1
|
Roh Y, Lee SB, Kim M, Kim MH, Kim HJ, Cho KO. Alleviation of hippocampal necroptosis and neuroinflammation by NecroX-7 treatment after acute seizures. Front Pharmacol 2023; 14:1187819. [PMID: 37601059 PMCID: PMC10433749 DOI: 10.3389/fphar.2023.1187819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Accepted: 07/24/2023] [Indexed: 08/22/2023] Open
Abstract
Temporal lobe epilepsy (TLE) is one of the most common neurological disorders, but still one-third of patients cannot be properly treated by current medication. Thus, we investigated the therapeutic effects of a novel small molecule, NecroX-7, in TLE using both a low [Mg2+]o-induced epileptiform activity model and a mouse model of pilocarpine-induced status epilepticus (SE). NecroX-7 post-treatment enhanced the viability of primary hippocampal neurons exposed to low [Mg2+]o compared to controls in an MTT assay. Application of NecroX-7 after pilocarpine-induced SE also reduced the number of degenerating neurons labelled with Fluoro-Jade B. Immunocytochemistry and immunohistochemistry showed that NecroX-7 post-treatment significantly alleviated ionized calcium-binding adaptor molecule 1 (Iba1) intensity and immunoreactive area, while the attenuation of reactive astrocytosis by glial fibrillary acidic protein (GFAP) staining was observed in cultured hippocampal neurons. However, NecroX-7-mediated morphologic changes of astrocytes were seen in both in vitro and in vivo models of TLE. Finally, western blot analysis demonstrated that NecroX-7 post-treatment after acute seizures could decrease the expression of mixed lineage kinase domain-like pseudokinase (MLKL) and phosphorylated MLKL (p-MLKL), markers for necroptosis. Taken all together, NecroX-7 has potential as a novel medication for TLE with its neuroprotective, anti-inflammatory, and anti-necroptotic effects.
Collapse
Affiliation(s)
- Yihyun Roh
- College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Su Bin Lee
- Department of Medical Laser, Graduate School, Dankook University, Cheonan, Republic of Korea
| | - Minseo Kim
- College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Mi-Hye Kim
- Department of Medical Laser, Graduate School, Dankook University, Cheonan, Republic of Korea
| | - Hee Jung Kim
- Department of Physiology, College of Medicine, Center for Human Risk Assessment, Dankook University, Cheonan, Republic of Korea
| | - Kyung-Ok Cho
- Department of Pharmacology, Catholic Neuroscience Institute, Institute for Aging and Metabolic Diseases, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- Department of Biomedicine and Health Sciences, The Catholic University of Korea, Seoul, Republic of Korea
| |
Collapse
|
2
|
Cienfuegos-Pecina E, Moreno-Peña DP, Torres-González L, Rodríguez-Rodríguez DR, Garza-Villarreal D, Mendoza-Hernández OH, Flores-Cantú RA, Samaniego Sáenz BA, Alarcon-Galvan G, Muñoz-Espinosa LE, Ibarra-Rivera TR, Saucedo AL, Cordero-Pérez P. Treatment with sodium ( S)-2-hydroxyglutarate prevents liver injury in an ischemia-reperfusion model in female Wistar rats. PeerJ 2021; 9:e12426. [PMID: 34824916 PMCID: PMC8592047 DOI: 10.7717/peerj.12426] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Accepted: 10/11/2021] [Indexed: 12/24/2022] Open
Abstract
Background Ischemia-reperfusion (IR) injury is one of the leading causes of early graft dysfunction in liver transplantation. Techniques such as ischemic preconditioning protect the graft through the activation of the hypoxia-inducible factors (HIF), which are downregulated by the EGLN family of prolyl-4-hydroxylases, a potential biological target for the development of strategies based on pharmacological preconditioning. For that reason, this study aims to evaluate the effect of the EGLN inhibitor sodium (S)-2-hydroxyglutarate [(S)-2HG] on liver IR injury in Wistar rats. Methods Twenty-eight female Wistar rats were divided into the following groups: sham (SH, n = 7), non-toxicity (HGTox, n = 7, 25 mg/kg of (S)-2HG, twice per day for two days), IR (n = 7, total liver ischemia: 20 minutes, reperfusion: 60 minutes), and (S)-2HG+IR (HGIR, n = 7, 25 mg/kg of (S)-2HG, twice per day for two days, total liver ischemia as the IR group). Serum ALT, AST, LDH, ALP, glucose, and total bilirubin were assessed. The concentrations of IL-1β, IL-6, TNF, malondialdehyde, superoxide dismutase, and glutathione peroxidase were measured in liver tissue, as well as the expression of Hmox1, Vegfa, and Pdk1, determined by RT-qPCR. Sections of liver tissue were evaluated histologically, assessing the severity of necrosis, sinusoidal congestion, and cytoplasmatic vacuolization. Results The administration of (S)-2HG did not cause any alteration in the assessed biochemical markers compared to SH. Preconditioning with (S)-2HG significantly ameliorated IR injury in the HGIR group, decreasing the serum activities of ALT, AST, and LDH, and the tissue concentrations of IL-1β and IL-6 compared to the IR group. IR injury decreased serum glucose compared to SH. There were no differences in the other biomarkers assessed. The treatment with (S)-2HG tended to decrease the severity of hepatocyte necrosis and sinusoidal congestion compared to the IR group. The administration of (S)-2HG did not affect the expression of Hmox1 but decreased the expression of both Vegfa and Pdk1 compared to the SH group, suggesting that the HIF-1 pathway is not involved in its mechanism of hepatoprotection. In conclusion, (S)-2HG showed a hepatoprotective effect, decreasing the levels of liver injury and inflammation biomarkers, without evidence of the involvement of the HIF-1 pathway. No hepatotoxic effect was observed at the tested dose.
Collapse
Affiliation(s)
- Eduardo Cienfuegos-Pecina
- Universidad Autónoma de Nuevo León. Liver Unit, Department of Internal Medicine, University Hospital "Dr. José E. González", Monterrey, Nuevo León, Mexico.,Universidad Autónoma de Nuevo León. Blood Bank, Department of Clinical Pathology, University Hospital "Dr. José E. González", Monterrey, Nuevo León, Mexico
| | - Diana P Moreno-Peña
- Universidad Autónoma de Nuevo León. Liver Unit, Department of Internal Medicine, University Hospital "Dr. José E. González", Monterrey, Nuevo León, Mexico
| | - Liliana Torres-González
- Universidad Autónoma de Nuevo León. Liver Unit, Department of Internal Medicine, University Hospital "Dr. José E. González", Monterrey, Nuevo León, Mexico
| | - Diana Raquel Rodríguez-Rodríguez
- Universidad Autónoma de Nuevo León. Liver Unit, Department of Internal Medicine, University Hospital "Dr. José E. González", Monterrey, Nuevo León, Mexico
| | - Diana Garza-Villarreal
- Universidad Autónoma de Nuevo León. Liver Unit, Department of Internal Medicine, University Hospital "Dr. José E. González", Monterrey, Nuevo León, Mexico
| | - Oscar H Mendoza-Hernández
- Universidad Autónoma de Nuevo León. Liver Unit, Department of Internal Medicine, University Hospital "Dr. José E. González", Monterrey, Nuevo León, Mexico
| | - Raul Alejandro Flores-Cantú
- Universidad Autónoma de Nuevo León. Liver Unit, Department of Internal Medicine, University Hospital "Dr. José E. González", Monterrey, Nuevo León, Mexico
| | - Brenda Alejandra Samaniego Sáenz
- Universidad Autónoma de Nuevo León. Liver Unit, Department of Internal Medicine, University Hospital "Dr. José E. González", Monterrey, Nuevo León, Mexico
| | - Gabriela Alarcon-Galvan
- Universidad de Monterrey, Basic Science Department, School of Medicine, Monterrey, Nuevo León, Mexico
| | - Linda E Muñoz-Espinosa
- Universidad Autónoma de Nuevo León. Liver Unit, Department of Internal Medicine, University Hospital "Dr. José E. González", Monterrey, Nuevo León, Mexico
| | - Tannya R Ibarra-Rivera
- Universidad Autónoma de Nuevo León. Department of Analytical Chemistry, School of Medicine, Monterrey, Nuevo León, Mexico
| | - Alma L Saucedo
- Universidad Autónoma de Nuevo León. Department of Analytical Chemistry, School of Medicine, Monterrey, Nuevo León, Mexico
| | - Paula Cordero-Pérez
- Universidad Autónoma de Nuevo León. Liver Unit, Department of Internal Medicine, University Hospital "Dr. José E. González", Monterrey, Nuevo León, Mexico
| |
Collapse
|
3
|
Kim G, Lee HS, Oh BJ, Kwon Y, Kim H, Ha S, Jin SM, Kim JH. Protective effect of a novel clinical-grade small molecule necrosis inhibitor against oxidative stress and inflammation during islet transplantation. Am J Transplant 2021; 21:1440-1452. [PMID: 32978875 DOI: 10.1111/ajt.16323] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 08/24/2020] [Accepted: 09/14/2020] [Indexed: 01/25/2023]
Abstract
Inhibition of mitochondrial reactive oxygen species (ROS) and subsequent damage-associated molecular patterns (DAMPs)-induced inflammatory responses could be a novel target in clinical islet transplantation. We investigated the protective effects of NecroX-7, a novel clinical-grade necrosis inhibitor that specifically targets mitochondrial ROS, against primary islet graft failure. Islets from heterozygote human islet amyloid polypeptide transgenic (hIAPP+/- ) mice and nonhuman primates (NHPs) were isolated or cultured with or without NecroX-7 in serum-deprived medium. Supplementation with NecroX-7 during hIAPP+/- mouse islet isolation markedly increased islet viability and adenosine triphosphate content, and attenuated ROS, transcription of c-Jun N-terminal kinases, high mobility group box 1, interleukin-1beta (IL-1 β ), IL-6, and tumor necrosis factor-alpha. Supplementation of NecroX-7 during serum-deprived culture also protected hIAPP+/- mouse and NHP islets against impaired viability, serum deprivation-induced ROS, proinflammatory response, and accumulation of toxic IAPP oligomer. Supplementation with NecroX-7 during isolation or serum-deprived culture of hIAPP+/- mouse and NHP islets also improved posttransplant glycemia in the recipient streptozotocin-induced diabetic hIAPP-/- mice and BALB/c-nu/nu mice, respectively. In conclusion, pretransplant administration of NecroX-7 during islet isolation and serum-deprived culture suppressed mitochondrial ROS injury, generation of DAMPs-induced proinflammatory responses, and accumulation of toxic IAPP oligomers ex vivo, and improved posttransplant glycemia in vivo.
