1
|
Li N, Shi J, Chen Z, Dong Z, Ma S, Li Y, Huang X, Li X. In silico prediction of drug-induced nephrotoxicity: current progress and pitfalls. Expert Opin Drug Metab Toxicol 2024:1-13. [PMID: 39360665 DOI: 10.1080/17425255.2024.2412629] [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: 02/17/2024] [Revised: 09/05/2024] [Accepted: 10/01/2024] [Indexed: 10/04/2024]
Abstract
INTRODUCTION Due to its role in absorption and metabolism, the kidney is an important target for drug toxicity. Drug-induced nephrotoxicity (DIN) presents a significant challenge in clinical practice and drug development. Conventional methods for assessing nephrotoxicity have limitations, highlighting the need for innovative approaches. In recent years, in silico methods have emerged as promising tools for predicting DIN. AREAS COVERED A literature search was performed using PubMed and Web of Science, from 2013 to February 2023 for this review. This review provides an overview of the current progress and pitfalls in the in silico prediction of DIN, which discusses the principles and methodologies of computational models. EXPERT OPINION Despite significant advancements, this review identified issues accentuates the pivotal imperatives of data fidelity, model optimization, interdisciplinary collaboration, and mechanistic comprehension in sculpting the vista of DIN prediction. Integration of multiple data sources and collaboration between disciplines are essential for improving predictive models. Ultimately, a holistic approach combining computational, experimental, and clinical methods will enhance our understanding and management of DIN.
Collapse
Affiliation(s)
- Na Li
- Department of Clinical Pharmacy, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Engineering and Technology Research Center for Pediatric Drug Development, Shandong Medicine and Health Key Laboratory of Clinical Pharmacy, Jinan, China
| | - Juan Shi
- Department of Clinical Pharmacy, The First People's Hospital of Jinan, Jinan, China
| | - Zhaoyang Chen
- Department of Clinical Pharmacy, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Engineering and Technology Research Center for Pediatric Drug Development, Shandong Medicine and Health Key Laboratory of Clinical Pharmacy, Jinan, China
| | - Zhonghua Dong
- Department of Clinical Pharmacy, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Engineering and Technology Research Center for Pediatric Drug Development, Shandong Medicine and Health Key Laboratory of Clinical Pharmacy, Jinan, China
| | - Shiyu Ma
- Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yan Li
- Department of Clinical Pharmacy, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Engineering and Technology Research Center for Pediatric Drug Development, Shandong Medicine and Health Key Laboratory of Clinical Pharmacy, Jinan, China
| | - Xin Huang
- Department of Clinical Pharmacy, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Engineering and Technology Research Center for Pediatric Drug Development, Shandong Medicine and Health Key Laboratory of Clinical Pharmacy, Jinan, China
| | - Xiao Li
- Department of Clinical Pharmacy, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Engineering and Technology Research Center for Pediatric Drug Development, Shandong Medicine and Health Key Laboratory of Clinical Pharmacy, Jinan, China
| |
Collapse
|
2
|
Sangwan M, Chaudhary H, Mehan S, Khan Z, Bahauddin AA, Alrehaili BD, Elbadawy HM, Almikhlafi MA, Narula AS, Kalfin R, Wanas H. Effect of mitochondrial coenzyme-Q10 precursor solanesol in gentamicin-induced experimental nephrotoxicity: Evidence from restoration of ETC-complexes and histopathological alterations. Pharmacol Res Perspect 2024; 12:e70022. [PMID: 39358913 PMCID: PMC11446958 DOI: 10.1002/prp2.70022] [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] [Scholar Register] [Received: 11/21/2023] [Revised: 09/13/2024] [Accepted: 09/17/2024] [Indexed: 10/04/2024] Open
Abstract
Nephrotoxicity occurs when the body is exposed to certain drugs or toxins. When kidney damage occurs, the kidney fails to eliminate excess urine and waste. Solanesol (C45H74O) is a tri-sesquiterpenoid alcohol first isolated from tobacco, and it is widely distributed in plants of the Solanaceae family. Solanesol (SNL) is an intermediate in the synthesis of coenzyme Q10 (CoQ10), an antioxidant which protects nerve cells. This study investigated the protective effect of SNL at doses of 30 and 60 mg/kg in gentamicin-induced nephrotoxicity in Wistar albino rats. Animals were distributed into six groups and administered 100 mg/kg gentamicin-intraperitoneal injection for 14 days. Biochemical assessments were performed on kidney homogenate, blood, and serum. Treatment with SNL was shown as lower serum levels of creatinine, blood urea nitrogen (BUN), thiobarbituric acid reactive substances (TBARS), and Tumor necrosis factor alpha)TNF-α ((p < .001). It also restored reduced glutathione (GSH) and mitochondrial complex enzymatic activity as protective measures against gentamicin-induced nephrotoxicity. SNL were shown to reduce inflammation and oxidative stress markers (p < .001). Histological findings furtherly augmented the protective effects of SNL. Long-term SNL therapy also restored mitochondrial electron transport chain complex enzymes, such as complex-I (p < .001). In conclusion, these findings suggest that SNL can represent a protective therapeutic option for drug-induced nephrotoxicity, a long-term adverse effect of aminoglycoside antibiotics such as gentamicin.
Collapse
Affiliation(s)
- Minakshi Sangwan
- Department of Pharmaceutical SciencePDM UniversityBahadurgarhHaryanaIndia
| | - Hema Chaudhary
- Department of Pharmaceutical SciencePDM UniversityBahadurgarhHaryanaIndia
- School of Medical and Allied SciencesK R Mangalam UniversityGurugramIndia
| | - Sidharth Mehan
- Division of Neuroscience, Department of PharmacologyISF College of Pharmacy (An Autonomous (College)MogaPunjabIndia
| | - Zuber Khan
- Division of Neuroscience, Department of PharmacologyISF College of Pharmacy (An Autonomous (College)MogaPunjabIndia
| | - Ammar A. Bahauddin
- Department of Pharmacology and ToxicologyCollege of PharmacyTaibah UniversityMedinaKingdom of Saudi Arabia
| | - Bandar D. Alrehaili
- Department of Pharmacology and ToxicologyCollege of PharmacyTaibah UniversityMedinaKingdom of Saudi Arabia
| | - Hossein M. Elbadawy
- Department of Pharmacology and ToxicologyCollege of PharmacyTaibah UniversityMedinaKingdom of Saudi Arabia
| | - Mohannad A. Almikhlafi
- Department of Pharmacology and ToxicologyCollege of PharmacyTaibah UniversityMedinaKingdom of Saudi Arabia
| | | | - Reni Kalfin
- Institute of NeurobiologyBulgarian Academy of SciencesSofiaBulgaria
- Department of HealthcareSouth‐West University BlagoevgradBlagoevgradBulgaria
| | - Hanna Wanas
- Department of Pharmacology and ToxicologyCollege of PharmacyTaibah UniversityMedinaKingdom of Saudi Arabia
- Department of Medical Pharmacology, Faculty of MedicineCairo UniversityGizaEgypt
| |
Collapse
|
3
|
Varda L, Ekart R, Lainscak M, Maver U, Bevc S. Clinical Properties and Non-Clinical Testing of Mineralocorticoid Receptor Antagonists in In Vitro Cell Models. Int J Mol Sci 2024; 25:9088. [PMID: 39201774 PMCID: PMC11354261 DOI: 10.3390/ijms25169088] [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: 07/17/2024] [Revised: 08/16/2024] [Accepted: 08/20/2024] [Indexed: 09/03/2024] Open
Abstract
Mineralocorticoid receptor antagonists (MRAs) are one of the renin-angiotensin-aldosterone system inhibitors widely used in clinical practice. While spironolactone and eplerenone have a long-standing profile in clinical medicine, finerenone is a novel agent within the MRA class. It has a higher specificity for mineralocorticoid receptors, eliciting less pronounced adverse effects. Although approved for clinical use in patients with chronic kidney disease and heart failure, intensive non-clinical research aims to further elucidate its mechanism of action, including dose-related selectivity. Within the field, animal models remain the gold standard for non-clinical testing of drug pharmacological and toxicological properties. Their role, however, has been challenged by recent advances in in vitro models, mainly through sophisticated analytical tools and developments in data analysis. Currently, in vitro models are gaining momentum as possible platforms for advanced pharmacological and pathophysiological studies. This article focuses on past, current, and possibly future in vitro cell models research with clinically relevant MRAs.
Collapse
Affiliation(s)
- Luka Varda
- Department of Dialysis, Clinic for Internal Medicine, University Medical Centre Maribor, Ljubljanska ulica 5, 2000 Maribor, Slovenia; (L.V.); (R.E.)
| | - Robert Ekart
- Department of Dialysis, Clinic for Internal Medicine, University Medical Centre Maribor, Ljubljanska ulica 5, 2000 Maribor, Slovenia; (L.V.); (R.E.)
- Department of Pharmacology, Faculty of Medicine, University of Maribor, Taborska ulica 5, 2000 Maribor, Slovenia;
| | - Mitja Lainscak
- Division of Cardiology, Murska Sobota General Hospital, Ulica Dr. Vrbnjaka 6, 9000 Murska Sobota, Slovenia;
- Faculty of Medicine, University of Ljubljana, Vrazov trg 2, 1000 Ljubljana, Slovenia
| | - Uroš Maver
- Department of Pharmacology, Faculty of Medicine, University of Maribor, Taborska ulica 5, 2000 Maribor, Slovenia;
- Institute of Biomedical Sciences, Faculty of Medicine, University of Maribor, Taborska ulica 8, 2000 Maribor, Slovenia
| | - Sebastjan Bevc
- Department of Pharmacology, Faculty of Medicine, University of Maribor, Taborska ulica 5, 2000 Maribor, Slovenia;
- Department of Nephrology, Clinic for Internal Medicine, University Medical Centre Maribor, Ljubljanska ulica 5, 2000 Maribor, Slovenia
| |
Collapse
|
4
|
Arakawa H, Higuchi D, Takahashi E, Matsushita K, Nedachi S, Peng H, Kadoguchi M, Morimura K, Araki A, Kondo M, Ishiguro N, Jimbo Y, Tamai I. Three-dimensional Culture of Human Proximal Tubular Epithelial Cells for an in vitro Evaluation of Drug-induced Kidney Injury. J Pharm Sci 2024:S0022-3549(24)00312-5. [PMID: 39153661 DOI: 10.1016/j.xphs.2024.08.009] [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: 05/21/2024] [Revised: 08/12/2024] [Accepted: 08/13/2024] [Indexed: 08/19/2024]
Abstract
Drug-induced kidney injury (DIKI) is the major cause of acute kidney injury (AKI). Renal proximal tubular epithelial cells (RPTECs) are the primary target sites of DIKI and express transporters involved in renal drug disposition. In the present study, we focused on three-dimensionally cultured human RPTECs (3D-RPTECs) with elevated expression of drug transporters to investigate their utility in DIKI evaluation. Intracellular ATP levels in 3D-RPTECs are reduced by tenofovir and cisplatin that are substrates of an organic anion transporter 1 and an organic cation transporter 2, respectively. In addition, 3D-RPTECs were exposed to 17 and 15 drugs that are positive and negative to RPTEC toxicity, respectively, for up to 28 d. The 20 % decreasing concentration of drugs for ATP amount (EC20) was obtained, and the ratio of EC20 values and clinical maximum concentration (Cmax) ≤100 were used as cut-off value to evaluate potential of DIKI. The sensitivities of 3D-RPTECs were 82.4 % and 88.2 % after 7 d and 28 d of drug exposure, respectively, and the specificities were 100 % and 93.3 %, respectively. The predictive performance of 3D-RPTECs was higher than that of two-dimensional cultured RPTECs and the kidney cell line HK-2. In conclusion, 3D-RPTECs are useful for in vitro evaluation of RPTEC injury by measuring intracellular ATP levels.
Collapse
Affiliation(s)
- Hiroshi Arakawa
- Faculty of Pharmaceutical Sciences, Institute of Medical Pharmaceutical and Health Sciences, Kanazawa University, Kakuma-machi, Kanazawa 920-1192, Japan.
| | - Daichi Higuchi
- Faculty of Pharmaceutical Sciences, Institute of Medical Pharmaceutical and Health Sciences, Kanazawa University, Kakuma-machi, Kanazawa 920-1192, Japan
| | - Etsushi Takahashi
- R &D Department, Precision Engineering Center, Industrial Division, Nikkiso Co. Ltd, Japan
| | - Kohei Matsushita
- Division of Pathology, National Institute of Health Sciences, Kawasaki, Kanagawa, Japan
| | - Shiho Nedachi
- Faculty of Pharmaceutical Sciences, Institute of Medical Pharmaceutical and Health Sciences, Kanazawa University, Kakuma-machi, Kanazawa 920-1192, Japan
| | - Hanwei Peng
- Faculty of Pharmaceutical Sciences, Institute of Medical Pharmaceutical and Health Sciences, Kanazawa University, Kakuma-machi, Kanazawa 920-1192, Japan
| | - Moeno Kadoguchi
- Faculty of Pharmaceutical Sciences, Institute of Medical Pharmaceutical and Health Sciences, Kanazawa University, Kakuma-machi, Kanazawa 920-1192, Japan
| | - Kaoru Morimura
- R &D Department, Precision Engineering Center, Industrial Division, Nikkiso Co. Ltd, Japan
| | - Ayano Araki
- R &D Department, Precision Engineering Center, Industrial Division, Nikkiso Co. Ltd, Japan
| | - Masayuki Kondo
- R &D Department, Precision Engineering Center, Industrial Division, Nikkiso Co. Ltd, Japan
| | - Naoki Ishiguro
- Pharmacokinetics and Non-Clinical Safety Department, Nippon Boehringer Ingelheim Co., Ltd., Kobe, Japan
| | - Yoichi Jimbo
- R &D Department, Precision Engineering Center, Industrial Division, Nikkiso Co. Ltd, Japan
| | - Ikumi Tamai
- Faculty of Pharmaceutical Sciences, Institute of Medical Pharmaceutical and Health Sciences, Kanazawa University, Kakuma-machi, Kanazawa 920-1192, Japan.
| |
Collapse
|
5
|
Lacueva-Aparicio A, Martínez-Gimeno L, Torcal P, Ochoa I, Giménez I. Advanced Kidney Models In Vitro Using the Established Cell Line Renal Proximal Tubular Epithelial/Telomerase Reverse Transcriptase1 for Nephrotoxicity Assays. Biomimetics (Basel) 2024; 9:446. [PMID: 39056887 PMCID: PMC11275192 DOI: 10.3390/biomimetics9070446] [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: 06/10/2024] [Revised: 07/15/2024] [Accepted: 07/19/2024] [Indexed: 07/28/2024] Open
Abstract
Nephrotoxicity stands as one of the most limiting effects in the development and validation of new drugs. The kidney, among the organs evaluated in toxicity assessments, has a higher susceptibility, with nephrotoxic potential frequently evading detection until late in clinical trials. Traditional cell culture, which has been widely used for decades, does not recapitulate the structure and complexity of the native tissue, which can affect cell function, and the response to cytotoxins does not resemble what occurs in the kidney. In the current study, we aimed to address these challenges by creating in vitro kidney models that faithfully biomimic the dynamics of the renal proximal tubule, using the well-established RPTEC/TERT1 cell line. For doing so, two models were developed, one recreating tubule-like structures (2.5D model) and the other using microfluidic technology (kidney-on-a-chip). The 2.5D model allowed tubular structures to be generated in the absence of hydrogels, and the kidney-on-a-chip model allowed shear stress to be applied to the cell culture, which is a physiological stimulus in the renal tissue. After characterization of both models, different nephrotoxic compounds such as cisplatin, tacrolimus, and daunorubicin were used to study cell responses after treatment. The developed models in our study could be a valuable tool for pre-clinical nephrotoxic testing of drugs and new compounds.
Collapse
Affiliation(s)
- Alodia Lacueva-Aparicio
- Renal and Cardiovascular Physiopathology (FISIOPREN), Aragon Health Science Institute, 50009 Zaragoza, Spain (I.G.)
- Tissue Microenvironment Lab (TME Lab), I3A, University of Zaragoza, 50018 Zaragoza, Spain;
| | - Laura Martínez-Gimeno
- Renal and Cardiovascular Physiopathology (FISIOPREN), Aragon Health Science Institute, 50009 Zaragoza, Spain (I.G.)
- Institute for Health Sciences of Aragon (IACS), 50009 Zaragoza, Spain
- Aragón Health Research Institute (IISAragón), 50009 Zaragoza, Spain
| | - Pilar Torcal
- Renal and Cardiovascular Physiopathology (FISIOPREN), Aragon Health Science Institute, 50009 Zaragoza, Spain (I.G.)
