre Hospitalier Universitaire de Dijon, Dijon, France
| | | | - Carlos M Orantes
- National Institute of Health, Ministry of Health of El Salvador, San Salvador, El Salvador
| | - Vincent Vuiblet
- Departments of Nephrology and Renal Pathology, Centre Hospitalier Universitaire de Reims, Reims, France
| | - Claire Rigothier
- Service Néphrologie, Transplantation, Dialyse et Aphérèses, Centre Hospitalier Universitaire de Bordeaux, Bordeaux, France
| | | | - Marc E De Broe
- Laboratory of Pathophysiology, University Antwerp, Antwerp, Belgium.
| |
Collapse
|
13
|
Hayward RS, Harding J, Molloy R, Land L, Longcroft-Neal K, Moore D, Ross JDC. Adverse effects of a single dose of gentamicin in adults: a systematic review. Br J Clin Pharmacol 2017; 84:223-238. [PMID: 28940715 DOI: 10.1111/bcp.13439] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2017] [Revised: 09/01/2017] [Accepted: 09/09/2017] [Indexed: 01/15/2023] Open
Abstract
AIMS To systematically review the frequency and type of adverse events associated with a single dose of intravenous or intramuscular gentamicin in adults, for any indication, in studies where a comparator was available. METHODS A review protocol was developed and registered (PROSPERO: CRD42013003229). Studies were eligible for review if they: recruited participants aged ≥16 years; used gentamicin intramuscularly or intravenously as a single one-off dose; compared gentamicin to another medication or placebo; and monitored adverse events. MEDLINE, EMBASE, Cochrane Library, trial registries, conference proceedings and other relevant databases were searched up to November 2016. Risk of bias was assessed on all included studies. RESULTS In total, 15 522 records were identified. After removal of duplicates, screening of title/abstracts for relevance and independent selection of full texts by two reviewers, 36 studies were included. Across all the included studies, 24 107 participants received a single one-off dose of gentamicin (doses ranged from 1 mg kg-1 to 480 mg per dose). Acute kidney injury was described in 2520 participants receiving gentamicin. The large majority of cases were reversible. There were no cases of ototoxicity reported in patients receiving gentamicin. A meta-analysis was not performed due to study heterogeneity. CONCLUSIONS A significant number of patients saw a transient rise in creatinine after a single dose of gentamicin at doses up to 480 mg. Persistent renal impairment and other adverse events were relatively rare.
Collapse
Affiliation(s)
- Rachel S Hayward
- Whittall Street Clinic, University Hospitals Birmingham NHS Trust, Birmingham, UK
| | - Jan Harding
- Whittall Street Clinic, University Hospitals Birmingham NHS Trust, Birmingham, UK
| | - Rob Molloy
- Whittall Street Clinic, University Hospitals Birmingham NHS Trust, Birmingham, UK
| | - Lucy Land
- Faculty of Health, Education and Life Sciences, Birmingham City University, Birmingham, UK
| | - Kate Longcroft-Neal
- Whittall Street Clinic, University Hospitals Birmingham NHS Trust, Birmingham, UK
| | - David Moore
- Institute of Applied Health Research, University of Birmingham, Birmingham, UK
| | - Jonathan D C Ross
- Whittall Street Clinic, University Hospitals Birmingham NHS Trust, Birmingham, UK
| |
Collapse
|
14
|
Abstract
Three pulmonary disease conditions result from the accumulation of phospholipids in the lung. These conditions are the human lung disease known as pulmonary alveolar proteinosis, the lipoproteinosis that arises in the lungs of rats during acute silicosis, and the phospholipidoses induced by numerous cationic amphiphilic therapeutic agents. In this paper, the status of phospholipid metabolism in the lungs during the process of each of these lung conditions has been reviewed and possible mechanisms for their establishment are discussed. Pulmonary alveolar proteinosis is characterized by the accumulation of tubular myelin-like multilamellated structures in the alveoli and distal airways of patients. These structures appear to be formed by a process of spontaneous assembly involving surfactant protein A and surfactant phospholipids. Structures similar to tubular myelin-like multilamellated structures can be seen in the alveoli of rats during acute silicosis and, as with the human condition, both surfactant protein A and surfactant phospholipids accumulate in the alveoli. Excessive accumulation of surfactant protein A and surfactant phospholipids in the alveoli could arise from their overproduction and hypersecretion by a subpopulation of Type II cells that are activated by silica, and possibly other agents. Phospholipidoses caused by cationic amphiphilic therapeutic agents arise as a result of their inhibition of phospholipid catabolism. Inhibition of phospholipases results in the accumulation of phospholipids in the cytoplasm of alveolar macrophages and other cells. While inhibition of phospholipases by these agents undoubtedly occurs, there are many anomalous features, such as the accumulation of extracellular phospholipids and surfactant protein A, that cannot be accounted for by this simplistic hypothesis.
Collapse
Affiliation(s)
- Gary E. R. Hook
- Biochemical Pathology Group, Laboratory of Pulmonary Pathobiology, National Institute of Environmental Health Sciences, P.O. Box 12233, Research Triangle Park, North Carolina 27709
| |
Collapse
|
15
|
Randjelovic P, Veljkovic S, Stojiljkovic N, Sokolovic D, Ilic I. Gentamicin nephrotoxicity in animals: Current knowledge and future perspectives. EXCLI JOURNAL 2017; 16:388-399. [PMID: 28507482 PMCID: PMC5427480 DOI: 10.17179/excli2017-165] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Accepted: 03/07/2017] [Indexed: 12/16/2022]
Abstract
Due to high relative blood flow the kidney is prone to drug-induced damage. Aminoglycoside type antibiotic gentamicin is one of the leading cause of drug-induced nephrotoxicity. In recent years gentamicin nephrotoxicity is significantly reduced by shifting to once daily dosage as well as by eliminating known risk factors. Application of gentamicin is still related to serious side effects which are reported more often compared to other antibiotics. Because gentamicin is still heavily used and is highly efficient in treating infections, it is important to find mechanisms to reduce its nephrotoxicity. This aim can only be achieved through better understanding of kidney metabolism of gentamicin. This problem has been extensively researched in the last 20 years. The experimental results have provided evidence for almost complete understanding of mechanisms responsible for gentamicin nephrotoxicity. We now have well described morphological, biochemical and functional changes in kidney due to gentamicin application. During the years, this model has become so popular that now it is used as an experimental model for nephrotoxicity per se. This situation can mislead an ordinary reader of scientific literature that we know everything about it and there is nothing new to discover here. But quite opposite is true. The precise and complete mechanism of gentamicin nephrotoxicity is still point of speculation and an unfinished story. With emerge of new and versatile technics in biomedicine we have an opportunity to reexamine old beliefs and discover new facts. This review focuses on current knowledge in this area and gives some future perspectives.
