1
|
Yahia R, Hassan GG, Abo-Youssef AM, Mahmoud HM. Piribedil and thymol mitigate vancomycin-evoked nephrotoxicity in rats through modulation of Keap-1/Nrf2/HO-1 and NF-κB/Bax/caspase 3 signalings. Drug Chem Toxicol 2025:1-16. [PMID: 40143539 DOI: 10.1080/01480545.2025.2481857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 02/18/2025] [Accepted: 03/15/2025] [Indexed: 03/28/2025]
Abstract
Nephrotoxicity is a sign in which endogenous or exogenous toxicants have damaged the kidney-specific detoxification and excretion processes. Vancomycin (VAN) exposure mostly causes kidney damage and a loss of body homeostasis regulation. This study aimed to investigate the protective effects of piribedil and thymol and its basic mechanisms against nephrotoxicity caused by VAN. Randomly, the animals were categorized into six groups (n = 8). For 7 d, Group I only received vehicles, Group II received piribedil (5 mg/kg/once daily, i.p.), Group III received thymol (25 mg/kg/once daily, i.p), Group IV was administered a single daily dose of VAN (200 mg/kg, i.p.), VAN+ piribedil was administered to Group V, and VAN + thymol was administered to Group VI. The findings showed that piribedil or thymol improved renal function parameters by an increase in serum albumin level in parallel to a decrease in serum creatinine and blood urea nitrogen (BUN) levels in addition to decreased levels of KIM-1 and serum cystatin C. Furthermore, enhanced oxidative stress biomarkers as GSH, myeloperoxidase (MPO), and malondialdehyde (MDA) as well as tumor necrosis factor-alpha (TNF-α) and interleukin-1beta (IL-1β), indicators of inflammatory mediators, were markedly reduced compared to VAN group. Moreover, piribedil or thymol markedly improved the histopathological aberrations provoked by VAN, increased the Nrf-2 and HO-1 renal protein expressions and reduced VAN-induced elevation of Keap-1 protein expression. In addition, NF-kB, Bax, and caspase 3 expression levels were considerably declined after piribedil or thymol co-treatment. These findings revealed that co-administration of piribedil or thymol with VAN may be a sensible therapeutic approach for reducing renal intoxication caused by VAN.
Collapse
Affiliation(s)
- Rania Yahia
- Department of Pharmacology, Egyptian Drug Authority, Cairo, Egypt
| | - Gehad Gamal Hassan
- Central Administration of Pharmaceutical Products, Egyptian Drug Authority, Cairo, Egypt
| | - Amira M Abo-Youssef
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Beni-Suef University, Beni-Suef, Egypt
| | - Heba M Mahmoud
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Beni-Suef University, Beni-Suef, Egypt
| |
Collapse
|
2
|
Alhumaidi R, Huang H, Saade MC, Clark AJ, Parikh SM. NAD + metabolism in acute kidney injury and chronic kidney disease transition. Trends Mol Med 2025:S1471-4914(24)00337-X. [PMID: 39757045 DOI: 10.1016/j.molmed.2024.12.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 11/21/2024] [Accepted: 12/09/2024] [Indexed: 01/07/2025]
Abstract
Disturbances in kidney tubular cell metabolism are increasingly recognized as a feature of acute kidney injury (AKI). In AKI, tubular epithelial cells undergo abnormal metabolic shifts that notably disrupt NAD+ metabolism. Recent advancements have highlighted the critical role of NAD+ metabolism in AKI, revealing that acute disruptions may lead to lasting cellular changes, thereby promoting the transition to chronic kidney disease (CKD). This review explores the molecular mechanisms underlying metabolic dysfunction in AKI, with a focus on NAD+ metabolism, and proposes several cellular processes through which acute aberrations in NAD+ may contribute to long-term changes in the kidney.
Collapse
Affiliation(s)
- Rahil Alhumaidi
- Division of Nephrology, Department of Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Huihui Huang
- Division of Nephrology, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Marie Christelle Saade
- Division of Nephrology, Department of Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Amanda J Clark
- Division of Nephrology, Department of Pediatrics, University of Texas Southwestern and Children's Medical Center, Dallas, TX, USA
| | - Samir M Parikh
- Division of Nephrology, Department of Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA; Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
3
|
El-Shoura EAM, Sharkawi SMZ, Abdelzaher LA, Abdel-Wahab BA, Ahmed YH, Abdel-Sattar AR. Reno-protective effect of fenofibrate and febuxostat against vancomycin-induced acute renal injury in rats: Targeting PPARγ/NF-κB/COX-II and AMPK/Nrf2/HO-1 signaling pathways. Immunopharmacol Immunotoxicol 2024; 46:509-520. [PMID: 38918173 DOI: 10.1080/08923973.2024.2373216] [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: 12/21/2023] [Accepted: 06/22/2024] [Indexed: 06/27/2024]
Abstract
BACKGROUND Vancomycin (VCM) is used clinically to treat serious infections caused by multi-resistant Gram-positive bacteria, although its use is severely constrained by nephrotoxicity. This study investigated the possible nephroprotective effect of febuxostat (FX) and/or fenofibrate (FENO) and their possible underlying mechanisms against VCM-induced nephrotoxicity in a rat model. METHODS Male Wistar rats were randomly allocated into five groups; Control, VCM, FX, FENO, and combination groups. Nephrotoxicity was evaluated histopathologically and biochemically. The oxidative stress biomarkers (SOD, MDA, GSH, total nitrite, GPx, MPO), the apoptotic marker, renal Bcl-2 associated X protein (Bax), and inflammatory and kidney injury markers (IL-1β, IL-6, TNF-α, Nrf2, OH-1, kappa-light-chain-enhancer of activated B cells (NF-κB), NADPH oxidase, Kim-1, COX-II, NGAL, Cys-C were also evaluated. RESULTS VCM resulted in significant elevation in markers of kidney damage, oxidative stress, apoptosis, and inflammatory markers. Co-administration of VCM with either/or FX and FENO significantly mitigated nephrotoxicity and associated oxidative stress, inflammatory and apoptotic markers. In comparison to either treatment alone, a more notable improvement was observed with the FX and FENO combination regimen. CONCLUSION Our findings show that FX, FENO, and their combination regimen have a nephroprotective impact on VCM-induced kidney injury by suppressing oxidative stress, apoptosis, and the inflammatory response. Renal recovery from VCM-induced injury was accomplished by activation of Nrf2/HO-1 signaling and inhibition of NF-κB expression. This study highlights the importance of FX and FENO as effective therapies for reducing nephrotoxicity in VCM-treated patients.
