1
|
Abady MM, Saadeldin IM, Han A, Bang S, Kang H, Seok DW, Kwon HJ, Cho J, Jeong JS. Modeling Lamotrigine-Induced Reprotoxicity in Porcine Endometrial Organoids: Integrated Multi-platform Profiling. Reprod Toxicol 2025:108926. [PMID: 40252710 DOI: 10.1016/j.reprotox.2025.108926] [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: 09/27/2024] [Revised: 02/26/2025] [Accepted: 04/16/2025] [Indexed: 04/21/2025]
Abstract
Lamotrigine, a newer generation anti-epileptic drug aimed at addressing reproductive complications, requires thorough evaluation of its effects on the endometrium. Using the three-dimensional endometrial organoid (EO) model provides a distinct advantage in modeling lamotrigine-induced toxicity, offering a more relevant physiological system. In this study, a porcine EO model was used and treated with lamotrigine to mimic and analyze drug-induced toxicity. Porcine uteri were processed and digested with collagenase, then combined with Matrigel and incubated with 5% CO2 environment, at 38°C. During passaging, cells were dissociated, treated with trypsin-EDTA, and subcultured, with the medium renewed every 2 to 3 days. Different analytical methods were employed to evaluate lamotrigine's impact on the endometrial organoids, covering aspects such as cell viability, morphology, replication, steroidogenesis, and metabolic changes. The results showed significant alterations in cell morphology with a decrease in number and size. Metabolite analysis revealed metabolic shifts in some amino acids, glucose and galactose, ranging from approximately 1.5 to 5 times, (p < 0.05), when compared to the control groups. Molecular assays indicated increased oxidative stress, activation of apoptotic pathway, and disrupted steroidogenesis, revealing lamotrigine as an active endocrine disruptor. Moreover, lamotrigine induced changes in specific miRNAs that regulate implantation, and epithelial-mesenchymal transition pathways. In conclusion, our study highlights the potential diverse impact of lamotrigine on the endometrial microenvironment, emphasizing the need for further investigations into its implications on reproductive health and embryo implantation.
Collapse
Affiliation(s)
- Mariam M Abady
- Organic Metrology Group, Division of Chemical and Material Metrology, Korea Research Institute of Standards and Science, 267 Gajeong-ro, Yuseong-gu, Daejeon 34113, Republic of Korea; Department of Bio-Analytical Science, University of Science and Technology, 217 Gajeong-ro, Yuseong-gu, Daejeon 34113, Republic of Korea; Department of Nutrition and Food Sciences, National Research Centre, Dokki, Cairo, 12622, Egypt
| | - Islam M Saadeldin
- Comparative Medicine Department, King Faisal Specialist Hospital and Research Centre, Riyadh 11211, Saudi Arabia; Laboratory of Theriogenology, College of Veterinary Medicine, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon, 34134, Republic of Korea; Research Institute of Veterinary Medicine, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon, 34134, Republic of Korea
| | - Ayeong Han
- Laboratory of Theriogenology, College of Veterinary Medicine, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon, 34134, Republic of Korea; Laboratory of Theriogenology, College of Veterinary Medicine, Seoul National University, 1, Gwanak-ro, Gwanak-gu, Seoul 08826, Republic of Korea
| | - Seonggyu Bang
- Laboratory of Theriogenology, College of Veterinary Medicine, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon, 34134, Republic of Korea; Laboratory of Theriogenology, College of Veterinary Medicine, Seoul National University, 1, Gwanak-ro, Gwanak-gu, Seoul 08826, Republic of Korea
| | - Heejae Kang
- Laboratory of Theriogenology, College of Veterinary Medicine, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon, 34134, Republic of Korea; Laboratory of Theriogenology, College of Veterinary Medicine, Seoul National University, 1, Gwanak-ro, Gwanak-gu, Seoul 08826, Republic of Korea
| | - Dong Wook Seok
- Organic Metrology Group, Division of Chemical and Material Metrology, Korea Research Institute of Standards and Science, 267 Gajeong-ro, Yuseong-gu, Daejeon 34113, Republic of Korea; Department of Bio-Analytical Science, University of Science and Technology, 217 Gajeong-ro, Yuseong-gu, Daejeon 34113, Republic of Korea
| | - Ha-Jeong Kwon
- Organic Metrology Group, Division of Chemical and Material Metrology, Korea Research Institute of Standards and Science, 267 Gajeong-ro, Yuseong-gu, Daejeon 34113, Republic of Korea
| | - Jongki Cho
- Laboratory of Theriogenology, College of Veterinary Medicine, Seoul National University, 1, Gwanak-ro, Gwanak-gu, Seoul 08826, Republic of Korea.
| | - Ji-Seon Jeong
- Organic Metrology Group, Division of Chemical and Material Metrology, Korea Research Institute of Standards and Science, 267 Gajeong-ro, Yuseong-gu, Daejeon 34113, Republic of Korea; Department of Bio-Analytical Science, University of Science and Technology, 217 Gajeong-ro, Yuseong-gu, Daejeon 34113, Republic of Korea.
| |
Collapse
|
2
|
Ribeiro M, Alvarenga L, Cardozo LFMF, Baptista BG, Nascimento D, Esgalhado M, Mafra D. Urolithin as a Metabolite of Ellagitannins and Ellagic Acid from Fruits and Nuts Produced by the Gut Microbiota: Its Role on Non-Communicable Diseases. Curr Nutr Rep 2025; 14:55. [PMID: 40180655 DOI: 10.1007/s13668-025-00645-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/14/2025] [Indexed: 04/05/2025]
Abstract
PURPOSE OF REVIEW This narrative review investigates how urolithins produced by the gut microbiota can regulate transcription factors (such as NRF2, NF-kB, and PPAR-γ) associated with senescence, inflammation, and imbalanced redox status. It also discusses the potential benefits of urolithins for patients with chronic diseases, including cardiovascular disease, cancer, diabetes, obesity, and chronic kidney disease. RECENT FINDINGS Studies have shown that urolithins have many health benefits, including anti-inflammatory, antioxidant, antimicrobial, and anti-atherosclerotic effects. They are also linked to improved mitochondrial function and imbalanced redox associated with activating the Nrf2/ARE pathway. Urolithins are metabolites produced by gut microbiota from ellagic acid and ellagitannins, polyphenols primarily found in nuts and fruits, including pomegranates and berries like raspberries, cloudberries, and blackberries.
Collapse
Affiliation(s)
- Marcia Ribeiro
- Graduate Program in Biological Sciences - Physiology, IBCCF, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro (RJ), Brazil.
- Unidade de Pesquisa Clínica-UPC, Rua Marquês de Paraná, 303/4 Andar, Niterói, RJ, Brazil.
- Federal Fluminense University, Niterói, Rio de Janeiro (RJ), Brazil.
| | - Livia Alvarenga
- Graduate Program in Biological Sciences - Physiology, IBCCF, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro (RJ), Brazil
- Graduate Program in Nutrition Sciences, Fluminense Federal University (UFF), Niterói, RJ, Brazil
| | - Ludmila F M F Cardozo
- Graduate Program in Nutrition Sciences, Fluminense Federal University (UFF), Niterói, RJ, Brazil
- Graduate Program in Cardiovascular Sciences, Fluminense Federal University (UFF), Niterói, RJ, Brazil
| | - Beatriz G Baptista
- Graduate Program in Medical Sciences, Fluminense Federal University (UFF), Niterói, RJ, Brazil
| | - Danielle Nascimento
- Graduate Program in Biological Sciences - Physiology, IBCCF, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro (RJ), Brazil
| | - Marta Esgalhado
- CBIOS - Universidade Lusófona's Research Center for Biosciences and Health Technologies, Lisbon, Portugal
| | - Denise Mafra
- Graduate Program in Biological Sciences - Physiology, IBCCF, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro (RJ), Brazil
- Graduate Program in Nutrition Sciences, Fluminense Federal University (UFF), Niterói, RJ, Brazil
- Graduate Program in Medical Sciences, Fluminense Federal University (UFF), Niterói, RJ, Brazil
| |
Collapse
|
3
|
Klatt OC, de Brouwer L, Hendriks F, Dehne EM, Ataç Wagegg B, Jennings P, Wilmes A. Human and rat renal proximal tubule in vitro models for ADME applications. Arch Toxicol 2025:10.1007/s00204-025-03987-4. [PMID: 40032686 DOI: 10.1007/s00204-025-03987-4] [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: 11/12/2024] [Accepted: 02/10/2025] [Indexed: 03/05/2025]
Abstract
The kidney is a major organ dictating excretion rates of chemicals and their metabolites from the body and thus renal clearance is frequently a major component of pharmaco-(toxico)-kinetic profiles. Within the nephron, the proximal tubule is the major site for xenobiotic reabsorption from glomerular filtrate and xenobiotic secretion from the blood into the lumen via the expression of multiple inward (lumen to interstitium) and outward transport systems (interstitium to lumen). While there exist several human proximal tubular cell culture options that could be utilized for modelling the proximal tubule component of renal clearance, they do not necessarily represent the full complement of xenobiotic transport processes of their in vivo counterparts. Here, we review available human and rat renal proximal tubule in vitro models, including subcellular fractions, immortalized cell lines, primary cell cultures, induced pluripotent stem cell (iPSC)-derived models and also consider more organotypic cell culture environments such as microporous growth supports, organoids and microfluidic systems. This review focuses on expression levels and function of human and rat renal transporters and phase I and II metabolizing enzymes in these models in order to critically assess their usefulness and to identify potential solutions to overcome identified limitations.
Collapse
Affiliation(s)
- Olivia C Klatt
- Department of Chemistry and Pharmaceutical Science, Amsterdam Institute of Molecular and Life Sciences, Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081 HZ, Amsterdam, The Netherlands
- Amsterdam Institute of Molecular and Life Sciences (AIMMS), Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Lenya de Brouwer
- Department of Chemistry and Pharmaceutical Science, Amsterdam Institute of Molecular and Life Sciences, Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081 HZ, Amsterdam, The Netherlands
- Amsterdam Institute of Molecular and Life Sciences (AIMMS), Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Femke Hendriks
- Department of Chemistry and Pharmaceutical Science, Amsterdam Institute of Molecular and Life Sciences, Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081 HZ, Amsterdam, The Netherlands
- Amsterdam Institute of Molecular and Life Sciences (AIMMS), Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | | | | | - Paul Jennings
- Department of Chemistry and Pharmaceutical Science, Amsterdam Institute of Molecular and Life Sciences, Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081 HZ, Amsterdam, The Netherlands.
- Amsterdam Institute of Molecular and Life Sciences (AIMMS), Vrije Universiteit Amsterdam, Amsterdam, Netherlands.
| | - Anja Wilmes
- Department of Chemistry and Pharmaceutical Science, Amsterdam Institute of Molecular and Life Sciences, Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081 HZ, Amsterdam, The Netherlands.
- Amsterdam Institute of Molecular and Life Sciences (AIMMS), Vrije Universiteit Amsterdam, Amsterdam, Netherlands.
| |
Collapse
|
4
|
Sakolish C, Tsai HHD, Lin HC, Bajaj P, Villenave R, Ferguson SS, Stanko JP, Becker RA, Hewitt P, Chiu WA, Rusyn I. Comparative Analysis of Proximal Tubule Cell Sources for In Vitro Studies of Renal Proximal Tubule Toxicity. Biomedicines 2025; 13:563. [PMID: 40149543 PMCID: PMC11940618 DOI: 10.3390/biomedicines13030563] [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: 01/24/2025] [Revised: 02/16/2025] [Accepted: 02/18/2025] [Indexed: 03/29/2025] Open
Abstract
Background/Objectives: The kidneys are essential for eliminating drugs and chemicals from the human body and renal epithelial cells are particularly vulnerable to damage caused by xenobiotics and their metabolites. Drug-induced kidney toxicity is a major cause of drug attrition during preclinical and clinical development and the ability to predict renal toxicity remains a pressing challenge, necessitating more predictive in vitro models. However, the abundance of commercially available renal proximal tubule epithelial cell (RPTEC) sources complicates the selection of the most predictive cell types. Methods: This study compared a wide range of RPTEC sources, including primary cells (Lonza) and various RPTEC lines from different vendors, such as ciPTECs (Cell4Pharma), TERT1/RPTECs (ATCC), and HEK293 (GenoMembrane), including OAT1-overexpressing variants. HepG2 cells were included for a comparison of organ specificity. The different cells were cultured in 96- or 384-well plates and exposed to 12 drugs for 72 h at a concentration yielding a response (0.3-300 µM) to evaluate their ability to predict clinical outcomes. The CellTiterGlo® assay was used to measure cell viability, and transcriptome data from unexposed cells was analyzed using the TempO-seq® S1500+ platform. Results: Gene expression data showed that the primary kidney cells most closely matched the transcriptome of the human kidney medulla, followed by the TERT1 and ciPTEC lines, with the HEK lines showing the lowest similarity. The RPTEC sources showed clustering by cell type, with OAT1 overexpression driving changes in metabolic, detoxification, and immune pathways, especially in TERT1 cells. Cell viability data were used to determine points of departure (PODs) which were compared to human serum Cmax values to assess safety margins. The TERT1 and ciPTEC RPTEC lines demonstrated the highest predictive performance for nephrotoxicity, with OAT1 overexpression significantly enhancing sensitivity, accuracy, and overall predictive power (MCC scores: 0.764 and 0.667, respectively). In contrast, HepG2 cells showed the lowest performance across all metrics, highlighting the critical role of cell type and transporter expression in nephrotoxicity prediction. Conclusions: This study highlights important differences among RPTEC sources and their utility in drug safety studies of the renal proximal tubule. We show that while improved cell options for renal proximal tubule are needed, OAT1-overexpressing RPTECs are a superior model to the background cell type.
Collapse
Affiliation(s)
- Courtney Sakolish
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX 77843, USA; (C.S.); (H.-H.D.T.); (H.-C.L.); (W.A.C.)
| | - Han-Hsuan D. Tsai
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX 77843, USA; (C.S.); (H.-H.D.T.); (H.-C.L.); (W.A.C.)
| | - Hsing-Chieh Lin
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX 77843, USA; (C.S.); (H.-H.D.T.); (H.-C.L.); (W.A.C.)
| | - Piyush Bajaj
- Global Investigative Toxicology, Preclinical Safety, Sanofi, Cambridge, MA 02141, USA;
| | - Remi Villenave
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., 4070 Basel, Switzerland;
| | - Stephen S. Ferguson
- Division of the National Toxicology Program, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA; (S.S.F.); (J.P.S.)
| | - Jason P. Stanko
- Division of the National Toxicology Program, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA; (S.S.F.); (J.P.S.)
| | | | - Philip Hewitt
- Chemical and Preclinical Safety, Merck Healthcare KGaA, 64293 Darmstadt, Germany;
| | - Weihsueh A. Chiu
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX 77843, USA; (C.S.); (H.-H.D.T.); (H.-C.L.); (W.A.C.)
| | - Ivan Rusyn
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX 77843, USA; (C.S.); (H.-H.D.T.); (H.-C.L.); (W.A.C.)
| |
Collapse
|
5
|
Coimbra S, Rocha S, Viana SD, Rebelo R, Rocha-Pereira P, Lousa I, Valente MJ, Catarino C, Belo L, Bronze-da-Rocha E, Reis F, Santos-Silva A. Gadoteric Acid and Gadolinium: Exploring Short- and Long-Term Effects on Healthy Animals. J Xenobiot 2025; 15:34. [PMID: 40126252 PMCID: PMC11932278 DOI: 10.3390/jox15020034] [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: 01/15/2025] [Revised: 02/13/2025] [Accepted: 02/19/2025] [Indexed: 03/25/2025] Open
Abstract
Regarding the safety of gadolinium (Gd (III))-based contrast agents, we aimed to evaluate the short- and long-term effects following a single exposure to gadoteric acid (DOTA) or to free Gd (III) using animal models. Biomarkers of kidney injury, inflammation, iron metabolism, dyslipidemia, hepatic and hematologic disturbances and kidney histopathological and differential gene expression (DGE) analyses were evaluated. In the short-term study, compared to the controls, exposure to Gd (III) was associated with higher inflammation; changes in lipid, iron and hepatic metabolisms; hematological alterations; and kidney damage. Exposure to DOTA revealed changes in hematological, lipid and hepatic biomarkers. In the long-term study, compared to the controls, exposure to Gd (III) or to DOTA showed much fewer changes than the short-term exposure. Comparing the kidney gene expression of Gd (III) or DOTA exposure versus the control, we found clearly different DGE patterns and a lower number of differently expressed genes in the long-term study, for both compounds. Our data show that a single-dose exposure to these compounds induces several short-term changes which over time return to normal or are sustained, although with less severity, especially in the case of DOTA.
