1
|
Saimoto Y, Kusakabe D, Morimoto K, Matsuoka Y, Kozakura E, Kato N, Tsunematsu K, Umeno T, Kiyotani T, Matsumoto S, Tsuji M, Hirayama T, Nagasawa H, Uchida K, Karasawa S, Jutanom M, Yamada KI. Lysosomal lipid peroxidation contributes to ferroptosis induction via lysosomal membrane permeabilization. Nat Commun 2025; 16:3554. [PMID: 40229298 PMCID: PMC11997074 DOI: 10.1038/s41467-025-58909-w] [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: 04/14/2024] [Accepted: 04/07/2025] [Indexed: 04/16/2025] Open
Abstract
Ferroptosis, a form of cell death instigated by iron-dependent lipid peroxidation reactions (LPO), is emerging as a promising therapeutic target for cancer. While the mechanisms governing LPO induction and suppression have gradually been unveiled, questions persist regarding the specific cellular location of LPO and the utilization of iron in driving cell death. A comprehensive understanding of these aspects holds significant potential for advancing therapeutic applications in disease management. Here, we show lysosomal LPO in the initiation of ferroptosis, leveraging the hidden abilities of fluorescent detection probes. Intra-lysosomal LPO triggers iron leakage, fostering cell-wide LPO by augmenting lysosomal membrane permeabilization (LMP). Conversely, cell lines with low susceptibility to ferroptosis do not exhibit LMP. This deficiency is rectified by the concurrent administration of chloroquine, leading to LMP induction and subsequent cell death. These findings underscore enhancing LMP induction efficacy as a strategic approach to surmount resistance to therapies in cancer.
Collapse
Affiliation(s)
- Yuma Saimoto
- Department of Molecular Pathobiology, Faculty of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Daiki Kusakabe
- Department of Molecular Pathobiology, Faculty of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Kazushi Morimoto
- Department of Molecular Pathobiology, Faculty of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Yuta Matsuoka
- Department of Molecular Pathobiology, Faculty of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
- Center for Cancer Immunotherapy and Immunobiology, Graduate School of Medicine, Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Eisho Kozakura
- Department of Molecular Pathobiology, Faculty of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Nao Kato
- Department of Molecular Pathobiology, Faculty of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Kayoko Tsunematsu
- Department of Molecular Pathobiology, Faculty of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Tomohiro Umeno
- Faculty of Pharmaceutical Sciences, Showa Pharmaceutical University, 3-3165 Higashi-Tamagawagakuen, Machida, 194-8543, Japan
| | - Tamiko Kiyotani
- Faculty of Pharmaceutical Sciences, Showa Pharmaceutical University, 3-3165 Higashi-Tamagawagakuen, Machida, 194-8543, Japan
| | - Shota Matsumoto
- Faculty of Pharmaceutical Sciences, Showa Pharmaceutical University, 3-3165 Higashi-Tamagawagakuen, Machida, 194-8543, Japan
| | - Mieko Tsuji
- Laboratory of Pharmaceutical and Medicinal Chemistry, Gifu Pharmaceutical University, 1-25-4 Daigaku-nishi, Gifu, 501-1196, Japan
| | - Tasuku Hirayama
- Laboratory of Pharmaceutical and Medicinal Chemistry, Gifu Pharmaceutical University, 1-25-4 Daigaku-nishi, Gifu, 501-1196, Japan
| | - Hideko Nagasawa
- Laboratory of Pharmaceutical and Medicinal Chemistry, Gifu Pharmaceutical University, 1-25-4 Daigaku-nishi, Gifu, 501-1196, Japan
| | - Koji Uchida
- Laboratory of Food Chemistry and Life Science, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1, Yayoi, Bunkyo-ku, Tokyo, 113-8657, Japan
| | - Satoru Karasawa
- Faculty of Pharmaceutical Sciences, Showa Pharmaceutical University, 3-3165 Higashi-Tamagawagakuen, Machida, 194-8543, Japan
| | - Mirinthorn Jutanom
- Department of Molecular Pathobiology, Faculty of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Ken-Ichi Yamada
- Department of Molecular Pathobiology, Faculty of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan.
| |
Collapse
|
2
|
Hu G, Li Y, Zang C, Yan H, Yang W, Hu Y. Development of a fluorescent probe based on the cyanine skeleton for the detection of PhSH. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2025; 330:125724. [PMID: 39805240 DOI: 10.1016/j.saa.2025.125724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 12/17/2024] [Accepted: 01/06/2025] [Indexed: 01/16/2025]
Abstract
In this study, a cyanine skeleton fluorescent parent core was designed based on the intramolecular charge transfer (ICT) principle, and 2, 4-dinitrofluorobenzene (DNFB) was used as the specific recognition site for phenylthiophene (PhSH). The probe showed a fluorescence transition from colorless to red under 410 nm excitation, which had the characteristics of fast response, high selectivity, low detection limit (55 nM), and the fluorescence intensity showed a positive linear correlation with PhSH concentration in the range of 0-100 μM (R2 = 0.9921). Furthermore, the recovery rate of PhSH detection in real water samples was 94 %-104 %, and the probe showed excellent fluorescence imaging effect in living cells.
Collapse
Affiliation(s)
- Guoxing Hu
- College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, No. 30, South Puzhu Road, Nanjing 211816, China; State Key Laboratory of Materials-Oriented Chemical Engineering, Nanjing Tech University, No. 30, South Puzhu Road, Nanjing 211816, China
| | - Yanni Li
- College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, No. 30, South Puzhu Road, Nanjing 211816, China; State Key Laboratory of Materials-Oriented Chemical Engineering, Nanjing Tech University, No. 30, South Puzhu Road, Nanjing 211816, China
| | - Chao Zang
- College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, No. 30, South Puzhu Road, Nanjing 211816, China; State Key Laboratory of Materials-Oriented Chemical Engineering, Nanjing Tech University, No. 30, South Puzhu Road, Nanjing 211816, China
| | - Haijun Yan
- Jiangsu Provincial Institute Of Materia Medica Co., Ltd., No. 30, South Puzhu Road, Nanjing 211816, China
| | - Wenge Yang
- School of Pharmaceutical Sciences, Nanjing Tech University, No. 30, South Puzhu Road, Nanjing 211816, China.
| | - Yonghong Hu
- College of Food Science and Light Industry, Nanjing Tech University, No. 30, South Puzhu Road, Nanjing 211816, China; State Key Laboratory of Materials-Oriented Chemical Engineering, Nanjing Tech University, No. 30, South Puzhu Road, Nanjing 211816, China.
| |
Collapse
|
3
|
Shiiba I, Ito N, Oshio H, Ishikawa Y, Nagao T, Shimura H, Oh KW, Takasaki E, Yamaguchi F, Konagaya R, Kadowaki H, Nishitoh H, Tanzawa T, Nagashima S, Sugiura A, Fujikawa Y, Umezawa K, Tamura Y, Il Lee B, Hirabayashi Y, Okazaki Y, Sawa T, Inatome R, Yanagi S. ER-mitochondria contacts mediate lipid radical transfer via RMDN3/PTPIP51 phosphorylation to reduce mitochondrial oxidative stress. Nat Commun 2025; 16:1508. [PMID: 39929810 PMCID: PMC11811300 DOI: 10.1038/s41467-025-56666-4] [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: 01/04/2024] [Accepted: 01/24/2025] [Indexed: 02/13/2025] Open
Abstract
The proximal domains of mitochondria and the endoplasmic reticulum (ER) are linked by tethering factors on each membrane, allowing the efficient transport of substances, including lipids and calcium, between them. However, little is known about the regulation and function of mitochondria-ER contacts (MERCs) dynamics under mitochondrial damage. In this study, we apply NanoBiT technology to develop the MERBiT system, which enables the measurement of reversible MERCs formation in living cells. Analysis using this system suggests that induction of mitochondrial ROS increases MERCs formation via RMDN3 (also known as PTPIP51)-VAPB tethering driven by RMDN3 phosphorylation. Disruption of this tethering caused lipid radical accumulation in mitochondria, leading to cell death. The lipid radical transfer activity of the TPR domain in RMDN3, as revealed by an in vitro liposome assay, suggests that RMDN3 transfers lipid radicals from mitochondria to the ER. Our findings suggest a potential role for MERCs in cell survival strategy by facilitating the removal of mitochondrial lipid radicals under mitochondrial damage.
Collapse
Grants
- 23H02691,20H04911,20H03454 MEXT | Japan Society for the Promotion of Science (JSPS)
- 22K15399, 22H05574, 24H01327 MEXT | Japan Society for the Promotion of Science (JSPS)
- 23K14185, 22K20637 MEXT | Japan Society for the Promotion of Science (JSPS)
- 22H05532 MEXT | Japan Society for the Promotion of Science (JSPS)
- 21H0207, 21H05267, 23K17979 MEXT | Japan Society for the Promotion of Science (JSPS)
- 21K06844 MEXT | Japan Society for the Promotion of Science (JSPS)
- JP17gm5010002, JP18gm5010002, JP19gm5010002, JP20gm5010002 Japan Agency for Medical Research and Development (AMED)
- JP19dm0207082 Japan Agency for Medical Research and Development (AMED)
- 23gm1610011h0001 Japan Agency for Medical Research and Development (AMED)
Collapse
Affiliation(s)
- Isshin Shiiba
- Laboratory of Molecular Biochemistry, Department of Life Science, Faculty of Science, Gakushuin University, Toshima, Tokyo, 171-8588, Japan.
| | - Naoki Ito
- Laboratory of Molecular Biochemistry, Department of Life Science, Faculty of Science, Gakushuin University, Toshima, Tokyo, 171-8588, Japan
| | - Hijiri Oshio
- Laboratory of Molecular Biochemistry, Department of Life Science, Faculty of Science, Gakushuin University, Toshima, Tokyo, 171-8588, Japan
| | - Yuto Ishikawa
- Laboratory of Molecular Biochemistry, Department of Life Science, Faculty of Science, Gakushuin University, Toshima, Tokyo, 171-8588, Japan
| | - Takahiro Nagao
- Department of Chemistry and Biotechnology, School of Engineering, The University of Tokyo, Tokyo, 113-8656, Japan
| | - Hiroki Shimura
- Laboratory of Regenerative Medicine, School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo, 192-0392, Japan
| | - Kyu-Wan Oh
- Research Institute, National Cancer Center, Goyang-si, Gyeonggi-do, Korea
| | - Eiki Takasaki
- Laboratory of Molecular Biochemistry, Department of Life Science, Faculty of Science, Gakushuin University, Toshima, Tokyo, 171-8588, Japan
| | - Fuya Yamaguchi
- Laboratory of Molecular Biochemistry, Department of Life Science, Faculty of Science, Gakushuin University, Toshima, Tokyo, 171-8588, Japan
| | - Ryoan Konagaya
- Laboratory of Molecular Biochemistry, Department of Life Science, Faculty of Science, Gakushuin University, Toshima, Tokyo, 171-8588, Japan
| | - Hisae Kadowaki
- Laboratory of Biochemistry and Molecular Biology, Faculty of Medicine, University of Miyazaki, 5200 Kihara, Kiyotake, Miyazaki, 889-1692, Japan
| | - Hideki Nishitoh
- Laboratory of Biochemistry and Molecular Biology, Faculty of Medicine, University of Miyazaki, 5200 Kihara, Kiyotake, Miyazaki, 889-1692, Japan
| | - Takehito Tanzawa
- Institute for Protein Research, Osaka University, Suita, Osaka, 565-0871, Japan
| | - Shun Nagashima
- Laboratory of Regenerative Medicine, School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo, 192-0392, Japan
| | - Ayumu Sugiura
- Diagnostics and Therapeutics of Intractable Diseases, Intractable Disease Research Center, Juntendo University, Graduate School of Medicine, Bunkyo-ku, Tokyo, Japan
| | - Yuuta Fujikawa
- School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji, Tokyo, 192-0392, Japan
| | - Keitaro Umezawa
- Research Team for Mechanism of Aging, Tokyo Metropolitan Institute of Gerontology, 35-2 Sakae-cho, Itabashi-ku, Tokyo, 173-0015, Japan
| | - Yasushi Tamura
- Faculty of Science, Yamagata University, 1-4-12 Kojirakawa-machi, Yamagata, Yamagata, 990-8560, Japan
| | - Byung Il Lee
- Research Institute, National Cancer Center, Goyang-si, Gyeonggi-do, Korea
| | - Yusuke Hirabayashi
- Department of Chemistry and Biotechnology, School of Engineering, The University of Tokyo, Tokyo, 113-8656, Japan
- Department of Bioengineering, School of Engineering, The University of Tokyo, Tokyo, 113-8656, Japan
| | - Yasushi Okazaki
- Diagnostics and Therapeutics of Intractable Diseases, Intractable Disease Research Center, Juntendo University, Graduate School of Medicine, Bunkyo-ku, Tokyo, Japan
| | - Tomohiro Sawa
- Department of Microbiology, Graduate School of Medical Sciences, Kumamoto University, 1-1-1 Honjo, Chuo-ku, Kumamoto, 860-8556, Japan
| | - Ryoko Inatome
- Laboratory of Molecular Biochemistry, Department of Life Science, Faculty of Science, Gakushuin University, Toshima, Tokyo, 171-8588, Japan
| | - Shigeru Yanagi
- Laboratory of Molecular Biochemistry, Department of Life Science, Faculty of Science, Gakushuin University, Toshima, Tokyo, 171-8588, Japan.
| |
Collapse
|
4
|
Doan VTH, Imai T, Kawazoe N, Chen G, Yoshitomi T. Regulation of antigen presentation and interleukin 10 production in murine dendritic cells via the oxidative stimulation of cell membrane using a polycation-porphyrin-conjugate-immobilized cell culture dish. Acta Biomater 2025; 193:231-241. [PMID: 39788307 DOI: 10.1016/j.actbio.2025.01.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 12/13/2024] [Accepted: 01/06/2025] [Indexed: 01/12/2025]
Abstract
Tolerogenic dendritic cells with professional antigen presentation via major histocompatibility complex molecules, co-stimulatory molecules (CD80/86), and interleukin 10 production have attracted significant attention as cellular therapies for autoimmune, allergic, and graft-versus-host diseases. In this study, we developed a cell culture dish equipped with polycation-porphyrin-conjugate-immobilized glass (PA-HP-G) to stimulate immature murine dendritic cell (iDCs). Upon irradiation with a red light at 635 nm toward the PA-HP-G surface, singlet oxygen was generated by the immobilized porphyrins on the PA-HP-G surface. When iDCs were cultured on the PA-HP-G surface, moderate light irradiation generated lipid radicals without excessive generation of reactive oxygen species in the cytoplasm and nucleus, which led to the oxidative stimulation of the iDC cell membrane without cell death. Light irradiation changed the morphology of dendritic cells on the PA-HP-G surface to a tree-like structure with dendrites, accelerated their maturation, and enhanced the antigen-presenting ability for the ovalbumin peptide via major histocompatibility complex class I molecules. Additionally, the antigen-presenting dendritic cells on the PA-HP-G surface produced the anti-inflammatory cytokine interleukin 10 upon light irradiation. These results indicated that upon moderate light irradiation, the PA-HP-G surface regulated the maturation of iDCs into tolerogenic dendritic cells. STATEMENT OF SIGNIFICANCE: • Cell culture dish is developed for selective oxidative stimulus of cell membrane. • 1O2 is generated from polycation/porphyrin-immobilized glass by light irradiation. • Lipid radicals are generated without generation of ROS in cytoplasm and nuclei. • Immature dendritic cells are maturated by oxidative stimulation of cell membrane. • Oxidative membrane stimulus enhances antigen-presentation and IL-10 secretion.
Collapse
Affiliation(s)
- Van Thi Hong Doan
- Research Center for Macromolecules and Biomaterials, National Institute for Materials Science, 1-1 Namiki, Tsukuba, Ibaraki 305-0044, Japan.
| | - Takashi Imai
- Department of Parasitology, National Institute of Infectious Diseases, 1-23-1 Toyama, Shinjuku-ku, Tokyo 162-8640 Japan; Antimicrobial Resistance Research Center, National Institute of Infectious Diseases, 4-2-1 Aobacho, Higashimurayama, Tokyo 189-0002, Japan; Leprosy Research Center, National Institute of Infectious Diseases, 4-2-1 Aobacho, Higashimurayama, Tokyo 189-0002, Japan
| | - Naoki Kawazoe
- Research Center for Macromolecules and Biomaterials, National Institute for Materials Science, 1-1 Namiki, Tsukuba, Ibaraki 305-0044, Japan
| | - Guoping Chen
- Research Center for Macromolecules and Biomaterials, National Institute for Materials Science, 1-1 Namiki, Tsukuba, Ibaraki 305-0044, Japan
| | - Toru Yoshitomi
- Research Center for Macromolecules and Biomaterials, National Institute for Materials Science, 1-1 Namiki, Tsukuba, Ibaraki 305-0044, Japan.
| |
Collapse
|
5
|
Hirata Y, Yamada Y, Taguchi S, Kojima R, Masumoto H, Kimura S, Niijima T, Toyama T, Kise R, Sato E, Uchida Y, Ito J, Nakagawa K, Taguchi T, Inoue A, Saito Y, Noguchi T, Matsuzawa A. Conjugated fatty acids drive ferroptosis through chaperone-mediated autophagic degradation of GPX4 by targeting mitochondria. Cell Death Dis 2024; 15:884. [PMID: 39643606 PMCID: PMC11624192 DOI: 10.1038/s41419-024-07237-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 11/04/2024] [Accepted: 11/08/2024] [Indexed: 12/09/2024]
Abstract
Conjugated fatty acids (CFAs) have been known for their anti-tumor activity. However, the mechanism of action remains unclear. Here, we identify CFAs as inducers of glutathione peroxidase 4 (GPX4) degradation through chaperone-mediated autophagy (CMA). CFAs, such as (10E,12Z)-octadecadienoic acid and α-eleostearic acid (ESA), induced GPX4 degradation, generation of mitochondrial reactive oxygen species (ROS) and lipid peroxides, and ultimately ferroptosis in cancer cell lines, including HT1080 and A549 cells, which were suppressed by either pharmacological blockade of CMA or genetic deletion of LAMP2A, a crucial molecule for CMA. Mitochondrial ROS were sufficient and necessary for CMA-dependent GPX4 degradation. Oral administration of an ESA-rich oil attenuated xenograft tumor growth of wild-type, but not that of LAMP2A-deficient HT1080 cells, accompanied by increased lipid peroxidation, GPX4 degradation and cell death. Our study establishes mitochondria as the key target of CFAs to trigger lipid peroxidation and GPX4 degradation, providing insight into ferroptosis-based cancer therapy.
