1
|
Takahashi K, Sato E, Yamakoshi S, Ogane M, Sekimoto A, Ishikawa T, Kisu K, Oe Y, Okamoto K, Miyazaki M, Tanaka T, Takahashi N. Nicotinamide ameliorates podocyte injury and albuminuria in adriamycin-induced nephropathy. Am J Physiol Renal Physiol 2025; 328:F501-F516. [PMID: 40033940 DOI: 10.1152/ajprenal.00297.2024] [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: 10/15/2024] [Revised: 11/06/2024] [Accepted: 02/26/2025] [Indexed: 03/05/2025] Open
Abstract
Podocytes are key components of the glomerular filtration barrier, and their injury leads to proteinuria, chronic kidney disease (CKD), and nephrotic syndrome. Effective treatments for these conditions are not well established, and prevention of podocyte injury is a crucial challenge. Nicotinamide (NAM), a form of vitamin B3, has been reported to exert beneficial effects in various renal disease models due to its antioxidant and anti-inflammatory properties and its ability to replenish nicotinamide adenine dinucleotide (NAD+). However, its impact on adriamycin (ADR)-induced nephropathy, a model of nephrotic syndrome caused by podocyte injury, remains unclear. We investigated the effects of NAM administration in a mouse model of ADR nephropathy. BALB/c mice were intravenously administered ADR to induce nephropathy. In the NAM-treated group, mice received 0.6% NAM in drinking water ad libitum starting 7 days before ADR administration. After 14 days, NAM treatment decreased albuminuria, glomerular sclerosis, and podocyte injury, and reduced inflammation and oxidative stress markers in the kidneys. NAM and NAD+ levels were decreased in ADR-treated kidneys, and the expression of the NAD+-consuming enzymes SIRT1 and poly(ADP-ribose) polymerase 1 (PARP-1) was decreased and increased, respectively. Nicotinamide N-methyltransferase expression was increased. NAM canceled these abnormalities. In cultured rat podocytes, NAD+ alleviated ADR-induced cytotoxicity, apoptosis, and inflammation. These findings suggest that NAM prevents ADR nephropathy and podocyte injury, likely through NAD+ replenishment.NEW & NOTEWORTHY Nephrotic syndrome can lead to end-stage kidney disease and cause severe complications. Currently, effective treatments for nephrotic syndrome have not been established, and new therapeutic approaches targeting podocyte injury are needed. Nicotinamide prevents podocyte injury in adriamycin-induced nephropathy in mice and ameliorates albuminuria, pathological changes, oxidative stress, and inflammation. Here, we provide evidence that pretreatment with nicotinamide can attenuate podocyte injury and subsequent nephropathy in mice.
Collapse
Affiliation(s)
- Kei Takahashi
- Department of Nephrology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Emiko Sato
- Department of Nephrology, Tohoku University Graduate School of Medicine, Sendai, Japan
- Division of Clinical Pharmacology and Therapeutics, Tohoku University Graduate School of Pharmaceutical Sciences, Sendai, Japan
| | - Seiko Yamakoshi
- Division of Clinical Pharmacology and Therapeutics, Tohoku University Graduate School of Pharmaceutical Sciences, Sendai, Japan
| | - Mizuki Ogane
- Division of Clinical Pharmacology and Therapeutics, Tohoku University Graduate School of Pharmaceutical Sciences, Sendai, Japan
| | - Akiyo Sekimoto
- Division of Clinical Pharmacology and Therapeutics, Tohoku University Graduate School of Pharmaceutical Sciences, Sendai, Japan
| | - Takamasa Ishikawa
- Infinity Lab, Inc., Tsuruoka, Japan
- Institute for Advanced Biosciences, Keio University, Tsuruoka, Japan
| | - Kiyomi Kisu
- Department of Nephrology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Yuji Oe
- Department of Nephrology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Koji Okamoto
- Department of Nephrology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Mariko Miyazaki
- Department of Nephrology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Tetsuhiro Tanaka
- Department of Nephrology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Nobuyuki Takahashi
- Department of Nephrology, Tohoku University Graduate School of Medicine, Sendai, Japan
- Division of Clinical Pharmacology and Therapeutics, Tohoku University Graduate School of Pharmaceutical Sciences, Sendai, Japan
| |
Collapse
|
2
|
Jiang YZ, Huang XR, Chang J, Zhou Y, Huang XT. SIRT1: An Intermediator of Key Pathways Regulating Pulmonary Diseases. J Transl Med 2024; 104:102044. [PMID: 38452903 DOI: 10.1016/j.labinv.2024.102044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 02/07/2024] [Accepted: 02/21/2024] [Indexed: 03/09/2024] Open
Abstract
Silent information regulator type-1 (SIRT1), a nicotinamide adenine dinucleotide+-dependent deacetylase, is a member of the sirtuins family and has unique protein deacetylase activity. SIRT1 participates in physiological as well as pathophysiological processes by targeting a wide range of protein substrates and signalings. In this review, we described the latest progress of SIRT1 in pulmonary diseases. We have introduced the basic information and summarized the prominent role of SIRT1 in several lung diseases, such as acute lung injury, acute respiratory distress syndrome, chronic obstructive pulmonary disease, lung cancer, and aging-related diseases.
Collapse
Affiliation(s)
- Yi-Zhu Jiang
- Xiangya Nursing School, Central South University, Changsha, China; Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan, China
| | - Xin-Ran Huang
- Xiangya Nursing School, Central South University, Changsha, China; Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan, China
| | - Jing Chang
- Xiangya Nursing School, Central South University, Changsha, China; Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan, China
| | - Yong Zhou
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan, China
| | - Xiao-Ting Huang
- Xiangya Nursing School, Central South University, Changsha, China.
| |
Collapse
|
3
|
Nicotinamide mononucleotide attenuates isoproterenol-induced cardiac fibrosis by regulating oxidative stress and Smad3 acetylation. Life Sci 2021; 274:119299. [PMID: 33675899 DOI: 10.1016/j.lfs.2021.119299] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Revised: 02/19/2021] [Accepted: 02/22/2021] [Indexed: 01/01/2023]
Abstract
AIMS Cardiac fibrosis is a pathological hallmark of progressive heart diseases currently lacking effective treatment. Nicotinamide mononucleotide (NMN), a member of the vitamin B3 family, is a defined biosynthetic precursor of nicotinamide adenine dinucleotide (NAD+). Its beneficial effects on cardiac diseases are known, but its effects on cardiac fibrosis and the underlying mechanism remain unclear. We aimed to elucidate the protective effect of NMN against cardiac fibrosis and its underlying mechanisms of action. MATERIALS AND METHODS Cardiac fibrosis was induced by isoproterenol (ISO) in mice. NMN was administered by intraperitoneal injection. In vitro, cardiac fibroblasts (CFs) were stimulated by transforming growth factor-beta (TGF-β) with or without NMN and sirtinol, a SIRT1 inhibitor. Levels of cardiac fibrosis, NAD+/SIRT1 alteration, oxidative stress, and Smad3 acetylation were evaluated by real-time polymerase chain reaction, western blots, immunohistochemistry staining, immunoprecipitation, and assay kits. KEY FINDINGS ISO treatment induced cardiac dysfunction, fibrosis, and hypertrophy in vivo, whereas NMN alleviated these changes. Additionally, NMN suppressed CFs activation stimulated by TGF-β in vitro. Mechanistically, NMN restored the NAD+/SIRT1 axis and inhibited the oxidative stress and Smad3 acetylation induced by ISO or TGF-β. However, the protective effects of NMN were partly antagonized by sirtinol in vitro. SIGNIFICANCE NMN could attenuate cardiac fibrosis in vivo and fibroblast activation in vitro by suppressing oxidative stress and Smad3 acetylation in a NAD+/SIRT1-dependent manner.
Collapse
|
4
|
Adiponectin enhances the bioenergetics of cardiac myocytes via an AMPK- and succinate dehydrogenase-dependent mechanism. Cell Signal 2021; 78:109866. [PMID: 33271223 PMCID: PMC9619024 DOI: 10.1016/j.cellsig.2020.109866] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 11/25/2020] [Accepted: 11/26/2020] [Indexed: 12/16/2022]
Abstract
Adiponectin is one of the most abundant circulating hormones, which through adenosine monophosphate-activated protein kinase (AMPK), enhances fatty acid and glucose oxidation, and exerts a cardioprotective effect. However, its effects on cellular bioenergetics have not been explored. We have previously reported that 5-aminoimidazole-4-carboxamide 1-β-D-ribofuranoside (AICAR, an AMPK activator) enhances mitochondrial respiration through a succinate dehydrogenase (SDH or complex II)-dependent mechanism in cardiac myocytes, leading us to predict that Adiponectin would exert a similar effect via activating AMPK. Our results show that Adiponectin enhances basal mitochondrial oxygen consumption rate (OCR), ATP production, and spare respiratory capacity (SRC), which were all abolished by the knockdown of AMPKγ1, inhibition of SDH complex assembly, via the knockdown of the SDH assembly factor 1 (Sdhaf1), or inhibition of SDH activity. Additionally, Adiponectin alleviated hypoxia-induced reductions in OCR and ATP production, in a Sdhaf1-dependent manner, whereas overexpression of Sdhaf1 confirmed its sufficiency for mediating these effects. Importantly, the levels of holoenzyme SDH under the various conditions correlated with OCR. We also show that the effects of Adiponectin, AMPK, Sdhaf1, as well as, SDH complex assembly all required sirtuin 3 (Sirt3). In conclusion, Adiponectin potentiates mitochondrial bioenergetics via promoting SDH complex assembly in an AMPK-, Sdhaf1-, and Sirt3-dependent fashion in cardiac myocytes.
