1
|
Mboni-Johnston IM, Hartmann S, Kroll C, Berndt C, Adjaye J, Schupp N. Impact of nephrotoxins and oxidants on survival and transport function of hiPSC-derived renal proximal tubular cells. Arch Toxicol 2025:10.1007/s00204-025-04015-1. [PMID: 40119912 DOI: 10.1007/s00204-025-04015-1] [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: 12/20/2024] [Accepted: 02/27/2025] [Indexed: 03/25/2025]
Abstract
Due to their role in excretion, renal proximal tubular cells are susceptible to damage by toxic metabolites and xenobiotics. The regenerative capacity of the kidney allows for the replacement of damaged cells, a process involving differentiation programs. However, kidney function tends to decline, suggesting that the replacement cells may not achieve full functionality. To understand possible causes of this decline, we investigated effects of nephrotoxins and oxidants on the differentiation of induced pluripotent stem cells (iPSC) into proximal tubular epithelial-like cells (PTELC). Proliferation, apoptosis, senescence, and expression of oxidative defense genes were analyzed in iPSC, differentiating and differentiated cells treated with cisplatin (CisPt, up to 45 µM), cyclosporin A (CycA, up to 12 µM), and the oxidants menadione (Mena, up to 50 µM) and tert-butylhydroquinone (tBHQ, up to 50 µM). We found that differentiating cells were most sensitive to oxidants and showed increased sensitivity to CisPt, whereas all differentiation stages showed similar sensitivity to CycA. Both oxidative stress and CisPt triggered apoptosis in all differentiation stages, whereas CycA mainly induced senescence. Treatment during differentiation resulted in long-term effects on gene expression in differentiated cells. While oxidants had no effect on transport function of differentiated cells, CisPt and CycA impaired albumin uptake. Our data suggest a substantial sensitivity of differentiating cells to nephrotoxins and oxidants, an aspect that could potentially interfere with regenerative processes.
Collapse
Affiliation(s)
- Isaac Musong Mboni-Johnston
- Institute of Toxicology, Medical Faculty, University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Moorenstraße 5, 40225, Düsseldorf, Germany
| | - Sören Hartmann
- Institute of Toxicology, Medical Faculty, University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Moorenstraße 5, 40225, Düsseldorf, Germany
| | - Christian Kroll
- Department of Neurology, Medical Faculty, University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, 40225, Düsseldorf, Germany
| | - Carsten Berndt
- Department of Neurology, Medical Faculty, University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, 40225, Düsseldorf, Germany
| | - James Adjaye
- Institute for Stem Cell Research and Regenerative Medicine, Medical Faculty, University Hospital Düsseldorf, University of Düsseldorf, 40225, Düsseldorf, Germany
- Zayed Centre for Research into Rare Diseases in Children (ZCR), EGA Institute for Women'S Health, University College London (UCL), 20 Guilford Street, London, WC1N 1DZ, UK
| | - Nicole Schupp
- Institute of Toxicology, Medical Faculty, University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Moorenstraße 5, 40225, Düsseldorf, Germany.
| |
Collapse
|
2
|
Carty JS, Bessho R, Zuchowski Y, Trapani JB, Davidoff O, Kobayashi H, Roland JT, Watts JA, Terker AS, Bock F, Arroyo JP, Haase VH. Disruption of mitochondrial electron transport impairs urinary concentration via AMPK-dependent suppression of aquaporin 2. JCI Insight 2024; 9:e182087. [PMID: 39361429 PMCID: PMC11601893 DOI: 10.1172/jci.insight.182087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Accepted: 10/02/2024] [Indexed: 10/05/2024] Open
Abstract
Urinary concentration is an energy-dependent process that minimizes body water loss by increasing aquaporin 2 (AQP2) expression in collecting duct (CD) principal cells. To investigate the role of mitochondrial (mt) ATP production in renal water clearance, we disrupted mt electron transport in CD cells by targeting ubiquinone (Q) binding protein QPC (UQCRQ), a subunit of mt complex III essential for oxidative phosphorylation. QPC-deficient mice produced less concentrated urine than controls, both at baseline and after type 2 vasopressin receptor stimulation with desmopressin. Impaired urinary concentration in QPC-deficient mice was associated with reduced total AQP2 protein levels in CD tubules, while AQP2 phosphorylation and membrane trafficking remained unaffected. In cultured inner medullary CD cells treated with mt complex III inhibitor antimycin A, the reduction in AQP2 abundance was associated with activation of 5' adenosine monophosphate-activated protein kinase (AMPK) and was reversed by treatment with AMPK inhibitor SBI-0206965. In summary, our studies demonstrated that the physiological regulation of AQP2 abundance in principal CD cells was dependent on mt electron transport. Furthermore, our data suggested that oxidative phosphorylation in CD cells was dispensable for maintaining water homeostasis under baseline conditions, but necessary for maximal stimulation of AQP2 expression and urinary concentration.