- Institute for Health Sciences of Aragon (IACS), 50009 Zaragoza, Spain
- Aragón Health Research Institute (IISAragón), 50009 Zaragoza, Spain
| | - Ignacio Ochoa
- Tissue Microenvironment Lab (TME Lab), I3A, University of Zaragoza, 50018 Zaragoza, Spain;
- Aragón Health Research Institute (IISAragón), 50009 Zaragoza, Spain
- School of Medicine, University of Zaragoza, 50009 Zaragoza, Spain
- CIBER in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 28029 Madrid, Spain
| | - Ignacio Giménez
- Renal and Cardiovascular Physiopathology (FISIOPREN), Aragon Health Science Institute, 50009 Zaragoza, Spain (I.G.)
- Institute for Health Sciences of Aragon (IACS), 50009 Zaragoza, Spain
- Aragón Health Research Institute (IISAragón), 50009 Zaragoza, Spain
- School of Medicine, University of Zaragoza, 50009 Zaragoza, Spain
| |
Collapse
|
6
|
Meijer T, da Costa Pereira D, Klatt OC, Buitenhuis J, Jennings P, Wilmes A. Characterization of Organic Anion and Cation Transport in Three Human Renal Proximal Tubular Epithelial Models. Cells 2024; 13:1008. [PMID: 38920639 PMCID: PMC11202273 DOI: 10.3390/cells13121008] [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] [Scholar Register] [Received: 05/24/2024] [Accepted: 06/05/2024] [Indexed: 06/27/2024] Open
Abstract
The polarised expression of specific transporters in proximal tubular epithelial cells is important for the renal clearance of many endogenous and exogenous compounds. Thus, ideally, the in vitro tools utilised for predictions would have a similar expression of apical and basolateral xenobiotic transporters as in vivo. Here, we assessed the functionality of organic cation and anion transporters in proximal tubular-like cells (PTL) differentiated from human induced pluripotent stem cells (iPSC), primary human proximal tubular epithelial cells (PTEC), and telomerase-immortalised human renal proximal tubular epithelial cells (RPTEC/TERT1). Organic cation and anion transport were studied using the fluorescent substrates 4-(4-(dimethylamino)styryl)-N-methylpyridinium iodide (ASP) and 6-carboxyfluorescein (6-CF), respectively. The level and rate of intracellular ASP accumulation in PTL following basolateral application were slightly lower but within a 3-fold range compared to primary PTEC and RPTEC/TERT1 cells. The basolateral uptake of ASP and its subsequent apical efflux could be inhibited by basolateral exposure to quinidine in all models. Of the three models, only PTL showed a modest preferential basolateral-to-apical 6-CF transfer. These results show that organic cation transport could be demonstrated in all three models, but more research is needed to improve and optimise organic anion transporter expression and functionality.
Collapse
Affiliation(s)
- Tamara Meijer
- Department of Chemistry and Pharmaceutical Sciences, Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands; (T.M.); (D.d.C.P.); (O.C.K.); (P.J.)
- Amsterdam Institute of Molecular and Life Sciences (AIMMS), Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
| | - Daniel da Costa Pereira
- Department of Chemistry and Pharmaceutical Sciences, Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands; (T.M.); (D.d.C.P.); (O.C.K.); (P.J.)
- Amsterdam Institute of Molecular and Life Sciences (AIMMS), Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
| | - Olivia C. Klatt
- Department of Chemistry and Pharmaceutical Sciences, Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands; (T.M.); (D.d.C.P.); (O.C.K.); (P.J.)
- Amsterdam Institute of Molecular and Life Sciences (AIMMS), Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
| | - Joanne Buitenhuis
- Department of Chemistry and Pharmaceutical Sciences, Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands; (T.M.); (D.d.C.P.); (O.C.K.); (P.J.)
- Amsterdam Institute of Molecular and Life Sciences (AIMMS), Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
| | - Paul Jennings
- Department of Chemistry and Pharmaceutical Sciences, Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands; (T.M.); (D.d.C.P.); (O.C.K.); (P.J.)
- Amsterdam Institute of Molecular and Life Sciences (AIMMS), Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
| | - Anja Wilmes
- Department of Chemistry and Pharmaceutical Sciences, Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands; (T.M.); (D.d.C.P.); (O.C.K.); (P.J.)
- Amsterdam Institute of Molecular and Life Sciences (AIMMS), Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
| |
Collapse
|
7
|
Yen NTH, Tien NTN, Anh NTV, Le QV, Eunsu C, Kim HS, Moon KS, Nguyen HT, Kim DH, Long NP. Cyclosporine A-induced systemic metabolic perturbations in rats: A comprehensive metabolome analysis. Toxicol Lett 2024; 395:50-59. [PMID: 38552811 DOI: 10.1016/j.toxlet.2024.03.009] [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: 11/07/2023] [Revised: 03/12/2024] [Accepted: 03/26/2024] [Indexed: 04/11/2024]
Abstract
A better understanding of cyclosporine A (CsA)-induced nephro- and hepatotoxicity at the molecular level is necessary for safe and effective use. Utilizing a sophisticated study design, this study explored metabolic alterations after long-term CsA treatment in vivo. Rats were exposed to CsA with 4, 10, and 25 mg/kg for 4 weeks and then sacrificed to obtain liver, kidney, urine, and serum for untargeted metabolomics analysis. Differential network analysis was conducted to explore the biological relevance of metabolites significantly altered by toxicity-induced disturbance. Dose-dependent toxicity was observed in all biospecimens. The toxic effects were characterized by alterations of metabolites related to energy metabolism and cellular membrane composition, which could lead to the cholestasis-induced accumulation of bile acids in the tissues. The unfavorable impacts were also demonstrated in the serum and urine. Intriguingly, phenylacetylglycine was increased in the kidney, urine, and serum treated with high doses versus controls. Differential correlation network analysis revealed the strong correlations of deoxycytidine and guanosine with other metabolites in the network, which highlighted the influence of repeated CsA exposure on DNA synthesis. Overall, prolonged CsA administration had system-level dose-dependent effects on the metabolome in treated rats, suggesting the need for careful usage and dose adjustment.
Collapse
Affiliation(s)
- Nguyen Thi Hai Yen
- Department of Pharmacology and PharmacoGenomics Research Center, Inje University College of Medicine, Busan 47392, Republic of Korea
| | - Nguyen Tran Nam Tien
- Department of Pharmacology and PharmacoGenomics Research Center, Inje University College of Medicine, Busan 47392, Republic of Korea
| | - Nguyen Thi Van Anh
- Department of Pharmacology and PharmacoGenomics Research Center, Inje University College of Medicine, Busan 47392, Republic of Korea
| | - Quoc-Viet Le
- Faculty of Pharmacy, Ton Duc Thang University, Ho Chi Minh City 700000, Vietnam
| | - Cho Eunsu
- Department of Pharmacology and PharmacoGenomics Research Center, Inje University College of Medicine, Busan 47392, Republic of Korea
| | - Ho-Sook Kim
- Department of Pharmacology and PharmacoGenomics Research Center, Inje University College of Medicine, Busan 47392, Republic of Korea
| | - Kyoung-Sik Moon
- Korea Institute of Toxicology, Daejeon 34114, Republic of Korea
| | - Huy Truong Nguyen
- Faculty of Pharmacy, Ton Duc Thang University, Ho Chi Minh City 700000, Vietnam
| | - Dong Hyun Kim
- Department of Pharmacology and PharmacoGenomics Research Center, Inje University College of Medicine, Busan 47392, Republic of Korea.
| | - Nguyen Phuoc Long
- Department of Pharmacology and PharmacoGenomics Research Center, Inje University College of Medicine, Busan 47392, Republic of Korea.
| |
Collapse
|
8
|
Jiao G, Niu Y, Wang B. Editorial: Pathogenic mechanisms, injury biomarkers, prophylaxis and treatment strategy of drug-induced nephrotoxicity. Front Med (Lausanne) 2024; 11:1412795. [PMID: 38741772 PMCID: PMC11090037 DOI: 10.3389/fmed.2024.1412795] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Accepted: 04/15/2024] [Indexed: 05/16/2024] Open
Affiliation(s)
- Guozheng Jiao
- Chemistry and Chemical Engineering Institute, Taishan University, Tai'an, Shandong, China
| | - Yimin Niu
- Department of Pharmacy, Zhongda Hospital, Southeast University, Nanjing, Jiangsu, China
| | - Bin Wang
- Institute of Nephrology, Zhongda Hospital, Southeast University, Nanjing, Jiangsu, China
| |
Collapse
|
9
|
Oishi H, Tabibzadeh N, Morizane R. Advancing preclinical drug evaluation through automated 3D imaging for high-throughput screening with kidney organoids. Biofabrication 2024; 16:10.1088/1758-5090/ad38df. [PMID: 38547531 PMCID: PMC11304660 DOI: 10.1088/1758-5090/ad38df] [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] [Scholar Register] [Received: 11/14/2023] [Accepted: 03/28/2024] [Indexed: 04/09/2024]
Abstract
High-throughput drug screening is crucial for advancing healthcare through drug discovery. However, a significant limitation arises from availablein vitromodels using conventional 2D cell culture, which lack the proper phenotypes and architectures observed in three-dimensional (3D) tissues. Recent advancements in stem cell biology have facilitated the generation of organoids-3D tissue constructs that mimic human organsin vitro. Kidney organoids, derived from human pluripotent stem cells, represent a significant breakthrough in disease representation. They encompass major kidney cell types organized within distinct nephron segments, surrounded by stroma and endothelial cells. This tissue allows for the assessment of structural alterations such as nephron loss, a characteristic of chronic kidney disease. Despite these advantages, the complexity of 3D structures has hindered the use of organoids for large-scale drug screening, and the drug screening pipelines utilizing these complexin vitromodels remain to be established for high-throughput screening. In this study, we address the technical limitations of kidney organoids through fully automated 3D imaging, aided by a machine-learning approach for automatic profiling of nephron segment-specific epithelial morphometry. Kidney organoids were exposed to the nephrotoxic agent cisplatin to model severe acute kidney injury. An U.S. Food and Drug Administration (FDA)-approved drug library was tested for therapeutic and nephrotoxicity screening. The fully automated pipeline of 3D image acquisition and analysis identified nephrotoxic or therapeutic drugs during cisplatin chemotherapy. The nephrotoxic potential of these drugs aligned with previousin vivoand human reports. Additionally, Imatinib, a tyrosine kinase inhibitor used in hematological malignancies, was identified as a potential preventive therapy for cisplatin-induced kidney injury. Our proof-of-concept report demonstrates that the automated screening process, using 3D morphometric assays with kidney organoids, enables high-throughput screening for nephrotoxicity and therapeutic assessment in 3D tissue constructs.
Collapse
Affiliation(s)
- Haruka Oishi
- Nephrology Division, Department of Medicine, Massachusetts General Hospital, Boston, MA, United States of America
| | - Nahid Tabibzadeh
- Nephrology Division, Department of Medicine, Massachusetts General Hospital, Boston, MA, United States of America
- Harvard Medical School, Boston, MA, United States of America
| | - Ryuji Morizane
- Nephrology Division, Department of Medicine, Massachusetts General Hospital, Boston, MA, United States of America
- Harvard Medical School, Boston, MA, United States of America
- Harvard Stem Cell Institute (HSCI), Cambridge, MA, United States of America
| |
Collapse
|
10
|
Huang W, Chen YY, He FF, Zhang C. Revolutionizing nephrology research: expanding horizons with kidney-on-a-chip and beyond. Front Bioeng Biotechnol 2024; 12:1373386. [PMID: 38605984 PMCID: PMC11007038 DOI: 10.3389/fbioe.2024.1373386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Accepted: 03/18/2024] [Indexed: 04/13/2024] Open
Abstract
Organs-on-a-chip (OoC) is a microengineered three-dimensional cell culture system developed for decades. Utilizing microfluidic technology, OoC cultivates cells on perfusable channels to construct in vitro organ models, enabling the simulation of organ-level functions under physiological and pathophysiological conditions. The superior simulation capabilities compared to traditional animal experiments and two-dimensional cell cultures, making OoC a valuable tool for in vitro research. Recently, the application of OoC has extended to the field of nephrology, where it replicates various functional units, including glomerulus-on-a-chip, proximal tubule-on-a-chip, distal tubule-on-a-chip, collecting duct-on-a-chip, and even the entire nephron-on-a-chip to precisely emulate the structure and function of nephrons. Moreover, researchers have integrated kidney models into multi-organ systems, establishing human body-on-a-chip platforms. In this review, the diverse functional kidney units-on-a-chip and their versatile applications are outlined, such as drug nephrotoxicity screening, renal development studies, and investigations into the pathophysiological mechanisms of kidney diseases. The inherent advantages and current limitations of these OoC models are also examined. Finally, the synergy of kidney-on-a-chip with other emerging biomedical technologies are explored, such as bioengineered kidney and bioprinting, and a new insight for chip-based renal replacement therapy in the future are prospected.
Collapse
Affiliation(s)
| | | | | | - Chun Zhang
- *Correspondence: Fang-Fang He, ; Chun Zhang,
| |
Collapse
|
11
|
Taguchi K, Sugahara S, Elias BC, Pabla NS, Canaud G, Brooks CR. IL-22 is secreted by proximal tubule cells and regulates DNA damage response and cell death in acute kidney injury. Kidney Int 2024; 105:99-114. [PMID: 38054920 PMCID: PMC11068062 DOI: 10.1016/j.kint.2023.09.020] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 08/29/2023] [Accepted: 09/15/2023] [Indexed: 12/07/2023]
Abstract
Acute kidney injury (AKI) affects over 13 million people worldwide annually and is associated with a 4-fold increase in mortality. Our lab and others have shown that DNA damage response (DDR) governs the outcome of AKI in a bimodal manner. Activation of DDR sensor kinases protects against AKI, while hyperactivation of DDR effector proteins, such as p53, induces cell death and worsens AKI. The factors that trigger DDR to switch from pro-repair to pro-cell death remain to be resolved. Here we investigated the role of interleukin 22 (IL-22), an IL-10 family member whose receptor (IL-22RA1) is expressed on proximal tubule cells (PTCs), in DDR activation and AKI. Using cisplatin and aristolochic acid (AA) induced nephropathy as models of DNA damage, we identified PTCs as a novel source of urinary IL-22. Functionally, IL-22 binding IL-22RA1 on PTCs amplified the DDR. Treating primary PTCs with IL-22 alone induced rapid activation of the DDR. The combination of IL-22 and either cisplatin- or AA-induced cell death in primary PTCs, while the same dose of cisplatin or AA alone did not. Global deletion of IL-22 protected against cisplatin- or AA-induced AKI, reduced expression of DDR components, and inhibited PTC cell death. To confirm PTC IL-22 signaling contributed to AKI, we knocked out IL-22RA1 specifically in kidney tubule cells. IL-22RA1ΔTub mice displayed reduced DDR activation, cell death, and kidney injury compared to controls. Thus, targeting IL-22 represents a novel therapeutic approach to prevent the negative consequences of the DDR activation while not interfering with repair of damaged DNA.
Collapse
Affiliation(s)
- Kensei Taguchi
- Division of Nephrology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Sho Sugahara
- Division of Nephrology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Bertha C Elias
- Division of Nephrology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Navjot S Pabla
- Division of Pharmaceutics and Pharmacology, College of Pharmacy & Comprehensive Cancer Center, Ohio State University, Columbus, Ohio, USA
| | - Guillaume Canaud
- Overgrowth Syndrome and Vascular Anomalies Unit, Hôpital Necker Enfants Malades, Université de Paris, Paris, France
| | - Craig R Brooks
- Division of Nephrology, Vanderbilt University Medical Center, Nashville, Tennessee, USA; Department of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee, USA.
| |
Collapse
|
12
|
Mboni-Johnston IM, Kouidrat NMZ, Hirsch C, Weber AG, Meißner A, Adjaye J, Schupp N. Sensitivity of Human Induced Pluripotent Stem Cells and Thereof Differentiated Kidney Proximal Tubular Cells towards Selected Nephrotoxins. Int J Mol Sci 2023; 25:81. [PMID: 38203251 PMCID: PMC10779191 DOI: 10.3390/ijms25010081] [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] [Scholar Register] [Received: 11/08/2023] [Revised: 12/11/2023] [Accepted: 12/14/2023] [Indexed: 01/12/2024] Open
Abstract
Proximal tubular epithelial cells (PTEC) are constantly exposed to potentially toxic metabolites and xenobiotics. The regenerative potential of the kidney enables the replacement of damaged cells either via the differentiation of stem cells or the re-acquisition of proliferative properties of the PTEC. Nevertheless, it is known that renal function declines, suggesting that the deteriorated cells are not replaced by fully functional cells. To understand the possible causes of this loss of kidney cell function, it is crucial to understand the role of toxins during the regeneration process. Therefore, we investigated the sensitivity and function of human induced pluripotent stem cells (hiPSC), hiPSC differentiating, and hiPSC differentiated into proximal tubular epithelial-like cells (PTELC) to known nephrotoxins. hiPSC were differentiated into PTELC, which exhibited similar morphology to PTEC, expressed prototypical PTEC markers, and were able to undergo albumin endocytosis. When treated with two nephrotoxins, hiPSC and differentiating hiPSC were more sensitive to cisplatin than differentiated PTELC, whereas all stages were equally sensitive to cyclosporin A. Both toxins also had an inhibitory effect on albumin uptake. Our results suggest a high sensitivity of differentiating cells towards toxins, which could have an unfavorable effect on regenerative processes. To study this, our model of hiPSC differentiating into PTELC appears suitable.
