1
|
Alqadi R, Alqumia A, Alhomoud IS, Alhowail A, Aldubayan M, Mohammed HA, Alhmoud H, Khan RA. Cyclosporine: Immunosuppressive effects, entwined toxicity, and clinical modulations of an organ transplant drug. Transpl Immunol 2025; 88:102147. [PMID: 39549927 DOI: 10.1016/j.trim.2024.102147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/21/2024] [Revised: 11/10/2024] [Accepted: 11/10/2024] [Indexed: 11/18/2024]
Abstract
The discovery and use of cyclosporine since its inception into the clinics in the '70s and up have played a crucial role in advancing transplant therapy, and containment of the immune-based rejections. The drug has improved the high rates of acute rejections and has supported early graft survival. However, the long-term survival of renal allografts is still less prevalent, and an in-depth analysis, as well as reported findings led us to believe that there is a chronic irreversible component to the drug, that is tackled through its metabolites, and that causes toxicity, which has led to new therapies, including monoclonal antibody-based medications. A recap of the immunosuppressive effects, and entwined toxicity of the drug, now relegated primarily to bone marrow early transplants, is being overviewed for the past protocols that were used to minimize, and avoid, or use this calcineurin inhibitor class of drug, cyclosporine, in combination with other drugs. The current review circumvents the cyclosporine's mechanism of action, pathophysiology, cytochrome roles, and other factors associated with acute and chronic toxicity developments. The review also attempts to find conclusive strategies reported in the recent studies to avoid its toxic side effects, and develop a safe-use strategy for the drug. Gastrointestinal decontamination, supporting the airway, monitoring for signs of respiratory insufficiency, monitoring for severe reactions, such as seizures, need for administration of oxygen, and avoiding the administration of drugs, that increase the blood levels of the cyclosporine, are beneficial interventions, when encountering cyclosporine toxicity cases. The constrained therapeutic outcomes have also led to redesign, and making use of combined formulations to reassess the pharmacokinetics of the drug.
Collapse
Affiliation(s)
- Razan Alqadi
- Department of Pharmacy, King Saud Hospital, Unaizah, Qassim 56249, Saudi Arabia
| | - Amal Alqumia
- Department of Pharmacy, King Fahd Specialist Hospital, Buraydah, Qassim 52719, Saudi Arabia
| | - Ibrahim S Alhomoud
- Department of Pharmacy Practice, College of Pharmacy, Qassim University, Qassim 51452, Saudi Arabia
| | - Ahmad Alhowail
- Department of Pharmacology and Toxicology, College of Pharmacy, Qassim University, Qassim 51452, Saudi Arabia
| | - Maha Aldubayan
- Department of Pharmacology and Toxicology, College of Pharmacy, Qassim University, Qassim 51452, Saudi Arabia
| | - Hamdoon A Mohammed
- Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, Qassim University, Qassim 51452, Saudi Arabia
| | - Hussam Alhmoud
- Department of Pharmacy Practice, College of Pharmacy, Qassim University, Qassim 51452, Saudi Arabia
| | - Riaz A Khan
- Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, Qassim University, Qassim 51452, Saudi Arabia.
| |
Collapse
|
2
|
Trillos-Almanza MC, Chvatal-Medina M, Connelly MA, Moshage H, Bakker SJL, de Meijer VE, Blokzijl H, Dullaart RPF. Circulating Trimethylamine-N-Oxide Is Elevated in Liver Transplant Recipients. Int J Mol Sci 2024; 25:6031. [PMID: 38892218 PMCID: PMC11172608 DOI: 10.3390/ijms25116031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 04/17/2024] [Revised: 05/28/2024] [Accepted: 05/29/2024] [Indexed: 06/21/2024] Open
Abstract
Liver transplant recipients (LTRs) have lower long-term survival rates compared with the general population. This underscores the necessity for developing biomarkers to assess post-transplantation mortality. Here we compared plasma trimethylamine-N-oxide (TMAO) levels with those in the general population, investigated its determinants, and interrogated its association with all-cause mortality in stable LTRs. Plasma TMAO was measured in 367 stable LTRs from the TransplantLines cohort (NCT03272841) and in 4837 participants from the population-based PREVEND cohort. TMAO levels were 35% higher in LTRs compared with PREVEND participants (4.3 vs. 3.2 µmol/L, p < 0.001). Specifically, TMAO was elevated in LTRs with metabolic dysfunction-associated steatotic liver disease, alcohol-associated liver disease, and polycystic liver disease as underlying etiology (p < 0.001 for each). Among LTRs, TMAO levels were independently associated with eGFR (std. β = -0.43, p < 0.001) and iron supplementation (std. β = 0.13, p = 0.008), and were associated with mortality (29 deaths during 8.6 years follow-up; log-rank test p = 0.017; hazard ratio of highest vs. lowest tertile 4.14, p = 0.007). In conclusion, plasma TMAO is likely elevated in stable LTRs, with impaired eGFR and iron supplementation as potential contributory factors. Our preliminary findings raise the possibility that plasma TMAO could contribute to increased mortality risk in such patients, but this need to be validated through a series of rigorous and methodical studies.
Collapse
Affiliation(s)
- Maria Camila Trillos-Almanza
- Department of Gastroenterology and Hepatology, University Medical Center Groningen, University of Groningen, P.O. Box 30.001, 9700 RB Groningen, The Netherlands; (M.C.-M.); (H.M.); (H.B.)
| | - Mateo Chvatal-Medina
- Department of Gastroenterology and Hepatology, University Medical Center Groningen, University of Groningen, P.O. Box 30.001, 9700 RB Groningen, The Netherlands; (M.C.-M.); (H.M.); (H.B.)
| | | | - Han Moshage
- Department of Gastroenterology and Hepatology, University Medical Center Groningen, University of Groningen, P.O. Box 30.001, 9700 RB Groningen, The Netherlands; (M.C.-M.); (H.M.); (H.B.)
| | - TransplantLines Investigators
- Groningen Institute for Organ Transplantation, University Medical Center Groningen, University of Groningen, 9700 AD Groningen, The Netherlands;
| | - Stephan J. L. Bakker
- Department of Internal Medicine, Division of Nephrology, University Medical Center Groningen, University of Groningen, P.O. Box 30.001, 9700 RB Groningen, The Netherlands;
| | - Vincent E. de Meijer
- Department of Surgery, Division of Hepato-Pancreato-Biliary Surgery and Liver Transplantation, University Medical Center Groningen, University of Groningen, P.O. Box 30.001, 9700 RB Groningen, The Netherlands;
| | - Hans Blokzijl
- Department of Gastroenterology and Hepatology, University Medical Center Groningen, University of Groningen, P.O. Box 30.001, 9700 RB Groningen, The Netherlands; (M.C.-M.); (H.M.); (H.B.)
| | - Robin P. F. Dullaart
- Department of Internal Medicine, Division of Endocrinology, University Medical Center Groningen, University of Groningen, P.O. Box 30.001, 9700 RB Groningen, The Netherlands;
| |
Collapse
|
3
|
Abstract
Major advances in scientific discovery and insights that stem from the development and use of new techniques and models can bring remarkable progress to conventional toxicology. Although animal testing is still considered as the "gold standard" in traditional toxicity testing, there is a necessity for shift from animal testing to alternative methods regarding the drug safety testing owing to the emerging state-of-art techniques and the proposal of 3Rs (replace, reduce, and refine) towards animal welfare. This review describes some recent research methods in drug discovery toxicology, including in vitro cell and organ-on-a-chip, imaging systems, model organisms (C. elegans, Danio rerio, and Drosophila melanogaster), and toxicogenomics in modern toxicology testing.
Collapse
Affiliation(s)
- Bowen Tang
- PTC Therapeutics Inc, South Plainfield, NJ, USA
| | - Vijay More
- PTC Therapeutics Inc, South Plainfield, NJ, USA
| |
Collapse
|
4
|
Burghelea D, Moisoiu T, Ivan C, Elec A, Munteanu A, Iancu ȘD, Truta A, Kacso TP, Antal O, Socaciu C, Elec FI, Kacso IM. The Use of Machine Learning Algorithms and the Mass Spectrometry Lipidomic Profile of Serum for the Evaluation of Tacrolimus Exposure and Toxicity in Kidney Transplant Recipients. Biomedicines 2022; 10:biomedicines10051157. [PMID: 35625894 PMCID: PMC9138871 DOI: 10.3390/biomedicines10051157] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 04/29/2022] [Revised: 05/13/2022] [Accepted: 05/14/2022] [Indexed: 12/10/2022] Open
Abstract
Tacrolimus has a narrow therapeutic window; a whole-blood trough target concentration of between 5 and 8 ng/mL is considered a safe level for stable kidney transplant recipients. Tacrolimus serum levels must be closely monitored to obtain a balance between maximizing efficacy and minimizing dose-related toxic effects. Currently, there is no specific tacrolimus toxicity biomarker except a graft biopsy. Our study aimed to identify specific serum metabolites correlated with tacrolinemia levels using serum high-precision liquid chromatography–mass spectrometry and standard laboratory evaluation. Three machine learning algorithms were used (Naïve Bayes, logistic regression, and Random Forest) in 19 patients with high tacrolinemia (8 ng/mL) and 23 patients with low tacrolinemia (5 ng/mL). Using a selected panel of five lipid metabolites (phosphatidylserine, phosphatidylglycerol, phosphatidylethanolamine, arachidyl palmitoleate, and ceramide), Mg2+, and uric acid, all three machine learning algorithms yielded excellent classification accuracies between the two groups. The highest classification accuracy was obtained by Naïve Bayes, with an area under the curve of 0.799 and a classification accuracy of 0.756. Our results show that using our identified five lipid metabolites combined with Mg2+ and uric acid serum levels may provide a novel tool for diagnosing tacrolimus toxicity in kidney transplant recipients. Further validation with targeted MS and biopsy-proven TAC toxicity is needed.