Collapse
Affiliation(s)
- Joshua S. Carty
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center and Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Ryoichi Bessho
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center and Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Yvonne Zuchowski
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center and Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Jonathan B. Trapani
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center and Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Olena Davidoff
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center and Vanderbilt University School of Medicine, Nashville, Tennessee, USA
- Research and Medical Services, Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville, Tennessee, USA
| | - Hanako Kobayashi
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center and Vanderbilt University School of Medicine, Nashville, Tennessee, USA
- Research and Medical Services, Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville, Tennessee, USA
| | - Joseph T. Roland
- Section of Surgical Sciences, Epithelial Biology Center, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Jason A. Watts
- Epigenetics and Stem Cell Laboratory, National Institute of Environmental Health Sciences, NIH, Research Triangle Park, North Carolina, USA
| | - Andrew S. Terker
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center and Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Fabian Bock
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center and Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Juan Pablo Arroyo
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center and Vanderbilt University School of Medicine, Nashville, Tennessee, USA
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Volker H. Haase
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center and Vanderbilt University School of Medicine, Nashville, Tennessee, USA
- Research and Medical Services, Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville, Tennessee, USA
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| |
Collapse
|
3
|
Nørregaard R, Mutsaers HAM, Frøkiær J, Kwon TH. Obstructive nephropathy and molecular pathophysiology of renal interstitial fibrosis. Physiol Rev 2023; 103:2827-2872. [PMID: 37440209 PMCID: PMC10642920 DOI: 10.1152/physrev.00027.2022] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 07/05/2023] [Accepted: 07/09/2023] [Indexed: 07/14/2023] Open
Abstract
The kidneys play a key role in maintaining total body homeostasis. The complexity of this task is reflected in the unique architecture of the organ. Ureteral obstruction greatly affects renal physiology by altering hemodynamics, changing glomerular filtration and renal metabolism, and inducing architectural malformations of the kidney parenchyma, most importantly renal fibrosis. Persisting pathological changes lead to chronic kidney disease, which currently affects ∼10% of the global population and is one of the major causes of death worldwide. Studies on the consequences of ureteral obstruction date back to the 1800s. Even today, experimental unilateral ureteral obstruction (UUO) remains the standard model for tubulointerstitial fibrosis. However, the model has certain limitations when it comes to studying tubular injury and repair, as well as a limited potential for human translation. Nevertheless, ureteral obstruction has provided the scientific community with a wealth of knowledge on renal (patho)physiology. With the introduction of advanced omics techniques, the classical UUO model has remained relevant to this day and has been instrumental in understanding renal fibrosis at the molecular, genomic, and cellular levels. This review details key concepts and recent advances in the understanding of obstructive nephropathy, highlighting the pathophysiological hallmarks responsible for the functional and architectural changes induced by ureteral obstruction, with a special emphasis on renal fibrosis.
Collapse
Affiliation(s)
- Rikke Nørregaard
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Department of Renal Medicine, Aarhus University Hospital, Aarhus, Denmark
| | | | - Jørgen Frøkiær
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Tae-Hwan Kwon
- Department of Biochemistry and Cell Biology, School of Medicine, Kyungpook National University, Taegu, Korea
| |
Collapse
|
4
|
Lv W, Liang L, Liu D, Li C, Jia L, Jin Y. Aquaporin 1 Facilitates Ferroptosis, M1 Polarization, Mitochondrial Dysfunction, and Autophagy Damage on Lipopolysaccharide-Induced Macrophage Through Down-Regulation of P53 Signaling Pathway. DNA Cell Biol 2023; 42:456-480. [PMID: 37379471 PMCID: PMC10457627 DOI: 10.1089/dna.2023.0016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 04/10/2023] [Accepted: 04/17/2023] [Indexed: 06/30/2023] Open
Abstract
This study was designed to investigate the role of aquaporin 1 (AQP1) in ferroptosis, macrophage polarization, mitochondrial dysfunction, and impaired autophagy of lipopolysaccharide (LPS)-stimulated RAW264.7 cells and explored the underlying mechanisms. Si-AQP1-mediated AQP1 silencing RAW264.7 cells was constructed. Si-P53-mediated P53 silencing or pcDNA-P53 overexpression RAW264.7 cells was constructed. Assays of ATP, reverse transcription-quantitative polymerase chain reaction (RT-qPCR), and Mitochondrial membrane potential (JC-1) staining were performed to evaluate mitochondrial biological function. Assays of flow cytometry, reactive oxygen species (ROS) staining, western blot (WB), RT-qPCR, malondialdehyde (MDA), glutathione (GSH), and total superoxide dismutase (SOD) were performed to detect cell ferroptosis, macrophage polarization, and impaired autophagy. The involvement of the P53 pathway was revealed by WB. The results showed that LPS (30 μg/mL) could induce ferroptosis, M1 polarization, mitochondrial dysfunction, and autophagy damage in RAW264.7 cells. Meanwhile, the expression of AQP1 was increased and the expression of P53 was decreased. In addition, Pifithrin-α (PIF; 15 μM), a P53 inhibitor, significantly aggravated ferroptosis, M1 polarization, mitochondrial dysfunction, and autophagy damage as well as up-regulation of AQP1 protein expression in LPS-induced RAW264.7 cells. Interestingly, this phenomenon was markedly alleviated by Kevetrin hydrochloride (70 μM), a P53 agonist. Mechanistically, silencing AQP1 significantly alleviated ferroptosis, M1 polarization, mitochondrial dysfunction, and autophagy damage by up-regulating the expression of P53 in LPS-stimulated RAW264.7 cells. Indeed, inhibition of P53 expression by PIF treatment dramatically reversed this effect on the basis of LPS+si-AQP1. Therefore, we concluded for the first time that AQP1 can promote ferroptosis, M1 polarization, mitochondrial dysfunction, and autophagy impairment by inhibiting the expression of P53 in LPS-stimulated RAW264.7 cells, and AQP1 or P53 may be considered as a crucial determiner that can regulate the biological behavior of RAW264.7 cells stimulated by LPS.