Collapse
|
5
|
Bansod S, Godugu C. Nimbolide ameliorates pancreatic inflammation and apoptosis by modulating NF-κB/SIRT1 and apoptosis signaling in acute pancreatitis model. Int Immunopharmacol 2020; 90:107246. [PMID: 33310297 DOI: 10.1016/j.intimp.2020.107246] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 11/25/2020] [Accepted: 11/25/2020] [Indexed: 12/12/2022]
Abstract
Acute pancreatitis (AP) is a potential gastrointestinal problem most commonly associated with pancreatic inflammation and acinar cells injury. Nimbolide (NB), isolated from the tree Azadirachta indica, possesses antioxidant and anti-inflammatory effects. Here, we aimed to investigate the pancreatic protective effects of NB in ameliorating cerulein-induced pancreatic inflammation and apoptosis in AP model and evaluate the potential mechanism of action. AP was induced in Swiss albino mice by six-hourly intraperitoneal exposures of cerulein (50 µg/kg/hr) and pre-treatment of NB (0.3 and 1 mg/kg) 7 days prior to the cerulein exposure. Various parameters associated with AP in plasma and pancreatic tissues were evaluated. Severity of AP was effectively ameliorated by NB as shown by reducing pancreatic edema, plasma amylase and lipase levels, MPO levels and in cerulein-induced histological damage. Further, the antioxidant effect of NB was associated with a significant inhibition of oxidative-nitrosative stress in Raw 264.7 cells and cerulein-induced AP mice. Moreover, NB suppressed proinflammatory cytokines, iNOS and nitrotyrosine expression. In addition, NB inhibited NF-κB activation and increased SIRT1 expression in cerulein challenged mice. Furthermore, NB also inhibited pancreatic apoptosis by downregulating cleaved caspase 3 and Bax while upregulating Bcl2 expression in cerulein-treated mice. Inhibition of pancreatic inflammation and apoptosis resulted in attenuation of cerulein-induced AP. These results suggest that NB exerts strong anti-pancreatitis effects against cerulein-induced AP by combating inflammatory and apoptosis signaling via SIRT1 activation.
Collapse
Affiliation(s)
- Sapana Bansod
- Department of Regulatory Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad, Telangana, India
| | - Chandraiah Godugu
- Department of Regulatory Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad, Telangana, India.
| |
Collapse
|
6
|
Abdullah A, Mohd Murshid N, Makpol S. Antioxidant Modulation of mTOR and Sirtuin Pathways in Age-Related Neurodegenerative Diseases. Mol Neurobiol 2020; 57:5193-5207. [PMID: 32865663 DOI: 10.1007/s12035-020-02083-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Accepted: 08/19/2020] [Indexed: 12/11/2022]
Abstract
In the human body, cell division and metabolism are expected to transpire uneventfully for approximately 25 years. Then, secondary metabolism and cell damage products accumulate, and ageing phenotypes are acquired, causing the progression of disease. Among these age-related diseases, neurodegenerative diseases have attracted considerable attention because of their irreversibility, the absence of effective treatment and their relationship with social and economic pressures. Mechanistic (formerly mammalian) target of rapamycin (mTOR), sirtuin (SIRT) and insulin/insulin growth factor 1 (IGF1) signalling pathways are among the most important pathways in ageing-associated conditions, such as neurodegeneration. These longevity-related pathways are associated with a diversity of various processes, including metabolism, cognition, stress reaction and brain plasticity. In this review, we discuss the roles of sirtuin and mTOR in ageing and neurodegeneration, with an emphasis on their regulation of autophagy, apoptosis and mitochondrial energy metabolism. The intervention of neurodegeneration using potential antioxidants, including vitamins, phytochemicals, resveratrol, herbals, curcumin, coenzyme Q10 and minerals, specifically aimed at retaining mitochondrial function in the treatment of Alzheimer's disease, Parkinson's disease and Huntington's disease is highlighted.
Collapse
Affiliation(s)
- Asmaa Abdullah
- Department of Biochemistry, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Level 17, Preclinical Building, Jalan Yaacob Latif, Bandar Tun Razak, Cheras, 56000, Kuala Lumpur, Malaysia
| | - Nuraqila Mohd Murshid
- Department of Biochemistry, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Level 17, Preclinical Building, Jalan Yaacob Latif, Bandar Tun Razak, Cheras, 56000, Kuala Lumpur, Malaysia
| | - Suzana Makpol
- Department of Biochemistry, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Level 17, Preclinical Building, Jalan Yaacob Latif, Bandar Tun Razak, Cheras, 56000, Kuala Lumpur, Malaysia.
| |
Collapse
|
7
|
Zuo W, Liu N, Zeng Y, Liu Y, Li B, Wu K, Xiao Y, Liu Q. CD38: A Potential Therapeutic Target in Cardiovascular Disease. Cardiovasc Drugs Ther 2020; 35:815-828. [PMID: 32472237 DOI: 10.1007/s10557-020-07007-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Substantial research has demonstrated the association between cardiovascular disease and the dysregulation of intracellular calcium, ageing, reduction in nicotinamide adenine dinucleotide NAD+ content, and decrease in sirtuin activity. CD38, which comprises the soluble type, type II, and type III, is the main NADase in mammals. This molecule catalyses the production of cyclic adenosine diphosphate ribose (cADPR), nicotinic acid adenine dinucleotide phosphate (NAADP), and adenosine diphosphate ribose (ADPR), which stimulate the release of Ca2+, accompanied by NAD+ consumption and decreased sirtuin activity. Therefore, the relationship between cardiovascular disease and CD38 has been attracting increased attention. In this review, we summarize the structure, regulation, function, targeted drug development, and current research on CD38 in the cardiac context. More importantly, we provide original views about the as yet elusive mechanisms of CD38 action in certain cardiovascular disease models. Based on our review, we predict that CD38 may serve as a novel therapeutic target in cardiovascular disease in the future.
Collapse
Affiliation(s)
- Wanyun Zuo
- Department of Cardiovascular Medicine, The Second Xiangya Hospital of Central South University, No. 139 Middle Renmin Road, Furong District, Changsha, 410011, Hunan, China
| | - Na Liu
- Department of Cardiovascular Medicine, The Second Xiangya Hospital of Central South University, No. 139 Middle Renmin Road, Furong District, Changsha, 410011, Hunan, China
| | - Yunhong Zeng
- Department of Cardiology, Hunan Children's Hospital, No. 86 Ziyuan Road, Yuhua District, Changsha, 410007, Hunan, China
| | - Yaozhong Liu
- Department of Cardiovascular Medicine, The Second Xiangya Hospital of Central South University, No. 139 Middle Renmin Road, Furong District, Changsha, 410011, Hunan, China
| | - Biao Li
- Department of Cardiovascular Medicine, The Second Xiangya Hospital of Central South University, No. 139 Middle Renmin Road, Furong District, Changsha, 410011, Hunan, China
| | - Keke Wu
- Department of Cardiovascular Medicine, The Second Xiangya Hospital of Central South University, No. 139 Middle Renmin Road, Furong District, Changsha, 410011, Hunan, China
| | - Yunbin Xiao
- Department of Cardiology, Hunan Children's Hospital, No. 86 Ziyuan Road, Yuhua District, Changsha, 410007, Hunan, China.
| | - Qiming Liu
- Department of Cardiovascular Medicine, The Second Xiangya Hospital of Central South University, No. 139 Middle Renmin Road, Furong District, Changsha, 410011, Hunan, China.