Collapse
Affiliation(s)
- Pavle Randjelovic
- University of Niš, Faculty of Medicine, Department of Physiology, Niš, Serbia
| | - Slavimir Veljkovic
- University of Niš, Faculty of Medicine, Department of Physiology, Niš, Serbia
| | - Nenad Stojiljkovic
- University of Niš, Faculty of Medicine, Department of Physiology, Niš, Serbia
| | - Dušan Sokolovic
- University of Niš, Faculty of Medicine, Department of Biochemistry, Niš, Serbia
| | - Ivan Ilic
- University of Niš, Faculty of Medicine, Institute of Pathology, Niš, Serbia
| |
Collapse
|
16
|
Keeling KM. Nonsense Suppression as an Approach to Treat Lysosomal Storage Diseases. Diseases 2016; 4:32. [PMID: 28367323 PMCID: PMC5370586 DOI: 10.3390/diseases4040032] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Accepted: 10/14/2016] [Indexed: 02/08/2023] Open
Abstract
In-frame premature termination codons (PTCs) (also referred to as nonsense mutations) comprise ~10% of all disease-associated gene lesions. PTCs reduce gene expression in two ways. First, PTCs prematurely terminate translation of an mRNA, leading to the production of a truncated polypeptide that often lacks normal function and/or is unstable. Second, PTCs trigger degradation of an mRNA by activating nonsense-mediated mRNA decay (NMD), a cellular pathway that recognizes and degrades mRNAs containing a PTC. Thus, translation termination and NMD are putative therapeutic targets for the development of treatments for genetic diseases caused by PTCs. Over the past decade, significant progress has been made in the identification of compounds with the ability to suppress translation termination of PTCs (also referred to as readthrough). More recently, NMD inhibitors have also been explored as a way to enhance the efficiency of PTC suppression. Due to their relatively low threshold for correction, lysosomal storage diseases are a particularly relevant group of diseases to investigate the feasibility of nonsense suppression as a therapeutic approach. In this review, the current status of PTC suppression and NMD inhibition as potential treatments for lysosomal storage diseases will be discussed.
Collapse
Affiliation(s)
- Kim M Keeling
- Department of Biochemistry and Molecular Genetics, Gregory Fleming Cystic Fibrosis Research Center, Comprehensive Arthritis, Musculoskeletal, Bone, and Autoimmunity Center, University of Alabama at Birmingham, Birmingham, AL 35294, USA; ; Tel.: +1-205-975-6585
| |
Collapse
|
17
|
Bukowy-Bieryllo Z, Dabrowski M, Witt M, Zietkiewicz E. Aminoglycoside-stimulated readthrough of premature termination codons in selected genes involved in primary ciliary dyskinesia. RNA Biol 2016; 13:1041-1050. [PMID: 27618201 PMCID: PMC5056772 DOI: 10.1080/15476286.2016.1219832] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Translational readthrough of premature termination codons (PTCs) induced by pharmacological compounds has proven to be an effective way of restoring functional protein expression and reducing symptoms in several genetic disorders. We tested the potential of different concentrations of several aminoglycosides (AAGs) for promoting PTC-readthrough in 5 genes involved in the pathogenesis of primary ciliary dyskinesia, an inherited disorder caused by the dysfunction of motile cilia and flagella. The efficiency of readthrough stimulation of PTCs cloned in dual reporter vectors was examined in 2 experimental settings: in vitro (transcription/translation system) and ex vivo (transiently transfected epithelial cell line). PTC-readthrough was observed in 5 of the 16 mutations analyzed. UGA codons were more susceptible to AAG-stimulated readthrough than UAG; no suppression of UAA was observed. The efficiency of PTC-readthrough in vitro (from less than 1% to ∼28% of the translation from the corresponding wild-type constructs) differed with the AAG type and concentration, and depended on the combination of AAG and PTC, indicating that each PTC has to be individually tested with a range of stimulating compounds. The maximal values of PTC suppression observed in the ex vivo experiments were, depending on AAG used, 3–5 times lower than the corresponding values in vitro, despite using AAG concentrations that were 2 orders of magnitude higher. This indicates that, while the in vitro system is sufficient to examine the readthrough-susceptibility of PTCs, it is not sufficient to test the compounds potential to stimulate PTC-readthrough in the living cells. Most of the tested compounds (except for G418) at their highest concentrations did not disturb ciliogenesis in the cultures of primary respiratory epithelial cells from healthy donors.
Collapse
Affiliation(s)
| | - Maciej Dabrowski
- a Institute of Human Genetics, Polish Academy of Sciences , Poznan , Poland
| | - Michał Witt
- a Institute of Human Genetics, Polish Academy of Sciences , Poznan , Poland
| | - Ewa Zietkiewicz
- a Institute of Human Genetics, Polish Academy of Sciences , Poznan , Poland
| |
Collapse
|
18
|
Cars O, Craig WA. Pharmacodynamics of Antibiotics-Consequences for Dosing: Proceedings of a Symposium Held in Stockholm, June 7–9, 1990. ACTA ACUST UNITED AC 2015. [DOI: 10.3109/inf.1990.22.suppl-74.01] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
19
|
Di-22:6-bis(monoacylglycerol)phosphate: A clinical biomarker of drug-induced phospholipidosis for drug development and safety assessment. Toxicol Appl Pharmacol 2014; 279:467-476. [PMID: 24967688 DOI: 10.1016/j.taap.2014.06.014] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2014] [Revised: 05/06/2014] [Accepted: 06/17/2014] [Indexed: 11/24/2022]
Abstract
The inability to routinely monitor drug-induced phospholipidosis (DIPL) presents a challenge in pharmaceutical drug development and in the clinic. Several nonclinical studies have shown di-docosahexaenoyl (22:6) bis(monoacylglycerol) phosphate (di-22:6-BMP) to be a reliable biomarker of tissue DIPL that can be monitored in the plasma/serum and urine. The aim of this study was to show the relevance of di-22:6-BMP as a DIPL biomarker for drug development and safety assessment in humans. DIPL shares many similarities with the inherited lysosomal storage disorder Niemann-Pick type C (NPC) disease. DIPL and NPC result in similar changes in lysosomal function and cholesterol status that lead to the accumulation of multi-lamellar bodies (myeloid bodies) in cells and tissues. To validate di-22:6-BMP as a biomarker of DIPL for clinical studies, NPC patients and healthy donors were classified by receiver operator curve analysis based on urinary di-22:6-BMP concentrations. By showing 96.7-specificity and 100-sensitivity to identify NPC disease, di-22:6-BMP can be used to assess DIPL in human studies. The mean concentration of di-22:6-BMP in the urine of NPC patients was 51.4-fold (p ≤ 0.05) above the healthy baseline range. Additionally, baseline levels of di-22:6-BMP were assessed in healthy non-medicated laboratory animals (rats, mice, dogs, and monkeys) and human subjects to define normal reference ranges for nonclinical/clinical studies. The baseline ranges of di-22:6-BMP in the plasma, serum, and urine of humans and laboratory animals were species dependent. The results of this study support the role of di-22:6-BMP as a biomarker of DIPL for pharmaceutical drug development and health care settings.