Collapse
Affiliation(s)
- Wuyang Lv
- Department of Clinical Laboratory, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Lei Liang
- Department of Clinical Laboratory, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Dongyang Liu
- Department of Clinical Laboratory, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Cuicui Li
- Department of Clinical Laboratory, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Liao Jia
- Department of Clinical Laboratory, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yingyu Jin
- Department of Clinical Laboratory, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
5
|
Liu M, Sun X, Chen B, Dai R, Xi Z, Xu H. Insights into Manganese Superoxide Dismutase and Human Diseases. Int J Mol Sci 2022; 23:ijms232415893. [PMID: 36555531 PMCID: PMC9786916 DOI: 10.3390/ijms232415893] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 12/09/2022] [Accepted: 12/12/2022] [Indexed: 12/15/2022] Open
Abstract
Redox equilibria and the modulation of redox signalling play crucial roles in physiological processes. Overproduction of reactive oxygen species (ROS) disrupts the body's antioxidant defence, compromising redox homeostasis and increasing oxidative stress, leading to the development of several diseases. Manganese superoxide dismutase (MnSOD) is a principal antioxidant enzyme that protects cells from oxidative damage by converting superoxide anion radicals to hydrogen peroxide and oxygen in mitochondria. Systematic studies have demonstrated that MnSOD plays an indispensable role in multiple diseases. This review focuses on preclinical evidence that describes the mechanisms of MnSOD in diseases accompanied with an imbalanced redox status, including fibrotic diseases, inflammation, diabetes, vascular diseases, neurodegenerative diseases, and cancer. The potential therapeutic effects of MnSOD activators and MnSOD mimetics are also discussed. Targeting this specific superoxide anion radical scavenger may be a clinically beneficial strategy, and understanding the therapeutic role of MnSOD may provide a positive insight into preventing and treating related diseases.
Collapse
Affiliation(s)
- Mengfan Liu
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
- Engineering Research Center, Shanghai Colleges for TCM New Drug Discovery, Shanghai 201203, China
| | - Xueyang Sun
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
- Engineering Research Center, Shanghai Colleges for TCM New Drug Discovery, Shanghai 201203, China
| | - Boya Chen
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Rongchen Dai
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
- Engineering Research Center, Shanghai Colleges for TCM New Drug Discovery, Shanghai 201203, China
| | - Zhichao Xi
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
- Engineering Research Center, Shanghai Colleges for TCM New Drug Discovery, Shanghai 201203, China
- Correspondence: (Z.X.); (H.X.)
| | - Hongxi Xu
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
- Engineering Research Center, Shanghai Colleges for TCM New Drug Discovery, Shanghai 201203, China
- Correspondence: (Z.X.); (H.X.)
| |
Collapse
|
6
|
Topiroxostat versus allopurinol in patients with chronic heart failure complicated by hyperuricemia: A prospective, randomized, open-label, blinded-end-point clinical trial. PLoS One 2022; 17:e0261445. [PMID: 35077456 PMCID: PMC8789120 DOI: 10.1371/journal.pone.0261445] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Accepted: 11/08/2021] [Indexed: 11/23/2022] Open
Abstract
Background The benefits of xanthine oxidase inhibitors to chronic heart failure (CHF) patients is controversial. We investigated the beneficial effects of a novel xanthine oxidoreductase inhibitor, topiroxostat, in patients with CHF and hyperuricemia (HU), in comparison to allopurinol. Methods and results The prospective, randomized open-label, blinded-end-point study was performed in 141 patients with CHF and HU at 4 centers. Patients were randomly assigned to either topiroxostat or allopurinol group to achieve target uric acid level ≤6.0 mg/dL. According to the protocol, 140 patients were followed up for 24 weeks. Percent change in ln (N-terminal-proB-type natriuretic peptide) at week 24 (primary endpoint) was comparable between topiroxostat and allopurinol groups (1.6±8.2 versus -0.4±8.0%; P = 0.17). In the limited number of patients with heart failure with reduced ejection fraction (HFrEF) (left ventricle ejection fraction <45%), ratio of peak early diastolic flow velocity at mitral valve leaflet to early diastolic mitral annular motion velocity (E/e’) decreased in topiroxostat group, but not in allopurinol group. Urinary 8-hydroxy-2’-deoxyguanosine and L-type fatty acid-binding protein levels increased and osmolality decreased significantly in allopurinol group, while these changes were less or absent in topiroxostat group. In allopurinol group HFrEF patients, additional to the increases in these urinary marker levels, urinary creatinine levels decreased, with no change in clearance, but not in topiroxostat group. Conclusions Compared with allopurinol, topiroxostat did not show great benefits in patients with CHF and HU. However, topiroxostat might have potential advantages of reducing left ventricular end-diastolic pressure, not worsening oxidative stress in proximal renal tubule, and renoprotection over allopurinol in HFrEF patients.