Collapse
Affiliation(s)
- Yusuke Hirata
- Laboratory of Health Chemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan.
| | - Yuto Yamada
- Laboratory of Health Chemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Soma Taguchi
- Laboratory of Health Chemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Ryota Kojima
- Laboratory of Health Chemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Haruka Masumoto
- Laboratory of Health Chemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Shinnosuke Kimura
- Laboratory of Health Chemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Takuya Niijima
- Laboratory of Health Chemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Takashi Toyama
- Laboratory of Molecular Biology and Metabolism, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Ryoji Kise
- Laboratory of Molecular and Cellular Biochemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Emiko Sato
- Division of Clinical Pharmacology and Therapeutics, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Yasunori Uchida
- Laboratory of Organelle Pathophysiology, Department of Integrative Life Sciences, Graduate School of Life Sciences, Tohoku University, Sendai, Japan
| | - Junya Ito
- Laboratory of Food Function Analysis, Graduate School of Agricultural Sciences, Tohoku University, Sendai, Japan
| | - Kiyotaka Nakagawa
- Laboratory of Food Function Analysis, Graduate School of Agricultural Sciences, Tohoku University, Sendai, Japan
| | - Tomohiko Taguchi
- Laboratory of Organelle Pathophysiology, Department of Integrative Life Sciences, Graduate School of Life Sciences, Tohoku University, Sendai, Japan
| | - Asuka Inoue
- Laboratory of Molecular and Cellular Biochemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
- Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Japan
| | - Yoshiro Saito
- Laboratory of Molecular Biology and Metabolism, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Takuya Noguchi
- Laboratory of Health Chemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Atsushi Matsuzawa
- Laboratory of Health Chemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan.
| |
Collapse
|
6
|
Lerksaipheng P, Paiboonsukwong K, Sanvarinda P, Luechapudiporn R, Yamada KI, Morales NP. Lipid radicals and oxidized cholesteryl esters in low- and high-density lipoproteins in patients with β-thalassemia: Effects of iron overload and iron chelation therapy. Free Radic Biol Med 2024; 224:618-629. [PMID: 39303953 DOI: 10.1016/j.freeradbiomed.2024.09.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2024] [Revised: 08/31/2024] [Accepted: 09/17/2024] [Indexed: 09/22/2024]
Abstract
Iron overload results in lipid peroxidation (LPO) and the oxidative modification of circulating lipoproteins, which contributes to cardiovascular complications in patients with β-thalassemia. Investigating LPO may provide opportunities for the development of novel therapeutic strategies; however, the chemical pathways underlying iron overload-induced LPO in β-thalassemia lipoproteins remain unclear. In this study, we identified various species of lipid radicals (L•), the key mediators of LPO, and oxidized cholesteryl esters (oxCE) derived from the in vitro oxidation of major core lipids, cholesteryl linoleate (CE18:2) and cholesteryl arachidonate (CE20:4); the levels of these radical products in low-density lipoproteins (LDL) and high-density lipoproteins (HDL) were measured and compared between β-thalassemia patients and healthy subjects by using a specific fluorescent probe for L• with a liquid chromatography-tandem mass spectrometric method. Our results demonstrated that iron overload substantially decreased the levels of CE18:2 and CE20:4 substrates and α-tocopherol, resulting in higher levels of full-length and short-chain truncated L• and oxCE products. In particular, CE epoxyallyl radicals (•CE-O) were observed in the lipoproteins of β-thalassemia, revealing the pathological roles of iron overload in the progression of LPO. In addition, we found that intermission for two weeks of iron chelators can increase the production of these oxidized products; therefore, suggesting the beneficial effects of iron chelators in preventing LPO progression. In conclusion, our findings partly revealed the primary chemical pathway by which the LPO of circulating lipoproteins is influenced by iron overload and affected by iron chelation therapy. Moreover, we found that •CE + O shows potential as a sensitive biomarker for monitoring LPO in individuals with β-thalassemia.
Collapse
Affiliation(s)
- Pakawit Lerksaipheng
- Department of Pharmacology, Faculty of Science, Mahidol University, Bangkok, Thailand.
| | - Kittiphong Paiboonsukwong
- Thalassemia Research Center, Institute of Molecular Biosciences, Mahidol University, Salaya Campus, Nakhon Pathom, Thailand.
| | - Pimtip Sanvarinda
- Department of Pharmacology, Faculty of Science, Mahidol University, Bangkok, Thailand.
| | - Rataya Luechapudiporn
- Department of Pharmacology and Physiology, Faculty of Pharmaceutical Science, Chulalongkorn University, Bangkok, Thailand.
| | - Ken-Ichi Yamada
- Department of Molecular Pathobiology, Graduate School of Pharmaceutical Science, Kyushu University, Fukuoka, 812-8582, Japan.
| | | |
Collapse
|
7
|
Takajo T, Saito K, Tsuchida K, Kato S, Nakagawa K, Okino A, Anzai K. Mechanism of lipid peroxidation of liposomes by cold atmospheric pressure plasma jet irradiation. J Clin Biochem Nutr 2024; 75:183-189. [PMID: 39583979 PMCID: PMC11579849 DOI: 10.3164/jcbn.24-72] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Accepted: 07/26/2024] [Indexed: 11/26/2024] Open
Abstract
Liposome lipid peroxidation induced by cold atmospheric pressure plasma jet (CAPPJ) irradiation was investigated. The formation of thiobarbituric acid reactive substances (TBARS), an indicator of lipid peroxidation final products, as a function of irradiation was observed. Lipid radicals, peroxidation reaction intermediates generated by CAPPJ irradiation, were confirmed by increased NBD-pen fluorescence intensity. Additionally, lipid peroxidation products, liposomal phosphatidylcholine (PC) isomers, were analyzed by LC-MS/MS. Products specific to singlet oxygen (1O2) oxidation, 16:0/10-hydroperoxy-8E,12Z-octadecanoic acid (10-8E,12Z-HpODE) PC and 16:0/12-9E,13E-HpODE PC, were not detected, but radical oxidation specific products 16:0/13-9E,11E-HpODE PC and 16:0/9-10E,12E-HpODE PC were. This suggests that during CAPPJ irradiation, radicals, rather than 1O2, are the primary reactive species of lipid peroxidation. This is also supported by the β-carotene quenching of 1O2 not suppressing TBARS and lipid radical generation. Also, neither TBARS formation nor lipid radical generation were suppressed by SOD, indicating that the superoxide radical (O2 •-) is not responsible for the lipid peroxidation reaction. As the CAPPJ irradiation of water produces large quantities of hydroxyl radical (•OH) and •OH scavengers decreased the amount of TBARS produced by CAPPJ irradiation, it is highly plausible that •OH is the primary species involved in CAPPJ-induced liposome lipid peroxidation.
Collapse
Affiliation(s)
- Tokuko Takajo
- Faculty of Pharmaceutical Sciences, Nihon Pharmaceutical University, 10281 Komuro, Ina-machi, Kitaadachi-gun, Saitama 362-0806, Japan
| | - Koichi Saito
- Faculty of Pharmaceutical Sciences, Nihon Pharmaceutical University, 10281 Komuro, Ina-machi, Kitaadachi-gun, Saitama 362-0806, Japan
| | - Kazunori Tsuchida
- Faculty of Pharmaceutical Sciences, Nihon Pharmaceutical University, 10281 Komuro, Ina-machi, Kitaadachi-gun, Saitama 362-0806, Japan
| | - Shunji Kato
- Graduate School of Agricultural Science, Tohoku University, Sendai, Miyagi 980-0845, Japan
| | - Kiyotaka Nakagawa
- Graduate School of Agricultural Science, Tohoku University, Sendai, Miyagi 980-0845, Japan
| | - Akitoshi Okino
- Laboratory for Future Interdisciplinary Research of Science and Technology, Institute of Innovative Research, Tokyo Institute of Technology, 4259 Nagatsuda, Midori-ku, Yokohama 226-8502, Japan
| | - Kazunori Anzai
- Faculty of Pharmaceutical Sciences, Nihon Pharmaceutical University, 10281 Komuro, Ina-machi, Kitaadachi-gun, Saitama 362-0806, Japan
| |
Collapse
|
8
|
Mori R, Abe M, Saimoto Y, Shinto S, Jodai S, Tomomatsu M, Tazoe K, Ishida M, Enoki M, Kato N, Yamashita T, Itabashi Y, Nakanishi I, Ohkubo K, Kaidzu S, Tanito M, Matsuoka Y, Morimoto K, Yamada KI. Construction of a screening system for lipid-derived radical inhibitors and validation of hit compounds to target retinal and cerebrovascular diseases. Redox Biol 2024; 73:103186. [PMID: 38744193 PMCID: PMC11109892 DOI: 10.1016/j.redox.2024.103186] [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: 04/16/2024] [Accepted: 05/07/2024] [Indexed: 05/16/2024] Open
Abstract
Recent studies have highlighted the indispensable role of oxidized lipids in inflammatory responses, cell death, and disease pathogenesis. Consequently, inhibitors targeting oxidized lipids, particularly lipid-derived radicals critical in lipid peroxidation, which are known as radical-trapping antioxidants (RTAs), have been actively pursued. We focused our investigation on nitroxide compounds that have rapid second-order reaction rate constants for reaction with lipid-derived radicals. A novel screening system was developed by employing competitive reactions between library compounds and a newly developed profluorescence nitroxide probe with lipid-derived radicals to identify RTA compounds. A PubMed search of the top hit compounds revealed their wide application as repositioned drugs. Notably, the inhibitory efficacy of methyldopa, selected from these compounds, against retinal damage and bilateral common carotid artery stenosis was confirmed in animal models. These findings underscore the efficacy of our screening system and suggest that it is an effective approach for the discovery of RTA compounds.
Collapse
Affiliation(s)
- Ryota Mori
- Department of Molecular Pathobiology, Faculty of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi Higashi-ku, Fukuoka, 812-8582, Japan
| | - Masami Abe
- Department of Molecular Pathobiology, Faculty of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi Higashi-ku, Fukuoka, 812-8582, Japan
| | - Yuma Saimoto
- Department of Molecular Pathobiology, Faculty of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi Higashi-ku, Fukuoka, 812-8582, Japan
| | - Saki Shinto
- Department of Molecular Pathobiology, Faculty of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi Higashi-ku, Fukuoka, 812-8582, Japan
| | - Sara Jodai
- Department of Molecular Pathobiology, Faculty of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi Higashi-ku, Fukuoka, 812-8582, Japan
| | - Manami Tomomatsu
- Department of Molecular Pathobiology, Faculty of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi Higashi-ku, Fukuoka, 812-8582, Japan
| | - Kaho Tazoe
- Department of Molecular Pathobiology, Faculty of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi Higashi-ku, Fukuoka, 812-8582, Japan
| | - Minato Ishida
- Department of Molecular Pathobiology, Faculty of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi Higashi-ku, Fukuoka, 812-8582, Japan
| | - Masataka Enoki
- Department of Molecular Pathobiology, Faculty of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi Higashi-ku, Fukuoka, 812-8582, Japan
| | - Nao Kato
- Department of Molecular Pathobiology, Faculty of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi Higashi-ku, Fukuoka, 812-8582, Japan
| | - Tomohiro Yamashita
- Department of Drug Discovery Structural Biology, Faculty of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi Higashi-ku, Fukuoka, 812-8582, Japan
| | - Yuki Itabashi
- Institute for Open and Transdisciplinary Research Initiatives, Osaka University, 1-6 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Ikuo Nakanishi
- Quantum RedOx Chemistry Team, Institute for Quantum Life Science (iQLS), Quantum Life and Medical Science Directorate (QLMS), National Institutes for Quantum Science and Technology (QST), 4-9-1 Anagawa, Inage-ku, Chiba, 263-8555, Japan
| | - Kei Ohkubo
- Institute for Open and Transdisciplinary Research Initiatives, Osaka University, 1-6 Yamadaoka, Suita, Osaka, 565-0871, Japan; Quantum RedOx Chemistry Team, Institute for Quantum Life Science (iQLS), Quantum Life and Medical Science Directorate (QLMS), National Institutes for Quantum Science and Technology (QST), 4-9-1 Anagawa, Inage-ku, Chiba, 263-8555, Japan; Institute for Advanced Co-Creation Studies, Osaka University, 1-6 Yamada-oka, Suita, Osaka, 565-0871, Japan
| | - Sachiko Kaidzu
- Department of Ophthalmology, Shimane University Faculty of Medicine, 89-1 Enya Izumo, Shimane, 693-8501, Japan
| | - Masaki Tanito
- Department of Ophthalmology, Shimane University Faculty of Medicine, 89-1 Enya Izumo, Shimane, 693-8501, Japan
| | - Yuta Matsuoka
- Department of Molecular Pathobiology, Faculty of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi Higashi-ku, Fukuoka, 812-8582, Japan
| | - Kazushi Morimoto
- Department of Molecular Pathobiology, Faculty of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi Higashi-ku, Fukuoka, 812-8582, Japan
| | - Ken-Ichi Yamada
- Department of Molecular Pathobiology, Faculty of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi Higashi-ku, Fukuoka, 812-8582, Japan.
| |
Collapse
|
9
|
Nguyen NT, Sennoune SR, Dharmalingam-Nandagopal G, Sivaprakasam S, Bhutia YD, Ganapathy V. Impact of Oncogenic Changes in p53 and KRAS on Macropinocytosis and Ferroptosis in Colon Cancer Cells and Anticancer Efficacy of Niclosamide with Differential Effects on These Two Processes. Cells 2024; 13:951. [PMID: 38891084 PMCID: PMC11171492 DOI: 10.3390/cells13110951] [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: 03/31/2024] [Revised: 05/25/2024] [Accepted: 05/29/2024] [Indexed: 06/21/2024] Open
Abstract
Mutations in p53 and KRAS are seen in most cases of colon cancer. The impact of these mutations on signaling pathways related to cancer growth has been studied in depth, but relatively less is known on their effects on amino acid transporters in cancer cells. This represents a significant knowledge gap because amino acid nutrition in cancer cells profoundly influences macropinocytosis and ferroptosis, two processes with opposing effects on tumor growth. Here, we used isogenic colon cancer cell lines to investigate the effects of p53 deletion and KRAS activation on two amino acid transporters relevant to macropinocytosis (SLC38A5) and ferroptosis (SLC7A11). Our studies show that the predominant effect of p53 deletion is to induce SLC7A11 with the resultant potentiation of antioxidant machinery and protection of cancer cells from ferroptosis, whereas KRAS activation induces not only SLC7A11 but also SLC38A5, thus offering protection from ferroptosis as well as improving amino acid nutrition in cancer cells via accelerated macropinocytosis. Niclosamide, an FDA-approved anti-helminthic, blocks the functions of SLC7A11 and SLC38A5, thus inducing ferroptosis and suppressing macropinocytosis, with the resultant effective reversal of tumor-promoting actions of oncogenic changes in p53 and KRAS. These findings underscore the potential of this drug in colon cancer treatment.
