1
|
Yu Y, Zhang L, Zhang D, Dai Q, Hou M, Chen M, Gao F, Liu XL. The role of ferroptosis in acute kidney injury: mechanisms and potential therapeutic targets. Mol Cell Biochem 2024:10.1007/s11010-024-05056-3. [PMID: 38943027 DOI: 10.1007/s11010-024-05056-3] [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: 04/07/2024] [Accepted: 06/18/2024] [Indexed: 06/30/2024]
Abstract
Acute kidney injury (AKI) is one of the most common and severe clinical renal syndromes with high morbidity and mortality. Ferroptosis is a form of programmed cell death (PCD), is characterized by iron overload, reactive oxygen species accumulation, and lipid peroxidation. As ferroptosis has been increasingly studied in recent years, it is closely associated with the pathophysiological process of AKI and provides a target for the treatment of AKI. This review offers a comprehensive overview of the regulatory mechanisms of ferroptosis, summarizes its role in various AKI models, and explores its interaction with other forms of cell death, it also presents research on ferroptosis in AKI progression to other diseases. Additionally, the review highlights methods for detecting and assessing AKI through the lens of ferroptosis and describes potential inhibitors of ferroptosis for AKI treatment. Finally, the review presents a perspective on the future of clinical AKI treatment, aiming to stimulate further research on ferroptosis in AKI.
Collapse
Affiliation(s)
- Yanxin Yu
- Yan'an Small Molecule Innovative Drug R&D Engineering Research Center, School of Medicine, Yan'an University, Yan'an, China
| | - Lei Zhang
- Yan'an Small Molecule Innovative Drug R&D Engineering Research Center, School of Medicine, Yan'an University, Yan'an, China
| | - Die Zhang
- Yan'an Small Molecule Innovative Drug R&D Engineering Research Center, School of Medicine, Yan'an University, Yan'an, China
| | - Qiangfang Dai
- Yan'an Small Molecule Innovative Drug R&D Engineering Research Center, School of Medicine, Yan'an University, Yan'an, China
| | - Mingzheng Hou
- Yan'an Small Molecule Innovative Drug R&D Engineering Research Center, School of Medicine, Yan'an University, Yan'an, China
| | - Meini Chen
- Yan'an Small Molecule Innovative Drug R&D Engineering Research Center, School of Medicine, Yan'an University, Yan'an, China
| | - Feng Gao
- Yan'an Small Molecule Innovative Drug R&D Engineering Research Center, School of Medicine, Yan'an University, Yan'an, China
| | - Xiao-Long Liu
- Yan'an Small Molecule Innovative Drug R&D Engineering Research Center, School of Medicine, Yan'an University, Yan'an, China.
| |
Collapse
|
2
|
Hassanein EHM, Ibrahim IM, Abd-Alhameed EK, Sharawi ZW, Jaber FA, Althagafy HS. Nrf2/HO-1 as a therapeutic target in renal fibrosis. Life Sci 2023; 334:122209. [PMID: 37890696 DOI: 10.1016/j.lfs.2023.122209] [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: 08/17/2023] [Revised: 10/17/2023] [Accepted: 10/23/2023] [Indexed: 10/29/2023]
Abstract
Chronic kidney disease (CKD) is one of the most prevalent chronic diseases and affects between 10 and 14 % of the world's population. The World Health Organization estimates that by 2040, the disease will be fifth in prevalence. End-stage CKD is characterized by renal fibrosis, which can eventually lead to kidney failure and death. Renal fibrosis develops due to multiple injuries and involves oxidative stress and inflammation. In the human body, nuclear factor erythroid 2-related factor 2 (Nrf2) plays an important role in the expression of antioxidant, anti-inflammatory, and cytoprotective genes, which prevents oxidative stress and inflammation damage. Heme oxygenase (HO-1) is an inducible homolog influenced by heme products and after exposure to cellular stress inducers such as oxidants, inflammatory chemokines/cytokines, and tissue damage as an outcome or downstream of Nrf2 activation. HO-1 is known for its antioxidative properties, which play an important role in regulating oxidative stress. In renal diseases-induced tissue fibrosis and xenobiotics-induced renal fibrosis, Nrf2/HO-1 has been targeted with promising results. This review summarizes these studies and highlights the interesting bioactive compounds that may assist in attenuating renal fibrosis mediated by HO-1 activation. In conclusion, Nrf2/HO-1 signal activation could have a renoprotective effect strategy against CKD caused by oxidative stress, inflammation, and consequent renal fibrosis.
Collapse
Affiliation(s)
- Emad H M Hassanein
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Al-Azhar University, Assiut, Egypt.
| | - Islam M Ibrahim
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Beni-Suef University, Beni-Suef, Egypt
| | - Esraa K Abd-Alhameed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Beni-Suef University, Beni-Suef, Egypt
| | - Zeina W Sharawi
- Biological Sciences Department, Faculty of Sciences, King AbdulAziz University, Jeddah, Saudi Arabia
| | - Fatima A Jaber
- Department of Biology, College of Science, University of Jeddah, P.O. Box 80327, Jeddah 21589, Saudi Arabia
| | - Hanan S Althagafy
- Department of Biochemistry, Faculty of Science, University of Jeddah, Jeddah, Saudi Arabia
| |
Collapse
|
3
|
Hamada S, Mae Y, Takata T, Hanada H, Kubo M, Taniguchi S, Iyama T, Sugihara T, Isomoto H. Five-Aminolevulinic Acid (5-ALA) Induces Heme Oxygenase-1 and Ameliorates Palmitic Acid-Induced Endoplasmic Reticulum Stress in Renal Tubules. Int J Mol Sci 2023; 24:10151. [PMID: 37373300 DOI: 10.3390/ijms241210151] [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: 04/21/2023] [Revised: 05/31/2023] [Accepted: 06/13/2023] [Indexed: 06/29/2023] Open
Abstract
Steatosis, or ectopic lipid deposition, is the fundamental pathophysiology of non-alcoholic steatohepatitis and chronic kidney disease. Steatosis in the renal tubule causes endoplasmic reticulum (ER) stress, leading to kidney injury. Thus, ER stress could be a therapeutic target in steatonephropathy. Five-aminolevulinic acid (5-ALA) is a natural product that induces heme oxygenase (HO)-1, which acts as an antioxidant. This study aimed to investigate the therapeutic potential of 5-ALA in lipotoxicity-induced ER stress in human primary renal proximal tubule epithelial cells. Cells were stimulated with palmitic acid (PA) to induce ER stress. Cellular apoptotic signals and expression of genes involved in the ER stress cascade and heme biosynthesis pathway were analyzed. The expression of glucose-regulated protein 78 (GRP78), a master regulator of ER stress, increased significantly, followed by increased cellular apoptosis. Administration of 5-ALA induced a remarkable increase in HO-1 expression, thus ameliorating PA-induced GRP78 expression and apoptotic signals. BTB and CNC homology 1 (BACH1), a transcriptional repressor of HO-1, was significantly downregulated by 5-ALA treatment. HO-1 induction attenuates PA-induced renal tubular injury by suppressing ER stress. This study demonstrates the therapeutic potential of 5-ALA against lipotoxicity through redox pathway.
Collapse
Affiliation(s)
- Shintaro Hamada
- Division of Gastroenterology and Nephrology, Faculty of Medicine, Tottori University, Tottori 683-8504, Japan
| | - Yukari Mae
- Division of Gastroenterology and Nephrology, Faculty of Medicine, Tottori University, Tottori 683-8504, Japan
| | - Tomoaki Takata
- Division of Gastroenterology and Nephrology, Faculty of Medicine, Tottori University, Tottori 683-8504, Japan
| | - Hinako Hanada
- Division of Gastroenterology and Nephrology, Faculty of Medicine, Tottori University, Tottori 683-8504, Japan
| | - Misaki Kubo
- Division of Gastroenterology and Nephrology, Faculty of Medicine, Tottori University, Tottori 683-8504, Japan
| | - Sosuke Taniguchi
- Division of Gastroenterology and Nephrology, Faculty of Medicine, Tottori University, Tottori 683-8504, Japan
| | - Takuji Iyama
- Division of Gastroenterology and Nephrology, Faculty of Medicine, Tottori University, Tottori 683-8504, Japan
| | - Takaaki Sugihara
- Division of Gastroenterology and Nephrology, Faculty of Medicine, Tottori University, Tottori 683-8504, Japan
| | - Hajime Isomoto
- Division of Gastroenterology and Nephrology, Faculty of Medicine, Tottori University, Tottori 683-8504, Japan
| |
Collapse
|
4
|
Horii S, Mori S, Ogata R, Nukaga S, Nishida R, Kishi S, Sasaki R, Ikemoto A, Owari T, Maesaka F, Honoki K, Miyake M, Tanaka Y, Fujimoto K, Fujiwara-Tani R, Kuniyasu H. 5-Aminolevrinic Acid Exhibits Dual Effects on Stemness in Human Sarcoma Cell Lines under Dark Conditions. Int J Mol Sci 2023; 24:ijms24076189. [PMID: 37047157 PMCID: PMC10094087 DOI: 10.3390/ijms24076189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 03/20/2023] [Accepted: 03/23/2023] [Indexed: 03/29/2023] Open
Abstract
5-aminolevulinic acid (ALA) is used for tumor-targeting phototherapy because it is converted to protoporphyrin IX (PPIX) upon excitation and induces phototoxicity. However, the effect of ALA on malignant cells under unexcited conditions is unclear. This information is essential when administering ALA systemically. We used sarcoma cell lines that usually arise deep in the body and are rarely exposed to light to examine the effects of ALA treatment under light (daylight lamp irradiation) and dark (dark room) conditions. ALA-treated human SW872 liposarcoma cells and human MG63 osteosarcoma cells cultured under light exhibited growth suppression and increased oxidative stress, while cells cultured in the dark showed no change. However, sphere-forming ability increased in the dark, and the expression of stem-cell-related genes was induced in dark, but not light, conditions. ALA administration increased heme oxygenase 1 (HO-1) expression in both cell types; when carbon monoxide (CO), a metabolite of HO-1, was administered to sarcoma cells via carbon-monoxide-releasing molecule 2 (CORM2), it enhanced sphere-forming ability. We also compared the concentration of biliverdin (BVD) (a co-product of HO-1 activity alongside CO) with sphere-forming ability when HO-1 activity was inhibited using ZnPPIX in the dark. Both cell types showed a peak in sphere-forming ability at 60–80 μM BVD. Furthermore, a cell death inhibitor assay revealed that the HO-1-induced suppression of sphere formation was rescued by apoptosis or ferroptosis inhibitors. These findings suggest that in the absence of excitation, ALA promotes HO-1 expression and enhances the stemness of sarcoma cells, although excessive HO-1 upregulation induces apoptosis and ferroptosis. Our data indicate that systemic ALA administration induces both enhanced stemness and cell death in malignant cells located in dark environments deep in the body and highlight the need to pay attention to drug delivery and ALA concentrations during phototherapy.