Collapse
Affiliation(s)
- Ehab A M El-Shoura
- Department of Clinical Pharmacy, Faculty of Pharmacy, Al-Azhar University, Assiut, Egypt
- Department of Pharmacy Practice, Faculty of Pharmacy, Horus University in Egypt, New Damietta, Egypt
| | - Souty M Z Sharkawi
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Beni-Suef University, Beni-Suef, Egypt
| | - Lobna A Abdelzaher
- Department of Pharmacology, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Basel A Abdel-Wahab
- Department of Pharmacology, College of Pharmacy, Najran University, Najran, Saudi Arabia
| | - Yasmine H Ahmed
- Department of Cytology and Histology, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt
| | | |
Collapse
|
4
|
Das K, Muthukumar A, Almuqbil M, Imran M, Rabaan AA, Halwani MA, Garout M, Alsaleh AA, Alissa M, Alwashmi ASS, Alshehri AA, Alsayyah A, Bhavani K, Mittal S, Gayathri R, Alomar NF, Rabbani SI, Basheeruddin Asdaq SM. Nephroprotective potential of Polyalthia longifolia roots against vancomycin-induced renal toxicity in experimental animals. Front Pharmacol 2023; 14:1107435. [PMID: 36755952 PMCID: PMC9900101 DOI: 10.3389/fphar.2023.1107435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Accepted: 01/09/2023] [Indexed: 01/24/2023] Open
Abstract
This study was done to investigate the possible nephroprotective effect of an ethanolic root extract of Polyalthia Longifolia (PL) on vancomycin-induced nephrotoxicity using curative and protective models. Vancomycin (150 mg/kg, intravenous) was given to healthy Wistar albino rats in the curative model before the start of treatment, whereas the protective group received vancomycin at the conclusion of the 10-day treatment procedure. Animals were divided into six groups for both models; group I served as the normal control, while groups II, III, IV, V, and VI were kept as toxic control, standard (selenium, 6 mg/kg), LDPL (low dose of PL 200 mg/kg), HDPL (high dose of PL 400 mg/kg), and HDPL + selenium (interactive) groups, respectively. Renal biomarkers [(uric acid, creatinine, blood urea nitrogen (BUN), serum proteins], and blood electrolyte levels were measured for all tested groups. When compared to the vancomycin group, the HDPL significantly (p < 0.01) showed greater effectiveness in lowering the BUN, potassium, and calcium levels. Additionally, in the curative model, there was a significant (p < 0.05) decrease in the blood levels of uric acid, creatinine, BUN, potassium, and calcium in the animals who received the combination of selenium and HDPL. Both LDPL and HDPL did not provide any distinguishable effect in the protective model, but groups that received HDPL with selenium did provide detectable protection by significantly lowering their levels of uric acid, BUN, serum potassium, and total serum protein in comparison to the vancomycin control group. These findings indicate that, whether administered before or after renal damage is induced, the Polyalthia longifolia root extract provided only modest protection to nephrons, which require selenium support to prevent vancomycin-induced kidney damage.
Collapse
Affiliation(s)
- Kuntal Das
- Nitte College of Pharmaceutical Science, Yelahanka, Bangalore, India
| | - A. Muthukumar
- Central Animal House, Department of Pharmacology, Al-Ameen College of Pharmacy, Bangalore, India,*Correspondence: Syed Mohammed Basheeruddin Asdaq, /; A. Muthukumar,
| | - Mansour Almuqbil
- Department of Clinical Pharmacy, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Mohd. Imran
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Northern Border University, Rafha, Saudi Arabia
| | - Ali A. Rabaan
- Molecular Diagnostic Laboratory, Johns Hopkins Aramco Healthcare, Dhahran, Saudi Arabia,College of Medicine, Alfaisal University, Riyadh, Saudi Arabia,Department of Public Health and Nutrition, The University of Haripur, Haripur, Pakistan
| | - Muhammad A. Halwani
- Department of Medical Microbiology, Faculty of Medicine, Al Baha University, Al Baha, Saudi Arabia
| | - Mohammed Garout
- Department of Community Medicine and Healthcare for Pilgrims, Faculty of Medicine, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Abdulmonem A. Alsaleh
- Clinical Laboratory Science Department, Mohammed Al-Mana College for Medical Sciences, Dammam, Saudi Arabia
| | - Mohammed Alissa
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Prince Sattam bin Abdulaziz University, Al-Kharj, Saudi Arabia
| | - Ameen S. S. Alwashmi
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah, Saudi Arabia
| | - Ahmad A. Alshehri
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Najran University, Najran, Saudi Arabia
| | - Ahmed Alsayyah
- Department of Pathology, College of Medicine, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| | | | - Swati Mittal
- Central Animal House, Department of Pharmacology, Al-Ameen College of Pharmacy, Bangalore, India
| | - R. Gayathri
- Department of Pharmaceutics, KMCH College of Pharmacy, Coimbatore, India
| | | | - Syed Imam Rabbani
- Department of Pharmacology and Toxicology, College of Pharmacy, Qassim University, Buraydah, Saudi Arabia
| | - Syed Mohammed Basheeruddin Asdaq
- Department of Pharmacy Practice, College of Pharmacy, AlMaarefa University, Riyadh, Saudi Arabia,*Correspondence: Syed Mohammed Basheeruddin Asdaq, /; A. Muthukumar,
| |
Collapse
|
5
|
Laou E, Mavridis T, Papagiannakis N, Pais G, Chighine A, Chang J, Locci E, D’Aloja E, Scheetz M, Chalkias A, Xanthos T. Blood Biomarkers and Metabolomic Profiling for the Early Diagnosis of Vancomycin-Associated Acute Kidney Injury: A Systematic Review and Meta-Analysis of Experimental Studies. J Pers Med 2022; 12:1397. [PMID: 36143182 PMCID: PMC9505572 DOI: 10.3390/jpm12091397] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Revised: 08/25/2022] [Accepted: 08/26/2022] [Indexed: 11/22/2022] Open
Abstract
BACKGROUND several blood-based biomarkers have been proposed for predicting vancomycin-associated kidney injury (VIKI). However, no systematic analysis has compared their prognostic value. OBJECTIVE this systematic review and meta-analysis was designed to investigate the role of blood biomarkers and metabolomic profiling as diagnostic and prognostic predictors in pre-clinical studies of VIKI. METHODS a systematic search of PubMed was conducted for relevant articles from January 2000 to May 2022. Animal studies that administered vancomycin and studied VIKI were eligible for inclusion. Clinical studies, reviews, and non-English literature were excluded. The primary outcome was to investigate the relationship between the extent of VIKI as measured by blood biomarkers and metabolomic profiling. Risk of bias was assessed with the CAMARADES checklist the SYRCLE's risk of bias tool. Standard meta-analysis methods (random-effects models) were used. RESULTS there were four studies for the same species, dosage, duration of vancomycin administration and measurement only for serum creatine and blood urea nitrogen in rats. A statistically significant increase was observed between serum creatinine in the vancomycin group compared to controls (pooled p = 0.037; Standardized Mean Difference: 2.93; 95% CI: 0.17 to 5.69; I2 = 92.11%). Serum BUN levels were not significantly different between control and vancomycin groups (pooled p = 0.11; SMD: 3.05; 95% CI: 0.69 to 6.8; I2 = 94.84%). We did not identify experimental studies using metabolomic analyses in animals with VIKI. CONCLUSIONS a total of four studies in rodents only described outcomes of kidney injury as defined by blood biomarkers. Blood biomarkers represented included serum creatinine and BUN. Novel blood biomarkers have not been explored.