Collapse
Affiliation(s)
- Isaac Musong Mboni-Johnston
- Institute of Toxicology, Medical Faculty and University Hospital, University of Düsseldorf, 40225 Düsseldorf, Germany; (I.M.M.-J.); (N.M.Z.K.); (C.H.); (A.M.)
| | - Nazih Mohamed Zakari Kouidrat
- Institute of Toxicology, Medical Faculty and University Hospital, University of Düsseldorf, 40225 Düsseldorf, Germany; (I.M.M.-J.); (N.M.Z.K.); (C.H.); (A.M.)
| | - Cornelia Hirsch
- Institute of Toxicology, Medical Faculty and University Hospital, University of Düsseldorf, 40225 Düsseldorf, Germany; (I.M.M.-J.); (N.M.Z.K.); (C.H.); (A.M.)
| | - Andreas Georg Weber
- Institute of Toxicology, Medical Faculty and University Hospital, University of Düsseldorf, 40225 Düsseldorf, Germany; (I.M.M.-J.); (N.M.Z.K.); (C.H.); (A.M.)
| | - Alexander Meißner
- Institute of Toxicology, Medical Faculty and University Hospital, University of Düsseldorf, 40225 Düsseldorf, Germany; (I.M.M.-J.); (N.M.Z.K.); (C.H.); (A.M.)
| | - James Adjaye
- Institute for Stem Cell Research and Regenerative Medicine, Medical Faculty and University Hospital, University of Düsseldorf, 40225 Düsseldorf, Germany;
- Zayed Centre for Research into Rare Diseases in Children (ZCR), EGA Institute for Women’s Health, University College London (UCL), 20 Guilford Street, London WC1N 1DZ, UK
| | - Nicole Schupp
- Institute of Toxicology, Medical Faculty and University Hospital, University of Düsseldorf, 40225 Düsseldorf, Germany; (I.M.M.-J.); (N.M.Z.K.); (C.H.); (A.M.)
| |
Collapse
|
13
|
Tang J, Yang N, Pan S, Ren P, Chen M, Jin J, He Q, Zeng Y. The renal damage and mechanisms relevant to antitumoral drugs. Front Oncol 2023; 13:1331671. [PMID: 38148845 PMCID: PMC10749913 DOI: 10.3389/fonc.2023.1331671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 11/22/2023] [Indexed: 12/28/2023] Open
Abstract
Over the past few decades, significant progress has been made in the development of drugs to combat cancer. It is unfortunate that these drugs can also lead to various kidney injuries and imbalances in electrolyte levels. Nephrotoxicity caused by chemotherapy drugs can impact different parts of the kidneys, including the glomeruli, renal tubules, interstitium, or renal microvessels. Despite the existing knowledge, our understanding of the mechanisms underlying the renal damage caused by antitumoral drugs remains incomplete. In this review, we aim to provide a comprehensive overview of the specific types of kidney injury and the mechanisms responsible for the drug-mediated renal damage, and briefly discuss possible prevention and treatment measures. Sensitive blood and urine biomarkers can provide clinicians with more information about kidney injury detection and reference value for subsequent treatment options. In addition, we emphasize that both oncologists and nephrologists have a responsibility to remain vigilant against the potential nephrotoxicity of the drugs. It's crucial for experts in both fields to collaborate in early detection, monitoring and prevention of kidney damage.
Collapse
Affiliation(s)
- Jiyu Tang
- The Second School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
- Department of Nephrology, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, Hangzhou, China
| | - Nan Yang
- Department of Nephrology, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, Hangzhou, China
- Jinzhou Medical University, Graduate School of Clinical Medicine, Jinzhou, China
| | - Shujun Pan
- School of Medicine, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Peiyao Ren
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Maosheng Chen
- Department of Nephrology, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, Hangzhou, China
| | - Juan Jin
- Department of Nephrology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Hangzhou, China
| | - Qiang He
- Department of Nephrology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Hangzhou, China
| | - Yuqun Zeng
- Department of Nephrology, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, Hangzhou, China
| |
Collapse
|
14
|
Hsiao HY, Yen TH, Wu FY, Cheng CM, Liu JW, Fan YT, Huang JJ, Nien CY. Delivery and Transcriptome Assessment of an In Vitro Three-Dimensional Proximal Tubule Model Established by Human Kidney 2 Cells in Clinical Gelatin Sponges. Int J Mol Sci 2023; 24:15547. [PMID: 37958530 PMCID: PMC10650118 DOI: 10.3390/ijms242115547] [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] [Scholar Register] [Received: 09/21/2023] [Revised: 10/10/2023] [Accepted: 10/14/2023] [Indexed: 11/15/2023] Open
Abstract
The high prevalence of kidney diseases and the low identification rate of drug nephrotoxicity in preclinical studies reinforce the need for representative yet feasible renal models. Although in vitro cell-based models utilizing renal proximal tubules are widely used for kidney research, many proximal tubule cell (PTC) lines have been indicated to be less sensitive to nephrotoxins, mainly due to altered expression of transporters under a two-dimensional culture (2D) environment. Here, we selected HK-2 cells to establish a simplified three-dimensional (3D) model using gelatin sponges as scaffolds. In addition to cell viability and morphology, we conducted a comprehensive transcriptome comparison and correlation analysis of 2D and 3D cultured HK-2 cells to native human PTCs. Our 3D model displayed stable and long-term growth with a tubule-like morphology and demonstrated a more comparable gene expression profile to native human PTCs compared to the 2D model. Many missing or low expressions of major genes involved in PTC transport and metabolic processes were restored, which is crucial for successful nephrotoxicity prediction. Consequently, we established a cost-effective yet more representative model for in vivo PTC studies and presented a comprehensive transcriptome analysis for the systematic characterization of PTC lines.
Collapse
Affiliation(s)
- Hui-Yi Hsiao
- Department of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan;
- Center for Tissue Engineering, Linkuo Chang Gung Memorial Hospital, Taoyuan 33305, Taiwan;
| | - Tzung-Hai Yen
- Department of Nephrology, Clinical Poison Center, Linkuo Chang Gung Memorial Hospital, Taoyuan 33305, Taiwan;
- Department of Nephrology, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
| | - Fang-Yu Wu
- Department of Life Science, National Central University, Taoyuan 32001, Taiwan; (F.-Y.W.); (Y.-T.F.)
| | - Chao-Min Cheng
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu 300193, Taiwan;
| | - Jia-Wei Liu
- Center for Tissue Engineering, Linkuo Chang Gung Memorial Hospital, Taoyuan 33305, Taiwan;
| | - Yu-Ting Fan
- Department of Life Science, National Central University, Taoyuan 32001, Taiwan; (F.-Y.W.); (Y.-T.F.)
| | - Jung-Ju Huang
- Division of Reconstructive Microsurgery, Department of Plastic and Reconstructive Surgery, Linkou Chang Gung Memorial Hospital, Taoyuan 33305, Taiwan;
| | - Chung-Yi Nien
- Department of Life Science, National Central University, Taoyuan 32001, Taiwan; (F.-Y.W.); (Y.-T.F.)
| |
Collapse
|
15
|
Zhang M, Li H, Huang L, Liu Y, Jiao XF, Zeng L, Jia ZJ, Cheng G, Zhang L, Zhang W. Drug-associated kidney injury in children: a disproportionality analysis of the FDA Adverse Event Reporting System. Eur J Pediatr 2023; 182:4655-4661. [PMID: 37561197 DOI: 10.1007/s00431-023-05146-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 07/27/2023] [Accepted: 08/01/2023] [Indexed: 08/11/2023]
Abstract
Drug-associated kidney injury is related to longer hospitalization and increased risk of chronic kidney disease and mortality. However, there is currently a lack of large population studies on drug-associated kidney injury in children. This study aimed to study perform data mining to generate hypotheses on drugs, which may deserve to be assessed as per their potential risk of increasing kidney injury in children. We extracted and analyzed reports on drugs associated with kidney injury in children in the FDA Adverse Event Reporting System (FAERS). We conducted a disproportionality analysis using proportional reporting ratio (PRR) to evaluate the association between drugs and kidney injury in children. Meanwhile, comparisons were performed with drug labels to identify drugs that, despite not having kidney injury currently mentioned in their labels, may potentially be associated with risks of kidney injury in children. A total of 6347 children had drug-associated kidney injury in the FAERS database. The top five drugs with the highest PRR were gentamicin (PRR = 12.28, N = 157 cases, Chi-Squared = 1602.77), piperacillin-tazobactam (PRR = 9.77, N = 129 cases, Chi-Squared = 1003.24), amlodipine (PRR = 8.98, N = 271 cases, Chi-Squared = 1861.46), vancomycin (PRR = 8.91, N = 295 cases, Chi-Squared = 1998.64), and ceftriaxone (PRR = 8.00, N = 251 cases, Chi-Squared = 1494.02). According to drug labels, 9 drugs (9/30) were classified as potential nephrotoxins. CONCLUSIONS Approximately one-third of drugs associated with kidney injury in children do not list kidney injury as a side effect in their drug labels. Future studies are therefore warranted to evaluate whether these drugs are associated with such a risk. WHAT IS KNOWN • Nephrotoxic drugs are an increasingly common cause of acute kidney injury in hospitalized children. • Currently, no study has systematically combed drugs associated with kidney injury in children. WHAT IS NEW • Approximately a third of drugs showing signals for potential kidney injury in children in data mining do not mention this side effect in their drug labels. • This study provides data on drugs needing further study to determine whether they might increase the risk of kidney injury in children.
Collapse
Affiliation(s)
- Miao Zhang
- Department of Pharmacy, West China Second University Hospital, Sichuan University, Chengdu, China
- Evidence-Based Pharmacy Center, West China Second University Hospital, Sichuan University, Chengdu, China
- NMPA Key Laboratory for Technical Research On Drug Products In Vitro and In Vivo Correlation, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Sichuan University, Chengdu, China
- West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Hailong Li
- Department of Pharmacy, West China Second University Hospital, Sichuan University, Chengdu, China
- Evidence-Based Pharmacy Center, West China Second University Hospital, Sichuan University, Chengdu, China
- NMPA Key Laboratory for Technical Research On Drug Products In Vitro and In Vivo Correlation, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Sichuan University, Chengdu, China
| | - Liang Huang
- Department of Pharmacy, West China Second University Hospital, Sichuan University, Chengdu, China
- Evidence-Based Pharmacy Center, West China Second University Hospital, Sichuan University, Chengdu, China
- NMPA Key Laboratory for Technical Research On Drug Products In Vitro and In Vivo Correlation, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Sichuan University, Chengdu, China
| | - Yan Liu
- Department of Pharmacy, West China Second University Hospital, Sichuan University, Chengdu, China
- Evidence-Based Pharmacy Center, West China Second University Hospital, Sichuan University, Chengdu, China
- NMPA Key Laboratory for Technical Research On Drug Products In Vitro and In Vivo Correlation, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Sichuan University, Chengdu, China
- West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Xue-Feng Jiao
- Department of Pharmacy, West China Second University Hospital, Sichuan University, Chengdu, China
- Evidence-Based Pharmacy Center, West China Second University Hospital, Sichuan University, Chengdu, China
- NMPA Key Laboratory for Technical Research On Drug Products In Vitro and In Vivo Correlation, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Sichuan University, Chengdu, China
| | - Linan Zeng
- Department of Pharmacy, West China Second University Hospital, Sichuan University, Chengdu, China
- Evidence-Based Pharmacy Center, West China Second University Hospital, Sichuan University, Chengdu, China
- NMPA Key Laboratory for Technical Research On Drug Products In Vitro and In Vivo Correlation, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Sichuan University, Chengdu, China
| | - Zhi-Jun Jia
- Department of Pharmacy, West China Second University Hospital, Sichuan University, Chengdu, China
- Evidence-Based Pharmacy Center, West China Second University Hospital, Sichuan University, Chengdu, China
- NMPA Key Laboratory for Technical Research On Drug Products In Vitro and In Vivo Correlation, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Sichuan University, Chengdu, China
- West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Guo Cheng
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Sichuan University, Chengdu, China
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
- Laboratory of Molecular Translational Medicine, Center for Translational Medicine, Sichuan University, Chengdu, China
| | - Lingli Zhang
- West China Biomedical Big Data Center, West China Hospital, Sichuan University, Chengdu, China.
- Medical Big Data Center, Sichuan University, Chengdu, China.
| | - Wei Zhang
- Department of Pharmacy, West China Second University Hospital, Sichuan University, Chengdu, China.
- Evidence-Based Pharmacy Center, West China Second University Hospital, Sichuan University, Chengdu, China.
- NMPA Key Laboratory for Technical Research On Drug Products In Vitro and In Vivo Correlation, Chengdu, China.
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Sichuan University, Chengdu, China.
- Chinese Evidence-Based Medicine Center, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
16
|
Huangfu Y, Wang J, Feng J, Zhang ZL. Distal renal tubular system-on-a-chip for studying the pathogenesis of influenza A virus-induced kidney injury. LAB ON A CHIP 2023; 23:4255-4264. [PMID: 37674367 DOI: 10.1039/d3lc00616f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/08/2023]
Abstract
Influenza A viruses typically cause acute respiratory infections in humans. However, virus-induced acute kidney injury (AKI) has dramatically increased mortality. The pathogenesis remains poorly understood due to limited disease models. Here, a distal renal tubular system-on-a-chip (dRTSC) was constructed to explore the pathogenesis. The renal tubule-vascular reabsorption interface was recapitulated by co-culturing the distal renal tubule and peritubular vessel with a collagen-coated porous membrane. To study the pathways of influenza virus entry into the kidney, dynamic tracking of fluorescence-labeled virus-infected blood vessels was performed. For the first time, the virus was shown to enter the kidney rapidly by cell-free transmission without disrupting the vascular barrier. Direct virus infection of renal tubules in dRTSC reveals disruption of tight junctions, microvilli formation, polar distribution of ion transporters, and sodium reabsorption function. This robust platform allows for a straightforward investigation of virus-induced AKI pathogenesis. The combination with single-virus tracking technology provides new insights into understanding influenza virus-induced extra-respiratory disease.
Collapse
Affiliation(s)
- Yueyue Huangfu
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, 430072, P.R. China.
| | - Ji Wang
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, 430072, P.R. China.
| | - Jiao Feng
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, 430072, P.R. China.
| | - Zhi-Ling Zhang
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, 430072, P.R. China.
| |
Collapse
|
17
|
Singh NK, Kim JY, Jang J, Kim YK, Cho DW. 3D Cell Printing of Advanced Vascularized Proximal Tubule-on-a-Chip for Drug Induced Nephrotoxicity Advancement. ACS APPLIED BIO MATERIALS 2023; 6:3750-3758. [PMID: 37606916 DOI: 10.1021/acsabm.3c00421] [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] [Indexed: 08/23/2023]
Abstract
Renal dysfunction due to drug-induced nephrotoxicity (DIN) affects >20% of the adult population worldwide. The vascularized proximal tubule is a complex structure that is often the primary site of drug-induced kidney injury. Herein, a vascularized proximal tubule-on-a-chip (Vas-POAC) was fabricated, demonstrating improved physiological emulation over earlier single-cell proximal tubule models. A perfusable model of vascularized proximal tubules permits the growth and proliferation of renal proximal tubule cells and adjacent endothelial cells under various conditions. An in vitro Vas-POAC showed mature expressions of the tubule and endothelial cell markers in the mature epithelium and endothelium lumens after 7 days of culture. Expression in the mature proximal tubule epithelium resembled the polarized expression of sodium-glucose cotransporter-2 and the de novo synthesis of ECM proteins. These perfusable Vas-POACs display significantly improved functional properties relative to the proximal tubules-on-a-chip (POAC), which lacks vascular components. Furthermore, the developed Vas-POAC model evaluated the cisplatin-induced nephrotoxicity and revealed enhanced drug receptivity compared to POAC. We further evaluated the capability of the developed proximal tubule model to act as a functional platform that targets screening drug doses that can cause renal proximal tubule injury in adults. Thus, our cell-printed models may prove valuable for screening, thoughtful mechanistic investigations of DIN, and discovery of drugs that interfere with tubule formation.