Collapse
|
5
|
Zhang D, Liu J, Ruan J, Jiang Z, Gong F, Lei W, Wang X, Zhao J, Meng Q, Xu M, Tang J, Li H. Combination of millet pepper and garlic water extracts improves the antioxidant capability of myofibrillar protein under malondialdehyde-induced oxidative modification. Lebensm Wiss Technol 2023. [DOI: 10.1016/j.lwt.2023.114472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
|
6
|
Nath KA, Singh RD, Croatt AJ, Adams CM. Heme Proteins and Kidney Injury: Beyond Rhabdomyolysis. KIDNEY360 2022; 3:1969-1979. [PMID: 36514409 PMCID: PMC9717624 DOI: 10.34067/kid.0005442022] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Accepted: 09/23/2022] [Indexed: 11/07/2022]
Abstract
Heme proteins, the stuff of life, represent an ingenious biologic strategy that capitalizes on the biochemical versatility of heme, and yet is one that avoids the inherent risks to cellular vitality posed by unfettered and promiscuously reactive heme. Heme proteins, however, may be a double-edged sword because they can damage the kidney in certain settings. Although such injury is often viewed mainly within the context of rhabdomyolysis and the nephrotoxicity of myoglobin, an increasing literature now attests to the fact that involvement of heme proteins in renal injury ranges well beyond the confines of this single disease (and its analog, hemolysis); indeed, through the release of the defining heme motif, destabilization of intracellular heme proteins may be a common pathway for acute kidney injury, in general, and irrespective of the underlying insult. This brief review outlines current understanding regarding processes underlying such heme protein-induced acute kidney injury (AKI) and chronic kidney disease (CKD). Topics covered include, among others, the basis for renal injury after the exposure of the kidney to and its incorporation of myoglobin and hemoglobin; auto-oxidation of myoglobin and hemoglobin; destabilization of heme proteins and the release of heme; heme/iron/oxidant pathways of renal injury; generation of reactive oxygen species and reactive nitrogen species by NOX, iNOS, and myeloperoxidase; and the role of circulating cell-free hemoglobin in AKI and CKD. Also covered are the characteristics of the kidney that render this organ uniquely vulnerable to injury after myolysis and hemolysis, and pathobiologic effects emanating from free, labile heme. Mechanisms that defend against the toxicity of heme proteins are discussed, and the review concludes by outlining the therapeutic strategies that have arisen from current understanding of mechanisms of renal injury caused by heme proteins and how such mechanisms may be interrupted.
Collapse
Affiliation(s)
- Karl A. Nath
- Division of Nephrology and Hypertension, Department of Medicine, Mayo Clinic, Rochester, Minnesota
| | - Raman Deep Singh
- Division of Nephrology and Hypertension, Department of Medicine, Mayo Clinic, Rochester, Minnesota
| | - Anthony J. Croatt
- Division of Nephrology and Hypertension, Department of Medicine, Mayo Clinic, Rochester, Minnesota
| | - Christopher M. Adams
- Division of Endocrinology, Metabolism and Nutrition, Department of Medicine, Mayo Clinic Rochester, Minnesota
| |
Collapse
|
7
|
Blocking Periostin Prevented Development of Inflammation in Rhabdomyolysis-Induced Acute Kidney Injury Mice Model. Cells 2022; 11:cells11213388. [DOI: 10.3390/cells11213388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 10/13/2022] [Accepted: 10/20/2022] [Indexed: 11/16/2022] Open
Abstract
Background: Rhabdomyolysis is the collapse of damaged skeletal muscle and the leakage of muscle-cell contents, such as electrolytes, myoglobin, and other sarcoplasmic proteins, into the circulation. The glomeruli filtered these products, leading to acute kidney injury (AKI) through several mechanisms, such as intratubular obstruction secondary to protein precipitation. The prognosis is highly mutable and depends on the underlying complications and etiologies. New therapeutic plans to reduce AKI are now needed. Up to now, several cellular pathways, with the nuclear factor kappa beta (NF-kB), as well as the proinflammatory effects on epithelial and tubular epithelial cells, have been recognized as the major pathway for the initiation of the matrix-producing cells in AKI. Recently, it has been mentioned that periostin (POSTN), an extracellular matrix protein, is involved in the development of inflammation through the modulation of the NF-kB pathway. However, how POSTN develops the inflammation protection in AKI by rhabdomyolysis is uncertain. This study aimed to investigate the role of POSTN in a rhabdomyolysis mice model of AKI induced by an intramuscular injection of 50% glycerol. Methods: In vivo, we performed an intramuscular injection of 50% glycerol (5 mg/kg body weight) to make rhabdomyolysis-induced AKI. We examined the expression level of POSTN through the progression of AKI after glycerol intramuscular injection for C57BL/6J wildtype (WT) mice. We sacrificed mice at 72 h after glycerol injection. We made periostin-null mice to examine the role of POSTN in acute renal failure. The role of periostin was further examined through in vitro methods. The development of renal inflammation is linked with the NF-kB pathway. To examine the POSTN function, we administrated hemin (100 μM) on NIH-3T3 fibroblast cells, and the following signaling pathways were examined. Results: The expression of periostin was highly increased, peaking at about 72 h after glycerol injection. The expression of inflammation-associated mRNAs such as monocyte chemotactic protein-1 (MCP-1), tumor necrosis factor-alpha (TNF-a) and IL-6, and tubular injury score in H-E staining were more reduced in POSTN-null mice than WT mice at 72 h after glycerol injection. Conclusion: POSTN was highly expressed in the kidney through rhabdomyolysis and was a positive regulator of AKI. Targeting POSTN might propose a new therapeutic strategy against the development of acute renal failure.
Collapse
|
8
|
Feng YL, Yang Y, Chen H. Small molecules as a source for acute kidney injury therapy. Pharmacol Ther 2022; 237:108169. [DOI: 10.1016/j.pharmthera.2022.108169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 02/28/2022] [Accepted: 03/07/2022] [Indexed: 10/18/2022]
|
9
|
V Ganesh G, Ganesan K, Xu B, Ramkumar KM. Nrf2 driven macrophage responses in diverse pathophysiological contexts: Disparate pieces from a shared molecular puzzle. Biofactors 2022; 48:795-812. [PMID: 35618963 DOI: 10.1002/biof.1867] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2022] [Accepted: 05/03/2022] [Indexed: 11/10/2022]
Abstract
The wide anatomical distribution of macrophages and their vast array of functions match various polarization states and their involvement in homeostasis and disease. The confluence of different cellular signaling networks, including direct involvement in inflammation, at the doorstep of the transcription factor Nuclear Factor- erythroid (NF-E2) p45-related factor 2 (Nrf2) activation raises the importance of deciphering the molecular circuitry at the background of multiple-discrete and antagonistic yet flexible and contextual pathways. While we primarily focus on wound healing and repair mechanisms that are affected in diabetic foot ulcers (DFUs), we strive to explore the striking similarities and differences in molecular events including inflammation, angiogenesis, and fibrosis during tissue injury and wound persistence that accumulates pro-inflammatory senescent macrophages, as a means to identify possible targets or cellular mediators to lessen DFU disease burden. In addition, the role of iron in the modulation of Nrf2 response in macrophages is crucial and reviewed here. Targeted approaches, unlike conventional treatments, in DFU management will require the review and re-assessment of mediators with relevance to other pathological conditions.