Collapse
Affiliation(s)
- Dan Burghelea
- Clinical Institute of Urology and Renal Transplantation, 400006 Cluj-Napoca, Romania; (D.B.); (T.M.); (A.E.); (A.M.); (O.A.)
- Department of Urology, “Iuliu Hatieganu” University of Medicine and Pharmacy Cluj-Napoca, 400012 Cluj-Napoca, Romania
| | - Tudor Moisoiu
- Clinical Institute of Urology and Renal Transplantation, 400006 Cluj-Napoca, Romania; (D.B.); (T.M.); (A.E.); (A.M.); (O.A.)
- Department of Urology, “Iuliu Hatieganu” University of Medicine and Pharmacy Cluj-Napoca, 400012 Cluj-Napoca, Romania
- Biomed Data Analytics SRL, 400696 Cluj-Napoca, Romania
| | - Cristina Ivan
- “Regina Maria” Hospital, 400117 Cluj-Napoca, Romania;
| | - Alina Elec
- Clinical Institute of Urology and Renal Transplantation, 400006 Cluj-Napoca, Romania; (D.B.); (T.M.); (A.E.); (A.M.); (O.A.)
| | - Adriana Munteanu
- Clinical Institute of Urology and Renal Transplantation, 400006 Cluj-Napoca, Romania; (D.B.); (T.M.); (A.E.); (A.M.); (O.A.)
| | - Ștefania D. Iancu
- Faculty of Physics, Babeș-Bolyai University, 400084 Cluj-Napoca, Romania;
| | - Anamaria Truta
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, “Iuliu Hațieganu” University of Medicine and Pharmacy Cluj-Napoca, 400337 Cluj-Napoca, Romania;
| | - Teodor Paul Kacso
- Department of Nephrology, “Iuliu Hatieganu” University of Medicine and Pharmacy Cluj-Napoca, 400012 Cluj-Napoca, Romania; (T.P.K.); (I.M.K.)
| | - Oana Antal
- Clinical Institute of Urology and Renal Transplantation, 400006 Cluj-Napoca, Romania; (D.B.); (T.M.); (A.E.); (A.M.); (O.A.)
- Department of Anesthesiology, “Iuliu Hatieganu” University of Medicine and Pharmacy Cluj-Napoca, 400012 Cluj-Napoca, Romania
| | - Carmen Socaciu
- Faculty of Food Science and Technology, University of Agricultural Science and Veterinary Medicine Cluj-Napoca, Calea Mănăştur 3–5, 400372 Cluj-Napoca, Romania;
| | - Florin Ioan Elec
- Clinical Institute of Urology and Renal Transplantation, 400006 Cluj-Napoca, Romania; (D.B.); (T.M.); (A.E.); (A.M.); (O.A.)
- Department of Urology, “Iuliu Hatieganu” University of Medicine and Pharmacy Cluj-Napoca, 400012 Cluj-Napoca, Romania
- Correspondence: ; Tel.: +40-756285972
| | - Ina Maria Kacso
- Department of Nephrology, “Iuliu Hatieganu” University of Medicine and Pharmacy Cluj-Napoca, 400012 Cluj-Napoca, Romania; (T.P.K.); (I.M.K.)
| |
Collapse
|
5
|
Sempio C, Almaraz-Quinones N, Jackson M, Zhao W, Wang GS, Liu Y, Leehey M, Knupp K, Klawitter J, Christians U, Klawitter J. Simultaneous Quantification of 17 Cannabinoids bY LC-MS-MS in Human Plasma. J Anal Toxicol 2021; 46:383-392. [PMID: 33754154 DOI: 10.1093/jat/bkab030] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 01/31/2021] [Revised: 03/09/2021] [Accepted: 03/22/2021] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND In recent years, the surge in use and of clinical trials involving tetrahydrocannabinol (THC) and cannabidiol (CBD) has increased the need for sensitive and specific analytical assays to measure said compounds in patients, to establish dose-effect relationships and to gain knowledge of their pharmacokinetics and metabolism. We developed and validated an online extraction high-performance liquid chromatography- tandem mass spectrometry (LC-MS/MS) method for simultaneous quantification of 17 cannabinoids and metabolites including THC and its metabolites, CBD and its metabolites and other minor cannabinoids in human plasma. METHODS CBD-glucuronide (CBD-gluc) standard was produced in-house by isolation of CBD-gluc from urine of patients using pure CBD oil. For calibration standards and quality control samples, human plasma was spiked with cannabinoids at varying concentrations within the working range of the respective compound and 200 µL was extracted using a simple one-step protein precipitation procedure. The extracts were analyzed using online trapping LC/LC-atmospheric pressure chemical ionization (APCI)-MS/MS running in the positive multiple reaction monitoring (MRM) mode. RESULTS The lower limit of quantification ranged from 0.78 ng/mL to 7.8 ng/mL and the upper limits of quantification were between 100 ng/mL and 2000 ng/mL. Inter-day analytical accuracy and imprecision ranged from 90.4 to 111% and from 3.1 to 17.4%, respectively. The analysis of plasma samples collected during clinical studies showed that (3R-trans)-Cannabidiol-7-oic Acid (7-CBD-COOH) was the major human metabolite with 5960% of CBD followed by 7-hydroxy-CBD (177%), CBD-gluc (157%) and 6α-hydroxy-CBD (39.8%); 6β-hydroxy-CBD was not detected in any of the samples. CONCLUSIONS In the present study, we developed and validated a robust LC-MS/MS assay for the simultaneous quantification of cannabinoids and their metabolites, which has been used to measure >5,000 samples in clinical studies. Moreover, we were able to quantify CBD-gluc and showed that 7-CBD-COOH, 7-hydroxy-CBD and CBD-gluc are the major CBD metabolites in human plasma.
Collapse
Affiliation(s)
- Cristina Sempio
- Department of Anesthesiology, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA
| | - Nohemi Almaraz-Quinones
- Department of Anesthesiology, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA
| | - Matthew Jackson
- Department of Anesthesiology, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA
| | - Wanzhu Zhao
- Department of Anesthesiology, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA
| | - George Sam Wang
- Section of Emergency Medicine and Medical Toxicology, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Children's Hospital Colorado, Aurora, CO, 80045, USA
| | - Ying Liu
- Department of Neurology, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Maureen Leehey
- Department of Neurology, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Kelly Knupp
- Department of Neurology and Pediatrics, University of Colorado, Anschutz Medical Campus, Children's Hospital Colorado, Aurora, CO 80045, USA
| | - Jelena Klawitter
- Department of Anesthesiology, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA
| | - Uwe Christians
- Department of Anesthesiology, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA
| | - Jost Klawitter
- Department of Anesthesiology, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA
| |
Collapse
|
6
|
Rivera-Velez SM, Navas J, Villarino NF. Applying metabolomics to veterinary pharmacology and therapeutics. J Vet Pharmacol Ther 2021; 44:855-869. [PMID: 33719079 DOI: 10.1111/jvp.12961] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 09/12/2020] [Accepted: 02/08/2021] [Indexed: 02/06/2023]
Abstract
Metabolomics is the large-scale study of low-molecular-weight substances in a biological system in a given physiological state at a given time point. Metabolomics can be applied to identify predictors of inter-individual variability in drug response, provide clinicians with data useful for decision-making processes in drug selection, and inform about the pharmacokinetics and pharmacodynamics of a drug. It is, therefore, an exceptional approach for gaining new understanding effects in the field of comparative veterinary pharmacology. However, the incorporation of metabolomics into veterinary pharmacology and toxicology is not yet widespread, and this is probably, at least in part, a result of its highly multidisciplinary nature. This article reviews the potential applications of metabolomics in veterinary pharmacology and therapeutics. It integrates key concepts for designing metabolomics studies and analyzing and interpreting metabolomics data, providing solid foundations for applying metabolomics to the study of drugs in all veterinary species.
Collapse
Affiliation(s)
- Sol M Rivera-Velez
- Molecular Determinants Core, Johns Hopkins All Children's Hospital, Saint Petersburg, Florida, USA
| | - Jinna Navas
- Program in Individualized Medicine, Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Washington State University, Pullman, Washington, USA
| | - Nicolas F Villarino
- Program in Individualized Medicine, Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Washington State University, Pullman, Washington, USA
| |
Collapse
|
7
|
Abstract
Early detection of graft injury after kidney transplantation is key to maintaining long-term good graft function. Graft injury could be due to a multitude of factors including ischaemia reperfusion injury, cell or antibody-mediated rejection, progressive interstitial fibrosis and tubular atrophy, infections and toxicity from the immunosuppressive drugs themselves. The current gold standard for assessing renal graft dysfunction is renal biopsy. However, biopsy is usually late when triggered by a change in serum creatinine and of limited utility in diagnosis of early injury when histological changes are equivocal. Therefore, there is a need for timely, objective and non-invasive diagnostic techniques with good early predictive value to determine graft injury and provide precision in titrating immunosuppression. We review potential novel plasma and urine biomarkers that offer sensitive new strategies for early detection and provide major insights into mechanisms of graft injury. This is a rapidly expanding field, but it is likely that a combination of biomarkers will be required to provide adequate sensitivity and specificity for detecting graft injury.
