1
|
Jiang Q, Chen X, Gong K, Xu Z, Chen L, Zhang F. M6a demethylase FTO regulates the oxidative stress, mitochondrial biogenesis of cardiomyocytes and PGC-1a stability in myocardial ischemia-reperfusion injury. Redox Rep 2025; 30:2454892. [PMID: 39869517 PMCID: PMC11774161 DOI: 10.1080/13510002.2025.2454892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2025] Open
Abstract
OBJECTIVE Myocardial ischemia-reperfusion injury (MIRI) is a highly complex disease with high morbidity and mortality. Studying the molecular mechanism of MIRI and discovering new targets are crucial for the future treatment of MIRI. METHODS We constructed the MIRI rat model and hypoxia/reoxygenation (H/R) injury cardiomyocytes model. RT-PCR and Western blot were used to investigate the expression of the fat mass and obesity-associated (FTO) gene. Electrocardiogram, echocardiography, triphenyltetrazolium chloride (TTC) staining and hematoxylin-eosin (HE) staining were used to assess the model and the effect of FTO overexpression. The generation of reactive oxygen species (ROS) and the levels of superoxide dismutase (SOD2), mitochondrial transcription factor (TFAM) and cytochrome c oxidase I (COXI) were detected to assess the oxidative stress and mitochondrial biogenesis. RNA immunoprecipitation (RIP) and RNA pulldown assays were used to identify the interaction of FTO and PGC-1a. The m6A dot blot, methylated RNA immunoprecipitation PCR (MeRIP-PCR) and RNA stability analysis were used to analyze the regulation of methylation of PGC-1a by FTO. RESULTS FTO was downregulated in MIRI rats and H/R induced cardiomyocytes. Overexpression of FTO inhibited ROS level and increased the expression of SOD2, TFAM and COXI in vitro and in vivo. In addition, PGC-1a was identified as a downstream target of FTO. FTO enhanced the stability of PGC-1a mRNA through removing the m6A modification. CONCLUSION Our study revealed the role of FTO regulates the oxidative stress and mitochondrial biogenesis via PGC-1a in MIRI, which may provide a new approach to mitigating MIRI.
Collapse
Affiliation(s)
- Qiong Jiang
- Department of Cardiology, Fujian Medical University Union Hospital, Fuzhou, Fujian, People’s Republic of China
- Fujian Institute of Coronary Heart Disease, Fuzhou, Fujian, People’s Republic of China
- Fujian Heart Medical Center, Fuzhou, Fujian, People’s Republic of China
| | - Xuehai Chen
- Department of Cardiology, Fujian Medical University Union Hospital, Fuzhou, Fujian, People’s Republic of China
- Fujian Institute of Coronary Heart Disease, Fuzhou, Fujian, People’s Republic of China
- Fujian Heart Medical Center, Fuzhou, Fujian, People’s Republic of China
| | - Kezeng Gong
- Department of Cardiology, Fujian Medical University Union Hospital, Fuzhou, Fujian, People’s Republic of China
- Fujian Institute of Coronary Heart Disease, Fuzhou, Fujian, People’s Republic of China
- Fujian Heart Medical Center, Fuzhou, Fujian, People’s Republic of China
| | - Zhe Xu
- Department of Cardiology, Fujian Medical University Union Hospital, Fuzhou, Fujian, People’s Republic of China
- Fujian Institute of Coronary Heart Disease, Fuzhou, Fujian, People’s Republic of China
- Fujian Heart Medical Center, Fuzhou, Fujian, People’s Republic of China
| | - Lianglong Chen
- Department of Cardiology, Fujian Medical University Union Hospital, Fuzhou, Fujian, People’s Republic of China
- Fujian Institute of Coronary Heart Disease, Fuzhou, Fujian, People’s Republic of China
- Fujian Heart Medical Center, Fuzhou, Fujian, People’s Republic of China
| | - Feilong Zhang
- Department of Cardiology, Fujian Medical University Union Hospital, Fuzhou, Fujian, People’s Republic of China
- Fujian Institute of Coronary Heart Disease, Fuzhou, Fujian, People’s Republic of China