Collapse
Affiliation(s)
- Gyuri Kim
- Division of Endocrinology and Metabolism, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Han Sin Lee
- Division of Endocrinology and Metabolism, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea.,Samsung Biomedical Research Institute, Samsung Medical Center, Seoul, Korea
| | - Bae Jun Oh
- Daegu-Gyeongbuk Medical Innovation Foundation, Daegu, Korea
| | - Youngsang Kwon
- Division of Endocrinology and Metabolism, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea.,Samsung Biomedical Research Institute, Samsung Medical Center, Seoul, Korea.,Department of Health Sciences and Technology, SAIHST (Samsung Advanced Institute for Health Sciences & Technology, Seoul, Korea
| | - Hyunjin Kim
- Division of Endocrinology and Metabolism, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea.,Samsung Biomedical Research Institute, Samsung Medical Center, Seoul, Korea
| | - Seungyeon Ha
- Division of Endocrinology and Metabolism, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea.,Samsung Biomedical Research Institute, Samsung Medical Center, Seoul, Korea.,Department of Health Sciences and Technology, SAIHST (Samsung Advanced Institute for Health Sciences & Technology, Seoul, Korea
| | - Sang-Man Jin
- Division of Endocrinology and Metabolism, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea.,Samsung Biomedical Research Institute, Samsung Medical Center, Seoul, Korea
| | - Jae Hyeon Kim
- Division of Endocrinology and Metabolism, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea.,Samsung Biomedical Research Institute, Samsung Medical Center, Seoul, Korea.,Department of Health Sciences and Technology, SAIHST (Samsung Advanced Institute for Health Sciences & Technology, Seoul, Korea
| |
Collapse
|
4
|
Chaudhary GR, Yadav PK, Yadav AK, Tiwari M, Gupta A, Sharma A, Sahu K, Pandey AN, Pandey AK, Chaube SK. Necrosis and necroptosis in germ cell depletion from mammalian ovary. J Cell Physiol 2018; 234:8019-8027. [PMID: 30341907 DOI: 10.1002/jcp.27562] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Accepted: 09/14/2018] [Indexed: 01/04/2023]
Abstract
The maximum number of germ cells is present during the fetal life in mammals. Follicular atresia results in rapid depletion of germ cells from the cohort of the ovary. At the time of puberty, only a few hundred (<1%) germ cells are either culminated into oocytes or further get eliminated during the reproductive life. Although apoptosis plays a major role, necrosis as well as necroptosis, might also be involved in germ cell elimination from the mammalian ovary. Both necrosis and necroptosis show similar morphological features and are characterized by an increase in cell volume, cell membrane permeabilization, and rupture that lead to cellular demise. Necroptosis is initiated by tumor necrosis factor and operated through receptor interacting protein kinase as well as mixed lineage kinase domain-like protein. The acetylcholinesterase, cytokines, starvation, and oxidative stress play important roles in necroptosis-mediated granulosa cell death. The granulosa cell necroptosis directly or indirectly induces susceptibility toward necroptotic or apoptotic cell death in oocytes. Indeed, prevention of necrosis and necroptosis pathways using their specific inhibitors could enhance growth/differentiation factor-9 expression, improve survivability as well as the meiotic competency of oocytes, and prevent decline of reproductive potential in several mammalian species and early onset of menopause in women. This study updates the information and focuses on the possible involvement of necrosis and necroptosis in germ cell depletion from the mammalian ovary.
Collapse
Affiliation(s)
- Govind R Chaudhary
- Cell Physiology Laboratory, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, India
| | - Pramod K Yadav
- Cell Physiology Laboratory, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, India
| | - Anil K Yadav
- Cell Physiology Laboratory, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, India
| | - Meenakshi Tiwari
- Cell Physiology Laboratory, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, India
| | - Anumegha Gupta
- Cell Physiology Laboratory, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, India
| | - Alka Sharma
- Cell Physiology Laboratory, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, India
| | - Kankshi Sahu
- Cell Physiology Laboratory, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, India
| | - Ashutosh N Pandey
- Cell Physiology Laboratory, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, India
| | - Ajai K Pandey
- Department of Kayachikitsa, Faculty of Ayurveda, Institute of Medical Science, Banaras Hindu University, Varanasi, India
| | - Shail K Chaube
- Cell Physiology Laboratory, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, India
| |
Collapse
|
5
|
Hwang IC, Kim JY, Kim JH, Lee JE, Seo JY, Lee JW, Park J, Yang HM, Kim SH, Cho HJ, Kim HS. Therapeutic Potential of a Novel Necrosis Inhibitor, 7-Amino-Indole, in Myocardial Ischemia-Reperfusion Injury. Hypertension 2018; 71:1143-1155. [PMID: 29661840 PMCID: PMC5959205 DOI: 10.1161/hypertensionaha.117.09405] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2017] [Revised: 04/04/2017] [Accepted: 03/08/2018] [Indexed: 12/20/2022]
Abstract
Supplemental Digital Content is available in the text. Opening of mitochondrial permeability transition pore and Ca2+ overload are main contributors to myocardial ischemia–reperfusion injury, which paradoxically causes a wide variety of myocardial damage. We investigated the protective role of a novel necrosis inhibitor (NecroX-7; NecX) against myocardial ischemia–reperfusion injury using in vitro and in vivo models. H9C2 rat cardiomyoblasts and neonatal cardiomyocytes were exposed to hypoxia–reoxygenation stress after pre-treatment with NecX, vitamin C, a combination of vitamin C and E, N-acetylcysteine, an apoptosis inhibitor (Z-VAD-fmk), or cyclosporine A. The main mechanism of cell death after hypoxia–reoxygenation stress was not apoptosis but necrosis, which was prevented by NecX. Protective effect of NecX was based on its potent reactive oxygen species scavenging activity, especially on mitochondrial reactive oxygen species. NecX preserved mitochondrial membrane potential through prevention of Ca2+ influx and inhibition of mitochondrial permeability transition pore opening, which was more potent than that by cyclosporine A. Using Sprague-Dawley rats exposed to myocardial ischemia for 45 minutes followed by reperfusion, we compared therapeutic efficacies of NecX with cyclosporine A, vitamin C, a combination of vitamin C and E, and 5% dextrose, each administered 5 minutes before reperfusion. NecX markedly inhibited myocardial necrosis and reduced fibrotic area to a greater extent than did cyclosporine A and other treated groups. In addition, NecX preserved systolic function and prevented pathological dilatory remodeling of left ventricle. The novel necrosis inhibitor has a significant protective effect against myocardial ischemia–reperfusion injury through inhibition of mitochondrial permeability transition pore opening, indicating that it is a promising candidate for cardioprotective adjunctive measure on top of reperfusion therapy.