Collapse
Affiliation(s)
| | | | | | | | | | - Vadivel Ganapathy
- Department of Cell Biology and Biochemistry, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; (N.T.N.); (S.R.S.); (G.D.-N.); (S.S.); (Y.D.B.)
| |
Collapse
|
10
|
Yamada N, Karasawa T, Ito J, Yamamuro D, Morimoto K, Nakamura T, Komada T, Baatarjav C, Saimoto Y, Jinnouchi Y, Watanabe K, Miura K, Yahagi N, Nakagawa K, Matsumura T, Yamada KI, Ishibashi S, Sata N, Conrad M, Takahashi M. Inhibition of 7-dehydrocholesterol reductase prevents hepatic ferroptosis under an active state of sterol synthesis. Nat Commun 2024; 15:2195. [PMID: 38472233 DOI: 10.1038/s41467-024-46386-6] [Citation(s) in RCA: 21] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Accepted: 02/23/2024] [Indexed: 03/14/2024] Open
Abstract
Recent evidence indicates ferroptosis is implicated in the pathophysiology of various liver diseases; however, the organ-specific regulation mechanism is poorly understood. Here, we demonstrate 7-dehydrocholesterol reductase (DHCR7), the terminal enzyme of cholesterol biosynthesis, as a regulator of ferroptosis in hepatocytes. Genetic and pharmacological inhibition (with AY9944) of DHCR7 suppress ferroptosis in human hepatocellular carcinoma Huh-7 cells. DHCR7 inhibition increases its substrate, 7-dehydrocholesterol (7-DHC). Furthermore, exogenous 7-DHC supplementation using hydroxypropyl β-cyclodextrin suppresses ferroptosis. A 7-DHC-derived oxysterol metabolite, 3β,5α-dihydroxycholest-7-en-6-one (DHCEO), is increased by the ferroptosis-inducer RSL-3 in DHCR7-deficient cells, suggesting that the ferroptosis-suppressive effect of DHCR7 inhibition is associated with the oxidation of 7-DHC. Electron spin resonance analysis reveals that 7-DHC functions as a radical trapping agent, thus protecting cells from ferroptosis. We further show that AY9944 inhibits hepatic ischemia-reperfusion injury, and genetic ablation of Dhcr7 prevents acetaminophen-induced acute liver failure in mice. These findings provide new insights into the regulatory mechanism of liver ferroptosis and suggest a potential therapeutic option for ferroptosis-related liver diseases.
Collapse
Affiliation(s)
- Naoya Yamada
- Division of Inflammation Research, Center for Molecular Medicine, Jichi Medical University, Shimotsuke, Tochigi, Japan.
- Division of Gastroenterological, General and Transplant Surgery, Department of Surgery, Jichi Medical University, Shimotsuke, Tochigi, Japan.
- Institute of Metabolism and Cell Death, Molecular Target and Therapeutics Center, Helmholtz Munich, Neuherberg, Bavaria, Germany.
| | - Tadayoshi Karasawa
- Division of Inflammation Research, Center for Molecular Medicine, Jichi Medical University, Shimotsuke, Tochigi, Japan.
| | - Junya Ito
- Laboratory of Food Function Analysis, Graduate School of Agricultural Science, Tohoku University, Sendai, Miyagi, Japan
| | - Daisuke Yamamuro
- Division of Endocrinology and Metabolism, Department of Medicine, Jichi Medical University, Shimotsuke, Tochigi, Japan
| | - Kazushi Morimoto
- Department of Molecular Pathobiology, Faculty of Pharmaceutical Sciences, Kyushu University, Fukuoka, Fukuoka, Japan
| | - Toshitaka Nakamura
- Institute of Metabolism and Cell Death, Molecular Target and Therapeutics Center, Helmholtz Munich, Neuherberg, Bavaria, Germany
| | - Takanori Komada
- Division of Inflammation Research, Center for Molecular Medicine, Jichi Medical University, Shimotsuke, Tochigi, Japan
| | - Chintogtokh Baatarjav
- Division of Inflammation Research, Center for Molecular Medicine, Jichi Medical University, Shimotsuke, Tochigi, Japan
| | - Yuma Saimoto
- Department of Molecular Pathobiology, Faculty of Pharmaceutical Sciences, Kyushu University, Fukuoka, Fukuoka, Japan
| | - Yuka Jinnouchi
- Department of Molecular Pathobiology, Faculty of Pharmaceutical Sciences, Kyushu University, Fukuoka, Fukuoka, Japan
| | - Kazuhisa Watanabe
- Division of Human Genetics, Center for Molecular Medicine, Jichi Medical University, Shimotsuke, Tochigi, Japan
| | - Kouichi Miura
- Division of Gastroenterology, Department of Medicine, Jichi Medical University, Shimotsuke, Tochigi, Japan
| | - Naoya Yahagi
- Division of Endocrinology and Metabolism, Department of Medicine, Jichi Medical University, Shimotsuke, Tochigi, Japan
| | - Kiyotaka Nakagawa
- Laboratory of Food Function Analysis, Graduate School of Agricultural Science, Tohoku University, Sendai, Miyagi, Japan
| | - Takayoshi Matsumura
- Division of Inflammation Research, Center for Molecular Medicine, Jichi Medical University, Shimotsuke, Tochigi, Japan
- Division of Human Genetics, Center for Molecular Medicine, Jichi Medical University, Shimotsuke, Tochigi, Japan
| | - Ken-Ichi Yamada
- Department of Molecular Pathobiology, Faculty of Pharmaceutical Sciences, Kyushu University, Fukuoka, Fukuoka, Japan
| | - Shun Ishibashi
- Division of Endocrinology and Metabolism, Department of Medicine, Jichi Medical University, Shimotsuke, Tochigi, Japan
| | - Naohiro Sata
- Division of Gastroenterological, General and Transplant Surgery, Department of Surgery, Jichi Medical University, Shimotsuke, Tochigi, Japan
| | - Marcus Conrad
- Institute of Metabolism and Cell Death, Molecular Target and Therapeutics Center, Helmholtz Munich, Neuherberg, Bavaria, Germany
| | - Masafumi Takahashi
- Division of Inflammation Research, Center for Molecular Medicine, Jichi Medical University, Shimotsuke, Tochigi, Japan.
| |
Collapse
|
11
|
Tsuruta K, Matsuoka M, Harada S, Enomoto A, Kumagai T, Yasuda S, Koumura T, Yamada KI, Imai H. Slowly progressive cell death induced by GPx4-deficiency occurs via MEK1/ERK2 activation as a downstream signal after iron-independent lipid peroxidation. J Clin Biochem Nutr 2024; 74:97-107. [PMID: 38510679 PMCID: PMC10948347 DOI: 10.3164/jcbn.23-101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2023] [Accepted: 10/28/2023] [Indexed: 03/22/2024] Open
Abstract
Glutathione peroxidase 4 (GPx4) is an antioxidant enzyme that reduces phospholipid hydroperoxide. Studies have reported that the loss of GPx4 activity through anticancer drugs leads to ferroptosis, an iron-dependent lipid peroxidation-induced cell death. In this study, we established Tamoxifen-inducible GPx4-deficient Mouse embryonic fibroblast (MEF) cells (ETK1 cells) and found that Tamoxifen-inducible gene disruption of GPx4 induces slow cell death at ~72 h. In contrast, RSL3- or erastin-induced ferroptosis occurred quickly within 24 h. Therefore, we investigated the differences in these mechanisms between GPx4 gene disruption-induced cell death and RSL3- or erastin-induced ferroptosis. We found that GPx4-deficiency induced lipid peroxidation at 24 h in Tamoxifen-treated ETK1 cells, which was not suppressed by iron chelators, although lipid peroxidation in RSL3- or erastin-treated cells induced ferroptosis that was inhibited by iron chelators. We revealed that GPx4-deficient cell death was MEK1-dependent but RSL3- or erastin-induced ferroptosis was not, although MEK1/2 inhibitors suppressed both GPx4-deficient cell death and RSL3- or erastin-induced ferroptosis. In GPx4-deficient cell death, the phosphorylation of MEK1/2 and ERK2 was observed 39 h after lipid peroxidation, but ERK1 was not phosphorylated. Selective inhibitors of ERK2 inhibited GPx4-deficient cell death but not in RSL3- or erastin-induced cell death. These findings suggest that iron-independent lipid peroxidation due to GPx4 disruption induced cell death via the activation of MEK1/ERK2 as a downstream signal of lipid peroxidation in Tamoxifen-treated ETK1 cells. This indicates that GPx4 gene disruption induces slow cell death and involves a different pathway from RSL3- and erastin-induced ferroptosis in ETK1 cells.
Collapse
Affiliation(s)
- Kahori Tsuruta
- Department of Hygienic Chemistry, School of Pharmaceutical Sciences, Kitasato University, 5-9-1 Shirokane, Minato-ku, Tokyo 108-8641, Japan
- Laboratory of Microbiology, School of Pharmaceutical Sciences, Kitasato University, 5-9-1 Shirokane, Minato-ku, Tokyo 108-8641, Japan
| | - Masaki Matsuoka
- Department of Hygienic Chemistry, School of Pharmaceutical Sciences, Kitasato University, 5-9-1 Shirokane, Minato-ku, Tokyo 108-8641, Japan
| | - Shinsaku Harada
- Department of Hygienic Chemistry, School of Pharmaceutical Sciences, Kitasato University, 5-9-1 Shirokane, Minato-ku, Tokyo 108-8641, Japan
| | - Ayaka Enomoto
- Department of Hygienic Chemistry, School of Pharmaceutical Sciences, Kitasato University, 5-9-1 Shirokane, Minato-ku, Tokyo 108-8641, Japan
| | - Takeshi Kumagai
- Laboratory of Clinical Pharmacy Research, School of Pharmaceutical Sciences, Kitasato University, 5-9-1 Shirokane, Minato-ku, Tokyo 108-8641, Japan
| | - Shu Yasuda
- Department of Hygienic Chemistry, School of Pharmaceutical Sciences, Kitasato University, 5-9-1 Shirokane, Minato-ku, Tokyo 108-8641, Japan
| | - Tomoko Koumura
- Department of Hygienic Chemistry, School of Pharmaceutical Sciences, Kitasato University, 5-9-1 Shirokane, Minato-ku, Tokyo 108-8641, Japan
| | - Ken-ichi Yamada
- Department of Molecular Pathobiology, Faculty of Pharmaceutical Sciences, Kyushu University, 3-1-1 Made, Higashi-ku, Fukuoka-shi, Fukuoka 812-8582, Japan
| | - Hirotaka Imai
- Department of Hygienic Chemistry, School of Pharmaceutical Sciences, Kitasato University, 5-9-1 Shirokane, Minato-ku, Tokyo 108-8641, Japan
- Medical Research Laboratories, School of Pharmaceutical Sciences, Kitasato University, 5-9-1 Shirokane, Minato-ku, Tokyo 108-8641, Japan
| |
Collapse
|
12
|
Liu J, Zheng Z, Wang N, Li G. Plasma-Droplet Fusion-Mass Spectrometry Reveals Sub-Millisecond Protein Unfolding Dynamics Induced by Reactive Oxygen Species. Anal Chem 2024; 96:2292-2296. [PMID: 38295309 DOI: 10.1021/acs.analchem.3c03419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2024]
Abstract
Investigating the connection between reactive oxygen species (ROS) and oxidative protein unfolding is critical to reveal the mechanisms underlying disease involving elevated ROS and protein misfolding. This could inform the development of therapeutics targeting cells based on their redox status. In this study, we developed a plasma-droplet fusion-mass spectrometry platform to rapidly assess protein resilience to ROS. This home-built system fuses ROS generated from the microplasma source with protein microdroplets from a tunable nanospray source. At the droplet-plasma intersection, ROS interact with proteins before entering the mass spectrometer for mass identification and structural characterization. Benefiting from the small-sized microdroplet with adjustable traveling velocity, the platform enables the first sub-millisecond kinetic study of ROS-induced protein unfolding, with a rate constant of approximately 1.81 ms-1. Capturing ROS-induced protein unfolding intermediates and the resultant ligand release dynamics can be extended to many more protein systems. We foresee broad applications for establishing previously undetected protein unfolding events when biologically impactful ROS are enriched in time and space with functional proteins and complexes.
Collapse
Affiliation(s)
- Jun Liu
- Tianjin Key Laboratory of Biosensing and Molecular Recognition, Research Center for Analytical Science, Frontiers Science Center for New Organic Matter, College of Chemistry, Nankai University, Tianjin 300071, China
| | - Zhen Zheng
- School of Pharmacy, Tianjin Medical University, Tianjin 300070, China
| | - Ning Wang
- Tianjin Key Laboratory of Biosensing and Molecular Recognition, Research Center for Analytical Science, Frontiers Science Center for New Organic Matter, College of Chemistry, Nankai University, Tianjin 300071, China
| | - Gongyu Li
- Tianjin Key Laboratory of Biosensing and Molecular Recognition, Research Center for Analytical Science, Frontiers Science Center for New Organic Matter, College of Chemistry, Nankai University, Tianjin 300071, China
| |
Collapse
|
13
|
Wu C, Zhang W, Yan F, Dai W, Fang F, Gao Y, Cui W. Amelioration effects of the soybean lecithin-gallic acid complex on iron-overload-induced oxidative stress and liver damage in C57BL/6J mice. PHARMACEUTICAL BIOLOGY 2023; 61:37-49. [PMID: 36573499 PMCID: PMC9809354 DOI: 10.1080/13880209.2022.2151632] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 09/30/2022] [Accepted: 11/19/2022] [Indexed: 06/17/2023]
Abstract
CONTEXT Gallic acid (GA) and lecithin showed important roles in antioxidant and drug delivery, respectively. A complex synthesized from GA and soybean lecithin (SL-GAC), significantly improved bioavailability of GA and pharmacological activities. However, the antioxidant activity of SL-GAC and its effect on iron-overload-induced liver injury remains unexplored. OBJECTIVE This study investigates the antioxidant properties of SL-GAC in vitro and in mice, and its remediating effects against liver injury by iron-overloaded. MATERIALS AND METHODS In vitro, free radical scavenging activity, lipid peroxidation inhibition, and ferric reducing power of SL-GAC were measured by absorbance photometry. In vivo, C57BL/6J mice were randomized into 4 groups: control, iron-overloaded, iron-overloaded + deferoxamine, and iron-overloaded + SL-GAC. Treatments with deferoxamine (150 mg/kg/intraperitioneally) and SL-GAC (200 mg/kg/orally) were given to the desired groups for 12 weeks, daily. Iron levels, oxidative stress, and biochemical parameters were determined by histopathological examination and molecular biological techniques. RESULTS In vitro, SL-GAC showed DPPH and ABTS free radicals scavenging activity with IC50 values equal to 24.92 and 128.36 μg/mL, respectively. In C57BL/6J mice, SL-GAC significantly reduced the levels of serum iron (22.82%), liver iron (50.29%), aspartate transaminase (25.97%), alanine transaminase (38.07%), gamma glutamyl transferase (42.11%), malondialdehyde (19.82%), total cholesterol (45.96%), triglyceride (34.90%), ferritin light chain (18.51%) and transferrin receptor (27.39%), while up-regulated the levels of superoxide dismutase (24.69%), and glutathione (11.91%). CONCLUSIONS These findings encourage the use of SL-GAC to treat liver injury induced by iron-overloaded. Further in vivo and in vitro studies are needed to validate its potential in clinical medicine.
Collapse
Affiliation(s)
- Caihong Wu
- Department of Nutrition and Food Hygiene, School of Public Health, Jilin University, Changchun, China
| | - Wenxin Zhang
- Department of Pathogenobiology, Jilin University Mycology Research Center, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Feifei Yan
- Department of Nutrition and Food Hygiene, School of Public Health, Jilin University, Changchun, China
| | - Wenwen Dai
- Department of Nutrition and Food Hygiene, School of Public Health, Jilin University, Changchun, China
| | - Fang Fang
- Department of Nutrition and Food Hygiene, School of Public Health, Jilin University, Changchun, China
| | - Yanli Gao
- Department of Pediatric Ultrosonic, The First Hospital of Jilin University, Changchun, China
| | - Weiwei Cui
- Department of Nutrition and Food Hygiene, School of Public Health, Jilin University, Changchun, China
| |
Collapse
|
14
|
Shi J, Xu W, Yu H, Wang X, Jin F, Zhang Q, Zhang H, Peng Q, Abdurahman A, Wang M. A Highly Luminescent Metallo-Supramolecular Radical Cage. J Am Chem Soc 2023; 145:24081-24088. [PMID: 37796113 DOI: 10.1021/jacs.3c07477] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/06/2023]
Abstract
Luminescent metal-radicals have recently received increasing attention due to their unique properties and promising applications in materials science. However, the luminescence of metal-radicals tends to be quenched after formation of metallo-complexes. It is challenging to construct metal-radicals with highly luminescent properties. Herein, we report a highly luminescent metallo-supramolecular radical cage (LMRC) constructed by the assembly of a tritopic terpyridinyl ligand RL with tris(2,4,6-trichlorophenyl)methyl (TTM) radical and Zn2+. Electrospray ionization-mass spectrometry (ESI-MS), traveling-wave ion mobility-mass spectrometry (TWIM-MS), X-ray crystallography, electron paramagnetic resonance (EPR) spectroscopy, and superconducting quantum interference device (SQUID) confirm the formation of a prism-like supramolecular radical cage. LMRC exhibits a remarkable photoluminescence quantum yield (PLQY) of 65%, which is 5 times that of RL; meanwhile, LMRC also shows high photostability. Notably, significant magnetoluminescence can be observed for the high-concentration LMRC (15 wt % doped in PMMA film); however, the magnetoluminescence of 0.1 wt % doped LMRC film vanishes, revealing negligible spin-spin interactions between two radical centers in LMRC.