Collapse
Affiliation(s)
- Eleni Laou
- Department of Anesthesiology, Faculty of Medicine, University of Thessaly, 41500 Larisa, Greece
| | - Theodoros Mavridis
- First Department of Neurology, Eginition University Hospital, Medical School, National and Kapodistrian University of Athens, 15772 Athens, Greece
| | - Nikolaos Papagiannakis
- First Department of Neurology, Eginition University Hospital, Medical School, National and Kapodistrian University of Athens, 15772 Athens, Greece
| | - Gwendolyn Pais
- Department of Pharmacy Practice, Pharmacometric Center of Excellence, Midwestern University Chicago College of Pharmacy, Downers Grove, IL 60515, USA
| | - Alberto Chighine
- Department of Medical Sciences and Public Health, Section of Legal Medicine, University of Cagliari, 09124 Cagliari, Italy
| | - Jack Chang
- Department of Pharmacy Practice, Pharmacometric Center of Excellence, Midwestern University Chicago College of Pharmacy, Downers Grove, IL 60515, USA
- Northwestern Memorial Hospital, Chicago, IL 60611, USA
| | - Emanuela Locci
- Department of Medical Sciences and Public Health, Section of Legal Medicine, University of Cagliari, 09124 Cagliari, Italy
| | - Ernesto D’Aloja
- Department of Medical Sciences and Public Health, Section of Legal Medicine, University of Cagliari, 09124 Cagliari, Italy
| | - Marc Scheetz
- Department of Pharmacy Practice, Pharmacometric Center of Excellence, Midwestern University Chicago College of Pharmacy, Downers Grove, IL 60515, USA
- Northwestern Memorial Hospital, Chicago, IL 60611, USA
| | - Athanasios Chalkias
- Department of Anesthesiology, Faculty of Medicine, University of Thessaly, 41500 Larisa, Greece
- Outcomes Research Consortium, Cleveland, OH 44195, USA
| | - Theodoros Xanthos
- School of Health sciences, University of West Attica, 12243 Athens, Greece
| |
Collapse
|
6
|
The Mechanism of Drug Nephrotoxicity and the Methods for Preventing Kidney Damage. Int J Mol Sci 2021; 22:ijms22116109. [PMID: 34204029 PMCID: PMC8201165 DOI: 10.3390/ijms22116109] [Citation(s) in RCA: 93] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 06/03/2021] [Accepted: 06/05/2021] [Indexed: 12/11/2022] Open
Abstract
Acute kidney injury (AKI) is a global health challenge of vast proportions, as approx. 13.3% of people worldwide are affected annually. The pathophysiology of AKI is very complex, but its main causes are sepsis, ischemia, and nephrotoxicity. Nephrotoxicity is mainly associated with the use of drugs. Drug-induced AKI accounts for 19-26% of all hospitalized cases. Drug-induced nephrotoxicity develops according to one of the three mechanisms: (1) proximal tubular injury and acute tubular necrosis (ATN) (a dose-dependent mechanism), where the cause is related to apical contact with drugs or their metabolites, the transport of drugs and their metabolites from the apical surface, and the secretion of drugs from the basolateral surface into the tubular lumen; (2) tubular obstruction by crystals or casts containing drugs and their metabolites (a dose-dependent mechanism); (3) interstitial nephritis induced by drugs and their metabolites (a dose-independent mechanism). In this article, the mechanisms of the individual types of injury will be described. Specific groups of drugs will be linked to specific injuries. Additionally, the risk factors for the development of AKI and the methods for preventing and/or treating the condition will be discussed.
Collapse
|
7
|
Blair M, Côté JM, Cotter A, Lynch B, Redahan L, Murray PT. Nephrotoxicity from Vancomycin Combined with Piperacillin-Tazobactam: A Comprehensive Review. Am J Nephrol 2021; 52:85-97. [PMID: 33735856 DOI: 10.1159/000513742] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Accepted: 12/11/2020] [Indexed: 12/13/2022]
Abstract
BACKGROUND Recent studies have identified the combination of vancomycin with piperacillin-tazobactam (VPT) to be associated with increased nephrotoxicity. Multiple, large cohort studies have found this widely used combination to have a higher risk of nephrotoxicity than other regimens in a variety of populations. SUMMARY This review summarizes the epidemiology and clinical features of VPT-associated acute kidney injury (AKI). Potential mechanisms involved in the pathogenesis of this phenomenon are also discussed. Key Message: VPT-associated nephrotoxicity is a recently recognized clinical entity. Clinical strategies to minimize the risk of toxicity in this setting include antimicrobial stewardship, monitoring of kidney function, and emerging data supporting the potential role for novel biomarkers in predicting and managing AKI.
Collapse
Affiliation(s)
- Matthew Blair
- Division of Medicine, Mater Misericordiae University Hospital, Dublin, Ireland
| | - Jean-Maxime Côté
- Service of Nephrology, Department of Medicine, Centre Hospitalier Universitaire de Montréal, Montréal, Québec, Canada
- Clinical Research Centre, University College Dublin, Dublin, Ireland
| | - Aoife Cotter
- Department of Infectious Diseases, Mater Misericordiae University Hospital, Dublin, Ireland
| | - Breda Lynch
- Department of Clinical Microbiology, Mater Misericordiae University Hospital, Dublin, Ireland
| | - Lynn Redahan
- Division of Medicine, Mater Misericordiae University Hospital, Dublin, Ireland
- Department of Renal Medicine, Mater Misericordiae University Hospital, Dublin, Ireland
| | - Patrick T Murray
- Division of Medicine, Mater Misericordiae University Hospital, Dublin, Ireland,
- Clinical Research Centre, University College Dublin, Dublin, Ireland,
- Department of Renal Medicine, Mater Misericordiae University Hospital, Dublin, Ireland,
| |
Collapse
|
8
|
Kidney biopsy findings in vancomycin-induced acute kidney injury: a pooled analysis. Int Urol Nephrol 2021; 54:137-148. [PMID: 33715061 DOI: 10.1007/s11255-021-02831-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Accepted: 02/28/2021] [Indexed: 02/07/2023]
Abstract
BACKGROUND Acute kidney injury represents a major adverse effect of vancomycin administration. The aim of the present study is to accumulate all biopsy-proven cases of vancomycin nephrotoxicity and assess the association of histopathological features with renal prognosis. METHODS Medline, Scopus, CENTRAL, Web of Science, and Clinicaltrials.gov were systematically searched from inception to 29 September 2020. All case reports/series providing individual data of patients with biopsy-proven vancomycin nephrotoxicity were held eligible. A time-to-event analysis was performed evaluating the effects of histological diagnosis on renal recovery. RESULTS Overall, 18 studies were included, comprising 21 patients. Acute tubulointerstitial nephritis was the predominant pattern in 9 patients and was associated with a significantly higher risk of permanent renal dysfunction (HR: 5.08, 95% CI: [1.05-24.50)] compared to acute tubular necrosis. Tubulitis and eosinophilic infiltration were the most common histopathological findings, while interstitial fibrosis was linked to significantly worse renal prognosis (HR: 5.55, 95% CI: 1.13-27.27). Immunofluorescence and electron microscopy features were non-specific. Obstruction by tubular casts composed of vancomycin aggregates and uromodulin has been identified as a new mechanism of nephrotoxicity. CONCLUSIONS Acute tubular necrosis and tubulointerstitial nephritis represent the main histological patterns of vancomycin-induced acute kidney injury. The presence of fibrosis in the context of interstitial inflammation may be linked to lower recovery rates and worse long-term renal outcomes. A novel cast nephropathy obstructive mechanism has been suggested, necessitating further confirmation. Large-scale studies should define the exact indications of kidney biopsy in cases with suspected vancomycin nephrotoxicity.