Collapse
|
8
|
Mussap M, Loddo C, Fanni C, Fanos V. Metabolomics in pharmacology - a delve into the novel field of pharmacometabolomics. Expert Rev Clin Pharmacol 2020; 13:115-134. [PMID: 31958027 DOI: 10.1080/17512433.2020.1713750] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/11/2022]
Abstract
Introduction: Pharmacometabolomics is an emerging science pursuing the application of precision medicine. Combining both genetic and environmental factors, the so-called pharmacometabolomic approach guides patient selection and stratification in clinical trials and optimizes personalized drug dosage, improving efficacy and safety.Areas covered: This review illustrates the progressive introduction of pharmacometabolomics as an innovative solution for enhancing the discovery of novel drugs and improving research and development (R&D) productivity of the pharmaceutical industry. An extended analysis on published pharmacometabolomics studies both in animal models and humans includes results obtained in several areas such as hepatology, gastroenterology, nephrology, neuropsychiatry, oncology, drug addiction, embryonic cells, neonatology, and microbiomics.Expert opinion: a tailored, individualized therapy based on the optimization of pharmacokinetics and pharmacodynamics, the improvement of drug efficacy, and the abolition of drug toxicity and adverse drug reactions is a key issue in precision medicine. Genetics alone has become insufficient for deciphring intra- and inter-individual variations in drug-response, since they originate both from genetic and environmental factors, including human microbiota composition. The association between pharmacogenomics and pharmacometabolomics may be considered the new strategy for an in-deep knowledge on changes and alterations in human and microbial metabolic pathways due to the action of a drug.
Collapse
Affiliation(s)
- Michele Mussap
- Laboratory Unit, Department of Surgical Sciences, University of Cagliari, Cagliari, Italy
| | | | - Claudia Fanni
- Division of Pediatrics, Rovigo Hospital, Rovigo, Italy
| | - Vassilios Fanos
- Neonatal Intensive Care Unit, Neonatal Pathology and Neonatal Section, Department of Surgical Sciences, University of Cagliari, Cagliari, Italy
| |
Collapse
|
9
|
Sborchia M, De Prez EG, Antoine MH, Bienfait L, Indra R, Valbuena G, Phillips DH, Nortier JL, Stiborová M, Keun HC, Arlt VM. The impact of p53 on aristolochic acid I-induced nephrotoxicity and DNA damage in vivo and in vitro. Arch Toxicol 2019; 93:3345-3366. [PMID: 31602497 PMCID: PMC6823306 DOI: 10.1007/s00204-019-02578-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/16/2019] [Accepted: 09/17/2019] [Indexed: 12/31/2022]
Abstract
Exposure to aristolochic acid (AA) is associated with human nephropathy and urothelial cancer. The tumour suppressor TP53 is a critical gene in carcinogenesis and frequently mutated in AA-induced urothelial tumours. We investigated the impact of p53 on AAI-induced nephrotoxicity and DNA damage in vivo by treating Trp53(+/+), Trp53(+/-) and Trp53(-/-) mice with 3.5 mg/kg body weight (bw) AAI daily for 2 or 6 days. Renal histopathology showed a gradient of intensity in proximal tubular injury from Trp53(+/+) to Trp53(-/-) mice, especially after 6 days. The observed renal injury was supported by nuclear magnetic resonance (NMR)-based metabonomic measurements, where a consistent Trp53 genotype-dependent trend was observed for urinary metabolites that indicate aminoaciduria (i.e. alanine), lactic aciduria (i.e. lactate) and glycosuria (i.e. glucose). However, Trp53 genotype had no impact on AAI-DNA adduct levels, as measured by 32P-postlabelling, in either target (kidney and bladder) or non-target (liver) tissues, indicating that the underlying mechanisms of p53-related AAI-induced nephrotoxicity cannot be explained by differences in AAI genotoxicity. Performing gas chromatography-mass spectrometry (GC-MS) on kidney tissues showed metabolic pathways affected by AAI treatment, but again Trp53 status did not clearly impact on such metabolic profiles. We also cultured primary mouse embryonic fibroblasts (MEFs) derived from Trp53(+/+), Trp53(+/-) and Trp53(-/-) mice and exposed them to AAI in vitro (50 µM for up to 48 h). We found that Trp53 genotype impacted on the expression of NAD(P)H:quinone oxidoreductase (Nqo1), a key enzyme involved in AAI bioactivation. Nqo1 induction was highest in Trp53(+/+) MEFs and lowest in Trp53(-/-) MEFs; and it correlated with AAI-DNA adduct formation, with lowest adduct levels being observed in AAI-exposed Trp53(-/-) MEFs. Overall, our results clearly demonstrate that p53 status impacts on AAI-induced renal injury, but the underlying mechanism(s) involved remain to be further explored. Despite the impact of p53 on AAI bioactivation and DNA damage in vitro, such effects were not observed in vivo.
Collapse
Affiliation(s)
- Mateja Sborchia
- Department of Analytical, Environmental and Forensic Sciences, MRC-PHE Centre for Environment and Health, King's College London, London, SE1 9NH, UK
| | - Eric G De Prez
- Laboratory of Experimental Nephrology, Department of Experimental Biochemistry, Faculty of Medicine, Université Libre de Bruxelles, 1070, Brussels, Belgium
| | - Marie-Hélène Antoine
- Laboratory of Experimental Nephrology, Department of Experimental Biochemistry, Faculty of Medicine, Université Libre de Bruxelles, 1070, Brussels, Belgium
| | - Lucie Bienfait
- Department of Pathology, Erasme University Hospital, 1070, Brussels, Belgium
| | - Radek Indra
- Department of Biochemistry, Faculty of Science, Charles University Prague, 128 40, Prague, Czech Republic
| | - Gabriel Valbuena
- Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, W12 0NN, UK
| | - David H Phillips
- Department of Analytical, Environmental and Forensic Sciences, MRC-PHE Centre for Environment and Health, King's College London, London, SE1 9NH, UK
| | - Joëlle L Nortier
- Laboratory of Experimental Nephrology, Department of Experimental Biochemistry, Faculty of Medicine, Université Libre de Bruxelles, 1070, Brussels, Belgium
| | - Marie Stiborová
- Department of Biochemistry, Faculty of Science, Charles University Prague, 128 40, Prague, Czech Republic
| | - Hector C Keun
- Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, W12 0NN, UK
| | - Volker M Arlt
- Department of Analytical, Environmental and Forensic Sciences, MRC-PHE Centre for Environment and Health, King's College London, London, SE1 9NH, UK.
| |
Collapse
|
10
|
Klawitter J, Klawitter J, Pennington A, Kirkpatrick B, Roda G, Kotecha NC, Thurman JM, Christians U. Cyclophilin D knockout protects the mouse kidney against cyclosporin A-induced oxidative stress. Am J Physiol Renal Physiol 2019; 317:F683-F694. [PMID: 31188033 DOI: 10.1152/ajprenal.00417.2018] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 01/05/2023] Open
Abstract
Mitochondrial dysfunction and oxidative stress have been implicated in cyclosporin A (CsA)-induced nephrotoxicity. CsA interacts with cyclophilin D (CypD), an essential component of the mitochondrial permeability transition pore and regulator of cell death processes. Controversial reports have suggested that CypD deletion may or may not protect cells against oxidative stress-induced cell death. In the present study, we treated wild-type (WT) mice and mice lacking CypD [peptidylprolyl isomerase F knockout (Ppif-/-) mice] with CsA to test the role and contribution of CypD to the widely described CsA-induced renal toxicity and oxidative stress. Our results showed an increase in the levels of several known uremic toxins as well as the oxidative stress markers PGF2α and 8-isoprostane in CsA-treated WT animals but not in Ppif-/- animals. Similarly, a decline in S-adenosylmethionine and the resulting methylation potential indicative of DNA hypomethylation were observed only in CsA-treated WT mice. This confirms previous reports of the protective effects of CypD deletion on the mouse kidney mediated through a stronger resistance of these animals to oxidative stress and DNA methylation damage. However, a negative effect of CsA on the glycolysis and overall energy metabolism in Ppif-/- mice also indicated that additional, CypD-parallel pathways are involved in the toxic effects of CsA on the kidney. In summary, CsA-mediated induction of oxidative stress is associated with CypD, with CypD deletion providing a protective effect, whereas the reduction of energy production observed upon CsA exposure did not depend on the animals' CypD status.