Collapse
Affiliation(s)
- Junjuan Shi
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun, Jilin 130012, China
| | - Wei Xu
- State Key Laboratory of Inorganic Synthesis and Preparative Chemistry, College of Chemistry, Jilin University, Changchun, Jilin 130012, China
| | - Hao Yu
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun, Jilin 130012, China
| | - Xing Wang
- Key Laboratory of Flexible Electronics (KLOFE), Institute of Advanced Materials (IAM) & School of Flexible Electronics (Future Technologies), Nanjing Tech University (Nanjing Tech), Nanjing, Jiangsu 211816, China
| | - Feng Jin
- Beijing National Laboratory for Condensed Matter Physics, Institute of Physics, Chinese Academy of Sciences, Beijing 100190, China
| | - Qingming Zhang
- Beijing National Laboratory for Condensed Matter Physics, Institute of Physics, Chinese Academy of Sciences, Beijing 100190, China
| | - Houyu Zhang
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun, Jilin 130012, China
| | - Qiming Peng
- Key Laboratory of Flexible Electronics (KLOFE), Institute of Advanced Materials (IAM) & School of Flexible Electronics (Future Technologies), Nanjing Tech University (Nanjing Tech), Nanjing, Jiangsu 211816, China
| | - Alim Abdurahman
- State Key Laboratory of Integrated Optoelectronics, College of Electronic Science and Engineering, Changchun, Jilin 130012, China
| | - Ming Wang
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun, Jilin 130012, China
| |
Collapse
|
15
|
Doan VTH, Komatsu Y, Matsui H, Kawazoe N, Chen G, Yoshitomi T. Singlet oxygen-generating cell-adhesive glass surfaces for the fundamental investigation of plasma membrane-targeted photodynamic therapy. Free Radic Biol Med 2023; 207:239-246. [PMID: 37499887 DOI: 10.1016/j.freeradbiomed.2023.07.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2023] [Revised: 07/23/2023] [Accepted: 07/24/2023] [Indexed: 07/29/2023]
Abstract
Recently, plasma membrane-targeted photodynamic therapy has attracted attention as an effective cancer immunotherapeutic strategy. However, the released photosensitizers do not only adhere to the plasma membrane but may also be internalized in the cytosol, in endosomes/lysosomes, hindering investigations of the effects of photosensitizers attached to the plasma membrane. In this study, we developed a cell culture dish with singlet oxygen-generating cell-adhesive glass surfaces that allows investigation of the effects of photosensitizers attached to the plasma membrane. For cell adhesion, poly[N-(3-aminopropyl)methacrylamide] conjugated with hematoporphyrin PA-HpD was immobilized on the glass surfaces. Singlet oxygen was produced from the PA-HpD-immobilized glass surface upon laser irradiation at 635 nm. When murine colon adenocarcinoma 26 (Colon-26) cells were cultured on the PA-HpD-immobilized surface, the cells were swollen and ruptured, leading to effective apoptotic cell death using laser irradiation at 635 nm. In addition, microvesicles of approximately 10 μm in diameter were released from the plasma membrane into the culture medium. These phenomena were due to the oxidation of lipids in the cellular membrane, caused by the plasma membrane-targeted photodynamic therapy. In contrast, these phenomena were not observed on poly[N-(3-aminopropyl)methacrylamide]-immobilized glass surfaces. These results indicate that cell culture dishes with singlet oxygen-generating cell-adhesive glass surfaces can be used to investigate fundamental mechanisms in plasma membrane-targeted photodynamic therapy.
Collapse
Affiliation(s)
- Van Thi Hong Doan
- Research Center for Macromolecules and Biomaterials, National Institute for Materials Science, 1-1 Namiki, Tsukuba, Ibaraki, 305-0044, Japan
| | - Yoshiki Komatsu
- Research Center for Macromolecules and Biomaterials, National Institute for Materials Science, 1-1 Namiki, Tsukuba, Ibaraki, 305-0044, Japan; Division of Gastroenterology, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Ibaraki, 305-8575, Japan
| | - Hirofumi Matsui
- Division of Gastroenterology, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Ibaraki, 305-8575, Japan
| | - Naoki Kawazoe
- Research Center for Macromolecules and Biomaterials, National Institute for Materials Science, 1-1 Namiki, Tsukuba, Ibaraki, 305-0044, Japan
| | - Guoping Chen
- Research Center for Macromolecules and Biomaterials, National Institute for Materials Science, 1-1 Namiki, Tsukuba, Ibaraki, 305-0044, Japan
| | - Toru Yoshitomi
- Research Center for Macromolecules and Biomaterials, National Institute for Materials Science, 1-1 Namiki, Tsukuba, Ibaraki, 305-0044, Japan.
| |
Collapse
|
16
|
Li J, Pan L, Pan W, Li N, Tang B. Recent progress of oxidative stress associated biomarker detection. Chem Commun (Camb) 2023. [PMID: 37194341 DOI: 10.1039/d3cc00878a] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/18/2023]
Abstract
Oxidative stress denotes the imbalance between the generation of reactive oxygen species (ROS) and antioxidant defenses in living organisms, participating in various pathophysiological processes and mediating the occurrence of diseases. Typically, the excessive production of ROS under oxidative stress elicits oxidative modification of biomacromolecules, including lipids, proteins and nucleic acids, leading to cell dysfunction and damage. Therefore, the analysis and detection of oxidative stress-associated biomarkers are of considerable importance to accurately reflect and evaluate the oxidative stress status. This review comprehensively elucidates the recent advances and applications of imaging probes for tracking and detecting oxidative stress-related biomarkers such as lipid peroxidation, and protein and DNA oxidation. The existing challenges and future development directions in this field are also discussed.
Collapse
Affiliation(s)
- Jingjing Li
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institute of Molecular and Nano Science, Shandong Normal University, Jinan 250014, P. R. China.
| | - Limeng Pan
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institute of Molecular and Nano Science, Shandong Normal University, Jinan 250014, P. R. China.
| | - Wei Pan
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institute of Molecular and Nano Science, Shandong Normal University, Jinan 250014, P. R. China.
| | - Na Li
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institute of Molecular and Nano Science, Shandong Normal University, Jinan 250014, P. R. China.
| | - Bo Tang
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institute of Molecular and Nano Science, Shandong Normal University, Jinan 250014, P. R. China.
| |
Collapse
|
17
|
Fujii J, Yamada KI. Defense systems to avoid ferroptosis caused by lipid peroxidation-mediated membrane damage. Free Radic Res 2023; 57:353-372. [PMID: 37551716 DOI: 10.1080/10715762.2023.2244155] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 07/27/2023] [Accepted: 07/31/2023] [Indexed: 08/09/2023]
Abstract
The presence of hydrogen peroxide along with ferrous iron produces hydroxyl radicals that preferably oxidize polyunsaturated fatty acids (PUFA) to alkyl radicals (L•). The reaction of L• with an oxygen molecule produces lipid peroxyl radical (LOO•) that collectively trigger chain reactions, which results in the accumulation of lipid peroxidation products (LOOH). Oxygenase enzymes, such as lipoxygenase, also stimulate the peroxidation of PUFA. The production of phospholipid hydroperoxides (P-LOOH) can result in the destruction of the architecture of cell membranes and ultimate cell death. This iron-dependent regulated cell death is generally referred to as ferroptosis. Radical scavengers, which include tocopherol and nitric oxide (•NO), react with lipid radicals and terminate the chain reaction. When tocopherol reductively detoxifies lipid radicals, the resultant tocopherol radicals are recycled via reduction by coenzyme Q or ascorbate. CoQ radicals are reduced back by the anti-ferroptotic enzyme FSP1. •NO reacts with lipid radicals and produces less reactive nitroso compounds. The resulting P-LOOH is reductively detoxified by the action of glutathione peroxidase 4 (GPX4) or peroxiredoxin 6 (PRDX6). The hydrolytic removal of LOOH from P-LOOH by calcium-independent phospholipase A2 leads the preservation of membrane structure. While the expression of such protective genes or the presence of these anti-oxidant compounds serve to maintain a healthy condition, tumor cells employ them to make themselves resistant to anti-tumor treatments. Thus, these defense mechanisms against ferroptosis are protective in ordinary cells but are also potential targets for cancer treatment.
Collapse
Affiliation(s)
- Junichi Fujii
- Department of Biochemistry and Molecular Biology, Graduate School of Medical Science, Yamagata University, Yamagata, Japan
| | - Ken-Ichi Yamada
- Faculty of Pharmaceutical Sciences, Physical Chemistry for Life Science Laboratory, Kyushu University, Fukuoka, Japan
| |
Collapse
|
18
|
Udo T, Matsuoka Y, Takahashi M, Izumi Y, Saito K, Tazoe K, Tanaka M, Naka H, Bamba T, Yamada KI. Structural Analysis of Intracellular Lipid Radicals by LC/MS/MS Using a BODIPY-Based Profluorescent Nitroxide Probe. Anal Chem 2023; 95:4585-4591. [PMID: 36847588 DOI: 10.1021/acs.analchem.2c04950] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/01/2023]
Abstract
Free radical-mediated lipid peroxidation (LPO) induces the formation of numerous lipid radicals, which contribute to the development of several oxidative diseases. To understand the mechanism of LPO in biological systems and the significance of these radicals, identifying the structures of individual lipid radicals is imperative. In this study, we developed an analytical method based on liquid chromatography coupled with tandem mass spectrometry (LC/MS/MS) and a profluorescent nitroxide probe, N-(1-oxyl-2,2,6-trimethyl-6-pentylpiperidin-4-yl)-3-(5,5-difluoro-1,3-dimethyl-3H,5H-5l4-dipyrrolo[1,2-c:2',1'-f][1,3,2]diazaborinin-7-yl)propanamide (BDP-Pen), for the detailed structural analysis of lipid radicals. The MS/MS spectra of BDP-Pen-lipid radical adducts showed product ions and thus allow the prediction of the lipid radical structures and individual detection of isomeric adducts. Using the developed technology, we separately detected the isomers of arachidonic acid (AA)-derived radicals generated in AA-treated HT1080 cells. This analytical system is a powerful tool for elucidating the mechanism of LPO in biological systems.
Collapse
Affiliation(s)
- Takumi Udo
- Department of Molecular Pathobiology, Faculty of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi Higashi-ku, Fukuoka 812-8582, Japan
| | - Yuta Matsuoka
- Department of Molecular Pathobiology, Faculty of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi Higashi-ku, Fukuoka 812-8582, Japan
| | - Masatomo Takahashi
- Division of Metabolomics, Medical Research Center for High Depth Omics, Medical Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi Higashi-ku, Fukuoka 812-8582, Japan
| | - Yoshihiro Izumi
- Division of Metabolomics, Medical Research Center for High Depth Omics, Medical Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi Higashi-ku, Fukuoka 812-8582, Japan
| | - Kota Saito
- Department of Molecular Pathobiology, Faculty of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi Higashi-ku, Fukuoka 812-8582, Japan
| | - Kaho Tazoe
- Department of Molecular Pathobiology, Faculty of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi Higashi-ku, Fukuoka 812-8582, Japan
| | - Moe Tanaka
- Department of Molecular Pathobiology, Faculty of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi Higashi-ku, Fukuoka 812-8582, Japan
| | - Hideto Naka
- Department of Molecular Pathobiology, Faculty of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi Higashi-ku, Fukuoka 812-8582, Japan
| | - Takeshi Bamba
- Division of Metabolomics, Medical Research Center for High Depth Omics, Medical Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi Higashi-ku, Fukuoka 812-8582, Japan
| | - Ken-Ichi Yamada
- Department of Molecular Pathobiology, Faculty of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi Higashi-ku, Fukuoka 812-8582, Japan
| |
Collapse
|
19
|
Azuma R, Yamasaki T, Emoto MC, Sato-Akaba H, Sano K, Munekane M, Fujii HG, Mukai T. Effect of relative configuration of TEMPO-type nitroxides on ascorbate reduction. Free Radic Biol Med 2023; 194:114-122. [PMID: 36442586 DOI: 10.1016/j.freeradbiomed.2022.11.033] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 11/10/2022] [Accepted: 11/22/2022] [Indexed: 11/28/2022]
Abstract
2,2,6,6-Tetramethylpiperidin-N-oxyl (TEMPO)-type nitroxides are susceptible to bioreduction, leading to a loss of radical properties. Although it has been reported that the steric and electronic environments around the N-O moiety of nitroxides affect the reduction, how the relative configuration of nitroxide derivatives alters it is unclear. In this study, we investigated the effect of diastereomers on the radical properties of C2- and C4-disubstituted TEMPO-type nitroxides. We succeeded in isolating the diastereomers of the studied nitroxides for the first time. In addition, we compared the reactivities of nitroxide derivatives with different substituents at the C2 and C4 positions toward ascorbate reduction. We found that the bulky substituents at both C2 and C4 and the electronic effect of C4 affected the reduction of the isomers. C2- and C4-disubstituted nitroxides were administered to mice for electron spin resonance imaging to assess bioreduction in the brain. Similar to the reactivity to reduction in vitro, a difference in the bioreduction of diastereomers was observed in brain tissues. Our research strongly indicates that bioreduction can be controlled by changing the relative configuration, which can be used in the design of nitroxide derivatives for biological applications.
Collapse
Affiliation(s)
- Risa Azuma
- Laboratory of Biophysical Chemistry, Kobe Pharmaceutical University, 4-19-1 Motoyamakita-machi, Higashinada-ku, Kobe, 658-8558, Japan
| | - Toshihide Yamasaki
- Laboratory of Biophysical Chemistry, Kobe Pharmaceutical University, 4-19-1 Motoyamakita-machi, Higashinada-ku, Kobe, 658-8558, Japan
| | - Miho C Emoto
- Department of Clinical Laboratory Science, School of Medical Technology, Health Sciences University of Hokkaido, Sapporo, Hokkaido, 002-8072, Japan
| | - Hideo Sato-Akaba
- Department of Systems Innovation, Graduate School of Engineering Science, Osaka University, Toyonaka, Osaka, 560-8531, Japan
| | - Kohei Sano
- Laboratory of Biophysical Chemistry, Kobe Pharmaceutical University, 4-19-1 Motoyamakita-machi, Higashinada-ku, Kobe, 658-8558, Japan
| | - Masayuki Munekane
- Laboratory of Biophysical Chemistry, Kobe Pharmaceutical University, 4-19-1 Motoyamakita-machi, Higashinada-ku, Kobe, 658-8558, Japan
| | - Hirotada G Fujii
- Advanced Research Promotion Center, Health Sciences University of Hokkaido, Ishikari, Hokkaido, 061-0293, Japan
| | - Takahiro Mukai
- Laboratory of Biophysical Chemistry, Kobe Pharmaceutical University, 4-19-1 Motoyamakita-machi, Higashinada-ku, Kobe, 658-8558, Japan.
| |
Collapse
|
20
|
Zhang F, Du T, Jiang L, Zhu L, Tian D. A combined “AIE + ESIPT” fluorescent probe for detection of lipase activity. Bioorg Chem 2022; 128:106026. [DOI: 10.1016/j.bioorg.2022.106026] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 07/09/2022] [Accepted: 07/11/2022] [Indexed: 01/12/2023]
|
21
|
Kinetics of lipid radical formation in lipoproteins from β-thalassemia: Implication of cholesteryl esters and α-tocopherol. Biomed Pharmacother 2022; 154:113624. [DOI: 10.1016/j.biopha.2022.113624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2022] [Revised: 08/27/2022] [Accepted: 08/29/2022] [Indexed: 11/21/2022] Open
|
22
|
Mishima E, Ito J, Wu Z, Nakamura T, Wahida A, Doll S, Tonnus W, Nepachalovich P, Eggenhofer E, Aldrovandi M, Henkelmann B, Yamada KI, Wanninger J, Zilka O, Sato E, Feederle R, Hass D, Maida A, Mourão ASD, Linkermann A, Geissler EK, Nakagawa K, Abe T, Fedorova M, Proneth B, Pratt DA, Conrad M. A non-canonical vitamin K cycle is a potent ferroptosis suppressor. Nature 2022; 608:778-783. [PMID: 35922516 PMCID: PMC9402432 DOI: 10.1038/s41586-022-05022-3] [Citation(s) in RCA: 369] [Impact Index Per Article: 123.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Accepted: 06/23/2022] [Indexed: 02/07/2023]
Abstract
Ferroptosis, a non-apoptotic form of cell death marked by iron-dependent lipid peroxidation1, has a key role in organ injury, degenerative disease and vulnerability of therapy-resistant cancers2. Although substantial progress has been made in understanding the molecular processes relevant to ferroptosis, additional cell-extrinsic and cell-intrinsic processes that determine cell sensitivity toward ferroptosis remain unknown. Here we show that the fully reduced forms of vitamin K—a group of naphthoquinones that includes menaquinone and phylloquinone3—confer a strong anti-ferroptotic function, in addition to the conventional function linked to blood clotting by acting as a cofactor for γ-glutamyl carboxylase. Ferroptosis suppressor protein 1 (FSP1), a NAD(P)H-ubiquinone reductase and the second mainstay of ferroptosis control after glutathione peroxidase-44,5, was found to efficiently reduce vitamin K to its hydroquinone, a potent radical-trapping antioxidant and inhibitor of (phospho)lipid peroxidation. The FSP1-mediated reduction of vitamin K was also responsible for the antidotal effect of vitamin K against warfarin poisoning. It follows that FSP1 is the enzyme mediating warfarin-resistant vitamin K reduction in the canonical vitamin K cycle6. The FSP1-dependent non-canonical vitamin K cycle can act to protect cells against detrimental lipid peroxidation and ferroptosis. Biochemical and lipidomic analyses identify an anti-ferroptotic function of vitamin K and reveal ferroptosis suppressor protein 1 (FSP1) as the enzyme mediating warfarin-resistant vitamin K reduction in the canonical vitamin K cycle.