Collapse
Affiliation(s)
- In-Chang Hwang
- From the Cardiovascular Center and Department of Internal Medicine, Seoul National University Hospital, Republic of Korea (I.-C.H., J.-H.K., J.P., H.-M.Y., H.-J.C., H.-S.K.).,National Leading Laboratory for Cardiovascular Stem Cell, Seoul National University College of Medicine, Republic of Korea (I.-C.H., J.-Y.K., J.-H.K., J.-E.L., J.-Y.S., J.-W.L., J.P., H.-M.Y., H.-J.C., H.-S.K.).,Strategic Center of Cell and Bio Therapy for Heart, Diabetes, and Cancer, Seoul National University Hospital, Republic of Korea (I.-C.H., J.-Y.K., J.-E.L., J.-W.L., H.-M.Y., H.-J.C., H.-S.K.)
| | - Ju-Young Kim
- From the Cardiovascular Center and Department of Internal Medicine, Seoul National University Hospital, Republic of Korea (I.-C.H., J.-H.K., J.P., H.-M.Y., H.-J.C., H.-S.K.).,National Leading Laboratory for Cardiovascular Stem Cell, Seoul National University College of Medicine, Republic of Korea (I.-C.H., J.-Y.K., J.-H.K., J.-E.L., J.-Y.S., J.-W.L., J.P., H.-M.Y., H.-J.C., H.-S.K.).,Strategic Center of Cell and Bio Therapy for Heart, Diabetes, and Cancer, Seoul National University Hospital, Republic of Korea (I.-C.H., J.-Y.K., J.-E.L., J.-W.L., H.-M.Y., H.-J.C., H.-S.K.)
| | - Ji-Hyun Kim
- National Leading Laboratory for Cardiovascular Stem Cell, Seoul National University College of Medicine, Republic of Korea (I.-C.H., J.-Y.K., J.-H.K., J.-E.L., J.-Y.S., J.-W.L., J.P., H.-M.Y., H.-J.C., H.-S.K.)
| | - Joo-Eun Lee
- National Leading Laboratory for Cardiovascular Stem Cell, Seoul National University College of Medicine, Republic of Korea (I.-C.H., J.-Y.K., J.-H.K., J.-E.L., J.-Y.S., J.-W.L., J.P., H.-M.Y., H.-J.C., H.-S.K.).,Strategic Center of Cell and Bio Therapy for Heart, Diabetes, and Cancer, Seoul National University Hospital, Republic of Korea (I.-C.H., J.-Y.K., J.-E.L., J.-W.L., H.-M.Y., H.-J.C., H.-S.K.)
| | - Ji-Yun Seo
- National Leading Laboratory for Cardiovascular Stem Cell, Seoul National University College of Medicine, Republic of Korea (I.-C.H., J.-Y.K., J.-H.K., J.-E.L., J.-Y.S., J.-W.L., J.P., H.-M.Y., H.-J.C., H.-S.K.).,Strategic Center of Cell and Bio Therapy for Heart, Diabetes, and Cancer, Seoul National University Hospital, Republic of Korea (I.-C.H., J.-Y.K., J.-E.L., J.-W.L., H.-M.Y., H.-J.C., H.-S.K.)
| | - Jae-Won Lee
- National Leading Laboratory for Cardiovascular Stem Cell, Seoul National University College of Medicine, Republic of Korea (I.-C.H., J.-Y.K., J.-H.K., J.-E.L., J.-Y.S., J.-W.L., J.P., H.-M.Y., H.-J.C., H.-S.K.)
| | - Jonghanne Park
- From the Cardiovascular Center and Department of Internal Medicine, Seoul National University Hospital, Republic of Korea (I.-C.H., J.-H.K., J.P., H.-M.Y., H.-J.C., H.-S.K.).,National Leading Laboratory for Cardiovascular Stem Cell, Seoul National University College of Medicine, Republic of Korea (I.-C.H., J.-Y.K., J.-H.K., J.-E.L., J.-Y.S., J.-W.L., J.P., H.-M.Y., H.-J.C., H.-S.K.)
| | - Han-Mo Yang
- From the Cardiovascular Center and Department of Internal Medicine, Seoul National University Hospital, Republic of Korea (I.-C.H., J.-H.K., J.P., H.-M.Y., H.-J.C., H.-S.K.).,National Leading Laboratory for Cardiovascular Stem Cell, Seoul National University College of Medicine, Republic of Korea (I.-C.H., J.-Y.K., J.-H.K., J.-E.L., J.-Y.S., J.-W.L., J.P., H.-M.Y., H.-J.C., H.-S.K.).,Strategic Center of Cell and Bio Therapy for Heart, Diabetes, and Cancer, Seoul National University Hospital, Republic of Korea (I.-C.H., J.-Y.K., J.-E.L., J.-W.L., H.-M.Y., H.-J.C., H.-S.K.)
| | - Soon-Ha Kim
- R&D Campus, LG Chem/Ltd., Daejeon, Republic of Korea (S.-H.K.)
| | - Hyun-Jai Cho
- From the Cardiovascular Center and Department of Internal Medicine, Seoul National University Hospital, Republic of Korea (I.-C.H., J.-H.K., J.P., H.-M.Y., H.-J.C., H.-S.K.).,National Leading Laboratory for Cardiovascular Stem Cell, Seoul National University College of Medicine, Republic of Korea (I.-C.H., J.-Y.K., J.-H.K., J.-E.L., J.-Y.S., J.-W.L., J.P., H.-M.Y., H.-J.C., H.-S.K.).,Strategic Center of Cell and Bio Therapy for Heart, Diabetes, and Cancer, Seoul National University Hospital, Republic of Korea (I.-C.H., J.-Y.K., J.-E.L., J.-W.L., H.-M.Y., H.-J.C., H.-S.K.)
| | - Hyo-Soo Kim
- From the Cardiovascular Center and Department of Internal Medicine, Seoul National University Hospital, Republic of Korea (I.-C.H., J.-H.K., J.P., H.-M.Y., H.-J.C., H.-S.K.) .,National Leading Laboratory for Cardiovascular Stem Cell, Seoul National University College of Medicine, Republic of Korea (I.-C.H., J.-Y.K., J.-H.K., J.-E.L., J.-Y.S., J.-W.L., J.P., H.-M.Y., H.-J.C., H.-S.K.).,and Department of Molecular Medicine and Biopharmaceutical Science, Seoul National University, Republic of Korea (H.-S.K.).,R&D Campus, LG Chem/Ltd., Daejeon, Republic of Korea (S.-H.K.)
| |
Collapse
|
6
|
Abstract
NecroX-5 is a derivative of cyclopentylamino carboxymethylthiazolylindole (NecroX), an inhibitor of necrosis/necroptosis. NecroX-5 has been shown to scavenge mitochondrial reactive oxygen and nitrogen species, and thus preventing necrotic cell death against various kinds of oxidative stress in several tissues, including the brain. To examine the effect of NecroX-5 on retinal degeneration (RD), RD was induced in Sprague-Dawley rats by an intraperitoneal injection of N-methyl-N-nitrosourea and in BALB/c mice by blue light-emitting diode exposure. Scotopic electroretinography recording was used to evaluate retinal function. For histological evaluation, hematoxylin and eosin staining, terminal deoxynucleotidyl transferase dUTP nick end labeling, and immunohistochemistry were performed. Electroretinography recordings showed that a-waves and b-waves were significantly reduced in both RD rats and mice, whereas the amplitudes of both waves were significantly increased in both NecroX-5-treated RD rats and mice compared with untreated RD animals. In hematoxylin and eosin staining and terminal deoxynucleotidyl transferase dUTP nick end labeling assay, the outer nuclear layer where photoreceptors reside appeared to be more preserved, and there were fewer apoptotic cells in NecroX-5-treated RD retinas than in untreated RD retinas. In addition, immunohistochemistry with antiglial fibrillary acidic protein and anti-8-hydroxy-2'-deoxyguanosine showed lower levels of retinal injury and oxidative stress in NecroX-5-treated RD retinas than in untreated RD retinas. These results indicated that NecroX-5 protects retinal neurons from experimentally induced RD, suggesting that NecroX-5 may have a potential for the treatment of RD as a medication.