Collapse
|
9
|
El Bohi KM, Abdel-Motal SM, Khalil SR, Abd-Elaal MM, Metwally MMM, ELhady WM. The efficiency of pomegranate (Punica granatum) peel ethanolic extract in attenuating the vancomycin-triggered liver and kidney tissues injury in rats. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2021; 28:7134-7150. [PMID: 33029776 DOI: 10.1007/s11356-020-10999-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Accepted: 09/25/2020] [Indexed: 06/11/2023]
Abstract
This study evaluated the potential of Punica granatum peel ethanol extract (PPEE) in attenuating the liver and kidney tissue injury induced by vancomycin (VM) treatment in rats. Fifty rats were distributed equally into five groups: control group, PPEE-administered group (100 mg/kg BW/day for 2 weeks; orally), VM-treated group (443.6 mg/kg BW, every alternate day for 2 weeks; intraperitoneally), pre-treated group, and concomitant-treated group. The biochemical response and the histopathology of the hepatic and renal tissue of the treated animals were assessed. The results showed that VM treatment induced substantial hepatotoxicity and nephrotoxicity, evidenced by a significant elevation in tissue injury and lipid oxidative (malondialdehyde) and inflammatory response (C-reactive protein) biomarkers, with lowered antioxidants and protein levels. Additionally, VM treatment induced various morphological, cytotoxic, vascular, and inflammatory perturbations as well as upregulation in the immune-expression of Caspase-3 and downregulation of BCL-2. Moreover, PPEE co-treatment was found to reduce the VM-induced toxicity by protecting the tissue against reactive oxygen species (ROS)-mediated oxidative damage, and inflammation as well as hinder the apoptotic cell death by modulating the expression of apoptosis-related proteins. Thus, we conclude that the PPEE administration showed more restoring efficacy when administered prior to VM medication.
Collapse
Affiliation(s)
- Khlood M El Bohi
- Forensic Medicine and Toxicology Department, Faculty of Veterinary Medicine, Zagazig University, Zagazig, 44511, Egypt
| | - Sabry M Abdel-Motal
- Department of Pharmacology, Faculty of Veterinary Medicine, Zagazig University, Zagazig, Egypt
| | - Samah R Khalil
- Forensic Medicine and Toxicology Department, Faculty of Veterinary Medicine, Zagazig University, Zagazig, 44511, Egypt.
| | - Mayar Mahmoud Abd-Elaal
- Department of Pharmacology, Faculty of Veterinary Medicine, Zagazig University, Zagazig, Egypt
| | - Mohamed M M Metwally
- Pathology Department, Faculty of Veterinary Medicine, Zagazig University, Zagazig, Egypt
| | - Walaa M ELhady
- Forensic Medicine and Toxicology Department, Faculty of Veterinary Medicine, Zagazig University, Zagazig, 44511, Egypt
| |
Collapse
|
10
|
Qu S, Dai C, Hao Z, Tang Q, Wang H, Wang J, Zhao H. Chlorogenic acid prevents vancomycin-induced nephrotoxicity without compromising vancomycin antibacterial properties. Phytother Res 2020; 34:3189-3199. [PMID: 32648634 DOI: 10.1002/ptr.6765] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Revised: 03/21/2020] [Accepted: 05/16/2020] [Indexed: 12/18/2022]
Abstract
Vancomycin (VCM) is an effective chemotherapeutic agent commonly used against gram-positive microorganisms but has serious nephrotoxic side effects that limit its effectiveness. New therapeutics and strategies are urgently needed to combat VCM associated nephrotoxicity. In this study, we determined the protective effect of chlorogenic acid (CA) in a rat model of VCM-induced nephrotoxicity. VCM administration led to markedly elevated blood urea nitrogen and serum creatinine levels that could be prevented with CA co-administration. VCM-mediated oxidative stress was also significantly attenuated by CA as reflected by decreased malondialdehyde and nitric oxide in VCM-treated kidneys. CA administration also prevented the VCM-mediated decrease in the renal antioxidative enzyme activities of glutathione reductase, glutathione peroxidase, and catalase and led to increased levels of reduced glutathione that had been depleted by VCM. Moreover, CA administration clearly inhibited VCM-induced expression of nuclear factor-kappa B, inducible nitric oxide synthase and the downstream pro-inflammatory mediators tumor necrosis factor-α and interleukins 1β and 6. Apoptotic markers were also markedly down-regulated with CA. Overall, CA treatment mitigated VCM-induced oxidative and nitrosative stresses and countered the apoptotic and inflammatory effects of VCM. Notably, CA did not affect the antibacterial activity of VCM in vitro.