- Fujian Heart Medical Center, Fuzhou, Fujian, People’s Republic of China
| |
Collapse
|
2
|
Duan H, Yang X, Cai S, Zhang L, Qiu Z, Wang J, Wang S, Li Z, Li X. Nrf2 mitigates sepsis-associated encephalopathy-induced hippocampus ferroptosis via modulating mitochondrial dynamic homeostasis. Int Immunopharmacol 2024; 143:113331. [PMID: 39396427 DOI: 10.1016/j.intimp.2024.113331] [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: 06/29/2024] [Revised: 09/24/2024] [Accepted: 10/04/2024] [Indexed: 10/15/2024]
Abstract
Sepsis-associated encephalopathy (SAE) is a serious neurological complication accompanied with acute and long-term cognitive dysfunction. Ferroptosis is a newly discovered type of cell death that is produced by iron-dependent lipid peroxidation. Emerging evidence suggests that ferroptosis is involved in SAE. The nuclear factor erythroid 2-related factor 2 (Nrf2) is a mitochondria related gene involved in ferroptosis. However, the role of Nrf2 in SAE and the mechanisms remains elusive. In this study, we found that Nrf2 knockout aggravated cognitive and emotional dysfunction and promoted caecal ligation and puncture (CLP)-induced brain injury and hippocampus ferroptosis as indicated by the increase of ROS, Fe2+ and the levels of proinflammatory cytokines. Meanwhile, the levels of glutathione peroxidase 4 (GPX4), SLC7A11 and glutathionewere downregulatedin Nrf2 knockout group. In vitro experiments showed that mitochondrial ROS, Fe2+ and the expression of Fis1 and Drp1 decreased, and the level of Mfn1 and Opa-1 increased after Nrf2 overexpression. The silence of Nrf2 increased the expression of ROS, MDA and Fe2+, while decreased glutathione, mitochondrial membrane potential (MMP) and cell viability in vitro, indicating Nrf2 improved LPS-induced mitochondrial dysfunction and mitigated hippocampal cells ferroptosis. These results suggest that Nrf2 could inhibit ferroptosis and neuroinflammation in hippocampus and reduce cognitive dysfunction in SAE mice, making it a potential therapeutic target in the treatment of SAE. The protective effects of Nrf2 on the brain may be mediated by maintaining mitochondrial dynamic homeostasis.
Collapse
Affiliation(s)
- Haifeng Duan
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan, China; Hubei Provincial Engineering Research Center of Minimally Invasive Cardiovascular Surgery, Wuhan, China; Wuhan Clinical Research Center for Minimally Invasive Treatment of Structural Heart Disease, Wuhan, China
| | - Xin Yang
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan, China; Hubei Provincial Engineering Research Center of Minimally Invasive Cardiovascular Surgery, Wuhan, China; Wuhan Clinical Research Center for Minimally Invasive Treatment of Structural Heart Disease, Wuhan, China
| | - Shuhan Cai
- Department of Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China; Clinical Research Center of Hubei Critical Care Medicine, Wuhan, China
| | - Lei Zhang
- Department of Anesthesiology, the First Clinical College of Hubei University of Medicine, Shiyan, Hubei, China
| | - Zebao Qiu
- Department of Anesthesiology, Suizhou Zengdu Hospital, Suizhou, Hubei, China
| | - Jin Wang
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Shun Wang
- Department of Ophthalmology, Zhongnan Hospital of Wuhan University, Wuhan, China.
| | - Zhi Li
- Department of Ophthalmology, Zhongnan Hospital of Wuhan University, Wuhan, China.
| | - Xinyi Li
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan, China; Hubei Provincial Engineering Research Center of Minimally Invasive Cardiovascular Surgery, Wuhan, China; Wuhan Clinical Research Center for Minimally Invasive Treatment of Structural Heart Disease, Wuhan, China.