| |
Collapse
|
8
|
Kiss A, Ráduly AP, Regdon Z, Polgár Z, Tarapcsák S, Sturniolo I, El-Hamoly T, Virág L, Hegedűs C. Targeting Nuclear NAD + Synthesis Inhibits DNA Repair, Impairs Metabolic Adaptation and Increases Chemosensitivity of U-2OS Osteosarcoma Cells. Cancers (Basel) 2020; 12:cancers12051180. [PMID: 32392755 PMCID: PMC7281559 DOI: 10.3390/cancers12051180] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 05/01/2020] [Accepted: 05/04/2020] [Indexed: 12/11/2022] Open
Abstract
Osteosarcoma (OS) is the most common bone tumor in children and adolescents. Modern OS treatment, based on the combination of neoadjuvant chemotherapy (cisplatin + doxorubicin + methotrexate) with subsequent surgical removal of the primary tumor and metastases, has dramatically improved overall survival of OS patients. However, further research is needed to identify new therapeutic targets. Here we report that expression level of the nuclear NAD synthesis enzyme, nicotinamide mononucleotide adenylyltransferase-1 (NMNAT1), increases in U-2OS cells upon exposure to DNA damaging agents, suggesting the involvement of the enzyme in the DNA damage response. Moreover, genetic inactivation of NMNAT1 sensitizes U-2OS osteosarcoma cells to cisplatin, doxorubicin, or a combination of these two treatments. Increased cisplatin-induced cell death of NMNAT1−/− cells showed features of both apoptosis and necroptosis, as indicated by the protective effect of the caspase-3 inhibitor z-DEVD-FMK and the necroptosis inhibitor necrostatin-1. Activation of the DNA damage sensor enzyme poly(ADP-ribose) polymerase 1 (PARP1), a major consumer of NAD+ in the nucleus, was fully blocked by NMNAT1 inactivation, leading to increased DNA damage (phospho-H2AX foci). The PARP inhibitor, olaparib, sensitized wild type but not NMNAT1−/− cells to cisplatin-induced anti-clonogenic effects, suggesting that impaired PARP1 activity is important for chemosensitization. Cisplatin-induced cell death of NMNAT1−/− cells was also characterized by a marked drop in cellular ATP levels and impaired mitochondrial respiratory reserve capacity, highlighting the central role of compromised cellular bioenergetics in chemosensitization by NMNAT1 inactivation. Moreover, NMNAT1 cells also displayed markedly higher sensitivity to cisplatin when grown as spheroids in 3D culture. In summary, our work provides the first evidence that NMNAT1 is a promising therapeutic target for osteosarcoma and possibly other tumors as well.
Collapse
Affiliation(s)
- Alexandra Kiss
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary, (A.K.)
- Doctoral School of Molecular Medicine, University of Debrecen, H-4032 Debrecen, Hungary
| | - Arnold Péter Ráduly
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary, (A.K.)
| | - Zsolt Regdon
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary, (A.K.)
| | - Zsuzsanna Polgár
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary, (A.K.)
| | - Szabolcs Tarapcsák
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary,
| | - Isotta Sturniolo
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary, (A.K.)
| | - Tarek El-Hamoly
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary, (A.K.)
- Drug Radiation Research Department, National Center for Radiation Research and Technology, Atomic Energy Authority, 113701 Cairo, Egypt
| | - László Virág
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary, (A.K.)
- MTA-DE Cell Biology and Signaling Research Group, H-4032 Debrecen, Hungary
| | - Csaba Hegedűs
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary, (A.K.)
| |
Collapse
|
9
|
Dimauro I, Paronetto MP, Caporossi D. Exercise, redox homeostasis and the epigenetic landscape. Redox Biol 2020; 35:101477. [PMID: 32127290 PMCID: PMC7284912 DOI: 10.1016/j.redox.2020.101477] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 02/12/2020] [Accepted: 02/23/2020] [Indexed: 02/07/2023] Open
Abstract
Physical exercise represents one of the strongest physiological stimuli capable to induce functional and structural modifications in all biological systems. Indeed, beside the traditional genetic mechanisms, physical exercise can modulate gene expression through epigenetic modifications, namely DNA methylation, post-translational histone modification and non-coding RNA transcripts. Initially considered as merely damaging molecules, it is now well recognized that both reactive oxygen (ROS) and nitrogen species (RNS) produced under voluntary exercise play an important role as regulatory mediators in signaling processes. While robust scientific evidences highlight the role of exercise-associated redox modifications in modulating gene expression through the genetic machinery, the understanding of their specific impact on epigenomic profile is still at an early stage. This review will provide an overview of the role of ROS and RNS in modulating the epigenetic landscape in the context of exercise-related adaptations. Physical exercise can modulate gene expression through epigenetic modifications. Epigenetic regulation of ROS/RNS generating, sensing and neutralizing enzymes can impact the cellular levels of ROS and RNS. ROS might act as modulators of epigenetic machinery, interfering with DNA methylation, hPTMs and ncRNAs expression. Redox homeostasis might hold a relevant role in the epigenetic landscape modulating exercise-related adaptations.
Collapse
Affiliation(s)
- Ivan Dimauro
- Unit of Biology and Genetics of Movement, Department of Movement, Human and Health Sciences, University of Rome Foro Italico, Piazza Lauro de Bosis 15, 00135, Rome, Italy
| | - Maria Paola Paronetto
- Unit of Biology and Genetics of Movement, Department of Movement, Human and Health Sciences, University of Rome Foro Italico, Piazza Lauro de Bosis 15, 00135, Rome, Italy; Laboratory of Cellular and Molecular Neurobiology, IRCCS Fondazione Santa Lucia, Via Del Fosso di Fiorano, Rome, Italy
| | - Daniela Caporossi
- Unit of Biology and Genetics of Movement, Department of Movement, Human and Health Sciences, University of Rome Foro Italico, Piazza Lauro de Bosis 15, 00135, Rome, Italy.
| |
Collapse
|
10
|
Liu Y, Jia S, Liang X, Dong M, Xu X, Lu C, Wei Y. Prognostic value of Sirtuin1 in acute ischemic stroke and its correlation with functional outcomes. Medicine (Baltimore) 2018; 97:e12959. [PMID: 30544370 PMCID: PMC6310560 DOI: 10.1097/md.0000000000012959] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND The blood-brain barrier is impaired in patients with stroke. The release of protein markers such as Sirtuin1 (SIRTl) into circulation may be useful to assess the prognosis of patients with cerebrovascular disease. In this study, we investigated the predictive value of SIRT1 levels in acute ischemic stroke (AIS) patients. METHODS In all, 101 AIS patients and 38 healthy controls were enrolled, and blood samples were collected within 72 hours of stroke onset. SIRT1 was analyzed using a commercially available enzyme-linked immunosorbent assay kit. On admission, neurological status was assessed by the standardized National Institutes of Health Stroke Scale (NIHSS). Functional outcomes were measured 1 year after admission using the modified Rankin scale. RESULTS Compared with the control group, SIRT1 was significantly increased in the AIS group (0.63 ± 0.75 vs 0.48 ± 0.80 ng/mL; P ≤ 0.05). However, there was no significant correlation between SIRT1 and NIHSS score at admission (r = -0.01, P = .920). In addition, with an unadjusted odds ratio of 0.862 (95% confidence interval 0.495-1.502), SIRT1 was not significantly correlated with functional outcomes. CONCLUSIONS Serum concentrations of SIRT1 have no significant predictive value for favorable functional outcome after acute stroke in our study.
Collapse
Affiliation(s)
- Yang Liu
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University
- Chongqing Key Laboratory of Neurobiology, Chongqing
| | - ShiYu Jia
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University
- Chongqing Key Laboratory of Neurobiology, Chongqing
| | - Xue Liang
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University
- Chongqing Key Laboratory of Neurobiology, Chongqing
| | - MeiXue Dong
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University
- Chongqing Key Laboratory of Neurobiology, Chongqing
| | - XiaoMin Xu
- Department of Neurology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - ChangQi Lu
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University
- Chongqing Key Laboratory of Neurobiology, Chongqing
| | - YouDong Wei
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University
- Chongqing Key Laboratory of Neurobiology, Chongqing
| |
Collapse
|
11
|
Zhou H, Wang H, Ni M, Yue S, Xia Y, Busuttil RW, Kupiec-Weglinski JW, Lu L, Wang X, Zhai Y. Glycogen synthase kinase 3β promotes liver innate immune activation by restraining AMP-activated protein kinase activation. J Hepatol 2018; 69:99-109. [PMID: 29452207 PMCID: PMC6291010 DOI: 10.1016/j.jhep.2018.01.036] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Revised: 01/08/2018] [Accepted: 01/30/2018] [Indexed: 12/22/2022]
Abstract
BACKGROUND & AIMS Glycogen synthase kinase 3β (Gsk3β [Gsk3b]) is a ubiquitously expressed kinase with distinctive functions in different types of cells. Although its roles in regulating innate immune activation and ischaemia and reperfusion injuries (IRIs) have been well documented, the underlying mechanisms remain ambiguous, in part because of the lack of cell-specific tools in vivo. METHODS We created a myeloid-specific Gsk3b knockout (KO) strain to study the function of Gsk3β in macrophages in a murine liver partial warm ischaemia model. RESULTS Compared with controls, myeloid Gsk3b KO mice were protected from IRI, with diminished proinflammatory but enhanced anti-inflammatory immune responses in livers. In bone marrow-derived macrophages, Gsk3β deficiency resulted in an early reduction of Tnf gene transcription but sustained increase of Il10 gene transcription on Toll-like receptor 4 stimulation in vitro. These effects were associated with enhanced AMP-activated protein kinase (AMPK) activation, which led to an accelerated and higher level of induction of the novel innate immune negative regulator small heterodimer partner (SHP [Nr0b2]). The regulatory function of Gsk3β on AMPK activation and SHP induction was confirmed in wild-type bone marrow-derived macrophages with a Gsk3 inhibitor. Furthermore, we found that this immune regulatory mechanism was independent of Gsk3β Ser9 phosphorylation and the phosphoinositide 3-kinase-Akt signalling pathway. In vivo, myeloid Gsk3β deficiency facilitated SHP upregulation by ischaemia-reperfusion in liver macrophages. Treatment of Gsk3b KO mice with either AMPK inhibitor or SHP small interfering RNA before the onset of liver ischaemia restored liver proinflammatory immune activation and IRI in these otherwise protected hosts. Additionally, pharmacological activation of AMPK protected wild-type mice from liver IRI, with reduced proinflammatory immune activation. Inhibition of the AMPK-SHP pathway by liver ischaemia was demonstrated in tumour resection patients. CONCLUSIONS Gsk3β promotes innate proinflammatory immune activation by restraining AMPK activation. LAY SUMMARY Glycogen synthase kinase 3β promotes macrophage inflammatory activation by inhibiting the immune regulatory signalling of AMP-activated protein kinase and the induction of small heterodimer partner. Therefore, therapeutic targeting of glycogen synthase kinase 3β enhances innate immune regulation and protects liver from ischaemia and reperfusion injury.