Collapse
Affiliation(s)
- Susana Coimbra
- UCIBIO i4HB, Faculdade de Farmácia, Universidade do Porto, 4050-313 Porto, Portugal
- UCIBIO i4HB, Translational Toxicology Research Laboratory, University Institute of Health Sciences (1H-TOXRUN, IUCS-CESPU), Avenida Central de Gandra 1317, 4585-116 Gandra, Portugal
| | - Susana Rocha
- UCIBIO i4HB, Faculdade de Farmácia, Universidade do Porto, 4050-313 Porto, Portugal
| | - Sofia D Viana
- Institute of Pharmacology & Experimental Therapeutics & Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3000-548 Coimbra, Portugal
- ESTESC-Coimbra Health School, Polytechnic Institute of Coimbra, 3046-854 Coimbra, Portugal
- H&TRC-Health and Technology Research Center, Coimbra Health School, Polytechnic University of Coimbra, 3046-854 Coimbra, Portugal
| | - Rute Rebelo
- UCIBIO i4HB, Faculdade de Farmácia, Universidade do Porto, 4050-313 Porto, Portugal
| | - Petronila Rocha-Pereira
- UCIBIO i4HB, Faculdade de Farmácia, Universidade do Porto, 4050-313 Porto, Portugal
- Health Science Research Centre, University of Beira Interior, 6201-506 Covilhã, Portugal
| | - Irina Lousa
- UCIBIO i4HB, Faculdade de Farmácia, Universidade do Porto, 4050-313 Porto, Portugal
| | - Maria João Valente
- National Food Institute, Technical University of Denmark, 2800 Kgs Lyngby, Denmark
| | - Cristina Catarino
- UCIBIO i4HB, Faculdade de Farmácia, Universidade do Porto, 4050-313 Porto, Portugal
| | - Luís Belo
- UCIBIO i4HB, Faculdade de Farmácia, Universidade do Porto, 4050-313 Porto, Portugal
| | - Elsa Bronze-da-Rocha
- UCIBIO i4HB, Faculdade de Farmácia, Universidade do Porto, 4050-313 Porto, Portugal
| | - Flávio Reis
- Institute of Pharmacology & Experimental Therapeutics & Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3000-548 Coimbra, Portugal
| | - Alice Santos-Silva
- UCIBIO i4HB, Faculdade de Farmácia, Universidade do Porto, 4050-313 Porto, Portugal
| |
Collapse
|
6
|
Ceccotti E, Semnani A, Bussolati B, Bruno S. Human kidney organoids for modeling the development of different diseases. Curr Top Dev Biol 2025; 163:364-393. [PMID: 40254349 DOI: 10.1016/bs.ctdb.2024.12.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/22/2025]
Abstract
The increasing incidence of kidney diseases has highlighted the need for in vitro experimental models to mimic disease development and to test new therapeutic approaches. Traditional two-dimensional in vitro experimental models are not fully able to recapitulate renal diseases. Instead, kidney organoids represent three-dimensional models that better mimic the human organ from both structural and functional points of view. Human pluripotent stem cells (PSCs), both embryonic and induced, are ideal sources for generating renal organoids. These organoids contain all renal cell types and the protocols to differentiate PSCs into renal organoids consist of three different stages that recapitulate embryonic development: mesodermal induction, nephron progenitor formation, and nephron differentiation. Recently it has been establish a renal organoid model where collecting ducts are also present. In this case, the presence of ureteric bud progenitor cells is essential. Renal organoids are particularly useful for studying genetic diseases, by introducing the specific mutations in PSCs by genome editing or generating organoids from patient-derived PSCs. Moreover, renal organoids represent promising models in toxicology studies and testing new therapeutic approaches. Renal organoids can be established also from adult stem cells. This type of organoid, named tubuloid, is composed only of epithelial cells and recapitulates the tissue repair process. The tubuloids can be generated from adult stem or progenitor cells, obtained from renal biopsies or urine, and are promising in vitro models for studying tubular functions, diseases, and regeneration. Tubuloids can be derived from patients and permit the study of genetic diseases, performing personalized drug screening and modeling renal pathologies.
Collapse
Affiliation(s)
- Elena Ceccotti
- Department of Medical Sciences, University of Torino, Corso Dogliotti, Torino, Italy
| | - Armina Semnani
- Department of Medical Sciences, University of Torino, Corso Dogliotti, Torino, Italy
| | - Benedetta Bussolati
- Department of Medical Sciences, University of Torino, Corso Dogliotti, Torino, Italy; Molecular Biotechnology Center "Guido Tarone", Via Nizza, Torino, Italy
| | - Stefania Bruno
- Department of Medical Sciences, University of Torino, Corso Dogliotti, Torino, Italy.
| |
Collapse
|
7
|
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
|
8
|
Choshi J, Hanser S, Mabhida SE, Mokoena H, Moetlediwa MT, Muvhulawa N, Sekgala MD, Nkambule BB, Mchiza ZJR, Ndwandwe D, Nqebelele U, Kengne AP, Dludla PV. A systematic review assessing the association of inflammatory markers with kidney dysfunction in people living with HIV on highly active antiretroviral therapy. BMC Infect Dis 2024; 24:776. [PMID: 39095687 PMCID: PMC11297709 DOI: 10.1186/s12879-024-09594-5] [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: 02/10/2024] [Accepted: 07/05/2024] [Indexed: 08/04/2024] Open
Abstract
Monitoring chronic diseases, particularly kidney disorders, in people living with HIV (PLWH) is of paramount importance. Here, a systematic search was conducted across electronic search engine and databases like PubMed, Scopus, and Google Scholar, from date of inception until December 2023, to identify pertinent studies reporting on any association between inflammation and kidney function in PLWH. Only six clinical studies in peer-reviewed journals met the inclusion criteria, involving 1467 participants aged 37 to 51, with approximately 17% being females. The report emphasizes the potential impact of highly active antiretroviral therapy (HAART) on kidney function in PLWH, highlighting the significance of monitoring inflammation markers as indicators of kidney function, even when HAART is effective. Acknowledging study limitations, particularly the scarcity of relevant research, the findings highlight a need for more research to inform on clinical guidance to optimize HIV management, particularly regarding kidney health and HAART regimens. Although very limited studies were evaluated, the study lays an important foundation for future research to uncover the complex relationship between HAART, inflammation markers, and kidney health in PLWH.
Collapse
Affiliation(s)
- Joel Choshi
- Department of Physiology and Environmental Health, University of Limpopo, Sovenga, 0727, South Africa.
| | - Sidney Hanser
- Department of Physiology and Environmental Health, University of Limpopo, Sovenga, 0727, South Africa
| | - Sihle E Mabhida
- Non-Communicable Diseases Research Unit, South African Medical Research Council, Tygerberg, 7505, South Africa
| | - Haskly Mokoena
- Department of Physiology and Environmental Health, University of Limpopo, Sovenga, 0727, South Africa
| | - Marakiya T Moetlediwa
- Department of Biochemistry, North-West University, Mafikeng Campus, Mmabatho, 2735, South Africa
| | - Ndivhuwo Muvhulawa
- Department of Biochemistry, North-West University, Mafikeng Campus, Mmabatho, 2735, South Africa
- Cochrane South Africa, South African Medical Research Council, Tygerberg, 7505, South Africa
| | - Machoene D Sekgala
- Non-Communicable Diseases Research Unit, South African Medical Research Council, Tygerberg, 7505, South Africa
| | - Bongani B Nkambule
- School of Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal, Durban, 4000, South Africa
| | - Zandile J R Mchiza
- Non-Communicable Diseases Research Unit, South African Medical Research Council, Tygerberg, 7505, South Africa
- School of Public Health, University of the Western Cape, Bellville, 7535, South Africa
| | - Duduzile Ndwandwe
- Cochrane South Africa, South African Medical Research Council, Tygerberg, 7505, South Africa
| | - Unati Nqebelele
- Non-Communicable Diseases Research Unit, South African Medical Research Council, Tygerberg, 7505, South Africa
- Department of Medicine, University of Cape Town, Cape Town, 7700, South Africa
- Department of Internal Medicine, University of the Witwatersrand, Johannesburg, 2050, South Africa
| | - André P Kengne
- Non-Communicable Diseases Research Unit, South African Medical Research Council, Tygerberg, 7505, South Africa
- Department of Medicine, University of Cape Town, Cape Town, 7700, South Africa
| | - Phiwayinkosi V Dludla
- Cochrane South Africa, South African Medical Research Council, Tygerberg, 7505, South Africa.
- Department of Biochemistry and Microbiology, University of Zululand, KwaDlangezwa, 3880, South Africa.
| |
Collapse
|
9
|
Tenebro CP, Marcial NBJM, Salcepuedes JJ, Torrecampo JC, Hernandez RD, Francisco JAP, Infante KMG, Belardo VJ, Paderes MC, Alvero RGY, Saludes JP, Dalisay DS. Visualization of renal rotenone accumulation after oral administration and in situ detection of kidney injury biomarkers via MALDI mass spectrometry imaging. Front Mol Biosci 2024; 11:1366278. [PMID: 39011141 PMCID: PMC11246995 DOI: 10.3389/fmolb.2024.1366278] [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: 01/06/2024] [Accepted: 06/04/2024] [Indexed: 07/17/2024] Open
Abstract
The examination of drug accumulation within complex biological systems offers valuable insights into the molecular aspects of drug metabolism and toxicity. Matrix-assisted laser desorption/ionization mass spectrometry imaging (MALDI MSI) is an innovative methodology that enables the spatial visualization and quantification of biomolecules as well as drug and its metabolites in complex biological system. Hence, this method provides valuable insights into the metabolic profile and any molecular changes that may occur as a result of drug treatment. The renal system is particularly vulnerable to adverse effects of drug-induced harm and toxicity. In this study, MALDI MSI was utilized to examine the spatial distribution of drug and renal metabolites within kidney tissues subsequent to a single oral dosage of the anticancer compound rotenone. The integration of ion mobility spectrometry with MALDI MSI enhanced the data acquisition and analysis, resulting to improved mass resolution. Subsequently, the MS/MS fragment ions of rotenone reference drug were detected and characterized using MALDI HDMS/MS imaging. Notably, drug accumulation was observed in the cortical region of the representative kidney tissue sections treated with rotenone. The histological examination of treated kidney tissues did not reveal any observable changes. Differential ion intensity of renal endogenous metabolites was observed between untreated and rotenone-treated tissues. In the context of treated kidney tissues, the ion intensity level of sphingomyelin (D18:1/16:0), a sphingolipid indicator of glomerular cell injury and renal damage, was found to be elevated significantly compared to untreated kidney tissues. Conversely, the ion intensities of choline, glycero-3-phosphocholine (GPC), inosine, and a lysophosphatidylcholine LysoPC(18:0) exhibited a significant decrease. The results of this study demonstrate the potential of MALDI MSI as a novel technique for investigating the in situ spatial distribution of drugs and renal endogenous molecules while preserving the anatomical integrity of the kidney tissue. This technique can be used to study drug-induced metabolism and toxicity in a dynamic manner.
Collapse
Affiliation(s)
- Chuckcris P Tenebro
- Center for Chemical Biology and Biotechnology, University of San Agustin, Iloilo City, Philippines
| | - Neaven Bon Joy M Marcial
- Center for Chemical Biology and Biotechnology, University of San Agustin, Iloilo City, Philippines
| | - Janine J Salcepuedes
- Center for Chemical Biology and Biotechnology, University of San Agustin, Iloilo City, Philippines
| | - Josie C Torrecampo
- Center for Chemical Biology and Biotechnology, University of San Agustin, Iloilo City, Philippines
| | - Rajelle D Hernandez
- Institute of Chemistry, University of the Philippines Diliman, Quezon City, Philippines
| | | | | | | | - Monissa C Paderes
- Institute of Chemistry, University of the Philippines Diliman, Quezon City, Philippines
| | | | - Jonel P Saludes
- Center for Natural Drug Discovery and Development, University of San Agustin, Iloilo City, Philippines
- Department of Chemistry, University of San Agustin, Iloilo City, Philippines
- Balik Scientist Program, Department of Science and Technology-Philippine Council for Health Research and Development, Taguig City, Philippines
| | - Doralyn S Dalisay
- Center for Chemical Biology and Biotechnology, University of San Agustin, Iloilo City, Philippines
- Balik Scientist Program, Department of Science and Technology-Philippine Council for Health Research and Development, Taguig City, Philippines
- Department of Biology, University of San Agustin, Iloilo City, Philippines
| |
Collapse
|
10
|
Khalil SM, Qin X, Hakenjos JM, Wang J, Hu Z, Liu X, Wang J, Maletic-Savatic M, MacKenzie KR, Matzuk MM, Li F. MALDI Imaging Mass Spectrometry Visualizes the Distribution of Antidepressant Duloxetine and Its Major Metabolites in Mouse Brain, Liver, Kidney, and Spleen Tissues. Drug Metab Dispos 2024; 52:673-680. [PMID: 38658163 PMCID: PMC11185819 DOI: 10.1124/dmd.124.001719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 04/10/2024] [Accepted: 04/11/2024] [Indexed: 04/26/2024] Open
Abstract
Imaging mass spectrometry (IMS) is a powerful tool for mapping the spatial distribution of unlabeled drugs and metabolites that may find application in assessing drug delivery, explaining drug efficacy, and identifying potential toxicity. This study focuses on determining the spatial distribution of the antidepressant duloxetine, which is widely prescribed despite common adverse effects (liver injury, constant headaches) whose mechanisms are not fully understood. We used high-resolution IMS with matrix-assisted laser desorption/ionization to examine the distribution of duloxetine and its major metabolites in four mouse organs where it may contribute to efficacy or toxicity: brain, liver, kidney, and spleen. In none of these tissues is duloxetine or its metabolites homogeneously distributed, which has implications for both efficacy and toxicity. We found duloxetine to be similarly distributed in spleen red pulp and white pulp but differentially distributed in different anatomic regions of the liver, kidney, and brain, with dose-dependent patterns. Comparison with hematoxylin and eosin staining of tissue sections reveals that the ion images of endogenous lipids help delineate anatomic regions in the brain and kidney, while heme ion images assist in differentiating regions within the spleen. These endogenous metabolites may serve as a valuable resource for examining the spatial distribution of other drugs in tissues when staining images are not available. These findings may facilitate future mechanistic studies of the therapeutic and adverse effects of duloxetine. In the current work, we did not perform absolute quantification of duloxetine, which will be reported in due course. SIGNIFICANCE STATEMENT: The study utilized imaging mass spectrometry to examine the spatial distribution of duloxetine and its primary metabolites in mouse brain, liver, kidney, and spleen. These results may pave the way for future investigations into the mechanisms behind duloxetine's therapeutic and adverse effects. Furthermore, the mass spectrometry images of specific endogenous metabolites such as heme could be valuable in analyzing the spatial distribution of other drugs within tissues in scenarios where histological staining images are unavailable.
Collapse
Affiliation(s)
- Saleh M Khalil
- Center for Drug Discovery, Department of Pathology and Immunology (S.M.K., X.Q., J.M.H., Jia.W., M.M.-S., K.R.M., M.M.M., F.L.), NMR and Drug Metabolism Core, Advanced Technology Cores (X.Q., J.M.H., Jia.W., K.R.M., F.L.), Department of Biochemistry and Molecular Pharmacology (Jin.W., K.R.M., M.M.M., F.L.), Department of Pediatrics (S.M.K., M.M.-S.), and Nephrology Division, Department of Medicine (Z.H.), Baylor College of Medicine, Houston, Texas; Jan and Dan Duncan Neurologic Research Institute, Texas Children's Hospital, Houston, Texas (M.M.-S.); and Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas (X.L.)
| | - Xuan Qin
- Center for Drug Discovery, Department of Pathology and Immunology (S.M.K., X.Q., J.M.H., Jia.W., M.M.-S., K.R.M., M.M.M., F.L.), NMR and Drug Metabolism Core, Advanced Technology Cores (X.Q., J.M.H., Jia.W., K.R.M., F.L.), Department of Biochemistry and Molecular Pharmacology (Jin.W., K.R.M., M.M.M., F.L.), Department of Pediatrics (S.M.K., M.M.-S.), and Nephrology Division, Department of Medicine (Z.H.), Baylor College of Medicine, Houston, Texas; Jan and Dan Duncan Neurologic Research Institute, Texas Children's Hospital, Houston, Texas (M.M.-S.); and Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas (X.L.)
| | - John M Hakenjos
- Center for Drug Discovery, Department of Pathology and Immunology (S.M.K., X.Q., J.M.H., Jia.W., M.M.-S., K.R.M., M.M.M., F.L.), NMR and Drug Metabolism Core, Advanced Technology Cores (X.Q., J.M.H., Jia.W., K.R.M., F.L.), Department of Biochemistry and Molecular Pharmacology (Jin.W., K.R.M., M.M.M., F.L.), Department of Pediatrics (S.M.K., M.M.-S.), and Nephrology Division, Department of Medicine (Z.H.), Baylor College of Medicine, Houston, Texas; Jan and Dan Duncan Neurologic Research Institute, Texas Children's Hospital, Houston, Texas (M.M.-S.); and Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas (X.L.)
| | - Jian Wang
- Center for Drug Discovery, Department of Pathology and Immunology (S.M.K., X.Q., J.M.H., Jia.W., M.M.-S., K.R.M., M.M.M., F.L.), NMR and Drug Metabolism Core, Advanced Technology Cores (X.Q., J.M.H., Jia.W., K.R.M., F.L.), Department of Biochemistry and Molecular Pharmacology (Jin.W., K.R.M., M.M.M., F.L.), Department of Pediatrics (S.M.K., M.M.-S.), and Nephrology Division, Department of Medicine (Z.H.), Baylor College of Medicine, Houston, Texas; Jan and Dan Duncan Neurologic Research Institute, Texas Children's Hospital, Houston, Texas (M.M.-S.); and Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas (X.L.)
| | - Zhaoyong Hu
- Center for Drug Discovery, Department of Pathology and Immunology (S.M.K., X.Q., J.M.H., Jia.W., M.M.-S., K.R.M., M.M.M., F.L.), NMR and Drug Metabolism Core, Advanced Technology Cores (X.Q., J.M.H., Jia.W., K.R.M., F.L.), Department of Biochemistry and Molecular Pharmacology (Jin.W., K.R.M., M.M.M., F.L.), Department of Pediatrics (S.M.K., M.M.-S.), and Nephrology Division, Department of Medicine (Z.H.), Baylor College of Medicine, Houston, Texas; Jan and Dan Duncan Neurologic Research Institute, Texas Children's Hospital, Houston, Texas (M.M.-S.); and Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas (X.L.)
| | - Xinli Liu
- Center for Drug Discovery, Department of Pathology and Immunology (S.M.K., X.Q., J.M.H., Jia.W., M.M.-S., K.R.M., M.M.M., F.L.), NMR and Drug Metabolism Core, Advanced Technology Cores (X.Q., J.M.H., Jia.W., K.R.M., F.L.), Department of Biochemistry and Molecular Pharmacology (Jin.W., K.R.M., M.M.M., F.L.), Department of Pediatrics (S.M.K., M.M.-S.), and Nephrology Division, Department of Medicine (Z.H.), Baylor College of Medicine, Houston, Texas; Jan and Dan Duncan Neurologic Research Institute, Texas Children's Hospital, Houston, Texas (M.M.-S.); and Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas (X.L.)