Collapse
|
20
|
Declèves AE, Zolkipli Z, Satriano J, Wang L, Nakayama T, Rogac M, Le TP, Nortier JL, Farquhar MG, Naviaux RK, Sharma K. Regulation of lipid accumulation by AMP-activated kinase [corrected] in high fat diet-induced kidney injury. Kidney Int 2014; 85:611-23. [PMID: 24304883 PMCID: PMC4244908 DOI: 10.1038/ki.2013.462] [Citation(s) in RCA: 173] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2013] [Revised: 09/18/2013] [Accepted: 09/19/2013] [Indexed: 11/08/2022]
Abstract
AMP-activated protein kinase (AMPK) is an important energy sensor that may be critical in regulating renal lipid accumulation. To evaluate the role of AMPK in mediating renal lipid accumulation, C57BL/6J mice were randomized to a standard diet, a high-fat diet, or a high-fat diet plus AICAR (an AMPK activator) for 14 weeks. Renal functional and structural studies along with electron microscopy were performed. Mice given the high-fat diet had proximal tubule injury with the presence of enlarged clear vacuoles, and multilaminar inclusions concurrent with an increase of tissue lipid and overloading of the lysosomal system. The margins of the clear vacuoles were positive for the endolysosomal marker, LAMP1, suggesting lysosome accumulation. Characterization of vesicles by special stains (Oil Red O, Nile Red, Luxol Fast Blue) and by electron microscopy showed they contained onion skin-like accumulations consistent with phospholipids. Moreover, cholesteryl esters and phosphatidylcholine-containing phospholipids were significantly increased in the kidneys of mice on a high-fat diet. AMPK activation with chronic AICAR treatment prevented the clinical and structural effects of high-fat diet. Thus, high-fat diet contributes to a dysfunction of the lysosomal system and altered lipid metabolism characterized by cholesterol and phospholipid accumulation in the kidney. AMPK activation normalizes the changes in renal lipid content despite chronic exposure to lipid challenge.
Collapse
Affiliation(s)
- Anne-Emilie Declèves
- Center for Renal Translational Medicine, Division of Nephrology-Hypertension, Department of Medicine, University of California, San Diego, California, USA
- Division of Nephrology-Hypertension, Veterans Affairs San Diego Healthcare System, Veterans Medical Research Foundation, San Diego, California, USA
- Laboratory of Experimental Nephrology, Faculty of Medicine, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Zarazuela Zolkipli
- The Mitochondrial and Metabolic Disease Center, Department of Medicine, University of California, San Diego, California, USA
| | - Joseph Satriano
- Center for Renal Translational Medicine, Division of Nephrology-Hypertension, Department of Medicine, University of California, San Diego, California, USA
- Division of Nephrology-Hypertension, Veterans Affairs San Diego Healthcare System, Veterans Medical Research Foundation, San Diego, California, USA
| | - Lin Wang
- The Mitochondrial and Metabolic Disease Center, Department of Medicine, University of California, San Diego, California, USA
| | - Tomohiro Nakayama
- The Mitochondrial and Metabolic Disease Center, Department of Medicine, University of California, San Diego, California, USA
| | - Mihael Rogac
- The Mitochondrial and Metabolic Disease Center, Department of Medicine, University of California, San Diego, California, USA
| | - Thuy P. Le
- The Mitochondrial and Metabolic Disease Center, Department of Medicine, University of California, San Diego, California, USA
| | - Joëlle L. Nortier
- Laboratory of Experimental Nephrology, Faculty of Medicine, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Marilyn G. Farquhar
- Department of Cellular and Molecular Medicine, Department of Medicine, University of California at San Diego, La Jolla, California, USA
| | - Robert K. Naviaux
- The Mitochondrial and Metabolic Disease Center, Department of Medicine, University of California, San Diego, California, USA
| | - Kumar Sharma
- Center for Renal Translational Medicine, Division of Nephrology-Hypertension, Department of Medicine, University of California, San Diego, California, USA
- Division of Nephrology-Hypertension, Veterans Affairs San Diego Healthcare System, Veterans Medical Research Foundation, San Diego, California, USA
| |
Collapse
|
21
|
Rahul, Jyoti S, Naz F, Siddique YH. Toxic effects of gentamicin in the third instar larvae of transgenic Drosophila melanogaster (hsp70-lacZ)Bg9. Toxicol Res (Camb) 2014. [DOI: 10.1039/c3tx50093d] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
|
22
|
Goracci L, Ceccarelli M, Bonelli D, Cruciani G. Modeling Phospholipidosis Induction: Reliability and Warnings. J Chem Inf Model 2013; 53:1436-46. [DOI: 10.1021/ci400113t] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Laura Goracci
- Laboratory
for Chemometrics and Cheminformatics, Chemistry
Department, University of Perugia, Via Elce di Sotto 8, I-06123 Perugia, Italy
| | - Martina Ceccarelli
- Laboratory
for Chemometrics and Cheminformatics, Chemistry
Department, University of Perugia, Via Elce di Sotto 8, I-06123 Perugia, Italy
| | - Daniela Bonelli
- Laboratory
for Chemometrics and Cheminformatics, Chemistry
Department, University of Perugia, Via Elce di Sotto 8, I-06123 Perugia, Italy
| | - Gabriele Cruciani
- Laboratory
for Chemometrics and Cheminformatics, Chemistry
Department, University of Perugia, Via Elce di Sotto 8, I-06123 Perugia, Italy
| |
Collapse
|
23
|
Li YC, Shih YM, Lee JA. Gentamicin caused renal injury deeply related to methylglyoxal and Nɛ-(carboxyethyl)lysine (CEL). Toxicol Lett 2013; 219:85-92. [DOI: 10.1016/j.toxlet.2013.01.024] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2012] [Revised: 01/18/2013] [Accepted: 01/28/2013] [Indexed: 01/18/2023]
|
24
|
Keeling KM, Wang D, Conard SE, Bedwell DM. Suppression of premature termination codons as a therapeutic approach. Crit Rev Biochem Mol Biol 2012; 47:444-63. [PMID: 22672057 PMCID: PMC3432268 DOI: 10.3109/10409238.2012.694846] [Citation(s) in RCA: 80] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
In this review, we describe our current understanding of translation termination and pharmacological agents that influence the accuracy of this process. A number of drugs have been identified that induce suppression of translation termination at in-frame premature termination codons (PTCs; also known as nonsense mutations) in mammalian cells. We discuss efforts to utilize these drugs to suppress disease-causing PTCs that result in the loss of protein expression and function. In-frame PTCs represent a genotypic subset of mutations that make up ~11% of all known mutations that cause genetic diseases, and millions of patients have diseases attributable to PTCs. Current approaches aimed at reducing the efficiency of translation termination at PTCs (referred to as PTC suppression therapy) have the goal of alleviating the phenotypic consequences of a wide range of genetic diseases. Suppression therapy is currently in clinical trials for treatment of several genetic diseases caused by PTCs, and preliminary results suggest that some patients have shown clinical improvements. While current progress is promising, we discuss various approaches that may further enhance the efficiency of this novel therapeutic approach.