| |
Collapse
|
3
|
Navarro E, Esteras N. Multitarget Effects of Nrf2 Signalling in the Brain: Common and Specific Functions in Different Cell Types. Antioxidants (Basel) 2024; 13:1502. [PMID: 39765831 PMCID: PMC11673142 DOI: 10.3390/antiox13121502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 12/05/2024] [Accepted: 12/06/2024] [Indexed: 01/11/2025] Open
Abstract
Nuclear factor erythroid 2-related factor 2 (Nrf2) is a crucial regulator of cellular defence mechanisms, essential for maintaining the brain's health. Nrf2 supports mitochondrial function and protects against oxidative damage, which is vital for meeting the brain's substantial energy and antioxidant demands. Furthermore, Nrf2 modulates glial inflammatory responses, playing a pivotal role in preventing neuroinflammation. This review explores these multifaceted functions of Nrf2 within the central nervous system, focusing on its activity across various brain cell types, including neurons, astrocytes, microglia, and oligodendrocytes. Due to the brain's vulnerability to oxidative stress and metabolic challenges, Nrf2 is emerging as a key therapeutic target to enhance resilience against oxidative stress, inflammation, mitochondrial dysfunction, and demyelination, which are central to many neurodegenerative diseases.
Collapse
Affiliation(s)
- Elisa Navarro
- Neurochemistry Research Institute, Department of Biochemistry and Molecular Biology, School of Medicine, Complutense University of Madrid, 28040 Madrid, Spain
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, 28040 Madrid, Spain
- Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), 28034 Madrid, Spain
| | - Noemí Esteras
- Neurochemistry Research Institute, Department of Biochemistry and Molecular Biology, School of Medicine, Complutense University of Madrid, 28040 Madrid, Spain
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, 28040 Madrid, Spain
- Group of Neurodegenerative Diseases, Hospital Universitario 12 de Octubre Research Institute (imas12), 28041 Madrid, Spain
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London WC1N 3BG, UK
| |
Collapse
|
4
|
Ma C, Li H, Lu S, Li X. Thyroid-associated ophthalmopathy and ferroptosis: a review of pathological mechanisms and therapeutic strategies. Front Immunol 2024; 15:1475923. [PMID: 39712031 PMCID: PMC11659143 DOI: 10.3389/fimmu.2024.1475923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Accepted: 11/21/2024] [Indexed: 12/24/2024] Open
Abstract
Thyroid-associated ophthalmopathy (TAO) is an inflammatory orbital disease associated with autoimmune thyroid disorders. Owing to the ambiguous nature of the pathogenesis, contemporary pharmacological treatment strategies predominantly involve the use of glucocorticoids and immunosuppressants. However, the adverse effects associated with these agents in clinical practice necessitate further investigation into the disease's pathogenesis and the identification of novel therapeutic targets and pharmacological interventions. Recent studies suggest that ferroptosis, a novel form of regulated cell death, may play a role in TAO pathogenesis. This review aims to explore the involvement of ferroptosis in TAO and evaluate its potential as a therapeutic target. Key topics include the epidemiology, clinical manifestations, and pathophysiology of TAO, along with the molecular mechanisms of ferroptosis. Evidence supporting ferroptosis in TAO and the therapeutic implications of targeting this pathway are also discussed, alongside challenges and future directions in this emerging research area.