| | - Jin Wang
- Center for Drug Discovery, Department of Pathology and Immunology (S.M.K., X.Q., J.M.H., Jia.W., M.M.-S., K.R.M., M.M.M., F.L.), NMR and Drug Metabolism Core, Advanced Technology Cores (X.Q., J.M.H., Jia.W., K.R.M., F.L.), Department of Biochemistry and Molecular Pharmacology (Jin.W., K.R.M., M.M.M., F.L.), Department of Pediatrics (S.M.K., M.M.-S.), and Nephrology Division, Department of Medicine (Z.H.), Baylor College of Medicine, Houston, Texas; Jan and Dan Duncan Neurologic Research Institute, Texas Children's Hospital, Houston, Texas (M.M.-S.); and Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas (X.L.)
| | - Mirjana Maletic-Savatic
- Center for Drug Discovery, Department of Pathology and Immunology (S.M.K., X.Q., J.M.H., Jia.W., M.M.-S., K.R.M., M.M.M., F.L.), NMR and Drug Metabolism Core, Advanced Technology Cores (X.Q., J.M.H., Jia.W., K.R.M., F.L.), Department of Biochemistry and Molecular Pharmacology (Jin.W., K.R.M., M.M.M., F.L.), Department of Pediatrics (S.M.K., M.M.-S.), and Nephrology Division, Department of Medicine (Z.H.), Baylor College of Medicine, Houston, Texas; Jan and Dan Duncan Neurologic Research Institute, Texas Children's Hospital, Houston, Texas (M.M.-S.); and Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas (X.L.)
| | - Kevin R MacKenzie
- Center for Drug Discovery, Department of Pathology and Immunology (S.M.K., X.Q., J.M.H., Jia.W., M.M.-S., K.R.M., M.M.M., F.L.), NMR and Drug Metabolism Core, Advanced Technology Cores (X.Q., J.M.H., Jia.W., K.R.M., F.L.), Department of Biochemistry and Molecular Pharmacology (Jin.W., K.R.M., M.M.M., F.L.), Department of Pediatrics (S.M.K., M.M.-S.), and Nephrology Division, Department of Medicine (Z.H.), Baylor College of Medicine, Houston, Texas; Jan and Dan Duncan Neurologic Research Institute, Texas Children's Hospital, Houston, Texas (M.M.-S.); and Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas (X.L.)
| | - Martin M Matzuk
- Center for Drug Discovery, Department of Pathology and Immunology (S.M.K., X.Q., J.M.H., Jia.W., M.M.-S., K.R.M., M.M.M., F.L.), NMR and Drug Metabolism Core, Advanced Technology Cores (X.Q., J.M.H., Jia.W., K.R.M., F.L.), Department of Biochemistry and Molecular Pharmacology (Jin.W., K.R.M., M.M.M., F.L.), Department of Pediatrics (S.M.K., M.M.-S.), and Nephrology Division, Department of Medicine (Z.H.), Baylor College of Medicine, Houston, Texas; Jan and Dan Duncan Neurologic Research Institute, Texas Children's Hospital, Houston, Texas (M.M.-S.); and Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas (X.L.)
| | - Feng Li
- Center for Drug Discovery, Department of Pathology and Immunology (S.M.K., X.Q., J.M.H., Jia.W., M.M.-S., K.R.M., M.M.M., F.L.), NMR and Drug Metabolism Core, Advanced Technology Cores (X.Q., J.M.H., Jia.W., K.R.M., F.L.), Department of Biochemistry and Molecular Pharmacology (Jin.W., K.R.M., M.M.M., F.L.), Department of Pediatrics (S.M.K., M.M.-S.), and Nephrology Division, Department of Medicine (Z.H.), Baylor College of Medicine, Houston, Texas; Jan and Dan Duncan Neurologic Research Institute, Texas Children's Hospital, Houston, Texas (M.M.-S.); and Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas (X.L.)
| |
Collapse
|
11
|
Addario G, Eussen D, Djudjaj S, Boor P, Moroni L, Mota C. 3D Printed Tubulointerstitium Chip as an In Vitro Testing Platform. Macromol Biosci 2024; 24:e2300440. [PMID: 37997523 DOI: 10.1002/mabi.202300440] [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: 09/27/2023] [Revised: 11/14/2023] [Indexed: 11/25/2023]
Abstract
Chronic kidney disease (CKD) ranks as the twelfth leading cause of death worldwide with limited treatment options. The development of in vitro models replicating defined segments of the kidney functional units, the nephrons, in a physiologically relevant and reproducible manner can facilitate drug testing. The aim of this study was to produce an in vitro organ-on-a-chip platform with extrusion-based three-dimensional (3D) printing. The manufacturing of the tubular platform was produced by printing sacrificial fibers with varying diameters, providing a suitable structure for cell adhesion and proliferation. The chip platform was seeded with primary murine tubular epithelial cells and human umbilical vein endothelial cells. The effect of channel geometry, its reproducibility, coatings for cell adhesion, and specific cell markers were investigated. The developed chip presents single and dual channels, mimicking segments of a renal tubule and the capillary network, together with an extracellular matrix gel analogue placed in the middle of the two channels, envisioning the renal tubulointerstitium in vitro. The 3D printed platform enables perfusable circular cross-section channels with fully automated, rapid, and reproducible manufacturing processes at low costs. This kidney tubulointerstitium on-a-chip provides the first step toward the production of more complex in vitro models for drug testing.
Collapse
Affiliation(s)
- Gabriele Addario
- Maastricht University, MERLN Institute for Technology-Inspired Regenerative Medicine, Complex Tissue Regeneration Department, Maastricht, 6229 ER, The Netherlands
| | - Daphne Eussen
- Maastricht University, MERLN Institute for Technology-Inspired Regenerative Medicine, Complex Tissue Regeneration Department, Maastricht, 6229 ER, The Netherlands
| | - Sonja Djudjaj
- Institute of Pathology, RWTH University of Aachen, 52074, Aachen, Germany
| | - Peter Boor
- Institute of Pathology, RWTH University of Aachen, 52074, Aachen, Germany
- Division of Nephrology, RWTH University of Aachen, 52074, Aachen, Germany
- Electron Microscopy Facility, RWTH University of Aachen, 52074, Aachen, Germany
| | - Lorenzo Moroni
- Maastricht University, MERLN Institute for Technology-Inspired Regenerative Medicine, Complex Tissue Regeneration Department, Maastricht, 6229 ER, The Netherlands
| | - Carlos Mota
- Maastricht University, MERLN Institute for Technology-Inspired Regenerative Medicine, Complex Tissue Regeneration Department, Maastricht, 6229 ER, The Netherlands
| |
Collapse
|
12
|
Valencia LJ, Tseng M, Chu ML, Yu L, Adedeji AO, Kiyota T. Zoledronic acid and ibandronate-induced nephrotoxicity in 2D and 3D proximal tubule cells derived from human and rat. Toxicol Sci 2024; 198:86-100. [PMID: 38059598 DOI: 10.1093/toxsci/kfad123] [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] [Indexed: 12/08/2023] Open
Abstract
Drug-induced proximal tubule (PT) injury remains a serious safety concern throughout drug development. Traditional in vitro 2-dimensional (2D) and preclinical in vivo models often fail to predict drug-related injuries presented in clinical trials. Various 3-dimensional (3D) microphysiological systems (MPSs) have been developed to mimic physiologically relevant properties, enabling them to be more predictive toward nephrotoxicity. To explore the capabilities of an MPS across species, we compared cytotoxicity in hRPTEC/TERT1s and rat primary proximal tubular epithelial cells (rPPTECs) following exposure to zoledronic acid and ibandronate (62.5-500 µM), and antibiotic polymyxin B (PMB) (50 and 250 µM, respectively). For comparison, we investigated cytotoxicity using 2D cultured hRPTEC/TERT1s and rPPTECs following exposure to the same drugs, including overlapping concentrations, as their 3D counterparts. Regardless of the in vitro model, bisphosphonate-exposed rPPTECs exhibited cytotoxicity quicker than hRPTEC/TERT1s. PMB was less sensitive toward nephrotoxicity in rPPTECs than hRPTEC/TERT1s, demonstrating differences in species sensitivity within both 3D and 2D models. Generally, 2D cultured cells experienced faster drug-induced cytotoxicity compared to the MPSs, suggesting that MPSs can be advantageous for longer-term drug-exposure studies, if warranted. Furthermore, ibandronate-exposed hRPTEC/TERT1s and rPPTECs produced higher levels of inflammatory and kidney injury biomarkers compared to zoledronic acid, indicating that ibandronate induces acute kidney injury, but also a potential protective response since ibandronate is less toxic than zoledronic acid. Our study suggests that the MPS model can be used for preclinical screening of compounds prior to animal studies and human clinical trials.
Collapse
Affiliation(s)
- Leslie J Valencia
- Investigative Toxicology, Department of Safety Assessment, Genentech Inc., South San Francisco, California 94080, USA
- Pathology, Department of Safety Assessment, Genentech Inc., South San Francisco, California 94080, USA
| | - Min Tseng
- Investigative Toxicology, Department of Safety Assessment, Genentech Inc., South San Francisco, California 94080, USA
| | - Mei-Lan Chu
- Pathology, Department of Safety Assessment, Genentech Inc., South San Francisco, California 94080, USA
| | - Lanlan Yu
- Investigative Toxicology, Department of Safety Assessment, Genentech Inc., South San Francisco, California 94080, USA
| | - Adeyemi O Adedeji
- Pathology, Department of Safety Assessment, Genentech Inc., South San Francisco, California 94080, USA
| | - Tomomi Kiyota
- Investigative Toxicology, Department of Safety Assessment, Genentech Inc., South San Francisco, California 94080, USA
| |
Collapse
|
13
|
Tsang YP, Hao T, Mao Q, Kelly EJ, Unadkat JD. Dysregulation of the mRNA Expression of Human Renal Drug Transporters by Proinflammatory Cytokines in Primary Human Proximal Tubular Epithelial Cells. Pharmaceutics 2024; 16:285. [PMID: 38399338 PMCID: PMC10893102 DOI: 10.3390/pharmaceutics16020285] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Revised: 01/31/2024] [Accepted: 02/07/2024] [Indexed: 02/25/2024] Open
Abstract
Proinflammatory cytokines, which are elevated during inflammation or infections, can affect drug pharmacokinetics (PK) due to the altered expression or activity of drug transporters and/or metabolizing enzymes. To date, such studies have focused on the effect of cytokines on the activity and/or mRNA expression of hepatic transporters and drug-metabolizing enzymes. However, many antibiotics and antivirals used to treat infections are cleared by renal transporters, including the basal organic cation transporter 2 (OCT2), organic anion transporters 1 and 3 (OAT1 and 3), the apical multidrug and toxin extrusion proteins 1 and 2-K (MATE1/2-K), and multidrug resistance-associated protein 2 and 4 (MRP2/4). Here, we determined the concentration-dependent effect of interleukin-6 (IL-6), IL-1β, tumor necrosis factor (TNF)-α, and interferon-γ (IFN-γ) on the mRNA expression of human renal transporters in freshly isolated primary human renal proximal tubular epithelial cells (PTECs, n = 3-5). PTECs were exposed to either a cocktail of cytokines, each at 0.01, 0.1, 1, or 10 ng/mL or individually at the same concentrations. Exposure to the cytokine cocktail for 48 h was found to significantly downregulate the mRNA expression, in a concentration-dependent manner, of OCT2, the organic anion transporting polypeptides 4C1 (OATP4C1), OAT4, MATE2-K, P-glycoprotein (P-gp), and MRP2 and upregulate the mRNA expression of the organic cation/carnitine transporter 1 (OCTN1) and MRP3. OAT1 and OAT3 also appeared to be significantly downregulated but only at 0.1 and 10 ng/mL, respectively, without a clear concentration-dependent trend. Among the cytokines, IL-1β appeared to be the most potent at down- and upregulating the mRNA expression of the transporters. Taken together, our results demonstrate for the first time that proinflammatory cytokines transcriptionally dysregulate renal drug transporters in PTECs. Such dysregulation could potentially translate into changes in transporter protein abundance or activity and alter renal transporter-mediated drug PK during inflammation or infections.
Collapse
Affiliation(s)
| | | | | | | | - Jashvant D. Unadkat
- Department of Pharmaceutics, University of Washington, Seattle, WA 98195, USA; (Y.P.T.); (T.H.); (E.J.K.)
| |
Collapse
|
14
|
Baker TK, Van Vleet TR, Mahalingaiah PK, Grandhi TSP, Evers R, Ekert J, Gosset JR, Chacko SA, Kopec AK. The Current Status and Use of Microphysiological Systems by the Pharmaceutical Industry: The International Consortium for Innovation and Quality Microphysiological Systems Affiliate Survey and Commentary. Drug Metab Dispos 2024; 52:198-209. [PMID: 38123948 DOI: 10.1124/dmd.123.001510] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 11/21/2023] [Accepted: 11/27/2023] [Indexed: 12/23/2023] Open
Abstract
Microphysiological systems (MPS) are comprised of one or multiple cell types of human or animal origins that mimic the biochemical/electrical/mechanical responses and blood-tissue barrier properties of the cells observed within a complex organ. The goal of incorporating these in vitro systems is to expedite and advance the drug discovery and development paradigm with improved predictive and translational capabilities. Considering the industry need for improved efficiency and the broad challenges of model qualification and acceptance, the International Consortium for Innovation and Quality (IQ) founded an IQ MPS working group in 2014 and Affiliate in 2018. This group connects thought leaders and end users, provides a forum for crosspharma collaboration, and engages with regulators to qualify translationally relevant MPS models. To understand how pharmaceutical companies are using MPS, the IQ MPS Affiliate conducted two surveys in 2019, survey 1, and 2021, survey 2, which differed slightly in the scope of definition of the complex in vitro models under question. The surveys captured demographics, resourcing, rank order for organs of interest, compound modalities tested, and MPS organ-specific questions, including nonclinical species needs and cell types. The major focus of this manuscript is on results from survey 2, where we specifically highlight the context of use for MPS within safety, pharmacology, or absorption, disposition, metabolism, and excretion and discuss considerations for including MPS data in regulatory submissions. In summary, these data provide valuable insights for developers, regulators, and pharma, offering a view into current industry practices and future considerations while highlighting key challenges impacting MPS adoption. SIGNIFICANCE STATEMENT: The application of microphysiological systems (MPS) represents a growing area of interest in the drug discovery and development framework. This study surveyed 20+ pharma companies to understand resourcing, current areas of application, and the key challenges and barriers to internal MPS adoption. These results will provide regulators, tech providers, and pharma industry leaders a starting point to assess the current state of MPS applications along with key learnings to effectively realize the potential of MPS as an emerging technology.
Collapse
Affiliation(s)
- Thomas K Baker
- Investigative Toxicology, Eli Lilly, Indianapolis, Indiana (T.K.B.); Investigative Toxicology and Pathology, AbbVie, Inc., Chicago, Illinois (T.R.V.F., P.K.M.); Complex In Vitro Models Group, GSK, Collegeville, Pennsylvania (T.S.P.G.); Preclinical Sciences and Translational Safety, Johnson & Johnson, Janssen Pharmaceuticals, Spring House, Pennsylvania (R.E.); UCB Pharma, Cambridge, Massachusetts (J.E.); Pharmacokinetics, Dynamics and Metabolism, Medicine Design, Pfizer, Inc., Cambridge, Massachusetts (J.R.G.); Research and Development, Bristol Myers Squibb Company, Princeton, New Jersey (S.A.C.); and Drug Safety Research & Development, Pfizer, Inc., Groton, Connecticut (A.K.K.) baker_thomas_k@lilly
| | - Terry R Van Vleet
- Investigative Toxicology, Eli Lilly, Indianapolis, Indiana (T.K.B.); Investigative Toxicology and Pathology, AbbVie, Inc., Chicago, Illinois (T.R.V.F., P.K.M.); Complex In Vitro Models Group, GSK, Collegeville, Pennsylvania (T.S.P.G.); Preclinical Sciences and Translational Safety, Johnson & Johnson, Janssen Pharmaceuticals, Spring House, Pennsylvania (R.E.); UCB Pharma, Cambridge, Massachusetts (J.E.); Pharmacokinetics, Dynamics and Metabolism, Medicine Design, Pfizer, Inc., Cambridge, Massachusetts (J.R.G.); Research and Development, Bristol Myers Squibb Company, Princeton, New Jersey (S.A.C.); and Drug Safety Research & Development, Pfizer, Inc., Groton, Connecticut (A.K.K.)
| | - Prathap Kumar Mahalingaiah
- Investigative Toxicology, Eli Lilly, Indianapolis, Indiana (T.K.B.); Investigative Toxicology and Pathology, AbbVie, Inc., Chicago, Illinois (T.R.V.F., P.K.M.); Complex In Vitro Models Group, GSK, Collegeville, Pennsylvania (T.S.P.G.); Preclinical Sciences and Translational Safety, Johnson & Johnson, Janssen Pharmaceuticals, Spring House, Pennsylvania (R.E.); UCB Pharma, Cambridge, Massachusetts (J.E.); Pharmacokinetics, Dynamics and Metabolism, Medicine Design, Pfizer, Inc., Cambridge, Massachusetts (J.R.G.); Research and Development, Bristol Myers Squibb Company, Princeton, New Jersey (S.A.C.); and Drug Safety Research & Development, Pfizer, Inc., Groton, Connecticut (A.K.K.)
| | - Taraka Sai Pavan Grandhi
- Investigative Toxicology, Eli Lilly, Indianapolis, Indiana (T.K.B.); Investigative Toxicology and Pathology, AbbVie, Inc., Chicago, Illinois (T.R.V.F., P.K.M.); Complex In Vitro Models Group, GSK, Collegeville, Pennsylvania (T.S.P.G.); Preclinical Sciences and Translational Safety, Johnson & Johnson, Janssen Pharmaceuticals, Spring House, Pennsylvania (R.E.); UCB Pharma, Cambridge, Massachusetts (J.E.); Pharmacokinetics, Dynamics and Metabolism, Medicine Design, Pfizer, Inc., Cambridge, Massachusetts (J.R.G.); Research and Development, Bristol Myers Squibb Company, Princeton, New Jersey (S.A.C.); and Drug Safety Research & Development, Pfizer, Inc., Groton, Connecticut (A.K.K.)