Collapse
Affiliation(s)
- Goutham V Ganesh
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science & Technology, Kattankulathur, Tamil Nadu, India
| | - Kumar Ganesan
- School of Chinese Medicine, LKS Faculty of Medicine, University of Hong Kong, Hong Kong
| | - Baojun Xu
- Food Science and Technology Programme, BNU-HKBU United International College, Zhuhai, Guangdong, China
| | - Kunka Mohanram Ramkumar
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science & Technology, Kattankulathur, Tamil Nadu, India
| |
Collapse
|
10
|
Chen J, Wang H, Wu Z, Gu H, Li C, Wang S, Liu G. Effects of 5-aminolevulinic acid on the inflammatory responses and antioxidative capacity in broiler chickens challenged with lipopolysaccharide. Animal 2022; 16:100575. [DOI: 10.1016/j.animal.2022.100575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 05/25/2022] [Accepted: 06/02/2022] [Indexed: 11/01/2022] Open
|
11
|
Elgendy O, Kitahara G, Taniguchi S, Osawa T. 5-Aminolevulinic acid combined with sodium ferrous citrate mitigates effects of heat stress on bovine oocyte developmental competence. J Reprod Dev 2022; 68:271-277. [PMID: 35705297 PMCID: PMC9334322 DOI: 10.1262/jrd.2021-145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
High summer temperatures have deleterious effects on oocyte developmental competence. The antioxidant and autophagy-related properties of 5-aminolevulinic acid (5-ALA) gives the compound a
broad range of biological activities. This study aimed to evaluate the effects of: 1) a high temperature-humidity index (THI) on the developmental competence of bovine oocytes, and 2) 5-ALA
administration in combination with sodium ferrous citrate (SFC) during in vitro maturation (IVM) on bovine oocyte developmental competence evaluated at high THI. Bovine
ovaries were collected from a local slaughterhouse at moderate environmental temperature (MT; THI of 56.2) and high environmental temperature (HT; THI of 76.7) periods; cumulus-oocyte
complexes (COCs) were aspirated from medium-sized follicles, matured in vitro for 22 h, fertilized, and cultured for 10 days. For COCs collected during the HT period, 0
(control), 0.01, 0.1, 0.5, or 1 µM 5-ALA was added to the maturation medium in combination with SFC at a molar ratio of 1:0.125. The results showed that HT adversely affected blastocyst and
hatching rates compared with MT. Adding 5-ALA/SFC (1 µM/0.125 µM) to the maturation medium of oocytes collected during the HT period improved cumulus cell expansion and blastocyst rates
compared with the no-addition control. In conclusion, this study showed that high THI can disrupt bovine oocyte developmental competence. Adding 5-ALA to SFC ameliorates this negative effect
of heat stress and improves subsequent embryo development.
Collapse
Affiliation(s)
- Omnia Elgendy
- Graduate School of Medicine and Veterinary Medicine, University of Miyazaki, Miyazaki 889-2192, Japan.,Department of Veterinary Sciences, Faculty of Agriculture, University of Miyazaki, Miyazaki 889-2192, Japan.,Department of Theriogenology, Faculty of Veterinary Medicine, Benha University, Qalyobia 3736, Egypt
| | - Go Kitahara
- Graduate School of Medicine and Veterinary Medicine, University of Miyazaki, Miyazaki 889-2192, Japan.,Department of Veterinary Sciences, Faculty of Agriculture, University of Miyazaki, Miyazaki 889-2192, Japan
| | - Shin Taniguchi
- Graduate School of Biosphere Science, Hiroshima University, Higashi-Hiroshima 739-8528, Japan.,One Health Business Department, Neopharma Japan Co., Ltd., Tokyo 102-0071, Japan
| | - Takeshi Osawa
- Graduate School of Medicine and Veterinary Medicine, University of Miyazaki, Miyazaki 889-2192, Japan.,Department of Veterinary Sciences, Faculty of Agriculture, University of Miyazaki, Miyazaki 889-2192, Japan
| |
Collapse
|
12
|
Cheng Y, Zhang J, Gao F, Xu Y, Wang C. Protective effects of 5-aminolevulinic acid against toxicity induced by alpha-cypermethrin to the liver-gut-microbiota axis in zebrafish. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2022; 234:113422. [PMID: 35305352 DOI: 10.1016/j.ecoenv.2022.113422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/13/2021] [Revised: 02/24/2022] [Accepted: 03/13/2022] [Indexed: 06/14/2023]
Abstract
To explore whether and how 5-aminolevulinic acid (ALA) can relieve the toxicity to the liver-gut-microbiota axis caused by alpha-cypermethrin (α-CP), adult zebrafish were exposed to α-CP (1.0 µg L-1) with or without 5.0 mg L-1 ALA supplementation. In the present work, the calculated LC50 of α-CP+ALA was 1.15 μg L-1, increasing about 1.16-fold compared with that of α-CP group (0.99 μg L-1), which indicated that ALA can alleviate the toxicity of α-CP. ALA also alleviated the histopathological lesions in the liver and gut induced by α-CP. Transcriptome sequencing of the liver showed that ALA rescues the differential expression of genes involved in the oxidation-reduction, heme metabolism, and complement activation pathways associated with dysfunctions induced by α-CP, and these findings were verified by RT-qPCR analysis and detection of the activities of enzymes in the liver-gut axis. The gut microbiota 16S rRNA sequencing results showed that α-CP alone induced gut microbial dysbiosis, which was efficiently antagonized by ALA due to decreasing the relative abundances of Cetobacterium and 3 major pathogens, and increasing the relative abundances of beneficial genera. Taken together, the results indicate that ALA might be a promising candidate for attenuating the adverse effects caused by pesticide-induced environmental pollution.
Collapse
Affiliation(s)
- Yi Cheng
- Innovation Center of Pesticide Research, Department of Applied Chemistry, College of Sciences, China Agricultural University, Beijing 100193, China.
| | - Jie Zhang
- Innovation Center of Pesticide Research, Department of Applied Chemistry, College of Sciences, China Agricultural University, Beijing 100193, China.
| | - Fei Gao
- Innovation Center of Pesticide Research, Department of Applied Chemistry, College of Sciences, China Agricultural University, Beijing 100193, China.
| | - Yong Xu
- Innovation Center of Pesticide Research, Department of Applied Chemistry, College of Sciences, China Agricultural University, Beijing 100193, China.
| | - Chengju Wang
- Innovation Center of Pesticide Research, Department of Applied Chemistry, College of Sciences, China Agricultural University, Beijing 100193, China.
| |
Collapse
|
13
|
Yuqiang C, Lisha Z, Jiejun W, Qin X, Niansong W. Pifithrin-α ameliorates glycerol induced rhabdomyolysis and acute kidney injury by reducing p53 activation. Ren Fail 2022; 44:473-481. [PMID: 35285384 PMCID: PMC8928845 DOI: 10.1080/0886022x.2022.2048857] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
Objectives Rhabdomyolysis is a series of symptoms caused by the dissolution of striped muscle, and acute kidney injury (AKI) is a potential complication of severe rhabdomyolysis. The underlying causes of AKI are remarkably complex and diverse. Here, we aim to investigate whether pifithrin-α protected against rhabdomyolysis-induced AKI and to determine the involved mechanisms. Methods Intramuscular injection in the right thigh caudal muscle of C57BL/6J mice with 7.5 ml/kg saline (Group A) or of the same volume 50% glycerol was used to induce rhabdomyolysis and subsequent AKI (Group B). Pifithrin-α was injected intraperitoneally 4 h before (Group C) or 4 h after (Group D) the glycerol injection. Serum creatine kinase, blood urea nitrogen, and creatinine were determined, and the renal cortex was histologically analyzed. Renal expression levels of interested mRNAs and proteins were determined and compared, too. Results Intramuscular injection of glycerol induced rhabdomyolysis and subsequent AKI in mice (Groups B–D). Renal function reduction and histologic injury of renal tubular epithelial cells were associated with increased p53 activation, oxidative stress, and inflammation. Notably, compared with pifithrin-α rescue therapy (Group D), pretreatment of pifithrin-α (Group C) protected the mice from severe injury more effectively. Conclusions Our present study suggests that p53 may be a therapeutic target of AKI caused by glycerol, and the inhibition of p53 can block glycerol-mediated AKI by using pharmacological agents instead of genetic inhibitory approaches, which further supports that p53 played a pivotal role in renal tubular injury when challenged with glycerol.
Collapse
Affiliation(s)
- Chen Yuqiang
- Department of Nephrology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Zhang Lisha
- Department of Emergency, Shanghai Punan Hospital, Pudong New District, Shanghai, China
| | - Wen Jiejun
- Department of Nephrology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Xue Qin
- Department of Nephrology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Wang Niansong
- Department of Nephrology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| |
Collapse
|
14
|
Kaur T, Singh D, Pathak D, Singh AP, Singh B. Umbelliferone attenuates glycerol-induced myoglobinuric acute kidney injury through peroxisome proliferator-activated receptor-γ agonism in rats. J Biochem Mol Toxicol 2021; 35:e22892. [PMID: 34409680 DOI: 10.1002/jbt.22892] [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: 01/29/2021] [Revised: 06/17/2021] [Accepted: 08/11/2021] [Indexed: 11/07/2022]
Abstract
Rhabdomyolysis is a clinical syndrome caused by damage to skeletal muscle, which consequently releases breakdown products into circulation and causes acute kidney injury (AKI) in humans. Intramuscular injection of glycerol mimics rhabdomyolysis and associated AKI. In this study, we explored the role of umbelliferone against glycerol-induced AKI in rats. Kidney function was assessed by measuring serum creatinine, urea, electrolytes, and microproteinuria. Renal oxidative stress was quantified using thiobarbituric acid reactive substances, superoxide anion generation, and reduced glutathione assay. Renal histological changes were determined using periodic acid Schiff and hematoxylin-eosin staining, and immunohistology of apoptotic markers (Bax, Bcl-2) was done. Serum creatine kinase was quantified to assess glycerol-induced muscle damage. Umbelliferone attenuated glycerol-induced change in biochemical parameters, oxidative stress, histological alterations, and renal apoptosis. Pretreatment with bisphenol A diglycidyl ether, a peroxisome proliferator-activated receptor-γ (PPAR-γ) antagonist, attenuated umbelliferone-mediated protection. It is concluded that umbelliferone attenuates glycerol-induced AKI possibly through PPAR-γ agonism in rats.