Collapse
|
7
|
Mitochondria-Targeted Antioxidants for the Treatment of Cardiovascular Disorders. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 982:621-646. [PMID: 28551810 DOI: 10.1007/978-3-319-55330-6_32] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
8
|
Park JH, Kim HK, Jung H, Kim KH, Kang MS, Hong JH, Yu BC, Park S, Seo SK, Choi IW, Kim SH, Kim N, Han J, Park SG. NecroX-5 prevents breast cancer metastasis by AKT inhibition via reducing intracellular calcium levels. Int J Oncol 2016; 50:185-192. [PMID: 27922686 DOI: 10.3892/ijo.2016.3789] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Accepted: 11/28/2016] [Indexed: 11/06/2022] Open
Abstract
A major goal of breast cancer research is to prevent the molecular events that lead to tumour metastasis. It is well-established that both cytoplasmic and mitochondrial reactive oxygen species (ROS) play important roles in cell migration and metastasis. Accordingly, this study examined the molecular mechanisms of the anti-metastatic effects of NecroX-5, a mitochondrial ROS scavenger. NecroX-5 inhibited lung cancer metastasis by ameliorating migration in a mouse model. In human cancer cells, the inhibition of migration by NecroX-5 is cell type-dependent. We observed that the effect of NecroX-5 correlated with a reduction in mitochondrial ROS, but mitochondrial ROS reduction by MitoQ did not inhibit cell migration. NecroX-5 decreased intracellular calcium concentration by blocking Ca2+ influx, which mediated the inhibition of cell migration, AKT downregulation and the reduction of mitochondrial ROS levels. However, the reduction of mitochondrial ROS was not associated with supressed migration and AKT downregulation. Our study demonstrates the potential of NecroX-5 as an inhibitor of breast cancer metastasis.
Collapse
Affiliation(s)
- Jin-Hee Park
- Department of Microbiology and Immunology, INJE University College of Medicine, Busan 614-735, Republic of Korea
| | - Hyoung Kyu Kim
- Department of Physiology, INJE University College of Medicine, Busan 614-735, Republic of Korea
| | - Hana Jung
- Department of Microbiology and Immunology, INJE University College of Medicine, Busan 614-735, Republic of Korea
| | - Ki Hyang Kim
- Department of Internal Medicine, INJE University College of Medicine, Busan 614-735, Republic of Korea
| | - Mi Seon Kang
- Department of Pathology, INJE University College of Medicine, Busan 614-735, Republic of Korea
| | - Jun Hyuk Hong
- Graduate School, Department of Preventive Medicine, Kosin University College of Medicine, Busan 602-702, Republic of Korea
| | - Byeng Chul Yu
- LG Life Science, Ltd., R&D Park, Daejeon 305-380, Republic of Korea
| | - Sungjae Park
- Department of Internal Medicine, INJE University College of Medicine, Busan 614-735, Republic of Korea
| | - Su-Kil Seo
- Department of Microbiology and Immunology, INJE University College of Medicine, Busan 614-735, Republic of Korea
| | - Il Whan Choi
- Department of Microbiology and Immunology, INJE University College of Medicine, Busan 614-735, Republic of Korea
| | - Soon Ha Kim
- LG Life Science, Ltd., R&D Park, Daejeon 305-380, Republic of Korea
| | - Nari Kim
- Department of Physiology, INJE University College of Medicine, Busan 614-735, Republic of Korea
| | - Jin Han
- Department of Physiology, INJE University College of Medicine, Busan 614-735, Republic of Korea
| | - Sae Gwang Park
- Department of Microbiology and Immunology, INJE University College of Medicine, Busan 614-735, Republic of Korea
| |
Collapse
|
9
|
Lee JH, Park KM, Lee YJ, Kim JH, Kim SH. A New Chemical Compound, NecroX-7, Acts as a Necrosis Modulator by Inhibiting High-Mobility Group Box 1 Protein Release During Massive Ischemia-Reperfusion Injury. Transplant Proc 2016; 48:3406-3414. [PMID: 27931589 DOI: 10.1016/j.transproceed.2016.09.046] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Revised: 08/21/2016] [Accepted: 09/14/2016] [Indexed: 12/13/2022]
Abstract
BACKGROUND Necrotic cell death is common in a wide variety of pathologic conditions, including ischemia-reperfusion (IR) injury. The aim of this study was to develop an IR injury-induced hepatic necrosis model in dogs by means of selective left hepatic inflow occlusion and to test the efficacy of a new chemical compound, NecroX-7, against the IR injury-induced hepatic damage. METHODS A group of male Beagle dogs received intravenous infusions of either vehicle or different doses of NecroX-7 (1.5, 4.5, or 13 mg/kg) for a 20-minute period before a 90-minute left hepatic inflow occlusion followed by reperfusion. RESULTS The gross morphology in the NecroX-7-treated groups after occlusion appeared to be less congested and less swollen than that in vehicle-treated control group. Circulating alanine transaminase and aspartate transaminase levels in the control group were elevated during the course of IR, and were effectively blocked in the 4.5 and 13 mg/kg NecroX-7-treated groups. The serum levels of high-mobility group box 1 protein showed a peak at 8 hours after occlusion in control group, and this elevation was significantly blunted by 4.5 mg/kg NecroX-7 treatment. Histologic analysis showed a marked ischemia or IR injury-induced hepatocytic degenerations, sinusoidal and portal vein congestions, and inflammatory cell infiltrations in the control group, whereas the treatment groups showed significantly diminished histopathology in a dose-dependent manner. CONCLUSIONS These results demonstrated that NecroX-7 attenuated the hepatocyte lethality caused by hepatic IR injury in a large animal setting. We conclude that NecroX-7 may provide a wide variety of therapeutic options for IR injury in human patients.
Collapse
Affiliation(s)
- J H Lee
- Department of Hepatobiliary and Pancreatic Surgery, Asan Medical Center, University of Ulsan, College of Medicine, Seoul, Korea
| | - K M Park
- Department of Hepatobiliary and Pancreatic Surgery, Asan Medical Center, University of Ulsan, College of Medicine, Seoul, Korea.
| | - Y J Lee
- Department of Hepatobiliary and Pancreatic Surgery, Asan Medical Center, University of Ulsan, College of Medicine, Seoul, Korea
| | - J H Kim
- Department of Pathology, Asan Medical Center, University of Ulsan, College of Medicine, Seoul, Korea
| | - S H Kim
- LG Life Sciences, Daejeon, Korea
| |
Collapse
|
10
|
Kopalli SR, Kang TB, Koppula S. Necroptosis inhibitors as therapeutic targets in inflammation mediated disorders - a review of the current literature and patents. Expert Opin Ther Pat 2016; 26:1239-1256. [PMID: 27568917 DOI: 10.1080/13543776.2016.1230201] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
INTRODUCTION Recent studies have shown substantial interplay between the apoptosis and necroptosis pathways. Necroptosis, a form of programmed cell death, has been found to stimulate the immune system contributing to the pathophysiology of several inflammation-mediated disorders. Determining the contribution of necroptotic signaling pathways to inflammation may lead to the development of selective and specific molecular target implicated necroptosis inhibitors. Areas covered: This review summarizes the recently published and patented necroptosis inhibitors as therapeutic targets in inflammation-mediated disorders. The role of several necroptosis inhibitors, focusing on specific signaling molecules, was discussed with particular attention to inflammation-mediated disorders. Data was obtained from Espacenet®, WIPO®, USPTO® patent websites, and other relevant sources (2006-2016). Expert opinion: Necroptosis inhibitors hold promise for treatment of inflammation-mediated clinical conditions in which necroptotic cell death plays a major role. Although necroptosis inhibitors reviewed in this survey showed inhibitory effects against several inflammation-mediated disorders, only a few have passed to the stage of clinical testing and need extensive research for therapeutic practice. Revisiting the existing drugs and developing novel necroptosis inhibiting agents as well as understanding their mechanism are essential. A detailed study of necroptosis function in animal models of inflammation may provide us an alternative strategy for the development of drug-like necroptosis inhibitors.