Collapse
Affiliation(s)
- Haoming Zhou
- Dumont-UCLA Transplant Center, Division of Liver and Pancreas Transplantation, Department of Surgery, David Geffen School of Medicine at the University of California, Los Angeles, CA, USA; Liver Transplantation Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Han Wang
- Dumont-UCLA Transplant Center, Division of Liver and Pancreas Transplantation, Department of Surgery, David Geffen School of Medicine at the University of California, Los Angeles, CA, USA; Liver Transplantation Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Ming Ni
- Dumont-UCLA Transplant Center, Division of Liver and Pancreas Transplantation, Department of Surgery, David Geffen School of Medicine at the University of California, Los Angeles, CA, USA; Liver Transplantation Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Shi Yue
- Liver Transplantation Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Yongxiang Xia
- Liver Transplantation Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Ronald W Busuttil
- Dumont-UCLA Transplant Center, Division of Liver and Pancreas Transplantation, Department of Surgery, David Geffen School of Medicine at the University of California, Los Angeles, CA, USA
| | - Jerzy W Kupiec-Weglinski
- Dumont-UCLA Transplant Center, Division of Liver and Pancreas Transplantation, Department of Surgery, David Geffen School of Medicine at the University of California, Los Angeles, CA, USA
| | - Ling Lu
- Liver Transplantation Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Xuehao Wang
- Liver Transplantation Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China.
| | - Yuan Zhai
- Dumont-UCLA Transplant Center, Division of Liver and Pancreas Transplantation, Department of Surgery, David Geffen School of Medicine at the University of California, Los Angeles, CA, USA.
| |
Collapse
|
12
|
Boslett J, Helal M, Chini E, Zweier JL. Genetic deletion of CD38 confers post-ischemic myocardial protection through preserved pyridine nucleotides. J Mol Cell Cardiol 2018; 118:81-94. [PMID: 29476764 PMCID: PMC6699759 DOI: 10.1016/j.yjmcc.2018.02.015] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Revised: 02/02/2018] [Accepted: 02/20/2018] [Indexed: 12/27/2022]
Abstract
Following the onset of ischemia/reperfusion (I/R), CD38 activation occurs and is associated with depletion of NAD(P)(H) in the heart as well as myocardial injury and endothelial dysfunction. Studies with pharmacological inhibitors suggest that the NADP+-hydrolyzing ability of CD38 can deplete the NAD(P)(H) pools. However, there is a need for more specific studies on the importance of CD38 and its role in the process of endothelial dysfunction and myocardial injury in the post-ischemic heart. Therefore, experiments were performed in hearts of mice with global gene knockout of CD38. Isolated perfused CD38-/- and wild type (WT) mouse hearts were studied to determine the link between CD38 activation, the levels of NADP(H), endothelial dysfunction, and myocardial injury after I/R. Genetic deletion of CD38 preserves the myocardial and endothelial NADP(H) pools compared to WT. Whole heart BH4 levels in CD38-/- hearts were also preserved. Post-ischemic levels of cGMP were greatly depleted in WT hearts, but preserved to near baseline levels in CD38-/- hearts. The preservation of these metabolite pools in CD38-/- hearts was accompanied by near full recovery of NOS-dependent coronary flow, while in WT hearts, severe impairment of endothelial function and NOS uncoupling occurred with decreased NO and enhanced superoxide generation. CD38-/- hearts also exhibited marked protection against I/R with preserved glutathione levels, increased recovery of left ventricular contractile function, decreased myocyte enzyme release, and decreased infarct size. Thus, CD38 activation causes post-ischemic depletion of NADP(H) within the heart, with severe depletion from the endothelium, resulting in endothelial dysfunction and myocardial injury.
Collapse
Affiliation(s)
- James Boslett
- Department of Internal Medicine, Davis Heart & Lung Research Institute, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Moustafa Helal
- Department of Internal Medicine, Davis Heart & Lung Research Institute, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Eduardo Chini
- Signal Transduction Laboratory, Kogod Aging Center, Department of Anesthesiology, Mayo Clinic College of Medicine, Rochester, MN 55905, USA
| | - Jay L Zweier
- Department of Internal Medicine, Davis Heart & Lung Research Institute, College of Medicine, The Ohio State University, Columbus, OH 43210, USA.
| |
Collapse
|
13
|
Kowalska M, Wegierek-Ciuk A, Brzoska K, Wojewodzka M, Meczynska-Wielgosz S, Gromadzka-Ostrowska J, Mruk R, Øvrevik J, Kruszewski M, Lankoff A. Genotoxic potential of diesel exhaust particles from the combustion of first- and second-generation biodiesel fuels-the FuelHealth project. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2017; 24:24223-24234. [PMID: 28889235 PMCID: PMC5655577 DOI: 10.1007/s11356-017-9995-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Accepted: 08/22/2017] [Indexed: 05/27/2023]
Abstract
Epidemiological data indicate that exposure to diesel exhaust particles (DEPs) from traffic emissions is associated with higher risk of morbidity and mortality related to cardiovascular and pulmonary diseases, accelerated progression of atherosclerotic plaques, and possible lung cancer. While the impact of DEPs from combustion of fossil diesel fuel on human health has been extensively studied, current knowledge of DEPs from combustion of biofuels provides limited and inconsistent information about its mutagenicity and genotoxicity, as well as possible adverse health risks. The objective of the present work was to compare the genotoxicity of DEPs from combustion of two first-generation fuels, 7% fatty acid methyl esters (FAME) (B7) and 20% FAME (B20), and a second-generation 20% FAME/hydrotreated vegetable oil (SHB: synthetic hydrocarbon biofuel) fuel. Our results revealed that particulate engine emissions from each type of biodiesel fuel induced genotoxic effects in BEAS-2B and A549 cells, manifested as the increased levels of single-strand breaks, the increased frequencies of micronuclei, or the deregulated expression of genes involved in DNA damage signaling pathways. We also found that none of the tested DEPs showed the induction of oxidative DNA damage and the gamma-H2AX-detectable double-strand breaks. The most pronounced differences concerning the tested particles were observed for the induction of single-strand breaks, with the greatest genotoxicity being associated with the B7-derived DEPs. The differences in other effects between DEPs from the different biodiesel blend percentage and biodiesel feedstock were also observed, but the magnitude of these variations was limited.
Collapse
Affiliation(s)
- Magdalena Kowalska
- Department of Radiobiology and Immunology, Institute of Biology, Jan Kochanowski University, 15 Swietokrzyska Str, 25-406, Kielce, Poland
| | - Aneta Wegierek-Ciuk
- Department of Radiobiology and Immunology, Institute of Biology, Jan Kochanowski University, 15 Swietokrzyska Str, 25-406, Kielce, Poland
| | - Kamil Brzoska
- Center for Radiobiology and Biological Dosimetry, Institute of Nuclear Chemistry and Technology, 16 Dorodna Str, 03-195, Warsaw, Poland
| | - Maria Wojewodzka
- Center for Radiobiology and Biological Dosimetry, Institute of Nuclear Chemistry and Technology, 16 Dorodna Str, 03-195, Warsaw, Poland
| | - Sylwia Meczynska-Wielgosz
- Center for Radiobiology and Biological Dosimetry, Institute of Nuclear Chemistry and Technology, 16 Dorodna Str, 03-195, Warsaw, Poland
| | - Joanna Gromadzka-Ostrowska
- Faculty of Human Nutrition and Consumer Science, Warsaw University of Life Sciences, 166 Nowoursynowska Str, 02-787, Warsaw, Poland
| | - Remigiusz Mruk
- Faculty of Production Engineering, Warsaw University of Life Sciences, 166 Nowoursynowska Str, 02-787, Warsaw, Poland
| | - Johan Øvrevik
- Domain of Infection Control and Environmental Health, Norwegian Institute of Public Health, P.O. Box 4404, Nydalen, 0403, Oslo, Norway
| | - Marcin Kruszewski
- Center for Radiobiology and Biological Dosimetry, Institute of Nuclear Chemistry and Technology, 16 Dorodna Str, 03-195, Warsaw, Poland
- Department of Molecular Biology and Translational Research, Institute of Rural Health, Jaczewskiego 2, 20-090, Lublin, Poland
- Faculty of Medicine, University of Information Technology and Management in Rzeszow, Sucharskiego 2, 35-225, Rzeszow, Poland
| | - Anna Lankoff
- Department of Radiobiology and Immunology, Institute of Biology, Jan Kochanowski University, 15 Swietokrzyska Str, 25-406, Kielce, Poland.