Collapse
Affiliation(s)
- Eikan Mishima
- Institute of Metabolism and Cell Death, Helmholtz Zentrum München, Neuherberg, Germany. .,Division of Nephrology, Rheumatology and Endocrinology, Tohoku University Graduate School of Medicine, Sendai, Japan.
| | - Junya Ito
- Laboratory of Food Function Analysis, Tohoku University, Sendai, Japan
| | - Zijun Wu
- Department of Chemistry and Biomolecular Science, University of Ottawa, Ottawa, Ontario, Canada
| | - Toshitaka Nakamura
- Institute of Metabolism and Cell Death, Helmholtz Zentrum München, Neuherberg, Germany
| | - Adam Wahida
- Institute of Metabolism and Cell Death, Helmholtz Zentrum München, Neuherberg, Germany
| | - Sebastian Doll
- Institute of Metabolism and Cell Death, Helmholtz Zentrum München, Neuherberg, Germany
| | - Wulf Tonnus
- Universitätsklinikum Carl Gustav Carus Dresden, Technische Universität Dresden, Dresden, Germany
| | - Palina Nepachalovich
- Institute of Bioanalytical Chemistry, Faculty of Chemistry and Mineralogy, Leipzig University, Leipzig, Germany.,Zentrum Membranbiochemie und Lipidforschung, Medizinische Fakultät Carl Gustav Carus, Technical University, Dresden, Germany
| | - Elke Eggenhofer
- Department of Surgery, University Hospital Regensburg, University of Regensburg, Regensburg, Germany
| | - Maceler Aldrovandi
- Institute of Metabolism and Cell Death, Helmholtz Zentrum München, Neuherberg, Germany
| | - Bernhard Henkelmann
- Institute of Metabolism and Cell Death, Helmholtz Zentrum München, Neuherberg, Germany
| | - Ken-Ichi Yamada
- Physical Chemistry for Life Science Laboratory, Faculty of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| | - Jonas Wanninger
- Institute of Metabolism and Cell Death, Helmholtz Zentrum München, Neuherberg, Germany
| | - Omkar Zilka
- Department of Chemistry and Biomolecular Science, University of Ottawa, Ottawa, Ontario, Canada
| | - Emiko Sato
- Division of Clinical Pharmacology and Therapeutics, Tohoku University Graduate School of Pharmaceutical Sciences and Faculty of Pharmaceutical Sciences, Sendai, Japan
| | - Regina Feederle
- Monoclonal Antibody Core Facility, Helmholtz Zentrum München, Neuherberg, Germany
| | - Daniela Hass
- Institute for Diabetes and Cancer, Helmholtz Zentrum München, Neuherberg, Germany
| | - Adriano Maida
- Institute for Diabetes and Cancer, Helmholtz Zentrum München, Neuherberg, Germany
| | | | - Andreas Linkermann
- Universitätsklinikum Carl Gustav Carus Dresden, Technische Universität Dresden, Dresden, Germany
| | - Edward K Geissler
- Department of Surgery, University Hospital Regensburg, University of Regensburg, Regensburg, Germany
| | - Kiyotaka Nakagawa
- Laboratory of Food Function Analysis, Tohoku University, Sendai, Japan
| | - Takaaki Abe
- Division of Nephrology, Rheumatology and Endocrinology, Tohoku University Graduate School of Medicine, Sendai, Japan.,Division of Medical Science, Tohoku University Graduate School of Biomedical Engineering, Sendai, Japan
| | - Maria Fedorova
- Institute of Bioanalytical Chemistry, Faculty of Chemistry and Mineralogy, Leipzig University, Leipzig, Germany.,Zentrum Membranbiochemie und Lipidforschung, Medizinische Fakultät Carl Gustav Carus, Technical University, Dresden, Germany
| | - Bettina Proneth
- Institute of Metabolism and Cell Death, Helmholtz Zentrum München, Neuherberg, Germany
| | - Derek A Pratt
- Department of Chemistry and Biomolecular Science, University of Ottawa, Ottawa, Ontario, Canada
| | - Marcus Conrad
- Institute of Metabolism and Cell Death, Helmholtz Zentrum München, Neuherberg, Germany.
| |
Collapse
|
23
|
A Two-Photon Fluorescent Probe for the Visual Detection of Peroxynitrite in Living Cells and Zebrafish. Molecules 2022; 27:molecules27154858. [PMID: 35956806 PMCID: PMC9369896 DOI: 10.3390/molecules27154858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2022] [Revised: 07/22/2022] [Accepted: 07/23/2022] [Indexed: 11/17/2022] Open
Abstract
Peroxynitrite (ONOO−), as an important reactive oxygen species (ROS), holds great potential to react with a variety of biologically active substances, leading to the occurrence of various diseases such as cancer and neurodegenerative diseases. In this work, we developed a novel mitochondria-localized fluorescent probe, HDBT-ONOO−, which was designed as a mitochondria-targeting two-photon fluorescence probe based on 1,8-naphthylimide fluorophore and the reactive group of 4-(bromomethyl)-benzene boronic acid pinacol ester. More importantly, the probe exhibited good biocompatibility, sensitivity, and selectivity, enabling its successful application in imaging the generation of intracellular and extracellular ONOO−. Furthermore, exogenous and endogenous ONOO− products in live zebrafish were visualized. It is greatly expected that the designed probe can serve as a useful imaging tool for clarifying the distribution and pathophysiological functions of ONOO− in cells and zebrafish.
Collapse
|
24
|
Jomen W, Ohtake T, Akita T, Suto D, Yagi H, Osawa Y, Kohgo Y. Iron chelator deferasirox inhibits NF-κB activity in hepatoma cells and changes sorafenib-induced programmed cell deaths. Biomed Pharmacother 2022; 153:113363. [PMID: 35834989 DOI: 10.1016/j.biopha.2022.113363] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 06/18/2022] [Accepted: 06/28/2022] [Indexed: 11/27/2022] Open
Abstract
OBJECTIVE The improvements of antitumor effects and tolerability on chemotherapy for advanced hepatocellular carcinoma (HCC) are warranted. Here, we aimed to elucidate the mechanism of the combining effect of tyrosine kinase inhibitor sorafenib (SOR) and iron chelator deferasirox (DFX) in human hepatoma cell lines, HepG2 and Huh-7. METHODS The types of programmed cell deaths (PCDs); necrosis/necroptosis and apoptosis, were evaluated by flow cytometry and fluorescent microscopy. Human cleaved caspase-3 was analyzed by ELISA for apoptosis. GSH assay was used for ferroptosis. PCDs inhibition was analyzed by adding apoptosis inhibitor Z-VAD-FMK, ferroptosis inhibitor ferrostatin-1, necroptosis inhibitor necrosulfonamide, respectively. The expression of NF-κB was quantified by Western blotting. RESULTS In SOR monotherapy, cleaved caspase-3 expression was increased in all concentrations, confirming the result that SOR induces apoptosis. In SOR monotherapy, GSH/GSSG ratio was decreased on concentration-dependent, showing that SOR also induced ferroptosis. Lipid Peroxidation caused by SOR, corresponding to ferroptosis, was suppressed by DFX. In fluorescence microscopy of SOR monotherapy, apoptosis was observed at a constant rate on all concentrations, while necroptosis and ferroptosis were increased on high concentration. In sorafenib and deferasirox combinations, sub G1 phase increased additively. In SOR and DFX combinations, the cytotoxic effects were not suppressed by ferrostatin-1, but suppressed by Z-VAD-FMK and necrosulfonamide. In each monotherapy, and SOR + DFX combinations, the expression of NF-κB in nucleus was suppressed. Regarding PCD by SOR and DFX combination, ferroptosis was suppressed and both apoptosis and necroptosis became dominant. CONCLUSION Suppression of NF-κB is possibly involved in the effect of DFX. As a result, SOR and DFX combination showed additive antitumor effects for HCC through the mechanism of programed cell deaths and NF-kB signal modification.
Collapse
Affiliation(s)
- Wataru Jomen
- Department of Clinical Medical Sciences, International University of Health and Welfare Graduate School of Medicine, Tokyo, Japan
| | - Takaaki Ohtake
- Department of Gastroenterology, International University of Health and Welfare School of Medicine, Narita, Japan; Department of Gastroenterology and Hepatology, International University of Health and Welfare Hospital, Nasushiobara, Japan.
| | - Takayuki Akita
- Department of Clinical Medical Sciences, International University of Health and Welfare Graduate School of Medicine, Tokyo, Japan; Department of Gastroenterology and Hepatology, International University of Health and Welfare Hospital, Nasushiobara, Japan
| | - Daisuke Suto
- Department of Gastroenterology, International University of Health and Welfare School of Medicine, Narita, Japan; Department of Gastroenterology and Hepatology, International University of Health and Welfare Hospital, Nasushiobara, Japan
| | - Hideki Yagi
- Department of Pharmaceutical, Faculty of Pharmacy, International University of Health and Welfare, Otawara, Japan
| | - Yosuke Osawa
- Department of Clinical Medical Sciences, International University of Health and Welfare Graduate School of Medicine, Tokyo, Japan; Department of Gastroenterology, International University of Health and Welfare School of Medicine, Narita, Japan; Department of Gastroenterology and Hepatology, International University of Health and Welfare Hospital, Nasushiobara, Japan
| | - Yutaka Kohgo
- Department of Clinical Medical Sciences, International University of Health and Welfare Graduate School of Medicine, Tokyo, Japan; Department of Gastroenterology, International University of Health and Welfare School of Medicine, Narita, Japan; Department of Gastroenterology and Hepatology, International University of Health and Welfare Hospital, Nasushiobara, Japan
| |
Collapse
|
25
|
Naguib M, Feldman N, Zarodkiewicz P, Shropshire H, Biamis C, El-Halfawy OM, McCain J, Dezanet C, Décout JL, Chen Y, Cosa G, Valvano MA. An evolutionary conserved detoxification system for membrane lipid-derived peroxyl radicals in Gram-negative bacteria. PLoS Biol 2022; 20:e3001610. [PMID: 35580139 PMCID: PMC9113575 DOI: 10.1371/journal.pbio.3001610] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Accepted: 03/18/2022] [Indexed: 11/30/2022] Open
Abstract
How double-membraned Gram-negative bacteria overcome lipid peroxidation is virtually unknown. Bactericidal antibiotics and superoxide ion stress stimulate the transcription of the Burkholderia cenocepacia bcnA gene that encodes a secreted lipocalin. bcnA gene orthologs are conserved in bacteria and generally linked to a conserved upstream gene encoding a cytochrome b561 membrane protein (herein named lcoA, lipocalin-associated cytochrome oxidase gene). Mutants in bcnA, lcoA, and in a gene encoding a conserved cytoplasmic aldehyde reductase (peroxidative stress-associated aldehyde reductase gene, psrA) display enhanced membrane lipid peroxidation. Compared to wild type, the levels of the peroxidation biomarker malondialdehyde (MDA) increase in the mutants upon exposure to sublethal concentrations of the bactericidal antibiotics polymyxin B and norfloxacin. Microscopy with lipid peroxidation-sensitive fluorescent probes shows that lipid peroxyl radicals accumulate at the bacterial cell poles and septum and peroxidation is associated with a redistribution of anionic phospholipids and reduced antimicrobial resistance in the mutants. We conclude that BcnA, LcoA, and PsrA are components of an evolutionary conserved, hitherto unrecognized peroxidation detoxification system that protects the bacterial cell envelope from lipid peroxyl radicals.
Collapse
Affiliation(s)
- Marwa Naguib
- Wellcome-Wolfson Institute for Experimental Medicine, Queen’s University Belfast, Belfast, United Kingdom
- Department of Microbiology and Immunology, Faculty of Pharmacy, Damanhour University, Damanhour, Egypt
| | - Nicolás Feldman
- Wellcome-Wolfson Institute for Experimental Medicine, Queen’s University Belfast, Belfast, United Kingdom
| | - Paulina Zarodkiewicz
- Wellcome-Wolfson Institute for Experimental Medicine, Queen’s University Belfast, Belfast, United Kingdom
| | - Holly Shropshire
- School of Life Sciences, University of Warwick, Coventry, United Kingdom
| | - Christina Biamis
- Wellcome-Wolfson Institute for Experimental Medicine, Queen’s University Belfast, Belfast, United Kingdom
| | - Omar M. El-Halfawy
- Department of Chemistry and Biochemistry, Faculty of Science, University of Regina, Regina, Saskatchewan, Canada
- Department of Microbiology and Immunology, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| | - Julia McCain
- Department of Chemistry and Quebec Center for Advanced Materials, McGill University, Montreal, Québec, Canada
| | - Clément Dezanet
- Department of Molecular Pharmacochemistry, Université Grenoble Alpes/CNRS, Grenoble, France
| | - Jean-Luc Décout
- Department of Molecular Pharmacochemistry, Université Grenoble Alpes/CNRS, Grenoble, France
| | - Yin Chen
- School of Life Sciences, University of Warwick, Coventry, United Kingdom
| | - Gonzalo Cosa
- Department of Chemistry and Quebec Center for Advanced Materials, McGill University, Montreal, Québec, Canada
| | - Miguel A. Valvano
- Wellcome-Wolfson Institute for Experimental Medicine, Queen’s University Belfast, Belfast, United Kingdom
| |
Collapse
|
26
|
Cui X, Zhang Z, Yang Y, Li S, Lee C. Organic radical materials in biomedical applications: State of the art and perspectives. EXPLORATION (BEIJING, CHINA) 2022; 2:20210264. [PMID: 37323877 PMCID: PMC10190988 DOI: 10.1002/exp.20210264] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 02/24/2022] [Indexed: 06/17/2023]
Abstract
Owing to their unique chemical reactivities and paramagnetism, organic radicals with unpaired electrons have found widespread exploration in physical, chemical, and biological fields. However, most radicals are too short-lived to be separated and only a few of them can maintain stable radical forms via stereochemical strategies. How to utilize these raw radicals for developing stable radical-containing materials have long been a research hotspot for many years. This perspective introduces fundamental characteristics of organic radical materials and highlights their applications in biomedical fields, particularly for bioimaging, biosensing, and photo-triggered therapies. Molecular design of these radical materials is considered with reference to their outstanding imaging and therapeutic performances. Various challenges currently limiting the wide applications of these organic radical materials and their future development are also discussed.
Collapse
Affiliation(s)
- Xiao Cui
- Department of ChemistryInstitution Center of Super‐Diamond and Advanced Films (COSDAF)City University of Hong KongKowloonHong Kong SARChina
| | - Zhen Zhang
- Department of ChemistryInstitution Center of Super‐Diamond and Advanced Films (COSDAF)City University of Hong KongKowloonHong Kong SARChina
| | - Yuliang Yang
- College of Pharmaceutical SciencesSoochow UniversitySuzhouChina
| | - Shengliang Li
- College of Pharmaceutical SciencesSoochow UniversitySuzhouChina
| | - Chun‐Sing Lee
- Department of ChemistryInstitution Center of Super‐Diamond and Advanced Films (COSDAF)City University of Hong KongKowloonHong Kong SARChina
| |
Collapse
|
27
|
Yamasaki T, Sano K, Mukai T. Redox Monitoring in Nuclear Medical Imaging. Antioxid Redox Signal 2022; 36:797-810. [PMID: 34847731 DOI: 10.1089/ars.2021.0246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Significance: The imbalance in redox homeostasis is known as oxidative stress, which is relevant to many diseases such as cancer, arteriosclerosis, and neurodegenerative disorders. Overproduction of reactive oxygen species (ROS) is one of the factors that trigger the redox state imbalance in vivo. The ROS have high reactivity and impair biomolecules, whereas antioxidants and antioxidant enzymes, such as ascorbate and glutathione, reduce the overproduction of ROS to rectify the redox imbalance. Owing to this, redox monitoring tools have been developed to understand the redox fluctuations in oxidative stress-related diseases. Recent Advances: In an attempt to monitor redox substances, including ROS and radical species, versatile modalities have been developed, such as electron spin resonance, chemiluminescence, and fluorescence. In particular, many fluorescent probes have been developed that are selective for ROS. This has significantly contributed to understanding the relevance of ROS in disease onset and progression. Critical Issues: To date, the dynamics of ROS and radical fluctuation in in vivo redox states remain unclear, and there are a few methods for the in vivo detection of redox fluctuations. Future Directions: In this review, we summarize the development of radiolabeled probes for monitoring redox-relevant species by nuclear medical imaging that is applicable in vivo. In the future, translational research is likely to be advanced through the development of highly sensitive and in vivo applicable detection methods, such as nuclear medical imaging, to clarify the underlying dynamics of ROS, radicals, and redox substances in many diseases. Antioxid. Redox Signal. 36, 797-810.