Collapse
Affiliation(s)
- Kim M. Keeling
- Dept. of Microbiology, University of Alabama at Birmingham, Birmingham, AL, USA
- Gregory Fleming James Cystic Fibrosis Research Center, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Dan Wang
- Dept. of Microbiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Sara E. Conard
- Dept. of Microbiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - David M. Bedwell
- Dept. of Microbiology, University of Alabama at Birmingham, Birmingham, AL, USA
- Gregory Fleming James Cystic Fibrosis Research Center, University of Alabama at Birmingham, Birmingham, AL, USA
| |
Collapse
|
25
|
Stojiljkovic N, Stoiljkovic M, Mihailovic D, Randjelovic P, Ilic S, Gocmanac-Ignjatovic M, Veljkovic M. Beneficial Effects of Calcium Oral Coadministration in Gentamicin-Induced Nephrotoxicity in Rats. Ren Fail 2012; 34:622-7. [DOI: 10.3109/0886022x.2012.664809] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
26
|
Thompson KL, Haskins K, Rosenzweig BA, Stewart S, Zhang J, Peters D, Knapton A, Rouse R, Mans D, Colatsky T. Comparison of the Diagnostic Accuracy of Di-22:6-Bis(monoacylglycerol)Phosphate and Other Urinary Phospholipids for Drug-Induced Phospholipidosis or Tissue Injury in the Rat. Int J Toxicol 2012; 31:14-24. [DOI: 10.1177/1091581811430167] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Cationic amphiphilic drugs and aminoglycoside antibiotics can induce phospholipidosis (PLD), an abnormal accumulation of phospholipids in lysosome-derived vesicles, in preclinical studies. The incidence of PLD in patients and its clinical relevance are difficult to assess without noninvasive biomarkers. Di-docosahexaenoyl bis(monoacylglycerol)phosphate (di-22:6-BMP) is a phospholipid that is enriched in lysosomal membranes and a proposed urinary biomarker of drug-induced PLD. The specificity of di-22:6-BMP for PLD was compared to other phospholipid species that can increase in urine with nephrotoxicity. Using liquid chromatography coupled to mass spectrometry, 12 phospholipids were assayed in the urine of rats treated with drugs that induced PLD or caused renal or skeletal muscle injury. In receiver operating curve analyses, urinary di-22:6-BMP was a significantly better predictor of PLD and the least predictive of tissue injury of the phospholipids assayed. The data provide evidence supporting the use of di-22:6-BMP as a urinary biomarker of PLD in rats.
Collapse
Affiliation(s)
- Karol L. Thompson
- Division of Drug Safety Research, Center for Drug Evaluation and Research, FDA, Silver Spring, MD, USA
| | - Kylie Haskins
- Division of Drug Safety Research, Center for Drug Evaluation and Research, FDA, Silver Spring, MD, USA
- Present address: Emergent Biosolutions, Rockville, MD, USA
| | - Barry A. Rosenzweig
- Division of Drug Safety Research, Center for Drug Evaluation and Research, FDA, Silver Spring, MD, USA
| | - Sharron Stewart
- Division of Drug Safety Research, Center for Drug Evaluation and Research, FDA, Silver Spring, MD, USA
| | - Jun Zhang
- Division of Drug Safety Research, Center for Drug Evaluation and Research, FDA, Silver Spring, MD, USA
| | - David Peters
- Division of Drug Safety Research, Center for Drug Evaluation and Research, FDA, Silver Spring, MD, USA
| | - Alan Knapton
- Division of Drug Safety Research, Center for Drug Evaluation and Research, FDA, Silver Spring, MD, USA
| | - Rodney Rouse
- Division of Drug Safety Research, Center for Drug Evaluation and Research, FDA, Silver Spring, MD, USA
| | - Daniel Mans
- Division of Pharmaceutical Analysis, CDER, FDA, St. Louis, MO, USA
| | - Thomas Colatsky
- Division of Drug Safety Research, Center for Drug Evaluation and Research, FDA, Silver Spring, MD, USA
| |
Collapse
|
27
|
Abstract
PURPOSE OF REVIEW Ingestions and iatrogenic administration of drugs are all too common causes of acute kidney injury. This review will discuss these preventable causes of acute kidney injury. RECENT FINDINGS Recent studies have examined the pathophysiology of acute kidney injury by several commonly used drugs. These studies have shown that drugs and toxins can cause acute kidney injury by altering renal hemodynamics, direct tubular injury or causing renal tubular obstruction. SUMMARY Knowledge of the drugs that cause acute kidney injury and how this injury is manifested can lead to improved diagnosis and treatment with the ultimate goal of prevention.