Collapse
Affiliation(s)
- Narendra K Singh
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), Pohang 37673, Republic of Korea
- Division of Biomaterials and Biomechanics, School of Dentistry, Oregon Health and Science University (OHSU), Portland, Oregon 97201, United States
- Cancer Early Detection Advanced Research Center (CEDAR), OHSU-Knight Cancer Institute, Portland, Oregon 97201, United States
| | - Jae Yun Kim
- School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology (POSTECH), Pohang 37673, Republic of Korea
| | - Jinah Jang
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), Pohang 37673, Republic of Korea
- School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology (POSTECH), Pohang 37673, Republic of Korea
- Department of Convergence IT Engineering, Pohang University of Science and Technology (POSTECH), Pohang 37673, Republic of Korea
- Institute for Convergence Research and Education in Advanced Technology, Yonsei University, Seoul 03722, Republic of Korea
| | - Yong Kyun Kim
- Cell Death Disease Research Center, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
- Department of Internal Medicine, College of Medicine, The Catholic University of Korea, St. Vincent's Hospital, Suwon 16247, Republic of Korea
- POSTECH-Catholic Biomedical Engineering Institute, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Dong-Woo Cho
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), Pohang 37673, Republic of Korea
- Institute for Convergence Research and Education in Advanced Technology, Yonsei University, Seoul 03722, Republic of Korea
| |
Collapse
|
18
|
Khalil NN, Petersen AP, Song CJ, Chen Y, Takamoto K, Kellogg AC, Chen EZ, McMahon AP, McCain ML. User-friendly microfluidic system reveals native-like morphological and transcriptomic phenotypes induced by shear stress in proximal tubule epithelium. APL Bioeng 2023; 7:036106. [PMID: 37584027 PMCID: PMC10424157 DOI: 10.1063/5.0143614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Accepted: 07/24/2023] [Indexed: 08/17/2023] Open
Abstract
Drug-induced nephrotoxicity is a leading cause of drug attrition, partly due to the limited relevance of pre-clinical models of the proximal tubule. Culturing proximal tubule epithelial cells (PTECs) under fluid flow to mimic physiological shear stress has been shown to improve select phenotypes, but existing flow systems are expensive and difficult to implement by non-experts in microfluidics. Here, we designed and fabricated an accessible and modular flow system for culturing PTECs under physiological shear stress, which induced native-like cuboidal morphology, downregulated pathways associated with hypoxia, stress, and injury, and upregulated xenobiotic metabolism pathways. We also compared the expression profiles of shear-dependent genes in our in vitro PTEC tissues to that of ex vivo proximal tubules and observed stronger clustering between ex vivo proximal tubules and PTECs under physiological shear stress relative to PTECs under negligible shear stress. Together, these data illustrate the utility of our user-friendly flow system and highlight the role of shear stress in promoting native-like morphological and transcriptomic phenotypes in PTECs in vitro, which is critical for developing more relevant pre-clinical models of the proximal tubule for drug screening or disease modeling.
Collapse
Affiliation(s)
- Natalie N. Khalil
- Alfred E. Mann Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles, California 90089, USA
| | - Andrew P. Petersen
- Alfred E. Mann Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles, California 90089, USA
| | | | - Yibu Chen
- USC Libraries Bioinformatics Service, University of Southern California, Los Angeles, California 90089, USA
| | - Kaelyn Takamoto
- Alfred E. Mann Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles, California 90089, USA
| | - Austin C. Kellogg
- Alfred E. Mann Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles, California 90089, USA
| | - Elaine Zhelan Chen
- Alfred E. Mann Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles, California 90089, USA
| | | | - Megan L. McCain
- Author to whom correspondence should be addressed:. Tel.: +1 2138210791. URL:https://livingsystemsengineering.usc.edu
| |
Collapse
|
19
|
Ishiguro N, Takahashi E, Arakawa H, Saito A, Kitagawa F, Kondo M, Morinaga G, Takatani M, Takahashi R, Kudo T, Mae SI, Kadoguchi M, Higuchi D, Nakazono Y, Tamai I, Osafune K, Jimbo Y. Improvement of Protein Expression Profile in Three-Dimensional Renal Proximal Tubular Epithelial Cell Spheroids Selected Based on OAT1 Gene Expression: A Potential In Vitro Tool for Evaluating Human Renal Proximal Tubular Toxicity and Drug Disposition. Drug Metab Dispos 2023; 51:1177-1187. [PMID: 37385755 DOI: 10.1124/dmd.122.001171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2022] [Revised: 06/05/2023] [Accepted: 06/21/2023] [Indexed: 07/01/2023] Open
Abstract
The proximal tubule plays an important role in the kidney and is a major site of drug interaction and toxicity. Analysis of kidney toxicity via in vitro assays is challenging, because only a few assays that reflect functions of drug transporters in renal proximal tubular epithelial cells (RPTECs) are available. In this study, we aimed to develop a simple and reproducible method for culturing RPTECs by monitoring organic anion transporter 1 (OAT1) as a selection marker. Culturing RPTECs in spherical cellular aggregates increased OAT1 protein expression, which was low in the conventional two-dimensional (2D) culture, to a level similar to that in human renal cortices. By proteome analysis, it was revealed that the expression of representative two proximal tubule markers was maintained and 3D spheroid culture improved the protein expression of approximately 7% of the 139 transporter proteins detected, and the expression of 2.3% of the 4,800 proteins detected increased by approximately fivefold that in human renal cortices. Furthermore, the expression levels of approximately 4,800 proteins in three-dimensional (3D) RPTEC spheroids (for 12 days) were maintained for over 20 days. Cisplatin and adefovir exhibited transporter-dependent ATP decreases in 3D RPTEC spheroids. These results indicate that the 3D RPTEC spheroids developed by monitoring OAT1 gene expression are a simple and reproducible in vitro experimental system with improved gene and protein expressions compared with 2D RPTECs and were more similar to that in human kidney cortices. Therefore, it can potentially be used for evaluating human renal proximal tubular toxicity and drug disposition. SIGNIFICANCE STATEMENT: This study developed a simple and reproducible spheroidal culture method with acceptable throughput using commercially available RPTECs by monitoring OAT1 gene expression. RPTECs cultured using this new method showed improved mRNA/protein expression profiles to those in 2D RPTECs and were more similar to those of human kidney cortices. This study provides a potential in vitro proximal tubule system for pharmacokinetic and toxicological evaluations during drug development.
Collapse
Affiliation(s)
- Naoki Ishiguro
- Pharmacokinetics and Non-Clinical Safety Department, Nippon Boehringer Ingelheim Company, Ltd., Kobe, Japan (N.I., A.S., G.M., M.T., R.T., T.K.); R&D Department, Industrial Division, Nikkiso Company, Ltd., Kanazawa, Japan (E.T., F.K., Ma.K., Y.J.); Faculty of Pharmaceutical Sciences, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan (H.A., Mo.K., D.H., Y.N., I.T.); and Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan (S.M., K.O.)
| | - Etsushi Takahashi
- Pharmacokinetics and Non-Clinical Safety Department, Nippon Boehringer Ingelheim Company, Ltd., Kobe, Japan (N.I., A.S., G.M., M.T., R.T., T.K.); R&D Department, Industrial Division, Nikkiso Company, Ltd., Kanazawa, Japan (E.T., F.K., Ma.K., Y.J.); Faculty of Pharmaceutical Sciences, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan (H.A., Mo.K., D.H., Y.N., I.T.); and Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan (S.M., K.O.)
| | - Hiroshi Arakawa
- Pharmacokinetics and Non-Clinical Safety Department, Nippon Boehringer Ingelheim Company, Ltd., Kobe, Japan (N.I., A.S., G.M., M.T., R.T., T.K.); R&D Department, Industrial Division, Nikkiso Company, Ltd., Kanazawa, Japan (E.T., F.K., Ma.K., Y.J.); Faculty of Pharmaceutical Sciences, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan (H.A., Mo.K., D.H., Y.N., I.T.); and Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan (S.M., K.O.)
| | - Asami Saito
- Pharmacokinetics and Non-Clinical Safety Department, Nippon Boehringer Ingelheim Company, Ltd., Kobe, Japan (N.I., A.S., G.M., M.T., R.T., T.K.); R&D Department, Industrial Division, Nikkiso Company, Ltd., Kanazawa, Japan (E.T., F.K., Ma.K., Y.J.); Faculty of Pharmaceutical Sciences, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan (H.A., Mo.K., D.H., Y.N., I.T.); and Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan (S.M., K.O.)
| | - Fumihiko Kitagawa
- Pharmacokinetics and Non-Clinical Safety Department, Nippon Boehringer Ingelheim Company, Ltd., Kobe, Japan (N.I., A.S., G.M., M.T., R.T., T.K.); R&D Department, Industrial Division, Nikkiso Company, Ltd., Kanazawa, Japan (E.T., F.K., Ma.K., Y.J.); Faculty of Pharmaceutical Sciences, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan (H.A., Mo.K., D.H., Y.N., I.T.); and Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan (S.M., K.O.)
| | - Masayuki Kondo
- Pharmacokinetics and Non-Clinical Safety Department, Nippon Boehringer Ingelheim Company, Ltd., Kobe, Japan (N.I., A.S., G.M., M.T., R.T., T.K.); R&D Department, Industrial Division, Nikkiso Company, Ltd., Kanazawa, Japan (E.T., F.K., Ma.K., Y.J.); Faculty of Pharmaceutical Sciences, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan (H.A., Mo.K., D.H., Y.N., I.T.); and Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan (S.M., K.O.)
| | - Gaku Morinaga
- Pharmacokinetics and Non-Clinical Safety Department, Nippon Boehringer Ingelheim Company, Ltd., Kobe, Japan (N.I., A.S., G.M., M.T., R.T., T.K.); R&D Department, Industrial Division, Nikkiso Company, Ltd., Kanazawa, Japan (E.T., F.K., Ma.K., Y.J.); Faculty of Pharmaceutical Sciences, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan (H.A., Mo.K., D.H., Y.N., I.T.); and Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan (S.M., K.O.)
| | - Masahito Takatani
- Pharmacokinetics and Non-Clinical Safety Department, Nippon Boehringer Ingelheim Company, Ltd., Kobe, Japan (N.I., A.S., G.M., M.T., R.T., T.K.); R&D Department, Industrial Division, Nikkiso Company, Ltd., Kanazawa, Japan (E.T., F.K., Ma.K., Y.J.); Faculty of Pharmaceutical Sciences, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan (H.A., Mo.K., D.H., Y.N., I.T.); and Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan (S.M., K.O.)
| | - Ryo Takahashi
- Pharmacokinetics and Non-Clinical Safety Department, Nippon Boehringer Ingelheim Company, Ltd., Kobe, Japan (N.I., A.S., G.M., M.T., R.T., T.K.); R&D Department, Industrial Division, Nikkiso Company, Ltd., Kanazawa, Japan (E.T., F.K., Ma.K., Y.J.); Faculty of Pharmaceutical Sciences, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan (H.A., Mo.K., D.H., Y.N., I.T.); and Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan (S.M., K.O.)
| | - Takashi Kudo
- Pharmacokinetics and Non-Clinical Safety Department, Nippon Boehringer Ingelheim Company, Ltd., Kobe, Japan (N.I., A.S., G.M., M.T., R.T., T.K.); R&D Department, Industrial Division, Nikkiso Company, Ltd., Kanazawa, Japan (E.T., F.K., Ma.K., Y.J.); Faculty of Pharmaceutical Sciences, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan (H.A., Mo.K., D.H., Y.N., I.T.); and Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan (S.M., K.O.)
| | - Shin-Ichi Mae
- Pharmacokinetics and Non-Clinical Safety Department, Nippon Boehringer Ingelheim Company, Ltd., Kobe, Japan (N.I., A.S., G.M., M.T., R.T., T.K.); R&D Department, Industrial Division, Nikkiso Company, Ltd., Kanazawa, Japan (E.T., F.K., Ma.K., Y.J.); Faculty of Pharmaceutical Sciences, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan (H.A., Mo.K., D.H., Y.N., I.T.); and Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan (S.M., K.O.)
| | - Moeno Kadoguchi
- Pharmacokinetics and Non-Clinical Safety Department, Nippon Boehringer Ingelheim Company, Ltd., Kobe, Japan (N.I., A.S., G.M., M.T., R.T., T.K.); R&D Department, Industrial Division, Nikkiso Company, Ltd., Kanazawa, Japan (E.T., F.K., Ma.K., Y.J.); Faculty of Pharmaceutical Sciences, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan (H.A., Mo.K., D.H., Y.N., I.T.); and Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan (S.M., K.O.)
| | - Daichi Higuchi
- Pharmacokinetics and Non-Clinical Safety Department, Nippon Boehringer Ingelheim Company, Ltd., Kobe, Japan (N.I., A.S., G.M., M.T., R.T., T.K.); R&D Department, Industrial Division, Nikkiso Company, Ltd., Kanazawa, Japan (E.T., F.K., Ma.K., Y.J.); Faculty of Pharmaceutical Sciences, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan (H.A., Mo.K., D.H., Y.N., I.T.); and Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan (S.M., K.O.)
| | - Yuya Nakazono
- Pharmacokinetics and Non-Clinical Safety Department, Nippon Boehringer Ingelheim Company, Ltd., Kobe, Japan (N.I., A.S., G.M., M.T., R.T., T.K.); R&D Department, Industrial Division, Nikkiso Company, Ltd., Kanazawa, Japan (E.T., F.K., Ma.K., Y.J.); Faculty of Pharmaceutical Sciences, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan (H.A., Mo.K., D.H., Y.N., I.T.); and Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan (S.M., K.O.)
| | - Ikumi Tamai
- Pharmacokinetics and Non-Clinical Safety Department, Nippon Boehringer Ingelheim Company, Ltd., Kobe, Japan (N.I., A.S., G.M., M.T., R.T., T.K.); R&D Department, Industrial Division, Nikkiso Company, Ltd., Kanazawa, Japan (E.T., F.K., Ma.K., Y.J.); Faculty of Pharmaceutical Sciences, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan (H.A., Mo.K., D.H., Y.N., I.T.); and Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan (S.M., K.O.)
| | - Kenji Osafune
- Pharmacokinetics and Non-Clinical Safety Department, Nippon Boehringer Ingelheim Company, Ltd., Kobe, Japan (N.I., A.S., G.M., M.T., R.T., T.K.); R&D Department, Industrial Division, Nikkiso Company, Ltd., Kanazawa, Japan (E.T., F.K., Ma.K., Y.J.); Faculty of Pharmaceutical Sciences, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan (H.A., Mo.K., D.H., Y.N., I.T.); and Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan (S.M., K.O.)
| | - Yoichi Jimbo
- Pharmacokinetics and Non-Clinical Safety Department, Nippon Boehringer Ingelheim Company, Ltd., Kobe, Japan (N.I., A.S., G.M., M.T., R.T., T.K.); R&D Department, Industrial Division, Nikkiso Company, Ltd., Kanazawa, Japan (E.T., F.K., Ma.K., Y.J.); Faculty of Pharmaceutical Sciences, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan (H.A., Mo.K., D.H., Y.N., I.T.); and Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan (S.M., K.O.)
| |
Collapse
|
20
|
Amarasiri SS, Attanayake AP, Arawwawala LD, Mudduwa LK, Jayatilaka KA. Barleria prionitis L. extracts ameliorate doxorubicin-induced acute kidney injury via modulation of oxidative stress, inflammation, and apoptosis. J Tradit Complement Med 2023; 13:500-510. [PMID: 37693098 PMCID: PMC10491988 DOI: 10.1016/j.jtcme.2023.05.007] [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: 05/26/2022] [Revised: 05/03/2023] [Accepted: 05/30/2023] [Indexed: 09/12/2023] Open
Abstract
Background and aim Doxorubicin (DOX) is a chemotherapeutic drug with potential nephrotoxic effects on patients who are on cancer chemotherapy. An interest has been observed in using natural products to ameliorate the potential side effects of DOX. The present study is to investigate the cellular mechanisms underlying the protective effects of Barleria prionitis L. (BP) (Acanthaceae) extracts, DOX-induced acute kidney injury (AKI). Experimental procedure Hexane (25 mg/kg/day), ethyl acetate (80 mg/kg/day), n-butanol (70 mg/kg/day), and water (120 mg/kg/day) extracts of BP, were administered to DOX-induced (5 mg/kg (2500 μL/kg), ip) Wistar rats for four consecutive weeks. At the end of the study, investigations were carried out for the assessment of biomarkers of nephrotoxicity, oxidative stress, inflammation, and apoptosis. Results Treatments with BP extracts significantly reversed DOX-induced elevations in serum and urine biochemical markers of nephrotoxicity (serum creatinine; 21-33%, blood urea nitrogen; 26-58%, β2-microglobulin; 19-22% and urine total protein; 47-67%). There was a reduction in the levels of tumor necrosis factor-α, interleukin-1β, and malondialdehyde in kidney homogenates of rats treated with the n-butanol extract (by 43, 62, and 24%) and water extract (by 57%, 85%, and 26%) (p < 0.05). Immunohistochemical expression of the pro-apoptotic B-cell associated X protein was reduced while the anti-apoptotic B-cell lymphoma gene product 2 protein was increased in kidney tissues after the treatments with BP extracts. Conclusions The selected BP extracts significantly ameliorated DOX-induced AKI. The findings would open new vistas for the development of a drug using the BP extracts to minimize DOX-induced AKI in cancer patients.