Collapse
Affiliation(s)
- Tajpreet Kaur
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, Amritsar, India
| | - Damanpreet Singh
- Pharmacology and Toxicology Laboratory, Dietetics and Nutrition Technology Division, CSIR-Institute of Himalayan Bioresource Technology, Palampur, India
| | - Devendra Pathak
- Department of Veterinary Anatomy, Guru Angad Dev Veterinary and Animal Sciences University, Ludhiana, India
| | - Amrit P Singh
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, Amritsar, India
| | - Balbir Singh
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, Amritsar, India
| |
Collapse
|
15
|
Ji SY, Cha HJ, Molagoda IMN, Kim MY, Kim SY, Hwangbo H, Lee H, Kim GY, Kim DH, Hyun JW, Kim HS, Kim S, Jin CY, Choi YH. Suppression of Lipopolysaccharide-Induced Inflammatory and Oxidative Response by 5-Aminolevulinic Acid in RAW 264.7 Macrophages and Zebrafish Larvae. Biomol Ther (Seoul) 2021; 29:685-696. [PMID: 33820881 PMCID: PMC8551728 DOI: 10.4062/biomolther.2021.030] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 03/11/2021] [Accepted: 03/15/2021] [Indexed: 11/05/2022] Open
Abstract
In this study, we investigated the inhibitory effect of 5-aminolevulinic acid (ALA), a heme precursor, on inflammatory and oxidative stress activated by lipopolysaccharide (LPS) in RAW 264.7 macrophages by estimating nitric oxide (NO), prostaglandin E2 (PGE2), cytokines, and reactive oxygen species (ROS). We also evaluated the molecular mechanisms through analysis of the expression of their regulatory genes, and further evaluated the anti-inflammatory and antioxidant efficacy of ALA against LPS in the zebrafish model. Our results indicated that ALA treatment significantly attenuated the LPS-induced release of pro-inflammatory mediators including NO and PGE2, which was associated with decreased inducible NO synthase and cyclooxygenase-2 expression. ALA also inhibited the LPS-induced expression of pro-inflammatory cytokines, such as tumor necrosis factor (TNF)-α, interleukin (IL)-1β, and IL-6, reducing their extracellular secretion. Additionally, ALA abolished ROS generation, improved the mitochondrial mass, and enhanced the expression of heme oxygenase-1 (HO-1) and the activation of nuclear translocation of nuclear factor-E2-related factor 2 (Nrf2) in LPS-stimulated RAW 264.7 macrophages. However, zinc protoporphyrin, a specific inhibitor of HO-1, reversed the ALA-mediated inhibition of pro-inflammatory cytokines production and activation of mitochondrial function in LPS-treated RAW 264.7 macrophages. Furthermore, ALA significantly abolished the expression of LPS-induced pro-inflammatory mediators and cytokines, and showed strong protective effects against NO and ROS production in zebrafish larvae. In conclusion, our findings suggest that ALA exerts LPS-induced anti-inflammatory and antioxidant effects by upregulating the Nrf2/HO-1 signaling pathway, and that ALA can be a potential functional agent to prevent inflammatory and oxidative damage.
Collapse
Affiliation(s)
- Seon Yeong Ji
- Anti-Aging Research Center, Dong-eui University, Busan 47340, Republic of Korea.,Department of Biochemistry, College of Korean Medicine, Dong-eui University, Busan 47227, Republic of Korea
| | - Hee-Jae Cha
- Department of Parasitology and Genetics, College of Medicine, Kosin University, Busan 49104, Republic of Korea
| | | | - Min Yeong Kim
- Anti-Aging Research Center, Dong-eui University, Busan 47340, Republic of Korea.,Department of Biochemistry, College of Korean Medicine, Dong-eui University, Busan 47227, Republic of Korea
| | - So Young Kim
- Anti-Aging Research Center, Dong-eui University, Busan 47340, Republic of Korea.,Department of Biochemistry, College of Korean Medicine, Dong-eui University, Busan 47227, Republic of Korea
| | - Hyun Hwangbo
- Anti-Aging Research Center, Dong-eui University, Busan 47340, Republic of Korea.,Department of Biochemistry, College of Korean Medicine, Dong-eui University, Busan 47227, Republic of Korea
| | - Hyesook Lee
- Anti-Aging Research Center, Dong-eui University, Busan 47340, Republic of Korea.,Department of Biochemistry, College of Korean Medicine, Dong-eui University, Busan 47227, Republic of Korea
| | - Gi-Young Kim
- Department of Marine Life Sciences, Jeju National University, Jeju 63243, Republic of Korea
| | - Do-Hyung Kim
- Department of Aquatic Life Medicine, Pukyong National University, Busan 48513, Republic of Korea
| | - Jin Won Hyun
- Department of Biochemistry, College of Medicine, Jeju National University, Jeju 63243, Republic of Korea
| | - Heui-Soo Kim
- Department of Biological Sciences, Pusan National University, Busan 46241, Republic of Korea
| | - Suhkmann Kim
- Department of Chemistry, Pusan National University, Busan 46241, Republic of Korea
| | - Cheng-Yun Jin
- School of Pharmaceutical Sciences, Zhengzhou University, Henan 450001, China
| | - Yung Hyun Choi
- Anti-Aging Research Center, Dong-eui University, Busan 47340, Republic of Korea.,Department of Biochemistry, College of Korean Medicine, Dong-eui University, Busan 47227, Republic of Korea
| |
Collapse
|
16
|
Sun T, Liu Q, Wang Y, Deng Y, Zhang D. MBD2 mediates renal cell apoptosis via activation of Tox4 during rhabdomyolysis-induced acute kidney injury. J Cell Mol Med 2021; 25:4562-4571. [PMID: 33764669 PMCID: PMC8107094 DOI: 10.1111/jcmm.16207] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 12/02/2020] [Accepted: 12/07/2020] [Indexed: 12/18/2022] Open
Abstract
Our study investigated the role of Methyl‐CpG–binding domain protein 2 (MBD2) in RM‐induced acute kidney injury (AKI) both in vitro and in vivo. MBD2 was induced by myoglobin in BUMPT cells and by glycerol in mice. MBD2 inhibition via MBD2 small interfering RNA and MBD2‐knockout (KO) attenuated RM‐induced AKI and renal cell apoptosis. The expression of TOX high mobility group box family member 4 (Tox4) induced by myoglobin was markedly reduced in MBD2‐KO mice. Chromatin immunoprecipitation analysis indicated that MBD2 directly bound to CpG islands in the Tox4 promoter region, thus preventing promoter methylation. Furthermore, siRNA inhibition of Tox4 attenuated myoglobin‐induced apoptosis in BUMPT cells. Finally, MBD2‐KO mice exhibited glycerol‐induced renal cell apoptosis by inactivation of Tox4. Altogether, our results suggested that MBD2 plays a role in RM‐induced AKI via the activation of Tox4 and represents a potential target for treatment of RM‐associated AKI.