Collapse
Affiliation(s)
| | - Tae-Bong Kang
- a College of Biomedical and Health Sciences , Konkuk University , Chungju , Republic of Korea
| | - Sushruta Koppula
- a College of Biomedical and Health Sciences , Konkuk University , Chungju , Republic of Korea
| |
Collapse
|
11
|
Grootaert MO, Schrijvers DM, Van Spaendonk H, Breynaert A, Hermans N, Van Hoof VO, Takahashi N, Vandenabeele P, Kim SH, De Meyer GR, Martinet W. NecroX-7 reduces necrotic core formation in atherosclerotic plaques of Apoe knockout mice. Atherosclerosis 2016; 252:166-174. [DOI: 10.1016/j.atherosclerosis.2016.06.045] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/24/2015] [Revised: 06/10/2016] [Accepted: 06/29/2016] [Indexed: 12/31/2022]
|
12
|
Degterev A, Linkermann A. Generation of small molecules to interfere with regulated necrosis. Cell Mol Life Sci 2016; 73:2251-67. [PMID: 27048812 PMCID: PMC11108466 DOI: 10.1007/s00018-016-2198-x] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Accepted: 03/18/2016] [Indexed: 12/16/2022]
Abstract
Interference with regulated necrosis for clinical purposes carries broad therapeutic relevance and, if successfully achieved, has a potential to revolutionize everyday clinical routine. Necrosis was interpreted as something that no clinician might ever be able to prevent due to the unregulated nature of this form of cell death. However, given our growing understanding of the existence of regulated forms of necrosis and the roles of key enzymes of these pathways, e.g., kinases, peroxidases, etc., the possibility emerges to identify efficient and selective small molecule inhibitors of pathologic necrosis. Here, we review the published literature on small molecule inhibition of regulated necrosis and provide an outlook on how combination therapy may be most effective in treatment of necrosis-associated clinical situations like stroke, myocardial infarction, sepsis, cancer and solid organ transplantation.
Collapse
Affiliation(s)
- Alexei Degterev
- Department of Developmental, Molecular & Chemical Biology, Sackler School of Graduate Biomedical Sciences, Tufts University School of Medicine, Boston, MA, 02111, USA.
| | - Andreas Linkermann
- Clinic for Nephrology and Hypertension, University-Hospital Schleswig-Holstein, Campus Kiel, Christian-Albrechts-University Kiel, Fleckenstr. 4, 24105, Kiel, Germany.
| |
Collapse
|
13
|
Thu VT, Kim HK, Long LT, Thuy TT, Huy NQ, Kim SH, Kim N, Ko KS, Rhee BD, Han J. NecroX-5 exerts anti-inflammatory and anti-fibrotic effects via modulation of the TNFα/Dcn/TGFβ1/Smad2 pathway in hypoxia/reoxygenation-treated rat hearts. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2016; 20:305-14. [PMID: 27162485 PMCID: PMC4860373 DOI: 10.4196/kjpp.2016.20.3.305] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/26/2016] [Revised: 03/04/2016] [Accepted: 03/04/2016] [Indexed: 12/12/2022]
Abstract
Inflammatory and fibrotic responses are accelerated during the reperfusion period, and excessive fibrosis and inflammation contribute to cardiac malfunction. NecroX compounds have been shown to protect the liver and heart from ischemia-reperfusion injury. The aim of this study was to further define the role and mechanism of action of NecroX-5 in regulating infl ammation and fi brosis responses in a model of hypoxia/reoxygenation (HR). We utilized HR-treated rat hearts and lipopolysaccharide (LPS)-treated H9C2 culture cells in the presence or absence of NecroX-5 (10 µmol/L) treatment as experimental models. Addition of NecroX-5 signifi cantly increased decorin (Dcn) expression levels in HR-treated hearts. In contrast, expression of transforming growth factor beta 1 (TGFβ1) and Smad2 phosphorylation (pSmad2) was strongly attenuated in NecroX-5-treated hearts. In addition, signifi cantly increased production of tumor necrosis factor alpha (TNFα), TGFβ1, and pSmad2, and markedly decreased Dcn expression levels, were observed in LPS-stimulated H9C2 cells. Interestingly, NecroX-5 supplementation effectively attenuated the increased expression levels of TNFα, TGFβ1, and pSmad2, as well as the decreased expression of Dcn. Thus, our data demonstrate potential antiinflammatory and anti-fibrotic effects of NecroX-5 against cardiac HR injuries via modulation of the TNFα/Dcn/TGFβ1/Smad2 pathway.
Collapse
Affiliation(s)
- Vu Thi Thu
- National Research Laboratory for Mitochondrial Signaling, Department of Physiology, Department of Health Sciences and Technology, BK21 Project Team, College of Medicine, Cardiovascular and Metabolic Disease Center, Inje University, Busan 47392, Korea.; VNU University of Science, Hanoi 120036, Vietnam
| | - Hyoung Kyu Kim
- National Research Laboratory for Mitochondrial Signaling, Department of Physiology, Department of Health Sciences and Technology, BK21 Project Team, College of Medicine, Cardiovascular and Metabolic Disease Center, Inje University, Busan 47392, Korea.; Department of Integrated Biomedical Science, College of Medicine, Inje University, Busan 47392, Korea
| | - Le Thanh Long
- National Research Laboratory for Mitochondrial Signaling, Department of Physiology, Department of Health Sciences and Technology, BK21 Project Team, College of Medicine, Cardiovascular and Metabolic Disease Center, Inje University, Busan 47392, Korea
| | | | | | - Soon Ha Kim
- Product Strategy and Development, LG Life Sciences Ltd., Seoul 03184, Korea
| | - Nari Kim
- National Research Laboratory for Mitochondrial Signaling, Department of Physiology, Department of Health Sciences and Technology, BK21 Project Team, College of Medicine, Cardiovascular and Metabolic Disease Center, Inje University, Busan 47392, Korea
| | - Kyung Soo Ko
- National Research Laboratory for Mitochondrial Signaling, Department of Physiology, Department of Health Sciences and Technology, BK21 Project Team, College of Medicine, Cardiovascular and Metabolic Disease Center, Inje University, Busan 47392, Korea
| | - Byoung Doo Rhee
- National Research Laboratory for Mitochondrial Signaling, Department of Physiology, Department of Health Sciences and Technology, BK21 Project Team, College of Medicine, Cardiovascular and Metabolic Disease Center, Inje University, Busan 47392, Korea
| | - Jin Han
- National Research Laboratory for Mitochondrial Signaling, Department of Physiology, Department of Health Sciences and Technology, BK21 Project Team, College of Medicine, Cardiovascular and Metabolic Disease Center, Inje University, Busan 47392, Korea
| |
Collapse
|
14
|
Thu VT, Kim HK, Long LT, Nyamaa B, Song IS, Thuy TT, Huy NQ, Marquez J, Kim SH, Kim N, Ko KS, Rhee BD, Han J. NecroX-5 protects mitochondrial oxidative phosphorylation capacity and preserves PGC1α expression levels during hypoxia/reoxygenation injury. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2016; 20:201-11. [PMID: 26937217 PMCID: PMC4770111 DOI: 10.4196/kjpp.2016.20.2.201] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Revised: 01/11/2016] [Accepted: 01/13/2016] [Indexed: 02/06/2023]
Abstract
Although the antioxidant and cardioprotective effects of NecroX-5 on various in vitro and in vivo models have been demonstrated, the action of this compound on the mitochondrial oxidative phosphorylation system remains unclear. Here we verify the role of NecroX-5 in protecting mitochondrial oxidative phosphorylation capacity during hypoxia-reoxygenation (HR). Necrox-5 treatment (10 µM) and non-treatment were employed on isolated rat hearts during hypoxia/reoxygenation treatment using an ex vivo Langendorff system. Proteomic analysis was performed using liquid chromatography-mass spectrometry (LC-MS) and non-labeling peptide count protein quantification. Real-time PCR, western blot, citrate synthases and mitochondrial complex activity assays were then performed to assess heart function. Treatment with NecroX-5 during hypoxia significantly preserved electron transport chain proteins involved in oxidative phosphorylation and metabolic functions. NecroX-5 also improved mitochondrial complex I, II, and V function. Additionally, markedly higher peroxisome proliferator-activated receptor-gamma coactivator-1α (PGC1α) expression levels were observed in NecroX-5-treated rat hearts. These novel results provide convincing evidence for the role of NecroX-5 in protecting mitochondrial oxidative phosphorylation capacity and in preserving PGC1α during cardiac HR injuries.