Collapse
Affiliation(s)
- Sachinthi S. Amarasiri
- Department of Medical Laboratory Science, Faculty of Allied Health Sciences, University of Ruhuna, Galle, Sri Lanka
| | - Anoja P. Attanayake
- Department of Biochemistry, Faculty of Medicine, University of Ruhuna, Galle, Sri Lanka
| | | | - Lakmini K.B. Mudduwa
- Department of Pathology, Faculty of Medicine, University of Ruhuna, Galle, Sri Lanka
| | | |
Collapse
|
21
|
Ellis C, Inaba K, Van de Vuurst C, Ghrayeb A, Cory TJ. Drug-drug interactions between COVID-19 therapeutics and antiretroviral treatment: the evidence to date. Expert Opin Drug Metab Toxicol 2023; 19:795-806. [PMID: 37800561 PMCID: PMC10841549 DOI: 10.1080/17425255.2023.2267970] [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] [Scholar Register] [Received: 04/20/2023] [Accepted: 10/04/2023] [Indexed: 10/07/2023]
Abstract
INTRODUCTION With new effective treatments for SARS-CoV-2, patient outcomes have greatly improved. However, new medications bring a risk of drug interactions with other medications. People living with HIV (PLWH) are at particular risk for these interactions due to heightened risk of immunosuppression, polypharmacy, and overlap in affected organs. It is critical to identify drug interactions are a significant barrier to care for PLWH. Establishing a better understanding of the pharmacologic relationships between COVID-19 therapies and antiretrovirals will improve patient-centered care in COVID-19. AREAS COVERED Potential drug-drug interactions between Human Immunodeficiency Virus (HIV) and COVID-19 treatments are detailed and reviewed here. The mechanisms seen in these interactions include alterations in metabolic enzymes, drug transporters, pharmacoenhancement, and organ toxicities. We also review the limitations and solutions that can be used to combat drug-drug interactions between these two disease states. EXPERT OPINION While current drug interactions are relatively mild between HIV and COVID-19 therapies, improvements in identifying these beforehand must take place as new therapies are approved. Antiretroviral therapy (ART) is essential in PLWH and must be maintained when treating COVID-19. As advancements in care occur, there is the possibility that newly approved drugs may have additional unknown interactions.
Collapse
Affiliation(s)
- Camden Ellis
- Department of Clinical Pharmacy and Translational Science, University of Tennessee Health Science Center College of Pharmacy, Memphis, USA
| | - Keita Inaba
- Department of Clinical Pharmacy and Translational Science, University of Tennessee Health Science Center College of Pharmacy, Memphis, USA
| | - Christine Van de Vuurst
- Department of Clinical Pharmacy and Translational Science, University of Tennessee Health Science Center College of Pharmacy, Memphis, USA
| | - Atheel Ghrayeb
- Department of Clinical Pharmacy and Translational Science, University of Tennessee Health Science Center College of Pharmacy, Memphis, USA
| | - Theodore James Cory
- Department of Clinical Pharmacy and Translational Science, University of Tennessee Health Science Center College of Pharmacy, Memphis, USA
| |
Collapse
|
22
|
Taguchi K, Sugahara S, Elias BC, Pabla N, Canaud G, Brooks CR. IL-22 promotes acute kidney injury through activation of the DNA damage response and cell death in proximal tubule cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.08.544134. [PMID: 37333314 PMCID: PMC10274795 DOI: 10.1101/2023.06.08.544134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
Acute kidney injury (AKI) affects over 13 million people world-wide annually and is associated with a fourfold increase in mortality. Our lab and others have shown that DNA damage response (DDR) governs the outcome of AKI in a bimodal manner. Activation of DDR sensor kinases protects against AKI, while hyperactivation of DDR effector proteins, such as p53, induces to cell death and worsens AKI. The factors that trigger the switch from pro-reparative to pro-cell death DDR remain to be resolved. Here we investigate the role of interleukin 22 (IL-22), an IL-10 family member whose receptor (IL-22RA1) is expressed on proximal tubule cells (PTCs), in DDR activation and AKI. Using cisplatin and aristolochic acid (AA) induced nephropathy as models of DNA damage, we identify PTCs as a novel source of urinary IL-22, making PTCs the only epithelial cells known to secret IL-22, to our knowledge. Functionally, IL-22 binding its receptor (IL-22RA1) on PTCs amplifies the DDR. Treating primary PTCs with IL-22 alone induces rapid activation of the DDR in vitro. The combination of IL-22 + cisplatin or AA treatment on primary PTCs induces cell death, while the same dose of cisplatin or AA alone does not. Global deletion of IL-22 protects against cisplatin or AA induced AKI. IL-22 deletion reduces expression of components of the DDR and inhibits PTC cell death. To confirm PTC IL-22 signaling contributes to AKI, we knocked out IL-22RA1 in renal epithelial cells by crossing IL-22RA1floxed mice with Six2-Cre mice. IL-22RA1 KO reduced DDR activation, cell death, and kidney injury. These data demonstrate that IL-22 promotes DDR activation in PTCs, switching pro-recovery DDR responses to a pro-cell death response and worsening AKI. Targeting IL-22 represents a novel therapeutic approach to prevent the negative consequences of the DDR activation while not interfering with the processes necessary for repair of damaged DNA.
Collapse
Affiliation(s)
- Kensei Taguchi
- Division of Nephrology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Sho Sugahara
- Division of Nephrology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Bertha C Elias
- Division of Nephrology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Navjot Pabla
- Division of Pharmaceutics and Pharmacology, College of Pharmacy & Comprehensive Cancer Center, Ohio State University, Columbus, OH, USA
| | - Guillaume Canaud
- Overgrowth Syndrome and Vascular Anomalies Unit, Hôpital Necker Enfants Malades, Université de Paris, Paris, France
| | - Craig R Brooks
- Division of Nephrology, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN, USA
| |
Collapse
|
23
|
Hou C, Gu Y, Yuan W, Zhang W, Xiu X, Lin J, Gao Y, Liu P, Chen X, Song L. Application of microfluidic chips in the simulation of the urinary system microenvironment. Mater Today Bio 2023; 19:100553. [PMID: 36747584 PMCID: PMC9898763 DOI: 10.1016/j.mtbio.2023.100553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 01/01/2023] [Accepted: 01/16/2023] [Indexed: 01/22/2023] Open
Abstract
The urinary system, comprising the kidneys, ureters, bladder, and urethra, has a unique mechanical and fluid microenvironment, which is essential to the urinary system growth and development. Microfluidic models, based on micromachining and tissue engineering technology, can integrate pathophysiological characteristics, maintain cell-cell and cell-extracellular matrix interactions, and accurately simulate the vital characteristics of human tissue microenvironments. Additionally, these models facilitate improved visualization and integration and meet the requirements of the laminar flow environment of the urinary system. However, several challenges continue to impede the development of a tissue microenvironment with controllable conditions closely resemble physiological conditions. In this review, we describe the biochemical and physical microenvironment of the urinary system and explore the feasibility of microfluidic technology in simulating the urinary microenvironment and pathophysiological characteristics in vitro. Moreover, we summarize the current research progress on adapting microfluidic chips for constructing the urinary microenvironment. Finally, we discuss the current challenges and suggest directions for future development and application of microfluidic technology in constructing the urinary microenvironment in vitro.
Collapse
Affiliation(s)
- Changhao Hou
- Department of Urology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
- Shanghai Eastern Institute of Urologic Reconstruction, Shanghai, China
| | - Yubo Gu
- Department of Urology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
- Shanghai Eastern Institute of Urologic Reconstruction, Shanghai, China
| | - Wei Yuan
- Department of Urology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
- Shanghai Eastern Institute of Urologic Reconstruction, Shanghai, China
| | - Wukai Zhang
- National Key Laboratory of Science and Technology on Micro/Nano Fabrication, Department of Micro/Nano Electronics, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Xianjie Xiu
- Department of Urology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
- Shanghai Eastern Institute of Urologic Reconstruction, Shanghai, China
| | - Jiahao Lin
- Department of Urology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
- Shanghai Eastern Institute of Urologic Reconstruction, Shanghai, China
| | - Yue Gao
- National Key Laboratory of Science and Technology on Micro/Nano Fabrication, Department of Micro/Nano Electronics, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Peichuan Liu
- National Key Laboratory of Science and Technology on Micro/Nano Fabrication, Department of Micro/Nano Electronics, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Xiang Chen
- National Key Laboratory of Science and Technology on Micro/Nano Fabrication, Department of Micro/Nano Electronics, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Lujie Song
- Department of Urology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
- Shanghai Eastern Institute of Urologic Reconstruction, Shanghai, China
| |
Collapse
|
24
|
Nanodrugs alleviate acute kidney injury: Manipulate RONS at kidney. Bioact Mater 2023; 22:141-167. [PMID: 36203963 PMCID: PMC9526023 DOI: 10.1016/j.bioactmat.2022.09.021] [Citation(s) in RCA: 38] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 08/12/2022] [Accepted: 09/19/2022] [Indexed: 02/06/2023] Open
Abstract
Currently, there are no clinical drugs available to treat acute kidney injury (AKI). Given the high prevalence and high mortality rate of AKI, the development of drugs to effectively treat AKI is a huge unmet medical need and a research hotspot. Although existing evidence fully demonstrates that reactive oxygen and nitrogen species (RONS) burst at the AKI site is a major contributor to AKI progression, the heterogeneity, complexity, and unique physiological structure of the kidney make most antioxidant and anti-inflammatory small molecule drugs ineffective because of the lack of kidney targeting and side effects. Recently, nanodrugs with intrinsic kidney targeting through the control of size, shape, and surface properties have opened exciting prospects for the treatment of AKI. Many antioxidant nanodrugs have emerged to address the limitations of current AKI treatments. In this review, we systematically summarized for the first time about the emerging nanodrugs that exploit the pathological and physiological features of the kidney to overcome the limitations of traditional small-molecule drugs to achieve high AKI efficacy. First, we analyzed the pathological structural characteristics of AKI and the main pathological mechanism of AKI: hypoxia, harmful substance accumulation-induced RONS burst at the renal site despite the multifactorial initiation and heterogeneity of AKI. Subsequently, we introduced the strategies used to improve renal targeting and reviewed advances of nanodrugs for AKI: nano-RONS-sacrificial agents, antioxidant nanozymes, and nanocarriers for antioxidants and anti-inflammatory drugs. These nanodrugs have demonstrated excellent therapeutic effects, such as greatly reducing oxidative stress damage, restoring renal function, and low side effects. Finally, we discussed the challenges and future directions for translating nanodrugs into clinical AKI treatment. AKI is a common clinical acute syndrome with high morbidity and mortality but without effective clinical drug available. Hypoxia and accumulation of toxic substances are key pathological features of various heterogeneous AKI. Excessive RONS is the core of the pathological mechanism of AKI. The development of nanodrugs is expected to achieve successful treatment in AKI.
Collapse
|
25
|
Zhang SY, Mahler GJ. A glomerulus and proximal tubule microphysiological system simulating renal filtration, reabsorption, secretion, and toxicity. LAB ON A CHIP 2023; 23:272-284. [PMID: 36514972 DOI: 10.1039/d2lc00887d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Microphysiological systems (MPS) are powerful predictive tools for assessing drug-induced kidney injuries. Previous MPS have examined single regions of the nephron, but lack simultaneous filtration, reabsorption, and secretion functionality. Here, we developed a partially open MPS that structurally and functionally recapitulated the glomerular filtration barrier, proximal tubular reabsorption, and secretion for seven days. The system introduced a recirculation circuit and an open filtrate output as a source of functional testing. As a proof-of-concept, a tri-culture of immortalized podocytes, umbilical vein endothelial cells, and proximal tubule (PCT) cells were housed in a single MPS: T-junction, glomerulus housing unit, and PCT chip. The MPS successfully retained blood serum protein, reabsorbed glucose, secreted creatinine, and expressed cell-type specific proteins (VE-cadherin, nephrin, and ZO-1). To simulate drug-induced kidney injuries, the system was perfused with cisplatin and adriamycin, and then tested using serum albumin filtration, glucose clearance, and lactate dehydrogenase release. The glomerulus and PCT MPS demonstrated a complex, dynamic microenvironment and recreated some in vivo-like functions in basal and drug-induced conditions, offering a novel prototype for preclinical testing.
Collapse
Affiliation(s)
- Stephanie Y Zhang
- Department of Biomedical Engineering, Binghamton University, PO Box 6000, Binghamton, NY, 13902, USA.
| | - Gretchen J Mahler
- Department of Biomedical Engineering, Binghamton University, PO Box 6000, Binghamton, NY, 13902, USA.
| |
Collapse
|
26
|
Muacevic A, Adler JR. Serious Multiorgan Toxicity Caused by Mixed Herbal Tea Ingestion: A Case Report. Cureus 2023; 15:e34000. [PMID: 36811058 PMCID: PMC9939026 DOI: 10.7759/cureus.34000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/19/2023] [Indexed: 01/22/2023] Open
Abstract
Recent years have witnessed a growing trend in the use of complementary and alternative herbal products. However, the ingestion of some herbal products may cause a wide spectrum of adverse effects. We report a case of multiorgan toxicity following the ingestion of mixed herbal tea. A 41-year-old woman presented to the nephrology clinic with complaints of nausea, vomiting, vaginal bleeding, and anuria. She had consumed a glass of mixed herbal tea three times a day after meals for three days, to lose weight. Initial clinical and laboratory findings showed serious multiorgan toxicity including hepatotoxicity, bone marrow toxicity, and nephrotoxicity. Although herbal preparations are marketed as natural products, they may cause various toxic effects. There should more efforts to raise public awareness about the possible toxic effects of herbal preparations. Clinicians should consider the ingestion of herbal remedies as an etiology when encountering patients with unexplained organ dysfunctions.
Collapse
|
27
|
Dorison A, Forbes TA, Little MH. What can we learn from kidney organoids? Kidney Int 2022; 102:1013-1029. [PMID: 35970244 DOI: 10.1016/j.kint.2022.06.032] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 06/15/2022] [Accepted: 06/24/2022] [Indexed: 12/14/2022]
Abstract
The ability to generate 3-dimensional models of the developing human kidney via the directed differentiation of pluripotent stem cells-termed kidney organoids-has been hailed as a major advance in experimental nephrology. Although these provide an opportunity to interrogate human development, model-specific kidney diseases facilitate drug screening and even deliver bioengineered tissue; most of these prophetic end points remain to be realized. Indeed, at present we are still finding out what we can learn and what we cannot learn from this approach. In this review, we will summarize the approaches available to generate models of the human kidney from stem cells, the existing successful applications of kidney organoids, their limitations, and remaining challenges.
Collapse
Affiliation(s)
- Aude Dorison
- Murdoch Children's Research Institute, Parkville, Melbourne, Australia; Department of Paediatrics, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, Melbourne, Australia; Novo Nordisk Foundation Centre for Stem Cell Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Thomas A Forbes
- Murdoch Children's Research Institute, Parkville, Melbourne, Australia; Department of Paediatrics, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, Melbourne, Australia; Department of Nephrology, Royal Children's Hospital, Parkville, Melbourne, Australia
| | - Melissa H Little
- Murdoch Children's Research Institute, Parkville, Melbourne, Australia; Department of Paediatrics, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, Melbourne, Australia; Novo Nordisk Foundation Centre for Stem Cell Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark.
| |
Collapse
|
28
|
Association of expression of GADD family genes and apoptosis in human kidney proximal tubular (HK-2) cells exposed to nephrotoxic drugs. Mol Cell Toxicol 2022. [DOI: 10.1007/s13273-022-00231-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
29
|
Aceves JO, Heja S, Kobayashi K, Robinson SS, Miyoshi T, Matsumoto T, Schäffers OJM, Morizane R, Lewis JA. 3D proximal tubule-on-chip model derived from kidney organoids with improved drug uptake. Sci Rep 2022; 12:14997. [PMID: 36056134 PMCID: PMC9440090 DOI: 10.1038/s41598-022-19293-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Accepted: 08/26/2022] [Indexed: 11/08/2022] Open
Abstract
Three-dimensional, organ-on-chip models that recapitulate kidney tissue are needed for drug screening and disease modeling. Here, we report a method for creating a perfusable 3D proximal tubule model composed of epithelial cells isolated from kidney organoids matured under static conditions. These organoid-derived proximal tubule epithelial cells (OPTECs) are seeded in cylindrical channels fully embedded within an extracellular matrix, where they form a confluent monolayer. A second perfusable channel is placed adjacent to each proximal tubule within these reusable multiplexed chips to mimic basolateral drug transport and uptake. Our 3D OPTEC-on-chip model exhibits significant upregulation of organic cation (OCT2) and organic anion (OAT1/3) transporters, which leads to improved drug uptake, compared to control chips based on immortalized proximal tubule epithelial cells. Hence, OPTEC tubules exhibit a higher normalized lactate dehydrogenase (LDH) release, when exposed to known nephrotoxins, cisplatin and aristolochic acid, which are diminished upon adding OCT2 and OAT1/3 transport inhibitors. Our integrated multifluidic platform paves the way for personalized kidney-on-chip models for drug screening and disease modeling.