| | - Raymond Evers
- Investigative Toxicology, Eli Lilly, Indianapolis, Indiana (T.K.B.); Investigative Toxicology and Pathology, AbbVie, Inc., Chicago, Illinois (T.R.V.F., P.K.M.); Complex In Vitro Models Group, GSK, Collegeville, Pennsylvania (T.S.P.G.); Preclinical Sciences and Translational Safety, Johnson & Johnson, Janssen Pharmaceuticals, Spring House, Pennsylvania (R.E.); UCB Pharma, Cambridge, Massachusetts (J.E.); Pharmacokinetics, Dynamics and Metabolism, Medicine Design, Pfizer, Inc., Cambridge, Massachusetts (J.R.G.); Research and Development, Bristol Myers Squibb Company, Princeton, New Jersey (S.A.C.); and Drug Safety Research & Development, Pfizer, Inc., Groton, Connecticut (A.K.K.)
| | - Jason Ekert
- Investigative Toxicology, Eli Lilly, Indianapolis, Indiana (T.K.B.); Investigative Toxicology and Pathology, AbbVie, Inc., Chicago, Illinois (T.R.V.F., P.K.M.); Complex In Vitro Models Group, GSK, Collegeville, Pennsylvania (T.S.P.G.); Preclinical Sciences and Translational Safety, Johnson & Johnson, Janssen Pharmaceuticals, Spring House, Pennsylvania (R.E.); UCB Pharma, Cambridge, Massachusetts (J.E.); Pharmacokinetics, Dynamics and Metabolism, Medicine Design, Pfizer, Inc., Cambridge, Massachusetts (J.R.G.); Research and Development, Bristol Myers Squibb Company, Princeton, New Jersey (S.A.C.); and Drug Safety Research & Development, Pfizer, Inc., Groton, Connecticut (A.K.K.)
| | - James R Gosset
- Investigative Toxicology, Eli Lilly, Indianapolis, Indiana (T.K.B.); Investigative Toxicology and Pathology, AbbVie, Inc., Chicago, Illinois (T.R.V.F., P.K.M.); Complex In Vitro Models Group, GSK, Collegeville, Pennsylvania (T.S.P.G.); Preclinical Sciences and Translational Safety, Johnson & Johnson, Janssen Pharmaceuticals, Spring House, Pennsylvania (R.E.); UCB Pharma, Cambridge, Massachusetts (J.E.); Pharmacokinetics, Dynamics and Metabolism, Medicine Design, Pfizer, Inc., Cambridge, Massachusetts (J.R.G.); Research and Development, Bristol Myers Squibb Company, Princeton, New Jersey (S.A.C.); and Drug Safety Research & Development, Pfizer, Inc., Groton, Connecticut (A.K.K.)
| | - Silvi A Chacko
- Investigative Toxicology, Eli Lilly, Indianapolis, Indiana (T.K.B.); Investigative Toxicology and Pathology, AbbVie, Inc., Chicago, Illinois (T.R.V.F., P.K.M.); Complex In Vitro Models Group, GSK, Collegeville, Pennsylvania (T.S.P.G.); Preclinical Sciences and Translational Safety, Johnson & Johnson, Janssen Pharmaceuticals, Spring House, Pennsylvania (R.E.); UCB Pharma, Cambridge, Massachusetts (J.E.); Pharmacokinetics, Dynamics and Metabolism, Medicine Design, Pfizer, Inc., Cambridge, Massachusetts (J.R.G.); Research and Development, Bristol Myers Squibb Company, Princeton, New Jersey (S.A.C.); and Drug Safety Research & Development, Pfizer, Inc., Groton, Connecticut (A.K.K.)
| | - Anna K Kopec
- Investigative Toxicology, Eli Lilly, Indianapolis, Indiana (T.K.B.); Investigative Toxicology and Pathology, AbbVie, Inc., Chicago, Illinois (T.R.V.F., P.K.M.); Complex In Vitro Models Group, GSK, Collegeville, Pennsylvania (T.S.P.G.); Preclinical Sciences and Translational Safety, Johnson & Johnson, Janssen Pharmaceuticals, Spring House, Pennsylvania (R.E.); UCB Pharma, Cambridge, Massachusetts (J.E.); Pharmacokinetics, Dynamics and Metabolism, Medicine Design, Pfizer, Inc., Cambridge, Massachusetts (J.R.G.); Research and Development, Bristol Myers Squibb Company, Princeton, New Jersey (S.A.C.); and Drug Safety Research & Development, Pfizer, Inc., Groton, Connecticut (A.K.K.)
| |
Collapse
|
15
|
Pisapia F, O’Brien D, Tasinato E, Garner KL, Brown CDA. Development of a Highly Differentiated Human Primary Proximal Tubule MPS Model (aProximate MPS Flow). Bioengineering (Basel) 2023; 11:7. [PMID: 38275575 PMCID: PMC10813028 DOI: 10.3390/bioengineering11010007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 12/14/2023] [Accepted: 12/19/2023] [Indexed: 01/27/2024] Open
Abstract
The kidney proximal tubule (PT) mediates renal drug elimination in vivo and is a major site of drug-induced toxicity. To reliably assess drug efficacy, it is crucial to construct a model in which PT functions are replicated. Current animal studies have proven poorly predictive of human outcome. To address this, we developed a physiologically relevant micro-physiological system (MPS) model of the human PT, the aProximate MPS Flow platform (Patent No: G001336.GB). In this model, primary human PT cells (hPTCs) are subjected to fluidic media flow and a shear stress of 0.01-0.2 Pa. We observe that these cells replicate the polarity of hPTCs and exhibit a higher expression of all the key transporters of SLC22A6 (OAT1), SLC22A8 (OAT3), SLC22A2 (OCT2), SLC47A1 (MATE1), SLC22A12 (URAT1), SLC2A9 (GLUT9), ABCB1 (MDR1), ABCC2 (MRP2), LRP2 (megalin), CUBN (cubilin), compared with cells grown under static conditions. Immunofluorescence microscopy confirmed an increase in OAT1, OAT3, and cilia protein expression. Increased sensitivity to nephrotoxic protein cisplatin was observed; creatinine and FITC-albumin uptake was significantly increased under fluidic shear stress conditions. Taken together, these data suggest that growing human PT cells under media flow significantly improves the phenotype and function of hPTC monolayers and has benefits to the utility and near-physiology of the model.
Collapse
Affiliation(s)
- Francesca Pisapia
- Newcells Biotech Ltd., The Biosphere, Draymans Way, Newcastle Helix, Newcastle upon Tyne NE4 5BX, UK; (D.O.); (E.T.); (C.D.A.B.)
| | - Donovan O’Brien
- Newcells Biotech Ltd., The Biosphere, Draymans Way, Newcastle Helix, Newcastle upon Tyne NE4 5BX, UK; (D.O.); (E.T.); (C.D.A.B.)
| | - Elena Tasinato
- Newcells Biotech Ltd., The Biosphere, Draymans Way, Newcastle Helix, Newcastle upon Tyne NE4 5BX, UK; (D.O.); (E.T.); (C.D.A.B.)
- Institute of Genetic Medicine, Newcastle University, International Centre for Life, Central Parkway, Newcastle upon Tyne NE1 3BZ, UK
| | - Kathryn L. Garner
- Newcells Biotech Ltd., The Biosphere, Draymans Way, Newcastle Helix, Newcastle upon Tyne NE4 5BX, UK; (D.O.); (E.T.); (C.D.A.B.)
| | - Colin D. A. Brown
- Newcells Biotech Ltd., The Biosphere, Draymans Way, Newcastle Helix, Newcastle upon Tyne NE4 5BX, UK; (D.O.); (E.T.); (C.D.A.B.)
| |
Collapse
|
16
|
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
|
17
|
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
|
18
|
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
|
19
|
Siricilla S, Hansen CJ, Rogers JH, De D, Simpson CL, Waterson AG, Sulikowski GA, Crockett SL, Boatwright N, Reese J, Paria BC, Newton J, Herington JL. Arrest of mouse preterm labor until term delivery by combination therapy with atosiban and mundulone, a natural product with tocolytic efficacy. Pharmacol Res 2023; 195:106876. [PMID: 37536638 PMCID: PMC10712649 DOI: 10.1016/j.phrs.2023.106876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 07/17/2023] [Accepted: 07/28/2023] [Indexed: 08/05/2023]
Abstract
There is a lack of FDA-approved tocolytics for the management of preterm labor (PL). In prior drug discovery efforts, we identified mundulone and mundulone acetate (MA) as inhibitors of in vitro intracellular Ca2+-regulated myometrial contractility. In this study, we probed the tocolytic potential of these compounds using human myometrial samples and a mouse model of preterm birth. In a phenotypic assay, mundulone displayed greater efficacy, while MA showed greater potency and uterine-selectivity in the inhibition of intracellular-Ca2+ mobilization. Cell viability assays revealed that MA was significantly less cytotoxic. Organ bath and vessel myography studies showed that only mundulone exerted inhibition of myometrial contractions and that neither compounds affected vasoreactivity of ductus arteriosus. A high-throughput combination screen identified that mundulone exhibits synergism with two clinical-tocolytics (atosiban and nifedipine), and MA displayed synergistic efficacy with nifedipine. Of these combinations, mundulone+atosiban demonstrated a significant improvement in the in vitro therapeutic index compared to mundulone alone. The ex vivo and in vivo synergism of mundulone+atosiban was substantiated, yielding greater tocolytic efficacy and potency on myometrial tissue and reduced preterm birth rates in a mouse model of PL compared to each single agent. Treatment with mundulone after mifepristone administration dose-dependently delayed the timing of delivery. Importantly, mundulone+atosiban permitted long-term management of PL, allowing 71% dams to deliver viable pups at term (>day 19, 4-5 days post-mifepristone exposure) without visible maternal and fetal consequences. Collectively, these studies provide a strong foundation for the development of mundulone as a single or combination tocolytic for management of PL.
Collapse
Affiliation(s)
- Shajila Siricilla
- Division of Neonatology, Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Christopher J Hansen
- Division of Neonatology, Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA; Department of Pharmacology, Vanderbilt University, Nashville, TN, USA
| | - Jackson H Rogers
- Division of Neonatology, Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Debasmita De
- Division of Neonatology, Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Carolyn L Simpson
- Division of Neonatology, Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Alex G Waterson
- Department of Pharmacology, Vanderbilt University, Nashville, TN, USA; Department of Chemistry, Vanderbilt University, Nashville, TN, USA; Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, TN, USA
| | - Gary A Sulikowski
- Department of Chemistry, Vanderbilt University, Nashville, TN, USA; Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, TN, USA
| | - Stacey L Crockett
- Division of Neonatology, Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Naoko Boatwright
- Division of Neonatology, Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Jeff Reese
- Division of Neonatology, Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA; Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA; Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN, USA
| | - Bibhash C Paria
- Division of Neonatology, Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - J Newton
- Division of Maternal Fetal Medicine, Department of Obstetrics and Gynecology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Jennifer L Herington
- Division of Neonatology, Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA; Department of Pharmacology, Vanderbilt University, Nashville, TN, USA.
| |
Collapse
|
20
|
Fenibo EO, Selvarajan R, Abia ALK, Matambo T. Medium-chain alkane biodegradation and its link to some unifying attributes of alkB genes diversity. THE SCIENCE OF THE TOTAL ENVIRONMENT 2023; 877:162951. [PMID: 36948313 DOI: 10.1016/j.scitotenv.2023.162951] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 03/14/2023] [Accepted: 03/15/2023] [Indexed: 05/06/2023]
Abstract
Hydrocarbon footprints in the environment, via biosynthesis, natural seepage, anthropogenic activities and accidents, affect the ecosystem and induce a shift in the healthy biogeochemical equilibrium that drives needed ecological services. In addition, these imbalances cause human diseases and reduce animal and microorganism diversity. Microbial bioremediation, which capitalizes on functional genes, is a sustainable mitigation option for cleaning hydrocarbon-impacted environments. This review focuses on the bacterial alkB functional gene, which codes for a non-heme di‑iron monooxygenase (AlkB) with a di‑iron active site that catalyzes C8-C16 medium-chain alkane metabolism. These enzymes are ubiquitous and share common attributes such as being controlled by global transcriptional regulators, being a component of most super hydrocarbon degraders, and their distributions linked to horizontal gene transfer (HGT) events. The phylogenetic approach used in the HGT detection suggests that AlkB tree topology clusters bacteria functionally and that a preferential gradient dictates gene distribution. The alkB gene also acts as a biomarker for bioremediation, although it is found in pristine environments and absent in some hydrocarbon degraders. For instance, a quantitative molecular method has failed to link alkB copy number to contamination concentration levels. This limitation may be due to AlkB homologues, which have other functions besides n-alkane assimilation. Thus, this review, which focuses on Pseudomonas putida GPo1 alkB, shows that AlkB proteins are diverse but have some unifying trends around hydrocarbon-degrading bacteria; it is erroneous to rely on alkB detection alone as a monitoring parameter for hydrocarbon degradation, alkB gene distribution are preferentially distributed among bacteria, and the plausible explanation for AlkB affiliation to broad-spectrum metabolism of hydrocarbons in super-degraders hitherto reported. Overall, this review provides a broad perspective of the ecology of alkB-carrying bacteria and their directed biodegradation pathways.
Collapse
Affiliation(s)
- Emmanuel Oliver Fenibo
- World Bank Africa Centre of Excellence, Centre for Oilfield Chemical Research, University of Port Harcourt, Port Harcourt 500272, Nigeria
| | - Ramganesh Selvarajan
- Laboratory of Extraterrestrial Ocean Systems (LEOS), Institute of Deep-Sea Science and Engineering, Chinese Academy of Sciences, Sanya, China; Department of Environmental Science, University of South Africa, Florida Campus, 1710, South Africa
| | - Akebe Luther King Abia
- Department of Environmental Science, University of South Africa, Florida Campus, 1710, South Africa; Environmental Research Foundation, Westville 3630, South Africa
| | - Tonderayi Matambo
- Institute for the Development of Energy for African Sustainability, University of South Africa, Roodepoort 1709, South Africa.
| |
Collapse
|
21
|
Siricilla S, Hansen CJ, Rogers JH, De D, Simpson CL, Waterson AG, Sulikowski GA, Crockett SL, Boatwright N, Reese J, Paria BC, Newton J, Herington JL. Arrest of mouse preterm labor until term delivery by combination therapy with atosiban and mundulone, a natural product with tocolytic efficacy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.06.543921. [PMID: 37333338 PMCID: PMC10274706 DOI: 10.1101/2023.06.06.543921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
Currently, there is a lack of FDA-approved tocolytics for the management of preterm labor (PL). In prior drug discovery efforts, we identified mundulone and its analog mundulone acetate (MA) as inhibitors of in vitro intracellular Ca 2+ -regulated myometrial contractility. In this study, we probed the tocolytic and therapeutic potential of these small molecules using myometrial cells and tissues obtained from patients receiving cesarean deliveries, as well as a mouse model of PL resulting in preterm birth. In a phenotypic assay, mundulone displayed greater efficacy in the inhibition of intracellular-Ca 2+ from myometrial cells; however, MA showed greater potency and uterine-selectivity, based IC 50 and E max values between myometrial cells compared to aorta vascular smooth muscle cells, a major maternal off-target site of current tocolytics. Cell viability assays revealed that MA was significantly less cytotoxic. Organ bath and vessel myography studies showed that only mundulone exerted concentration-dependent inhibition of ex vivo myometrial contractions and that neither mundulone or MA affected vasoreactivity of ductus arteriosus, a major fetal off-target of current tocolytics. A high-throughput combination screen of in vitro intracellular Ca 2+ -mobilization identified that mundulone exhibits synergism with two clinical-tocolytics (atosiban and nifedipine), and MA displayed synergistic efficacy with nifedipine. Of these synergistic combinations, mundulone + atosiban demonstrated a favorable in vitro therapeutic index (TI)=10, a substantial improvement compared to TI=0.8 for mundulone alone. The ex vivo and in vivo synergism of mundulone and atosiban was substantiated, yielding greater tocolytic efficacy and potency on isolated mouse and human myometrial tissue and reduced preterm birth rates in a mouse model of PL compared to each single agent. Treatment with mundulone 5hrs after mifepristone administration (and PL induction) dose-dependently delayed the timing of delivery. Importantly, mundulone in combination with atosiban (FR 3.7:1, 6.5mg/kg + 1.75mg/kg) permitted long-term management of PL after induction with 30 μg mifepristone, allowing 71% dams to deliver viable pups at term (> day 19, 4-5 days post-mifepristone exposure) without any visible maternal and fetal consequences. Collectively, these studies provide a strong foundation for the future development of mundulone as a stand-alone single- and/or combination-tocolytic therapy for management of PL.
Collapse
|
22
|
Feitor JF, Brazaca LC, Lima AM, Ferreira VG, Kassab G, Bagnato VS, Carrilho E, Cardoso DR. Organ-on-a-Chip for Drug Screening: A Bright Future for Sustainability? A Critical Review. ACS Biomater Sci Eng 2023; 9:2220-2234. [PMID: 37014814 DOI: 10.1021/acsbiomaterials.2c01454] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/05/2023]
Abstract
Globalization has raised concerns about spreading diseases and emphasized the need for quick and efficient methods for drug screening. Established drug efficacy and toxicity approaches have proven obsolete, with a high failure rate in clinical trials. Organ-on-a-chip has emerged as an essential alternative to outdated techniques, precisely simulating important characteristics of organs and predicting drug pharmacokinetics more ethically and efficiently. Although promising, most organ-on-a-chip devices are still manufactured using principles and materials from the micromachining industry. The abusive use of plastic for traditional drug screening methods and device production should be considered when substituting technologies so that the compensation for the generation of plastic waste can be projected. This critical review outlines recent advances for organ-on-a-chip in the industry and estimates the possibility of scaling up its production. Moreover, it analyzes trends in organ-on-a-chip publications and provides suggestions for a more sustainable future for organ-on-a-chip research and production.