Collapse
Affiliation(s)
- Jelena Klawitter
- Clinical Research and Development, Department of Anesthesiology, University of Colorado Denver, Aurora, Colorado
- Division of Nephrology and Hypertension, Department of Medicine, University of Colorado Denver, Aurora, Colorado
| | - Jost Klawitter
- Clinical Research and Development, Department of Anesthesiology, University of Colorado Denver, Aurora, Colorado
| | - Alexander Pennington
- Clinical Research and Development, Department of Anesthesiology, University of Colorado Denver, Aurora, Colorado
| | - Bruce Kirkpatrick
- Clinical Research and Development, Department of Anesthesiology, University of Colorado Denver, Aurora, Colorado
| | - Galen Roda
- Clinical Research and Development, Department of Anesthesiology, University of Colorado Denver, Aurora, Colorado
| | - Nidhi C Kotecha
- Clinical Research and Development, Department of Anesthesiology, University of Colorado Denver, Aurora, Colorado
| | - Joshua M Thurman
- Division of Nephrology and Hypertension, Department of Medicine, University of Colorado Denver, Aurora, Colorado
| | - Uwe Christians
- Clinical Research and Development, Department of Anesthesiology, University of Colorado Denver, Aurora, Colorado
| |
Collapse
|
11
|
Barin-Le Guellec C, Largeau B, Bon D, Marquet P, Hauet T. Ischemia/reperfusion-associated tubular cells injury in renal transplantation: Can metabolomics inform about mechanisms and help identify new therapeutic targets? Pharmacol Res 2018; 129:34-43. [PMID: 29309901 DOI: 10.1016/j.phrs.2017.12.032] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 09/29/2017] [Revised: 12/29/2017] [Accepted: 12/29/2017] [Indexed: 12/31/2022]
Abstract
Tubular cells are central targets of ischemia-reperfusion (I/R) injury in kidney transplantation. Inflammation and metabolic disturbances occurring within these cells are deleterious by themselves but also favor secondary events, such as activation of immune response. It is critical to have an in depth understanding of the mechanisms governing tubular cells response to I/R if one wants to define pertinent biomarkers or to elaborate targeted therapeutic interventions. As oxidative damage was shown to be central in the patho-physiological mechanisms, the impact of I/R on proximal tubular cells metabolism has been widely studied, contrary to its effects on expression and activity of membrane transporters of the proximal tubular cells. Yet, temporal modulation of transporters over ischemia and reperfusion periods appears to play a central role, not only in the induction of cells injury but also in graft function recovery. Metabolomics in cell models or diverse biofluids has the potential to provide large pictures of biochemical consequences of I/R. Metabolomic studies conducted in experimental models of I/R or in transplanted patients indeed retrieved metabolites belonging to the pathways known to be particularly affected. Interestingly, they also revealed that metabolic disturbances and transporters activities are in very close mutual interplay. As well as helping to select diagnostic biomarkers, such analyses could also contribute to identify new pharmacological targets and to set up innovative nephroprotective strategies for the future. Even if various therapeutic approaches have been evaluated for a long time to prevent or treat I/R injuries, metabolomics has helped identifying new ones, those related to membrane transporters seeming to be of particular interest. However, considering the very complex and multifactorial effects of I/R in the context of kidney transplantation, all tracks must be followed if one wants to prevent or limit its deleterious consequences.
Collapse
Affiliation(s)
- Chantal Barin-Le Guellec
- INSERM UMR 1248, IPPRITT, Limoges, France; CHU Tours, Laboratory of Biochemistry and Molecular Biology, Tours, France; FHU SUPORT, Limoges, Poitiers, Tours, France.
| | - Bérenger Largeau
- CHU Tours, Laboratory of Biochemistry and Molecular Biology, Tours, France
| | - Delphine Bon
- FHU SUPORT, Limoges, Poitiers, Tours, France; University of Poitiers, Poitiers, France; INSERM UMR 1082, IRTOMIT, Poitiers, France; CHU Poitiers, Laboratory of Biochemistry, Poitiers, France
| | - Pierre Marquet
- INSERM UMR 1248, IPPRITT, Limoges, France; FHU SUPORT, Limoges, Poitiers, Tours, France; University of Limoges, Faculty of Medicine, Limoges, France; CHU Limoges, Department of Pharmacology, Toxicology & Pharmacovigilance, Limoges, France
| | - Thierry Hauet
- FHU SUPORT, Limoges, Poitiers, Tours, France; University of Poitiers, Poitiers, France; INSERM UMR 1082, IRTOMIT, Poitiers, France; CHU Poitiers, Laboratory of Biochemistry, Poitiers, France
| |
Collapse
|
12
|
Martin-Lorenzo M, Gonzalez-Calero L, Ramos-Barron A, Sanchez-Niño MD, Gomez-Alamillo C, García-Segura JM, Ortiz A, Arias M, Vivanco F, Alvarez-Llamas G. Urine metabolomics insight into acute kidney injury point to oxidative stress disruptions in energy generation and H 2S availability. J Mol Med (Berl) 2017; 95:1399-1409. [PMID: 28975359 DOI: 10.1007/s00109-017-1594-5] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 06/02/2017] [Revised: 08/03/2017] [Accepted: 09/12/2017] [Indexed: 11/26/2022]
Abstract
Acute kidney injury (AKI) is one of the main complications in acute care medicine and a risk factor for chronic kidney disease (CKD). AKI incidence has increased; however, its diagnosis has limitations and physiopathological mechanisms are underexplored. We investigated urine samples, aiming to identify major metabolite changes during human AKI evolution. Metabolic signatures found were further explored for a potential link to severity of injury. Twenty-four control subjects and 38 hospitalized patients with AKI were recruited and urine samples were collected at the time of diagnosis, during follow-up and at discharge. Nuclear magnetic resonance (NMR) was used in a first discovery phase for identifying potential metabolic differences. Target metabolites of interest were confirmed by liquid chromatography-mass spectrometry (LC-MS/MS) in an independent group. Underlying metabolic defects were further explored by kidney transcriptomics of murine toxic AKI. Urinary 2-hydroxybutyric acid, pantothenic acid, and hippuric acid were significantly downregulated and urinary N-acetylneuraminic acid, phosphoethanolamine, and serine were upregulated during AKI. Hippuric acid, phosphoethanolamine, and serine showed further downregulation/upregulation depending on the metabolite in acute tubular necrosis (ATN) AKI compared to prerenal AKI. Kidney transcriptomics disclosed decreased expression of cystathionase, cystathionine-β-synthase, and ethanolamine-phosphate cytidylyltransferase, and increased N-acetylneuraminate synthase as the potentially underlying cause of changes in urinary metabolites. A urinary metabolite panel identified AKI patients and provided insight into intrarenal events. A urine fingerprint made up of six metabolites may be related to pathophysiological changes in oxidative stress, energy generation, and H2S availability associated with AKI. KEY MESSAGES The urinary metabolome reflects AKI evolution and severity of injury. Kidney transcriptomics revealed enzymatic expression changes. Enzymatic expression changes may be the potentially underlying cause of changes in urine metabolites. Identified metabolite changes link oxidative stress, energy generation, and H2S availability to AKI.
Collapse
Affiliation(s)
- Marta Martin-Lorenzo
- Department of Immunology, IIS-Fundacion Jimenez Diaz-UAM, REDinREN, Madrid, Spain
| | | | - Angeles Ramos-Barron
- Nephrology Department, Hospital Valdecilla, Universidad de Cantabria, Instituto de Investigación Marqués de Valdecilla, IDIVAL, Santander, Cantabria, Spain
| | - Maria D Sanchez-Niño
- Department of Nephrology/IRSIN, IIS-Fundación Jiménez Díaz-UAM, REDinREN, Madrid, Spain
| | - Carlos Gomez-Alamillo
- Nephrology Department, Hospital Valdecilla, Universidad de Cantabria, Instituto de Investigación Marqués de Valdecilla, IDIVAL, Santander, Cantabria, Spain
| | - Juan Manuel García-Segura
- CAI-RMN, Universidad Complutense, Madrid, Spain
- Department of Biochemistry and Molecular Biology I, Universidad Complutense, Madrid, Spain
| | - Alberto Ortiz
- Department of Nephrology/IRSIN, IIS-Fundación Jiménez Díaz-UAM, REDinREN, Madrid, Spain
| | - Manuel Arias
- Nephrology Department, Hospital Valdecilla, Universidad de Cantabria, Instituto de Investigación Marqués de Valdecilla, IDIVAL, Santander, Cantabria, Spain
| | - Fernando Vivanco
- Department of Immunology, IIS-Fundacion Jimenez Diaz-UAM, REDinREN, Madrid, Spain
- Department of Biochemistry and Molecular Biology I, Universidad Complutense, Madrid, Spain
| | | |
Collapse
|
13
|
Duquesne M, Declèves AE, De Prez E, Nortier J, Colet J. Interest of metabonomic approach in environmental nephrotoxicants: Application to aristolochic acid exposure. Food Chem Toxicol 2017; 108:19-29. [DOI: 10.1016/j.fct.2017.07.015] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 04/13/2017] [Revised: 06/23/2017] [Accepted: 07/06/2017] [Indexed: 01/10/2023]
|
14
|
DCoH: A novel biomarker for diagnosing acute kidney injury. Med Hypotheses 2016; 95:27-30. [PMID: 27692160 DOI: 10.1016/j.mehy.2016.08.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 05/07/2016] [Accepted: 08/14/2016] [Indexed: 11/21/2022]
Abstract
Initial diagnosis of acute kidney injury (AKI) is usually based on measuring serum-creatinine and blood urea nitrogen levels; however such measurements are still poor in identifying renal injuries at initial stages. These standard matrices are not enough to monitor the outcome and progression of AKI. The prognosis prevents proper treatment and timely delay in providing putative therapeutic agents. The cost effective therapies to get delayed, patient health gets compromised and ultimately requires renal transplant due to end-stage renal disease, which is another major problematic factor due to shortage kidney donors. To establish effective therapies for AKI the need will be facilitated by developing and identifying reliable, sensitive biomarkers which can be detected early during all stages of AKI, even during preclinical and clinical studies. Although reaching to human clinical trials takes years of thorough evaluations, preliminary studies should be carried out effectively by: (a) Employing cell culture analysis, (b) use of AKI animal models, studying various gene regulated networks, and biomarkers, and (c) patient serum sampling and testing. As elevated phenylalanine are indicative of AKI onset within 4h, its levels is controlled, 4a-Hydroxy-tetrahydrobiopterin dehydratase/dimerization cofactor of HNF-1 (DCoH). There is a possibility of targeting DCoH to the current bedside list of biomarkers involved in AKI onset.