- Center for Radiobiology and Biological Dosimetry, Institute of Nuclear Chemistry and Technology, 16 Dorodna Str, 03-195, Warsaw, Poland.
| |
Collapse
|
14
|
Sirt1-Sirt3 axis regulates human blood-brain barrier permeability in response to ischemia. Redox Biol 2017; 14:229-236. [PMID: 28965081 PMCID: PMC5633840 DOI: 10.1016/j.redox.2017.09.016] [Citation(s) in RCA: 105] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Revised: 09/12/2017] [Accepted: 09/21/2017] [Indexed: 12/22/2022] Open
Abstract
Sirtuin1 (Sirt1) and Sirtuin3 (Sirt3) are two well-characterized members of the silent information regulator 2 (Sir2) family of proteins. Both Sirt1 and Sirt3 have been shown to play vital roles in resistance to cellular stress, but the interaction between these two sirtuins has not been fully determined. In this study, we investigated the role of Sirt1-Sirt3 axis in blood-brain barrier (BBB) permeability after ischemia in vitro. Human brain microvascular endothelial cells and astrocytes were co-cultured to model the BBB in vitro and oxygen and glucose deprivation (OGD) was performed to mimic ischemia. The results of transepithelial electrical resistance (TEER) showed that suppression of Sirt1 via siRNA or salermide significantly decreased BBB permeability, whereas Sirt3 knockdown increased BBB permeability. In addition, Sirt1 was shown to regulate Sirt3 expression after OGD through inhibiting the AMPK-PGC1 pathway. Application of the AMPK inhibitor compound C partially prevented the effects of Sirt1-Sirt3 axis on BBB permeability after OGD. The results of flow cytometry and cytochrome c release demonstrated that Sirt1 and Sirt3 exert opposite effects on OGD-induced apoptosis. Furthermore, suppression of Sirt1 was shown to attenuate mitochondrial reactive oxygen species (ROS) generation, which contribute to the Sirt1-Sirt3 axis-induced regulation of BBB permeability and cell damage. In summary, these findings demonstrate that the Sirt1-Sirt3 axis might act as an important modulator in BBB physiology, and could be a therapeutic target for ischemic stroke via regulating mitochondrial ROS generation.
Collapse
|
15
|
NAD + augmentation ameliorates acute pancreatitis through regulation of inflammasome signalling. Sci Rep 2017; 7:3006. [PMID: 28592850 PMCID: PMC5462749 DOI: 10.1038/s41598-017-03418-0] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Accepted: 04/27/2017] [Indexed: 12/12/2022] Open
Abstract
Acute pancreatitis (AP) is a complicated disease without specific drug therapy. The cofactor nicotinamide adenine dinucleotide (NAD+) is an important regulator of cellular metabolism and homeostasis. However, it remains unclear whether modulation of NAD+ levels has an impact on caerulein-induced AP. Therefore, in this study, we investigated the effect of increased cellular NAD+ levels on caerulein-induced AP. We demonstrated for the first time that the activities and expression of SIRT1 were suppressed by reduction of intracellular NAD+ levels and the p53-microRNA-34a pathway in caerulein-induced AP. Moreover, we confirmed that the increase of cellular NAD+ by NQO1 enzymatic action using the substrate β-Lapachone suppressed caerulein-induced AP with down-regulating TLR4-mediated inflammasome signalling, and thereby reducing the inflammatory responses and pancreatic cell death. These results suggest that pharmacological stimulation of NQO1 could be a promising therapeutic strategy to protect against pathological tissue damage in AP.
Collapse
|
16
|
Hill LJ, Williams AC. Meat Intake and the Dose of Vitamin B 3 - Nicotinamide: Cause of the Causes of Disease Transitions, Health Divides, and Health Futures? Int J Tryptophan Res 2017; 10:1178646917704662. [PMID: 28579801 PMCID: PMC5419340 DOI: 10.1177/1178646917704662] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Accepted: 03/15/2017] [Indexed: 12/26/2022] Open
Abstract
Meat and vitamin B3 - nicotinamide - intake was high during hunter-gatherer times. Intake then fell and variances increased during and after the Neolithic agricultural revolution. Health, height, and IQ deteriorated. Low dietary doses are buffered by 'welcoming' gut symbionts and tuberculosis that can supply nicotinamide, but this co-evolved homeostatic metagenomic strategy risks dysbioses and impaired resistance to pathogens. Vitamin B3 deficiency may now be common among the poor billions on a low-meat diet. Disease transitions to non-communicable inflammatory disorders (but longer lives) may be driven by positive 'meat transitions'. High doses of nicotinamide lead to reduced regulatory T cells and immune intolerance. Loss of no longer needed symbiotic 'old friends' compounds immunological over-reactivity to cause allergic and auto-immune diseases. Inhibition of nicotinamide adenine dinucleotide consumers and loss of methyl groups or production of toxins may cause cancers, metabolic toxicity, or neurodegeneration. An optimal dosage of vitamin B3 could lead to better health, but such a preventive approach needs more equitable meat distribution. Some people may require personalised doses depending on genetic make-up or, temporarily, when under stress.
Collapse
Affiliation(s)
- Lisa J Hill
- Neuroscience and Ophthalmology Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
| | - Adrian C Williams
- Department of Neurology, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| |
Collapse
|
17
|
Monocyte-derived extracellular Nampt-dependent biosynthesis of NAD(+) protects the heart against pressure overload. Sci Rep 2015; 5:15857. [PMID: 26522369 PMCID: PMC4629142 DOI: 10.1038/srep15857] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2015] [Accepted: 10/05/2015] [Indexed: 12/31/2022] Open
Abstract
Nicotinamide phosphoribosyltransferase (Nampt) catalyzes the rate-limiting step in the salvage pathway for nicotinamide adenine dinucleotide (NAD+) biosynthesis, and thereby regulates the deacetylase activity of sirtuins. Here we show accommodative regulation of myocardial NAD+ by monocyte-derived extracellular Nampt (eNampt), which is essential for hemodynamic compensation to pressure overload. Although intracellular Nampt (iNampt) expression was decreased in pressure-overloaded hearts, myocardial NAD+ concentration and Sirt1 activity were preserved. In contrast, iNampt was up-regulated in spleen and monocytes, and circulating eNampt protein and nicotinamide mononucleotide (NMN), a key precursor of NAD+, were significantly increased. Pharmacological inhibition of Nampt by FK866 or depletion of monocytes/macrophages by clodronate liposomes disrupted the homeostatic mechanism of myocardial NAD+ levels and NAD+-dependent Sirt1 activity, leading to susceptibility to cardiomyocyte apoptosis and cardiac decompensation in pressure-overloaded mice. These biochemical and hemodynamic defects were prevented by systemic administration of NMN. Our studies uncover a crucial role of monocyte-derived eNampt in myocardial adaptation to pressure overload, and highlight a potential intervention controlling myocardial NAD+ against heart failure.
Collapse
|
18
|
Li J, Dou X, Li S, Zhang X, Zeng Y, Song Z. Nicotinamide ameliorates palmitate-induced ER stress in hepatocytes via cAMP/PKA/CREB pathway-dependent Sirt1 upregulation. BIOCHIMICA ET BIOPHYSICA ACTA 2015; 1853:2929-2936. [PMID: 26352206 PMCID: PMC5445659 DOI: 10.1016/j.bbamcr.2015.09.003] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/20/2015] [Revised: 08/14/2015] [Accepted: 09/03/2015] [Indexed: 02/05/2023]
Abstract
Nicotinamide (NAM) is the amide of nicotinic acid and a predominant precursor for NAD(+) biosynthesis via the salvage pathway. Sirt1 is a NAD(+)-dependent deacetylase, playing an important role in regulating cellular functions. Although hepatoprotective effect of NAM has been reported, the underlying mechanism remains elusive. ER stress, induced by saturated fatty acids, in specific palmitate, plays a pathological role in the development of nonalcoholic fatty liver disease. This study aims to determine the effect of NAM on palmitate-induced ER stress in hepatocytes and to elucidate molecular mechanisms behind. Both HepG2 cells and primary mouse hepatocytes were exposed to palmitate (conjugated to BSA at a 2:1 M ratio), NAM, or their combination for different durations. Cellular NAD(+) level, Sirt1 expression/activity, ER stress, as well as cAMP/PKA/CREB pathway activation were determined. NAM increased Sirt1 expression and enzymatic activity, which contributes to the ameliorative effect of NAM on palmitate-triggered ER stress. NAM increased intracellular NAD(+) level in hepatocytes, however, blocking the salvage pathway, a pathway for NAD(+) synthesis from NAM, only partially prevented NAM-induced Sirt1 upregulation while completely prevented NAD+ increase in response to NAM. Further mechanistic investigations revealed that NAM elevated intracellular cAMP level via suppressing PDE activity, leading to downstream PKA and CREB activation. Importantly, cAMP/PKA/CREB pathway blockade abolished not only NAM-induced Sirt1 upregulation, but also its protective effect against ER stress. Our results demonstrate that NAM protects hepatocytes against palmitate-induced ER stress in hepatocytes via upregulating Sirt1. Activation of the cAMP/PKA/CREB pathway plays a key role in NAM-induced Sirt1 upregulation.