Collapse
Affiliation(s)
- Jéssica F Feitor
- Instituto de Química de São Carlos, Universidade de São Paulo, 13566-590 São Carlos, SP, Brazil
| | - Laís C Brazaca
- Instituto de Química de São Carlos, Universidade de São Paulo, 13566-590 São Carlos, SP, Brazil
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, 02138 Massachusetts, United States
| | - Amanda M Lima
- Instituto de Química de São Carlos, Universidade de São Paulo, 13566-590 São Carlos, SP, Brazil
| | - Vinícius G Ferreira
- Instituto de Química de São Carlos, Universidade de São Paulo, 13566-590 São Carlos, SP, Brazil
| | - Giulia Kassab
- Instituto de Física de São Carlos, Universidade de São Paulo, 13566-590 São Carlos, SP, Brazil
| | - Vanderlei S Bagnato
- Instituto de Física de São Carlos, Universidade de São Paulo, 13566-590 São Carlos, SP, Brazil
| | - Emanuel Carrilho
- Instituto de Química de São Carlos, Universidade de São Paulo, 13566-590 São Carlos, SP, Brazil
- Instituto Nacional de Ciência e Tecnologia de Bioanalítica-INCTBio, 13083-970 Campinas, SP, Brazil
| | - Daniel R Cardoso
- Instituto de Química de São Carlos, Universidade de São Paulo, 13566-590 São Carlos, SP, Brazil
| |
Collapse
|
23
|
de Almeida KA, de Moura FR, Lima JV, Garcia EM, Muccillo-Baisch AL, Ramires PF, Penteado JO, da Luz Mathias M, Dias D, da Silva Júnior FMR. Oxidative damage in the Vesper mouse (Calomys laucha) exposed to a simulated oil spill-a multi-organ study. ECOTOXICOLOGY (LONDON, ENGLAND) 2023; 32:502-511. [PMID: 37118609 DOI: 10.1007/s10646-023-02657-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 04/20/2023] [Indexed: 05/11/2023]
Abstract
Small wild mammals have been used to measure the damage caused by exposure to oil-contaminated soil, including deer mice. However, the study of toxic effects of crude oil using oxidative damage biomarkers in the wild rodent Calomys laucha (Vesper mouse) is absent. This investigation aimed to evaluate the effects of acute exposure to contaminated soil with different concentrations of crude oil (0, 1, 2, 4 and 8% w/w), simulating an accidental spill, using oxidative stress biomarkers in the liver, kidneys, lungs, testes, paw muscle, and lymphocytes of C. laucha. Animals exposed to the contaminated soil showed increases in lipid peroxidation and protein carbonylation at the highest exposure concentrations in most organ homogenates analyzed and also in blood cells, but responses to total antioxidant capacity were tissue-dependent. These results showed that acute exposure to oil-contaminated soil caused oxidative damage in C. laucha and indicate these small mammals may be susceptible to suffer the impacts of such contamination in its occurrence region, threatening the species' survival.
Collapse
Affiliation(s)
- Krissia Aparecida de Almeida
- LEFT - Laboratório de Ensaios Farmacológicos e Toxicológicos, Instituto de Ciências Biológicas, Universidade Federal do Rio Grande - FURG, Av. Itália, km 8, Campus Carreiros, Rio Grande, RS, CEP 96203-900, Brazil
| | - Fernando Rafael de Moura
- LEFT - Laboratório de Ensaios Farmacológicos e Toxicológicos, Instituto de Ciências Biológicas, Universidade Federal do Rio Grande - FURG, Av. Itália, km 8, Campus Carreiros, Rio Grande, RS, CEP 96203-900, Brazil
- Programa de Pós Graduação em Ciências da Saúde, Universidade Federal do Rio Grande - FURG, Rua Visconde de Paranaguá, 102, Rio Grande, RS, CEP 96203-900, Brazil
| | - Juliane Ventura Lima
- Programa de Pós Graduação em Ciências Fisiológicas, Universidade Federal do Rio Grande - FURG, Av. Itália, km 8, Campus Carreiros, Rio Grande, RS, CEP 96203-900, Brazil
| | - Edariane Menestrino Garcia
- LEFT - Laboratório de Ensaios Farmacológicos e Toxicológicos, Instituto de Ciências Biológicas, Universidade Federal do Rio Grande - FURG, Av. Itália, km 8, Campus Carreiros, Rio Grande, RS, CEP 96203-900, Brazil
| | - Ana Luíza Muccillo-Baisch
- LEFT - Laboratório de Ensaios Farmacológicos e Toxicológicos, Instituto de Ciências Biológicas, Universidade Federal do Rio Grande - FURG, Av. Itália, km 8, Campus Carreiros, Rio Grande, RS, CEP 96203-900, Brazil
- Programa de Pós Graduação em Ciências da Saúde, Universidade Federal do Rio Grande - FURG, Rua Visconde de Paranaguá, 102, Rio Grande, RS, CEP 96203-900, Brazil
| | - Paula Florencio Ramires
- LEFT - Laboratório de Ensaios Farmacológicos e Toxicológicos, Instituto de Ciências Biológicas, Universidade Federal do Rio Grande - FURG, Av. Itália, km 8, Campus Carreiros, Rio Grande, RS, CEP 96203-900, Brazil
- Programa de Pós Graduação em Ciências da Saúde, Universidade Federal do Rio Grande - FURG, Rua Visconde de Paranaguá, 102, Rio Grande, RS, CEP 96203-900, Brazil
| | - Julia Oliveira Penteado
- LEFT - Laboratório de Ensaios Farmacológicos e Toxicológicos, Instituto de Ciências Biológicas, Universidade Federal do Rio Grande - FURG, Av. Itália, km 8, Campus Carreiros, Rio Grande, RS, CEP 96203-900, Brazil
- Programa de Pós Graduação em Ciências da Saúde, Universidade Federal do Rio Grande - FURG, Rua Visconde de Paranaguá, 102, Rio Grande, RS, CEP 96203-900, Brazil
| | - Maria da Luz Mathias
- Department of Animal Biology, Faculty of Sciences of the University of Lisbon & CESAM - Centre for Environmental and Marine Studies, Campo Grande, 1749-016, Lisbon, Portugal
| | - Deodália Dias
- Department of Animal Biology, Faculty of Sciences of the University of Lisbon & CESAM - Centre for Environmental and Marine Studies, Campo Grande, 1749-016, Lisbon, Portugal
| | - Flavio Manoel Rodrigues da Silva Júnior
- LEFT - Laboratório de Ensaios Farmacológicos e Toxicológicos, Instituto de Ciências Biológicas, Universidade Federal do Rio Grande - FURG, Av. Itália, km 8, Campus Carreiros, Rio Grande, RS, CEP 96203-900, Brazil.
- Programa de Pós Graduação em Ciências da Saúde, Universidade Federal do Rio Grande - FURG, Rua Visconde de Paranaguá, 102, Rio Grande, RS, CEP 96203-900, Brazil.
| |
Collapse
|
24
|
Toropov AA, Barnes DA, Toropova AP, Roncaglioni A, Irvine AR, Masereeuw R, Benfenati E. CORAL Models for Drug-Induced Nephrotoxicity. TOXICS 2023; 11:293. [PMID: 37112520 PMCID: PMC10142465 DOI: 10.3390/toxics11040293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 03/20/2023] [Accepted: 03/21/2023] [Indexed: 06/19/2023]
Abstract
Drug-induced nephrotoxicity is a major cause of kidney dysfunction with potentially fatal consequences. The poor prediction of clinical responses based on preclinical research hampers the development of new pharmaceuticals. This emphasises the need for new methods for earlier and more accurate diagnosis to avoid drug-induced kidney injuries. Computational predictions of drug-induced nephrotoxicity are an attractive approach to facilitate such an assessment and such models could serve as robust and reliable replacements for animal testing. To provide the chemical information for computational prediction, we used the convenient and common SMILES format. We examined several versions of so-called optimal SMILES-based descriptors. We obtained the highest statistical values, considering the specificity, sensitivity and accuracy of the prediction, by applying recently suggested atoms pairs proportions vectors and the index of ideality of correlation, which is a special statistical measure of the predictive potential. Implementation of this tool in the drug development process might lead to safer drugs in the future.
Collapse
Affiliation(s)
- Andrey A. Toropov
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, 20156 Milano, Italy; (A.P.T.); (A.R.); (E.B.)
| | - Devon A. Barnes
- Utrecht Institute for Pharmaceutical Sciences, div. Pharmacology, Universiteitsweg 99, 3584 CG Utrecht, The Netherlands; (D.A.B.); (A.R.I.); (R.M.)
| | - Alla P. Toropova
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, 20156 Milano, Italy; (A.P.T.); (A.R.); (E.B.)
| | - Alessandra Roncaglioni
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, 20156 Milano, Italy; (A.P.T.); (A.R.); (E.B.)
| | - Alasdair R. Irvine
- Utrecht Institute for Pharmaceutical Sciences, div. Pharmacology, Universiteitsweg 99, 3584 CG Utrecht, The Netherlands; (D.A.B.); (A.R.I.); (R.M.)
| | - Rosalinde Masereeuw
- Utrecht Institute for Pharmaceutical Sciences, div. Pharmacology, Universiteitsweg 99, 3584 CG Utrecht, The Netherlands; (D.A.B.); (A.R.I.); (R.M.)
| | - Emilio Benfenati
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, 20156 Milano, Italy; (A.P.T.); (A.R.); (E.B.)
| |
Collapse
|
25
|
Méndez L, Muñoz S, Barros L, Miralles-Pérez B, Romeu M, Ramos-Romero S, Torres JL, Medina I. Combined Intake of Fish Oil and D-Fagomine Prevents High-Fat High-Sucrose Diet-Induced Prediabetes by Modulating Lipotoxicity and Protein Carbonylation in the Kidney. Antioxidants (Basel) 2023; 12:antiox12030751. [PMID: 36978999 PMCID: PMC10045798 DOI: 10.3390/antiox12030751] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2023] [Revised: 03/12/2023] [Accepted: 03/17/2023] [Indexed: 03/30/2023] Open
Abstract
Obesity has been recognized as a major risk factor for chronic kidney disease, insulin resistance being an early common metabolic feature in patients suffering from this syndrome. This study aims to investigate the mechanism underlying the induction of kidney dysfunction and the concomitant onset of insulin resistance by long-term high-fat and sucrose diet feeding in Sprague Dawley rats. To achieve this goal, our study analyzed renal carbonylated protein patterns, ectopic lipid accumulation and fatty acid profiles and correlated them with biometrical and biochemical measurements and other body redox status parameters. Rats fed the obesogenic diet developed a prediabetic state and incipient kidney dysfunction manifested in increased plasma urea concentration and superior levels of renal fat deposition and protein carbonylation. An obesogenic diet increased renal fat by preferentially promoting the accumulation of saturated fat, arachidonic, and docosahexaenoic fatty acids while decreasing oleic acid. Renal lipotoxicity was accompanied by selectively higher carbonylation of proteins involved in the blood pH regulation, i.e., bicarbonate reclamation and synthesis, amino acid, and glucose metabolisms, directly related to the onset of insulin resistance. This study also tested the combination of antioxidant properties of fish oil with the anti-diabetic properties of buckwheat D-Fagomine to counteract diet-induced renal alterations. Results demonstrated that bioactive compounds combined attenuated lipotoxicity, induced more favorable lipid profiles and counteracted the excessive carbonylation of proteins associated with pH regulation in the kidneys, resulting in an inhibition of the progression of the prediabetes state and kidney disease.
Collapse
Affiliation(s)
- Lucía Méndez
- Instituto de Investigaciones Marinas-Consejo Superior de Investigaciones Científicas (IIM-CSIC), Eduardo Cabello 6, E-36208 Vigo, Spain
| | - Silvia Muñoz
- Instituto de Investigaciones Marinas-Consejo Superior de Investigaciones Científicas (IIM-CSIC), Eduardo Cabello 6, E-36208 Vigo, Spain
| | - Lorena Barros
- Instituto de Investigaciones Marinas-Consejo Superior de Investigaciones Científicas (IIM-CSIC), Eduardo Cabello 6, E-36208 Vigo, Spain
| | - Bernat Miralles-Pérez
- Unidad de Farmacología, Facultad de Medicina y Ciencias de la Salud, Universidad Rovira i Virgili, Sant Llorenç 21, E-43201 Reus, Spain
| | - Marta Romeu
- Unidad de Farmacología, Facultad de Medicina y Ciencias de la Salud, Universidad Rovira i Virgili, Sant Llorenç 21, E-43201 Reus, Spain
| | - Sara Ramos-Romero
- Instituto de Química Avanzada de Catalunya-Consejo Superior de Investigaciones Científicas (IQAC-CSIC), Jordi Girona 18-26, E-08034 Barcelona, Spain
- Departamento de Biología Celular, Fisiología e Inmunología, Facultad de Biología, Universidad de Barcelona, E-08028 Barcelona, Spain
| | - Josep Lluís Torres
- Instituto de Química Avanzada de Catalunya-Consejo Superior de Investigaciones Científicas (IQAC-CSIC), Jordi Girona 18-26, E-08034 Barcelona, Spain
| | - Isabel Medina
- Instituto de Investigaciones Marinas-Consejo Superior de Investigaciones Científicas (IIM-CSIC), Eduardo Cabello 6, E-36208 Vigo, Spain
| |
Collapse
|
26
|
Jiang S, Wan F, Lian H, Lu Z, Li X, Cao D, Jiang Y, Li J. Friend or foe? The dual role of triptolide in the liver, kidney, and heart. Biomed Pharmacother 2023; 161:114470. [PMID: 36868013 DOI: 10.1016/j.biopha.2023.114470] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2023] [Revised: 02/23/2023] [Accepted: 02/28/2023] [Indexed: 03/05/2023] Open
Abstract
Triptolide, a controversial natural compound due to its significant pharmacological activities and multiorgan toxicity, has gained much attention since it was isolated from the traditional Chinese herb Tripterygium wilfordii Hook F. However, in addition to its severe toxicity, triptolide also presents powerful therapeutic potency in the same organs, such as the liver, kidney, and heart, which corresponds to the Chinese medicine theory of You Gu Wu Yun (anti-fire with fire) and deeply interested us. To determine the possible mechanisms involved in the dual role of triptolide, we reviewed related articles about the application of triptolide in both physiological and pathological conditions. Inflammation and oxidative stress are the two main ways triptolide exerts different roles, and the cross-talk between NF-κB and Nrf2 may be one of the mechanisms responsible for the dual role of triptolide and may represent the scientific connotation of You Gu Wu Yun. For the first time, we present a review of the dual role of triptolide in the same organ and propose the possible scientific connotation of the Chinese medicine theory of You Gu Wu Yun, hoping to promote the safe and efficient use of triptolide and other controversial medicines.
Collapse
Affiliation(s)
- Shiyuan Jiang
- Department of Histology and Embryology, School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Feng Wan
- Department of Anatomy, School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Hui Lian
- Department of Histology and Embryology, School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Zhihao Lu
- Department of Histology and Embryology, School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Xueming Li
- Department of Histology and Embryology, School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Dan Cao
- Department of Histology and Embryology, School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Yangyu Jiang
- Department of Histology and Embryology, School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Jian Li
- Department of Histology and Embryology, School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 102488, China.
| |
Collapse
|
27
|
Chen J, Song Y, Liu Y, Chen W, Cen Y, You M, Yang G. DBP and BaP co-exposure induces kidney injury via promoting pyroptosis of renal tubular epithelial cells in rats. CHEMOSPHERE 2023; 314:137714. [PMID: 36592837 DOI: 10.1016/j.chemosphere.2022.137714] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 12/19/2022] [Accepted: 12/29/2022] [Indexed: 06/17/2023]
Abstract
Dibutyl phthalate (DBP) and benzo(a)pyrene (BaP) are widespread environmental and foodborne contaminants that have detrimental effects on human health. Although people are often simultaneously exposed to DBP and BaP via the intake of polluted food and water, the combined effects on the kidney and potential mechanisms remain unclear. Hence, we treated rats with DBP and BaP for 90 days to investigate their effects on kidney histopathology and function. We also investigated the levels of paramount proteins and genes involved in pyroptosis and TLR4/NF-κB p65 signaling in the kidney. Our research showed that combined exposure to DBP and BaP triggered more severe histopathological and renal function abnormalities than in those exposed to DBP or BaP alone. Simultaneously, combined exposure to DBP and BaP enhanced the excretion of IL-1β and IL-18, along with the release of LDH in rat renal tubular epithelial cells (RTECs). Moreover, combined exposure to DBP and BaP increased the expression of pyroptosis marker molecules, including NLRP3, ASC, cleaved-Caspase-1, and GSDMD. Meanwhile, the combination of DBP and BaP activated TLR4/NF-κB signaling in the kidney. Taken together, the combined exposure to DBP and BaP causes more severe kidney injury than that caused by DBP or BaP exposure separately. In addition, pyroptosis of RTECs regulated by TLR4/NF-κB signaling may add to the kidney damage triggered by combined exposure to DBP and BaP.