Collapse
|
15
|
Xing W, Gu L, Zhang X, Xu J, Lu H. A metabolic profiling analysis of the nephrotoxicity of acyclovir in rats using ultra performance liquid chromatography/mass spectrometry. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2016; 46:234-240. [PMID: 27497730 DOI: 10.1016/j.etap.2016.07.018] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Academic Contribution Register] [Received: 03/17/2016] [Revised: 06/14/2016] [Accepted: 07/27/2016] [Indexed: 06/06/2023]
Abstract
Acyclovir (ACV) exposure is a common cause of acute kidney injury (AKI). The toxicity mechanism of ACV has always been a matter of debate. The present study investigated into the time-effect relationship and dose-effect relationship of ACV-induced nephrotoxicity in rats using metabonomics. Twenty-four rats were randomly divided into four groups: a 0.9% NaCl solution group, and 100, 300, and 600mg/kg ACV-treated groups; the ACV or vehicle solution was administered with a single intravenous injection. Urine was collected at different time periods (12h before administration, and 0-6h, 7-12h, and 13-24h after administration). Routine urinalysis was conducted by a urine automatic analyzer. Renal markers, including urine urea nitrogen, urine creatinine, and urinary N-acetyl-β-d-glucosaminidase (NAG) activity, were determined using established protocols. Urinary metabolites were evaluated using ultra performance liquid chromatography/mass spectrometry (UPLC/MS). In the ACV-treated rats, increased levels of protein (PRO), occult blood (BLD), white blood cell (WBC), and NAG activity in urine were observed, while the urine creatinine and urea nitrogen levels showed a decrease compared with the control. Moreover, urine metabolites significantly changed after the treatment with ACV, and all the effects induced by ACV were dose-time dependent. Finally, 4 metabolites (guanine, 4-guanidinobutyric acid, creatinine, and urea) were identified, which can be used for further research on the mechanism of ACV-induced nephrotoxicity.
Collapse
Affiliation(s)
- Wenmin Xing
- Zhejiang Provincial Key Lab of Geriatrics and Geriatrics Institute of Zhejiang Province, Zhejiang Hospital, Hangzhou 310013, China
| | - Lili Gu
- School of Pharmacology, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Xinyue Zhang
- Institute of Materia Medica, Zhejiang Academy of Medical Sciences, Hangzhou 310013, China
| | - Jiadong Xu
- School of Pharmacology, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Hong Lu
- School of Pharmacology, Zhejiang Chinese Medical University, Hangzhou 310053, China.
| |
Collapse
|
16
|
Leberkühne LJ, Ebtehaj S, Dimova LG, Dikkers A, Dullaart RPF, Bakker SJL, Tietge UJF. The predictive value of the antioxidative function of HDL for cardiovascular disease and graft failure in renal transplant recipients. Atherosclerosis 2016; 249:181-5. [PMID: 27107804 DOI: 10.1016/j.atherosclerosis.2016.04.008] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 05/22/2015] [Revised: 04/05/2016] [Accepted: 04/06/2016] [Indexed: 11/19/2022]
Abstract
BACKGROUND Protection of low-density lipoproteins (LDL) against oxidative modification is a key anti-atherosclerotic property of high-density lipoproteins (HDL). This study evaluated the predictive value of the HDL antioxidative function for cardiovascular mortality, all-cause mortality and chronic graft failure in renal transplant recipients (RTR). METHODS The capacity of HDL to inhibit native LDL oxidation was determined in vitro in a prospective cohort of renal transplant recipients (RTR, n = 495, median follow-up 7.0 years). RESULTS The HDL antioxidative functionality was significantly higher in patients experiencing graft failure (57.4 ± 9.7%) than in those without (54.2 ± 11.3%; P = 0.039), while there were no differences for cardiovascular and all-cause mortality. Specifically glomerular filtration rate (P = 0.001) and C-reactive protein levels (P = 0.006) associated independently with antioxidative functionality in multivariate linear regression analyses. Cox regression analysis demonstrated a significant relationship between antioxidative functionality of HDL and graft failure in age-adjusted analyses, but significance was lost following adjustment for baseline kidney function and inflammatory load. No significant association was found between HDL antioxidative functionality and cardiovascular and all-cause mortality. CONCLUSION This study demonstrates that the antioxidative function of HDL (i) does not predict cardiovascular or all-cause mortality in RTR, but (ii) conceivably contributes to the development of graft failure, however, not independent of baseline kidney function and inflammatory load.
Collapse
Affiliation(s)
- Lynn J Leberkühne
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713GZ Groningen, The Netherlands
| | - Sanam Ebtehaj
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713GZ Groningen, The Netherlands
| | - Lidiya G Dimova
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713GZ Groningen, The Netherlands
| | - Arne Dikkers
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713GZ Groningen, The Netherlands
| | - Robin P F Dullaart
- Department of Endocrinology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713GZ Groningen, The Netherlands
| | - Stephan J L Bakker
- Department of Internal Medicine, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713GZ Groningen, The Netherlands
| | - Uwe J F Tietge
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713GZ Groningen, The Netherlands.
| |
Collapse
|
17
|
Bonneau E, Tétreault N, Robitaille R, Boucher A, De Guire V. Metabolomics: Perspectives on potential biomarkers in organ transplantation and immunosuppressant toxicity. Clin Biochem 2016; 49:377-84. [DOI: 10.1016/j.clinbiochem.2016.01.006] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 09/11/2015] [Revised: 12/23/2015] [Accepted: 01/07/2016] [Indexed: 12/27/2022]
|
18
|
Barrios C, Spector TD, Menni C. Blood, urine and faecal metabolite profiles in the study of adult renal disease. Arch Biochem Biophys 2015; 589:81-92. [PMID: 26476344 DOI: 10.1016/j.abb.2015.10.006] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 04/15/2015] [Revised: 10/08/2015] [Accepted: 10/09/2015] [Indexed: 01/04/2023]
Abstract
Chronic kidney disease (CKD) is a major public health burden and to date traditional biomarkers of renal function (such as serum creatinine and cystatin C) are unable to identify at-risk individuals before the disease process is well under way. To help preventive strategies and maximize the potential for effective interventions, it is important to characterise the molecular changes that take place in the development of renal damage. Metabolomics is a promising tool to identify markers of renal disease since the kidneys are involved in the handling of major biochemical classes of metabolites. These metabolite levels capture a snap-shot of the metabolic profile of the individual, allowing for the potential identification of early biomarkers, and the monitoring of real-time kidney function. In this review, we describe the current status of the identification of blood/urine/faecal metabolic biomarkers in different entities of kidney diseases including: acute kidney injury, chronic kidney disease, renal transplant, diabetic nephropathy and other disorders.
Collapse
Affiliation(s)
- Clara Barrios
- Department of Twin Research and Genetic Epidemiology, King's College London, London, UK; Department of Nephrology, Hospital del Mar. Institut Mar d'Investigacions Mediques, Barcelona, Spain
| | - Tim D Spector
- Department of Twin Research and Genetic Epidemiology, King's College London, London, UK
| | - Cristina Menni
- Department of Twin Research and Genetic Epidemiology, King's College London, London, UK.
| |
Collapse
|
19
|
Yan SK, Liu RH, Jin HZ, Liu XR, Ye J, Shan L, Zhang WD. "Omics" in pharmaceutical research: overview, applications, challenges, and future perspectives. Chin J Nat Med 2015; 13:3-21. [PMID: 25660284 DOI: 10.1016/s1875-5364(15)60002-4] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 10/13/2014] [Indexed: 12/18/2022]
Abstract
In the post-genomic era, biological studies are characterized by the rapid development and wide application of a series of "omics" technologies, including genomics, proteomics, metabolomics, transcriptomics, lipidomics, cytomics, metallomics, ionomics, interactomics, and phenomics. These "omics" are often based on global analyses of biological samples using high through-put analytical approaches and bioinformatics and may provide new insights into biological phenomena. In this paper, the development and advances in these omics made in the past decades are reviewed, especially genomics, transcriptomics, proteomics and metabolomics; the applications of omics technologies in pharmaceutical research are then summarized in the fields of drug target discovery, toxicity evaluation, personalized medicine, and traditional Chinese medicine; and finally, the limitations of omics are discussed, along with the future challenges associated with the multi-omics data processing, dynamics omics analysis, and analytical approaches, as well as amenable solutions and future prospects.