Collapse
Affiliation(s)
- Jeffrey O Aceves
- Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - Szilvia Heja
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - Kenichi Kobayashi
- Nephrology Division, Massachusetts General Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Sanlin S Robinson
- Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - Tomoya Miyoshi
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Renal Division, Brigham and Women's Hospital, Boston, MA, USA
| | - Takuya Matsumoto
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
- Nephrology Division, Massachusetts General Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Renal Division, Brigham and Women's Hospital, Boston, MA, USA
| | - Olivier J M Schäffers
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Renal Division, Brigham and Women's Hospital, Boston, MA, USA
| | - Ryuji Morizane
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA.
- Nephrology Division, Massachusetts General Hospital, Boston, MA, USA.
- Department of Medicine, Harvard Medical School, Boston, MA, USA.
- Renal Division, Brigham and Women's Hospital, Boston, MA, USA.
| | - Jennifer A Lewis
- Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA.
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA.
| |
Collapse
|
30
|
Phytochemical Classification of Medicinal Plants Used in the Treatment of Kidney Disease Based on Traditional Persian Medicine. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:8022599. [PMID: 35958915 PMCID: PMC9357710 DOI: 10.1155/2022/8022599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/19/2022] [Revised: 05/09/2022] [Accepted: 07/01/2022] [Indexed: 12/02/2022]
Abstract
Methods This review was focused on studying the various secondary metabolites in model plants of Iranian herbal medicine known as treatment of kidney diseases in traditional Persian medicine textbooks including Makhzan-ol-Advieh, The Canon of Medicine, and Taghvim al-Abdan fi Tadbir al-Ensan. Results Secondary metabolites of 94 medical plants belonging to 42 families were reported with their scientific and family name. Conclusion Although herbal medicines are gaining rapid popularity among people and the pharmaceutical industry, the understandings of the phytochemical and therapeutic properties of medicinal plant are important for developing effective nephroprotective medicines. Therefore, the relationship between traditional uses and biological properties should be clearly verified through further studies.
Collapse
|
31
|
Wang Q, Lu J, Fan K, Xu Y, Xiong Y, Sun Z, Zhai M, Zhang Z, Zhang S, Song Y, Luo J, You M, Guo M, Zhang X. High-throughput "read-on-ski" automated imaging and label-free detection system for toxicity screening of compounds using personalised human kidney organoids. J Zhejiang Univ Sci B 2022; 23:564-577. [PMID: 35794686 DOI: 10.1631/jzus.b2100701] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Organoid models are used to study kidney physiology, such as the assessment of nephrotoxicity and underlying disease processes. Personalized human pluripotent stem cell-derived kidney organoids are ideal models for compound toxicity studies, but there is a need to accelerate basic and translational research in the field. Here, we developed an automated continuous imaging setup with the "read-on-ski" law of control to maximize temporal resolution with minimum culture plate vibration. High-accuracy performance was achieved: organoid screening and imaging were performed at a spatial resolution of 1.1 μm for the entire multi-well plate under 3 min. We used the in-house developed multi-well spinning device and cisplatin-induced nephrotoxicity model to evaluate the toxicity in kidney organoids using this system. The acquired images were processed via machine learning-based classification and segmentation algorithms, and the toxicity in kidney organoids was determined with 95% accuracy. The results obtained by the automated "read-on-ski" imaging device, combined with label-free and non-invasive algorithms for detection, were verified using conventional biological procedures. Taking advantage of the close-to-in vivo-kidney organoid model, this new development opens the door for further application of scaled-up screening using organoids in basic research and drug discovery.
Collapse
Affiliation(s)
- Qizheng Wang
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Jun Lu
- Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China.,Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou 510320, China
| | - Ke Fan
- Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
| | - Yiwei Xu
- Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
| | - Yucui Xiong
- Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
| | - Zhiyong Sun
- Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China.,Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou 510320, China
| | - Man Zhai
- Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
| | - Zhizhong Zhang
- Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China.,Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou 510320, China
| | - Sheng Zhang
- Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
| | - Yan Song
- Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
| | - Jianzhong Luo
- Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
| | - Mingliang You
- Hangzhou Cancer Institute, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou Cancer Hospital, Zhejiang University School of Medicine, Hangzhou 310002, China
| | - Meijin Guo
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, China. ,
| | - Xiao Zhang
- Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China. .,Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou 510320, China.
| |
Collapse
|
32
|
Piossek F, Beneke S, Schlichenmaier N, Mucic G, Drewitz S, Dietrich DR. Physiological oxygen and co-culture with human fibroblasts facilitate in vivo-like properties in human renal proximal tubular epithelial cells. Chem Biol Interact 2022; 361:109959. [DOI: 10.1016/j.cbi.2022.109959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 04/08/2022] [Accepted: 04/12/2022] [Indexed: 11/28/2022]
|
33
|
Galateanu B, Hudita A, Biru EI, Iovu H, Zaharia C, Simsensohn E, Costache M, Petca RC, Jinga V. Applications of Polymers for Organ-on-Chip Technology in Urology. Polymers (Basel) 2022; 14:1668. [PMID: 35566836 PMCID: PMC9105302 DOI: 10.3390/polym14091668] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Revised: 04/05/2022] [Accepted: 04/18/2022] [Indexed: 02/07/2023] Open
Abstract
Organ-on-chips (OOCs) are microfluidic devices used for creating physiological organ biomimetic systems. OOC technology brings numerous advantages in the current landscape of preclinical models, capable of recapitulating the multicellular assemblage, tissue-tissue interaction, and replicating numerous human pathologies. Moreover, in cancer research, OOCs emulate the 3D hierarchical complexity of in vivo tumors and mimic the tumor microenvironment, being a practical cost-efficient solution for tumor-growth investigation and anticancer drug screening. OOCs are compact and easy-to-use microphysiological functional units that recapitulate the native function and the mechanical strain that the cells experience in the human bodies, allowing the development of a wide range of applications such as disease modeling or even the development of diagnostic devices. In this context, the current work aims to review the scientific literature in the field of microfluidic devices designed for urology applications in terms of OOC fabrication (principles of manufacture and materials used), development of kidney-on-chip models for drug-toxicity screening and kidney tumors modeling, bladder-on-chip models for urinary tract infections and bladder cancer modeling and prostate-on-chip models for prostate cancer modeling.
Collapse
Affiliation(s)
- Bianca Galateanu
- Department of Biochemistry and Molecular Biology, University of Bucharest, 91-95 Splaiul Independentei Street, 050095 Bucharest, Romania; (B.G.); (M.C.)
| | - Ariana Hudita
- Department of Biochemistry and Molecular Biology, University of Bucharest, 91-95 Splaiul Independentei Street, 050095 Bucharest, Romania; (B.G.); (M.C.)
| | - Elena Iuliana Biru
- Advanced Polymer Materials Group, Department of Bioresources and Polymer Science, University Politehnica of Bucharest, 1-7 Gh. Polizu Street, 011061 Bucharest, Romania; (H.I.); (C.Z.)
| | - Horia Iovu
- Advanced Polymer Materials Group, Department of Bioresources and Polymer Science, University Politehnica of Bucharest, 1-7 Gh. Polizu Street, 011061 Bucharest, Romania; (H.I.); (C.Z.)
- Academy of Romanian Scientists, Ilfov Street, 50044 Bucharest, Romania
| | - Catalin Zaharia
- Advanced Polymer Materials Group, Department of Bioresources and Polymer Science, University Politehnica of Bucharest, 1-7 Gh. Polizu Street, 011061 Bucharest, Romania; (H.I.); (C.Z.)
| | - Eliza Simsensohn
- “Carol Davila” University of Medicine and Pharmacy Bucharest, 050474 Bucharest, Romania; (E.S.); (R.-C.P.); (V.J.)
| | - Marieta Costache
- Department of Biochemistry and Molecular Biology, University of Bucharest, 91-95 Splaiul Independentei Street, 050095 Bucharest, Romania; (B.G.); (M.C.)
| | - Razvan-Cosmin Petca
- “Carol Davila” University of Medicine and Pharmacy Bucharest, 050474 Bucharest, Romania; (E.S.); (R.-C.P.); (V.J.)
| | - Viorel Jinga
- “Carol Davila” University of Medicine and Pharmacy Bucharest, 050474 Bucharest, Romania; (E.S.); (R.-C.P.); (V.J.)
| |
Collapse
|
34
|
Becerir T, Tokgün O, İnci K, Girişgen İ, Yuksel S. Therapeutic Effect of Teneligliptin in Drug-Induced Nephrotoxicity: An In-Vitro Study. Cureus 2022; 14:e23871. [PMID: 35530894 PMCID: PMC9074376 DOI: 10.7759/cureus.23871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/06/2022] [Indexed: 11/23/2022] Open
Abstract
Background Drug-induced nephrotoxicity is an important side effect of many commonly used drugs. In this study, we planned to evaluate the effects of teneligliptin (TG), which is a dipeptidyl peptidase-4 (DPP-4) inhibitor, on cell healing by creating nephrotoxicity models in human renal proximal tubule cell and human embryonic kidney epithelial cells cell lines in-vitro with cisplatin, vancomycin, and gentamicin. Methodology First, we determined the 50% inhibitory concentration doses of nephrotoxic drugs and the nephroprotective dose of TG. Then, we analyzed the difference in cell viability, apoptosis, and oxidative stress (reactive oxygen and nitrogen species (ROS/RNS) production) between TG-treated and untreated cells after nephrotoxicity occurred. Moreover, we evaluated the expression of kidney injury molecule-1 (KIM-1) and neutrophil gelatinase-associated lipocalin (NGAL) in cells. Results We found that when cell lines were treated after toxicity was induced with TG, cell viability increased, apoptosis and ROS/RNS production were significantly decreased, and expressions of KIM-1 and NGAL were significantly reduced. Conclusions This study showed that TG has positive effects on the recovery of drug-induced nephrotoxicity in an in-vitro setting.
Collapse
|
35
|
Ma K, Bai Y, Li J, Ren Z, Li J, Zhang J, Shan A. Lactobacillus rhamnosus GG ameliorates deoxynivalenol-induced kidney oxidative damage and mitochondrial injury in weaned piglets. Food Funct 2022; 13:3905-3916. [PMID: 35285834 DOI: 10.1039/d2fo00185c] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Deoxynivalenol (DON) is a common mycotoxin that pollutes food crops and adversely affects the health of animals, even humans. Lactobacillus rhamnosus GG (LGG) can alleviate intestinal injury, and anti-inflammatory and antioxidant effects. However, the potential of LGG in alleviating kidney injury induced by DON in piglets remains to be studied. The objective of this study was to investigate the adverse effect of DON on kidney injury and the protective ability of LGG. A total of twenty-seven weaned piglets were divided into three groups: CON group, DON group (3.11 mg kg-1 feed) and LGG + DON group (LGG powder 1 g kg-1 + DON 3.15 mg kg-1 feed). DON increased the MDA content, and decreased antioxidant enzyme activity (GSH-Px) and total antioxidant capacity (P < 0.05). Meanwhile, DON activated the Nrf2 antioxidant pathway. However, LGG supplementation alleviated the damage of DON to the kidney antioxidant system of piglets. Notably, DON significantly reduced the Sirt3 expression (P < 0.05), which was alleviated by LGG addition. The expression of mitochondrial biogenesis related factors such as VDAC1 and Cyt C was up-regulated by DON (P < 0.05), and LGG could improve mitochondrial ultrastructural abnormalities and mitochondrial dysfunction. In addition, LGG mitigated DON-induced mitochondrial fusion inhibition, and prevented DON-mediated mitochondrial autophagy. In conclusion, LGG play a protective role in DON-induced kidney toxicity, and dietary intervention may be a strategy to reduce mycotoxins.
Collapse
Affiliation(s)
- Kaidi Ma
- Institute of Animal Nutrition, Northeast Agricultural University, No. 600, Changjiang Road, Harbin 150030, P. R. China.
| | - Yongsong Bai
- Institute of Animal Nutrition, Northeast Agricultural University, No. 600, Changjiang Road, Harbin 150030, P. R. China.
| | - Jibo Li
- Institute of Animal Nutrition, Northeast Agricultural University, No. 600, Changjiang Road, Harbin 150030, P. R. China.
| | - Zhongshuai Ren
- College of Animal Science, Jilin University, Changchun 130062, P. R. China.
| | - Jianping Li
- Institute of Animal Nutrition, Northeast Agricultural University, No. 600, Changjiang Road, Harbin 150030, P. R. China.
| | - Jing Zhang
- College of Animal Science, Jilin University, Changchun 130062, P. R. China.
| | - Anshan Shan
- Institute of Animal Nutrition, Northeast Agricultural University, No. 600, Changjiang Road, Harbin 150030, P. R. China.
| |
Collapse
|
36
|
Bejoy J, Qian ES, Woodard LE. Tissue Culture Models of AKI: From Tubule Cells to Human Kidney Organoids. J Am Soc Nephrol 2022; 33:487-501. [PMID: 35031569 PMCID: PMC8975068 DOI: 10.1681/asn.2021050693] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
AKI affects approximately 13.3 million people around the world each year, causing CKD and/or mortality. The mammalian kidney cannot generate new nephrons after postnatal renal damage and regenerative therapies for AKI are not available. Human kidney tissue culture systems can complement animal models of AKI and/or address some of their limitations. Donor-derived somatic cells, such as renal tubule epithelial cells or cell lines (RPTEC/hTERT, ciPTEC, HK-2, Nki-2, and CIHP-1), have been used for decades to permit drug toxicity screening and studies into potential AKI mechanisms. However, tubule cell lines do not fully recapitulate tubular epithelial cell properties in situ when grown under classic tissue culture conditions. Improving tissue culture models of AKI would increase our understanding of the mechanisms, leading to new therapeutics. Human pluripotent stem cells (hPSCs) can be differentiated into kidney organoids and various renal cell types. Injury to human kidney organoids results in renal cell-type crosstalk and upregulation of kidney injury biomarkers that are difficult to induce in primary tubule cell cultures. However, current protocols produce kidney organoids that are not mature and contain off-target cell types. Promising bioengineering techniques, such as bioprinting and "kidney-on-a-chip" methods, as applied to kidney nephrotoxicity modeling advantages and limitations are discussed. This review explores the mechanisms and detection of AKI in tissue culture, with an emphasis on bioengineered approaches such as human kidney organoid models.
Collapse
Affiliation(s)
- Julie Bejoy
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Eddie S. Qian
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Lauren E. Woodard
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee,Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville, Tennessee,Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee
| |
Collapse
|
37
|
Youhanna S, Kemas AM, Preiss L, Zhou Y, Shen JX, Cakal SD, Paqualini FS, Goparaju SK, Shafagh RZ, Lind JU, Sellgren CM, Lauschke VM. Organotypic and Microphysiological Human Tissue Models for Drug Discovery and Development-Current State-of-the-Art and Future Perspectives. Pharmacol Rev 2022; 74:141-206. [PMID: 35017176 DOI: 10.1124/pharmrev.120.000238] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 10/12/2021] [Indexed: 12/11/2022] Open
Abstract
The number of successful drug development projects has been stagnant for decades despite major breakthroughs in chemistry, molecular biology, and genetics. Unreliable target identification and poor translatability of preclinical models have been identified as major causes of failure. To improve predictions of clinical efficacy and safety, interest has shifted to three-dimensional culture methods in which human cells can retain many physiologically and functionally relevant phenotypes for extended periods of time. Here, we review the state of the art of available organotypic culture techniques and critically review emerging models of human tissues with key importance for pharmacokinetics, pharmacodynamics, and toxicity. In addition, developments in bioprinting and microfluidic multiorgan cultures to emulate systemic drug disposition are summarized. We close by highlighting important trends regarding the fabrication of organotypic culture platforms and the choice of platform material to limit drug absorption and polymer leaching while supporting the phenotypic maintenance of cultured cells and allowing for scalable device fabrication. We conclude that organotypic and microphysiological human tissue models constitute promising systems to promote drug discovery and development by facilitating drug target identification and improving the preclinical evaluation of drug toxicity and pharmacokinetics. There is, however, a critical need for further validation, benchmarking, and consolidation efforts ideally conducted in intersectoral multicenter settings to accelerate acceptance of these novel models as reliable tools for translational pharmacology and toxicology. SIGNIFICANCE STATEMENT: Organotypic and microphysiological culture of human cells has emerged as a promising tool for preclinical drug discovery and development that might be able to narrow the translation gap. This review discusses recent technological and methodological advancements and the use of these systems for hit discovery and the evaluation of toxicity, clearance, and absorption of lead compounds.