Collapse
Affiliation(s)
- Jing Chen
- School of Public Health, The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, Guizhou, 550025, China
| | - Yawen Song
- School of Public Health, The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, Guizhou, 550025, China
| | - Yining Liu
- School of Public Health, The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, Guizhou, 550025, China
| | - Wenyan Chen
- School of Public Health, The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, Guizhou, 550025, China
| | - Yanli Cen
- School of Public Health, The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, Guizhou, 550025, China
| | - Mingdan You
- School of Public Health, The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, Guizhou, 550025, China.
| | - Guanghong Yang
- Guizhou Provincial Center for Disease Control and Prevention, Guiyang, Guizhou, 550004, China; School of Public Health, Guizhou Medical University, Guiyang, Guizhou, 550025, China.
| |
Collapse
|
28
|
Pearson A, Haenni D, Bouitbir J, Hunt M, Payne BAI, Sachdeva A, Hung RKY, Post FA, Connolly J, Nlandu-Khodo S, Jankovic N, Bugarski M, Hall AM. Integration of High-Throughput Imaging and Multiparametric Metabolic Profiling Reveals a Mitochondrial Mechanism of Tenofovir Toxicity. FUNCTION (OXFORD, ENGLAND) 2022; 4:zqac065. [PMID: 36654930 PMCID: PMC9840465 DOI: 10.1093/function/zqac065] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 12/14/2022] [Accepted: 12/20/2022] [Indexed: 12/26/2022]
Abstract
Nephrotoxicity is a major cause of kidney disease and failure in drug development, but understanding of cellular mechanisms is limited, highlighting the need for better experimental models and methodological approaches. Most nephrotoxins damage the proximal tubule (PT), causing functional impairment of solute reabsorption and systemic metabolic complications. The antiviral drug tenofovir disoproxil fumarate (TDF) is an archetypal nephrotoxin, inducing mitochondrial abnormalities and urinary solute wasting, for reasons that were previously unclear. Here, we developed an automated, high-throughput imaging pipeline to screen the effects of TDF on solute transport and mitochondrial morphology in human-derived RPTEC/TERT1 cells, and leveraged this to generate realistic models of functional toxicity. By applying multiparametric metabolic profiling-including oxygen consumption measurements, metabolomics, and transcriptomics-we elucidated a highly robust molecular fingerprint of TDF exposure. Crucially, we identified that the active metabolite inhibits complex V (ATP synthase), and that TDF treatment causes rapid, dose-dependent loss of complex V activity and expression. Moreover, we found evidence of complex V suppression in kidney biopsies from humans with TDF toxicity. Thus, we demonstrate an effective and convenient experimental approach to screen for disease relevant functional defects in kidney cells in vitro, and reveal a new paradigm for understanding the pathogenesis of a substantial cause of nephrotoxicity.
Collapse
Affiliation(s)
- Adam Pearson
- Institute of Anatomy, University of Zurich, Winterthurerstrasse 190, CH-8057 Zurich, Switzerland
| | - Dominik Haenni
- Center for Microscopy and Image Analysis, University of Zurich, Winterthurerstrasse 190, CH-8057 Zurich, Switzerland
| | - Jamal Bouitbir
- Division of Molecular and Systems Toxicology, Department of Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 50, CH-4056 Basel, Switzerland
| | - Matthew Hunt
- Wellcome Centre for Mitochondrial Research, Newcastle University, Framlington Place, Newcastle upon Tyne, NE2 4HH, UK
| | - Brendan A I Payne
- Wellcome Centre for Mitochondrial Research, Newcastle University, Framlington Place, Newcastle upon Tyne, NE2 4HH, UK,Department of Infection and Tropical Medicine, Royal Victoria Infirmary, Queen Victoria Road, Newcastle upon Tyne NE1 4LP, UK
| | - Ashwin Sachdeva
- Genito-Urinary Cancer Research Group, Division of Cancer Sciences, University of Manchester, Manchester, M20 4GJ, UK,Department of Surgery, The Christie Hospital NHS Foundation Trust, 550 Wilmslow Road, Manchester M20 4BX, UK
| | - Rachel K Y Hung
- King’s College Hospital and School of Immunology & Microbial Sciences, King’s College London, London, SE5 8AF, UK
| | - Frank A Post
- King’s College Hospital and School of Immunology & Microbial Sciences, King’s College London, London, SE5 8AF, UK
| | - John Connolly
- UCL Centre for Nephrology, Royal Free Hospital, Rowland Hill Street, London NW3 2PF, UK
| | - Stellor Nlandu-Khodo
- Institute of Physiology, University of Zurich, Winterthurerstrasse 190, CH-8057 Zurich, Switzerland
| | - Nevena Jankovic
- Institute of Anatomy, University of Zurich, Winterthurerstrasse 190, CH-8057 Zurich, Switzerland
| | - Milica Bugarski
- Institute of Anatomy, University of Zurich, Winterthurerstrasse 190, CH-8057 Zurich, Switzerland
| | | |
Collapse
|
29
|
Vidal Yucha SE, Quackenbush D, Chu T, Lo F, Sutherland JJ, Kuzu G, Roberts C, Luna F, Barnes SW, Walker J, Kuss P. "3D, human renal proximal tubule (RPTEC-TERT1) organoids 'tubuloids' for translatable evaluation of nephrotoxins in high-throughput". PLoS One 2022; 17:e0277937. [PMID: 36409750 PMCID: PMC9678317 DOI: 10.1371/journal.pone.0277937] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Accepted: 11/07/2022] [Indexed: 11/22/2022] Open
Abstract
The importance of human cell-based in vitro tools to drug development that are robust, accurate, and predictive cannot be understated. There has been significant effort in recent years to develop such platforms, with increased interest in 3D models that can recapitulate key aspects of biology that 2D models might not be able to deliver. We describe the development of a 3D human cell-based in vitro assay for the investigation of nephrotoxicity, using RPTEC-TERT1 cells. These RPTEC-TERT1 proximal tubule organoids 'tubuloids' demonstrate marked differences in physiologically relevant morphology compared to 2D monolayer cells, increased sensitivity to nephrotoxins observable via secreted protein, and with a higher degree of similarity to native human kidney tissue. Finally, tubuloids incubated with nephrotoxins demonstrate altered Na+/K+-ATPase signal intensity, a potential avenue for a high-throughput, translatable nephrotoxicity assay.
Collapse
Affiliation(s)
- Sarah E. Vidal Yucha
- Novartis Institutes for BioMedical Research-San Diego, La Jolla, CA, United States of America
- * E-mail:
| | - Doug Quackenbush
- Novartis Institutes for BioMedical Research-San Diego, La Jolla, CA, United States of America
| | - Tiffany Chu
- Novartis Institutes for BioMedical Research-San Diego, La Jolla, CA, United States of America
| | - Frederick Lo
- Novartis Institutes for BioMedical Research-San Diego, La Jolla, CA, United States of America
| | - Jeffrey J. Sutherland
- Novartis Institutes for BioMedical Research-Cambridge, Cambridge, MA, United States of America
| | - Guray Kuzu
- Novartis Institutes for BioMedical Research-San Diego, La Jolla, CA, United States of America
| | - Christopher Roberts
- Novartis Institutes for BioMedical Research-San Diego, La Jolla, CA, United States of America
| | - Fabio Luna
- Novartis Institutes for BioMedical Research-San Diego, La Jolla, CA, United States of America
| | - S. Whitney Barnes
- Novartis Institutes for BioMedical Research-San Diego, La Jolla, CA, United States of America
| | - John Walker
- Novartis Institutes for BioMedical Research-San Diego, La Jolla, CA, United States of America
| | - Pia Kuss
- Novartis Institutes for BioMedical Research-San Diego, La Jolla, CA, United States of America
| |
Collapse
|
30
|
Lourenço D, Lopes R, Pestana C, Queirós AC, João C, Carneiro EA. Patient-Derived Multiple Myeloma 3D Models for Personalized Medicine-Are We There Yet? Int J Mol Sci 2022; 23:12888. [PMID: 36361677 PMCID: PMC9657251 DOI: 10.3390/ijms232112888] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 10/19/2022] [Accepted: 10/22/2022] [Indexed: 12/03/2023] Open
Abstract
Despite the wide variety of existing therapies, multiple myeloma (MM) remains a disease with dismal prognosis. Choosing the right treatment for each patient remains one of the major challenges. A new approach being explored is the use of ex vivo models for personalized medicine. Two-dimensional culture or animal models often fail to predict clinical outcomes. Three-dimensional ex vivo models using patients' bone marrow (BM) cells may better reproduce the complexity and heterogeneity of the BM microenvironment. Here, we review the strengths and limitations of currently existing patient-derived ex vivo three-dimensional MM models. We analyze their biochemical and biophysical properties, molecular and cellular characteristics, as well as their potential for drug testing and identification of disease biomarkers. Furthermore, we discuss the remaining challenges and give some insight on how to achieve a more biomimetic and accurate MM BM model. Overall, there is still a need for standardized culture methods and refined readout techniques. Including both myeloma and other cells of the BM microenvironment in a simple and reproducible three-dimensional scaffold is the key to faithfully mapping and examining the relationship between these players in MM. This will allow a patient-personalized profile, providing a powerful tool for clinical and research applications.
Collapse
Affiliation(s)
- Diana Lourenço
- Myeloma Lymphoma Research Group—Champalimaud Experimental Clinical Research Programme of Champalimaud Foundation, 1400-038 Lisbon, Portugal
- Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Raquel Lopes
- Myeloma Lymphoma Research Group—Champalimaud Experimental Clinical Research Programme of Champalimaud Foundation, 1400-038 Lisbon, Portugal
- Faculty of Medicine, University of Lisbon, 1649-028 Lisbon, Portugal
| | - Carolina Pestana
- Myeloma Lymphoma Research Group—Champalimaud Experimental Clinical Research Programme of Champalimaud Foundation, 1400-038 Lisbon, Portugal
- Centre of Statistics and Its Applications, Faculty of Sciences, University of Lisbon, 1749-016 Lisbon, Portugal
| | - Ana C. Queirós
- Myeloma Lymphoma Research Group—Champalimaud Experimental Clinical Research Programme of Champalimaud Foundation, 1400-038 Lisbon, Portugal
| | - Cristina João
- Myeloma Lymphoma Research Group—Champalimaud Experimental Clinical Research Programme of Champalimaud Foundation, 1400-038 Lisbon, Portugal
- Faculty of Medical Sciences, NOVA Medical School, 1169-056 Lisbon, Portugal
- Hemato-Oncology Department of Champalimaud Foundation, 1400-038 Lisbon, Portugal
| | - Emilie Arnault Carneiro
- Myeloma Lymphoma Research Group—Champalimaud Experimental Clinical Research Programme of Champalimaud Foundation, 1400-038 Lisbon, Portugal
| |
Collapse
|
31
|
García-Niño WR, Ibarra-Lara L, Cuevas-Magaña MY, Sánchez-Mendoza A, Armada E. Protective activities of ellagic acid and urolithins against kidney toxicity of environmental pollutants: A review. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2022; 95:103960. [PMID: 35995378 DOI: 10.1016/j.etap.2022.103960] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 08/07/2022] [Accepted: 08/17/2022] [Indexed: 06/15/2023]
Abstract
Oxidative stress and inflammation are two possible mechanisms related to nephrotoxicity caused by environmental pollutants. Ellagic acid, a powerful antioxidant phytochemical, may have great relevance in mitigating pollutant-induced nephrotoxicity and preventing the progression of kidney disease. This review discusses the latest findings on the protective effects of ellagic acid, its metabolic derivatives, the urolithins, against kidney toxicity caused by heavy metals, pesticides, mycotoxins, and organic air pollutants. We describe the chelating, antioxidant, anti-inflammatory, antifibrotic, antiautophagic, and antiapoptotic properties of ellagic acid to attenuate nephrotoxicity. Furthermore, we present the molecular targets and signaling pathways that are regulated by these antioxidants, and suggest some others that should be explored. Nevertheless, the number of reports is still limited to establish the efficacy of ellagic acid against kidney damage induced by environmental pollutants. Therefore, additional preclinical studies on this topic are required, as well as the development of well-designed clinical trials.
Collapse
Affiliation(s)
- Wylly Ramsés García-Niño
- Department of Cardiovascular Biomedicine, National Institute of Cardiology Ignacio Chávez, Mexico City 14080, Mexico.
| | - Luz Ibarra-Lara
- Department of Pharmacology, National Institute of Cardiology Ignacio Chávez, Mexico City 14080, Mexico
| | - Mayra Yael Cuevas-Magaña
- Department of Cardiovascular Biomedicine, National Institute of Cardiology Ignacio Chávez, Mexico City 14080, Mexico
| | - Alicia Sánchez-Mendoza
- Department of Pharmacology, National Institute of Cardiology Ignacio Chávez, Mexico City 14080, Mexico
| | - Elisabeth Armada
- Department of Plant Molecular Biology, Institute of Biotechnology, National Autonomous University of Mexico, Cuernavaca 62210, Morelos, Mexico
| |
Collapse
|
32
|
Apical Medium Flow Influences the Morphology and Physiology of Human Proximal Tubular Cells in a Microphysiological System. BIOENGINEERING (BASEL, SWITZERLAND) 2022; 9:bioengineering9100516. [PMID: 36290484 PMCID: PMC9598399 DOI: 10.3390/bioengineering9100516] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Accepted: 09/16/2022] [Indexed: 12/28/2022]
Abstract
There is a lack of physiologically relevant in vitro human kidney models for disease modelling and detecting drug-induced effects given the limited choice of cells and difficulty implementing quasi-physiological culture conditions. We investigated the influence of fluid shear stress on primary human renal proximal tubule epithelial cells (RPTECs) cultured in the micro-physiological Vitrofluid device. This system houses cells seeded on semipermeable membranes and can be connected to a regulable pump that enables controlled, unidirectional flow. After 7 days in culture, RPTECs maintained physiological characteristics such as barrier integrity, protein uptake ability, and expression of specific transporters (e.g., aquaporin-1). Exposure to constant apical side flow did not cause cytotoxicity, cell detachment, or intracellular reactive oxygen species accumulation. However, unidirectional flow profoundly affected cell morphology and led to primary cilia lengthening and alignment in the flow direction. The dynamic conditions also reduced cell proliferation, altered plasma membrane leakiness, increased cytokine secretion, and repressed histone deacetylase 6 and kidney injury molecule 1 expression. Cells under flow also remained susceptible to colistin-induced toxicity. Collectively, the results suggest that dynamic culture conditions in the Vitrofluid system promote a more differentiated phenotype in primary human RPTECs and represent an improved in vitro kidney model.
Collapse
|
33
|
Kim H, Lee JB, Kim K, Sung GY. Effect of shear stress on the proximal tubule-on-a-chip for multi-organ microphysiological system. J IND ENG CHEM 2022. [DOI: 10.1016/j.jiec.2022.08.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
|
34
|
Gao P, Shen X, Zhang X, Jiang C, Zhang S, Zhou X, Schüssler-Fiorenza Rose SM, Snyder M. Precision environmental health monitoring by longitudinal exposome and multi-omics profiling. Genome Res 2022; 32:1199-1214. [PMID: 35667843 PMCID: PMC9248886 DOI: 10.1101/gr.276521.121] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 04/18/2022] [Indexed: 11/24/2022]
Abstract
Conventional environmental health studies have primarily focused on limited environmental stressors at the population level, which lacks the power to dissect the complexity and heterogeneity of individualized environmental exposures. Here, as a pilot case study, we integrated deep-profiled longitudinal personal exposome and internal multi-omics to systematically investigate how the exposome shapes a single individual's phenome. We annotated thousands of chemical and biological components in the personal exposome cloud and found they were significantly correlated with thousands of internal biomolecules, which was further cross-validated using corresponding clinical data. Our results showed that agrochemicals and fungi predominated in the highly diverse and dynamic personal exposome, and the biomolecules and pathways related to the individual's immune system, kidney, and liver were highly associated with the personal external exposome. Overall, this data-driven longitudinal monitoring study shows the potential dynamic interactions between the personal exposome and internal multi-omics, as well as the impact of the exposome on precision health by producing abundant testable hypotheses.
Collapse
Affiliation(s)
- Peng Gao
- Department of Genetics, Stanford University School of Medicine, Stanford, California 94304, USA
| | - Xiaotao Shen
- Department of Genetics, Stanford University School of Medicine, Stanford, California 94304, USA
| | - Xinyue Zhang
- Department of Genetics, Stanford University School of Medicine, Stanford, California 94304, USA
| | - Chao Jiang
- Life Sciences Institute, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Sai Zhang
- Department of Genetics, Stanford University School of Medicine, Stanford, California 94304, USA
| | - Xin Zhou
- Department of Genetics, Stanford University School of Medicine, Stanford, California 94304, USA
| | | | - Michael Snyder
- Department of Genetics, Stanford University School of Medicine, Stanford, California 94304, USA
| |
Collapse
|
35
|
Liu C, Zhou S, Bai W, Shi L, Li X. Protective effect of food derived nutrients on cisplatin nephrotoxicity and its mechanism. Food Funct 2022; 13:4839-4860. [PMID: 35416186 DOI: 10.1039/d1fo04391a] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Platinum-based metal complexes, especially cisplatin (cis-diamminedichloroplatinum II, CDDP), possess strong anticancer properties and a broad anticancer spectrum. However, the clinical application of CDDP has been limited by its side effects including nephrotoxicity, ototoxicity, and neurotoxicity. Furthermore, the therapeutic effects of current clinical protocols are imperfect. Accordingly, it is essential to identify key targets and effective clinical protocols to restrict CDDP-induced nephrotoxicity. Herein, we first analyzed the relevant molecular mechanisms during the process of CDDP-induced nephrotoxicity including oxidative stress, apoptosis, and inflammation. Evidence from current studies was collected and potential targets and clinical protocols are summarized. The evidence indicates an efficacious role of nutrition-based substances in CDDP-induced renal injury.
Collapse
Affiliation(s)
- Chaofan Liu
- Institute of Food Safety and Nutrition, Jinan University, Guangzhou 510632, PR China.
| | - Sajin Zhou
- Institute of Food Safety and Nutrition, Jinan University, Guangzhou 510632, PR China.
| | - Weibin Bai
- Institute of Food Safety and Nutrition, Jinan University, Guangzhou 510632, PR China.
| | - Lei Shi
- Institute of Food Safety and Nutrition, Jinan University, Guangzhou 510632, PR China.
| | - Xiaoling Li
- Institute of Food Safety and Nutrition, Jinan University, Guangzhou 510632, PR China.
| |
Collapse
|
36
|
Chang X, Tan YM, Allen DG, Bell S, Brown PC, Browning L, Ceger P, Gearhart J, Hakkinen PJ, Kabadi SV, Kleinstreuer NC, Lumen A, Matheson J, Paini A, Pangburn HA, Petersen EJ, Reinke EN, Ribeiro AJS, Sipes N, Sweeney LM, Wambaugh JF, Wange R, Wetmore BA, Mumtaz M. IVIVE: Facilitating the Use of In Vitro Toxicity Data in Risk Assessment and Decision Making. TOXICS 2022; 10:232. [PMID: 35622645 PMCID: PMC9143724 DOI: 10.3390/toxics10050232] [Citation(s) in RCA: 59] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 04/24/2022] [Indexed: 02/04/2023]
Abstract
During the past few decades, the science of toxicology has been undergoing a transformation from observational to predictive science. New approach methodologies (NAMs), including in vitro assays, in silico models, read-across, and in vitro to in vivo extrapolation (IVIVE), are being developed to reduce, refine, or replace whole animal testing, encouraging the judicious use of time and resources. Some of these methods have advanced past the exploratory research stage and are beginning to gain acceptance for the risk assessment of chemicals. A review of the recent literature reveals a burst of IVIVE publications over the past decade. In this review, we propose operational definitions for IVIVE, present literature examples for several common toxicity endpoints, and highlight their implications in decision-making processes across various federal agencies, as well as international organizations, including those in the European Union (EU). The current challenges and future needs are also summarized for IVIVE. In addition to refining and reducing the number of animals in traditional toxicity testing protocols and being used for prioritizing chemical testing, the goal to use IVIVE to facilitate the replacement of animal models can be achieved through their continued evolution and development, including a strategic plan to qualify IVIVE methods for regulatory acceptance.