Collapse
Affiliation(s)
- Shi-Kai Yan
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Run-Hui Liu
- School of Pharmacy, Second Military Medical University, Shanghai 200433, China
| | - Hui-Zi Jin
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Xin-Ru Liu
- School of Pharmacy, Second Military Medical University, Shanghai 200433, China
| | - Ji Ye
- School of Pharmacy, Second Military Medical University, Shanghai 200433, China
| | - Lei Shan
- School of Pharmacy, Second Military Medical University, Shanghai 200433, China
| | - Wei-Dong Zhang
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China; School of Pharmacy, Second Military Medical University, Shanghai 200433, China; Shanghai Institute of Pharmaceutical Industry, Shanghai 200040, China.
| |
Collapse
|
20
|
Kienana M, Lydie ND, Jean-Michel H, Binta D, Matthias B, Patrick E, Hélène B, Chantal LG. Elucidating time-dependent changes in the urinary metabolome of renal transplant patients by a combined (1)H NMR and GC-MS approach. MOLECULAR BIOSYSTEMS 2015; 11:2493-510. [PMID: 26161811 DOI: 10.1039/c5mb00108k] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Academic Contribution Register] [Indexed: 12/12/2022]
Abstract
Urine metabolomic profiling can identify biochemical alterations resulting from various injuries affecting the graft outcome after renal transplantation. Here, we aimed to describe in depth the metabolite content of urines of renal transplant patients and to link it with the major injury factors acting at critical stages following transplantation. Morning urine samples were prospectively collected from 38 kidney transplant patients at 7 days (D7), 3 months (M3) and 12 months (M12) after transplantation. Twenty-five patients were treated with tacrolimus (Tac) and thirteen patients with cyclosporine (CsA). (1)H-NMR (proton nuclear magnetic resonance) and gas chromatography-mass spectrometry (GC-MS) were used to examine the overall metabolomic signature of each sample. Multivariate analysis was performed to study the changes in the metabolic profile over time and their dependency on the type of calcineurin inhibitor (CNI) administered to patients. Biological pathways affected by transplantation were identified using a metabolomics pathway analysis (MetPA) web-tool. The metabolic profile of urine samples clearly varied with time. Markers of medullary injury, tubule cell oxidative metabolism and impaired tubular reabsorption or secretion were present at D7. Differences in metabolic profiles became less marked as time passed on, urine content being quite similar at M3 and M12. The metabolite profile tended to differ between patients receiving Tac and those receiving CsA but no clear discriminating profiles can be found. The combination of (1)H-NMR and GC-MS for the analysis of urine metabolomic profiles is a very useful method to study patho-physiological alterations in kidney transplant patients over time.
Collapse
Affiliation(s)
- Muhrez Kienana
- Cellules dendritiques, immuno-intervention et greffes, EA4245, Université François Rabelais, Faculté de médecine, bâtiment Vialle, 10 Boulevard Tonnellé, 37032 Tours Cedex 1, France.
| | | | | | | | | | | | | | | |
Collapse
|
21
|
Klepacki J, Klawitter J, Klawitter J, Thurman JM, Christians U. A high-performance liquid chromatography-tandem mass spectrometry-based targeted metabolomics kidney dysfunction marker panel in human urine. Clin Chim Acta 2015; 446:43-53. [PMID: 25871999 DOI: 10.1016/j.cca.2015.04.005] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 02/16/2015] [Revised: 03/28/2015] [Accepted: 04/03/2015] [Indexed: 02/04/2023]
Abstract
BACKGROUND Previous studies have examined and documented fluctuations in urine metabolites in response to disease processes and drug toxicity affecting glomerular filtration, tubule cell metabolism, reabsorption, oxidative stress, purine degradation, active secretion and kidney amino acylase activity representative of diminished renal function. However, a high-throughput assay that incorporates metabolites that are surrogate markers for such changes into a kidney dysfunction panel has yet to be described. METHODS A high-performance liquid chromatography-tandem mass spectrometry (HPLC-MS/MS) assay for the quantification of ten metabolites associated with the Krebs cycle, purine degradation, and oxidative stress in human urine was developed and validated. Normal values were assessed in healthy adult (n=120) and pediatric (n=36) individuals. In addition, 9 pediatric renal transplant recipients patients were evaluated before and after initial dosing of the immunosuppressant tacrolimus in a proof-of-concept study. RESULTS The assay met all predefined acceptance criteria. The lower limit of quantification ranged from 0.1 to 1000 μmol/l. Inter-day trueness and imprecisions ranged from 91.4-112.9% and 1.5-12.4%, respectively. The total assay run time was 5.5 minutes. Concentrations of glucose, sorbitol, and trimethylamine oxide (TMAO) were elevated in pediatric renal transplant patients (n=9) prior to transplantation as well as before and immediately after initial dosing of tacrolimus. One month post-transplant urine metabolite patterns matched those of healthy children (n=36). CONCLUSIONS The LC-MS/MS assay will provide the basis for further large-scale clinical studies to explore these analytes as molecular markers for the patients with renal insufficiency.
Collapse
Affiliation(s)
- Jacek Klepacki
- iC42 Clinical Research and Development, Department of Anesthesiology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
| | - Jost Klawitter
- iC42 Clinical Research and Development, Department of Anesthesiology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Jelena Klawitter
- iC42 Clinical Research and Development, Department of Anesthesiology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA; Department of Renal Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Joshua M Thurman
- Department of Renal Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Uwe Christians
- iC42 Clinical Research and Development, Department of Anesthesiology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| |
Collapse
|
22
|
Xu JD, Xing WM, Yuan TJ, Chen J, Lu H. Metabolic changes in the urine of andrographolide sodium bisulfite-treated rats. Hum Exp Toxicol 2015; 35:162-9. [DOI: 10.1177/0960327115579429] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/17/2022]
Abstract
In recent years, andrographolide sodium bisulfite (ASB) has been reported to cause acute renal failure frequently in clinical practice. We hypothesized that changes in metabolic profile could have occurred after administration of ASB. To investigate the metabolic changes caused by ASB-induced nephrotoxicity, metabonomics method was utilized to depict the urine metabolic characteristics and find the specific urine biomarkers associated with ASB-induced nephrotoxicity. Sprague-Dawley rats were randomly assigned into three experimental groups. They received a single daily injection of vehicle (0.9% sodium chloride solution) or ASB at a dose of 100 or 600 mg kg−1 day−1 for 7 days. Twelve-hour urine was collected after the last administration. The routine urinalysis was measured by a urine automatic analyzer while urinary metabolites were evaluated using gas chromatography/mass spectrometry. The acquired data were processed by multivariate principal component analysis (PCA), partial least squares discriminant analysis (PLS-DA), and orthogonal PLS-DA. After 7-day administration of ASB, the positive urine samples in protein, occult blood, and ketones were increased, presenting dose dependence. The PCA and PLS-DA models were capable of distinguishing the difference between ASB-treated group and control. Biomarkers such as 1,5-anhydroglucitol, d-erythro-sphingosine, and 2-ketoadipate were identified as the most influential factors in ASB-induced nephrotoxicity.
Collapse
Affiliation(s)
- JD Xu
- School of Pharmacology, Zhejiang Chinese Medical University, Hangzhou, China
| | - WM Xing
- School of Pharmacology, Zhejiang Chinese Medical University, Hangzhou, China
| | - TJ Yuan
- School of Pharmacology, Zhejiang Chinese Medical University, Hangzhou, China
| | - J Chen
- School of Pharmacology, Zhejiang Chinese Medical University, Hangzhou, China
| | - H Lu
- School of Pharmacology, Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
23
|
Fernando M, Peake PW, Endre ZH. Biomarkers of calcineurin inhibitor nephrotoxicity in transplantation. Biomark Med 2014; 8:1247-62. [DOI: 10.2217/bmm.14.86] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/23/2022] Open
Abstract
Over 35 years of use has demonstrated the revolutionary therapeutic benefits of calcineurin inhibitors (CNI) in not only preventing transplant rejection, but also the renal and nonrenal toxicity of CNI. Acute reversible and insidious irreversible forms of CNI nephrotoxicity have been identified, with ischemia from an imbalance between vasoconstrictors and vasodilators playing an important role. The ongoing search to define toxicity pathways has been enriched by ‘Omics’ studies. Changes in proteins including those involved in activation of pro-inflammatory responses, oxidative stress, ER stress and the unfolded protein response have been identified, and these may serve as biomarkers of toxicity. However, the current standard of CNI toxicity, histology, lacks specificity, which creates challenges for biomarker validation. This review focuses on progress in nephrotoxic pathway identification of CNI and biomarker validation.