Collapse
Affiliation(s)
- Vu Thi Thu
- National Research Laboratory for Mitochondrial Signaling, Department of Physiology, College of Medicine, Cardiovascular and Metabolic Disease Center, Inje University, Busan 47392, Korea.; Faculty of Biology, VNU University of Science, Hanoi 120036, Vietnam
| | - Hyoung Kyu Kim
- National Research Laboratory for Mitochondrial Signaling, Department of Physiology, College of Medicine, Cardiovascular and Metabolic Disease Center, Inje University, Busan 47392, Korea
| | - Le Thanh Long
- National Research Laboratory for Mitochondrial Signaling, Department of Physiology, College of Medicine, Cardiovascular and Metabolic Disease Center, Inje University, Busan 47392, Korea
| | - Bayalagmaa Nyamaa
- National Research Laboratory for Mitochondrial Signaling, Department of Physiology, College of Medicine, Cardiovascular and Metabolic Disease Center, Inje University, Busan 47392, Korea
| | - In-Sung Song
- National Research Laboratory for Mitochondrial Signaling, Department of Physiology, College of Medicine, Cardiovascular and Metabolic Disease Center, Inje University, Busan 47392, Korea
| | - To Thanh Thuy
- Faculty of Biology, VNU University of Science, Hanoi 120036, Vietnam
| | - Nguyen Quang Huy
- Faculty of Biology, VNU University of Science, Hanoi 120036, Vietnam
| | - Jubert Marquez
- National Research Laboratory for Mitochondrial Signaling, Department of Physiology, College of Medicine, Cardiovascular and Metabolic Disease Center, Inje University, Busan 47392, Korea
| | - Soon Ha Kim
- Product Strategy and Development, LG Life Sciences Ltd, Seoul 03184, Korea
| | - Nari Kim
- National Research Laboratory for Mitochondrial Signaling, Department of Physiology, College of Medicine, Cardiovascular and Metabolic Disease Center, Inje University, Busan 47392, Korea
| | - Kyung Soo Ko
- National Research Laboratory for Mitochondrial Signaling, Department of Physiology, College of Medicine, Cardiovascular and Metabolic Disease Center, Inje University, Busan 47392, Korea
| | - Byoung Doo Rhee
- National Research Laboratory for Mitochondrial Signaling, Department of Physiology, College of Medicine, Cardiovascular and Metabolic Disease Center, Inje University, Busan 47392, Korea
| | - Jin Han
- National Research Laboratory for Mitochondrial Signaling, Department of Physiology, College of Medicine, Cardiovascular and Metabolic Disease Center, Inje University, Busan 47392, Korea
| |
Collapse
|
15
|
Jin S, Kim S, Seo H, Jeong J, Ahn K, Kim J, Choi D, Park J, Lee J, Choi S, Seong I, Kim S, Suh K, Jeong JO. Beneficial Effects of Necrosis Modulator, Indole Derivative NecroX-7, on Renal Ischemia-Reperfusion Injury in Rats. Transplant Proc 2016; 48:199-204. [DOI: 10.1016/j.transproceed.2015.12.018] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2015] [Accepted: 12/22/2015] [Indexed: 01/03/2023]
|
16
|
Im KI, Kim N, Lim JY, Nam YS, Lee ES, Kim EJ, Kim HJ, Kim SH, Cho SG. The Free Radical Scavenger NecroX-7 Attenuates Acute Graft-versus-Host Disease via Reciprocal Regulation of Th1/Regulatory T Cells and Inhibition of HMGB1 Release. THE JOURNAL OF IMMUNOLOGY 2015; 194:5223-32. [PMID: 25911749 DOI: 10.4049/jimmunol.1402609] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/16/2014] [Accepted: 03/27/2015] [Indexed: 01/27/2023]
Abstract
Graft-versus-host disease (GVHD) is a major complication associated with allogeneic hematopoietic stem cell transplantation. Despite the prominent role of the adaptive immune system, the importance of controlling the innate immune system in the pathogenesis of GVHD has recently been rediscovered. High-mobility group box 1 (HMGB1) is a crucial damage-associated molecular pattern signal that functions as a potent innate immune mediator in GVHD. In the present study, we investigated treatment of experimental GVHD through HMGB1 blockade using the compound cyclopentylamino carboxymethylthiazolylindole (NecroX)-7. Treated animals significantly attenuated GVHD-related mortality and inhibited severe tissue damage. These protective effects correlated with the decrease in HMGB1 expression and lower levels of reactive oxidative stress. Additionally, NecroX-7 inhibited the HMGB1-induced release of TNF and IL-6, as well as the expression of TLR-4 and receptor for advanced glycation end products. We also observed increased regulatory T cell numbers, which may be associated with regulation of differentiation signals independent of HMGB1. Taken together, these data indicate that NecroX-7 protects mice against lethal GVHD by reciprocal regulation of regulatory T/Th1 cells, attenuating systemic HMGB1 accumulation and inhibiting HMGB1-mediated inflammatory response. Our results indicate the possibility of a new use for a clinical drug that is effective for the treatment of GVHD.
Collapse
Affiliation(s)
- Keon-Il Im
- Institute for Translational Research and Molecular Imaging, Catholic Research Institute of Medical Science, Catholic University of Korea, Seoul 137-701, Korea; Laboratory of Immune Regulation, Convergent Research Consortium for Immunologic Disease, Catholic University of Korea College of Medicine, St. Mary's Hospital, Seoul 137-701, Korea
| | - Nayoun Kim
- Institute for Translational Research and Molecular Imaging, Catholic Research Institute of Medical Science, Catholic University of Korea, Seoul 137-701, Korea; Laboratory of Immune Regulation, Convergent Research Consortium for Immunologic Disease, Catholic University of Korea College of Medicine, St. Mary's Hospital, Seoul 137-701, Korea
| | - Jung-Yeon Lim
- Institute for Translational Research and Molecular Imaging, Catholic Research Institute of Medical Science, Catholic University of Korea, Seoul 137-701, Korea; Laboratory of Immune Regulation, Convergent Research Consortium for Immunologic Disease, Catholic University of Korea College of Medicine, St. Mary's Hospital, Seoul 137-701, Korea
| | - Young-Sun Nam
- Institute for Translational Research and Molecular Imaging, Catholic Research Institute of Medical Science, Catholic University of Korea, Seoul 137-701, Korea; Laboratory of Immune Regulation, Convergent Research Consortium for Immunologic Disease, Catholic University of Korea College of Medicine, St. Mary's Hospital, Seoul 137-701, Korea
| | - Eun-Sol Lee
- Institute for Translational Research and Molecular Imaging, Catholic Research Institute of Medical Science, Catholic University of Korea, Seoul 137-701, Korea; Laboratory of Immune Regulation, Convergent Research Consortium for Immunologic Disease, Catholic University of Korea College of Medicine, St. Mary's Hospital, Seoul 137-701, Korea
| | - Eun-Jung Kim
- Laboratory of Immune Regulation, Convergent Research Consortium for Immunologic Disease, Catholic University of Korea College of Medicine, St. Mary's Hospital, Seoul 137-701, Korea
| | - Hyoung Jin Kim
- Strategy and Development, LG Life Sciences Ltd., Seoul 2305-738, Korea; and
| | - Soon Ha Kim
- Strategy and Development, LG Life Sciences Ltd., Seoul 2305-738, Korea; and
| | - Seok-Goo Cho
- Institute for Translational Research and Molecular Imaging, Catholic Research Institute of Medical Science, Catholic University of Korea, Seoul 137-701, Korea; Laboratory of Immune Regulation, Convergent Research Consortium for Immunologic Disease, Catholic University of Korea College of Medicine, St. Mary's Hospital, Seoul 137-701, Korea; Department of Hematology, Catholic Blood and Marrow Transplantation Center, Seoul St. Mary's Hospital, Catholic University of Korea College of Medicine, Seoul 137-701, Korea
| |
Collapse
|
17
|
Chung HK, Kim YK, Park JH, Ryu MJ, Chang JY, Hwang JH, Lee CH, Kim SH, Kim HJ, Kweon GR, Kim KS, Shong M. The indole derivative NecroX-7 improves nonalcoholic steatohepatitis in ob/ob mice through suppression of mitochondrial ROS/RNS and inflammation. Liver Int 2015; 35:1341-53. [PMID: 25443620 DOI: 10.1111/liv.12741] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2014] [Accepted: 11/13/2014] [Indexed: 02/13/2023]
Abstract
BACKGROUND & AIMS Nonalcoholic steatohepatitis (NASH) is associated with cirrhosis and hepatocellular carcinoma. Reactive oxygen species (ROS) and reactive nitrogen species (RNS) play key roles in the development of the disease. However, the therapeutic target of NASH has not been fully defined and new treatments are needed. We investigated the protective effects of the antioxidant indole-derived NecroX-7 in a NASH mouse model using leptin-deficient ob/ob and methionine- and choline-deficient (MCD) diet-fed ob/ob mice. METHODS Six-week-old male mice were divided into three groups: ob/+ mice, ob/ob mice treated with vehicle and ob/ob mice treated daily with NecroX-7 (20 mg/kg) for 4 weeks. To study the effects of NecroX-7 in a fibrosis model, NASH was induced by feeding ob/ob mice an MCD diet. The effects of NecroX-7 on NASH progression were evaluated using biochemical, histological and molecular markers. RESULTS NecroX-7-treated ob/ob mice had a marked decrease in serum aspartate aminotransferase and alanine transaminase compared with vehicle-treated controls. Interestingly, hepatic steatosis and lipid peroxidation were significantly improved by NecroX-7 treatment. NecroX-7 inhibited tert-butylhydroperoxide- and H2 O2 -induced mitochondrial ROS/RNS in primary hepatocytes and attenuated mitochondrial dysfunction in vitro and in vivo. Furthermore, NecroX-7-treated mice exhibited fewer infiltrating macrophages and reduced hepatic tumour necrosis factor-alpha expression. Hepatic fibrosis in MCD-fed ob/ob mice was significantly decreased by NecroX-7 treatment. CONCLUSIONS NecroX-7 treatment improved hepatic steatosis and fibrosis in murine NASH models. These effects occurred through the suppression of whole-cell ROS/RNS and inflammatory responses and suggest that NecroX-7 has a potential therapeutic benefit in steatohepatitis.