Collapse
Affiliation(s)
- Sonia Youhanna
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (S.Y., A.M.K., L.P., Y.Z., J.X.S., S.K.G., R.Z.S., C.M.S., V.M.L.); Department of Drug Metabolism and Pharmacokinetics (DMPK), Merck KGaA, Darmstadt, Germany (L.P.); Department of Health Technology, Technical University of Denmark, Lyngby, Denmark (S.D.C., J.U.L.); Synthetic Physiology Laboratory, Department of Civil Engineering and Architecture, University of Pavia, Pavia, Italy (F.S.P.); Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden (Z.S.); and Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.)
| | - Aurino M Kemas
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (S.Y., A.M.K., L.P., Y.Z., J.X.S., S.K.G., R.Z.S., C.M.S., V.M.L.); Department of Drug Metabolism and Pharmacokinetics (DMPK), Merck KGaA, Darmstadt, Germany (L.P.); Department of Health Technology, Technical University of Denmark, Lyngby, Denmark (S.D.C., J.U.L.); Synthetic Physiology Laboratory, Department of Civil Engineering and Architecture, University of Pavia, Pavia, Italy (F.S.P.); Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden (Z.S.); and Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.)
| | - Lena Preiss
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (S.Y., A.M.K., L.P., Y.Z., J.X.S., S.K.G., R.Z.S., C.M.S., V.M.L.); Department of Drug Metabolism and Pharmacokinetics (DMPK), Merck KGaA, Darmstadt, Germany (L.P.); Department of Health Technology, Technical University of Denmark, Lyngby, Denmark (S.D.C., J.U.L.); Synthetic Physiology Laboratory, Department of Civil Engineering and Architecture, University of Pavia, Pavia, Italy (F.S.P.); Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden (Z.S.); and Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.)
| | - Yitian Zhou
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (S.Y., A.M.K., L.P., Y.Z., J.X.S., S.K.G., R.Z.S., C.M.S., V.M.L.); Department of Drug Metabolism and Pharmacokinetics (DMPK), Merck KGaA, Darmstadt, Germany (L.P.); Department of Health Technology, Technical University of Denmark, Lyngby, Denmark (S.D.C., J.U.L.); Synthetic Physiology Laboratory, Department of Civil Engineering and Architecture, University of Pavia, Pavia, Italy (F.S.P.); Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden (Z.S.); and Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.)
| | - Joanne X Shen
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (S.Y., A.M.K., L.P., Y.Z., J.X.S., S.K.G., R.Z.S., C.M.S., V.M.L.); Department of Drug Metabolism and Pharmacokinetics (DMPK), Merck KGaA, Darmstadt, Germany (L.P.); Department of Health Technology, Technical University of Denmark, Lyngby, Denmark (S.D.C., J.U.L.); Synthetic Physiology Laboratory, Department of Civil Engineering and Architecture, University of Pavia, Pavia, Italy (F.S.P.); Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden (Z.S.); and Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.)
| | - Selgin D Cakal
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (S.Y., A.M.K., L.P., Y.Z., J.X.S., S.K.G., R.Z.S., C.M.S., V.M.L.); Department of Drug Metabolism and Pharmacokinetics (DMPK), Merck KGaA, Darmstadt, Germany (L.P.); Department of Health Technology, Technical University of Denmark, Lyngby, Denmark (S.D.C., J.U.L.); Synthetic Physiology Laboratory, Department of Civil Engineering and Architecture, University of Pavia, Pavia, Italy (F.S.P.); Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden (Z.S.); and Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.)
| | - Francesco S Paqualini
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (S.Y., A.M.K., L.P., Y.Z., J.X.S., S.K.G., R.Z.S., C.M.S., V.M.L.); Department of Drug Metabolism and Pharmacokinetics (DMPK), Merck KGaA, Darmstadt, Germany (L.P.); Department of Health Technology, Technical University of Denmark, Lyngby, Denmark (S.D.C., J.U.L.); Synthetic Physiology Laboratory, Department of Civil Engineering and Architecture, University of Pavia, Pavia, Italy (F.S.P.); Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden (Z.S.); and Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.)
| | - Sravan K Goparaju
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (S.Y., A.M.K., L.P., Y.Z., J.X.S., S.K.G., R.Z.S., C.M.S., V.M.L.); Department of Drug Metabolism and Pharmacokinetics (DMPK), Merck KGaA, Darmstadt, Germany (L.P.); Department of Health Technology, Technical University of Denmark, Lyngby, Denmark (S.D.C., J.U.L.); Synthetic Physiology Laboratory, Department of Civil Engineering and Architecture, University of Pavia, Pavia, Italy (F.S.P.); Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden (Z.S.); and Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.)
| | - Reza Zandi Shafagh
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (S.Y., A.M.K., L.P., Y.Z., J.X.S., S.K.G., R.Z.S., C.M.S., V.M.L.); Department of Drug Metabolism and Pharmacokinetics (DMPK), Merck KGaA, Darmstadt, Germany (L.P.); Department of Health Technology, Technical University of Denmark, Lyngby, Denmark (S.D.C., J.U.L.); Synthetic Physiology Laboratory, Department of Civil Engineering and Architecture, University of Pavia, Pavia, Italy (F.S.P.); Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden (Z.S.); and Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.)
| | - Johan Ulrik Lind
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (S.Y., A.M.K., L.P., Y.Z., J.X.S., S.K.G., R.Z.S., C.M.S., V.M.L.); Department of Drug Metabolism and Pharmacokinetics (DMPK), Merck KGaA, Darmstadt, Germany (L.P.); Department of Health Technology, Technical University of Denmark, Lyngby, Denmark (S.D.C., J.U.L.); Synthetic Physiology Laboratory, Department of Civil Engineering and Architecture, University of Pavia, Pavia, Italy (F.S.P.); Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden (Z.S.); and Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.)
| | - Carl M Sellgren
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (S.Y., A.M.K., L.P., Y.Z., J.X.S., S.K.G., R.Z.S., C.M.S., V.M.L.); Department of Drug Metabolism and Pharmacokinetics (DMPK), Merck KGaA, Darmstadt, Germany (L.P.); Department of Health Technology, Technical University of Denmark, Lyngby, Denmark (S.D.C., J.U.L.); Synthetic Physiology Laboratory, Department of Civil Engineering and Architecture, University of Pavia, Pavia, Italy (F.S.P.); Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden (Z.S.); and Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.)
| | - Volker M Lauschke
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (S.Y., A.M.K., L.P., Y.Z., J.X.S., S.K.G., R.Z.S., C.M.S., V.M.L.); Department of Drug Metabolism and Pharmacokinetics (DMPK), Merck KGaA, Darmstadt, Germany (L.P.); Department of Health Technology, Technical University of Denmark, Lyngby, Denmark (S.D.C., J.U.L.); Synthetic Physiology Laboratory, Department of Civil Engineering and Architecture, University of Pavia, Pavia, Italy (F.S.P.); Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden (Z.S.); and Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.)
| |
Collapse
|
38
|
Novel PHD2/HDACs hybrid inhibitors protect against cisplatin-induced acute kidney injury. Eur J Med Chem 2022; 230:114115. [PMID: 35033824 DOI: 10.1016/j.ejmech.2022.114115] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 12/21/2021] [Accepted: 01/09/2022] [Indexed: 12/15/2022]
Abstract
Acute kidney injury (AKI) is associated with high morbidity and mortality. Cisplatin is a common chemotherapeutic, but its nephrotoxicity-driven AKI limits its clinical application. Currently, there are no specific and satisfactory therapies in the clinic for AKI. Inhibitors of hypoxia-inducible factor prolyl hydroxylase 2 (HIF-PHD2) or histone deacetylase (HDACs) had shown renoprotective effects against AKI in preclinical studies. This study aimed to develop a novel therapeutic to prevent AKI progression by targeting PHD2 and HDACs simultaneously. We designed and synthesized a series of PHD2/HDACs hybrid inhibitors. The initial drug activity screening identified a candidate compound 31c, which exhibited potent inhibitory activities against PHD2 and HDAC1/2/6. Cellular analyses further showed that 31c did not affect cisplatin's antitumor activity in cancer cells but strongly protected cisplatin-induced toxicity on HK-2 cells. In vivo studies with the cisplatin-induced AKI mouse model demonstrated that 31c remarkably alleviated kidney dysfunction with suppressed plasma BUN/SCr and increased EPO levels. The potent renoprotective effects of 31c on AKI were confirmed by significant improvements in pathological kidney conditions in the mouse model. These results suggest that the novel PHD2/HDACs hybrid inhibitor, 31c, has a clinical potential as the renoprotective agent for the treatment/prevention of cisplatin-induced AKI for various cancers.
Collapse
|
39
|
Kim HR, Park JH, Lee SH, Kwack SJ, Lee J, Kim S, Yoon S, Kim KB, Lee BM, Kacew S, Kim HS. Using intracellular metabolic profiling to identify novel biomarkers of cisplatin-induced acute kidney injury in NRK-52E cells. JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART A 2022; 85:29-42. [PMID: 34445936 DOI: 10.1080/15287394.2021.1969305] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
The aim of this study was to investigate changes in the intracellular metabolism resulting from cisplatin (CDDP)-induced nephrotoxicity in normal kidney tubular epithelial NRK-52E cells. Cytotoxicity, cell cycle analysis, and apoptotic cell death were all evaluated in NRK-52E cells treated with CDDP. Subsequently, proton nuclear magnetic resonance (1H-NMR) spectroscopy was used to investigate cellular metabolic profiles. CDDP-induced nephrotoxicity was determined in vivo model. Cytotoxicity in the NRK-52E cells significantly rose following treatment with CDDP and these increases were found to be concentration-dependent. Both p53 and Bax protein expression was increased in CDDP-treated NRK-52E cells, correlating with enhanced cellular apoptosis. In addition, a number of metabolites were altered in both media and cell lysates in these cells. In cell lysates, citrate, creatinine, and acetate levels were dramatically reduced following treatment with 20 µM CDDP concentrations, while glutamate level was elevated. Lactate and acetate levels were significantly increased in culture media but citrate concentrations were reduced following high 20 µM CDDP concentrations incubation. In addition, excretion of clusterin, calbindin, neutrophil gelatinase-associated lipocalin (NGAL), kidney injury molecule-1 (KIM-1), selenium binding protein 1 (SBP1), and pyruvate kinase M2 (PKM2) into the culture media was significantly increased in CDDP-treated cells while expression of acetyl CoA synthetase 1 (AceCS1) was markedly reduced in these cells. These findings suggest that acetate-dependent metabolic pathway may be a reliable and useful biomarker for detecting CDDP-induced nephrotoxicity. Taken together, data demonstrate that the discovery of novel biomarkers by metabolite profiling in target cells may contribute to the detection of nephrotoxicity and new drug development.
Collapse
Affiliation(s)
- Hae Ri Kim
- Division of Toxicology, School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
| | - Jae Hyeon Park
- Division of Toxicology, School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
| | - Song Hee Lee
- Division of Toxicology, School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
| | - Seung Jun Kwack
- Department of Biochemistry and Health Science, Changwon National University, Gyeongnam, Republic of Korea
| | - Jaewon Lee
- Department of Neuroscience, College of Pharmacy, Pusan National University, Busan, Republic of Korea
| | - Suhkmann Kim
- Department of Chemistry and Chemistry Institute of Functional Materials, Pusan National University, Busan, Republic of Korea
| | - Sungpil Yoon
- Division of Toxicology, School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
| | - Kyu-Bong Kim
- Department of Toxicology, College of Pharmacy, Dankook University, Chungnam, Republic of Korea
| | - Byung Mu Lee
- Division of Toxicology, School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
| | - Sam Kacew
- Department of Cellular and Molecular Medicine, McLaughlin Centre for Population Health Risk Assessment, University of Ottawa, Ottawa, Canada
| | - Hyung Sik Kim
- Division of Toxicology, School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
| |
Collapse
|
40
|
Tragiannidis A, Gkampeta A, Vousvouki M, Vasileiou E, Groll AH. Antifungal agents and the kidney: pharmacokinetics, clinical nephrotoxicity, and interactions. Expert Opin Drug Saf 2021; 20:1061-1074. [PMID: 33896310 DOI: 10.1080/14740338.2021.1922667] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
INTRODUCTION Invasive fungal infections continue to be important causes of morbidity and mortality in severely ill and immunocompromised patient populations. The past three decades have seen a considerable expansion in antifungal drug research, resulting in the clinical development of different classes of antifungal agents with different pharmacologic properties. Among drug-specific characteristics of antifungal agents, renal disposition and nephrotoxicity are important clinical considerations as many patients requiring antifungal therapy have compromised organ functions or are receiving other potentially nephrotoxic medications. AREAS COVERED The present article reviews incidence, severity and mechanisms of nephrotoxicity associated with antifungal agents used for prevention and treatment of invasive fungal diseases by discussing distribution, metabolism, elimination and drug-related adverse events in the context of safety data from phase II and III clinical studies. EXPERT OPINION Based on the available data amphotericin B deoxycholate has the highest relative potential for nephrotoxicity, followed by the lipid formulations of amphotericin B, and, to a much lesser extent and by indirect mechanisms, the antifungal triazoles.
Collapse
Affiliation(s)
- Athanasios Tragiannidis
- Childhood & Adolescent Hematology Oncology Unit, 2nd Pediatric Department, Faculty of Health Sciences, Aristotle University of Thessaloniki, AHEPA Hospital, Greece
- Infectious Disease Research Program, Center for Bone Marrow Transplantation and Department of Pediatric Hematology/Oncology, University Children's Hospital Münster, Münster, Germany
| | - Anastasia Gkampeta
- Childhood & Adolescent Hematology Oncology Unit, 2nd Pediatric Department, Faculty of Health Sciences, Aristotle University of Thessaloniki, AHEPA Hospital, Greece
| | - Maria Vousvouki
- Childhood & Adolescent Hematology Oncology Unit, 2nd Pediatric Department, Faculty of Health Sciences, Aristotle University of Thessaloniki, AHEPA Hospital, Greece
| | - Eleni Vasileiou
- Childhood & Adolescent Hematology Oncology Unit, 2nd Pediatric Department, Faculty of Health Sciences, Aristotle University of Thessaloniki, AHEPA Hospital, Greece
| | - Andreas H Groll
- Infectious Disease Research Program, Center for Bone Marrow Transplantation and Department of Pediatric Hematology/Oncology, University Children's Hospital Münster, Münster, Germany
| |
Collapse
|
41
|
Schoch S, Sen V, Brenner W, Hartwig A, Köberle B. In Vitro Nephrotoxicity Studies of Established and Experimental Platinum-Based Compounds. Biomedicines 2021; 9:biomedicines9081033. [PMID: 34440237 PMCID: PMC8394219 DOI: 10.3390/biomedicines9081033] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 08/12/2021] [Accepted: 08/13/2021] [Indexed: 11/16/2022] Open
Abstract
Cisplatin is one of the most commonly used drugs for the treatment of various solid cancers. However, its efficacy is restricted by severe side effects, especially dose-limiting nephrotoxicity. New platinum-based compounds are designed to overcome this limitation. Previous investigations showed that the platinum(IV)–nitroxyl complex PN149 is highly cytotoxic in various tumor cell lines. In the present study, investigations with PN149 were extended to normal human kidney tubule epithelia. Coincident with higher intracellular platinum accumulation, the cytotoxicity of PN149 in the proximal tubule epithelial cell line ciPTEC was more pronounced compared to the established platinum chemotherapeutics cisplatin, carboplatin and oxaliplatin. Quantitative gene expression profiling revealed the induction of ROS-inducible and anti-oxidative genes, suggesting an oxidative stress response by PN149. However, in contrast to cisplatin, no pro-inflammatory response was observed. Genes coding for distinct DNA damage response factors and genes related to apoptosis were up-regulated, indicating the activation of the DNA damage response system and induction of the apoptotic cascade by PN149. Altogether, a comparable transcriptional response was observed for PN149 and the platinum chemotherapeutics. However, the lack of inflammatory activity, which is a possible cause contributing to toxicity in human renal proximal tubule epithelia, might indicate the reduced nephrotoxic potential of PN149.