Collapse
Affiliation(s)
- Jiaxin Li
- Department of Kinesiology and Nutrition, University of Illinois at Chicago, Chicago, IL60612, USA; Department of Liver Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, PR China
| | - Xiaobing Dou
- Department of Kinesiology and Nutrition, University of Illinois at Chicago, Chicago, IL60612, USA; College of Life Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, PR China
| | - Songtao Li
- Department of Kinesiology and Nutrition, University of Illinois at Chicago, Chicago, IL60612, USA
| | - Ximei Zhang
- Department of Kinesiology and Nutrition, University of Illinois at Chicago, Chicago, IL60612, USA
| | - Yong Zeng
- Department of Liver Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, PR China
| | - Zhenyuan Song
- Department of Kinesiology and Nutrition, University of Illinois at Chicago, Chicago, IL60612, USA; College of Life Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, PR China.
| |
Collapse
|
19
|
Kim HJ, Pandit A, Oh GS, Shen A, Lee SB, Khadka D, Lee S, Shim H, Yang SH, Cho EY, Kwak TH, Choe SK, Park R, So HS. Dunnione ameliorates cisplatin ototoxicity through modulation of NAD(+) metabolism. Hear Res 2015; 333:235-246. [PMID: 26341473 DOI: 10.1016/j.heares.2015.08.017] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2015] [Revised: 08/25/2015] [Accepted: 08/27/2015] [Indexed: 12/14/2022]
Abstract
Ototoxicity is an important issue in patients receiving cisplatin chemotherapy. Numerous studies have demonstrated that cisplatin-induced ototoxicity is related to oxidative stress and DNA damage. However, the precise mechanism underlying cisplatin-associated ototoxicity is still unclear. The cofactor nicotinamide adenine dinucleotide (NAD(+)) has emerged as an important regulator of energy metabolism and cellular homeostasis. Here, we demonstrate that the levels and activities of sirtuin-1 (SIRT1) are suppressed by the reduction of intracellular NAD(+) levels in cisplatin-mediated ototoxicity. We provide evidence that the decreases in SIRT1 activity and expression facilitated by increasing poly(ADP-ribose) polymerase-1 (PARP-1) activation and microRNA-34a levels through cisplatin-mediated p53 activation aggravate the associated ototoxicity. Furthermore, we show that the induction of cellular NAD(+) levels using dunnione, which targets intracellular NQO1, prevents the toxic effects of cisplatin through the regulation of PARP-1 and SIRT1 activity. These results suggest that direct modulation of cellular NAD(+) levels by pharmacological agents could be a promising therapeutic approach for protection from cisplatin-induced ototoxicity.
Collapse
Affiliation(s)
- Hyung-Jin Kim
- Center for Metabolic Function Regulation & Department of Microbiology, Wonkwang University School of Medicine, Iksan, Jeonbuk, 570-749, Republic of Korea
| | - Arpana Pandit
- Center for Metabolic Function Regulation & Department of Microbiology, Wonkwang University School of Medicine, Iksan, Jeonbuk, 570-749, Republic of Korea
| | - Gi-Su Oh
- Center for Metabolic Function Regulation & Department of Microbiology, Wonkwang University School of Medicine, Iksan, Jeonbuk, 570-749, Republic of Korea
| | - AiHua Shen
- Center for Metabolic Function Regulation & Department of Microbiology, Wonkwang University School of Medicine, Iksan, Jeonbuk, 570-749, Republic of Korea
| | - Su-Bin Lee
- Center for Metabolic Function Regulation & Department of Microbiology, Wonkwang University School of Medicine, Iksan, Jeonbuk, 570-749, Republic of Korea
| | - Dipendra Khadka
- Center for Metabolic Function Regulation & Department of Microbiology, Wonkwang University School of Medicine, Iksan, Jeonbuk, 570-749, Republic of Korea
| | - SeungHoon Lee
- Center for Metabolic Function Regulation & Department of Microbiology, Wonkwang University School of Medicine, Iksan, Jeonbuk, 570-749, Republic of Korea
| | - Hyeok Shim
- Department of Internal Medicine, Wonkwang University School of Medicine, Iksan, Jeonbuk, 570-749, Republic of Korea
| | - Sei-Hoon Yang
- Department of Internal Medicine, Wonkwang University School of Medicine, Iksan, Jeonbuk, 570-749, Republic of Korea
| | - Eun-Young Cho
- Department of Internal Medicine, Wonkwang University School of Medicine, Iksan, Jeonbuk, 570-749, Republic of Korea
| | - Tae Hwan Kwak
- PAEAN Biotechnology, 160 Techno-2 Street, Yuseong-gu, Daejeon, 305-500, Republic of Korea
| | - Seong-Kyu Choe
- Center for Metabolic Function Regulation & Department of Microbiology, Wonkwang University School of Medicine, Iksan, Jeonbuk, 570-749, Republic of Korea
| | - Raekil Park
- Center for Metabolic Function Regulation & Department of Microbiology, Wonkwang University School of Medicine, Iksan, Jeonbuk, 570-749, Republic of Korea
| | - Hong-Seob So
- Center for Metabolic Function Regulation & Department of Microbiology, Wonkwang University School of Medicine, Iksan, Jeonbuk, 570-749, Republic of Korea.
| |
Collapse
|
20
|
Mitochondrial complex II is a source of the reserve respiratory capacity that is regulated by metabolic sensors and promotes cell survival. Cell Death Dis 2015. [PMID: 26225774 PMCID: PMC4650745 DOI: 10.1038/cddis.2015.202] [Citation(s) in RCA: 195] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The survival of a cell depends on its ability to meet its energy requirements. We hypothesized that the mitochondrial reserve respiratory capacity (RRC) of a cell is a critical component of its bioenergetics that can be utilized during an increase in energy demand, thereby, enhancing viability. Our goal was to identify the elements that regulate and contribute to the development of RRC and its involvement in cell survival. The results show that activation of metabolic sensors, including pyruvate dehydrogenase and AMP-dependent kinase, increases cardiac myocyte RRC via a Sirt3-dependent mechanism. Notably, we identified mitochondrial complex II (cII) as a target of these metabolic sensors and the main source of RRC. Moreover, we show that RRC, via cII, correlates with enhanced cell survival after hypoxia. Thus, for the first time, we show that metabolic sensors via Sirt3 maximize the cellular RRC through activating cII, which enhances cell survival after hypoxia.
Collapse
|
21
|
Pereira YCL, do Nascimento GC, Iyomasa DM, Iyomasa MM. Muscle characterization of reactive oxygen species in oral diseases. Acta Odontol Scand 2015; 73:81-6. [PMID: 25205230 DOI: 10.3109/00016357.2014.954267] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
IMPORTANCE AND OBJECTIVE Reactive Oxygen Species (ROS) are oxygen-derived molecules that are unstable and highly reactive. They are important signaling mediators of biological processes. In contrast, excessive ROS generation, defective oxidant scavenging or both have been implicated in the pathogenesis of several conditions. This biological paradox of ROS function contributes to the integrity of cells and tissues. So, the aim of this review was examined for published literature related to 'reactive oxygen species and dentistry and muscle'. MATERIALS AND METHODS A PubMed search was performed by using the following key words: 'reactive oxygen species and dentistry and muscle'. RESULTS Involvement of ROS in pathologic conditions can be highlighted in oral diseases like periodontitis, orofacial pain, temporomandibular disorders and oral cancer. Also, several studies have correlated the increase in ROS production with the initiation of the muscle fatigue process and the process of muscle injury. However, studies evaluating the relation of ROS and orofacial muscles, which can prove very important to understand the fatigue muscle in this region during oral movements, have not yet been conducted. CONCLUSIONS It is concluded that the data on skeletal muscles, especially those of mastication, are not commonly published in this data source; therefore, further studies in this field are strongly recommended.
Collapse
|
22
|
Oh GS, Kim HJ, Shen A, Lee SB, Khadka D, Pandit A, So HS. Cisplatin-induced Kidney Dysfunction and Perspectives on Improving Treatment Strategies. Electrolyte Blood Press 2014; 12:55-65. [PMID: 25606044 PMCID: PMC4297704 DOI: 10.5049/ebp.2014.12.2.55] [Citation(s) in RCA: 155] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2014] [Accepted: 12/05/2014] [Indexed: 01/14/2023] Open
Abstract
Cisplatin is one of the most widely used and highly effective drug for the treatment of various solid tumors; however, it has dose-dependent side effects on the kidney, cochlear, and nerves. Nephrotoxicity is the most well-known and clinically important toxicity. Numerous studies have demonstrated that several mechanisms, including oxidative stress, DNA damage, and inflammatory responses, are closely associated with cisplatin-induced nephrotoxicity. Even though the establishment of cisplatin-induced nephrotoxicity can be alleviated by diuretics and pre-hydration of patients, the prevalence of cisplatin nephrotoxicity is still high, occurring in approximately one-third of patients who have undergone cisplatin therapy. Therefore it is imperative to develop treatments that will ameliorate cisplatin-nephrotoxicity. In this review, we discuss the mechanisms of cisplatin-induced renal toxicity and the new strategies for protecting the kidneys from the toxic effects without lowering the tumoricidal activity.