Collapse
Affiliation(s)
- Hyo Kyun Chung
- Research Center for Endocrine & Metabolic Diseases, Chungnam National University School of Medicine, Daejeon, Korea
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
CHO YOUNGSIK. Perspectives on the therapeutic modulation of an alternative cell death, programmed necrosis (Review). Int J Mol Med 2014; 33:1401-6. [DOI: 10.3892/ijmm.2014.1716] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2013] [Accepted: 03/21/2014] [Indexed: 11/05/2022] Open
|
19
|
Lee SR, Lee SJ, Kim SH, Ko KS, Rhee BD, Xu Z, Kim N, Han J. NecroX-5 suppresses sodium nitroprusside-induced cardiac cell death through inhibition of JNK and caspase-3 activation. Cell Biol Int 2014; 38:702-7. [PMID: 24446382 DOI: 10.1002/cbin.10242] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2013] [Accepted: 01/07/2014] [Indexed: 12/12/2022]
Abstract
Although sodium nitroprusside (SNP) is an effective hypotensive drug and is often used in pediatric intensive care units and to treat acute heart failure, clinical application of SNP is limited by its cardiotoxicity. NecroX-5 (NX-5) was recently developed and has the capacity to inhibit necrotic cell death. No current literature addresses whether NX-5 suppresses SNP-induced cell death or its mechanism of action. We have investigated the protective role of NX-5 against SNP-induced cell death in cardiomyocyte-like H9c2 cells. SNP treatment induced severe cell death, possibly through phosphorylation of stress-activated protein kinase/c-Jun NH₂-terminal kinase (JNK) and activation of the apoptotic signaling pathway, including downregulation of Bcl-2 and cleavage of caspase-3. However, NX-5 suppresses SNP-induced cell death through inhibition of JNK activation and suppression of both downregulation of Bcl-2 protein expression and caspase-3 cleavage. These findings will provide insights and facilitate development of antidotes to SNP toxicity in cardiac cells.
Collapse
Affiliation(s)
- Sung Ryul Lee
- National Research Laboratory for Mitochondrial Signaling, Department of Physiology, College of Medicine, Cardiovascular and Metabolic Disease Center, Inje University, 633-165 Gaegeum-dong Busanjin-gu, Busan, 614 735, Republic of Korea
| | | | | | | | | | | | | | | |
Collapse
|
20
|
Patent Highlights. Pharm Pat Anal 2013. [DOI: 10.4155/ppa.13.59] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Snapshot of key developments in the patent literature accompanied by explanatory synopses
Collapse
|
21
|
Protective role of NecroX-5 against neomycin-induced hair cell damage in zebrafish. Arch Toxicol 2013; 88:435-41. [PMID: 24030356 DOI: 10.1007/s00204-013-1124-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2013] [Accepted: 08/22/2013] [Indexed: 10/26/2022]
Abstract
NecroX-5, one of the derivatives of NecroX series compounds, is a mitochondrial reactive oxygen species and reactive nitrogen species scavenger that inhibits cell death against various kinds of oxidative stresses. The objective of the present study was to evaluate the effects of NecroX-5 on neomycin-induced ototoxicity in transgenic zebrafish (Brn3C: EGFP). Five days post-fertilization, zebrafish larvae were exposed to 125 μM neomycin and one of the following NecroX-5 concentrations for 1 h: 10, 25, 50, and 75 μM. Hair cells within the neuromasts of the supraorbital (SO1 and SO2), otic (O1), and occipital (OC1) lateral lines were analyzed using fluorescence microscopy (n = 10). The terminal deoxynucleotidyl transferase-mediated dUTP-biotin nick end labeling (TUNEL) assay and 2-[4-(dimethylamino) styryl]-N-ethylpyridiniumiodide (DASPEI) assay were performed for evaluation of apoptosis and mitochondrial damage. Ultrastructural changes were evaluated using scanning electron microscopy. NecroX-5 decreased neomycin-induced hair cell loss in the neuromasts (NecroX-5 50 μM: 13.4 ± 2.0 cells, 125 μM neomycin only: 8.1 ± 1.2 cells; n = 10, P < 0.05) and decreased the TUNEL reaction. The ultrastructural analysis showed that the structures of mitochondria and hair cells within the neuromasts were preserved in zebrafish exposed to 125 μM neomycin and 50 μM NecroX-5. NecroX-5 decreased apoptosis and mitochondrial damage. In conclusion, NecroX-5 attenuated neomycin-induced hair cell loss in zebrafish.
Collapse
|
22
|
Park JH, Seo KS, Tadi S, Ahn BH, Lee JU, Heo JY, Han J, Song MS, Kim SH, Yim YH, Choi HS, Shong M, Kweon G. An indole derivative protects against acetaminophen-induced liver injury by directly binding to N-acetyl-p-benzoquinone imine in mice. Antioxid Redox Signal 2013; 18:1713-22. [PMID: 23121402 PMCID: PMC3619205 DOI: 10.1089/ars.2012.4677] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
AIMS Acetaminophen (APAP)-induced liver injury is mainly due to the excessive formation of reactive oxygen species (ROS) and reactive nitrogen species (RNS) through the formation of a reactive intermediate, N-acetyl-p-benzoquinone imine (NAPQI), in both humans and rodents. Here, we show that the indole-derived synthetic compound has a protective effect against APAP-induced liver injury in C57Bl/6 mice model. RESULTS NecroX-7 decreased tert-butylhydroperoxide (t-BHP)- and APAP-induced cell death and ROS/RNS formation in HepG2 human hepatocarcinoma and primary mouse hepatocytes. In mice, NecroX-7 decreased APAP-induced phosphorylation of c-Jun N-terminal kinase (JNK) and 3-nitrotyrosine (3-NT) formation, and also protected mice from APAP-induced liver injury and lethality by binding directly to NAPQI. The binding of NecroX-7 to NAPQI did not require any of cofactors or proteins. NecroX-7 could only scavenge NAPQI when hepatocellular GSH levels were very low. INNOVATION NecroX-7 is an indole-derived potent antioxidant molecule, which can be bound to some types of radicals and especially NAPQI. It is well known that the NAPQI is a major intermediate of APAP, which causes necrosis of hepatocytes in rodents and humans. Thus, blocking NAPQI formation or eliminating NAPQI are novel strategies for the treatment or prevention of APAP-induced liver injury instead of GSH replenishment. CONCLUSION Our data suggest that the indole-derivative, NecroX-7, directly binds to NAPQI when hepatic GSH levels are very low and the NAPQI-NecroX-7 complex is secreted to the blood from the liver. NecroX-7 shows more preventive and similar therapeutic effects against APAP-induced liver injury when compared to the effect of N-acetylcysteine in C57Bl/6 mice.