Collapse
|
28
|
Gauzit R. Actualités en antibiothérapie — Aminosides toujours et encore: bon usage et suivi thérapeutique. ACTA ACUST UNITED AC 2010. [DOI: 10.1007/s13546-010-0106-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
29
|
Lopez-Novoa JM, Quiros Y, Vicente L, Morales AI, Lopez-Hernandez FJ. New insights into the mechanism of aminoglycoside nephrotoxicity: an integrative point of view. Kidney Int 2010; 79:33-45. [PMID: 20861826 DOI: 10.1038/ki.2010.337] [Citation(s) in RCA: 414] [Impact Index Per Article: 29.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Nephrotoxicity is one of the most important side effects and therapeutical limitations of aminoglycoside antibiotics, especially gentamicin. Despite rigorous patient monitoring, nephrotoxicity appears in 10-25% of therapeutic courses. Traditionally, aminoglycoside nephrotoxicity has been considered to result mainly from tubular damage. Both lethal and sub-lethal alterations in tubular cells handicap reabsorption and, in severe cases, may lead to a significant tubular obstruction. However, a reduced glomerular filtration is necessary to explain the symptoms of the disease. Reduced filtration is not solely the result of tubular obstruction and tubular malfunction, resulting in tubuloglomerular feedback activation; renal vasoconstriction and mesangial contraction are also crucial to fully explain aminoglycoside nephrotoxicity. This review critically presents an integrative view on the interactions of tubular, glomerular, and vascular effects of gentamicin, in the context of the most recent information available. Moreover, it discusses therapeutic perspectives for prevention of aminoglycoside nephrotoxicity derived from the pathophysiological knowledge.
Collapse
Affiliation(s)
- Jose M Lopez-Novoa
- Unidad de Fisiopatología Renal y Cardiovascular, Departamento de Fisiología y Farmacología, Universidad de Salamanca, Salamanca, Spain.
| | | | | | | | | |
Collapse
|
30
|
Quiros Y, Vicente-Vicente L, Morales AI, Lopez-Novoa JM, Lopez-Hernandez FJ. An Integrative Overview on the Mechanisms Underlying the Renal Tubular Cytotoxicity of Gentamicin. Toxicol Sci 2010; 119:245-56. [DOI: 10.1093/toxsci/kfq267] [Citation(s) in RCA: 162] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
|
31
|
Tengstrand EA, Miwa GT, Hsieh FY. Bis(monoacylglycerol)phosphate as a non-invasive biomarker to monitor the onset and time-course of phospholipidosis with drug-induced toxicities. Expert Opin Drug Metab Toxicol 2010; 6:555-70. [DOI: 10.1517/17425251003601961] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
|
32
|
Hanumegowda UM, Wenke G, Regueiro-Ren A, Yordanova R, Corradi JP, Adams SP. Phospholipidosis as a Function of Basicity, Lipophilicity, and Volume of Distribution of Compounds. Chem Res Toxicol 2010; 23:749-55. [DOI: 10.1021/tx9003825] [Citation(s) in RCA: 80] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Umesh M. Hanumegowda
- Departments of Discovery Toxicology, Discovery Analytical Sciences, Discovery Chemistry, and Bioinformatics, Bristol-Myers Squibb Research and Development, 5 Research Parkway, Wallingford, Connecticut 06492
| | - Gottfried Wenke
- Departments of Discovery Toxicology, Discovery Analytical Sciences, Discovery Chemistry, and Bioinformatics, Bristol-Myers Squibb Research and Development, 5 Research Parkway, Wallingford, Connecticut 06492
| | - Alicia Regueiro-Ren
- Departments of Discovery Toxicology, Discovery Analytical Sciences, Discovery Chemistry, and Bioinformatics, Bristol-Myers Squibb Research and Development, 5 Research Parkway, Wallingford, Connecticut 06492
| | - Roumyana Yordanova
- Departments of Discovery Toxicology, Discovery Analytical Sciences, Discovery Chemistry, and Bioinformatics, Bristol-Myers Squibb Research and Development, 5 Research Parkway, Wallingford, Connecticut 06492
| | - John P. Corradi
- Departments of Discovery Toxicology, Discovery Analytical Sciences, Discovery Chemistry, and Bioinformatics, Bristol-Myers Squibb Research and Development, 5 Research Parkway, Wallingford, Connecticut 06492
| | - Stephen P. Adams
- Departments of Discovery Toxicology, Discovery Analytical Sciences, Discovery Chemistry, and Bioinformatics, Bristol-Myers Squibb Research and Development, 5 Research Parkway, Wallingford, Connecticut 06492
| |
Collapse
|
33
|
Oszlánczi Á, Bóta A, Klumpp E. Influence of aminoglycoside antibiotics on the thermal behaviour and structural features of DPPE–DPPG model membranes. Colloids Surf B Biointerfaces 2010; 75:141-8. [DOI: 10.1016/j.colsurfb.2009.08.027] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2009] [Accepted: 08/16/2009] [Indexed: 11/16/2022]
|
34
|
Mueller EW, Boucher BA. The Use of Extended-Interval Aminoglycoside Dosing Strategies for the Treatment of Moderate-to-Severe Infections Encountered in Critically Ill Surgical Patients. Surg Infect (Larchmt) 2009; 10:563-70. [DOI: 10.1089/sur.2007.080] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- Eric W. Mueller
- Department of Pharmacy Services, The University Hospital, Cincinnati, Ohio
| | - Bradley A. Boucher
- Department of Clinical Pharmacy, The University of Tennessee Health Science Center, Memphis, Tennessee
| |
Collapse
|
35
|
Sweileh WM. A prospective comparative study of gentamicin- and amikacin-induced nephrotoxicity in patients with normal baseline renal function. Fundam Clin Pharmacol 2009; 23:515-20. [DOI: 10.1111/j.1472-8206.2009.00702.x] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
36
|
Ziai SA, Salehian P, Mahmoudian M. Study of Serum and Tissues Angiotensin Converting Enzyme (ACE) Activity in Rat with Gentamicin Induced Renal Toxicity. Ren Fail 2009; 25:923-33. [PMID: 14669851 DOI: 10.1081/jdi-120026027] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
PURPOSE In this research ACE activity (as a marker of epithelial injury) was studied in rats with gentamicin induced renal toxicity. METHODS Male Sprague-Dawley rats were sacrificed 1, 3, 5, and 7 days after gentamicin injection, 100 mg/kg/day for 1, 3, 5, and 7 consecutive days. ACE activity was measured in serum, kidney and lung. These data were compared with normal saline-treated rats. Histological scoring of renal cortical pathology was performed on days 1, 3, 5, and 7. RESULTS Treatment of rats with gentamicin resulted in renal damage evidenced by proteinuria, polyuria, and decreased creatinine clearance. The damage to the kidney proximal tubule was evident by (a) the histological analysis at light microscopy and (b) the augmentation in the urinary excretion of N-acetyl-beta-D-glucosaminidase (NAG). Kidney ACE activity decreased while lung and serum ACE activity didn't change until day 7. Lung ACE activity increased significantly on day 7. Kidney and serum ACE activity increased too. Blood pressure increased significantly on day 7. This corresponded well with the lung ACE activity increment. CONCLUSION These data suggest that kidney ACE activity decreased significantly just one day after gentamicin administration and prior to kidney NAG decrease.