Collapse
Affiliation(s)
- Mangalee Fernando
- Department of Nephrology, Prince of Wales Hospital, Barker St., Randwick, Sydney, NSW, Australia
- Prince of Wales Clinical School, University of New South Wales, Sydney, NSW, Australia
| | - Philip W Peake
- Department of Nephrology, Prince of Wales Hospital, Barker St., Randwick, Sydney, NSW, Australia
- Prince of Wales Clinical School, University of New South Wales, Sydney, NSW, Australia
| | - Zoltan H Endre
- Department of Nephrology, Prince of Wales Hospital, Barker St., Randwick, Sydney, NSW, Australia
- Prince of Wales Clinical School, University of New South Wales, Sydney, NSW, Australia
| |
Collapse
|
24
|
Abstract
Nephrotoxicity or renal toxicity can be a result of hemodynamic changes, direct injury to cells and tissue, inflammatory tissue injury, and/or obstruction of renal excretion. Nephrotoxicity is frequently induced by a wide spectrum of therapeutic drugs and environ mental pollutants. Knowledge of the complex molecular and pathophysiologic mechanisms leading to nephrotoxicity remains limited, in part, by research that historically focused on single or relatively few risk markers. As such, current kidney injury biomarkers are inadequate in terms of sensitivity and specificity. In contrast, metabolomics enables screening of a vast array of metabolites simultaneously using NMR and MS to assess their role in nephrotoxicity development and progression. A more comprehensive understanding of these biochemical pathways would also provide valuable insight to disease mechanisms critical for drug development and treatment.
Collapse
|
25
|
Assessing the Metabolic Effects of Calcineurin Inhibitors in Renal Transplant Recipients by Urine Metabolic Profiling. Transplantation 2014; 98:195-201. [DOI: 10.1097/tp.0000000000000039] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/14/2022]
|
26
|
Klawitter J, Klawitter J, Schmitz V, Shokati T, Epshtein E, Thurman JM, Christians U. Mycophenolate mofetil enhances the negative effects of sirolimus and tacrolimus on rat kidney cell metabolism. PLoS One 2014; 9:e86202. [PMID: 24497939 PMCID: PMC3907404 DOI: 10.1371/journal.pone.0086202] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 04/23/2013] [Accepted: 12/08/2013] [Indexed: 12/12/2022] Open
Abstract
Background and Purpose Mycophenolate mofetil (MMF) per se is not known to have negative effects on the kidney. MMF alone or in combination with sirolimus, can be the basis of calcineurin inhibitor (CNI)-free, kidney sparing drug protocols. However, long-term outcomes in patients on MMF/SRL seem to be inferior to those treated with regimens that include the CNI tacrolimus (TAC) due to an increased risk of allo-immune reactions. Interestingly, potential enhancement of the negative effects of SRL and TAC on the kidney by MMF has never been considered. Experimental Approach It was our aim to study the effects of TAC, SRL and MMF alone and evaluate their interactions when combined on the rat kidney. For this purpose we used a comprehensive molecular marker approach including measurements of urinary 8-isoprostane concentrations (oxidative stress marker) and changes of urinary metabolite patterns (1H-NMR spectroscopy) and comparing these markers to renal function (glomerular filtration rate (GFR)) and morphologic alterations (histology). Key Results While MMF alone did not impact GFR, its interaction with SRL and TAC led to a significant decrease of rats’ renal function. The decline went in parallel with a significant increase in urinary isoprostane concentrations and an enhancement of negative effects on urinary metabolite patterns. Conclusions In broad summary, the present study showed that MMF may enhance the negative effects of TAC on kidney function and may even display nephrotoxic properties when combined with SRL.
Collapse
Affiliation(s)
- Jelena Klawitter
- Department of Anesthesiology, University of Colorado, Aurora, Colorado, United States of America
- Division of Renal Diseases and Hypertension, University of Colorado, Aurora, Colorado, United States of America
- * E-mail:
| | - Jost Klawitter
- Department of Anesthesiology, University of Colorado, Aurora, Colorado, United States of America
| | - Volker Schmitz
- Department of Anesthesiology, University of Colorado, Aurora, Colorado, United States of America
- Department of General-, Visceral- and Transplantation Surgery, Charité, Campus Virchow, Berlin, Germany
| | - Touraj Shokati
- Department of Anesthesiology, University of Colorado, Aurora, Colorado, United States of America
| | - Ekaterina Epshtein
- Department of Anesthesiology, University of Colorado, Aurora, Colorado, United States of America
| | - Joshua M. Thurman
- Division of Renal Diseases and Hypertension, University of Colorado, Aurora, Colorado, United States of America
| | - Uwe Christians
- Department of Anesthesiology, University of Colorado, Aurora, Colorado, United States of America
| |
Collapse
|
27
|
Atzler D, Schwedhelm E, Zeller T. Integrated genomics and metabolomics in nephrology. Nephrol Dial Transplant 2013; 29:1467-74. [DOI: 10.1093/ndt/gft492] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 01/14/2023] Open
|
28
|
|
29
|
Liu BC, Song X, Lu XY, Fang CZ, Wei SP, Alli AA, Eaton DC, Shen BZ, Li XQ, Ma HP. Lovastatin attenuates effects of cyclosporine A on tight junctions and apoptosis in cultured cortical collecting duct principal cells. Am J Physiol Renal Physiol 2013; 305:F304-13. [PMID: 23720343 DOI: 10.1152/ajprenal.00074.2013] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 01/19/2023] Open
Abstract
We used mouse cortical collecting duct principal cells (mpkCCDc14 cell line) as a model to determine whether statins reduce the harmful effects of cyclosporine A (CsA) on the distal nephron. The data showed that treatment of cells with CsA increased transepithelial resistance and that the effect of CsA was abolished by lovastatin. Scanning ion conductance microscopy showed that CsA significantly increased the height of cellular protrusions near tight junctions. In contrast, lovastatin eliminated the protrusions and even caused a modest depression between cells. Western blot analysis and confocal microscopy showed that lovastatin also abolished CsA-induced elevation of both zonula occludens-1 and cholesterol in tight junctions. In contrast, a high concentration of CsA induced apoptosis, which was also attenuated by lovastatin, elevated intracellular ROS via activation of NADPH oxidase, and increased the expression of p47phox. Sustained treatment of cells with lovastatin also induced significant apoptosis, which was attenuated by CsA, but did not elevate intracellular ROS. These results indicate that both CsA and lovastatin are harmful to principal cells of the distal tubule, but via ROS-dependent and ROS-independent apoptotic pathways, respectively, and that they counteract probably via mobilization of cellular cholesterol levels.
Collapse
Affiliation(s)
- Bing-Chen Liu
- Department of Cardiology, The Fourth Affiliated Hospital, Harbin Medical University, Harbin, Heilongjiang, People's Republic of China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Bohra R, Klepacki J, Klawitter J, Klawitter J, Thurman J, Christians U. Proteomics and metabolomics in renal transplantation-quo vadis? Transpl Int 2013; 26:225-41. [PMID: 23350848 PMCID: PMC4006577 DOI: 10.1111/tri.12003] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 04/15/2012] [Revised: 05/07/2012] [Accepted: 10/07/2012] [Indexed: 12/13/2022]
Abstract
The improvement of long-term transplant organ and patient survival remains a critical challenge following kidney transplantation. Proteomics and biochemical profiling (metabolomics) may allow for the detection of early changes in cell signal transduction regulation and biochemistry with high sensitivity and specificity. Hence, these analytical strategies hold the promise to detect and monitor disease processes and drug effects before histopathological and pathophysiological changes occur. In addition, they will identify enriched populations and enable individualized drug therapy. However, proteomics and metabolomics have not yet lived up to such high expectations. Renal transplant patients are highly complex, making it difficult to establish cause-effect relationships between surrogate markers and disease processes. Appropriate study design, adequate sample handling, storage and processing, quality and reproducibility of bioanalytical multi-analyte assays, data analysis and interpretation, mechanistic verification, and clinical qualification (=establishment of sensitivity and specificity in adequately powered prospective clinical trials) are important factors for the success of molecular marker discovery and development in renal transplantation. However, a newly developed and appropriately qualified molecular marker can only be successful if it is realistic that it can be implemented in a clinical setting. The development of combinatorial markers with supporting software tools is an attractive goal.