Collapse
|
7
|
Matsumoto AK, Maes M, Michelin AP, Soares AE, Semeão LDO, Godeny P, Venturini D, Barbosa DS, Delfino VDA. Vitamin D deficiency is not associated with increased oxidative stress in chronic kidney disease pre-dialysis patients. J Bras Nefrol 2020; 42:420-428. [PMID: 32406474 PMCID: PMC7860644 DOI: 10.1590/2175-8239-jbn-2019-0156] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Accepted: 03/04/2020] [Indexed: 12/02/2022] Open
Abstract
INTRODUCTION The progressive decline in 25-hydroxyvitamin D [25(OH)D] in chronic kidney disease (CKD) limits the kidney ability of synthesizing the vitamin. Vitamin D deficiency as defined by KDIGO (25(OH)D <20 ng/mL) is prevalent in CKD patients and associated to oxidative stress (OS). We studied a possible association between vitamin D deficiency and OS in pre-dialysis patients. METHODS A cross-sectional study with 206 CKD patients was carried out. Laboratory tests for 25(OH)D, 1,25(OH)2D, inflammatory markers, and OS were added to routine tests including creatinine, albumin, calcium, phosphorus, alkaline phosphatase, iPTH, glucose, hemoglobin, uric acid, total cholesterol, LDL, HDL, and triglycerides. RESULTS Vitamin D deficiency was present in 55 CKD patients and normal vitamin D levels were seen in 149 patients. There was a significant association between vitamin D and estimated glomerular filtration rate (eGRF). Homocysteine levels were best predicted by eGRF, sex, and age; high sensitivity C-reactive protein (hsCRP) by staging and BMI; nitric oxide metabolites (NOx) were increased in late disease; leptin was influenced by BMI and higher in women than man; and adiponectin levels were higher in women. CONCLUSIONS OS biomarkers were not correlated with vitamin D deficiency but increased NOx were seen in stages 4-5 CKD patients. Even though a relatively large number of CKD patients was included and a broad number of OS and inflammatory biomarkers were used in this studied we failed to find an association between vitamin D levels and eGRF. More studies are needed to evaluate the influence of vitamin D status in OS in pre-dialysis CKD patients.
Collapse
Affiliation(s)
- Andressa Keiko Matsumoto
- Universidade Estadual de Londrina, Departamento de Patologia, Análises Clínicas e Toxicológicas, Londrina, PR, Brasil
| | - Michael Maes
- Deakin University, IMPACT Research Center, Geelong, Australia
- Chulalongkorn University, Faculty of Medicine, Department of Psychiatry, Bangkok, Thailand
- Universidade Estadual de Londrina, Centro de Ciências da Saúde, Programa de Pós-Graduação em Ciências da Saúde, Londrina, PR, Brasil
| | - Ana Paula Michelin
- Universidade Estadual de Londrina, Departamento de Patologia, Análises Clínicas e Toxicológicas, Londrina, PR, Brasil
| | - Abel Esteves Soares
- Universidade Estadual de Londrina, Departamento de Medicina Interna, Seção de Nefrologia, Londrina, PR, Brasil
| | - Laura de Oliveira Semeão
- Universidade Estadual de Londrina, Departamento de Patologia, Análises Clínicas e Toxicológicas, Londrina, PR, Brasil
| | - Paula Godeny
- Universidade Estadual de Londrina, Departamento de Patologia, Análises Clínicas e Toxicológicas, Londrina, PR, Brasil
| | - Danielle Venturini
- Universidade Estadual de Londrina, Departamento de Patologia, Análises Clínicas e Toxicológicas, Londrina, PR, Brasil
| | - Décio Sabbatini Barbosa
- Universidade Estadual de Londrina, Departamento de Patologia, Análises Clínicas e Toxicológicas, Londrina, PR, Brasil
| | | |
Collapse
|
8
|
Uribe P, Cárcamo C, Navarro E, Sepúlveda J, Zambrano F, Schulz M, Sánchez R. Protective effect of the superoxide dismutase mimetic MnTBAP during sperm vitrification process. Andrologia 2020; 52:e13665. [PMID: 32539179 DOI: 10.1111/and.13665] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Revised: 04/21/2020] [Accepted: 05/07/2020] [Indexed: 12/31/2022] Open
Abstract
Sperm cryopreservation is widely used in assisted reproduction and male infertility therapy; however, it induces oxidative stress affecting sperm quality. This work evaluated the effect of the antioxidant MnTBAP during vitrification steps in human spermatozoa. First, the effect of MnTBAP on viability and ROS production was evaluated. Then, the spermatozoa were vitrified in straws with the vitrification, warming and post-warming incubation media separately supplemented with MnTBAP. An untreated control was included. The sperm viability, ROS production, total and progressive motility were evaluated. The results showed that the direct exposure of spermatozoa to MnTBAP significantly decreases the ROS levels in comparison with the untreated control without affecting the viability. The supplementation of the vitrification medium with MnTBAP did not affect the parameters analysed. However, the supplementation of the warming and incubation post-warming media resulted in a decrease in ROS production and maintained viability and motility for 4 hr after warming with concentrations up to 100 μM of MnTBAP. Higher concentrations of MnTBAP caused a decrease in total motility. In conclusion, the use of MnTBAP during the warming or post-warming incubation media has beneficial effect decreasing ROS levels and maintaining the viability and motility during the vitrification procedure.