Collapse
Affiliation(s)
- Seyed Ali Ziai
- Department of Pharmacology, Institute of Medicinal Plants ACECR, Tehran, Iran
| | | | | |
Collapse
|
37
|
Bervoets AR, Behets GJ, Schryvers D, Roels F, Yang Z, Verberckmoes SC, Damment SJ, Dauwe S, Mubiana VK, Blust R, De Broe ME, D'Haese PC. Hepatocellular transport and gastrointestinal absorption of lanthanum in chronic renal failure. Kidney Int 2009; 75:389-98. [DOI: 10.1038/ki.2008.571] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
38
|
Structural and calorimetrical studies of the effect of different aminoglycosides on DPPC liposomes. Colloids Surf B Biointerfaces 2009; 69:116-21. [DOI: 10.1016/j.colsurfb.2008.11.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2008] [Revised: 11/17/2008] [Accepted: 11/18/2008] [Indexed: 11/22/2022]
|
39
|
Nioi P, Pardo IDR, Snyder RD. Monitoring the accumulation of fluorescently labeled phospholipids in cell cultures provides an accurate screen for drugs that induce phospholipidosis. Drug Chem Toxicol 2009; 31:515-28. [PMID: 18850360 DOI: 10.1080/01480540802391229] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
A large number of cationic amphiphilic drugs (CADs) are known to cause phospholipidosis (PLD) in vivo. In the present study, we have built upon our previous findings to further qualify the use of a fluorescently labeled phospholipid-based cell-culture assay to detect PLD-inducing drugs. In this paper, we demonstrate that 12 PLD-negative compounds and 11 drugs known to cause PLD in vivo are all correctly identified by using this assay. Interestingly, we found that in cells treated with certain CADs, the fluorescent phospholipid was sequestered in a very specific punctate pattern, which overlapped strongly with the staining pattern seen with a lysosomal marker protein. Our data also show that false positives can be generated with the fluorescence assay when compounds are used at concentrations that cause a >30% decrease in cell number in this assay. Confocal microscopy demonstrated that the staining pattern of fluorescent phospholipids in these cases may be differentiated from those of true positives by the fact that diffuse, rather than punctuate, fluorescence is observed. These studies confirm and expand our previous results showing that the fluorescent phospholipid assay is a highly sensitive, specific tool for detecting PLD-inducing drugs, if care is taken to rule out cytotoxicity-related artifact.
Collapse
Affiliation(s)
- Paul Nioi
- The Schering-Plough Research Institute, Summit, New Jersey 07901, USA.
| | | | | |
Collapse
|
40
|
Du M, Keeling KM, Fan L, Liu X, Bedwell DM. Poly-L-aspartic acid enhances and prolongs gentamicin-mediated suppression of the CFTR-G542X mutation in a cystic fibrosis mouse model. J Biol Chem 2009; 284:6885-92. [PMID: 19136563 DOI: 10.1074/jbc.m806728200] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Aminoglycosides such as gentamicin have the ability to suppress translation termination at premature stop mutations, leading to a partial restoration of protein expression and function. This observation led to studies showing that this approach may provide a viable treatment for patients with genetic diseases such as cystic fibrosis that are caused by premature stop mutations. Although aminoglycoside treatment is sometimes associated with harmful side effects, several studies have shown that the co-administration of polyanions such as poly-L-aspartic acid (PAA) can both reduce toxicity and increase the intracellular aminoglycoside concentration. In the current study we examined how the co-administration of gentamicin with PAA influenced the readthrough of premature stop codons in cultured cells and a cystic fibrosis mouse model. Using a dual luciferase readthrough reporter system in cultured cells, we found that the co-administration of gentamicin with PAA increased readthrough 20-40% relative to cells treated with the same concentration of gentamicin alone. Using a Cftr-/- hCFTR-G542X mouse model, we found that PAA also increased the in vivo nonsense suppression induced by gentamicin. Following the withdrawal of gentamicin, PAA significantly prolonged the time interval during which readthrough could be detected, as shown by short circuit current measurements and immunofluorescence. Because the use of gentamicin to suppress disease-causing nonsense mutations will require their long term administration, the ability of PAA to reduce toxicity and increase both the level and duration of readthrough has important implications for this promising therapeutic approach.
Collapse
Affiliation(s)
- Ming Du
- Department of Microbiology, Gregory Fleming James Cystic Fibrosis Research Center, University of Alabama at Birmingham, Birmingham, Alabama 35294-2170, USA
| | | | | | | | | |
Collapse
|
41
|
|
42
|
Schurdak ME, Vernetti LA, Abel SJ, Thiffault C. Adaptation of an In Vitro Phospholipidosis Assay to an Automated Image Analysis System. Toxicol Mech Methods 2008; 17:77-86. [DOI: 10.1080/15376510600860185] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
43
|
Guimond A, Viau E, Aubé P, Renzi PM, Paquet L, Ferrari N. Advantageous toxicity profile of inhaled antisense oligonucleotides following chronic dosing in non-human primates. Pulm Pharmacol Ther 2008; 21:845-54. [PMID: 18761414 DOI: 10.1016/j.pupt.2008.08.001] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2008] [Accepted: 08/09/2008] [Indexed: 10/21/2022]
Abstract
TPI ASM8 and TPI 1100 are two products containing modified phosphorothioate antisense oligonucleotides (AONs), which are undergoing development for the treatment of asthma and chronic obstructive pulmonary disease (COPD), respectively. TPI ASM8 is comprised of two AONs, one targeting the human chemokine receptor 3 (CCR3) and the other targeting the common beta-chain of the IL-3/IL-5/GM-CSF receptors. TPI 1100 is also a dual-AON compound targeting the phosphodiesterase (PDE) 4 and 7 isotypes. For both products, the AONs are present in a 1:1 ratio by weight. Both products will be administered by inhalation to patients, and TPI ASM8 is currently undergoing Phase 2 clinical trials. As part of the safety assessment of both products, the toxicity and disposition (i.e., pharmacokinetics of the AON components in plasma and tissues) were investigated in 14-day inhalation studies in monkeys at doses ranging from 0.05 to 2.5mg/kg/day. Results indicated that both products were safe and well tolerated at all dose levels. Reversible treatment-related alterations were only observed at the high dose levels tested and were limited to changes in the respiratory tract which were characterized primarily by the presence of alveolar macrophages in the absence of a generalized inflammatory response. Plasma pharmacokinetic profiles showed very low plasma concentrations, and no plasma accumulation was observed after repeated doses. While significant amounts of the AONs of both TPI ASM8 and TPI 1100 were measured in trachea and lung, only limited amounts of the AONs could be measured in kidney and liver, which, in combination with the low plasma level data, is indicative of very low systemic exposure. Taken together, these results demonstrate that these two new AON-based products are safe and that delivery via the inhaled route achieves localized deposition in the pulmonary tract with very limited systemic exposure and reduced toxicity compared to other routes of AON administration.