Collapse
Affiliation(s)
- Chao Ma
- Department of Ophthalmology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Haoyu Li
- Department of Ophthalmology, the Second Xiangya Hospital of Central South University, Changsha, Hunan, China
- Hunan Clinical Research Centre of Ophthalmic Disease, Changsha, Hunan, China
| | - Shuwen Lu
- Department of Ophthalmology, the First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Xian Li
- Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom
| |
Collapse
|
5
|
Wang J, Chen Y, Yuan H, Zhang X, Febbraio M, Pan Y, Huang S, Liu Z. Mitochondrial biogenesis disorder and oxidative damage promote refractory apical periodontitis in rat and human. Int Endod J 2024; 57:1326-1342. [PMID: 38881187 DOI: 10.1111/iej.14106] [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: 12/07/2023] [Revised: 05/20/2024] [Accepted: 05/24/2024] [Indexed: 06/18/2024]
Abstract
AIM To elucidate whether mitochondrial biogenesis disorder and damage from oxidative stress promote refractory apical periodontitis (RAP) in rat and human. METHODOLOGY Twenty Enterococcus faecalis-induced RAPs were established in the maxillary first molars of male Wistar rats. Concurrently, 12 periapical lesion specimens from patients presenting with RAP were obtained by apicoectomy. Radiographic examination and histologic analysis were conducted to evaluate periapical bone tissue destruction and morphological changes. The expression of key regulators of mitochondrial biogenesis, PGC-1α and Nrf2, were detected by immunohistochemistry and double immunofluorescence staining, Western blot and real-time PCR were also assayed. Mitochondrial ROS (mtROS) was identified by MitoSOX staining. Mitochondrial function was detected by the quantification of ATP production, mitochondrial DNA (mtDNA) copy number and activities of mitochondrial respiratory chain complexes. Furthermore, mitochondrial oxidative stress was evaluated by the determination of 3-nitrotyrosine (3-NT), 4-hydroxy-2-nonenal (4-HNE) and 8-hydroxy-deoxyguanosine (8-OHdG) expression levels, as well as malondialdehyde (MDA) expression and antioxidant capacity. Student's t-test was performed to determine significance between the groups; p < .05 was considered significant. RESULTS In the maxilla, significantly more bone resorption, greater number of periapical apoptotic cells and Tartrate-resistant acid phosphatase (TRAP)-positive multinucleated cells were observed in the RAP group compared with the control group (p < .01). PGC-1α and Nrf2 were significantly reduced in rat and human RAP lesions compared to the control group (p < .01) at both the mRNA and protein levels. Double immunofluorescence analysis of PGC-1α or Nrf2 with TOMM20 also indicated that mitochondrial biogenesis was impaired in RAP group (p < .01). Additionally, mitochondrial dysfunction was observed in RAP group, as reflected by increased mtROS, decreased ATP production, reduced mtDNA copy number and complexes of the mitochondrial respiratory chain. Finally, the expression levels of mitochondrial oxidative stress markers, 3-NT, 4-HNE and 8-OHdG, were significantly increased in the RAP group (p < .01). Consistent with this, systemic oxidative damage was also present in the progression of RAP, including increased MDA expression and decreased antioxidant activity (p < .01). CONCLUSIONS Mitochondrial biogenesis disorder and damage from oxidative stress contribute to the development of RAP.
Collapse
Affiliation(s)
- Jun Wang
- School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, China
- Department of Prosthodontics, School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, China
| | - Yuge Chen
- School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, China
- Department of Dentistry, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Huina Yuan
- School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, China
- Department of Orthodontics, School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, China
| | - Xuejia Zhang
- School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, China
- Department of Dentistry, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Maria Febbraio
- Department of Dentistry, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Yihuai Pan
- School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, China
- Department of Endodontics, School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, China
| | - Shengbin Huang
- School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, China
- Department of Prosthodontics, School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, China
| | - Zhongfang Liu
- School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, China
- Department of Endodontics, School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
6
|
Zhong YJ, Liu LL, Zhao Y, Feng Z, Liu Y. Elucidating the molecular mechanisms behind the therapeutic impact of median nerve stimulation on cognitive dysfunction post-traumatic brain injury. Exp Gerontol 2024; 194:112500. [PMID: 38901771 DOI: 10.1016/j.exger.2024.112500] [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: 11/05/2023] [Revised: 05/22/2024] [Accepted: 06/17/2024] [Indexed: 06/22/2024]