Collapse
Affiliation(s)
- Ji-Hoon Park
- Department of Biochemistry, School of Medicine, Chungnam National University, Daejeon, Korea
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Affiliation(s)
- Ji-Hoon Park
- Department of Biochemistry, School of Medicine, Chungnam National University, Daejeon, Korea
| | - Gi Ryang Kweon
- Department of Biochemistry, School of Medicine, Chungnam National University, Daejeon, Korea
| |
Collapse
|
24
|
Kim HJ, Yoon KA, Lee MK, Kim SH, Lee IK, Kim SY. A novel small molecule, NecroX-7, inhibits osteoclast differentiation by suppressing NF-κB activity and c-Fos expression. Life Sci 2012; 91:928-934. [PMID: 23000100 DOI: 10.1016/j.lfs.2012.09.009] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2012] [Revised: 08/30/2012] [Accepted: 09/07/2012] [Indexed: 12/25/2022]
Abstract
AIMS Osteoclasts, the unique bone-resorbing polykaryons, are responsible for many bone-destructive diseases, such as osteoporosis and rheumatoid arthritis. Hence, the regulation of osteoclast formation is considered a potential therapeutic approach for these diseases. In this study, we investigated the effect of a novel small compound, C(25)H(32)N(4)O(4)S(2) (NecroX-7) on osteoclast formation. MAIN METHODS We analyzed the effects of NecoX-7 on receptor activator of nuclear factor kappa B ligand (RANKL)-induced osteoclast differentiation in vitro and LPS-induced bone loss in vivo. KEY FINDINGS We observed that NecroX-7 suppressed osteoclast formation from primary bone marrow macrophages (BMMs) in a dose-dependent manner. NecroX-7 significantly inhibited the NF-κB signaling pathway without affecting the activation of the mitogen-activated protein kinases (MAPKs) JNK, p38, and ERK in response to RANKL. In addition, NecroX-7 strongly attenuated the induction of c-Fos and nuclear factor of activated T cells c1 (NFATc1), which are crucial transcription factors for osteoclast differentiation. Mirroring the down-regulation of c-Fos and NFATc1, the expression of osteoclastogenic markers, such as tartrate-resistant acid phosphatase (TRAP) and cathepsin K, was also reduced by the addition of NecroX-7. Furthermore, confirming the in vitro anti-osteoclastogenic effect, NecroX-7 inhibited lipopolysaccharide (LPS)-induced bone loss in vivo. SIGNIFICANCE Our data imply that NecroX-7 is useful as a therapeutic drug for the treatment of bone resorption-associated diseases.
Collapse
Affiliation(s)
- Hyun-Ju Kim
- Skeletal Diseases Genome Research Center, Kyungpook National University and Hospital, Daegu 700-412, Republic of Korea.
| | | | | | | | | | | |
Collapse
|
25
|
Park MK, Lee BD, Chae SW, Chi J, Kwon SK, Song JJ. Protective effect of NecroX, a novel necroptosis inhibitor, on gentamicin-induced ototoxicity. Int J Pediatr Otorhinolaryngol 2012; 76:1265-9. [PMID: 22704672 DOI: 10.1016/j.ijporl.2012.05.016] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2012] [Revised: 05/14/2012] [Accepted: 05/15/2012] [Indexed: 12/19/2022]
Abstract
INTRODUCTION NecroX is a novel necrosis and necroptosis inhibitor that shows scavenger activity against mitochondrial reactive oxygen species (ROS) and cytoprotective activity against various insults. These findings raise the possibility of its protective effect in ototoxicity. This study was performed to investigate the protective effect of NecroX on gentamicin (GM)-induced hair cell loss in neonatal mouse cochlea cultures. MATERIALS AND METHODS The protective effects of NecroX were measured by phalloidin staining of cultures from postnatal day 2-3 mice with GM-induced hair cell loss. Terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) staining was used to detect apoptosis. The radical-scavenging activity of NecroX was assessed using the 1,1-diphenyl-2-picrylhydrazyl (DPPH) assay. RESULTS NecroX showed a significant and concentration-dependent protective effect against GM-induced hair cell loss, and hair cells retained their stereocilia well. NecroX decreased GM-induced apoptosis of hair cells as assessed by TUNEL staining. Additionally, NecroX showed direct radical scavenging activity in the DPPH assay. CONCLUSIONS In this study, we demonstrated the protective effect of NecroX on GM-induced hair cell loss in neonatal cochlea cultures, and suggest that it may be of therapeutic use in the treatment of drug-induced ototoxicity.
Collapse
Affiliation(s)
- Moo Kyun Park
- Department of Otolaryngology - Head and Neck Surgery, Soonchunhyang University College of Medicine, Seoul, South Korea
| | | | | | | | | | | |
Collapse
|
26
|
Park J, Park E, Ahn BH, Kim HJ, Park JH, Koo SY, Kwak HS, Park HS, Kim DW, Song M, Yim HJ, Seo DO, Kim SH. NecroX-7 prevents oxidative stress-induced cardiomyopathy by inhibition of NADPH oxidase activity in rats. Toxicol Appl Pharmacol 2012; 263:1-6. [DOI: 10.1016/j.taap.2012.05.014] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2011] [Revised: 05/15/2012] [Accepted: 05/23/2012] [Indexed: 10/28/2022]
|
27
|
Thu VT, Kim HK, Long LT, Lee SR, Hanh TM, Ko TH, Heo HJ, Kim N, Kim SH, Ko KS, Rhee BD, Han J. NecroX-5 prevents hypoxia/reoxygenation injury by inhibiting the mitochondrial calcium uniporter. Cardiovasc Res 2012; 94:342-50. [DOI: 10.1093/cvr/cvs122] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
|
28
|
Lee DW, Lee TK, Cho IS, Park HE, Jin S, Cho HJ, Kim SH, Oh S, Kim HS. Creation of myocardial fibrosis by transplantation of fibroblasts primed with survival factors. Am J Physiol Heart Circ Physiol 2011; 301:H1004-14. [PMID: 21685267 DOI: 10.1152/ajpheart.00156.2011] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
One of the major obstacles in the creation of myocardial fibrosis using fibroblasts is massive cell death after cell injection. To overcome this problem, a method that delivers fibroblasts primed with survival factors was studied. Cardiac fibroblasts were isolated from wild-type male C57BL/6 mice. Female mice were randomly placed into the following three groups: 1) fibroblasts transfected with β-galactosidase-containing adenovirus (control group), 2) fibroblasts treated with a necrosis inhibitor (NI group), and 3) fibroblasts transfected with Akt-containing adenovirus (Akt group). Pretreated cells were transplanted into the recipient heart by direct injection after a thoracotomy. Quantitative real-time PCR and morphometric analysis were performed to investigate the effects of survival factor priming on the induction of cell engraftment and fibrosis. In addition, a canine model was used to investigate the development of fibrosis and conduction modification using autologous dermal fibroblasts. The NI and Akt groups showed a better engraftment rate: 13 (NI group) and 7 (Akt group) times greater at 21 days compared with the control group. Increased fibrosis and conduction delay were also observed in the NI and Akt groups compared with the control group. Survival factor priming increased cellular engraftment and enhanced the efficacy of cell transplantation. Delivery of fibroblasts primed with survival factors might be a promising approach to develop conduction modification as a novel strategy to treat arrhythmias.
Collapse
Affiliation(s)
- Dong-Won Lee
- Department of Internal Medicine, Seoul National University Hospital, Seoul Korea
| | | | | | | | | | | | | | | | | |
Collapse
|