Collapse
Affiliation(s)
- Xiaoqing Chang
- Inotiv-RTP, 601 Keystone Park Drive, Suite 200, Morrisville, NC 27560, USA; (X.C.); (D.G.A.); (S.B.); (L.B.); (P.C.)
| | - Yu-Mei Tan
- U.S. Environmental Protection Agency, Office of Pesticide Programs, 109 T.W. Alexander Drive, Durham, NC 27709, USA;
| | - David G. Allen
- Inotiv-RTP, 601 Keystone Park Drive, Suite 200, Morrisville, NC 27560, USA; (X.C.); (D.G.A.); (S.B.); (L.B.); (P.C.)
| | - Shannon Bell
- Inotiv-RTP, 601 Keystone Park Drive, Suite 200, Morrisville, NC 27560, USA; (X.C.); (D.G.A.); (S.B.); (L.B.); (P.C.)
| | - Paul C. Brown
- U.S. Food and Drug Administration, Center for Drug Evaluation and Research, 10903 New Hampshire Avenue, Silver Spring, MD 20903, USA; (P.C.B.); (A.J.S.R.); (R.W.)
| | - Lauren Browning
- Inotiv-RTP, 601 Keystone Park Drive, Suite 200, Morrisville, NC 27560, USA; (X.C.); (D.G.A.); (S.B.); (L.B.); (P.C.)
| | - Patricia Ceger
- Inotiv-RTP, 601 Keystone Park Drive, Suite 200, Morrisville, NC 27560, USA; (X.C.); (D.G.A.); (S.B.); (L.B.); (P.C.)
| | - Jeffery Gearhart
- The Henry M. Jackson Foundation, Air Force Research Laboratory, 711 Human Performance Wing, Wright-Patterson Air Force Base, OH 45433, USA;
| | - Pertti J. Hakkinen
- National Library of Medicine, National Center for Biotechnology Information, 8600 Rockville Pike, Bethesda, MD 20894, USA;
| | - Shruti V. Kabadi
- U.S. Food and Drug Administration, Center for Food Safety and Applied Nutrition, Office of Food Additive Safety, 5001 Campus Drive, HFS-275, College Park, MD 20740, USA;
| | - Nicole C. Kleinstreuer
- National Institute of Environmental Health Sciences, National Toxicology Program Interagency Center for the Evaluation of Alternative Toxicological Methods, P.O. Box 12233, Research Triangle Park, NC 27709, USA;
| | - Annie Lumen
- U.S. Food and Drug Administration, National Center for Toxicological Research, 3900 NCTR Road, Jefferson, AR 72079, USA;
| | - Joanna Matheson
- U.S. Consumer Product Safety Commission, Division of Toxicology and Risk Assessment, 5 Research Place, Rockville, MD 20850, USA;
| | - Alicia Paini
- European Commission, Joint Research Centre (JRC), 21027 Ispra, Italy;
| | - Heather A. Pangburn
- Air Force Research Laboratory, 711 Human Performance Wing, 2729 R Street, Area B, Building 837, Wright-Patterson Air Force Base, OH 45433, USA;
| | - Elijah J. Petersen
- U.S. Department of Commerce, National Institute of Standards and Technology, 100 Bureau Drive, Gaithersburg, MD 20899, USA;
| | - Emily N. Reinke
- U.S. Army Public Health Center, 8252 Blackhawk Rd., Aberdeen Proving Ground, MD 21010, USA;
| | - Alexandre J. S. Ribeiro
- U.S. Food and Drug Administration, Center for Drug Evaluation and Research, 10903 New Hampshire Avenue, Silver Spring, MD 20903, USA; (P.C.B.); (A.J.S.R.); (R.W.)
| | - Nisha Sipes
- U.S. Environmental Protection Agency, Center for Computational Toxicology and Exposure, 109 TW Alexander Dr., Research Triangle Park, NC 27711, USA; (N.S.); (J.F.W.); (B.A.W.)
| | - Lisa M. Sweeney
- UES, Inc., 4401 Dayton-Xenia Road, Beavercreek, OH 45432, Assigned to Air Force Research Laboratory, 711 Human Performance Wing, Wright-Patterson Air Force Base, OH 45433, USA;
| | - John F. Wambaugh
- U.S. Environmental Protection Agency, Center for Computational Toxicology and Exposure, 109 TW Alexander Dr., Research Triangle Park, NC 27711, USA; (N.S.); (J.F.W.); (B.A.W.)
| | - Ronald Wange
- U.S. Food and Drug Administration, Center for Drug Evaluation and Research, 10903 New Hampshire Avenue, Silver Spring, MD 20903, USA; (P.C.B.); (A.J.S.R.); (R.W.)
| | - Barbara A. Wetmore
- U.S. Environmental Protection Agency, Center for Computational Toxicology and Exposure, 109 TW Alexander Dr., Research Triangle Park, NC 27711, USA; (N.S.); (J.F.W.); (B.A.W.)
| | - Moiz Mumtaz
- Agency for Toxic Substances and Disease Registry, Office of the Associate Director for Science, 1600 Clifton Road, S102-2, Atlanta, GA 30333, USA
| |
Collapse
|
37
|
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: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [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
|
38
|
Unwin RJ. Toxic nephropathy: Adverse renal effects caused by drugs. Eur J Intern Med 2022; 96:20-25. [PMID: 34607721 DOI: 10.1016/j.ejim.2021.09.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 08/29/2021] [Accepted: 09/15/2021] [Indexed: 01/02/2023]
Abstract
This is a brief overview of toxic nephropathy, which is an increasingly recognised problem with the continual introduction of new drugs and novel drug modalities, especially in oncology, and the risks associated with polypharmacy in many patients; although it is important to remember that it may not always be caused by a drug. It is also important to note that several possibly harmful drugs are now available without prescription ('over-the-counter') and can be purchased easily over the internet, including some poorly characterised herbal remedies. Knowing exactly what our patients are taking as medication is not always easy and patients often fail to mention drugs that may not have been prescribed by a doctor or recommended by a pharmacist. Moreover, patients with several comorbidities often require care from more than one doctor in other specialties, which can also lead to drug prescribing in isolation. This article will summarise some key aspects of drug nephrotoxicity and provide a few clinical pointers to consider, bearing in mind that there is rarely any antidote available, and effective treatment relies on early detection, prompt drug withdrawal, and supportive care. This short review is intended only as a primer to highlight some of the more practical aspects of toxic nephropathy; its content is based on a lecture delivered during the 2021 European Congress of Internal Medicine.
Collapse
Affiliation(s)
- Robert J Unwin
- Department of Renal Medicine, Royal Free Hospital Trust, University College London, Rowland Hill Street, London NW3 2PF, UK.
| |
Collapse
|
39
|
Yu P, Duan Z, Liu S, Pachon I, Ma J, Hemstreet GP, Zhang Y. Drug-Induced Nephrotoxicity Assessment in 3D Cellular Models. MICROMACHINES 2021; 13:mi13010003. [PMID: 35056167 PMCID: PMC8780064 DOI: 10.3390/mi13010003] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 12/11/2021] [Accepted: 12/17/2021] [Indexed: 12/19/2022]
Abstract
The kidneys are often involved in adverse effects and toxicity caused by exposure to foreign compounds, chemicals, and drugs. Early predictions of these influences are essential to facilitate new, safe drugs to enter the market. However, in current drug treatments, drug-induced nephrotoxicity accounts for 1/4 of reported serious adverse reactions, and 1/3 of them are attributable to antibiotics. Drug-induced nephrotoxicity is driven by multiple mechanisms, including altered glomerular hemodynamics, renal tubular cytotoxicity, inflammation, crystal nephropathy, and thrombotic microangiopathy. Although the functional proteins expressed by renal tubules that mediate drug sensitivity are well known, current in vitro 2D cell models do not faithfully replicate the morphology and intact renal tubule function, and therefore, they do not replicate in vivo nephrotoxicity. The kidney is delicate and complex, consisting of a filter unit and a tubular part, which together contain more than 20 different cell types. The tubular epithelium is highly polarized, and maintaining cellular polarity is essential for the optimal function and response to environmental signals. Cell polarity depends on the communication between cells, including paracrine and autocrine signals, as well as biomechanical and chemotaxis processes. These processes affect kidney cell proliferation, migration, and differentiation. For drug disposal research, the microenvironment is essential for predicting toxic reactions. This article reviews the mechanism of drug-induced kidney injury, the types of nephrotoxicity models (in vivo and in vitro models), and the research progress related to drug-induced nephrotoxicity in three-dimensional (3D) cellular culture models.
Collapse
Affiliation(s)
- Pengfei Yu
- Difficult & Complicated Liver Diseases and Artificial Liver Center, Fourth Department of Liver Disease, Beijing Youan Hospital, Capital Medical University, Beijing 100069, China; (P.Y.); (Z.D.); (S.L.)
- Beijing Municipal Key Laboratory of Liver Failure and Artificial Liver Treatment Research, Beijing Youan Hospital, Capital Medical University, Beijing 100069, China
| | - Zhongping Duan
- Difficult & Complicated Liver Diseases and Artificial Liver Center, Fourth Department of Liver Disease, Beijing Youan Hospital, Capital Medical University, Beijing 100069, China; (P.Y.); (Z.D.); (S.L.)
- Beijing Municipal Key Laboratory of Liver Failure and Artificial Liver Treatment Research, Beijing Youan Hospital, Capital Medical University, Beijing 100069, China
| | - Shuang Liu
- Difficult & Complicated Liver Diseases and Artificial Liver Center, Fourth Department of Liver Disease, Beijing Youan Hospital, Capital Medical University, Beijing 100069, China; (P.Y.); (Z.D.); (S.L.)
- Beijing Municipal Key Laboratory of Liver Failure and Artificial Liver Treatment Research, Beijing Youan Hospital, Capital Medical University, Beijing 100069, China
| | - Ivan Pachon
- Wake Forest Institute for Regenerative Medicine, Wake Forest University Health Sciences, Winston-Salem, NC 27157, USA;
| | - Jianxing Ma
- Department of Biochemistry, Wake Forest University Health Sciences, Winston-Salem, NC 27157, USA;
| | | | - Yuanyuan Zhang
- Wake Forest Institute for Regenerative Medicine, Wake Forest University Health Sciences, Winston-Salem, NC 27157, USA;
- Correspondence: ; Tel.: +1-336-713-1189
| |
Collapse
|
40
|
Leta B, Kenenisa C, Wondimnew T, Sime T. Evaluation of Renoprotective Effects of Our Locally Grown Green Coffee Beans against Cisplatin-Induced Nephrotoxicity in Swiss Albino Mice. Int J Nephrol 2021; 2021:2805068. [PMID: 34676116 PMCID: PMC8526242 DOI: 10.1155/2021/2805068] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 09/18/2021] [Accepted: 09/29/2021] [Indexed: 12/29/2022] Open
Abstract
INTRODUCTION Nephrotoxicity is the most common and severe side effect of cisplatin. Cisplatin causes nephrotoxicity through free radical production and debilitating cellular antioxidant capacity. Coffee is a commonly consumed drink and its ingredients have antioxidant roles that could bring benefits to patients affected by nephrotoxicity. Thus, the present study aimed to investigate the renoprotective effects of our locally grown green coffee beans against cisplatin-induced nephrotoxicity in Swiss albino mice. METHODS The posttest only control group design was employed on a total of thirty male Swiss albino mice. The mice were divided into five groups: group I (normal control group) received distilled water; group II (negative control group) received distilled water; and groups III-V (treatment groups) received 100, 200, and 300 mg/kg BW/day of green coffee bean extract for 14 days, respectively. Nephrotoxicity was induced in groups II-V by a single intraperitoneal injection of cisplatin (7.5 mg/kg). All mice were sacrificed after 14 days and blood was drawn to evaluate kidney function tests (serum creatinine and serum blood urea nitrogen). Besides, body weight, relative kidney weight, and kidney histopathology were investigated. RESULT Our results showed that treatment of cisplatin alone (group II mice) significantly increased serum creatinine, serum blood urea nitrogen, relative kidney weight, and pathological damage to the kidney with a decrease in final body weight. However, low-dose green coffee beans (group III), medium-dose green coffee beans (group IV), and high-dose green coffee beans (group V) mice showed a significant dose-dependent decrease in serum creatinine, serum blood urea nitrogen, and relative kidney weight. Furthermore, the dose-dependent treatment with green coffee bean extract prevented the decrease in body weight gain and pathological damage to the kidney in mice. CONCLUSION Our locally grown green coffee beans brought a dose-dependent ameliorative effect and a promising preventive approach against cisplatin-induced kidney damage in mice.
Collapse
Affiliation(s)
- Bati Leta
- Department of Biomedical Sciences, Faculty of Medical Sciences, Institute of Health Sciences, Jimma University, Jimma, Ethiopia
| | - Chala Kenenisa
- Department of Biomedical Sciences, Faculty of Medical Sciences, Institute of Health Sciences, Jimma University, Jimma, Ethiopia
| | - Tesaka Wondimnew
- Department of Biomedical Sciences, Faculty of Medical Sciences, Institute of Health Sciences, Jimma University, Jimma, Ethiopia
| | - Tariku Sime
- Department of Biomedical Sciences, Faculty of Medical Sciences, Institute of Health Sciences, Jimma University, Jimma, Ethiopia
| |
Collapse
|
41
|
Kojima M, Degawa M. Androgen-Dependent Differences in the Amounts of CYP mRNAs in the Pig Kidney. Biol Pharm Bull 2021; 44:1120-1128. [PMID: 34334497 DOI: 10.1248/bpb.b21-00333] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
We previously reported androgen-dependent sex and breed differences in the amounts of mRNAs of CYP isoforms in the pig liver. To clarify whether there are such sex and breed differences in the kidney, we examined the amounts of several CYP mRNAs in the kidney using both sexes of 5-month-old Landrace, Meishan and/or their crossbred F1 (LM and ML) pigs. Significant sex differences in the amounts of several CYP mRNAs were found: male < female for CYP2A19 and CYP3A29; and male > female for CYP4A24/25 in all the breeds. Sex differences in the amount of CYP2B22 mRNA (male < female) and in CYP2C33 and CYP2C49 mRNAs (male > female) were also observed in all the breeds except Landrace pigs. Furthermore, a significant sex difference (male < female) in CYP3A46 mRNA was only found in LM and ML pigs. No significant sex differences were found in either Landrace or Meishan pigs for CYP1A1, CYP1A2 and CYP4B1 mRNAs. The amounts of CYP2C33 and CYP4A24/25 mRNAs in males were higher in Meishan pigs than in Landrace pigs. Additional experiments using pigs treated by castration and/or testosterone propionate indicated that sex and breed differences in the amounts of those CYP mRNAs were, at least in part, dependent on the levels of serum testosterone. Furthermore, the effects of androgen on the amounts of CYP mRNAs in the kidney did not necessarily correlate with those in the liver, suggesting that there is a tissue-selective factor responsible for the androgen-related expression of CYP genes.
Collapse
Affiliation(s)
- Misaki Kojima
- Animal Genome Unit, Institute of Livestock and Grassland Science, National Agriculture and Food Research Organization (NARO)
| | - Masakuni Degawa
- Laboratory of Molecular Toxicology, School of Pharmaceutical Sciences, University of Shizuoka
| |
Collapse
|
42
|
Chen WY, Evangelista EA, Yang J, Kelly EJ, Yeung CK. Kidney Organoid and Microphysiological Kidney Chip Models to Accelerate Drug Development and Reduce Animal Testing. Front Pharmacol 2021; 12:695920. [PMID: 34381363 PMCID: PMC8350564 DOI: 10.3389/fphar.2021.695920] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Accepted: 07/13/2021] [Indexed: 01/17/2023] Open
Abstract
Kidneys are critical for the elimination of many drugs and metabolites via the urine, filtering waste and maintaining proper fluid and electrolyte balance. Emerging technologies incorporating engineered three-dimensional (3D) in vitro cell culture models, such as organoids and microphysiological systems (MPS) culture platforms, have been developed to replicate nephron function, leading to enhanced efficacy, safety, and toxicity evaluation of new drugs and environmental exposures. Organoids are tiny, self-organized three-dimensional tissue cultures derived from stem cells that can include dozens of cell types to replicate the complexity of an organ. In contrast, MPS are highly controlled fluidic culture systems consisting of isolated cell type(s) that can be used to deconvolute mechanism and pathophysiology. Both systems, having their own unique benefits and disadvantages, have exciting applications in the field of kidney disease modeling and therapeutic discovery and toxicology. In this review, we discuss current uses of both hPSC-derived organoids and MPS as pre-clinical models for studying kidney diseases and drug induced nephrotoxicity. Examples such as the use of organoids to model autosomal dominant polycystic kidney disease, and the use of MPS to predict renal clearance and nephrotoxic concentrations of novel drugs are briefly discussed. Taken together, these novel platforms allow investigators to elaborate critical scientific questions. While much work needs to be done, utility of these 3D cell culture technologies has an optimistic outlook and the potential to accelerate drug development while reducing the use of animal testing.