Collapse
Affiliation(s)
- Toshihide Yamasaki
- Laboratory of Biophysical Chemistry, Kobe Pharmaceutical University, Kobe, Japan
| | - Kohei Sano
- Laboratory of Biophysical Chemistry, Kobe Pharmaceutical University, Kobe, Japan
| | - Takahiro Mukai
- Laboratory of Biophysical Chemistry, Kobe Pharmaceutical University, Kobe, Japan
| |
Collapse
|
28
|
Nishizawa H, Yamanaka M, Igarashi K. Ferroptosis: regulation by competition between NRF2 and BACH1 and propagation of the death signal. FEBS J 2022; 290:1688-1704. [PMID: 35107212 DOI: 10.1111/febs.16382] [Citation(s) in RCA: 75] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Revised: 01/25/2022] [Accepted: 01/31/2022] [Indexed: 12/15/2022]
Abstract
Ferroptosis is triggered by a chain of intracellular labile iron-dependent peroxidation of cell membrane phospholipids. Ferroptosis is important not only as a cause of ischaemic and neurodegenerative diseases but also as a mechanism of cancer suppression, and a better understanding of its regulatory mechanism is required. It has become clear that ferroptosis is finely controlled by two oxidative stress-responsive transcription factors, NRF2 (NF-E2-related factor 2) and BACH1 (BTB and CNC homology 1). NRF2 and BACH1 inhibit and promote ferroptosis, respectively, by activating or suppressing the expression of genes in the major regulatory pathways of ferroptosis: intracellular labile iron metabolism, the GSH (glutathione) -GPX4 (glutathione peroxidase 4) pathway and the FSP1 (ferroptosis suppressor protein 1)-CoQ (coenzyme Q) pathway. In addition to this, NRF2 and BACH1 control ferroptosis through the regulation of lipid metabolism and cell differentiation. This multifaceted regulation of ferroptosis by NRF2 and BACH1 is considered to have been acquired during the evolution of multicellular organisms, allowing the utilization of ferroptosis for maintaining homeostasis, including cancer suppression. In terms of cell-cell interaction, it has been revealed that ferroptosis has the property of propagating to surrounding cells along with lipid peroxidation. The regulation of ferroptosis by NRF2 and BACH1 and the propagation phenomenon could be used to realize anticancer cell therapy in the future. In this review, these points will be summarized and discussed.
Collapse
Affiliation(s)
- Hironari Nishizawa
- Department of Biochemistry, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Mie Yamanaka
- Department of Biochemistry, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Kazuhiko Igarashi
- Department of Biochemistry, Tohoku University Graduate School of Medicine, Sendai, Japan.,Center for Regulatory Epigenome and Diseases, Tohoku University Graduate School of Medicine, Sendai, Japan
| |
Collapse
|
29
|
Fluorescence imaging for visualizing the bioactive molecules of lipid peroxidation within biological systems. Trends Analyt Chem 2022. [DOI: 10.1016/j.trac.2021.116484] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
30
|
Yasukawa K. Redox-Based Theranostics of Gastric Ulcers Using Nitroxyl Radicals. Antioxid Redox Signal 2022; 36:160-171. [PMID: 34498915 DOI: 10.1089/ars.2021.0104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Significance: Redox-based theranostics involves redox monitoring and therapeutics that normalize redox imbalance. It may be a promising approach to markedly improve a patient's quality of life through streamlined treatment. Nitroxyl radicals are useful for both redox monitoring and treating gastric ulcers in rodents. Recent Advances: Redox monitoring using in vivo electron paramagnetic resonance (EPR) spectroscopy in a gastric ulcer rat model showed the production of reactive oxygen species in the whole stomach. A combination of Overhauser-enhanced magnetic resonance imaging (MRI) and nitroxyl radicals provided high-resolution images of redox imbalance in the stomach of rats with a gastric ulcer. Treatment with nitroxyl radicals was effective to treat ulcers that were formed using model experiments of Helicobacter pylori and mental stress as well as nonsteroidal anti-inflammatory drugs. Critical Issues: For redox monitoring using Overhauser-enhanced MRI, the EPR irradiation power that is delivered to subjects must be within the range of the specific absorption rate regulation to protect against microwave damage regardless of a decrease in image contrast. The effect of long-term treatment with a nitroxyl radical in patients with a gastric ulcer remains unclear. Future Directions: Further research on redox-based theranostics in redox-related diseases, including gastric ulcers, would be accelerated by improving the redox imager and by developing functional nitroxyl radicals that localize in the target organ, tissue, or cell and that have specific reactivity for the redox-related biomolecule.
Collapse
Affiliation(s)
- Keiji Yasukawa
- Laboratory of Advanced Pharmacology, Faculty of Pharmaceutical Sciences, Daiichi University of Pharmacy, Fukuoka, Japan
| |
Collapse
|
31
|
Yamauchi K, Matsuoka Y, Takahashi M, Izumi Y, Naka H, Taniguchi Y, Kawai K, Bamba T, Yamada KI. Detection and structural analysis of pyrimidine-derived radicals generated on DNA using a profluorescent nitroxide probe. Chem Commun (Camb) 2021; 58:56-59. [PMID: 34897335 DOI: 10.1039/d1cc04998d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The oxidative damage of DNA is associated with aging and the development of various diseases. Although nucleoside-derived radicals play an important role in DNA oxidation, their analysis methods are limited. Herein, we propose a fluorometric detection and structural analysis of radicals on the surface of oxidatively damaged DNA using a profluorescent nitroxide probe combined with liquid chromatography-fluorometry and high-resolution tandem mass spectrometry.
Collapse
Affiliation(s)
- Kosho Yamauchi
- Physical Chemistry for Life Science Laboratory, Faculty of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi Higashi-ku, Fukuoka 812-8582, Japan.
| | - Yuta Matsuoka
- Physical Chemistry for Life Science Laboratory, Faculty of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi Higashi-ku, Fukuoka 812-8582, Japan.
| | - Masatomo Takahashi
- Metabolomics Laboratory, Research Center for Transomics Medicine, Medical Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi Higashi-ku, Fukuoka 812-8582, Japan
| | - Yoshihiro Izumi
- Metabolomics Laboratory, Research Center for Transomics Medicine, Medical Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi Higashi-ku, Fukuoka 812-8582, Japan
| | - Hideto Naka
- Physical Chemistry for Life Science Laboratory, Faculty of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi Higashi-ku, Fukuoka 812-8582, Japan.
| | - Yosuke Taniguchi
- Frontier in Biofunction of Nucleic Acid and Organic Chemistry, Faculty of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi Higashi-ku, Fukuoka 812-8582, Japan
| | - Kazuaki Kawai
- Department of Environmental Oncology, Institute of Industrial Ecological Sciences, University of Occupational and Environmental Health, 1-1 Iseigaoka, Yahatanishi-ku, Kitakyushu, 807-8555, Japan
| | - Takeshi Bamba
- Metabolomics Laboratory, Research Center for Transomics Medicine, Medical Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi Higashi-ku, Fukuoka 812-8582, Japan
| | - Ken-Ichi Yamada
- Physical Chemistry for Life Science Laboratory, Faculty of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi Higashi-ku, Fukuoka 812-8582, Japan.
| |
Collapse
|
32
|
Saito Y. Diverse cytoprotective actions of vitamin E isoforms- role as peroxyl radical scavengers and complementary functions with selenoproteins. Free Radic Biol Med 2021; 175:121-129. [PMID: 34481936 DOI: 10.1016/j.freeradbiomed.2021.08.234] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 08/25/2021] [Accepted: 08/26/2021] [Indexed: 02/05/2023]
Abstract
Vitamin E, a generic term for tocopherol (T) and tocotrienol (T3), is one of the most potent lipid-soluble antioxidants in the body. It is classified into T and T3 based on the difference in the side chain structure. T and T3 have four isoforms: α-, β-, γ-, and δ, which have different chroman rings. Both T and T3 exhibit a similar ability to scavenge free radicals, and the extent of this ability depends on the difference in the chroman structure. However, they display unique cytoprotective activities in cultured cells depending on the difference in the side chain structure. The cytoprotective effects of vitamin E have received much attention in the prevention of ferroptosis, which is a distinct form of cell death involving iron-dependent lipid peroxidation. This review focuses on the cytoprotective actions of vitamin E isoforms against oxidative stress, particularly the difference between T and T3 and its relation to cellular uptake and distribution. Moreover, the molecular mechanism for cytoprotection of vitamin E oxidation products is explained, and the complementary role of vitamin E and selenoproteins to prevent lipid peroxidation and ferroptosis is described. Furthermore, the evaluation of vitamin E's radical scavenging activity in vivo using oxidative stress markers is discussed, particularly based on kinetic data and the physiological molar ratio of vitamin E to substrates, and the limited role of vitamin E as a peroxyl radical scavenger is described. The future directions and unresolved issues related to vitamin E and lipid peroxidation are also discussed.
Collapse
Affiliation(s)
- Yoshiro Saito
- Laboratory of Molecular Biology and Metabolism, Graduate School of Pharmaceutical Sciences, Tohoku University C301, 6-3 Aoba, Aramaki, Aoba-ku, Sendai, Miyagi 980-8578, Japan.
| |
Collapse
|
33
|
Matsuoka Y, Yamada KI. Detection and structural analysis of lipid-derived radicals in vitro and in vivo. Free Radic Res 2021; 55:441-449. [PMID: 33504242 DOI: 10.1080/10715762.2021.1881500] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Lipids can be oxidized by reactive oxygen species, resulting in lipid peroxidation and the formation of reactive metabolites such as lipid-derived electrophiles. These products have been reported to induce inflammation, angiogenesis, and ferroptosis. Lipid peroxidation can produce many different products, each of which performs a different function, and which can be challenging to detect in vivo. The initial products of lipid oxidation are lipid-derived radicals, which can cause extensive chain reactions leading to lipid peroxidation. Hence, the ability to detect lipid radicals may provide information about this important class of molecules and the mechanism by which they cause cellular and tissue damage in a wide range of oxidative conditions. In this review, we report recent scientific advances in the detection of lipid-derived radicals in vitro and in cultured cells. We also introduce the possibility of visualization and structural analysis of lipid-derived radicals generated not only in in cells but also in animal tissue samples from oxidative disease models, using fluorescence-based lipid radicals' detection probes. We anticipate that the various innovative techniques summarized in this paper will be applied and further developed to clarify the role of lipid peroxidation in the pathogenesis of oxidative stress-associated diseases.
Collapse
Affiliation(s)
- Yuta Matsuoka
- Physical Chemistry for Life Science Laboratory, Faculty of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| | - Ken-Ichi Yamada
- Physical Chemistry for Life Science Laboratory, Faculty of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| |
Collapse
|
34
|
Azuma R, Yamasaki T, Sano K, Munekane M, Matsuoka Y, Yamada KI, Mukai T. A radioiodinated nitroxide probe with improved stability against bioreduction for in vivo detection of lipid radicals. Free Radic Biol Med 2021; 163:297-305. [PMID: 33359688 DOI: 10.1016/j.freeradbiomed.2020.12.028] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 12/15/2020] [Accepted: 12/18/2020] [Indexed: 10/22/2022]
Abstract
It is well known that lipid carbon radicals (lipid radicals) are the origin of lipid peroxidation and are involved in various diseases such as cancer. Therefore, the in vivo detection of lipid radicals would be expected to lead to early diagnosis of these diseases. However, there are no methods for measuring lipid radicals in vivo. Nitroxides are known to be highly reactive with lipid radicals, but they tend to be reduced in vivo. Focusing on the excellent detection sensitivity of nuclear medical imaging, we have developed a radioiodinated nitroxide derivative with resistance to bioreduction for the in vivo detection of lipid radicals. The desired compound was obtained successfully and was highly stable against bioreduction while maintaining high reactivity toward lipid radicals. The I-125 labeling was efficacious with radiochemical yields of 84-87% and radiochemical purities of >99%. A cellular uptake assay showed that the radioiodinated compound was significantly taken up by cells under lipid radical-producing conditions compared to that in the absence of lipid radical production. A biodistribution study indicated that the radioiodinated compound accumulated more in organs where lipid peroxidation was promoted than the methoxyamine derivative, which lost reactivity to lipid radicals. These results indicated that the developed probe became trapped in cells or organs by reacting with lipid radicals. Thus, the radioiodinated nitroxide is a candidate probe for in vivo detection of lipid radicals.
Collapse
Affiliation(s)
- Risa Azuma
- Laboratory of Biophysical Chemistry, Kobe Pharmaceutical University, 4-19-1 Motoyama-kita, Higashinada, Kobe, 658-8558, Japan
| | - Toshihide Yamasaki
- Laboratory of Biophysical Chemistry, Kobe Pharmaceutical University, 4-19-1 Motoyama-kita, Higashinada, Kobe, 658-8558, Japan
| | - Kohei Sano
- Laboratory of Biophysical Chemistry, Kobe Pharmaceutical University, 4-19-1 Motoyama-kita, Higashinada, Kobe, 658-8558, Japan
| | - Masayuki Munekane
- Laboratory of Biophysical Chemistry, Kobe Pharmaceutical University, 4-19-1 Motoyama-kita, Higashinada, Kobe, 658-8558, Japan
| | - Yuta Matsuoka
- Physical Chemistry for Life Science Laboratory, Faculty of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Ken-Ichi Yamada
- Physical Chemistry for Life Science Laboratory, Faculty of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan; AMED-CREST, Japan Agency for Medical Research and Development, Tokyo, Japan
| | - Takahiro Mukai
- Laboratory of Biophysical Chemistry, Kobe Pharmaceutical University, 4-19-1 Motoyama-kita, Higashinada, Kobe, 658-8558, Japan.
| |
Collapse
|
35
|
Matsuoka Y. [Developments of Profluorescent Nitroxide Probes for Highly Sensitive and Selective Detection of Biological Redox Molecules]. YAKUGAKU ZASSHI 2021; 141:1297-1304. [PMID: 34853201 DOI: 10.1248/yakushi.21-00149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Disruption of the redox balance in vivo is closely involved in the development of various diseases associated with oxidative stress. Therefore, methods for the in vivo analysis of antioxidants and free radicals are essential to elucidate the pathogenic mechanisms of such diseases. Although profluorescent nitroxide probes can be used to evaluate redox molecules with high sensitivity, these probes have low selectivity. Recently, we developed two profluorescent nitroxide probes, 15-((9-(ethylimino)-10-methyl-9Hbenzo[a]phenoxazin-5-yl)amino)-3,11-dioxa-7-azadispiro-hexadecan-7-yloxyl (Nile-DiPy) and 2,2,6-trimethyl-4-(4-nitrobenzo[1,2,5]oxadiazol-7-ylamino)-6-pentylpiperidine-1-oxyl (NBD-Pen), which had high sensitivity and selectivity toward ascorbic acid and lipid-derived radicals, respectively. These probes can react sensitively and selectively to each target molecule and can be used in animal experiments. In this paper, we review the design strategies and application of these profluorescent nitroxide probes.
Collapse
Affiliation(s)
- Yuta Matsuoka
- Physical Chemistry for Life Science Laboratory, Faculty of Pharmaceutical Sciences, Kyushu University
| |
Collapse
|
36
|
Hagihara R, Umeno T, Ueki S, Yoshihara D, Fuchi Y, Usui K, Sakuma M, Yamada KI, Karasawa S. Push-Pull Bisnaphthyridylamine Supramolecular Nanoparticles: Polarity-Induced Aggregation and Crystallization-Induced Emission Enhancement and Fluorescence Resonance Energy Transfer. Chemistry 2020; 27:3039-3046. [PMID: 32935395 DOI: 10.1002/chem.202003854] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 09/10/2020] [Indexed: 11/07/2022]
Abstract
Emissive push-pull-type bisnaphthyridylamine derivatives (BNA-X: X=Me, Et, Bzl, Ph, BuBr, and BuTEMPO) aggregate in aqueous methanol. Furthermore, a two-step emission and aggregation process is controllable by varying the methanol-to-water ratio. At 2:3 MeOH/H2 O, crystallization-induced emission enhancement (CIEE) occurs via formation of an emissive crystal phase, whereas, at 1:9 MeOH/H2 O, aggregation-induced emission enhancement (AIEE) occurs, induced by emissive supramolecular nanoparticles (NPs). For BNA-Ph, the emission quantum yield was 25 times higher in aqueous methanol than that in pure methanol. Despite the high hydrophobicity of BNA-X (C log P=6.1-8.0), the spherical NPs were monodisperse (polydispersity indices <0.2). Moreover, the emissive NPs exhibited fluorescence resonance energy transfer (FRET) with pyrene; however, for BNA-X bearing the TEMPO radical (BNA-BuTEMPO), no FRET was observed because of quenching. In particular, the BNA-BuTEMPO NPs have a slow rotational correlation time (1.3 ns), suggesting applications as magnetic resonance imaging contrast agents with large relaxivity.
Collapse
Affiliation(s)
- Ryusuke Hagihara
- Graduate School of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Tomohiro Umeno
- Faculty of Pharmaceutical Sciences, Showa Pharmaceutical University, 3-3165 Higashi-Tamagawagakuen, Machida, Tokyo, 194-8543, Japan
| | - Shoji Ueki
- Kagawa School of Pharmaceutical Sciences, Tokushima Bunri University, 1314-1 Shido, Sanuki City, Kagawa, 769-2193, Japan
| | - Daisuke Yoshihara
- Materials Open Laboratory (BUNSEKI-NEXT), Institute of Systems Information Technologies and Nanotechnologies (ISIT), 4-1 Kyudaishinmachi, Nishi-ku, Fukuoka, 819-0388, Japan
| | - Yasufumi Fuchi
- Faculty of Pharmaceutical Sciences, Showa Pharmaceutical University, 3-3165 Higashi-Tamagawagakuen, Machida, Tokyo, 194-8543, Japan
| | - Kazuteru Usui
- Faculty of Pharmaceutical Sciences, Showa Pharmaceutical University, 3-3165 Higashi-Tamagawagakuen, Machida, Tokyo, 194-8543, Japan
| | - Masaomi Sakuma
- Faculty of Pharmaceutical Sciences, Showa Pharmaceutical University, 3-3165 Higashi-Tamagawagakuen, Machida, Tokyo, 194-8543, Japan
| | - Ken-Ichi Yamada
- Graduate School of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Satoru Karasawa
- Faculty of Pharmaceutical Sciences, Showa Pharmaceutical University, 3-3165 Higashi-Tamagawagakuen, Machida, Tokyo, 194-8543, Japan
| |
Collapse
|
37
|
Shiraishi R, Matsumoto S, Fuchi Y, Naganuma T, Yoshihara D, Usui K, Yamada KI, Karasawa S. Characterization and Water-Proton Longitudinal Relaxivities of Liposome-Type Radical Nanoparticles Prepared via a Supramolecular Approach. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2020; 36:5280-5286. [PMID: 32321252 DOI: 10.1021/acs.langmuir.0c00610] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
For the construction of metal-free magnetic resonance imaging (MRI) contrast agents, radical-based nanoparticles (RNPs) are promising materials because they allow the water-proton longitudinal relaxivity (r1) to be enhanced not only by paramagnetic resonance effects but also by prolonging the rotational correlation times (τR). However, the τR effect is limited because the radical units are often located within the central hydrophobic core of oil-in-water (o/w) emulsions, resulting in a lack of water molecules surrounding the radical units. In this study, to construct supramolecular RNPs that have high r1 values, we designed a liposome-type RNP in which the radical units are located at positions with sufficient surrounding water molecules. Using this strategy, PRO1 with a PROXYL framework was prepared by introducing hydrophilic groups on both sides of the radical unit. The RNP composed of PRO1 formed spherical nanoparticles approximately 100 nm in size and yielded a higher r1 value (0.26 mM-1 s-1) compared to those of small radical species and similar supramolecular o/w emulsion-type nanoparticles (0.17 mM-1 s-1 in PRO2).