Collapse
Affiliation(s)
- Pamela Uribe
- Center of Excellence in Translational Medicine - Scientific and Technological Bioresource Nucleus (CEMT - BIOREN), Universidad de La Frontera, Temuco, Chile.,Department of Internal Medicine, Faculty of Medicine, Universidad de La Frontera, Temuco, Chile
| | - Constanza Cárcamo
- Center of Excellence in Translational Medicine - Scientific and Technological Bioresource Nucleus (CEMT - BIOREN), Universidad de La Frontera, Temuco, Chile
| | - Eliana Navarro
- Center of Excellence in Translational Medicine - Scientific and Technological Bioresource Nucleus (CEMT - BIOREN), Universidad de La Frontera, Temuco, Chile
| | - Josefa Sepúlveda
- Center of Excellence in Translational Medicine - Scientific and Technological Bioresource Nucleus (CEMT - BIOREN), Universidad de La Frontera, Temuco, Chile
| | - Fabiola Zambrano
- Center of Excellence in Translational Medicine - Scientific and Technological Bioresource Nucleus (CEMT - BIOREN), Universidad de La Frontera, Temuco, Chile.,Department of Preclinical Sciences, Faculty of Medicine, Universidad de La Frontera, Temuco, Chile
| | - Mabel Schulz
- Center of Excellence in Translational Medicine - Scientific and Technological Bioresource Nucleus (CEMT - BIOREN), Universidad de La Frontera, Temuco, Chile.,Department of Preclinical Sciences, Faculty of Medicine, Universidad de La Frontera, Temuco, Chile
| | - Raúl Sánchez
- Center of Excellence in Translational Medicine - Scientific and Technological Bioresource Nucleus (CEMT - BIOREN), Universidad de La Frontera, Temuco, Chile.,Department of Preclinical Sciences, Faculty of Medicine, Universidad de La Frontera, Temuco, Chile
| |
Collapse
|
9
|
Bayat E, Rahpeima Z, Dastghaib S, Gholizadeh F, Erfani M, Asadikaram G, Mokarram P. Stevia rebaudiana extract attenuate metabolic disorders in diabetic rats via modulation of glucose transport and antioxidant signaling pathways and aquaporin-2 expression in two extrahepatic tissues. J Food Biochem 2020; 44:e13252. [PMID: 32515037 DOI: 10.1111/jfbc.13252] [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] [Received: 07/02/2019] [Revised: 03/27/2020] [Accepted: 03/31/2020] [Indexed: 12/11/2022]
Abstract
Today, plant-based therapies have been attracted attention to overcome diabetes complications. This study was an attempt to evaluate whether antidiabetic and nephroprotective effects of Stevia Rebaudiana Bertoni (SRB) can be exerted via upregulation of GLUT-4, SNAP23, and Stx4 in skeletal muscles or modulation of AQP2 mRNA expression and antioxidant signaling pathway activity (Nrf2/Keap1) in kidneys. To achieve this aim, diabetes was induced via STZ-nicotinamide (STZ-NA). Diabetes increased the level of Blood Urea Nitrogen (BUN), serum creatinine, Fasting Blood Sugar (FBS), and Keap1 mRNA expression, which was coincide with reduction in mRNA levels of Nrf2, GLUT4, SNAP23, and Stx4. SRB and metformin compensate mentioned variables. However, SRB extract was more effective than metformin to increase the levels of GLUT4 and Nrf2 mRNA. It seems that SRB might attenuate the diabetic complications via manipulating the glucose uptake components in peripheral tissues and might exert the nephroprotective effects by modulation of AQP2, and Nrf2/Keap1 mRNA expression. PRACTICAL APPLICATIONS: Synthetic antidiabetic drugs have been only partially successful in controlling the diabetic complications. Moreover, use of these drugs is associated with a number of adverse effects. Over the past few years, a renewed attention has been paid to the prevention and treatment of diabetes using medicinal plants and functional foods. SRB that have been known as natural sweetener for centuries, is a such natural agent that has high source of various phytochemicals with antidiabetic, renal protective, antitumor, and antioxidant properties. In the current study, possible molecular mechanisms of insulin-mimetic and nephroprotective effects of SRB extract was evaluated in diabetic rats. Due to powerful antihyperglycemic and nephroprotective effects of SRB extract that were showed in this study and previous studies, hence the fact that SRB is to be highlighted for future research as a new therapeutic agent for diabetes.
Collapse
Affiliation(s)
- Elahe Bayat
- Department of Biochemistry, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Zahra Rahpeima
- Department of Biochemistry, School of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Sanaz Dastghaib
- Department of Biochemistry, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran.,Endocrinology and Metabolism Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Fatemeh Gholizadeh
- Department of Biochemistry, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mehran Erfani
- Department of Biochemistry, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Gholamreza Asadikaram
- Department of Biochemistry, School of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Pooneh Mokarram
- Department of Biochemistry, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran.,Autophagy Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
10
|
He Y, Bao YT, Chen HS, Chen YT, Zhou XJ, Yang YX, Li CY. The Effect of Shen Qi Wan Medicated Serum on NRK-52E Cells Proliferation and Migration by Targeting Aquaporin 1 (AQP1). Med Sci Monit 2020; 26:e922943. [PMID: 32491998 PMCID: PMC7293146 DOI: 10.12659/msm.922943] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2020] [Accepted: 03/17/2020] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Shen Qi Wan (SQW) as a well-known formula for the amelioration of kidney yang deficiency syndrome (KYDS), and it has been widely employed in traditional Chinese medicine (TCM). This study aimed to investigate the effect and underlying mechanism of SQW medicated serum on proliferation and migration in NRK-52E cells. MATERIAL AND METHODS We employed the real-time cell analysis (RTCA) system to investigate the effect of SQW medicated serum on proliferation and migration in NRK-52E cells. In addition, the migration was further investigated by using a wound-healing assay. The mRNA and protein expression level of aquaporin 1 (AQP1) of NRK-52E cells with SQW medicated serum-treated were quantified by real-time quantitative polymerase chain reaction (q-PCR) and western blot assay, respectively. Furthermore, NRK-52E cells were transfected with lentivirus AQP1-RNAi to assess migratory cell abilities in vitro. RESULTS The migratory abilities of NRK-52E cells were significantly increased after SQW medicated serum treatment (P<0.05), and no significant difference in cell proliferation. In addition, SQW medicated serum was significantly upregulated the mRNA and protein expression level of AQP1 in NRK-52E cells (P<0.05). Additionally, the in vitro metastasis test proved that knockdown of AQP1 suppressed migratory abilities according to RTCA and wound healing test while was reversed by SQW medicated serum (P<0.05). CONCLUSIONS Our study demonstrates that SQW medicated serum effectively promotes the migration of NRK-52E cells by increasing AQP1 expression, and AQP1 may be as a therapeutic target of SQW for renal injury treatment under KYDS.