Collapse
Affiliation(s)
- Shaoqi Qu
- College of Veterinary Medicine, China Agricultural University, Beijing, China.,Agricultural Bio-pharmaceutical Laboratory, Qingdao Agricultural University, Qingdao, China
| | - Cunchun Dai
- Agricultural Bio-pharmaceutical Laboratory, Qingdao Agricultural University, Qingdao, China
| | - Zhihui Hao
- College of Veterinary Medicine, China Agricultural University, Beijing, China.,Agricultural Bio-pharmaceutical Laboratory, Qingdao Agricultural University, Qingdao, China
| | - Qihe Tang
- Agricultural Bio-pharmaceutical Laboratory, Qingdao Agricultural University, Qingdao, China
| | - Haixia Wang
- Agricultural Bio-pharmaceutical Laboratory, Qingdao Agricultural University, Qingdao, China
| | - Jinquan Wang
- College of Animal Medicine, Xinjiang Agricultural University, Wulumuqi, China
| | - Hongqiong Zhao
- College of Animal Medicine, Xinjiang Agricultural University, Wulumuqi, China
| |
Collapse
|
11
|
Pais GM, Liu J, Zepcan S, Avedissian SN, Rhodes NJ, Downes KJ, Moorthy GS, Scheetz MH. Vancomycin-Induced Kidney Injury: Animal Models of Toxicodynamics, Mechanisms of Injury, Human Translation, and Potential Strategies for Prevention. Pharmacotherapy 2020; 40:438-454. [PMID: 32239518 PMCID: PMC7331087 DOI: 10.1002/phar.2388] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Revised: 02/21/2020] [Accepted: 03/02/2020] [Indexed: 12/13/2022]
Abstract
Vancomycin is a recommended therapy in multiple national guidelines. Despite the common use, there is a poor understanding of the mechanistic drivers and potential modifiers of vancomycin-mediated kidney injury. In this review, historic and contemporary rates of vancomycin-induced kidney injury (VIKI) are described, and toxicodynamic models and mechanisms of toxicity from preclinical studies are reviewed. Aside from known clinical covariates that worsen VIKI, preclinical models have demonstrated that various factors impact VIKI, including dose, route of administration, and thresholds for pharmacokinetic parameters. The degree of acute kidney injury (AKI) is greatest with the intravenous route and higher doses that produce larger maximal concentrations and areas under the concentration curve. Troughs (i.e., minimum concentrations) have less of an impact. Mechanistically, preclinical studies have identified that VIKI is a result of drug accumulation in proximal tubule cells, which triggers cellular oxidative stress and apoptosis. Yet, there are several gaps in the knowledge that may represent viable targets to make vancomycin therapy less toxic. Potential strategies include prolonging infusions and lowering maximal concentrations, administration of antioxidants, administering agents that decrease cellular accumulation, and reformulating vancomycin to alter the renal clearance mechanism. Based on preclinical models and mechanisms of toxicity, we propose potential strategies to lessen VIKI.
Collapse
Affiliation(s)
- Gwendolyn M. Pais
- Department of Pharmacy Practice, Chicago College of Pharmacy, Midwestern University, Downers Grove, Illinois
- Pharmacometrics Center of Excellence, Midwestern University Chicago College of Pharmacy, Downers Grove, Illinois
| | - Jiajun Liu
- Department of Pharmacy Practice, Chicago College of Pharmacy, Midwestern University, Downers Grove, Illinois
- Pharmacometrics Center of Excellence, Midwestern University Chicago College of Pharmacy, Downers Grove, Illinois
| | - Sanja Zepcan
- Chicago College of Pharmacy, Midwestern University, Downers Grove, Illinois
| | - Sean N. Avedissian
- Antiviral Pharmacology Laboratory, University of Nebraska Medical Center (UNMC) Center for Drug Discovery, UNMC, Omaha, Nebraska
- College of Pharmacy, University of Nebraska Medical Center, Omaha, Nebraska
| | - Nathaniel J. Rhodes
- Department of Pharmacy Practice, Chicago College of Pharmacy, Midwestern University, Downers Grove, Illinois
- Pharmacometrics Center of Excellence, Midwestern University Chicago College of Pharmacy, Downers Grove, Illinois
| | - Kevin J. Downes
- Division of Infectious Diseases, The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
- Department of Pediatrics, The University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Ganesh S. Moorthy
- Division of Critical Care, Department of Anesthesiology and Critical Care, The University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Marc H. Scheetz
- Department of Pharmacy Practice, Chicago College of Pharmacy, Midwestern University, Downers Grove, Illinois
- Pharmacometrics Center of Excellence, Midwestern University Chicago College of Pharmacy, Downers Grove, Illinois
| |
Collapse
|
12
|
Qu S, Dai C, Guo H, Wang C, Hao Z, Tang Q, Wang H, Zhang Y. Rutin attenuates vancomycin‐induced renal tubular cell apoptosis via suppression of apoptosis, mitochondrial dysfunction, and oxidative stress. Phytother Res 2019; 33:2056-2063. [DOI: 10.1002/ptr.6391] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Revised: 02/13/2019] [Accepted: 04/23/2019] [Indexed: 12/30/2022]
Affiliation(s)
- Shaoqi Qu
- Agricultural Bio‐pharmaceutical LaboratoryQingdao Agricultural University Qingdao 266109 China
| | - Cunchun Dai
- Agricultural Bio‐pharmaceutical LaboratoryQingdao Agricultural University Qingdao 266109 China
| | - Hui Guo
- Agricultural Bio‐pharmaceutical LaboratoryQingdao Agricultural University Qingdao 266109 China
| | - Cuncai Wang
- Agricultural Bio‐pharmaceutical LaboratoryQingdao Agricultural University Qingdao 266109 China
| | - Zhihui Hao
- Agricultural Bio‐pharmaceutical LaboratoryQingdao Agricultural University Qingdao 266109 China
| | - Qihe Tang
- College of Chemistry and Pharmaceutical SciencesQingdao Agricultural University Qingdao China
| | - Haixia Wang
- College of Chemistry and Pharmaceutical SciencesQingdao Agricultural University Qingdao China
| | - Yanping Zhang
- College of Chemistry and Pharmaceutical SciencesQingdao Agricultural University Qingdao China
| |
Collapse
|
13
|
Rutin Attenuates Vancomycin-Induced Nephrotoxicity by Ameliorating Oxidative Stress, Apoptosis, and Inflammation in Rats. Antimicrob Agents Chemother 2018; 63:AAC.01545-18. [PMID: 30397060 DOI: 10.1128/aac.01545-18] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2018] [Accepted: 10/25/2018] [Indexed: 01/12/2023] Open
Abstract
Nephrotoxicity is the major limiting factor for the clinical use of vancomycin (VCM) for treatment of serious infections caused by multiresistant Gram-positive bacteria. This study investigated the renal protective activity of rutin in a rat model of VCM-induced kidney injury in male Wistar rats. VCM administered intraperitoneally at 200 mg/kg twice daily for 7 successive days resulted in significant elevation of blood urea nitrogen and creatinine, as well as urinary N-acetyl-β-D-glucosaminidase. Coadministration of VCM with oral rutin at 150 mg/kg significantly reduced these markers of kidney damage. Rutin also significantly attenuated VCM-induced oxidative stress, inflammatory cell infiltration, apoptosis, and decreased interleukin-1β and tumor necrosis factor alpha levels (all P < 0.05 or 0.01) in kidneys. Renal recovery from VCM injury was achieved by rutin through increases in Nrf2 and HO-1 and a decrease in NF-κB expression. Our results demonstrated a protective effect of rutin on VCM-induced kidney injury through suppression of oxidative stress, apoptosis, and downregulation of the inflammatory response. This study highlights a role for oral rutin as an effective intervention to ameliorate nephrotoxicity in patients undergoing VCM therapy.