Collapse
Affiliation(s)
- Tianshi Sun
- Department of Spine Surgery, The Third Xiangya Hospital of Central South University, Changsha, China.,Department of Emergency Medicine, Second Xiangya Hospital of Central South University, Changsha, China.,Emergency Medicine and Difficult Diseases Institute, Second Xiangya Hospital of Central South University, Changsha, China
| | - Qing Liu
- Department of Spine Surgery, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Yifan Wang
- Department of Emergency Medicine, Second Xiangya Hospital of Central South University, Changsha, China.,Emergency Medicine and Difficult Diseases Institute, Second Xiangya Hospital of Central South University, Changsha, China
| | - Youwen Deng
- Department of Spine Surgery, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Dongshan Zhang
- Department of Emergency Medicine, Second Xiangya Hospital of Central South University, Changsha, China.,Emergency Medicine and Difficult Diseases Institute, Second Xiangya Hospital of Central South University, Changsha, China.,Department of Nephrology, Second Xiangya Hospital of Central South University, Changsha, China
| |
Collapse
|
17
|
Sharmin MM, Islam MA, Yamamoto I, Taniguchi S, Yonekura S. 5-ALA Attenuates the Palmitic Acid-Induced ER Stress and Apoptosis in Bovine Mammary Epithelial Cells. Molecules 2021; 26:molecules26041183. [PMID: 33672109 PMCID: PMC7926617 DOI: 10.3390/molecules26041183] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2020] [Revised: 02/19/2021] [Accepted: 02/19/2021] [Indexed: 11/24/2022] Open
Abstract
The conservation of mammary gland physiology by maintaining the maximum number of mammary epithelial cells (MECs) is of the utmost importance for the optimum amount of milk production. In a state of negative energy balance, palmitic acid (PA) reduces the number of bovine MECs. However, there is no effective strategy against PA-induced apoptosis of MECs. In the present study, 5-aminolevulinic acid (5-ALA) was established as a remedial agent against PA-induced apoptosis of MAC-T cells (an established line of bovine MECs). In PA-treated cells, the apoptosis-related genes BCL2 and BAX were down- and upregulated, respectively. The elevated expression of major genes of the unfolded protein response (UPR), such as CHOP, a proapoptotic marker (C/EBP homologous protein), reduced the viability of PA-treated MAC-T cells. In contrast, 5-ALA pretreatment increased and decreased BCL2 and BAX expression, respectively. Moreover, cleaved caspase-3 protein expression was significantly reduced in the 5-ALA-pretreated group in comparison with the PA group. The downregulation of major UPR-related genes, including CHOP, extended the viability of MAC-T cells pretreated with 5-ALA and also reduced the enhanced intensity of the PA-induced expression of phospho-protein kinase R-like ER kinase. Moreover, the enhanced expression of HO-1 (antioxidant gene heme oxygenase) by 5-ALA reduced PA-induced oxidative stress (OxS). HO-1 is not only protective against OxS but also effective against ER stress. Collectively, these findings offer new insights into the protective effects of 5-ALA against PA-induced apoptosis of bovine MECs.
Collapse
Affiliation(s)
- Mst Mamuna Sharmin
- Department of Biomedical Engineering, Graduate School of Medicine, Science and Technology, Shinshu University, Minamiminowa, Kamiina-gun, Nagano 399-4598, Japan; (M.M.S.); (M.A.I.)
| | - Md Aminul Islam
- Department of Biomedical Engineering, Graduate School of Medicine, Science and Technology, Shinshu University, Minamiminowa, Kamiina-gun, Nagano 399-4598, Japan; (M.M.S.); (M.A.I.)
| | - Itsuki Yamamoto
- Department of Biomedical Engineering, Graduate School of Science and Technology, Shinshu University, Minamiminowa, Kamiina, Nagano 399-4598, Japan;
| | - Shin Taniguchi
- Neopharma Japan Co., Ltd., Tokyo 102-0071, Japan;
- Graduate School of Biosphere Science, Hiroshima University, Higashi-Hiroshima 739-8528, Japan
| | - Shinichi Yonekura
- Department of Biomedical Engineering, Graduate School of Medicine, Science and Technology, Shinshu University, Minamiminowa, Kamiina-gun, Nagano 399-4598, Japan; (M.M.S.); (M.A.I.)
- Department of Biomedical Engineering, Graduate School of Science and Technology, Shinshu University, Minamiminowa, Kamiina, Nagano 399-4598, Japan;
- Department of Biomolecular Innovation, Institute for Biomedical Sciences, Shinshu University, 8304 Minamiminowa, Kamiina, Nagano 399-4598, Japan
- Correspondence: ; Tel.: +81-265-77-1443
| |
Collapse
|
18
|
Hejazian SM, Hosseiniyan Khatibi SM, Barzegari A, Pavon-Djavid G, Razi Soofiyani S, Hassannejhad S, Ahmadian E, Ardalan M, Zununi Vahed S. Nrf-2 as a therapeutic target in acute kidney injury. Life Sci 2020; 264:118581. [PMID: 33065149 DOI: 10.1016/j.lfs.2020.118581] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 09/27/2020] [Accepted: 10/05/2020] [Indexed: 02/06/2023]
Abstract
Multifaceted cellular pathways exhibit a crucial role in the preservation of homeostasis at the molecular, cellular, and organism levels. One of the most important of these protective cascades is Nuclear factor E2-related factor (Nrf-2) that regulates the expression of several genes responsible for cellular detoxification, antioxidant function, anti-inflammation, drug/xenobiotic transportation, and stress-related factors. A growing body of evidence provides information regarding the protective role of Nrf-2 against a number of kidney diseases. Acute kidney injury (AKI) is a substantial clinical problem that causes a huge social burden. In the kidneys, Nrf-2 exerts a dynamic role in improving the injury triggered by inflammation and oxidative stress. Understanding of the exact molecular mechanisms underlying AKI is vital in order to determine the equilibrium between renal adaptation and malfunction and thus reduce disease progression. This review highlights the role of Nrf-2 targeting against AKI and provides evidence that targeting Nrf-2 to prevail oxidative damage and its consequences might exhibit protective effects in kidney diseases.
Collapse
Affiliation(s)
- Seyyedeh Mina Hejazian
- Kidney Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Abolfazl Barzegari
- Research Center for Pharmaceutical Nanotechnology, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Graciela Pavon-Djavid
- INSERM U1148, Laboratory for Vascular Translational Science, Cardiovascular Bioengineering, Université Sorbonne Paris Nord, Paris, France
| | | | - Sina Hassannejhad
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran; Research Development and Coordination Center (RDCC), Faculty of Medicine, Tabriz University of Medical Sciences, Iran
| | - Elham Ahmadian
- Kidney Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | | |
Collapse
|
19
|
Wei W, Ma N, Fan X, Yu Q, Ci X. The role of Nrf2 in acute kidney injury: Novel molecular mechanisms and therapeutic approaches. Free Radic Biol Med 2020; 158:1-12. [PMID: 32663513 DOI: 10.1016/j.freeradbiomed.2020.06.025] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 05/24/2020] [Accepted: 06/11/2020] [Indexed: 12/13/2022]
Abstract
Acute kidney injury (AKI) is a common clinical syndrome that is related to high morbidity and mortality. Oxidative stress, including the production of reactive oxygen species (ROS), appears to be the main element in the occurrence of AKI and the cause of the progression of chronic kidney disease (CKD) into end-stage renal disease (ESRD). Nuclear factor erythroid 2 related factor 2 (Nrf2) is a significant regulator of redox balance that has been shown to improve kidney disease by eliminating ROS. To date, researchers have found that the use of Nrf2-activated compounds can effectively reduce ROS, thereby preventing or retarding the progression of various types of AKI. In this review, we summarized the molecular mechanisms of Nrf2 and ROS in AKI and described the latest findings on the therapeutic potential of Nrf2 activators in various types of AKI.
Collapse
Affiliation(s)
- Wei Wei
- Department of Urology, The First Hospital, Jilin University, Changchun, China
| | - Ning Ma
- Department of Urology, The First Hospital, Jilin University, Changchun, China
| | - Xiaoye Fan
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun, China
| | - Qinlei Yu
- Jilin Provincial Animal Disease Control Center, 4510 Xi'an Road, Changchun, 130062, China
| | - Xinxin Ci
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun, China.
| |
Collapse
|
20
|
Islam MA, Noguchi Y, Taniguchi S, Yonekura S. Protective effects of 5-aminolevulinic acid on heat stress in bovine mammary epithelial cells. Anim Biosci 2020; 34:1006-1013. [PMID: 32898952 PMCID: PMC8100485 DOI: 10.5713/ajas.20.0349] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Accepted: 07/27/2020] [Indexed: 02/06/2023] Open
Abstract
OBJECTIVE Cells have increased susceptibility to activation of apoptosis when suffering heat stress (HS). An effective supplementation strategy to mimic heat-induced apoptosis of bovine mammary epithelial cells (MECs) is necessary to maintain optimal milk production. This study aimed to investigate possible protective effects of the anti-apoptotic activity of 5-aminolevulinic acid (5-ALA) against HS-induced damage of bovine MECs. METHODS Bovine MECs were pretreated with or without 5-ALA at concentrations of 10, 100, and 500 μM for 24 h followed by HS (42.5°C for 24 h and 48 h). Cell viability was measured with 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assays. Real-time quantitative polymerase chain reaction and Western blotting were used to explore the regulation of genes associated with apoptosis, oxidative stress, and endoplasmic reticulum (ER) stress genes. RESULTS We found that 5-ALA induces cytoprotection via inhibition of apoptosis markers after HS-induced damage. Pretreatment of bovine MECs with 5-ALA resulted in dramatic upregulation of mRNA for nuclear factor erythroid-derived 2-like factor 2, heme oxygenase-1, and NAD(P)H quinone oxidoreductase 1, all of which are antioxidant stress genes. Moreover, 5-ALA pretreatment significantly suppressed HS-induced ER stress-associated markers, glucose-regulated protein 78, and C/EBP homologous protein expression levels. CONCLUSION 5-ALA can ameliorate the ER stress in heat stressed bovine MEC via enhancing the expression of antioxidant gene.