Collapse
Affiliation(s)
- Ryoma Shiraishi
- Faculty of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi Higashi-ku, Fukuoka 812-8582, Japan
| | - Shota Matsumoto
- Faculty of Pharmaceutical Sciences, Showa Pharmaceutical University, 3-3165 Higashi-tamagawagakuen, Machida, Tokyo 194-0042, Japan
| | - Yasufumi Fuchi
- Faculty of Pharmaceutical Sciences, Showa Pharmaceutical University, 3-3165 Higashi-tamagawagakuen, Machida, Tokyo 194-0042, Japan
| | - Tatsuya Naganuma
- Development Department Director, Japan REDOX Limited, 4-29-49-805 Chiyo Hakata-ku, Fukuoka 812-0044, Japan
| | - Daisuke Yoshihara
- Materials Open Laboratory (BUNSEKI-NEXT), Institute of Systems Information Technologies and Nanotechnologies (ISIT), 4-1 Kyudaishinmachi, Nishi-ku, Fukuoka 819-0388, Japan
| | - Kazuteru Usui
- Faculty of Pharmaceutical Sciences, Showa Pharmaceutical University, 3-3165 Higashi-tamagawagakuen, Machida, Tokyo 194-0042, Japan
| | - Ken-Ichi Yamada
- Faculty of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi Higashi-ku, Fukuoka 812-8582, Japan
| | - Satoru Karasawa
- Faculty of Pharmaceutical Sciences, Showa Pharmaceutical University, 3-3165 Higashi-tamagawagakuen, Machida, Tokyo 194-0042, Japan
- PRESTO, Japan Science and Technology Agency, 4-1-8 Honcho, Kawaguchi 332-0012, Japan
| |
Collapse
|
38
|
Saito K, Matsuoka Y, Yamada KI. Reaction targets of antioxidants in azo-initiator or lipid hydroperoxide induced lipid peroxidation. Free Radic Res 2020; 54:301-310. [PMID: 32338088 DOI: 10.1080/10715762.2020.1761020] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
Lipid peroxidation (LPO) is reported to be involved in the pathogenesis of several oxidative diseases, and several therapeutic approaches using antioxidants have been proposed. LPO is thought to progress via a complicated series of multistep reactions suggesting that the activity of each antioxidant may be different, and depends on the reacting molecules. Hence, in this study, we evaluated the inhibitory mechanisms of several antioxidants toward arachidonic acid (AA) peroxidation induced by the azo initiator 2,2'-azobis(2-amidinopropane) dihydrochloride (AAPH) or a lipid hydroperoxide, hydroperoxyoctadecadienoic acid (HpODE)/hemin. Edaravone, ferrostatin-1, TEMPO and trolox effectively inhibited the production of malondialdehyde (MDA) and several oxidised AAs generated in the AAPH-induced LPO because of their scavenging ability toward lipid peroxyl radicals. In contrast, ebselen and ferrostatin-1 showed strong antioxidative activity in the HpODE/hemin-induced peroxidation. Under this condition, ebselen and ferrostatin-1 were thought to reduce HpODE and its derived alkoxyl radicals to the corresponding lipid alcohols. In conclusion, we found that each antioxidant had different antioxidative activities that prevented the progression of LPO. We expect that these findings will contribute to the design of novel therapeutic strategies using an appropriate antioxidant targeted to each step of the development of oxidative stress diseases.
Collapse
Affiliation(s)
- Kota Saito
- Physical Chemistry for Life Science Laboratory, Faculty of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| | - Yuta Matsuoka
- Physical Chemistry for Life Science Laboratory, Faculty of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan.,AMED-CREST, Japan Agency for Medical Research and Development, Tokyo, Japan
| | - Ken-Ichi Yamada
- Physical Chemistry for Life Science Laboratory, Faculty of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan.,AMED-CREST, Japan Agency for Medical Research and Development, Tokyo, Japan
| |
Collapse
|
39
|
Matsuoka Y, Izumi Y, Takahashi M, Bamba T, Yamada KI. Method for Structural Determination of Lipid-Derived Radicals. Anal Chem 2020; 92:6993-7002. [DOI: 10.1021/acs.analchem.0c00053] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Yuta Matsuoka
- Physical Chemistry for Life Science Laboratory, Faculty of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
- AMED-CREST, Japan Agency for Medical Research and Development, 1-7-1 Ohtemachi, Chiyoda-ku, Tokyo 100-0004, Japan
| | - Yoshihiro Izumi
- AMED-CREST, Japan Agency for Medical Research and Development, 1-7-1 Ohtemachi, Chiyoda-ku, Tokyo 100-0004, Japan
- Division of Metabolomics, Medical Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Masatomo Takahashi
- Division of Metabolomics, Medical Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Takeshi Bamba
- AMED-CREST, Japan Agency for Medical Research and Development, 1-7-1 Ohtemachi, Chiyoda-ku, Tokyo 100-0004, Japan
- Division of Metabolomics, Medical Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Ken-ichi Yamada
- Physical Chemistry for Life Science Laboratory, Faculty of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
- AMED-CREST, Japan Agency for Medical Research and Development, 1-7-1 Ohtemachi, Chiyoda-ku, Tokyo 100-0004, Japan
| |
Collapse
|
40
|
Yamasaki T, Azuma R, Sano K, Munekane M, Matsuoka Y, Yamada KI, Mukai T. Radioiodinated Nitroxide Derivative for the Detection of Lipid Radicals. ACS Med Chem Lett 2020; 11:45-48. [PMID: 31938462 DOI: 10.1021/acsmedchemlett.9b00416] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Accepted: 12/03/2019] [Indexed: 11/29/2022] Open
Abstract
Thus far, no accurate measurement technology has been developed to detect lipid alkyl radicals (lipid radicals), which cause lipid peroxidation. Therefore, we aimed to develop a nuclear medical imaging probe that can be taken up in the lipophilic site in cells such as biological membranes, by reacting specifically with the lipid radicals generated there. We designed and synthesized 4-(4-[125I]iodobenzamido)-2,2,6,6-tetramethylpiperidine-1-oxyl, which shows high reactivity to lipid radicals with a high radiochemical yield and purity. Intracellular retention was found to increase significantly when lipid radicals were produced.
Collapse
Affiliation(s)
- Toshihide Yamasaki
- Laboratory of Biophysical Chemistry, Kobe Pharmaceutical University, 4-19-1 Motoyama-kita, Higashinada, Kobe 658-8558, Japan
| | - Risa Azuma
- Laboratory of Biophysical Chemistry, Kobe Pharmaceutical University, 4-19-1 Motoyama-kita, Higashinada, Kobe 658-8558, Japan
| | - Kohei Sano
- Laboratory of Biophysical Chemistry, Kobe Pharmaceutical University, 4-19-1 Motoyama-kita, Higashinada, Kobe 658-8558, Japan
| | - Masayuki Munekane
- Laboratory of Biophysical Chemistry, Kobe Pharmaceutical University, 4-19-1 Motoyama-kita, Higashinada, Kobe 658-8558, Japan
| | - Yuta Matsuoka
- Physical Chemistry for Life Science Laboratory, Faculty of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Ken-ichi Yamada
- Physical Chemistry for Life Science Laboratory, Faculty of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
- AMED-CREST, Japan Agency for Medical Research and Development, Tokyo 100-0004, Japan
| | - Takahiro Mukai
- Laboratory of Biophysical Chemistry, Kobe Pharmaceutical University, 4-19-1 Motoyama-kita, Higashinada, Kobe 658-8558, Japan
| |
Collapse
|
41
|
Mishima E, Sato E, Ito J, Yamada KI, Suzuki C, Oikawa Y, Matsuhashi T, Kikuchi K, Toyohara T, Suzuki T, Ito S, Nakagawa K, Abe T. Drugs Repurposed as Antiferroptosis Agents Suppress Organ Damage, Including AKI, by Functioning as Lipid Peroxyl Radical Scavengers. J Am Soc Nephrol 2019; 31:280-296. [PMID: 31767624 DOI: 10.1681/asn.2019060570] [Citation(s) in RCA: 117] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Accepted: 10/17/2019] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Ferroptosis, nonapoptotic cell death mediated by free radical reactions and driven by the oxidative degradation of lipids, is a therapeutic target because of its role in organ damage, including AKI. Ferroptosis-causing radicals that are targeted by ferroptosis suppressors have not been unequivocally identified. Because certain cytochrome P450 substrate drugs can prevent lipid peroxidation via obscure mechanisms, we evaluated their antiferroptotic potential and used them to identify ferroptosis-causing radicals. METHODS Using a cell-based assay, we screened cytochrome P450 substrate compounds to identify drugs with antiferroptotic activity and investigated the underlying mechanism. To evaluate radical-scavenging activity, we used electron paramagnetic resonance-spin trapping methods and a fluorescence probe for lipid radicals, NBD-Pen, that we had developed. We then assessed the therapeutic potency of these drugs in mouse models of cisplatin-induced AKI and LPS/galactosamine-induced liver injury. RESULTS We identified various US Food and Drug Administration-approved drugs and hormones that have antiferroptotic properties, including rifampicin, promethazine, omeprazole, indole-3-carbinol, carvedilol, propranolol, estradiol, and thyroid hormones. The antiferroptotic drug effects were closely associated with the scavenging of lipid peroxyl radicals but not significantly related to interactions with other radicals. The elevated lipid peroxyl radical levels were associated with ferroptosis onset, and known ferroptosis suppressors, such as ferrostatin-1, also functioned as lipid peroxyl radical scavengers. The drugs exerted antiferroptotic activities in various cell types, including tubules, podocytes, and renal fibroblasts. Moreover, in mice, the drugs ameliorated AKI and liver injury, with suppression of tissue lipid peroxidation and decreased cell death. CONCLUSIONS Although elevated lipid peroxyl radical levels can trigger ferroptosis onset, some drugs that scavenge lipid peroxyl radicals can help control ferroptosis-related disorders, including AKI.
Collapse
Affiliation(s)
- Eikan Mishima
- Divisions of Nephrology, Endocrinology, and Vascular Medicine and
| | - Emiko Sato
- Divisions of Nephrology, Endocrinology, and Vascular Medicine and.,Department of Clinical Pharmacology and Therapeutics, Tohoku University Graduate School of Pharmaceutical Sciences, Sendai, Japan
| | - Junya Ito
- Food and Biodynamic Chemistry Laboratory, Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
| | - Ken-Ichi Yamada
- Physical Chemistry for Life Science Laboratory, Faculty of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| | - Chitose Suzuki
- Divisions of Nephrology, Endocrinology, and Vascular Medicine and
| | | | | | - Koichi Kikuchi
- Divisions of Nephrology, Endocrinology, and Vascular Medicine and
| | | | - Takehiro Suzuki
- Divisions of Nephrology, Endocrinology, and Vascular Medicine and
| | - Sadayoshi Ito
- Divisions of Nephrology, Endocrinology, and Vascular Medicine and.,Katta Public General Hospital, Shiroishi, Japan; and
| | - Kiyotaka Nakagawa
- Food and Biodynamic Chemistry Laboratory, Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
| | - Takaaki Abe
- Divisions of Nephrology, Endocrinology, and Vascular Medicine and.,Department of Medical Science, Tohoku University Graduate School of Biomedical Engineering, Sendai, Japan.,Department of Clinical Biology and Hormonal Regulation, Tohoku University Graduate School of Medicine, Sendai, Japan
| |
Collapse
|
42
|
Sakuma M, Fuchi Y, Usui K, Karasawa S. Photophysical Properties of Emissive Pyrido[3,2‐
c
]carbazole Derivatives and Apoptosis Induction: Development towards Theranostic Agents in Response to Light Stimulus. Chem Asian J 2019; 14:3938-3945. [DOI: 10.1002/asia.201901200] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Indexed: 12/12/2022]
Affiliation(s)
- Masaomi Sakuma
- Faculty of Pharmaceutical Sciences Showa Pharmaceutical University 3-3165 Higashi-Tamagawagakuen Machida Tokyo 194-8543 Japan
| | - Yasufumi Fuchi
- Faculty of Pharmaceutical Sciences Showa Pharmaceutical University 3-3165 Higashi-Tamagawagakuen Machida Tokyo 194-8543 Japan
| | - Kazuteru Usui
- Faculty of Pharmaceutical Sciences Showa Pharmaceutical University 3-3165 Higashi-Tamagawagakuen Machida Tokyo 194-8543 Japan
| | - Satoru Karasawa
- Faculty of Pharmaceutical Sciences Showa Pharmaceutical University 3-3165 Higashi-Tamagawagakuen Machida Tokyo 194-8543 Japan
| |
Collapse
|
43
|
Yuan Z, Xu M, Wu T, Zhang X, Shen Y, Ernest U, Gui L, Wang F, He Q, Chen H. Design and synthesis of NQO1 responsive fluorescence probe and its application in bio-imaging for cancer diagnosis. Talanta 2019; 198:323-329. [PMID: 30876568 DOI: 10.1016/j.talanta.2019.02.009] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Revised: 01/27/2019] [Accepted: 02/03/2019] [Indexed: 12/31/2022]
Abstract
As an over-expressed flavoprotein in several kinds of tumor cells, NAD(P)H: quinone oxidoreductase-1 (NQO1) is considered as a potent biomarker in early-stage cancer diagnosis. Developing a fast, selective and sensitive method of monitoring NQO1 on cellular level will greatly promote cancer diagnosis in clinical practice. In this paper, a fast NQO1 responsive fluorescence probe SYZ-30 containing quinone acid and 7-nitro-2,1,3-benzoxadiazole (NBD) fluorophore is constructed. The probe could selectively respond to NQO1 and rapidly emit strong fluorescence in vitro within only 5 min. Notably, the peak fluorescent intensities at 550 nm showed a linear relationship with NQO1 concentrations in the range of 3-30 ng/ml and limit of detection (LOD) was 0.0667 ng/ml. Furthermore, it was validated that the probe has good biocompatibility and could be applied for bio-imaging in NQO1 over-expressed cancer cells, together with its mitochondria targeting ability. Importantly, confocal fluorescence imaging confirmed the NQO1 detection ability on cellular level, which can be used for real-time detection of several cancer subtypes like adenocarcinoma. To conclude, the probe is rapidly responsive, highly sensitive and selective which will potentially become a practical tool for early cancer detection and diagnosis.
Collapse
Affiliation(s)
- Zhenwei Yuan
- Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, 24 Tongjia Lane, Gulou District, Nanjing 210009, China
| | - Mingjun Xu
- Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, 24 Tongjia Lane, Gulou District, Nanjing 210009, China
| | - Tianze Wu
- High School Affiliated to Nanjing University, 83 Gulou Street, Gulou District, Nanjing 210009, China
| | - Xinyi Zhang
- Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, 24 Tongjia Lane, Gulou District, Nanjing 210009, China
| | - Yuanzhi Shen
- Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, 24 Tongjia Lane, Gulou District, Nanjing 210009, China
| | - Umeorah Ernest
- Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, 24 Tongjia Lane, Gulou District, Nanjing 210009, China
| | - Lijuan Gui
- Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, 24 Tongjia Lane, Gulou District, Nanjing 210009, China
| | - Fei Wang
- Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, 24 Tongjia Lane, Gulou District, Nanjing 210009, China
| | - Qing He
- Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, 24 Tongjia Lane, Gulou District, Nanjing 210009, China
| | - Haiyan Chen
- Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, 24 Tongjia Lane, Gulou District, Nanjing 210009, China.
| |
Collapse
|
44
|
Greene LE, Lincoln R, Cosa G. Spatio-temporal monitoring of lipid peroxyl radicals in live cell studies combining fluorogenic antioxidants and fluorescence microscopy methods. Free Radic Biol Med 2018; 128:124-136. [PMID: 29649566 DOI: 10.1016/j.freeradbiomed.2018.04.006] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Revised: 04/05/2018] [Accepted: 04/06/2018] [Indexed: 12/28/2022]
Abstract
Lipid peroxidation of polyunsaturated fatty acids in cells may occur via their catalytic autoxidation through peroxyl radicals under oxidative stress conditions. Lipid peroxidation is related to a number of pathologies, and may be invoked in new forms of regulated cell death, yet it may also have beneficial roles in cell signaling cascades. Antioxidants are a natural line of defense against lipid peroxidation, and may accordingly impact the biological outcome associated with the redox chemistry of lipid peroxidation. Critical to unraveling the physiological and pathological role of lipid peroxidation is the development of novel probes with the partition, chemical sensitivity and more importantly, molecular specificity, enabling the spatial and temporal imaging of peroxyl radicals in the lipid membranes of live cells, reporting on the redox status of the cell membrane. This review describes our recent progress to visualize lipid peroxidation in model membrane systems and in live cell studies. Our work portrays the mechanistic insight leading to the development of a highly sensitive probe to monitor lipid peroxyl radicals (LOO•). It also describes technical aspects including reagents and fluorescence microscopy methodologies to consider in order to achieve the much sought after monitoring of rates of lipid peroxyl radical production in live cell studies, be it under oxidative stress but also under cell homeostasis. This review seeks to bring attention to the study of lipid redox reactions and to lay the groundwork for the adoption of fluorogenic antioxidant probeshancement and maximum intensity recorded in turn provide a benchmark to estimate, when compared to the control BODIPY dye lacking the intramolecular PeT based switch, the overall exte and related fluorescence microscopy methods toward gaining rich spatiotemporal information on lipid peroxidation in live cells.