Collapse
|
14
|
Uckun Z, Guzel S, Canacankatan N, Yalaza C, Kibar D, Coskun Yilmaz B. Potential protective effects of naringenin against vancomycin-induced nephrotoxicity via reduction on apoptotic and oxidative stress markers in rats. Drug Chem Toxicol 2018; 43:104-111. [PMID: 30257567 DOI: 10.1080/01480545.2018.1512612] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Abstract
Vancomycin (VCM), a glycopeptide antibiotic, is a drug widely used in severe infections. However, VCM induce notable nephrotoxic side effects. Naringenin (NAR) is a natural of flavonoid and are known as strongly antioxidant, nefroprotective, antiapoptotic, and anti-inflammatory. The purpose of this study was to determine the potential protective effects of NAR against VCM-induced nephrotoxicity by measuring apoptotic and oxidative stress markers and evaluating histopathological alterations in rats. For this purpose, we used male Wistar albino rats that divided into seven groups: (i) Control [saline, intraperitoneally (i.p.)], (ii) carboxymethyl cellulose (0.5% CMC, orally), (iii) VCM (400 mg/kg, i.p.), (iv) NAR100 (100 mg/kg, orally), (v) VCM + NAR25 (25 mg/kg, orally), (vi) VCM + NAR50 (50 mg/kg, orally), and (vii) VCM + NAR100 (100 mg/kg, orally) groups. VCM administration was started one day after the first treatment of NAR and continued across 7-day. Caspase-3, -8, and-9 activities and malondialdehyde (MDA) and nitric oxide (NO) levels were measured by colorimetric methods in the kidney tissues, creatinine, and blood urea nitrogen (BUN) levels were analyzed based on ELISA in serum. Caspase-3 and -8 activities, NO levels, serum creatinine and BUN levels were significantly higher in VCM group in comparison with VCM + NAR (25, 50, and 100) groups (p < 0.05). Caspase-9 activity and MDA were significantly higher in VCM group compared to VCM + NAR (25 and 50) groups (p < 0.05). Histopathological alterations in VCM group were significantly diminished by administration of NAR, especially NAR 25. In conclusion, NAR 25 and 50 mg have more potent protective effects on VCM-induced nephrotoxicity compared to NAR 100 mg.
Collapse
Affiliation(s)
- Zuhal Uckun
- Department of Pharmaceutical Toxicology, Mersin University, Mersin, Turkey
| | - Sevda Guzel
- Department of Pharmacognosy, Mersin University, Mersin, Turkey
| | | | - Cem Yalaza
- Department of Medical Services and Techniques, Toros University Vocational School, Mersin, Turkey
| | - Deniz Kibar
- Department of Histology and Embryology, Mersin University, Mersin, Turkey
| | - Banu Coskun Yilmaz
- Department of Histology and Embryology, Mersin University, Mersin, Turkey
| |
Collapse
|
15
|
Velez J, Obadan N, Kaushal A, Alzubaidi M, Bhasin B, Sachdev S, Karakala N, Arthur J, Nesbit R, Phadke G. Vancomycin-Associated Acute Kidney Injury with a Steep Rise in Serum Creatinine. Nephron Clin Pract 2018; 139:131-142. [DOI: 10.1159/000487149] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Accepted: 01/24/2018] [Indexed: 01/09/2023] Open
|
16
|
Kers J, Leemans JC, Linkermann A. An Overview of Pathways of Regulated Necrosis in Acute Kidney Injury. Semin Nephrol 2018; 36:139-52. [PMID: 27339380 DOI: 10.1016/j.semnephrol.2016.03.002] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Necrosis is the predominant form of regulated cell death in acute kidney injury (AKI) and represents results in the formation of casts that appear in the urine sedimentation, referred to as muddy brown casts, which are part of the diagnosis of AKI. Pathologists referred to this typical feature as acute tubular necrosis. We are only beginning to understand the dynamics and the molecular pathways that underlie such typical necrotic morphology. In this review, we provide an overview of candidate pathways and summarize the emerging evidence for the relative contribution of these pathways of regulated necrosis, such as necroptosis, ferroptosis, mitochondrial permeability transition-mediated regulated necrosis, parthanatos, and pyroptosis. Inhibitors of each of these pathways are available, and clinical trials may be started after the detection of the most promising drug targets, which will be discussed here. With the global burden of AKI in mind, inhibitiors of regulated necrosis represent promising means to prevent this disease.
Collapse
Affiliation(s)
- Jesper Kers
- Department of Pathology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands.
| | - Jaklien C Leemans
- Department of Pathology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Andreas Linkermann
- Clinic for Nephrology and Hypertension, Christian-Albrechts-University Kiel, Kiel, Germany
| |
Collapse
|
17
|
Wang J, Li H, Qiu S, Dong Z, Xiang X, Zhang D. MBD2 upregulates miR-301a-5p to induce kidney cell apoptosis during vancomycin-induced AKI. Cell Death Dis 2017; 8:e3120. [PMID: 29022913 PMCID: PMC5682674 DOI: 10.1038/cddis.2017.509] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Revised: 08/14/2017] [Accepted: 08/24/2017] [Indexed: 02/07/2023]
Abstract
Despite DNA methylation occurred in acute kidney injury (AKI), how it influenced progression of AKI remains unclear. Methyl-CpG-binding domain protein 2 (MBD2), a protein readers of methylation, was used to analyze the impact of DNA methylation on vancomycin (VAN)-induced AKI. Here, in cultured human kidney tubular epithelial cells (HK-2), we show that knockdown of MBD2 by siRNA attenuated VAN-induced apoptosis, caspase activity, and the expression of BAX and cleaved caspase 3. Interestingly, knockdown of MBD2 by siRNA was associated with the suppression of miR-301a-5p. Mechanistic studies confirmed MBD2 binds to these methylated CpG elements of miR-301a-5p promoter, and then activates miR-301a-5p promoter by suppressing methylation. Furthermore, anti-miR-301a-5p significantly blocked VAN-induced apoptosis and caspase activity in HK-2 cells, which was accompanied by downregulation of p53, and upregulation of MITF, HDGF and MDM-4 together. The latter genes were further identified as target genes of miR-301a-5p, and silencing of MDM-4 promoted p53 accumulation. In vivo, mice with MBD2 knockout (MBD2-KO) were counteracted to VAN-induced AKI, indicated by the analysis of renal function, histology, apoptosis and inflammation. MBD2-KO also significantly suppressed the expression of miR-301a-5p, p53, BAX and cleaved caspase 3, and restored the expression of MDM-4, MITF and HDGF. Finally, in vivo inhibition of miR-301a-5p also ameliorated VAN-induced AKI. Together, these results show the novel MBD2/miR-301a-5p/MITF, HDGF and MDM-4/p53 pathway in VAN-induced AKI.