Collapse
Affiliation(s)
- Ying He
- College of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, P.R. China
- Department of Traditional Chinese Medicine (TCM) Pharmacy, Zhejiang Integrated Traditional and Western Medicine Hospital, Hangzhou, Zhejiang, P.R. China
| | - Yu Ting Bao
- College of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, P.R. China
| | - Hong Shu Chen
- Department of Traditional Chinese Medicine (TCM) Pharmacy, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, P.R. China
| | - Yi Tao Chen
- College of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, P.R. China
| | - Xiao Jie Zhou
- College of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, P.R. China
| | - Yuan Xiao Yang
- School of Basic Medical Sciences and Forensic Medicine, Hangzhou Medical College, Hangzhou, Zhejiang, P.R. China
| | - Chang Yu Li
- College of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, P.R. China
| |
Collapse
|
11
|
Cell organelles as targets of mammalian cadmium toxicity. Arch Toxicol 2020; 94:1017-1049. [PMID: 32206829 DOI: 10.1007/s00204-020-02692-8] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Accepted: 02/25/2020] [Indexed: 02/07/2023]
Abstract
Ever increasing environmental presence of cadmium as a consequence of industrial activities is considered a health hazard and is closely linked to deteriorating global health status. General animal and human cadmium exposure ranges from ingestion of foodstuffs sourced from heavily polluted hotspots and cigarette smoke to widespread contamination of air and water, including cadmium-containing microplastics found in household water. Cadmium is promiscuous in its effects and exerts numerous cellular perturbations based on direct interactions with macromolecules and its capacity to mimic or displace essential physiological ions, such as iron and zinc. Cell organelles use lipid membranes to form complex tightly-regulated, compartmentalized networks with specialized functions, which are fundamental to life. Interorganellar communication is crucial for orchestrating correct cell behavior, such as adaptive stress responses, and can be mediated by the release of signaling molecules, exchange of organelle contents, mechanical force generated through organelle shape changes or direct membrane contact sites. In this review, cadmium effects on organellar structure and function will be critically discussed with particular consideration to disruption of organelle physiology in vertebrates.
Collapse
|
12
|
Somparn P, Boonkrai C, Charngkaew K, Chomanee N, Hodge KG, Fenton RA, Pisitkun T, Khositseth S. Bilateral ureteral obstruction is rapidly accompanied by ER stress and activation of autophagic degradation of IMCD proteins, including AQP2. Am J Physiol Renal Physiol 2020; 318:F135-F147. [PMID: 31736351 PMCID: PMC7054639 DOI: 10.1152/ajprenal.00113.2019] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
After the release of bilateral ureteral obstruction (BUO), postobstructive diuresis from an impaired urine concentration mechanism is associated with reduced aquaporin 2 (AQP2) abundance in the inner medullary collecting duct (IMCD). However, the underlying molecular mechanism of this AQP2 reduction is incompletely understood. To elucidate the mechanisms responsible for this phenomenon, we studied molecular changes in IMCDs isolated from rats with 4-h BUO or sham operation at the early onset of AQP2 downregulation using mass spectrometry-based proteomic analysis. Two-hundred fifteen proteins had significant changes in abundances, with 65% of them downregulated in the IMCD of 4-h BUO rats compared with sham rats. Bioinformatic analysis revealed that significantly changed proteins were associated with functional Gene Ontology terms, including “cell-cell adhesion,” “cell-cell adherens junction,” “mitochondrial inner membrane,” “endoplasmic reticulum chaperone complex,” and the KEGG pathway of glycolysis/gluconeogenesis. Targeted liquid chromatography-tandem mass spectrometry or immunoblot analysis confirmed the changes in 19 proteins representative of each predominant cluster, including AQP2. Electron microscopy demonstrated disrupted tight junctions, disorganized adherens junctions, swollen mitochondria, enlargement of the endoplasmic reticulum lumen, and numerous autophagosomes/lysosomes in the IMCD of rats with 4-h BUO. AQP2 and seven proteins chosen as representative of the significantly altered clusters had a significant increase in immunofluorescence-based colocalization with autophagosomes/lysosomes. Immunogold electron microscopy confirmed colocalization of AQP2 with the autophagosome marker microtubule-associated protein 1A/1B-light chain 3 and the lysosomal marker cathepsin D in IMCD cells of rats with 4-h BUO. We conclude that enhanced autophagic degradation of AQP2 and other critical proteins, as well as endoplasmic reticulum stress in the IMCD, are initiated shortly after BUO.