Collapse
Affiliation(s)
- Gi-Su Oh
- Center for Metabolic Function Regulation, Department of Microbiology, Wonkwang University School of Medicine, Iksan, Jeonbuk, Korea
| | - Hyung-Jin Kim
- Center for Metabolic Function Regulation, Department of Microbiology, Wonkwang University School of Medicine, Iksan, Jeonbuk, Korea
| | - AiHua Shen
- Center for Metabolic Function Regulation, Department of Microbiology, Wonkwang University School of Medicine, Iksan, Jeonbuk, Korea
| | - Su Bin Lee
- Center for Metabolic Function Regulation, Department of Microbiology, Wonkwang University School of Medicine, Iksan, Jeonbuk, Korea
| | - Dipendra Khadka
- Center for Metabolic Function Regulation, Department of Microbiology, Wonkwang University School of Medicine, Iksan, Jeonbuk, Korea
| | - Arpana Pandit
- Center for Metabolic Function Regulation, Department of Microbiology, Wonkwang University School of Medicine, Iksan, Jeonbuk, Korea
| | - Hong-Seob So
- Center for Metabolic Function Regulation, Department of Microbiology, Wonkwang University School of Medicine, Iksan, Jeonbuk, Korea
| |
Collapse
|
23
|
Chapalamadugu KC, Panguluri SK, Bennett ES, Kolliputi N, Tipparaju SM. High level of oxygen treatment causes cardiotoxicity with arrhythmias and redox modulation. Toxicol Appl Pharmacol 2014; 282:100-7. [PMID: 25447406 DOI: 10.1016/j.taap.2014.10.019] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2014] [Revised: 10/24/2014] [Accepted: 10/29/2014] [Indexed: 01/08/2023]
Abstract
Hyperoxia exposure in mice leads to cardiac hypertrophy and voltage-gated potassium (Kv) channel remodeling. Because redox balance of pyridine nucleotides affects Kv function and hyperoxia alters cellular redox potential, we hypothesized that hyperoxia exposure leads to cardiac ion channel disturbances and redox changes resulting in arrhythmias. In the present study, we investigated the electrical changes and redox abnormalities caused by 72h hyperoxia treatment in mice. Cardiac repolarization changes were assessed by acquiring electrocardiogram (ECG) and cardiac action potentials (AP). Biochemical assays were employed to identify the pyridine nucleotide changes, Kv1.5 expression and myocardial injury. Hyperoxia treatment caused marked bradycardia, arrhythmia and significantly prolonged (ms) the, RR (186.2 ± 10.7 vs. 146.4 ± 6.2), PR (46.8 ± 3.1 vs. 39.3 ± 1.6), QRS (10.8 ± 0.6 vs. 8.5 ± 0.2), QTc (57.1 ± 3.5 vs. 40 ± 1.4) and JT (13.4 ± 2.1 vs. 7.0 ± 0.5) intervals, when compared with normoxia group. Hyperoxia treatment also induced significant increase in cardiac action potential duration (APD) (ex-APD90; 73.8 ± 9.5 vs. 50.9 ± 3.1 ms) and elevated levels of serum markers of myocardial injury; cardiac troponin I (TnI) and lactate dehydrogenase (LDH). Hyperoxia exposure altered cardiac levels of mRNA/protein expression of; Kv1.5, Kvβ subunits and SiRT1, and increased ratios of reduced pyridine nucleotides (NADH/NAD & NADPH/NADP). Inhibition of SiRT1 in H9C2 cells using Splitomicin resulted in decreased SiRT1 and Kv1.5 expression, suggesting that SiRT1 may mediate Kv1.5 downregulation. In conclusion, the cardiotoxic effects of hyperoxia exposure involve ion channel disturbances and redox changes resulting in arrhythmias.
Collapse
Affiliation(s)
- Kalyan C Chapalamadugu
- Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, Tampa, FL, USA
| | - Siva K Panguluri
- Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, Tampa, FL, USA
| | - Eric S Bennett
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Narasaiah Kolliputi
- Division of Allergy and Immunology, Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Srinivas M Tipparaju
- Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, Tampa, FL, USA.
| |
Collapse
|
24
|
Kim HJ, Oh GS, Shen A, Lee SB, Choe SK, Kwon KB, Lee S, Seo KS, Kwak TH, Park R, So HS. Augmentation of NAD(+) by NQO1 attenuates cisplatin-mediated hearing impairment. Cell Death Dis 2014; 5:e1292. [PMID: 24922076 PMCID: PMC4611728 DOI: 10.1038/cddis.2014.255] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2014] [Revised: 04/18/2014] [Accepted: 05/09/2014] [Indexed: 12/20/2022]
Abstract
Cisplatin (cis-diaminedichloroplatinum-II) is an extensively used chemotherapeutic agent, and one of its most adverse effects is ototoxicity. A number of studies have demonstrated that these effects are related to oxidative stress and DNA damage. However, the precise mechanism underlying cisplatin-associated ototoxicity is still unclear. The cofactor nicotinamide adenine dinucleotide (NAD(+)) has emerged as a key regulator of cellular energy metabolism and homeostasis. Here, we demonstrate for the first time that, in cisplatin-mediated ototoxicity, the levels and activities of SIRT1 are suppressed by the reduction of intracellular NAD(+) levels. We provide evidence that the decrease in SIRT1 activity and expression facilitated by increasing poly(ADP-ribose) transferase (PARP)-1 activation and microRNA-34a through p53 activation aggravates cisplatin-mediated ototoxicity. Moreover, we show that the induction of cellular NAD(+) levels using β-lapachone (β-Lap), whose intracellular target is NQO1, prevents the toxic effects of cisplatin through the regulation of PARP-1 and SIRT1 activity. These results suggest that direct modulation of cellular NAD(+) levels by pharmacological agents could be a promising therapeutic approach for protection from cisplatin-induced ototoxicity.
Collapse
Affiliation(s)
- H-J Kim
- Center for Metabolic Function Regulation, Department of Microbiology, Wonkwang University School of Medicine, Jeonbuk, Republic of Korea
| | - G-S Oh
- Center for Metabolic Function Regulation, Department of Microbiology, Wonkwang University School of Medicine, Jeonbuk, Republic of Korea
| | - A Shen
- 1] Center for Metabolic Function Regulation, Department of Microbiology, Wonkwang University School of Medicine, Jeonbuk, Republic of Korea [2] BK21plus Program and Department of Smart Life-Care Convergence, Wonkwang University Graduate School, Jeonbuk, Republic of Korea
| | - S-B Lee
- 1] Center for Metabolic Function Regulation, Department of Microbiology, Wonkwang University School of Medicine, Jeonbuk, Republic of Korea [2] BK21plus Program and Department of Smart Life-Care Convergence, Wonkwang University Graduate School, Jeonbuk, Republic of Korea
| | - S-K Choe
- Center for Metabolic Function Regulation, Department of Microbiology, Wonkwang University School of Medicine, Jeonbuk, Republic of Korea
| | - K-B Kwon
- 1] Center for Metabolic Function Regulation, Department of Microbiology, Wonkwang University School of Medicine, Jeonbuk, Republic of Korea [2] BK21plus Program and Department of Smart Life-Care Convergence, Wonkwang University Graduate School, Jeonbuk, Republic of Korea [3] Department of Oriental Medical Physiology, College of Korean Medicine, Wonkwang University, Jeonbuk, Republic of Korea
| | - S Lee
- Life Science Research Center, KT&G Life Sciences, Suwon, Republic of Korea
| | - K-S Seo
- Life Science Research Center, KT&G Life Sciences, Suwon, Republic of Korea
| | - T H Kwak
- Life Science Research Center, KT&G Life Sciences, Suwon, Republic of Korea
| | - R Park
- 1] Center for Metabolic Function Regulation, Department of Microbiology, Wonkwang University School of Medicine, Jeonbuk, Republic of Korea [2] BK21plus Program and Department of Smart Life-Care Convergence, Wonkwang University Graduate School, Jeonbuk, Republic of Korea
| | - H-S So
- 1] Center for Metabolic Function Regulation, Department of Microbiology, Wonkwang University School of Medicine, Jeonbuk, Republic of Korea [2] BK21plus Program and Department of Smart Life-Care Convergence, Wonkwang University Graduate School, Jeonbuk, Republic of Korea
| |
Collapse
|
25
|
Affiliation(s)
- Bradford G Hill
- From the Institute of Molecular Cardiology, Department of Medicine, Diabetes and Obesity Center, Department of Biochemistry and Molecular Biology, and Department of Physiology and Biophysics, University of Louisville, KY
| |
Collapse
|
26
|
Christodoulou MS, Thomas A, Poulain S, Vidakovic M, Lahtela-Kakkonen M, Matulis D, Bertrand P, Bartova E, Blanquart C, Mikros E, Fokialakis N, Passarella D, Benhida R, Martinet N. Can we use the epigenetic bioactivity of caloric restriction and phytochemicals to promote healthy ageing? MEDCHEMCOMM 2014. [DOI: 10.1039/c4md00268g] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Why is it relevant to propose epigenetic “Nutricures” to prevent diseases linked with ageing?