Collapse
Affiliation(s)
- Juan Wang
- Department of Emergency Medicine, Second Xiangya Hospital, Central South University, Changsha, Hunan, People’s Republic of China
- Emergency Medicine and Difficult Diseases Institute, Second Xiangya Hospital, Central South University, Changsha, Hunan, People’s Republic of China
| | - Huiling Li
- Department of Ophthalmology, Second Xiangya Hospital, Central South University, Changsha, Hunan, People’s Republic of China
| | - Shuangfa Qiu
- Department of Emergency Medicine, Second Xiangya Hospital, Central South University, Changsha, Hunan, People’s Republic of China
- Emergency Medicine and Difficult Diseases Institute, Second Xiangya Hospital, Central South University, Changsha, Hunan, People’s Republic of China
| | - Zheng Dong
- Department of Nephrology, Second Xiangya Hospital, Central South University, Changsha, Hunan, People’s Republic of China
- Department of Cellular Biology and Anatomy, Medical College of Georgia at Georgia Regents University and Charlie Norwood VA Medical Center, Augusta, GA, USA
| | - Xudong Xiang
- Department of Emergency Medicine, Second Xiangya Hospital, Central South University, Changsha, Hunan, People’s Republic of China
- Emergency Medicine and Difficult Diseases Institute, Second Xiangya Hospital, Central South University, Changsha, Hunan, People’s Republic of China
| | - Dongshan Zhang
- Department of Emergency Medicine, Second Xiangya Hospital, Central South University, Changsha, Hunan, People’s Republic of China
- Emergency Medicine and Difficult Diseases Institute, Second Xiangya Hospital, Central South University, Changsha, Hunan, People’s Republic of China
- Department of Nephrology, Second Xiangya Hospital, Central South University, Changsha, Hunan, People’s Republic of China
| |
Collapse
|
18
|
Celik-Ozenci C, Kuscu N, Gungor-Ordueri NE, Tasatargil A, Sahin P, Durmus H. Inhibition of poly(ADP-ribose) polymerase may have preventive potential for varicocoele-associated testicular damage in rats. Andrology 2016; 5:362-369. [DOI: 10.1111/andr.12305] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Revised: 09/27/2016] [Accepted: 10/28/2016] [Indexed: 01/29/2023]
Affiliation(s)
- C. Celik-Ozenci
- Department of Histology and Embryology; Akdeniz University School of Medicine; Antalya Turkey
| | - N. Kuscu
- Department of Histology and Embryology; Akdeniz University School of Medicine; Antalya Turkey
| | - N. E. Gungor-Ordueri
- Department of Histology and Embryology; Biruni University School of Medicine; Istanbul Turkey
| | - A. Tasatargil
- Department of Pharmacology; Akdeniz University School of Medicine; Antalya Turkey
| | - P. Sahin
- Department of Histology and Embryology; Akdeniz University School of Medicine; Antalya Turkey
| | - H. Durmus
- Department of Oncology; Sana Klinikum Hameln-Pyrmont; Hameln Germany
| |
Collapse
|
19
|
Abstract
In recent times the use of larger doses of vancomycin aimed at curbing the increasing incidence of resistant strains of Staphylococcus aureus has led to a wider report of acute kidney injury (AKI). Apart from biological plausibility, causality is implied by the predictive association of AKI with larger doses, longer duration, and graded plasma concentrations of vancomycin. AKI is more likely to occur with the concurrent use of nephrotoxic agents, and in critically ill patients who are susceptible to poor renal perfusion. Although most vancomycin-induced AKI cases are mild and therefore reversible, their occurrence may be associated with greater incidence of end-stage kidney disease and higher mortality rate. The strategy for its prevention includes adequate renal perfusion and therapeutic drug monitoring in high-risk individuals. In the near future, there is feasibility of renoprotective use of antioxidative substances in the delivery of vancomycin.
Collapse
|
20
|
Liu SB, Liu J, Liu DW, Wang XT, Yang RL. Inhibition of Poly-(ADP-Ribose) Polymerase Protects the Kidney in a Canine Model of Endotoxic Shock. Nephron Clin Pract 2015; 130:281-92. [PMID: 26184635 DOI: 10.1159/000435815] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2014] [Accepted: 06/08/2015] [Indexed: 11/19/2022] Open
Abstract
Poly-(ADP-ribose) polymerases (PARPs), a super family of enzymes, play important roles in preserving genomic integrity, regulating transcriptions, protecting telomeres and determining cell fate. PARP overactivation leads to metabolic disorder and cell injury via depletion of energy substance. However, it is still unclear whether PARP overactivation happens during acute kidney injury (AKI) caused by endotoxic shock (ES). Here, we built a canine model of lipopolysaccharide-induced ES to explore the role of PARP during the development AKI. We also used an intravenous injection of 3-aminobenzamide (3-AB) to further explore whether PARP inhibition rescues the kidney from injury. Cell fate and energy metabolism were detected to explore the underlying mechanisms. As a result, Western blot and immunohistochemistry assays showed PARP overactivation in the very early phase of ES. Through PARP inhibition by 3-AB, we observed significant improvement of systemic hemodynamics, renal hemodynamics, renal oxygen metabolism and renal tubular cell apoptosis. These findings indicated that overactivation of PARP plays an important role in septic AKI. Inhibition of PARP overactivation may protect renal function against hemodynamic disorder, renal metabolism disturbance and renal cell apoptosis during endotoxic AKI.
Collapse
Affiliation(s)
- Si-bo Liu
- Surgical Intensive Care Unit, Dalian Municipal Central Hospital, Dalian City, China
| | | | | | | | | |
Collapse
|
21
|
Zhou TB, Jiang ZP. Role of poly (ADP-ribose)-polymerase and its signaling pathway with renin-angiotensin aldosterone system in renal diseases. J Recept Signal Transduct Res 2014; 34:143-148. [PMID: 24303937 DOI: 10.3109/10799893.2013.865748] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Poly(ADP-ribose) polymerase (PARP), a ubiquitous, chromatin-bound enzyme, plays a crucial role in many processes, including DNA repair, cell death, metabolism, and inflammatory responses, by activating DNA repair pathways responsible for cellular survival. Renin-angiotensin-aldosterone system (RAAS) genes encode renin, angiotensinogen, angiotensin-converting enzyme, angiotensin type-1 receptor and aldosterone synthase gene. RAAS is a hormone system which acts on multiple physiologic pathways primarily by regulating blood pressure, electrolyte and fluid homeostasis in mammals, but also by local autocrine and paracrine actions. The current status quo of scientific evidence shows that there might be a signaling pathway between PARP and RAAS. Herein, we review the role of PARP and its signaling pathways with RAAS in renal diseases.