Collapse
Affiliation(s)
- Poorichaya Somparn
- Center of Excellent in Systems Biology, Research Affairs, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand.,Translational Research in Inflammation and Immunology Research Unit, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Chatikorn Boonkrai
- Center of Excellent in Systems Biology, Research Affairs, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Komgrid Charngkaew
- Department of Pathology, Faculty of Medicine, Siriraj Hospital, Mahidol University, Thailand
| | - Nusara Chomanee
- Department of Pathology, Faculty of Medicine, Siriraj Hospital, Mahidol University, Thailand
| | - Kenneth G Hodge
- Center of Excellent in Systems Biology, Research Affairs, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Robert A Fenton
- Department of Biomedicine and Center for Interactions of Proteins in Epithelial Transport, Aarhus University, Aarhus, Denmark
| | - Trairak Pisitkun
- Center of Excellent in Systems Biology, Research Affairs, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand.,Epithelial Systems Biology Laboratory, National Heart, Lung, and Blood Institute, Bethesda, Maryland
| | - Sookkasem Khositseth
- Department of Pediatrics, Faculty of Medicine, Thammasat University, Pathumthani, Thailand
| |
Collapse
|
13
|
Zhou B, Gao M, Feng X, Huang L, Huang Q, Kootala S, Larsson TE, Zheng L, Bowden T. Carbazate modified dextrans as scavengers for carbonylated proteins. Carbohydr Polym 2019; 232:115802. [PMID: 31952601 DOI: 10.1016/j.carbpol.2019.115802] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Revised: 12/23/2019] [Accepted: 12/27/2019] [Indexed: 12/15/2022]
Abstract
A series of biocompatible and non- toxic polysaccharide molecules have been successfully fabricated and explored their potential application for scavenging the carbonyl species in vitro. These macromolecules were dextrans with different hydrazide substitution ratios determined by TNBS assay, NMR and FTIR characterization. The colorimetric assay had demonstrated that these macromolecules could effectively scavenge acrolein, oxidized bovine serum albumin (BSA) in buffer solutions as well as carbonyl proteins from serum. The scavengers could achieve twice more scavenging effects for modified dextrans with high molecular weight (Mw = 100,000) than those of low ones (Mw = 40,000) with the same substitution ratio. Protein gel electrophoresis confirmed that the formation of the complex between carbonyls and modified dextrans resulted in appearance of slower bands. It also revealed that such macromolecules could protect cultured cells against the toxicity of acrolein or its derivatives. The proposed macromolecules indicated a very promising capability as scavengers for oxidative stress plus its derivatives without side effects.
Collapse
Affiliation(s)
- Bo Zhou
- Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China; Guangxi Collaborative Innovation Center for Biomedicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China
| | - Ming Gao
- Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China; Guangxi Collaborative Innovation Center for Biomedicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China.
| | - Xianjing Feng
- Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China; Guangxi Collaborative Innovation Center for Biomedicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China; Pharmaceutical College, Guangxi Medical University, Nanning, 530021, China
| | - Lanli Huang
- Guangxi Collaborative Innovation Center for Biomedicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China; Pharmaceutical College, Guangxi Medical University, Nanning, 530021, China
| | - Quanxin Huang
- Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China; Guangxi Collaborative Innovation Center for Biomedicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China
| | - Sujit Kootala
- Department of Chemistry - Ångström Laboratory, Uppsala University, Box 538, SE-751 21, Uppsala, Sweden
| | - Tobias E Larsson
- Department of Clinical Science, Intervention and Technology, Karolinska Institutet, SE-141 86, Stockholm, Sweden; Department of Nephrology, Karolinska University Hospital, Stockholm, Sweden
| | - Li Zheng
- Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China; Guangxi Collaborative Innovation Center for Biomedicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China.
| | - Tim Bowden
- Department of Chemistry - Ångström Laboratory, Uppsala University, Box 538, SE-751 21, Uppsala, Sweden.
| |
Collapse
|
14
|
Gonzalez-Vicente A, Hong N, Garvin JL. Effects of reactive oxygen species on renal tubular transport. Am J Physiol Renal Physiol 2019; 317:F444-F455. [PMID: 31215804 DOI: 10.1152/ajprenal.00604.2018] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Reactive oxygen species (ROS) play a critical role in regulating nephron transport both via transcellular and paracellular pathways under physiological and pathological circumstances. Here, we review the progress made in the past ~10 yr in understanding how ROS regulate solute and water transport in individual nephron segments. Our knowledge in this field is still rudimentary, with basic information lacking. This is most obvious when looking at the reported disparate effects of superoxide ([Formula: see text]) and H2O2 on proximal nephron transport, where there are no easy explanations as to how to reconcile the data. Similarly, we know almost nothing about the regulation of transport in thin descending and ascending limbs, information that is likely critical to understanding the urine concentrating mechanism. In the thick ascending limb, there is general agreement that ROS enhance transcellular reabsorption of NaCl, but we know very little about their effects on the paracellular pathway and therefore Ca2+ and Mg2+ transport. In the distal convoluted tubule, precious little is known. In the collecting duct, there is general agreement that ROS stimulate the epithelial Na+ channel.