Collapse
|
27
|
Schluesener JK, Schluesener H. Plant polyphenols in the treatment of age-associated diseases: revealing the pleiotropic effects of icariin by network analysis. Mol Nutr Food Res 2013; 58:49-60. [PMID: 24311544 DOI: 10.1002/mnfr.201300409] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2013] [Revised: 10/18/2013] [Accepted: 10/30/2013] [Indexed: 12/15/2022]
Abstract
Polyphenols are a broad class of compounds. Some are ingested in substantial quantities from nutritional sources, more are produced by medicinal plants, and some of them are taken as drugs. It is becoming clear, that a single polyphenol is impacting several cellular pathways. Thus, a network approach is becoming feasible, describing the interaction of a single polyphenol with cellular networks. Here we have selected icariin to draw a prototypic network of icariin activities. Icariin appears to be a promising drug to treat major age-related diseases, like neurodegeneration, memory and depressive disorders, chronic inflammation, diabetes, and osteoporosis. It interacts with several relevant pathways, like PDE, TGF-ß, MAPK, PPAR, NOS, IGF, Sirtuin, and others. Such networks will be useful to future comparative studies of complex effects of polyphenols.
Collapse
Affiliation(s)
- Jan Kevin Schluesener
- Division of Immunopathology of the Nervous System, Department of Neuropathology, Institute of Pathology and Neuropathology, University of Tuebingen, Tuebingen, Germany
| | | |
Collapse
|
28
|
Oh GS, Kim HJ, Choi JH, Shen A, Choe SK, Karna A, Lee SH, Jo HJ, Yang SH, Kwak TH, Lee CH, Park R, So HS. Pharmacological activation of NQO1 increases NAD⁺ levels and attenuates cisplatin-mediated acute kidney injury in mice. Kidney Int 2013; 85:547-60. [PMID: 24025646 PMCID: PMC3944666 DOI: 10.1038/ki.2013.330] [Citation(s) in RCA: 118] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2013] [Revised: 06/10/2013] [Accepted: 06/13/2013] [Indexed: 12/12/2022]
Abstract
Cisplatin is a widely used chemotherapeutic agent for the treatment of various tumors. In addition to its antitumor activity, cisplatin affects normal cells and may induce adverse effects, such as ototoxicity, nephrotoxicity, and neuropathy. Various mechanisms, such as DNA adduct formation, mitochondrial dysfunction, oxidative stress, and inflammatory responses, are critically involved in cisplatin-induced adverse effects. As NAD+ is a cofactor for various enzymes associated with cellular homeostasis, we studied the effects of increased NAD+ levels by means of NAD(P)H:quinone oxidoreductase 1 (NQO1) activation using a known pharmacological activator (β-lapachone) in wild-type and NQO1−/− mice on cisplatin-induced renal dysfunction in vivo. The intracellular NAD+/NADH ratio in renal tissues was significantly increased in wild-type mice co-treated with cisplatin and β-lapachone compared with the ratio in mice treated with cisplatin alone. Inflammatory cytokines and biochemical markers for renal damage were significantly attenuated by β-lapachone co-treatment compared with those in the cisplatin alone group. Notably, the protective effects of β-lapachone in wild-type mice were completely abrogated in NQO1−/− mice. Moreover, β-lapachone enhanced the tumoricidal action of cisplatin in a xenograft tumor model. Thus, intracellular regulation of NAD+ levels through NQO1 activation might be a promising therapeutic target for the protection of cisplatin-induced acute kidney injury.
Collapse
Affiliation(s)
- Gi-Su Oh
- Center for Metabolic Function Regulation, Department of Microbiology, Wonkwang University School of Medicine, Iksan, Republic of Korea
| | - Hyung-Jin Kim
- Center for Metabolic Function Regulation, Department of Microbiology, Wonkwang University School of Medicine, Iksan, Republic of Korea
| | - Jae-Hyuck Choi
- Center for Metabolic Function Regulation, Department of Microbiology, Wonkwang University School of Medicine, Iksan, Republic of Korea
| | - Aihua Shen
- Center for Metabolic Function Regulation, Department of Microbiology, Wonkwang University School of Medicine, Iksan, Republic of Korea
| | - Seong-Kyu Choe
- Center for Metabolic Function Regulation, Department of Microbiology, Wonkwang University School of Medicine, Iksan, Republic of Korea
| | - Anzani Karna
- Center for Metabolic Function Regulation, Department of Microbiology, Wonkwang University School of Medicine, Iksan, Republic of Korea
| | - Seung Hoon Lee
- Life Science Research Center, KT&G Life Sciences, Suwon, Republic of Korea
| | - Hyang-Jeong Jo
- Department of Pathology, Kunsan Medical Center of Wonkwang University Hospital, Kunsan, Republic of Korea
| | - Sei-Hoon Yang
- Department of Internal Medicine, Wonkwang University School of Medicine, Iksan, Republic of Korea
| | - Tae Hwan Kwak
- Life Science Research Center, KT&G Life Sciences, Suwon, Republic of Korea
| | - Chul-Ho Lee
- Laboratory Animal Center, Korea Research Institute of Bioscience and Biotechnology, University of Science and Technology, Daejeon, Republic of Korea
| | - Raekil Park
- Center for Metabolic Function Regulation, Department of Microbiology, Wonkwang University School of Medicine, Iksan, Republic of Korea
| | - Hong-Seob So
- Center for Metabolic Function Regulation, Department of Microbiology, Wonkwang University School of Medicine, Iksan, Republic of Korea
| |
Collapse
|
29
|
SIRT1 Regulation Modulates Stroke Outcome. Transl Stroke Res 2013; 4:663-71. [DOI: 10.1007/s12975-013-0277-y] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2013] [Revised: 07/19/2013] [Accepted: 07/23/2013] [Indexed: 12/15/2022]
|
30
|
Abstract
In contrast to their role in cell types with higher energy demands, mitochondria in endothelial cells primarily function in signaling cellular responses to environmental cues. This article provides an overview of key aspects of mitochondrial biology in endothelial cells, including subcellular location, biogenesis, dynamics, autophagy, reactive oxygen species production and signaling, calcium homeostasis, regulated cell death, and heme biosynthesis. In each section, we introduce key concepts and then review studies showing the importance of that mechanism to endothelial control of vasomotor tone, angiogenesis, and/or inflammatory activation. We particularly highlight the small number of clinical and translational studies that have investigated each mechanism in human subjects. Finally, we review interventions that target different aspects of mitochondrial function and their effects on endothelial function. The ultimate goal of such research is the identification of new approaches for therapy. The reviewed studies make it clear that mitochondria are important in endothelial physiology and pathophysiology. A great deal of work will be needed, however, before mitochondria-directed therapies are available for the prevention and treatment of cardiovascular disease.
Collapse
Affiliation(s)
- Matthew A Kluge
- Evans Department of Medicine, Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA 02118, USA
| | | | | |
Collapse
|
31
|
Effects of caloric restriction on cardiac oxidative stress and mitochondrial bioenergetics: potential role of cardiac sirtuins. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2013; 2013:528935. [PMID: 23577224 PMCID: PMC3614061 DOI: 10.1155/2013/528935] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/10/2013] [Accepted: 02/18/2013] [Indexed: 01/25/2023]
Abstract
The biology of aging has not been fully clarified, but the free radical theory of aging is one of the strongest aging theories proposed to date. The free radical theory has been expanded to the oxidative stress theory, in which mitochondria play a central role in the development of the aging process because of their critical roles in bioenergetics, oxidant production, and regulation of cell death. A decline in cardiac mitochondrial function associated with the accumulation of oxidative damage might be responsible, at least in part, for the decline in cardiac performance with age. In contrast, lifelong caloric restriction can attenuate functional decline with age, delay the onset of morbidity, and extend lifespan in various species. The effect of caloric restriction appears to be related to a reduction in cellular damage induced by reactive oxygen species. There is increasing evidence that sirtuins play an essential role in the reduction of mitochondrial oxidative stress during caloric restriction. We speculate that cardiac sirtuins attenuate the accumulation of oxidative damage associated with age by modifying specific mitochondrial proteins posttranscriptionally. Therefore, the distinct role of each sirtuin in the heart subjected to caloric restriction should be clarified to translate sirtuin biology into clinical practice.
Collapse
|
32
|
Circulation Research thematic synopsis: mitochondria. Circ Res 2013; 112:e55-67. [PMID: 23493305 DOI: 10.1161/circresaha.113.301165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|