Collapse
Affiliation(s)
- Md Aminul Islam
- Graduate School of Medicine, Science and Technology, Shinshu University, Kamiina, Nagano 399-4598, Japan
| | | | - Shin Taniguchi
- Neopharma Japan Co., Ltd. Tokyo 102-0071, Japan.,Graduate School of Biosphere Science, Hiroshima University, Higashi-Hiroshima 739-8528, Japan
| | - Shinichi Yonekura
- Graduate School of Medicine, Science and Technology, Shinshu University, Kamiina, Nagano 399-4598, Japan.,Department of Biomolecular Innovation, Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research, Shinshu University, Kamiina, Nagano 399-4598, Japan
| |
Collapse
|
21
|
A prospective study of oral 5-aminolevulinic acid to prevent adverse events in patients with localized prostate cancer undergoing low-dose-rate brachytherapy: Protocol of the AMBER study. Contemp Clin Trials Commun 2020; 19:100593. [PMID: 32637724 PMCID: PMC7327239 DOI: 10.1016/j.conctc.2020.100593] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 06/07/2020] [Accepted: 06/14/2020] [Indexed: 12/14/2022] Open
Abstract
Background Radiotherapy is one of the most frequently selected treatment options for patients with prostate cancer. However, adverse effects related to the irradiated surrounding normal organs are significant clinical concerns. Specifically, genitourinary and gastrointestinal toxicities can lead to a dramatically reduced quality of life. The aim of this clinical trial is to determine the efficacy of oral 5-aminolevulinic acid (ALA) phosphate with sodium ferrous citrate (SFC) in patients treated with low-dose-rate brachytherapy (LDR-BT) using an iodine-125 seed source. Methods The AMBER study is a prospective, single-center trial in patients with localized prostate cancer undergoing LDR-BT. Patients who undergo supplementary extra-beam radiotherapy are excluded, whereas those who undergo pre-implantation short-term (4–6 months) androgen deprivation therapy to decrease the prostate volume and/or improve oncological outcomes are included. After the screening and registration, the patients will be instructed to take capsules of ALA-SFC twice a day (200 mg and 229.42 mg per day) for 6 months from the day of seed implantation (prescribed radiation dose of 160 Gy). Patient data will be collected before the implantation; during oral ALA-SFC treatment; and 1, 3, 6, 9, and 12 month(s) after seed implantation. The primary endpoint of this trial is the urinary frequency 3 months after seed implantation. At each visit, the 24-h urinary frequency, total voided volume, and mean voided volume on a frequency volume chart and other patient-reported outcomes are recorded. The data of the trial cases will be compared with those of historical controls, who are consecutive patients undergoing LDR-BT without supplementary extra-beam radiotherapy between January 2016 and January 2019. The number of subjects has been set to be 50 for trial cases and 150 for the historical control cases. Pre- and post-treatment clinicopathologic factors are compared between two groups. Discussion The goal of this trial is to determine the potential benefit of ALA-SFC in patients who undergo LDR-BT. To the best of our knowledge, this is the first study investigating the potential clinical benefit of oral ALA-SFC after radiotherapy. More evidence from a further randomized controlled trial is needed to change the standard of care and lead to better post-radiotherapy management. Trial registration This clinical trial was prospectively registered with the Japan Registry of Clinical Trials on 5 December 2019. The reference number is jRCTs051190077, nara0013 (Certified Review Board of Nara Medical University).
Collapse
Key Words
- 5-Aminolevulinic acid
- ALA, 5-aminolevulinic acid
- Adverse event
- CTCAE, Common Toxicity Criteria for Adverse Events
- EBRT, extra-beam radiotherapy
- EPIC, Expanded Prostate Cancer Index Composite
- GI, gastrointestinal
- GU, genitourinary
- I-125, iodine-125
- IPSS, International Prostate Symptom Score
- J-POPS, Japanese nationwide prospective cohort study
- LDR-BT, low-dose-rate brachytherapy
- Low-dose-rate brachytherapy
- OABSS, overactive bladder symptom score
- PCa, prostate cancer
- PRO, patient reported outcome
- PSA, prostate-specific antigen
- Prostate cancer
- QOL, quality of life
- Radioprotection
- Radiotherapy
- SFC, sodium ferrous citrate
- SHIM, Sexual Health Inventory for Men
- Urinary frequency
Collapse
|
22
|
Taguchi K, Ogaki S, Nagasaki T, Yanagisawa H, Nishida K, Maeda H, Enoki Y, Matsumoto K, Sekijima H, Ooi K, Ishima Y, Watanabe H, Fukagawa M, Otagiri M, Maruyama T. Carbon Monoxide Rescues the Developmental Lethality of Experimental Rat Models of Rhabdomyolysis-Induced Acute Kidney Injury. J Pharmacol Exp Ther 2020; 372:355-365. [PMID: 31924689 DOI: 10.1124/jpet.119.262485] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Accepted: 12/26/2019] [Indexed: 02/05/2023] Open
Abstract
Many victims, after being extricated from a collapsed building as the result of a disaster, suffer from disaster nephrology, a term that is referred to as the crush syndrome (CS). Recommended treatments, which include dialysis or the continuous administration of massive amounts of fluid are not usually easy in cases of such mass natural disasters. In the present study, we examined the therapeutic performance of a biomimetic carbon monoxide (CO) delivery system, CO-enriched red blood cells (CO-RBCs), on experimental animal models of an acute kidney injury (AKI) induced by traumatic and nontraumatic rhabdomyolysis, including CS and rhabdomyolysis with massive hemorrhage shock. A single CO-RBC treatment was found to effectively suppress the pathogenesis of AKI with the mortality in these model rats being improved. In addition, in further studies using glycerol-induced rhabdomyolysis model rats, the pathogenesis of which is similar to that for the CS, AKI and mortality were also reduced as the result of a CO-RBC treatment. Furthermore, CO-RBCs were found to have renoprotective effects via the suppression of subsequent heme protein-associated renal oxidative injury; the oxidation of myoglobin in the kidneys, the generation of reactive oxygen species by free heme produced from degraded-cytochrome P450 and hemoglobin-associated renal injury. Because CO-RBCs can be prepared and used at both hospitals and at a disaster site, these findings suggest that CO-RBCs have the potential for use as a novel cell therapy against both nontraumatic and traumatic rhabdomyolysis including CS-induced AKI. SIGNIFICANCE STATEMENT: After mass natural and man-made disasters, people who are trapped in collapsed buildings are in danger of acute kidney injury (AKI), including crush syndrome (CS)-related AKI. This paper reports that carbon monoxide-enriched red blood cells (CO-RBCs), which can be prepared at both hospitals and disaster sites, dramatically suppressed the pathogenesis of CS-related AKI, thus improving mortality via suppressing heme protein-associated renal injuries. CO-RBCs have the potential for serving as a practical therapeutic agent against disaster nephrology associated with the CS.
Collapse
Affiliation(s)
- Kazuaki Taguchi
- Division of Pharmacodynamics, Faculty of Pharmacy, Keio University, Tokyo, Japan (K.T., Y.E., K.M.); Department of Pharmacokinetics, Faculty of Pharmaceutical Sciences (K.T., M.O.) and DDS Research Institute (M.O.), Sojo University, Kumamoto, Japan; Department of Biopharmaceutics, Graduate School of Pharmaceutical Sciences (S.O., T.N., H.Y., K.N., H.M., H.W., T.M.) and Center for Clinical Pharmaceutical Sciences, Graduate School of Pharmaceutical Sciences (H.W., T.M.), Kumamoto University, Kumamoto, Japan; Laboratory of Clinical Pharmacology, Faculty of Pharmaceutical Sciences, Suzuka University of Medical Science, Mie, Japan (H.S., K.O.); Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University, Tokushima, Japan (Y.I.); and Division of Nephrology, Endocrinology and Metabolism, Tokai University School of Medicine, Isehara, Japan (M.F.)
| | - Shigeru Ogaki
- Division of Pharmacodynamics, Faculty of Pharmacy, Keio University, Tokyo, Japan (K.T., Y.E., K.M.); Department of Pharmacokinetics, Faculty of Pharmaceutical Sciences (K.T., M.O.) and DDS Research Institute (M.O.), Sojo University, Kumamoto, Japan; Department of Biopharmaceutics, Graduate School of Pharmaceutical Sciences (S.O., T.N., H.Y., K.N., H.M., H.W., T.M.) and Center for Clinical Pharmaceutical Sciences, Graduate School of Pharmaceutical Sciences (H.W., T.M.), Kumamoto University, Kumamoto, Japan; Laboratory of Clinical Pharmacology, Faculty of Pharmaceutical Sciences, Suzuka University of Medical Science, Mie, Japan (H.S., K.O.); Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University, Tokushima, Japan (Y.I.); and Division of Nephrology, Endocrinology and Metabolism, Tokai University School of Medicine, Isehara, Japan (M.F.)
| | - Taisei Nagasaki
- Division of Pharmacodynamics, Faculty of Pharmacy, Keio University, Tokyo, Japan (K.T., Y.E., K.M.); Department of Pharmacokinetics, Faculty of Pharmaceutical Sciences (K.T., M.O.) and DDS Research Institute (M.O.), Sojo University, Kumamoto, Japan; Department of Biopharmaceutics, Graduate School of Pharmaceutical Sciences (S.O., T.N., H.Y., K.N., H.M., H.W., T.M.) and Center for Clinical Pharmaceutical Sciences, Graduate School of Pharmaceutical Sciences (H.W., T.M.), Kumamoto University, Kumamoto, Japan; Laboratory of Clinical Pharmacology, Faculty of Pharmaceutical Sciences, Suzuka University of Medical Science, Mie, Japan (H.S., K.O.); Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University, Tokushima, Japan (Y.I.); and Division of Nephrology, Endocrinology and Metabolism, Tokai University School of Medicine, Isehara, Japan (M.F.)