Collapse
Affiliation(s)
- Agustin Gonzalez-Vicente
- Department of Inflammation and Immunity, Cleveland Clinic, Lerner Research Institute, Cleveland, Ohio
| | - Nancy Hong
- Department of Physiology and Biophysics, School of Medicine, Case Western Reserve University Cleveland, Ohio
| | - Jeffrey L Garvin
- Department of Physiology and Biophysics, School of Medicine, Case Western Reserve University Cleveland, Ohio
| |
Collapse
|
15
|
Li W, Cao Z, Yu W, Yao X, Ting R, Cheng F. Acute Kidney Injury Induced by Pneumoperitoneum Pressure Via a Mitochondrial Injury-dependent Mechanism in a Rabbit Model of Different Degrees of Hydronephrosis. Urology 2019; 127:134.e1-134.e7. [PMID: 30790647 DOI: 10.1016/j.urology.2019.02.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Revised: 01/23/2019] [Accepted: 02/01/2019] [Indexed: 11/18/2022]
Abstract
OBJECTIVE To clarify the effect of mitochondrial injury during laparoscopic surgery of the kidney in different degrees of hydronephrosis in rabbit model. METHODS A total of 90 rabbits were randomly allocated into 3 groups (groups PN, PM, and PS, ie, rabbits without, with mild and with severe hydronephrosis, respectively). The rabbits in the PM group (n = 30) and PS group (n = 30) underwent surgical procedures that induced mild and severe left hydronephrosis, respectively. The rabbits in all the groups were then allocated into 5 subgroups and were subjected to intra-abdominal pressures of 0, 6, 9, 12, and 15 mmHg. Changes in the mitochondrial membrane potential and mitochondrial electron microstructure were observed. The apoptosis proteins cytochrome C, apoptosis-inducing factor, caspase-3, and caspase-9 were measured by western blot analysis. RESULTS As the degrees of hydronephrosis increased, histopathological changes such as the decrease in mitochondrial membrane potential and mitochondrial vacuolization along with increased expression of apoptosis proteins, cytochrome C, apoptosis-inducing factor, caspase-3 gained statistically significance at lower intra-abdominal pressures (In PN and PM groups at 15 mmHg, and in PS group at 9 mmHg; for all P <.01). CONCLUSION Mitochondrial injury plays an important role during acute kidney injury induced by pneumoperitoneal pressure in different degrees of hydronephrosis in the rabbit model.
Collapse
Affiliation(s)
- Wei Li
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Zhixiu Cao
- Department of Urology, Wuhan No.1 Hospital, Wuhan, Hubei, China
| | - Weimin Yu
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Xiaobing Yao
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Rao Ting
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Fan Cheng
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China.
| |
Collapse
|
16
|
Batinic-Haberle I, Tovmasyan A, Spasojevic I. Mn Porphyrin-Based Redox-Active Drugs: Differential Effects as Cancer Therapeutics and Protectors of Normal Tissue Against Oxidative Injury. Antioxid Redox Signal 2018; 29:1691-1724. [PMID: 29926755 PMCID: PMC6207162 DOI: 10.1089/ars.2017.7453] [Citation(s) in RCA: 104] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
SIGNIFICANCE After approximatelty three decades of research, two Mn(III) porphyrins (MnPs), MnTE-2-PyP5+ (BMX-010, AEOL10113) and MnTnBuOE-2-PyP5+ (BMX-001), have progressed to five clinical trials. In parallel, another similarly potent metal-based superoxide dismutase (SOD) mimic-Mn(II)pentaaza macrocycle, GC4419-has been tested in clinical trial on application, identical to that of MnTnBuOE-2-PyP5+-radioprotection of normal tissue in head and neck cancer patients. This clearly indicates that Mn complexes that target cellular redox environment have reached sufficient maturity for clinical applications. Recent Advances: While originally developed as SOD mimics, MnPs undergo intricate interactions with numerous redox-sensitive pathways, such as those involving nuclear factor κB (NF-κB) and nuclear factor E2-related factor 2 (Nrf2), thereby impacting cellular transcriptional activity. An increasing amount of data support the notion that MnP/H2O2/glutathione (GSH)-driven catalysis of S-glutathionylation of protein cysteine, associated with modification of protein function, is a major action of MnPs on molecular level. CRITICAL ISSUES Differential effects of MnPs on normal versus tumor cells/tissues, which support their translation into clinic, arise from differences in their accumulation and redox environment of such tissues. This in turn results in different yields of MnP-driven modifications of proteins. Thus far, direct evidence for such modification of NF-κB, mitogen-activated protein kinases (MAPK), phosphatases, Nrf2, and endogenous antioxidative defenses was provided in tumor, while indirect evidence shows the modification of NF-κB and Nrf2 translational activities by MnPs in normal tissue. FUTURE DIRECTIONS Studies that simultaneously explore differential effects in same animal are lacking, while they are essential for understanding of extremely intricate interactions of metal-based drugs with complex cellular networks of normal and cancer cells/tissues.
Collapse
Affiliation(s)
- Ines Batinic-Haberle
- 1 Department of Radiation Oncology, Duke University School of Medicine , Durham, North Carolina
| | - Artak Tovmasyan
- 1 Department of Radiation Oncology, Duke University School of Medicine , Durham, North Carolina
| | - Ivan Spasojevic
- 2 Department of Medicine, Duke University School of Medicine , Durham, North Carolina.,3 PK/PD Core Laboratory, Pharmaceutical Research Shared Resource, Duke Cancer Institute , Durham, North Carolina
| |
Collapse
|