Collapse
Affiliation(s)
- Alain Guimond
- Topigen Pharmaceuticals Inc., 2901 East Rachel Street, Suite 13, Montreal, Quebec, Canada H1W 4A4
| | | | | | | | | | | |
Collapse
|
44
|
Pelletier DJ, Gehlhaar D, Tilloy-Ellul A, Johnson TO, Greene N. Evaluation of a Published in Silico Model and Construction of a Novel Bayesian Model for Predicting Phospholipidosis Inducing Potential. J Chem Inf Model 2007; 47:1196-205. [PMID: 17428028 DOI: 10.1021/ci6004542] [Citation(s) in RCA: 78] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The identification of phospholipidosis (PPL) during preclinical testing in animals is a recognized problem in the pharmaceutical industry. Depending on the intended indication and dosing regimen, PPL can delay or stop development of a compound in the drug discovery process. Therefore, for programs and projects where a PPL finding would have adverse impact on the success of the project, it would be desirable to be able to rapidly identify and screen out those compounds with the potential to induce PPL as early as possible. Currently, electron microscopy is the gold standard method for identifying phospholipidosis, but it is low-throughput and resource-demanding. Therefore, a low-cost, high-throughput screening strategy is required to overcome these limitations and be applicable in the drug discovery cycle. A recent publication by Ploemen et al. (Exp. Toxicol. Pathol. 2004, 55, 347-55) describes a method using the computed physicochemical properties pKa and ClogP as part of a simple calculation to determine a compound's potential to induce PPL. We have evaluated this method using a set of 201 compounds, both public and proprietary, with known in vivo PPL-inducing ability and have found the overall concordance to be 75%. We have proposed simple modifications to the model rules, which improve the model's concordance to 80%. Finally, we describe the development of a Bayesian model using the same compound set and found its overall concordance to be 83%.
Collapse
Affiliation(s)
- Dennis J Pelletier
- Toxicoinformatics Group, Pfizer Global Research, Groton, Connecticut 06340, USA.
| | | | | | | | | |
Collapse
|
45
|
Reasor MJ, Hastings KL, Ulrich RG. Drug-induced phospholipidosis: issues and future directions. Expert Opin Drug Saf 2006; 5:567-83. [PMID: 16774494 DOI: 10.1517/14740338.5.4.567] [Citation(s) in RCA: 168] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Numerous drugs containing a cationic amphiphilic structure are capable of inducing phospholipidosis in cells under conditions of in vivo administration or ex vivo incubation. The principal characteristics of this condition include the reversible accumulation of polar phospholipids in association with the development of unicentric or multicentric lamellated bodies within cells. There is an abundance of data providing an understanding of potential mechanisms for the induction of phospholipidosis; however, the process is likely to be complex and may differ from one drug to another. The functional consequences of the presence of this condition on cellular or tissue function are not well understood. The general consensus is that the condition is an adaptive response rather than a toxicological manifestation; however, additional studies to examine this question are needed. Until this issue is resolved, concerns about phospholipidosis will continue to exist at regulatory agencies. Procedures for the screening of potential phospholipogenic candidate compounds are available. In contrast, a clear need exists for the identification of valid biomarkers to assess the development of phospholipidosis in preclinical and clinical studies.
Collapse
Affiliation(s)
- Mark J Reasor
- Robert C Byrd Health Sciences Center of West Virginia University, Department of Physiology and Pharmacology, P.O. Box 9229, Morgantown, WV 26506, USA.
| | | | | |
Collapse
|
46
|
Servais H, Jossin Y, Van Bambeke F, Tulkens PM, Mingeot-Leclercq MP. Gentamicin causes apoptosis at low concentrations in renal LLC-PK1 cells subjected to electroporation. Antimicrob Agents Chemother 2006; 50:1213-21. [PMID: 16569831 PMCID: PMC1426926 DOI: 10.1128/aac.50.4.1213-1221.2006] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Gentamicin accumulates in the lysosomes of kidney proximal tubular cells and causes apoptosis at clinically relevant doses. Gentamicin-induced apoptosis can be reproduced with cultured renal cells, but only at high extracellular concentrations (1 to 3 mM; 0.4 to 1.2 g/liter) because of its low level of uptake. We recently showed that gentamicin-induced apoptosis in LLC-PK1 cells involves a rapid (2-h) permeabilization of lysosomes and activation of the mitochondrial pathway of apoptosis (10 h). We now examine whether the delivery of gentamicin to the cytosol by electroporation would sensitize LLC-PK1 cells to apoptosis. Cells were subjected to eight pulses (1 ms) at 800 V/cm (square waves) in the presence of gentamicin (3 microM to 3 mM; 1.2 mg/liter to 1.2 g/liter); returned to gentamicin-free medium; and examined at 8 h for their Bax (a marker of mitochondrial pathway activation) contents by Western blotting and competitive reverse transcriptase PCR and at 24 h for apoptosis by 4',6'-diamidino-2'-phenylindole staining (confirmed by electron microscopy) and for necrosis (by determination of lactate dehydrogenase release). Nonelectroporated cells were incubated with gentamicin for 8 and 24 h. Significant increases in Bax levels (8 h) and apoptosis (24 h) were detected with 0.03 mM (13.2 mg/liter) gentamicin in electroporated cells compared with those achieved with 2 mM (928 mg/liter) in incubated cells. The increase in the Bax level was not associated with an increase in the level of its mRNA but was associated with the accumulation of ubiquitinated forms (probably as a result of impairment of its degradation by the proteasome). Assay of cell-associated gentamicin showed a marked, immediate, but transient accumulation in electroporated cells, whereas a slow, steady uptake was detected in incubated cells. The data indicate that cytosolic gentamicin triggers apoptosis. Sequestration of gentamicin in lysosomes would, to some extent, protect against apoptosis.