| | - Hiroki Yanagisawa
- Division of Pharmacodynamics, Faculty of Pharmacy, Keio University, Tokyo, Japan (K.T., Y.E., K.M.); Department of Pharmacokinetics, Faculty of Pharmaceutical Sciences (K.T., M.O.) and DDS Research Institute (M.O.), Sojo University, Kumamoto, Japan; Department of Biopharmaceutics, Graduate School of Pharmaceutical Sciences (S.O., T.N., H.Y., K.N., H.M., H.W., T.M.) and Center for Clinical Pharmaceutical Sciences, Graduate School of Pharmaceutical Sciences (H.W., T.M.), Kumamoto University, Kumamoto, Japan; Laboratory of Clinical Pharmacology, Faculty of Pharmaceutical Sciences, Suzuka University of Medical Science, Mie, Japan (H.S., K.O.); Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University, Tokushima, Japan (Y.I.); and Division of Nephrology, Endocrinology and Metabolism, Tokai University School of Medicine, Isehara, Japan (M.F.)
| | - Kento Nishida
- Division of Pharmacodynamics, Faculty of Pharmacy, Keio University, Tokyo, Japan (K.T., Y.E., K.M.); Department of Pharmacokinetics, Faculty of Pharmaceutical Sciences (K.T., M.O.) and DDS Research Institute (M.O.), Sojo University, Kumamoto, Japan; Department of Biopharmaceutics, Graduate School of Pharmaceutical Sciences (S.O., T.N., H.Y., K.N., H.M., H.W., T.M.) and Center for Clinical Pharmaceutical Sciences, Graduate School of Pharmaceutical Sciences (H.W., T.M.), Kumamoto University, Kumamoto, Japan; Laboratory of Clinical Pharmacology, Faculty of Pharmaceutical Sciences, Suzuka University of Medical Science, Mie, Japan (H.S., K.O.); Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University, Tokushima, Japan (Y.I.); and Division of Nephrology, Endocrinology and Metabolism, Tokai University School of Medicine, Isehara, Japan (M.F.)
| | - Hitoshi Maeda
- Division of Pharmacodynamics, Faculty of Pharmacy, Keio University, Tokyo, Japan (K.T., Y.E., K.M.); Department of Pharmacokinetics, Faculty of Pharmaceutical Sciences (K.T., M.O.) and DDS Research Institute (M.O.), Sojo University, Kumamoto, Japan; Department of Biopharmaceutics, Graduate School of Pharmaceutical Sciences (S.O., T.N., H.Y., K.N., H.M., H.W., T.M.) and Center for Clinical Pharmaceutical Sciences, Graduate School of Pharmaceutical Sciences (H.W., T.M.), Kumamoto University, Kumamoto, Japan; Laboratory of Clinical Pharmacology, Faculty of Pharmaceutical Sciences, Suzuka University of Medical Science, Mie, Japan (H.S., K.O.); Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University, Tokushima, Japan (Y.I.); and Division of Nephrology, Endocrinology and Metabolism, Tokai University School of Medicine, Isehara, Japan (M.F.)
| | - Yuki Enoki
- Division of Pharmacodynamics, Faculty of Pharmacy, Keio University, Tokyo, Japan (K.T., Y.E., K.M.); Department of Pharmacokinetics, Faculty of Pharmaceutical Sciences (K.T., M.O.) and DDS Research Institute (M.O.), Sojo University, Kumamoto, Japan; Department of Biopharmaceutics, Graduate School of Pharmaceutical Sciences (S.O., T.N., H.Y., K.N., H.M., H.W., T.M.) and Center for Clinical Pharmaceutical Sciences, Graduate School of Pharmaceutical Sciences (H.W., T.M.), Kumamoto University, Kumamoto, Japan; Laboratory of Clinical Pharmacology, Faculty of Pharmaceutical Sciences, Suzuka University of Medical Science, Mie, Japan (H.S., K.O.); Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University, Tokushima, Japan (Y.I.); and Division of Nephrology, Endocrinology and Metabolism, Tokai University School of Medicine, Isehara, Japan (M.F.)
| | - Kazuaki Matsumoto
- Division of Pharmacodynamics, Faculty of Pharmacy, Keio University, Tokyo, Japan (K.T., Y.E., K.M.); Department of Pharmacokinetics, Faculty of Pharmaceutical Sciences (K.T., M.O.) and DDS Research Institute (M.O.), Sojo University, Kumamoto, Japan; Department of Biopharmaceutics, Graduate School of Pharmaceutical Sciences (S.O., T.N., H.Y., K.N., H.M., H.W., T.M.) and Center for Clinical Pharmaceutical Sciences, Graduate School of Pharmaceutical Sciences (H.W., T.M.), Kumamoto University, Kumamoto, Japan; Laboratory of Clinical Pharmacology, Faculty of Pharmaceutical Sciences, Suzuka University of Medical Science, Mie, Japan (H.S., K.O.); Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University, Tokushima, Japan (Y.I.); and Division of Nephrology, Endocrinology and Metabolism, Tokai University School of Medicine, Isehara, Japan (M.F.)
| | - Hidehisa Sekijima
- Division of Pharmacodynamics, Faculty of Pharmacy, Keio University, Tokyo, Japan (K.T., Y.E., K.M.); Department of Pharmacokinetics, Faculty of Pharmaceutical Sciences (K.T., M.O.) and DDS Research Institute (M.O.), Sojo University, Kumamoto, Japan; Department of Biopharmaceutics, Graduate School of Pharmaceutical Sciences (S.O., T.N., H.Y., K.N., H.M., H.W., T.M.) and Center for Clinical Pharmaceutical Sciences, Graduate School of Pharmaceutical Sciences (H.W., T.M.), Kumamoto University, Kumamoto, Japan; Laboratory of Clinical Pharmacology, Faculty of Pharmaceutical Sciences, Suzuka University of Medical Science, Mie, Japan (H.S., K.O.); Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University, Tokushima, Japan (Y.I.); and Division of Nephrology, Endocrinology and Metabolism, Tokai University School of Medicine, Isehara, Japan (M.F.)
| | - Kazuya Ooi
- Division of Pharmacodynamics, Faculty of Pharmacy, Keio University, Tokyo, Japan (K.T., Y.E., K.M.); Department of Pharmacokinetics, Faculty of Pharmaceutical Sciences (K.T., M.O.) and DDS Research Institute (M.O.), Sojo University, Kumamoto, Japan; Department of Biopharmaceutics, Graduate School of Pharmaceutical Sciences (S.O., T.N., H.Y., K.N., H.M., H.W., T.M.) and Center for Clinical Pharmaceutical Sciences, Graduate School of Pharmaceutical Sciences (H.W., T.M.), Kumamoto University, Kumamoto, Japan; Laboratory of Clinical Pharmacology, Faculty of Pharmaceutical Sciences, Suzuka University of Medical Science, Mie, Japan (H.S., K.O.); Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University, Tokushima, Japan (Y.I.); and Division of Nephrology, Endocrinology and Metabolism, Tokai University School of Medicine, Isehara, Japan (M.F.)
| | - Yu Ishima
- Division of Pharmacodynamics, Faculty of Pharmacy, Keio University, Tokyo, Japan (K.T., Y.E., K.M.); Department of Pharmacokinetics, Faculty of Pharmaceutical Sciences (K.T., M.O.) and DDS Research Institute (M.O.), Sojo University, Kumamoto, Japan; Department of Biopharmaceutics, Graduate School of Pharmaceutical Sciences (S.O., T.N., H.Y., K.N., H.M., H.W., T.M.) and Center for Clinical Pharmaceutical Sciences, Graduate School of Pharmaceutical Sciences (H.W., T.M.), Kumamoto University, Kumamoto, Japan; Laboratory of Clinical Pharmacology, Faculty of Pharmaceutical Sciences, Suzuka University of Medical Science, Mie, Japan (H.S., K.O.); Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University, Tokushima, Japan (Y.I.); and Division of Nephrology, Endocrinology and Metabolism, Tokai University School of Medicine, Isehara, Japan (M.F.)
| | - Hiroshi Watanabe
- Division of Pharmacodynamics, Faculty of Pharmacy, Keio University, Tokyo, Japan (K.T., Y.E., K.M.); Department of Pharmacokinetics, Faculty of Pharmaceutical Sciences (K.T., M.O.) and DDS Research Institute (M.O.), Sojo University, Kumamoto, Japan; Department of Biopharmaceutics, Graduate School of Pharmaceutical Sciences (S.O., T.N., H.Y., K.N., H.M., H.W., T.M.) and Center for Clinical Pharmaceutical Sciences, Graduate School of Pharmaceutical Sciences (H.W., T.M.), Kumamoto University, Kumamoto, Japan; Laboratory of Clinical Pharmacology, Faculty of Pharmaceutical Sciences, Suzuka University of Medical Science, Mie, Japan (H.S., K.O.); Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University, Tokushima, Japan (Y.I.); and Division of Nephrology, Endocrinology and Metabolism, Tokai University School of Medicine, Isehara, Japan (M.F.)