Collapse
Affiliation(s)
- Rahul Bohra
- iC42 Clinical Research & Development, Department of Anesthesiology, University of Colorado Denver, Aurora, Colorado, USA
| | - Jacek Klepacki
- iC42 Clinical Research & Development, Department of Anesthesiology, University of Colorado Denver, Aurora, Colorado, USA
| | - Jelena Klawitter
- iC42 Clinical Research & Development, Department of Anesthesiology, University of Colorado Denver, Aurora, Colorado, USA
- Renal Medicine, University of Colorado Denver, Aurora, USA
| | - Jost Klawitter
- iC42 Clinical Research & Development, Department of Anesthesiology, University of Colorado Denver, Aurora, Colorado, USA
| | - Joshua Thurman
- Renal Medicine, University of Colorado Denver, Aurora, USA
| | - Uwe Christians
- iC42 Clinical Research & Development, Department of Anesthesiology, University of Colorado Denver, Aurora, Colorado, USA
| |
Collapse
|
31
|
Everolimus and sirolimus in combination with cyclosporine have different effects on renal metabolism in the rat. PLoS One 2012; 7:e48063. [PMID: 23118926 PMCID: PMC3485290 DOI: 10.1371/journal.pone.0048063] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 04/02/2012] [Accepted: 09/19/2012] [Indexed: 01/05/2023] Open
Abstract
Enhancement of calcineurin inhibitor nephrotoxicity by sirolimus (SRL) is limiting the clinical use of this drug combination. We compared the dose-dependent effects of the structurally related everolimus (EVL) and sirolimus (SRL) alone, and in combination with cyclosporine (CsA), on the rat kidney. Lewis rats were treated by oral gavage for 28 days using a checkerboard dosing format (0, 3.0, 6.0 and 10.0 CsA and 0, 0.5, 1.5 and 3.0 mg/kg/day SRL or EVL, n = 4/dose combination). After 28 days, oxidative stress, energy charge, kidney histologies, glomerular filtration rates, and concentrations of the immunosuppressants were measured along with 1H-magnetic resonance spectroscopy (MRS) and gas chromatography- mass spectrometry profiles of cellular metabolites in urine. The combination of CsA with SRL led to higher urinary glucose concentrations and decreased levels of urinary Krebs cycle metabolites when compared to controls, suggesting that CsA+SRL negatively impacted proximal tubule metabolism. Unsupervised principal component analysis of MRS spectra distinguished unique urine metabolite patterns of rats treated with CsA+SRL from those treated with CsA+EVL and the controls. SRL, but not EVL blood concentrations were inversely correlated with urine Krebs cycle metabolite concentrations. Interestingly, the higher the EVL concentration, the closer urine metabolite patterns resembled those of controls, while in contrast, the combination of the highest doses of CsA+SRL showed the most significant differences in metabolite patterns. Surprisingly in this rat model, EVL and SRL in combination with CsA had different effects on kidney biochemistry, suggesting that further exploration of EVL in combination with low dose calcineurin inhibitors may be of potential benefit.
Collapse
|
32
|
Corona G, Rizzolio F, Giordano A, Toffoli G. Pharmaco-metabolomics: an emerging "omics" tool for the personalization of anticancer treatments and identification of new valuable therapeutic targets. J Cell Physiol 2012; 227:2827-31. [PMID: 22105661 DOI: 10.1002/jcp.24003] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 01/17/2023]
Abstract
In the post-genomics era, metabolomics represents a new "omics" approach that in the last decade has received increased attention in the field of oncology. Metabolomics is based on the holistic study of the metabolic profile that characterizes a specific phenotype in a biological system. The metabolic profile provides a readout of the metabolic state of an individual that cannot be obtained directly from DNA genotyping, gene expression, or proteomic profiling analyses. The translational value of metabonomics in the oncology field has been demonstrated by the identification of diagnostic and prognostic biomarkers. The so-called pharmaco-metabolomic approach that is currently emerging aims to identify the individual metabolomic characteristics able to predict drug effectiveness and/or toxicity. This review presents the potential role of pharmaco-metabolomics in the future of anticancer pharmacology to achieve customized anticancer treatments and new, targeted therapeutic approaches.
Collapse
Affiliation(s)
- Giuseppe Corona
- Experimental and Clinical Pharmacology Division, IRCCS, National Cancer Institute, Aviano, Pordenone, Italy
| | | | | | | |
Collapse
|
33
|
Abstract
Acute kidney injury (AKI) is a common problem in both the inpatient and outpatient setting and often results from drug toxicities. Traditional methods of identifying AKI, through measurement of blood urea nitrogen and serum creatinine, are problematic in that they are slow to detect decreases in glomerular filtration rate (GFR) and are influenced by a variety of factors that are not related to GFR changes. The problems inherent in a creatinine-based diagnosis of AKI have impeded the development of proper therapeutics in AKI and posed problems in evaluating nephrotoxicity of drugs and other chemical exposures. In recent years, a number of new biomarkers of AKI with more favorable test characteristics than creatinine have been identified and studied in a variety of experimental and clinical settings. This review will consider the most well-established biomarkers and appraise the literature, with particular attention given to the use of biomarkers in identifying toxin-mediated AKI.
Collapse
|
34
|
Metabolomics of oxidative stress in recent studies of endogenous and exogenously administered intermediate metabolites. Int J Mol Sci 2011; 12:6469-501. [PMID: 22072900 PMCID: PMC3210991 DOI: 10.3390/ijms12106469] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 08/18/2011] [Revised: 09/13/2011] [Accepted: 09/21/2011] [Indexed: 11/19/2022] Open
Abstract
Aerobic metabolism occurs in a background of oxygen radicals and reactive oxygen species (ROS) that originate from the incomplete reduction of molecular oxygen in electron transfer reactions. The essential role of aerobic metabolism, the generation and consumption of ATP and other high energy phosphates, sustains a balance of approximately 3000 essential human metabolites that serve not only as nutrients, but also as antioxidants, neurotransmitters, osmolytes, and participants in ligand-based and other cellular signaling. In hypoxia, ischemia, and oxidative stress, where pathological circumstances cause oxygen radicals to form at a rate greater than is possible for their consumption, changes in the composition of metabolite ensembles, or metabolomes, can be associated with physiological changes. Metabolomics and metabonomics are a scientific disciplines that focuse on quantifying dynamic metabolome responses, using multivariate analytical approaches derived from methods within genomics, a discipline that consolidated innovative analysis techniques for situations where the number of biomarkers (metabolites in our case) greatly exceeds the number of subjects. This review focuses on the behavior of cytosolic, mitochondrial, and redox metabolites in ameliorating or exacerbating oxidative stress. After reviewing work regarding a small number of metabolites—pyruvate, ethyl pyruvate, and fructose-1,6-bisphosphate—whose exogenous administration was found to ameliorate oxidative stress, a subsequent section reviews basic multivariate statistical methods common in metabolomics research, and their application in human and preclinical studies emphasizing oxidative stress. Particular attention is paid to new NMR spectroscopy methods in metabolomics and metabonomics. Because complex relationships connect oxidative stress to so many physiological processes, studies from different disciplines were reviewed. All, however, shared the common goal of ultimately developing “omics”-based, diagnostic tests to help influence therapies.
Collapse
|
35
|
Gieser G, Harigaya H, Colangelo PM, Burckart G. Biomarkers in solid organ transplantation. Clin Pharmacol Ther 2011; 90:217-20. [PMID: 21772300 DOI: 10.1038/clpt.2011.75] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/09/2022]
Abstract
This workshop was organized by the US Food and Drug Administration (FDA) Office of Clinical Pharmacology Review Team supporting the Division of Special Pathogen and Transplant Products in the Center for Drug Evaluation and Research. The main goal of the workshop was to enhance the knowledge base regarding biomarkers in solid-organ transplantation via presentation and discussion of scientific findings.
Collapse
Affiliation(s)
- G Gieser
- Office of Clinical Pharmacology, Center for Drug Evaluation, US Food and Drug Administration, Silver Spring, Maryland, USA.
| | | | | | | |
Collapse
|
36
|
Christians U, Klawitter J, Klawitter J, Brunner N, Schmitz V. Biomarkers of immunosuppressant organ toxicity after transplantation: status, concepts and misconceptions. Expert Opin Drug Metab Toxicol 2011; 7:175-200. [PMID: 21241200 DOI: 10.1517/17425255.2011.544249] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/16/2022]
Abstract
INTRODUCTION A major challenge in transplantation is improving long-term organ transplant and patient survival. Immunosuppressants protect the transplant organ from alloimmune reactions, but sometimes also exhibit limiting side effects. The key to improving long-term outcome following transplantation is the selection of the correct immunosuppressive regimen for an individual patient for minimizing toxicity while maintaining immunosuppressive efficacy. AREAS COVERED Proteomics and metabolomics have the potential to develop sensitive and specific diagnostic tools for monitoring early changes in cell signal transduction, regulation and biochemical pathways. Here, we review the steps required for the development of molecular markers from discovery, mechanistic and clinical qualification to regulatory approval, and present a critical discussion of the current status of molecular marker development as relevant for the management and individualization of immunosuppressive drug regimens. EXPERT OPINION Although metabolomics and proteomics-based studies have yielded several candidate molecular markers, most published studies are poorly designed, statistically underpowered and/or often have not gone beyond the discovery stage. Most molecular marker candidates are still at an early stage. Due to the high complexity of and the resources required for diagnostic marker development, initiatives and consortia organized and supported by funding agencies and regulatory agencies will be critical.
Collapse
Affiliation(s)
- Uwe Christians
- University of Colorado, Department of Anesthesiology, 1999 North Fitzsimons Parkway, Bioscience East, Suite 100, Aurora, CO 80045-7503, USA.
| | | | | | | | | |
Collapse
|
37
|
Following dynamic biological processes through NMR-based metabonomics: A new tool in nanomedicine? J Control Release 2011; 153:34-9. [DOI: 10.1016/j.jconrel.2011.03.008] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 01/11/2011] [Accepted: 03/08/2011] [Indexed: 01/09/2023]
|
38
|
Robertson DG, Watkins PB, Reily MD. Metabolomics in toxicology: preclinical and clinical applications. Toxicol Sci 2010; 120 Suppl 1:S146-70. [PMID: 21127352 DOI: 10.1093/toxsci/kfq358] [Citation(s) in RCA: 130] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/29/2022] Open
Affiliation(s)
- Donald G Robertson
- Applied and Investigative Metabolomics, Bristol-Myers Squibb Co., Princeton, New Jersey 08543, USA.
| | | | | |
Collapse
|