Collapse
Affiliation(s)
- Hélène Servais
- Unité de Pharmacologie Cellulaire et Moléculaire, Université Catholique de Louvain, 7370 avenue E. Mounier 73, B-1200 Brussels, Belgium
| | | | | | | | | |
Collapse
|
47
|
Du M, Keeling KM, Fan L, Liu X, Kovaçs T, Sorscher E, Bedwell DM. Clinical doses of amikacin provide more effective suppression of the human CFTR-G542X stop mutation than gentamicin in a transgenic CF mouse model. J Mol Med (Berl) 2006; 84:573-82. [PMID: 16541275 DOI: 10.1007/s00109-006-0045-5] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2005] [Accepted: 01/09/2006] [Indexed: 10/24/2022]
Abstract
Mutations in the cystic fibrosis transmembrane conductance regulator (CFTR) gene cause the disease cystic fibrosis. We previously reported that gentamicin administration suppressed a CFTR premature stop mutation in a Cftr-/- mouse model carrying a human CFTR-G542X (hCFTR-G542X) transgene, resulting in the appearance of hCFTR protein and function. However, the high doses used in that study resulted in peak serum levels well beyond the levels typically administered to humans. To address this problem, we identified doses of both gentamicin and amikacin that resulted in peak serum levels within their accepted clinical ranges. We then asked whether these doses could suppress the hCFTR-G542X mutation in the Cftr-/- hCFTR-G542X mouse model. Our results indicate that low doses of each compound restored some hCFTR protein expression and function, as shown by immunofluorescence and short-circuit current measurements. However, we found that amikacin suppressed the hCFTR-G542X premature stop mutation more effectively than gentamicin when administered at these clinically relevant doses. Because amikacin is also less toxic than gentamicin, it may represent a superior choice for suppression therapy in patients that carry a premature stop mutation in the CFTR gene.
Collapse
Affiliation(s)
- Ming Du
- Department of Microbiology, The University of Alabama at Birmingham, 35294-2170, USA
| | | | | | | | | | | | | |
Collapse
|
48
|
Hirukawa S, Olson KA, Tsuji T, Hu GF. Neamine inhibits xenografic human tumor growth and angiogenesis in athymic mice. Clin Cancer Res 2006; 11:8745-52. [PMID: 16361562 DOI: 10.1158/1078-0432.ccr-05-1495] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE We have previously shown that the aminoglycoside antibiotic neomycin blocks the nuclear translocation of angiogenin and inhibits its angiogenic activity. However, neomycin has not been considered as a favorable drug candidate for clinical development because of its known nephrotoxicity and ototoxicity. The aim of this study is to determine whether neamine, a nontoxic derivative of neomycin, possesses antitumor activity. EXPERIMENTAL DESIGN The effect of neamine on the nuclear translocation of angiogenin was examined by means of immunofluorescence and Western blotting. The antitumor activity of neamine was determined with three different animal models. RESULTS Neamine effectively blocked the nuclear translocation of angiogenin in endothelial cells and inhibited angiogenin-induced cell proliferation. It inhibited the establishment of human tumor xenografts in athymic mice in both ectopic and orthotopic tumor models. It also inhibited the progression of established human tumor transplants, whereas the structurally related antibiotic paromomycin had no effect. Immunohistochemical staining showed that both angiogenesis and cancer cell proliferation are inhibited by neamine. CONCLUSION These results suggest that the nontoxic aminoglycoside antibiotic neamine is an effective inhibitor of nuclear translocation of angiogenin and may serve as an inhibitor for angiogenin-induced angiogenesis and cancer progression.
Collapse
Affiliation(s)
- Saori Hirukawa
- Department of Pathology, Harvard Medical School, Boston, Massachusetts 02115, USA
| | | | | | | |
Collapse
|
49
|
Cittanova ML. [Which are the factors of toxic and pharmacological risk implicated in occurrence of acute renal failure during the perioperative period?]. ANNALES FRANCAISES D'ANESTHESIE ET DE REANIMATION 2005; 24:167-76. [PMID: 15737503 DOI: 10.1016/j.annfar.2004.12.024] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/01/2023]
Affiliation(s)
- M-L Cittanova
- GARHPA, hôpital privé d'Antony, 1, rue Velpeau, 92160 Antony, France.
| |
Collapse
|
50
|
Rougier F, Ducher M, Maurin M, Corvaisier S, Claude D, Jelliffe R, Maire P. Aminoglycoside dosages and nephrotoxicity: quantitative relationships. Clin Pharmacokinet 2003; 42:493-500. [PMID: 12739987 DOI: 10.2165/00003088-200342050-00007] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
OBJECTIVE To develop a model that relates the probability of occurrence of nephrotoxicity to the cumulative area under the curve (AUC) of amikacin serum concentration. DESIGN AND PATIENTS This was a retrospective study of two groups of patients in whom nephrotoxicity was observed after administration of amikacin. The first group consisted of patients treated with once-daily administration (ODA) [n = 13]. The second group consisted of patients treated with twice-daily administration (TDA) [n = 22]. MAIN OUTCOME MEASURES The probability of nephrotoxicity occurrence. RESULTS The model is a powerful tool to represent and describe the influence of the dosage regimen on aminoglycoside nephrotoxicity. The onset of nephrotoxicity is delayed in the ODA group (p = 0.01) for the same total daily dose among the two groups. The cumulative serum AUC values at onset of nephrotoxicity were greater for the ODA group (p = 0.029). In addition, for the same probability of nephrotoxicity occurrence (50%), the cumulative AUC for the ODA dosage regimen is 2 613 mg. h/L versus only 1 521 mg. h/L for the TDA dosage regimen. The difference in nephrotoxicity between ODA and TDA is greatest for a cumulative AUC of 2 495 mg. h/L, which corresponds to standard therapy with amikacin 900 mg/day during a 7-day period, i.e. 15 mg/kg/day for a 60kg patient with normal renal function (initial creatinine clearance >80 mL/min). For an AUC above 2 495 mg. h/L, the difference in nephrotoxicity decreases slowly to zero. This result means that ODA is especially justified when the treatment is administered over a short duration, i.e. less than 7 days. CONCLUSIONS The utility of selecting ODA in order to obtain less nephrotoxicity in comparison with TDA is therefore not established when the treatment is prolonged. In clinical use, the choice of the dosage regimen is not clear-cut, and both expected efficacy and expected toxicity must be taken into account in order to obtain an overall optimisation of each patient's therapy.
Collapse
Affiliation(s)
- Florent Rougier
- UMR CNRS 5558-ADCAPT Service Pharmaceutique, Hôpital Antoine Charial, Francheville, France
| | | | | | | | | | | | | |
Collapse
|