| | - Masafumi Fukagawa
- Division of Pharmacodynamics, Faculty of Pharmacy, Keio University, Tokyo, Japan (K.T., Y.E., K.M.); Department of Pharmacokinetics, Faculty of Pharmaceutical Sciences (K.T., M.O.) and DDS Research Institute (M.O.), Sojo University, Kumamoto, Japan; Department of Biopharmaceutics, Graduate School of Pharmaceutical Sciences (S.O., T.N., H.Y., K.N., H.M., H.W., T.M.) and Center for Clinical Pharmaceutical Sciences, Graduate School of Pharmaceutical Sciences (H.W., T.M.), Kumamoto University, Kumamoto, Japan; Laboratory of Clinical Pharmacology, Faculty of Pharmaceutical Sciences, Suzuka University of Medical Science, Mie, Japan (H.S., K.O.); Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University, Tokushima, Japan (Y.I.); and Division of Nephrology, Endocrinology and Metabolism, Tokai University School of Medicine, Isehara, Japan (M.F.)
| | - Masaki Otagiri
- Division of Pharmacodynamics, Faculty of Pharmacy, Keio University, Tokyo, Japan (K.T., Y.E., K.M.); Department of Pharmacokinetics, Faculty of Pharmaceutical Sciences (K.T., M.O.) and DDS Research Institute (M.O.), Sojo University, Kumamoto, Japan; Department of Biopharmaceutics, Graduate School of Pharmaceutical Sciences (S.O., T.N., H.Y., K.N., H.M., H.W., T.M.) and Center for Clinical Pharmaceutical Sciences, Graduate School of Pharmaceutical Sciences (H.W., T.M.), Kumamoto University, Kumamoto, Japan; Laboratory of Clinical Pharmacology, Faculty of Pharmaceutical Sciences, Suzuka University of Medical Science, Mie, Japan (H.S., K.O.); Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University, Tokushima, Japan (Y.I.); and Division of Nephrology, Endocrinology and Metabolism, Tokai University School of Medicine, Isehara, Japan (M.F.)
| | - Toru Maruyama
- Division of Pharmacodynamics, Faculty of Pharmacy, Keio University, Tokyo, Japan (K.T., Y.E., K.M.); Department of Pharmacokinetics, Faculty of Pharmaceutical Sciences (K.T., M.O.) and DDS Research Institute (M.O.), Sojo University, Kumamoto, Japan; Department of Biopharmaceutics, Graduate School of Pharmaceutical Sciences (S.O., T.N., H.Y., K.N., H.M., H.W., T.M.) and Center for Clinical Pharmaceutical Sciences, Graduate School of Pharmaceutical Sciences (H.W., T.M.), Kumamoto University, Kumamoto, Japan; Laboratory of Clinical Pharmacology, Faculty of Pharmaceutical Sciences, Suzuka University of Medical Science, Mie, Japan (H.S., K.O.); Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University, Tokushima, Japan (Y.I.); and Division of Nephrology, Endocrinology and Metabolism, Tokai University School of Medicine, Isehara, Japan (M.F.)
| |
Collapse
|
23
|
Liu C, Fujino M, Zhu S, Isaka Y, Ito H, Takahashi K, Nakajima M, Tanaka T, Zhu P, Li X. 5-ALA/SFC enhances HO-1 expression through the MAPK/Nrf2 antioxidant pathway and attenuates murine tubular epithelial cell apoptosis. FEBS Open Bio 2019; 9:1928-1938. [PMID: 31495071 PMCID: PMC6823284 DOI: 10.1002/2211-5463.12729] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Revised: 08/16/2019] [Accepted: 09/03/2019] [Indexed: 12/15/2022] Open
Abstract
Cyclosporin A (CsA) is a common immunosuppressant, but its use is limited as it can cause chronic kidney injury. Oxidative stress and apoptosis play a key role in CsA-induced nephrotoxicity. This study investigated the protective effect of 5-aminolevulinic acid and iron (5-ALA/SFC) on CsA-induced injury in murine proximal tubular epithelial cells (mProx24). 5-ALA/SFC significantly inhibited apoptosis in CsA-treated mProx24 cells with increases in heme oxygenase (HO)-1, nuclear factor E2-related factor 2 (Nrf2), and p38, and Erk-1/2 phosphorylation. Moreover, 5-ALA/SFC suppressed production of reactive oxygen species in CsA-exposed cells and inhibition of HO-1 suppressed the protective effects of 5-ALA/SFC. In summary, 5-ALA/SFC may have potential for development into a treatment for the anti-nephrotoxic/apoptotic effects of CsA.
Collapse
Affiliation(s)
- Chi Liu
- Division of Transplantation ImmunologyNational Research Institute for Child Health and DevelopmentTokyoJapan
| | - Masayuki Fujino
- Division of Transplantation ImmunologyNational Research Institute for Child Health and DevelopmentTokyoJapan
- AIDS Research CenterNational Institute of Infectious DiseasesTokyoJapan
| | - Shuoji Zhu
- Guangdong Cardiovascular InstituteGuangdong Academy of Medical SciencesGuangdong Provincial People's HospitalGuangzhouChina
| | - Yoshitaka Isaka
- Department of NephrologyOsaka University Graduate School of MedicineJapan
| | | | | | | | | | - Ping Zhu
- Guangdong Cardiovascular InstituteGuangdong Academy of Medical SciencesGuangdong Provincial People's HospitalGuangzhouChina
| | - Xiao‐Kang Li
- Division of Transplantation ImmunologyNational Research Institute for Child Health and DevelopmentTokyoJapan
| |
Collapse
|
24
|
Kamiya A, Hara T, Tsuda M, Tsuru E, Kuroda Y, Ota U, Karashima T, Fukuhara H, Inoue K, Ishizuka M, Nakajima M, Tanaka T. 5-Aminolevulinic acid with ferrous iron improves early renal damage and hepatic steatosis in high fat diet-induced obese mice. J Clin Biochem Nutr 2018; 64:59-65. [PMID: 30705513 PMCID: PMC6348406 DOI: 10.3164/jcbn.18-35] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Accepted: 06/11/2018] [Indexed: 01/22/2023] Open
Abstract
5-Aminolevulinic acid, a natural amino acid, activates mitochondrial respiration and induces heme oxygenase-1 expression. Obesity and type 2 diabetes mellitus are associated with age-related mitochondrial respiration defect, oxidative stress and inflammation. The aim of this study is to investigate the effects of 5-aminolevulinic acid with sodium ferrous citrate on early renal damage and hepatic steatosis. 7-Month-old C57BL/6 mice were fed with a standard diet or high fat diet for 9 weeks, which were orally administered 300 mg/kg 5-aminolevulinic acid combined with 47 mg/kg sodium ferrous citrate (5-aminolevulinic acid/sodium ferrous citrate) or vehicle for the last 5 weeks. We observed that 5-aminolevulinic acid/sodium ferrous citrate significantly decreased body weight, fat weight, hepatic lipid deposits and improved levels of blood glucose and oral glucose tolerance test. In addition, 5-aminolevulinic acid/sodium ferrous citrate suppressed increased glomerular tuft area in high fat diet-fed mice, which was associated with increased heme oxygenase-1 protein expression. Our findings demonstrate additional evidence that 5-aminolevulinic acid/sodium ferrous citrate could improve glucose and lipid metabolism in diabetic mice. 5-Aminolevulinic acid/sodium ferrous citrate has potential application in obesity or type 2 diabetes mellitus-associated disease such as diabetic nephropathy and nonalcoholic fatty liver disease.
Collapse
Affiliation(s)
- Atsuko Kamiya
- SBI Pharmaceuticals Co. Ltd., 1-6-1 Roppongi, Minato-ku, Tokyo 106-6020, Japan
| | - Takeshi Hara
- SBI Pharmaceuticals Co. Ltd., 1-6-1 Roppongi, Minato-ku, Tokyo 106-6020, Japan
| | - Masayuki Tsuda
- Institute for Laboratory Animal Research, Kochi Medical School, Kohasu, Oko, Nankoku, Kochi 783-8505, Japan
| | - Emi Tsuru
- Institute for Laboratory Animal Research, Kochi Medical School, Kohasu, Oko, Nankoku, Kochi 783-8505, Japan
| | - Yasushi Kuroda
- SBI Pharmaceuticals Co. Ltd., 1-6-1 Roppongi, Minato-ku, Tokyo 106-6020, Japan
| | - Urara Ota
- SBI Pharmaceuticals Co. Ltd., 1-6-1 Roppongi, Minato-ku, Tokyo 106-6020, Japan
| | - Takashi Karashima
- Department of Urology, Kochi Medical School, Kohasu, Oko, Nankoku, Kochi 783-8505, Japan
| | - Hideo Fukuhara
- Department of Urology, Kochi Medical School, Kohasu, Oko, Nankoku, Kochi 783-8505, Japan
| | - Keiji Inoue
- Department of Urology, Kochi Medical School, Kohasu, Oko, Nankoku, Kochi 783-8505, Japan
| | - Masahiro Ishizuka
- SBI Pharmaceuticals Co. Ltd., 1-6-1 Roppongi, Minato-ku, Tokyo 106-6020, Japan
| | - Motowo Nakajima
- SBI Pharmaceuticals Co. Ltd., 1-6-1 Roppongi, Minato-ku, Tokyo 106-6020, Japan
| | - Tohru Tanaka
- SBI Pharmaceuticals Co. Ltd., 1-6-1 Roppongi, Minato-ku, Tokyo 106-6020, Japan
| |
Collapse
|