Collapse
Affiliation(s)
- Lana E Greene
- Department of Chemistry and Quebec Center for Advanced Materials (QCAM/CQMF), McGill University, 801 Sherbrooke Street West, Montreal, QC, Canada H3A 0B8
| | - Richard Lincoln
- Department of Chemistry and Quebec Center for Advanced Materials (QCAM/CQMF), McGill University, 801 Sherbrooke Street West, Montreal, QC, Canada H3A 0B8
| | - Gonzalo Cosa
- Department of Chemistry and Quebec Center for Advanced Materials (QCAM/CQMF), McGill University, 801 Sherbrooke Street West, Montreal, QC, Canada H3A 0B8.
| |
Collapse
|
45
|
Haley HMS, Hill AG, Greenwood AI, Woerly EM, Rienstra CM, Burke MD. Peridinin Is an Exceptionally Potent and Membrane-Embedded Inhibitor of Bilayer Lipid Peroxidation. J Am Chem Soc 2018; 140:15227-15240. [PMID: 30388000 PMCID: PMC6452872 DOI: 10.1021/jacs.8b06933] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Antilipoperoxidant protein dysfunction is associated with many human diseases, suggesting that bilayer lipid peroxidation may contribute broadly to pathogenesis. Small molecule inhibitors of this membrane-localized chemistry could in theory enable better understanding and/or treatment of such diseases, but currently available compounds have important limitations. Many biological questions thus remain unanswered, and clinical trials have largely been disappointing. Enabled by efficient, building block-based syntheses of three atypical carotenoid natural products produced by microorganisms that thrive in environments of extreme oxidative stress, we found that peridinin is a potent inhibitor of nonenzymatic bilayer lipid peroxidation in liposomes and in primary human endothelial cells. We also found that peridinin blocks monocyte-endothelial cell adhesion, a key step in atherogenesis. A series of frontier solid-state NMR experiments with a site-specifically 13C-labeled isotopolog synthesized using the same MIDA boronate building block-based total synthesis approach revealed that peridinin is completely embedded within and physically spans the hydrophobic core of POPC membranes, maximizing its effective molarity at the site of the targeted lipid peroxidation reactions. Alternatively, the widely used carotenoid astaxanthin is significantly less potent and was found to primarily localize extramembranously. Peridinin thus represents a promising and biophysically well-characterized starting point for the development of small molecule antilipoperoxidants that serve as more effective biological probes and/or therapeutics.
Collapse
Affiliation(s)
- Hannah M. S. Haley
- Department of Chemistry, University of Illinois at Urbana–Champaign, Urbana, Illinois 61801, United States
| | - Adam G. Hill
- Department of Chemistry, University of Illinois at Urbana–Champaign, Urbana, Illinois 61801, United States
| | - Alexander I. Greenwood
- Department of Chemistry, University of Illinois at Urbana–Champaign, Urbana, Illinois 61801, United States
- Nuclear Magnetic Resonance (NMR) Facility in Applied Science and Physics, William & Mary, Williamsburg, Virginia 23185, United States (A.I.G.)
| | - Eric M. Woerly
- Department of Chemistry, University of Illinois at Urbana–Champaign, Urbana, Illinois 61801, United States
- Eli Lilly and Company, Indianapolis, Indiana 46225, United States (E.M.W.)
| | - Chad M. Rienstra
- Department of Chemistry, University of Illinois at Urbana–Champaign, Urbana, Illinois 61801, United States
- Department of Biochemistry, University of Illinois at Urbana–Champaign, Urbana, Illinois 61801, United States
- Center for Biophysics and Computational Biology, University of Illinois at Urbana–Champaign, Urbana, Illinois 61801, United States
| | - Martin D. Burke
- Department of Chemistry, University of Illinois at Urbana–Champaign, Urbana, Illinois 61801, United States
- Department of Biochemistry, University of Illinois at Urbana–Champaign, Urbana, Illinois 61801, United States
- Carle Illinois College of Medicine, University of Illinois at Urbana–Champaign, Champaign, Illinois 61821, United States
| |
Collapse
|
46
|
Dharmarwardana M, Martins AF, Chen Z, Palacios PM, Nowak CM, Welch RP, Li S, Luzuriaga MA, Bleris L, Pierce BS, Sherry AD, Gassensmith JJ. Nitroxyl Modified Tobacco Mosaic Virus as a Metal-Free High-Relaxivity MRI and EPR Active Superoxide Sensor. Mol Pharm 2018; 15:2973-2983. [PMID: 29771534 PMCID: PMC6078806 DOI: 10.1021/acs.molpharmaceut.8b00262] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Superoxide overproduction is known to occur in multiple disease states requiring critical care; yet, noninvasive detection of superoxide in deep tissue remains a challenge. Herein, we report a metal-free magnetic resonance imaging (MRI) and electron paramagnetic resonance (EPR) active contrast agent prepared by "click conjugating" paramagnetic organic radical contrast agents (ORCAs) to the surface of tobacco mosaic virus (TMV). While ORCAs are known to be reduced in vivo to an MRI/EPR silent state, their oxidation is facilitated specifically by reactive oxygen species-in particular, superoxide-and are largely unaffected by peroxides and molecular oxygen. Unfortunately, single molecule ORCAs typically offer weak MRI contrast. In contrast, our data confirm that the macromolecular ORCA-TMV conjugates show marked enhancement for T1 contrast at low field (<3.0 T) and T2 contrast at high field (9.4 T). Additionally, we demonstrated that the unique topology of TMV allows for a "quenchless fluorescent" bimodal probe for concurrent fluorescence and MRI/EPR imaging, which was made possible by exploiting the unique inner and outer surface of the TMV nanoparticle. Finally, we show TMV-ORCAs do not respond to normal cellular respiration, minimizing the likelihood for background, yet still respond to enzymatically produced superoxide in complicated biological fluids like serum.
Collapse
Affiliation(s)
- Madushani Dharmarwardana
- Department of Chemistry and Biochemistry, University of Texas at Dallas, 800 West Campbell Road, Richardson, TX 75080-3021, USA
| | - André F. Martins
- Department of Chemistry and Biochemistry, University of Texas at Dallas, 800 West Campbell Road, Richardson, TX 75080-3021, USA
- Advanced Imaging Research Center, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, Texas 75390, USA
| | - Zhuo Chen
- Department of Chemistry and Biochemistry, University of Texas at Dallas, 800 West Campbell Road, Richardson, TX 75080-3021, USA
| | - Philip M. Palacios
- Department of Chemistry and Biochemistry, College of Sciences, The University of Texas at Arlington, Arlington, Texas 76019, USA
| | - Chance M. Nowak
- Department of Biological Sciences, University of Texas at Dallas, 800 West Campbell Road, Richardson, TX 75080-3021, USA
| | - Raymond P. Welch
- Department of Chemistry and Biochemistry, University of Texas at Dallas, 800 West Campbell Road, Richardson, TX 75080-3021, USA
| | - Shaobo Li
- Department of Chemistry and Biochemistry, University of Texas at Dallas, 800 West Campbell Road, Richardson, TX 75080-3021, USA
| | - Michael A. Luzuriaga
- Department of Chemistry and Biochemistry, University of Texas at Dallas, 800 West Campbell Road, Richardson, TX 75080-3021, USA
| | - Leonidas Bleris
- Department of Biological Sciences, University of Texas at Dallas, 800 West Campbell Road, Richardson, TX 75080-3021, USA
| | - Brad S. Pierce
- Department of Chemistry and Biochemistry, College of Sciences, The University of Texas at Arlington, Arlington, Texas 76019, USA
| | - A. Dean Sherry
- Department of Chemistry and Biochemistry, University of Texas at Dallas, 800 West Campbell Road, Richardson, TX 75080-3021, USA
- Advanced Imaging Research Center, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, Texas 75390, USA
| | - Jeremiah J. Gassensmith
- Department of Chemistry and Biochemistry, University of Texas at Dallas, 800 West Campbell Road, Richardson, TX 75080-3021, USA
- Department of Bioengineering, University of Texas at Dallas, 800 West Campbell Road, Richardson, TX 75080-3021, USA
| |
Collapse
|
47
|
Griesser M, Shah R, Van Kessel AT, Zilka O, Haidasz EA, Pratt DA. The Catalytic Reaction of Nitroxides with Peroxyl Radicals and Its Relevance to Their Cytoprotective Properties. J Am Chem Soc 2018; 140:3798-3808. [PMID: 29451786 DOI: 10.1021/jacs.8b00998] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Sterically-hindered nitroxides such as 2,2,6,6-tetramethylpiperidin- N-oxyl (TEMPO) have long been ascribed antioxidant activity that is thought to underlie their chemopreventive and anti-aging properties. However, the most commonly invoked reactions in this context-combination with an alkyl radical to give a redox inactive alkoxyamine or catalysis of superoxide dismutation-are unlikely to be relevant under (most) physiological conditions. Herein, we characterize the kinetics and mechanisms of the reactions of TEMPO, as well as an N-arylnitroxide and an N, N-diarylnitroxide, with alkylperoxyl radicals, the propagating species in lipid peroxidation. In each of aqueous solution and lipid bilayers, they are found to be significantly more reactive than Vitamin E, Nature's premier radical-trapping antioxidant (RTA). Inhibited autoxidations of THF in aqueous buffers reveal that nitroxides reduce peroxyl radicals by electron transfer with rate constants ( k ≈ 106 to >107 M-1 s-1) that correlate with the standard potentials of the nitroxides ( E° ≈ 0.75-0.95 V vs NHE) and that this activity is catalytic in nitroxide. Regeneration of the nitroxide occurs by a two-step process involving hydride transfer from the substrate to the nitroxide-derived oxoammonium ion followed by H-atom transfer from the resultant hydroxylamine to a peroxyl radical. This reactivity extends from aqueous solution to phosphatidylcholine liposomes, where added NADPH can be used as a hydride donor to promote nitroxide recycling, as well as to cell culture, where the nitroxides are shown to be potent inhibitors of lipid peroxidation-associated cell death (ferroptosis). These insights have enabled the identification of the most potent nitroxide RTA and anti-ferroptotic agent yet described: phenoxazine- N-oxyl.
Collapse
Affiliation(s)
- Markus Griesser
- Department of Chemistry and Biomolecular Sciences , University of Ottawa , Ottawa , Ontario K1N 6N5 , Canada
| | - Ron Shah
- Department of Chemistry and Biomolecular Sciences , University of Ottawa , Ottawa , Ontario K1N 6N5 , Canada
| | - Antonius T Van Kessel
- Department of Chemistry and Biomolecular Sciences , University of Ottawa , Ottawa , Ontario K1N 6N5 , Canada
| | - Omkar Zilka
- Department of Chemistry and Biomolecular Sciences , University of Ottawa , Ottawa , Ontario K1N 6N5 , Canada
| | - Evan A Haidasz
- Department of Chemistry and Biomolecular Sciences , University of Ottawa , Ottawa , Ontario K1N 6N5 , Canada
| | - Derek A Pratt
- Department of Chemistry and Biomolecular Sciences , University of Ottawa , Ottawa , Ontario K1N 6N5 , Canada
| |
Collapse
|
48
|
Shinto S, Matsuoka Y, Yamato M, Yamada KI. Antioxidant nitroxides protect hepatic cells from oxidative stress-induced cell death. J Clin Biochem Nutr 2018; 62:132-138. [PMID: 29610552 PMCID: PMC5874234 DOI: 10.3164/jcbn.17-60] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Accepted: 11/09/2017] [Indexed: 12/21/2022] Open
Abstract
Oxidative stress causes cell death and induces many kinds of disease, including liver disease. Nitroxides are known to react catalytically with free radicals. In this study, the cell protective activities of nitroxides were compared with those of other antioxidants. Nitroxides showed much greater inhibition of hydrogen peroxide-induced cell death than other antioxidants in a hepatic cell line and in primary hepatocytes. The intracellular oxidative stress level at 24 h after hydrogen peroxide stimulation was significantly decreased by nitroxides, but not by other antioxidants. To clarify the mechanism of cell protection by nitroxides, we investigated whether nitroxides inhibited DNA damage and mitogen-activated protein kinase pathway activation. We found that nitroxides reduced caspase-3 activation and may have ultimately inhibited cell death. In conclusion, nitroxides are very useful for attenuating cell damage due to oxidative stress. Nitroxides are thus a potential therapeutic agent for oxidative stress-related diseases.
Collapse
Affiliation(s)
- Saki Shinto
- Physical Chemistry for Life Science Laboratory, Faculty of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi Higashi-ku, Fukuoka 812-8582, Japan
| | - Yuta Matsuoka
- Physical Chemistry for Life Science Laboratory, Faculty of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi Higashi-ku, Fukuoka 812-8582, Japan
| | - Mayumi Yamato
- Physical Chemistry for Life Science Laboratory, Faculty of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi Higashi-ku, Fukuoka 812-8582, Japan
| | - Ken-Ichi Yamada
- Physical Chemistry for Life Science Laboratory, Faculty of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi Higashi-ku, Fukuoka 812-8582, Japan
| |
Collapse
|
49
|
Hirayama A, Oowada S, Ito H, Matsui H, Ueda A, Aoyagi K. Clinical significance of redox effects of Kampo formulae, a traditional Japanese herbal medicine: comprehensive estimation of multiple antioxidative activities. J Clin Biochem Nutr 2017; 62:39-48. [PMID: 29371753 PMCID: PMC5773833 DOI: 10.3164/jcbn.17-59] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Accepted: 08/27/2017] [Indexed: 12/27/2022] Open
Abstract
To clarify the clinical significance of the redox-controlling effects of Kampo, a traditional Japanese herbal medicine, we determined the scavenging activities of various reactive oxygen species in clinically used Kampo formulae using an electron spin resonance-based technique. Formulae containing Rhei Rhizoma (i.e., mashiningan and daiobotanpito) showed high scavenging activity against the alkoxyl radical, and crude extract quantity was significantly correlated with scavenging activity. Hydroxyl radical scavenging activity was positively correlated with the quantity of Zingiberis Rhizoma. Strong hydroxyl radical scavenging activity was also found in formulae containing both Bupleuri Radix and Scutellariae Radix, a widely used anti-inflammatory combination. Formulae containing a clinically common combination of Scutellariae Radix, Coptidis Rhizoma, and Phellodendri Cortex induced high superoxide scavenging activity. Singlet oxygen scavenging activity was high in formulae containing Bupleuri Radix and Glycyrrhizae Radix. In contrast, formulae containing Rehmanniae Radix showed generally low reactive oxygen species scavenging activities, and the quantity of Rehmanniae Radix was negatively correlated with hydroxyl radical and singlet oxygen scavenging activities. These results indicate that the antioxidative effects of Kampo formulae are not uniform but complexly varied against multiple reactive oxygen species. Some formulae have almost no antioxidant effects but may act as pro-oxidants.
Collapse
Affiliation(s)
- Aki Hirayama
- Center for Integrative Medicine, Tsukuba University of Technology, 4-12-7 Kasuga, Tsukuba, Ibaraki 305-8521, Japan
| | - Shigeru Oowada
- Asao Clinic, 1-8-10 Manpukuji, Kawasaki, Kanagawa 215-0004, Japan
| | - Hiromu Ito
- Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan
| | - Hirofumi Matsui
- Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan
| | - Atsushi Ueda
- Tsukuba University Hospital Hitachi Medical Education and Research Center, 2-1-1 Jyonancho, Hitachi, Ibaraki 317-0077, Japan
| | - Kazumasa Aoyagi
- Center for Integrative Medicine, Tsukuba University of Technology, 4-12-7 Kasuga, Tsukuba, Ibaraki 305-8521, Japan
| |
Collapse
|
50
|
Jabeen E, Janjua NK, Ahmed S, Domínguez-Álvarez E, Jacob C. A selective and sensitive monitoring of the OH radical using flavonoid-modified electrodes. Electrochim Acta 2017. [DOI: 10.1016/j.electacta.2017.10.065] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|