Collapse
Affiliation(s)
- Sarah Schoch
- Department of Laboratory Medicine, Lund University, Scheelevägen 2, 223 81 Lund, Sweden;
| | - Vasily Sen
- Institute of Problems of Chemical Physics, Russian Academy of Sciences, Chernogolovka, 142432 Moscow, Russia;
| | - Walburgis Brenner
- Clinic for Obstetrics and Women’s Health, Department of Urology and Pediatric Urology, University Medical Center Mainz, Langenbeckstreet 1, 55131 Mainz, Germany;
| | - Andrea Hartwig
- Karlsruhe Institute of Technology, Department of Food Chemistry and Toxicology, Adenauerring 20, 76131 Karlsruhe, Germany;
| | - Beate Köberle
- Karlsruhe Institute of Technology, Department of Food Chemistry and Toxicology, Adenauerring 20, 76131 Karlsruhe, Germany;
- Correspondence: ; Tel.: +49-721-608-42933
| |
Collapse
|
42
|
Mukherjee K, Chio TI, Gu H, Sackett DL, Bane SL, Sever S. A Novel Fluorogenic Assay for the Detection of Nephrotoxin-Induced Oxidative Stress in Live Cells and Renal Tissue. ACS Sens 2021; 6:2523-2528. [PMID: 34214393 PMCID: PMC8314269 DOI: 10.1021/acssensors.1c00422] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
![]()
Drug-induced kidney
injury frequently leads to aborted clinical
trials and drug withdrawals. Sufficiently sensitive sensors capable
of detecting mild signs of chemical insult in cell-based screening
assays are critical to identifying and eliminating potential toxins
in the preclinical stage. Oxidative stress is a common early manifestation
of chemical toxicity, and biomolecule carbonylation is an irreversible
repercussion of oxidative stress. Here, we present a novel fluorogenic
assay using a sensor, TFCH, that responds to biomolecule carbonylation
and efficiently detects modest forms of renal injury with much greater
sensitivity than standard assays for nephrotoxins. We demonstrate
that this sensor can be deployed in live kidney cells and in renal
tissue. Our robust assay may help inform preclinical decisions to
recall unsafe drug candidates. The application of this sensor in identifying
and analyzing diverse pathologies is envisioned.
Collapse
Affiliation(s)
- Kamalika Mukherjee
- Division of Nephrology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02114, United States
| | - Tak Ian Chio
- Department of Chemistry, Binghamton University, State University of New York, Binghamton, New York 13902, United States
| | - Han Gu
- Department of Chemistry, Binghamton University, State University of New York, Binghamton, New York 13902, United States
| | - Dan L. Sackett
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Susan L. Bane
- Department of Chemistry, Binghamton University, State University of New York, Binghamton, New York 13902, United States
| | - Sanja Sever
- Division of Nephrology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02114, United States
| |
Collapse
|
43
|
Shi HH, Wang CC, Ding L, Mao XZ, Xue CH, Yanagita T, Zhang TT, Wang YM. Comparative evaluation of phosphatidylcholine and phosphatidylserine with different fatty acids on nephrotoxicity in vancomycin-induced mice. Biosci Biotechnol Biochem 2021; 85:1873-1884. [PMID: 34196365 DOI: 10.1093/bbb/zbab105] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Accepted: 06/01/2021] [Indexed: 01/06/2023]
Abstract
Phospholipids reportedly alleviate drug-induced acute kidney injury. However, no study has compared the effect of phospholipids with different fatty acids and polar heads on drug-induced nephrotoxicity. In the present study, we aimed to compare the possible nephroprotection afforded by phosphatidylcholine and phosphatidylserine with different fatty acids in a mouse model of vancomycin-induced nephrotoxicity. Pretreatment with phospholipids rich in docosahexaenoic acid (DHA) or eicosapentaenoic acid (EPA) doubled the survival time when compared with the model group. Moreover, phospholipids rich in DHA/EPA significantly reduced the serum levels of renal function biomarkers and ameliorated kidney pathologies. In terms of alleviating renal damage, no significant differences were observed between different polar heads in DHA-enriched phospholipids, while phosphatidylserine from soybean was better than phosphatidylcholine in mitigating renal injury. Furthermore, DHA/EPA-enriched phospholipids inhibited vancomycin-induced nephrotoxicity mainly by inhibiting apoptosis and oxidative stress. These results provide a scientific basis for phospholipids as potential ingredients to prevent acute kidney injury.
Collapse
Affiliation(s)
- Hao-Hao Shi
- College of Food Science and Engineering, Ocean University of China, Qingdao, Shandong, China
| | - Cheng-Cheng Wang
- College of Food Science and Engineering, Ocean University of China, Qingdao, Shandong, China
| | - Lin Ding
- College of Food Science and Engineering, Ocean University of China, Qingdao, Shandong, China
| | - Xiang-Zhao Mao
- College of Food Science and Engineering, Ocean University of China, Qingdao, Shandong, China
| | - Chang-Hu Xue
- College of Food Science and Engineering, Ocean University of China, Qingdao, Shandong, China.,Laboratory for Marine Drugs and Bioproducts, Pilot National Laboratory for Marine Science and Technology (Qingdao), Qingdao, Shandong Province, P. R. China
| | - Teruyoshi Yanagita
- Laboratory of Nutrition Biochemistry, Department of Applied Biochemistry and Food Science, Saga University, Saga, Japan
| | - Tian-Tian Zhang
- College of Food Science and Engineering, Ocean University of China, Qingdao, Shandong, China
| | - Yu-Ming Wang
- College of Food Science and Engineering, Ocean University of China, Qingdao, Shandong, China.,Laboratory for Marine Drugs and Bioproducts, Pilot National Laboratory for Marine Science and Technology (Qingdao), Qingdao, Shandong Province, P. R. China
| |
Collapse
|
44
|
Protective effects of Cinnamomum zeylanicum and Zingiber officinale extract against CCl4-induced acute kidney injury in rats. PHYSIOLOGY AND PHARMACOLOGY 2021. [DOI: 10.52547/phypha.26.2.6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|
45
|
Effective exposure of chemicals in in vitro cell systems: A review of chemical distribution models. Toxicol In Vitro 2021; 73:105133. [DOI: 10.1016/j.tiv.2021.105133] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 02/11/2021] [Accepted: 02/25/2021] [Indexed: 12/23/2022]
|
46
|
Yu XA, Bai X, Zhang R, Zhang Y, Hu Y, Lu M, Yu BY, Liu S, Tian J. A nanosensor for precise discrimination of nephrotoxic drug mechanisms via dynamic fluorescence fingerprint strategy. Anal Chim Acta 2021; 1160:338447. [PMID: 33894967 DOI: 10.1016/j.aca.2021.338447] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 03/19/2021] [Accepted: 03/20/2021] [Indexed: 11/18/2022]
Abstract
Drug-induced kidney injury causes structural or functional abnormalities of kidney, seriously affecting clinical practice and drug discovery. However, rapid and effective identification of nephrotoxic drug mechanisms is yet a challenging task arising from the complexity and diversity of various nephrotoxic mechanisms. Herein, we have constructed a polydopamine-polyethyleneimine/quantum dots sensor to instantaneously read out the nephrotoxic drugs mechanisms based on the disparate cell surface phenotypes. Cell surface components induced by multiple nephrotoxic drugs can change the fluorescence emission of multicolor quantum dots, generating their corresponding fluorescent fingerprints. The fluorescence response signatures induced by different nephrotoxic agents are gained with 84% accuracy via linear discriminant analysis. Furthermore, taking the time-toxicity relationship into consideration, dynamic fluorescent fingerprint is obtained through continuous monitoring the progress of renal cell damage, achieving 100% precise classification for nephrotoxic mechanisms of four types of antibiotics. Notably, the fluorescent fingerprint-based high-throughput sensor has been demonstrated by successfully distinguishing nephrotoxic drugs in seconds, employing a promising protocol to discriminate the specific mechanism of nephrotoxic drugs, as well as drug safety evaluation.
Collapse
Affiliation(s)
- Xie-An Yu
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Research Center for Traceability and Standardization of TCMs, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 211198, China; Shenzhen Institute for Drug Control, Shenzhen, 518057, China
| | - Xuefei Bai
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Research Center for Traceability and Standardization of TCMs, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 211198, China
| | - Ran Zhang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Research Center for Traceability and Standardization of TCMs, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 211198, China
| | - Ying Zhang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Research Center for Traceability and Standardization of TCMs, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 211198, China
| | - Yiting Hu
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Research Center for Traceability and Standardization of TCMs, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 211198, China
| | - Mi Lu
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Research Center for Traceability and Standardization of TCMs, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 211198, China
| | - Bo-Yang Yu
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Research Center for Traceability and Standardization of TCMs, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 211198, China.
| | - Shijia Liu
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, China.
| | - Jiangwei Tian
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Research Center for Traceability and Standardization of TCMs, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 211198, China.
| |
Collapse
|
47
|
Vriend J, Vormann MK, Lanz HL, Joore J, Trietsch SJ, Russel FG, Jacobsen B, Roth A, Lu S, Polli JW, Naidoo AA, Masereeuw R, Wilmer MJ, Suter-Dick L. Nephroscreen: A robust and versatile renal tubule-on-a-chip platform for nephrotoxicity assessment. CURRENT OPINION IN TOXICOLOGY 2021. [DOI: 10.1016/j.cotox.2021.03.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
48
|
Implementation of a Human Renal Proximal Tubule on a Chip for Nephrotoxicity and Drug Interaction Studies. J Pharm Sci 2021; 110:1601-1614. [PMID: 33545187 DOI: 10.1016/j.xphs.2021.01.028] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 01/26/2021] [Accepted: 01/26/2021] [Indexed: 12/18/2022]
Abstract
Proximal tubule epithelial cells (PTEC) are susceptible to drug-induced kidney injury (DIKI). Cell-based, two-dimensional (2D) in vitro PTEC models are often poor predictors of DIKI, probably due to the lack of physiological architecture and flow. Here, we assessed a high throughput, 3D microfluidic platform (Nephroscreen) for the detection of DIKI in pharmaceutical development. This system was established with four model nephrotoxic drugs (cisplatin, tenofovir, tobramycin and cyclosporin A) and tested with eight pharmaceutical compounds. Measured parameters included cell viability, release of lactate dehydrogenase (LDH) and N-acetyl-β-d-glucosaminidase (NAG), barrier integrity, release of specific miRNAs, and gene expression of toxicity markers. Drug-transporter interactions for P-gp and MRP2/4 were also determined. The most predictive read outs for DIKI were a combination of cell viability, LDH and miRNA release. In conclusion, Nephroscreen detected DIKI in a robust manner, is compatible with automated pipetting, proved to be amenable to long-term experiments, and was easily transferred between laboratories. This proof-of-concept-study demonstrated the usability and reproducibility of Nephroscreen for the detection of DIKI and drug-transporter interactions. Nephroscreen it represents a valuable tool towards replacing animal testing and supporting the 3Rs (Reduce, Refine and Replace animal experimentation).
Collapse
|
49
|
Hashem KS, Abdelazem AZ, Mohammed MA, Nagi AM, Aboulhoda BE, Mohammed ET, Abdel-Daim MM. Thymoquinone alleviates mitochondrial viability and apoptosis in diclofenac-induced acute kidney injury (AKI) via regulating Mfn2 and miR-34a mRNA expressions. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2021; 28:10100-10113. [PMID: 33165700 DOI: 10.1007/s11356-020-11313-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Accepted: 10/18/2020] [Indexed: 06/11/2023]
Abstract
The current study was prepared to assess the underlying mechanism of diclofenac (Diclo)-stimulated renal oxidative damage (50 mg/kg/day for two consecutive days I.P) and antioxidative, and antiapoptotic effects of Thymoquinone (20 mg/kg/day for 21 days P.O). Exposure of rats to Diclo significantly increased serum urea and creatinine, decreased GSH, catalase, and total antioxidant capacity with a concomitant increase of lipid peroxidation. Diclo significantly decreased renal mitochondrial viability %, increased DNA fragmentation %, caspase 3 activity, and cytochrome C (Cyt C) concentration. Molecular investigations revealed that Diclo administration caused a significant reduction of mitofusin-2 (Mfn2) and increase of microRNA-34a (miR-34a) mRNA expressions with a concomitant decrease of Nrf2 and HO-1 mRNA expressions/protein levels and increase of NF-κB mRNA expressions. Thymoquinone restored renal oxidative/antioxidant redox. Thymoquinone significantly increased the renal mitochondrial viability % and reduced renal DNA fragmentation %, caspase 3 activity, and Cyt C. Moreover, thymoquinone modulated renal Mfn2 and miR-34a as compared to Diclo group. Our findings were confirmed by immunohistochemical assays for detecting the iNOS and NOX4 in renal tissue as well as histopathological investigations. Obtained results demonstrated that thymoquinone possess a potential antioxidant, antiapoptotic defense and exhibited a strong nephroprotective activity against Diclo-induced toxicity.
Collapse
Affiliation(s)
- Khalid Shaaban Hashem
- Biochemistry Department, Faculty of Veterinary Medicine, Beni-Suef University, Beni-Suef, 62511, Egypt.
| | - Ahmed Zakaria Abdelazem
- Biotechnology and Life Sciences Department, Faculty of Postgraduate Studies for Advanced Sciences, Beni-Suef University, Beni-Suef, Egypt
| | | | - Amr M Nagi
- Biotechnology and Life Sciences Department, Faculty of Postgraduate Studies for Advanced Sciences, Beni-Suef University, Beni-Suef, Egypt
| | - Basma Emad Aboulhoda
- Department of Anatomy and Embryology, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Eman T Mohammed
- Biochemistry Department, Faculty of Veterinary Medicine, Beni-Suef University, Beni-Suef, 62511, Egypt
| | - Mohamed M Abdel-Daim
- Department of Zoology, College of Science, King Saud University, P.O. Box 2455, Riyadh, 11451, Saudi Arabia
- Pharmacology Department, Faculty of Veterinary Medicine, Suez Canal University, 41522, Ismailia, Egypt
| |
Collapse
|
50
|
Mody H, Ramakrishnan V, Chaar M, Lezeau J, Rump A, Taha K, Lesko L, Ait-Oudhia S. A Review on Drug-Induced Nephrotoxicity: Pathophysiological Mechanisms, Drug Classes, Clinical Management, and Recent Advances in Mathematical Modeling and Simulation Approaches. Clin Pharmacol Drug Dev 2020; 9:896-909. [PMID: 33025766 DOI: 10.1002/cpdd.879] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Accepted: 09/08/2020] [Indexed: 12/13/2022]
Abstract
A variety of marketed drugs belonging to various therapeutic classes are known to cause nephrotoxicity. Nephrotoxicity can manifest itself in several forms depending on the specific site involved as well as the underlying pathophysiological mechanisms. As they often coexist with other pathophysiological conditions, the steps that can be taken to treat them are often limited. Thus, drug-induced nephrotoxicity remains a major clinical challenge. Prior knowledge of risk factors associated with special patient populations and specific classes of drugs, combined with early diagnosis, therapeutic drug monitoring with dose adjustments, as well as timely prospective treatments are essential to prevent and manage them better. Most incident drug-induced renal toxicity is reversible only if diagnosed at an early stage and treated promptly. Hence, diagnosis at an early stage is the need of the hour to counter it. Significant recent advances in the identification of novel early biomarkers of nephrotoxicity are not beyond limitations. In such a scenario, mathematical modeling and simulation (M&S) approaches may help to better understand and predict toxicities in a clinical setting. This review summarizes pathophysiological mechanisms of drug-induced nephrotoxicity, classes of nephrotoxic drugs, management, prevention, and diagnosis in clinics. Finally, it also highlights some of the recent advancements in mathematical M&S approaches that could be used to better understand and predict drug-induced nephrotoxicity.
Collapse
Affiliation(s)
- Hardik Mody
- Center for Pharmacometrics and Systems Pharmacology, Department of Pharmaceutics, College of Pharmacy, University of Florida, Gainesville, Florida, USA
| | - Vidya Ramakrishnan
- Department of Pharmaceutical Sciences, University at Buffalo, SUNY, Buffalo, New York, USA
| | - Maher Chaar
- Center for Pharmacometrics and Systems Pharmacology, Department of Pharmaceutics, College of Pharmacy, University of Florida, Gainesville, Florida, USA
| | - Jovin Lezeau
- Center for Pharmacometrics and Systems Pharmacology, Department of Pharmaceutics, College of Pharmacy, University of Florida, Gainesville, Florida, USA
| | - Adrian Rump
- Center for Pharmacometrics and Systems Pharmacology, Department of Pharmaceutics, College of Pharmacy, University of Florida, Gainesville, Florida, USA
| | - Kareem Taha
- Center for Pharmacometrics and Systems Pharmacology, Department of Pharmaceutics, College of Pharmacy, University of Florida, Gainesville, Florida, USA
| | - Lawrence Lesko
- Center for Pharmacometrics and Systems Pharmacology, Department of Pharmaceutics, College of Pharmacy, University of Florida, Gainesville, Florida, USA
| | | |
Collapse
|