Collapse
Affiliation(s)
- Wei-Yang Chen
- Department of Pharmacy, School of Pharmacy, University of Washington, Seattle, WA, United States
| | - Eric A Evangelista
- Department of Pharmacy, School of Pharmacy, University of Washington, Seattle, WA, United States
| | - Jade Yang
- Department of Pharmaceutics, University of Washington School of Pharmacy, Seattle, WA, United States
| | - Edward J Kelly
- Department of Pharmaceutics, University of Washington School of Pharmacy, Seattle, WA, United States
- Kidney Research Institute, University of Washington School of Medicine, Seattle, WA, United States
| | - Catherine K Yeung
- Department of Pharmacy, School of Pharmacy, University of Washington, Seattle, WA, United States
- Kidney Research Institute, University of Washington School of Medicine, Seattle, WA, United States
| |
Collapse
|
43
|
Bondue T, Arcolino FO, Veys KRP, Adebayo OC, Levtchenko E, van den Heuvel LP, Elmonem MA. Urine-Derived Epithelial Cells as Models for Genetic Kidney Diseases. Cells 2021; 10:cells10061413. [PMID: 34204173 PMCID: PMC8230018 DOI: 10.3390/cells10061413] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 05/28/2021] [Accepted: 06/02/2021] [Indexed: 12/11/2022] Open
Abstract
Epithelial cells exfoliated in human urine can include cells anywhere from the urinary tract and kidneys; however, podocytes and proximal tubular epithelial cells (PTECs) are by far the most relevant cell types for the study of genetic kidney diseases. When maintained in vitro, they have been proven extremely valuable for discovering disease mechanisms and for the development of new therapies. Furthermore, cultured patient cells can individually represent their human sources and their specific variants for personalized medicine studies, which are recently gaining much interest. In this review, we summarize the methodology for establishing human podocyte and PTEC cell lines from urine and highlight their importance as kidney disease cell models. We explore the well-established and recent techniques of cell isolation, quantification, immortalization and characterization, and we describe their current and future applications.
Collapse
Affiliation(s)
- Tjessa Bondue
- Department of Development and Regeneration, KU Leuven, 3000 Leuven, Belgium; (T.B.); (F.O.A.); (K.R.P.V.); (O.C.A.); (E.L.); (L.P.v.d.H.)
| | - Fanny O. Arcolino
- Department of Development and Regeneration, KU Leuven, 3000 Leuven, Belgium; (T.B.); (F.O.A.); (K.R.P.V.); (O.C.A.); (E.L.); (L.P.v.d.H.)
| | - Koenraad R. P. Veys
- Department of Development and Regeneration, KU Leuven, 3000 Leuven, Belgium; (T.B.); (F.O.A.); (K.R.P.V.); (O.C.A.); (E.L.); (L.P.v.d.H.)
- Department of Pediatrics, Division of Pediatric Nephrology, University Hospitals Leuven, 3000 Leuven, Belgium
| | - Oyindamola C. Adebayo
- Department of Development and Regeneration, KU Leuven, 3000 Leuven, Belgium; (T.B.); (F.O.A.); (K.R.P.V.); (O.C.A.); (E.L.); (L.P.v.d.H.)
- Centre for Molecular and Vascular Biology, Department of Cardiovascular Sciences, KU Leuven, 3000 Leuven, Belgium
| | - Elena Levtchenko
- Department of Development and Regeneration, KU Leuven, 3000 Leuven, Belgium; (T.B.); (F.O.A.); (K.R.P.V.); (O.C.A.); (E.L.); (L.P.v.d.H.)
- Department of Pediatrics, Division of Pediatric Nephrology, University Hospitals Leuven, 3000 Leuven, Belgium
| | - Lambertus P. van den Heuvel
- Department of Development and Regeneration, KU Leuven, 3000 Leuven, Belgium; (T.B.); (F.O.A.); (K.R.P.V.); (O.C.A.); (E.L.); (L.P.v.d.H.)
- Department of Pediatric Nephrology, Radboud University Medical Center, 6500 Nijmegen, The Netherlands
| | - Mohamed A. Elmonem
- Department of Clinical and Chemical Pathology, Faculty of Medicine, Cairo University, Cairo 11628, Egypt
- Correspondence:
| |
Collapse
|
44
|
Shokri Z, Seidi F, Karami S, Li C, Saeb MR, Xiao H. Laccase immobilization onto natural polysaccharides for biosensing and biodegradation. Carbohydr Polym 2021; 262:117963. [DOI: 10.1016/j.carbpol.2021.117963] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 02/21/2021] [Accepted: 03/16/2021] [Indexed: 12/20/2022]
|
45
|
Human reconstructed kidney models. In Vitro Cell Dev Biol Anim 2021; 57:133-147. [PMID: 33594607 DOI: 10.1007/s11626-021-00548-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Accepted: 01/12/2021] [Indexed: 02/07/2023]
Abstract
The human kidney, which consists of up to 2 million nephrons, is critical for blood filtration, electrolyte balance, pH regulation, and fluid balance in the body. Animal experiments, particularly mice and rats, combined with advances in genetically modified technology have been the primary mechanism to study kidney injury in recent years. Mouse or rat kidneys, however, differ substantially from human kidneys at the anatomical, histological, and molecular levels. These differences combined with increased regulatory hurdles and shifting attitudes towards animal testing by non-specialists have led scientists to develop new and more relevant models of kidney injury. Although in vitro tissue culture studies are a valuable tool to study kidney injury and have yielded a great deal of insight, they are not a perfect model. Perhaps, the biggest limitation of tissue culture is that it cannot replicate the complex architecture, consisting of multiple cell types, of the kidney, and the interplay between these cells. Recent studies have found that pluripotent stem cells (PSCs), which are capable of differentiation into any cell type, can be used to generate kidney organoids. Organoids recapitulate the multicellular relationships and microenvironments of complex organs like kidney. Kidney organoids have been used to successfully model nephrotoxin-induced tubular and glomerular disease as well as complex diseases such as chronic kidney disease (CKD), which involves multiple cell types. In combination with genetic engineering techniques, such as CRISPR-Cas9, genetic diseases of the kidney can be reproduced in organoids. Thus, organoid models have the potential to predict drug toxicity and enhance drug discovery for human disease more accurately than animal models.
Collapse
|
46
|
Wang H, Liang Y, Yin Y, Zhang J, Su W, White AM, Bin Jiang, Xu J, Zhang Y, Stewart S, Lu X, He X. Carbon nano-onion-mediated dual targeting of P-selectin and P-glycoprotein to overcome cancer drug resistance. Nat Commun 2021; 12:312. [PMID: 33436622 PMCID: PMC7803730 DOI: 10.1038/s41467-020-20588-0] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Accepted: 12/09/2020] [Indexed: 02/06/2023] Open
Abstract
The transmembrane P-glycoprotein (P-gp) pumps that efflux drugs are a major mechanism of cancer drug resistance. They are also important in protecting normal tissue cells from poisonous xenobiotics and endogenous metabolites. Here, we report a fucoidan-decorated silica-carbon nano-onion (FSCNO) hybrid nanoparticle that targets tumor vasculature to specifically release P-gp inhibitor and anticancer drug into tumor cells. The tumor vasculature targeting capability of the nanoparticle is demonstrated using multiple models. Moreover, we reveal the superior light absorption property of nano-onion in the near infrared region (NIR), which enables triggered drug release from the nanoparticle at a low NIR power. The released inhibitor selectively binds to P-gp pumps and disables their function, which improves the bioavailability of anticancer drug inside the cells. Furthermore, free P-gp inhibitor significantly increases the systemic toxicity of a chemotherapy drug, which can be resolved by delivering them with FSCNO nanoparticles in combination with a short low-power NIR laser irradiation.
Collapse
Affiliation(s)
- Hai Wang
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, 20742, USA.
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, 100190, Beijing, China.
- University of Chinese Academy of Sciences, 100049, Beijing, China.
| | - Yutong Liang
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, 20742, USA
| | - Yue Yin
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, 100190, Beijing, China
| | - Jie Zhang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, 100190, Beijing, China
| | - Wen Su
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, 100190, Beijing, China
| | - Alisa M White
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, 20742, USA
| | - Bin Jiang
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, 20742, USA
| | - Jiangsheng Xu
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, 20742, USA
| | - Yuntian Zhang
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, 20742, USA
| | - Samantha Stewart
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, 20742, USA
| | - Xiongbin Lu
- Department of Medical and Molecular Genetics and Melvin and Bren Simon Cancer Center, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Xiaoming He
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, 20742, USA.
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland, Baltimore, MD, 21201, USA.
- Robert E. Fischell Institute for Biomedical Devices, University of Maryland, College Park, MD, 20742, USA.
| |
Collapse
|
47
|
Gao R, Niu X, Zhu L, Qi R, He L. iTRAQ quantitative proteomic analysis differentially expressed proteins and signal pathways in henoch-schönlein purpura nephritis. Am J Transl Res 2020; 12:7908-7922. [PMID: 33437369 PMCID: PMC7791518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Accepted: 11/03/2020] [Indexed: 06/12/2023]
Abstract
Henoch-Schönlein purpura nephritis (HSPN) has been considered as a major cause of chronic renal failure in children and a condition which can worsen clinical outcomes in adults. At present, the molecular mechanisms of HSPN are still unclear. In this study, iTRAQ quantitative proteomic analysis was performed on renal tissues collected from patients with HSPN and compared with those of patients after nephrectomy (controls). A total of 149 differentially expressed proteins (DEPs) were detected, of which, 97 being upregulated and 52 down-regulated. Protein functions and classifications were analyzed using Gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG). In addition, protein domains. expressive hierarchical clustering analysis and protein-protein interaction (PPI) analysis were also conducted for DEPs. The results of bioinformatics analysis indicated that DEPs were enriched in lipid metabolism and the adherens junction pathway. Among these proteins, CDC42 and CTNNB1 were identified as potential candidates involved in the pathogenesis of HSPN. Immunohistochemistry and real-time PCR further demonstrated that CDC42 and CTNNB1 were up-regulated in HSPN patients. These results provide new and important insights into some underlying molecular pathogenesis of HSPN.
Collapse
Affiliation(s)
- Ran Gao
- Department of Hematology, No. 1 Hospital of China Medical UniversityShenyang 110001, Liaoning, China
| | - Xueli Niu
- Department of Dermatology, No. 1 Hospital of China Medical University and Key Laboratory of Immunodermatology, Ministry of Health and Ministry of EducationShenyang 110001, Liaoning, China
| | - Lili Zhu
- Department of Dermatology, The People’s Hospital of China Medical University and The People’s Hospital of Liaoning ProvinceShenyang 110016, Liaoning, China
| | - Ruiqun Qi
- Department of Dermatology, No. 1 Hospital of China Medical University and Key Laboratory of Immunodermatology, Ministry of Health and Ministry of EducationShenyang 110001, Liaoning, China
| | - Liang He
- Department of Thyroid Surgery, No. 1 Hospital of China Medical UniversityShenyang 110001, Liaoning, China
| |
Collapse
|
48
|
Bowman CM, Ma F, Mao J, Chen Y. Examination of Physiologically-Based Pharmacokinetic Models of Rosuvastatin. CPT-PHARMACOMETRICS & SYSTEMS PHARMACOLOGY 2020; 10:5-17. [PMID: 33220025 PMCID: PMC7825190 DOI: 10.1002/psp4.12571] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Accepted: 10/19/2020] [Indexed: 12/14/2022]
Abstract
Physiologically‐based pharmacokinetic (PBPK) modeling is increasingly used to predict drug disposition and drug–drug interactions (DDIs). However, accurately predicting the pharmacokinetics of transporter substrates and transporter‐mediated DDIs (tDDIs) is still challenging. Rosuvastatin is a commonly used substrate probe in DDI risk assessment for new molecular entities (NMEs) that are potential organic anion transporting polypeptide 1B or breast cancer resistance protein transporter inhibitors, and as such, several rosuvastatin PBPK models have been developed to try to predict the clinical DDI and support NME drug labeling. In this review, we examine five representative PBPK rosuvastatin models, discuss common challenges that the models have come across, and note remaining gaps. These shared learnings will help with the continuing efforts of rosuvastatin model validation, provide more information to understand transporter‐mediated drug disposition, and increase confidence in tDDI prediction.
Collapse
Affiliation(s)
- Christine M Bowman
- Department of Drug Metabolism and Pharmacokinetics, Genentech, Inc., South San Francisco, California, USA
| | - Fang Ma
- Department of Drug Metabolism and Pharmacokinetics, Genentech, Inc., South San Francisco, California, USA
| | - Jialin Mao
- Department of Drug Metabolism and Pharmacokinetics, Genentech, Inc., South San Francisco, California, USA
| | - Yuan Chen
- Department of Drug Metabolism and Pharmacokinetics, Genentech, Inc., South San Francisco, California, USA
| |
Collapse
|
49
|
Huynh KM, Wong ACY, Wu B, Horschman M, Zhao H, Brooks JD. Sprr2f protects against renal injury by decreasing the level of reactive oxygen species in female mice. Am J Physiol Renal Physiol 2020; 319:F876-F884. [PMID: 33017192 DOI: 10.1152/ajprenal.00318.2020] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Renal injury leads to chronic kidney disease, with which women are not only more likely to be diagnosed than men but have poorer outcomes as well. We have previously shown that expression of small proline-rich region 2f (Sprr2f), a member of the small proline-rich region (Sprr) gene family, is increased several hundredfold after renal injury using a unilateral ureteral obstruction (UUO) mouse model. To better understand the role of Sprr2f in renal injury, we generated a Sprr2f knockout (Sprr2f-KO) mouse model using CRISPR-Cas9 technology. Sprr2f-KO female mice showed greater renal damage after UUO compared with wild-type (Sprr2f-WT) animals, as evidenced by higher hydroxyproline levels and denser collagen staining, indicating a protective role of Sprr2f during renal injury. Gene expression profiling by RNA sequencing identified 162 genes whose expression levels were significantly different between day 0 and day 5 after UUO in Sprr2f-KO mice. Of the 162 genes, 121 genes were upregulated after UUO and enriched with those involved in oxidation-reduction, a phenomenon not observed in Sprr2f-WT animals, suggesting a protective role of Sprr2f in UUO through defense against oxidative damage. Consistently, bilateral ischemia-reperfusion injury resulted in higher serum blood urea nitrogen levels and higher tissue reactive oxygen species in Sprr2f-KO compared with Sprr2f-WT female mice. Moreover, cultured renal epithelial cells from Sprr2f-KO female mice showed lower viability after oxidative damage induced by menadione compared with Sprr2f-WT cells that could be rescued by supplementation with reduced glutathione, suggesting that Sprr2f induction after renal damage acts as a defense against reactive oxygen species.
Collapse
Affiliation(s)
- Kieu My Huynh
- Department of Urology, School of Medicine, Stanford University, Stanford, California
| | - Anny Chuu-Yun Wong
- Department of Urology, School of Medicine, Stanford University, Stanford, California
| | - Bo Wu
- Department of Urology, School of Medicine, Stanford University, Stanford, California
| | - Marc Horschman
- Department of Urology, School of Medicine, Stanford University, Stanford, California
| | - Hongjuan Zhao
- Department of Urology, School of Medicine, Stanford University, Stanford, California
| | - James D Brooks
- Department of Urology, School of Medicine, Stanford University, Stanford, California
| |
Collapse
|
50
|
Tang H, Abouleila Y, Si L, Ortega-Prieto AM, Mummery CL, Ingber DE, Mashaghi A. Human Organs-on-Chips for Virology. Trends Microbiol 2020; 28:934-946. [PMID: 32674988 PMCID: PMC7357975 DOI: 10.1016/j.tim.2020.06.005] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 06/03/2020] [Accepted: 06/19/2020] [Indexed: 02/03/2023]
Abstract
While conventional in vitro culture systems and animal models have been used to study the pathogenesis of viral infections and to facilitate development of vaccines and therapeutics for viral diseases, models that can accurately recapitulate human responses to infection are still lacking. Human organ-on-a-chip (Organ Chip) microfluidic culture devices that recapitulate tissue–tissue interfaces, fluid flows, mechanical cues, and organ-level physiology have been developed to narrow the gap between in vitro experimental models and human pathophysiology. Here, we describe how recent developments in Organ Chips have enabled re-creation of complex pathophysiological features of human viral infections in vitro. Microfluidic Organ Chip culture devices are emerging alternatives to conventional in vitro and animal models due to their ability to replicate many structural and functional features of human physiology and disease states. Recent innovations demonstrate that Organ Chip technology is a promising strategy for virology studies where there have been successes in reproducing various viral disease phenotypes. Organ Chips have enabled investigation of many aspects of viral infection, including virus–host interactions, viral therapy-resistance evolution, and development of new antiviral therapeutics, as well as underlying pathogenesis. As Organ Chip-based assays provide accessibility to study virus-induced diseases in real time and at high resolution, they can open new avenues to uncover viral pathogenesis in a human-relevant environment and may eventually enable development of novel therapeutics and vaccines.
Collapse
Affiliation(s)
- Huaqi Tang
- Medical Systems Biophysics and Bioengineering, Leiden Academic Centre for Drug Research, Leiden University, Einsteinweg 55, 2333 CC, Leiden, The Netherlands
| | - Yasmine Abouleila
- Medical Systems Biophysics and Bioengineering, Leiden Academic Centre for Drug Research, Leiden University, Einsteinweg 55, 2333 CC, Leiden, The Netherlands
| | - Longlong Si
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA 02115, USA
| | | | - Christine L Mummery
- Department of Anatomy and Embryology, Leiden University Medical Center, Einthovenweg 20, 2333 ZD, Leiden, The Netherlands
| | - Donald E Ingber
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA 02115, USA; Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA; Vascular Biology Program and Department of Surgery, Harvard Medical School and Boston Children's Hospital, Boston, MA 02115, USA
| | - Alireza Mashaghi
- Medical Systems Biophysics and Bioengineering, Leiden Academic Centre for Drug Research, Leiden University, Einsteinweg 55, 2333 CC, Leiden, The Netherlands.
| |
Collapse
|