Collapse
Affiliation(s)
- Tian-Biao Zhou
- Department of Nephrology, The Sixth Affiliated Hospital of Sun Yat-Sen University , Guangzhou , China
| | | |
Collapse
|
22
|
Konishi H, Morita Y, Mizumura M, Iga I, Nagai K. Difference in nephrotoxicity of vancomycin administered once daily and twice daily in rats. J Chemother 2014; 25:273-8. [PMID: 24070134 DOI: 10.1179/1973947812y.0000000067] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
We compared the degree of nephrotoxicity of vancomycin (VCM) administered once daily and twice daily in rats. VCM was intraperitoneally administered once daily to rats at a dose of 400 mg/kg (VCM-1-treated) or administered at a dose of 200 mg/kg twice daily at 12-hour intervals (VCM-2-treated) for 7 consecutive days. Creatinine clearance was decreased more markedly in VCM-1 rats relative to VCM-2 rats, although there was no significant difference in renal accumulation of VCM between the two groups. Renal superoxide dismutase activity was lower in VCM-1 rats than that in VCM-2 rats. The magnitude of histological change in kidney tissue was in agreement with the degree of alterations in the abovementioned biochemical values. These results suggest that the nephrotoxic effect of once-daily VCM administration is more pronounced than that of the twice-daily treatment. Our findings provide fundamental evidence for the advantage in choosing a divided VCM administration to attenuate nephrotoxicity.
Collapse
|
23
|
Prevention of vancomycin induced nephrotoxicity: a review of preclinical data. Eur J Clin Pharmacol 2012; 69:747-54. [DOI: 10.1007/s00228-012-1406-3] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2012] [Accepted: 09/04/2012] [Indexed: 12/28/2022]
|
24
|
Dalaklioglu S, Sahin P, Ordueri EG, Celik-Ozenci C, Tasatargil A. Potential role of poly(ADP-ribose) polymerase (PARP) activation in methotrexate-induced nephrotoxicity and tubular apoptosis. Int J Toxicol 2012; 31:430-40. [PMID: 22914891 DOI: 10.1177/1091581812457430] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Nephrotoxicity is one of the serious dose-limiting complications of methotrexate (MTX) when used in the treatment of various malignancies and nononcological diseases. The aim of this study was to investigate the role of poly(adenosine diphosphate ribose) polymerase (PARP) activity in MTX-induced nephrotoxicity. Rats were divided into 4 groups as control, MTX treated (MTX, 7 mg/kg per d, intraperitoneally [ip], once daily for 3 consecutive days), MTX plus 1,5-isoquinelinediol (ISO, a PARP inhibitor, 3 mg/kg per d, i.p.) treated, or ISO treated. Histopathology of kidneys was evaluated by light microscopy. Terminal deoxynucleotidyl transferase deoxyuridine triphosphate nick end labeling assay was used to analyze apoptosis in kidney sections. Blood urea nitrogen (BUN), serum creatinine, and urinary N-acetyl-β-d-glucosaminidase (NAG) were used as biochemical markers of MTX-induced renal injury. Our results showed that MTX administration significantly increased BUN, serum creatinine, and urinary NAG levels. The PARP-1 and PAR (a product of PARP activity) expression and apoptotic cell death were also markedly increased in renal tubules after MTX administration. The ISO treatment attenuated MTX-induced renal injury, as indicated by BUN and serum creatinine levels, urinary NAG excretion, and renal histology. The PARP inhibitor treatment reduced PARP-1 and PAR expression to levels similar to that of controls. These results revealed that ISO may have a protective effect against the nephrotoxic effects of MTX by inhibiting PARP activation. This is the first study that demonstrates the role of PARP activation in MTX-induced nephrotoxicity and tubular apoptosis.
Collapse
|
25
|
Htike NL, Santoro J, Gilbert B, Elfenbein IB, Teehan G. Biopsy-proven vancomycin-associated interstitial nephritis and acute tubular necrosis. Clin Exp Nephrol 2011; 16:320-4. [PMID: 22086124 DOI: 10.1007/s10157-011-0559-1] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2011] [Accepted: 10/25/2011] [Indexed: 10/15/2022]
Abstract
Fewer than ten biopsy-proven case reports exist on vancomycin-associated interstitial nephritis (VAIN) and vancomycin-associated acute tubular necrosis (VAATN). Among these, several are confounded by the use of other potentially offending drugs. We report a case of isolated VAIN/VAATN in a patient on no other potentially nephrotoxic agents other than vancomycin. The patient received intravenous vancomycin for coagulase-negative staphylococcus bacteremia. Her baseline serum creatinine of 0.9 mg/dL increased to 9.6 mg/dL after 1 week of therapy during which vancomycin levels peaked at 141 μg/mL. Renal biopsy revealed acute interstitial nephritis with lymphocytic and eosinophilic infiltrate and acute tubular necrosis. Upon discontinuation of vancomycin and administration of prednisone complete renal recovery ensued over a period of 4 weeks.
Collapse
Affiliation(s)
- Naing Lin Htike
- Lankenau Medical Center, 100 E Lancaster Ave, Lankenau Medical Office Building West, Suite 130, Wynnewood, PA 19096, USA.
| | | | | | | | | |
Collapse
|
26
|
Giansanti V, Donà F, Tillhon M, Scovassi AI. PARP inhibitors: new tools to protect from inflammation. Biochem Pharmacol 2010; 80:1869-77. [PMID: 20417190 DOI: 10.1016/j.bcp.2010.04.022] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2010] [Revised: 04/12/2010] [Accepted: 04/13/2010] [Indexed: 10/19/2022]
Abstract
Poly(ADP-ribosylation) consists in the conversion of β-NAD(+) into ADP-ribose, which is then bound to acceptor proteins and further used to form polymers of variable length and structure. The correct turnover of poly(ADP-ribose) is ensured by the concerted action of poly(ADP-ribose) polymerase (PARP) and poly(ADP-ribose) glycohydrolase (PARG) enzymes, which are responsible for polymer synthesis and degradation, respectively. Despite the positive role of poly(ADP-ribosylation) in sensing and repairing DNA damage, generated also by ROS, PARP over-activation could allow NAD depletion and consequent necrosis, thus leading to an inflammatory condition in many diseases. In this respect, inhibition of PARP enzymes could exert a protective role towards a number of pathological conditions; i.e. the combined treatment of tumors with PARP inhibitors/anticancer agents proved to have a beneficial effect in cancer therapy. Thus, pharmacological inactivation of poly(ADP-ribosylation) could represent a novel therapeutic strategy to limit cellular injury and to attenuate the inflammatory processes that characterize many disorders.
Collapse
Affiliation(s)
- Vincenzo Giansanti
- Istituto di Genetica Molecolare CNR, Via Abbiategrasso 207, I-27100 Pavia, Italy
| | | | | | | |
Collapse
|