Abstract
OBJECTIVE Ferroptosis represents a form of regulated cellular death dependent upon iron and lipid peroxidation derivatives, holding considerable implications for cerebral and neurologic pathologies. In the present study, we endeavored to elucidate the molecular mechanisms governing ferroptosis and appraise the therapeutic value of electrical stimulation of median nerve in addressing cognitive impairments following traumatic brain injury (TBI), employing a rodent model. METHODS In this study, we established a rat model to investigate the cognitive impairments resulting from TBI, followed by the application of median nerve stimulation (MNS). Initially, rats received an intraperitoneal injection of Erastin (2 mg/kg) prior to undergoing MNS. After 24 h of MNS treatment, the rats were subjected to an open field test to evaluate their cognitive and motor functions. Subsequently, we conducted biochemical assays to measure the serum levels of GSH, MDA and SOD. The structural integrity and cellular morphology of hippocampal tissue were examined through H&E staining, Nissl staining and transmission electron microscopy. Additionally, we assessed the expression levels of proteins crucial for neuronal health and function in the hippocampus, including VEGF, SLC7A11, GPX4, Nrf2, α-syn, NEUN and PSD95. RESULTS Compared to the control group, rats in the stimulation group demonstrated enhanced mobility, reduced levels of tissue damage, a decrease in MDA concentration, and increased levels of GSH and SOD. Additionally, there was a significant upregulation in the expression of proteins critical for cellular defense and neuronal health, including GPX4, SLC7A11, Nrf2, VEGF, α-syn, NEUN, and PSD95 proteins. Conversely, rats in the Erastin group demonstrated decreased mobility, exacerbated pathological tissue damage, elevated MDA concentration, and decreased levels of GSH and SOD. There was also a notable decrease in the expression of GPX4, SLC7CA11, Nrf2, and VEGF proteins. The expression levels of α-syn, NEUN, and PSD95 were similarly diminished in the Erastin group. Each of these findings was statistically significant, indicating that MNS exerts neuroprotective effect in the hippocampal tissue of rats with TBI by inhibiting the ferroptosis pathway. CONCLUSION (1) MNS may enhance the cognitive and behavioral performance of rats after TBI; (2) MNS can play a neuroprotective role by promoting the expression of nerve injury-related proteins, alleviating oxidative stress and ferroptosis process; (3) MNS may inhibit ferroptosis of neuronal cells by activating Nrf2/ GPX4 signaling pathway, thereby improving cognitive impairment in TBI rats.
Collapse
Affiliation(s)
- Ying-Jun Zhong
- Department of Rehabilitation Medicine, The 1(st) Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330000, China
| | - Ling-Ling Liu
- Department of Rehabilitation Medicine, The 1(st) Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330000, China
| | - Yue Zhao
- Department of Rehabilitation Medicine, The 1(st) Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330000, China
| | - Zhen Feng
- Department of Rehabilitation Medicine, The 1(st) Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330000, China.
| | - Yuan Liu
- Department of Orthopedics, The 1(st) Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330000, China.
| |
Collapse
|
7
|
Chen YL, Wu JM, Chen KY, Wu MH, Yang PJ, Lee PC, Chen PD, Kuo TC, Yeh SL, Lin MT. Intravenous calcitriol administration improves the liver redox status and attenuates ferroptosis in mice with high-fat diet-induced obesity complicated with sepsis. Biomed Pharmacother 2024; 177:116926. [PMID: 38906016 DOI: 10.1016/j.biopha.2024.116926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 06/05/2024] [Accepted: 06/09/2024] [Indexed: 06/23/2024] Open
Abstract
Obesity aggravates ferroptosis, and vitamin D (VD) may inhibit ferroptosis. We hypothesized that weight reduction and/or calcitriol administration have benefits against the sepsis-induced liver redox imbalance and ferroptosis in obese mice. Mice were fed a high-fat diet for 11 weeks, then half of the mice continued to consume the diet, while the other half were transferred to a low-energy diet for 5 weeks. After feeding the respective diets for 16 weeks, sepsis was induced by cecal ligation and puncture (CLP). Septic mice were divided into four experimental groups: OS group, obese mice injected with saline; OD group, obese mice with calcitriol; WS group, weight-reduction mice with saline; and WD group, weight-reduction mice with calcitriol. Mice in the respective groups were euthanized at 12 or 24 h after CLP. Results showed that the OS group had the highest inflammatory mediators and lipid peroxide levels in the liver. Calcitriol treatment reduced iron content, enhanced the reduced glutathione/oxidized glutathione ratio, upregulated nuclear factor erythroid 2-related factor 2, ferroptosis-suppressing protein 1, and solute carrier family 7 member 11 expression levels. Also, mitochondrion-associated nicotinamide adenine dinucleotide phosphate oxidase 1, peroxisome proliferator-activated receptor-γ coactivator 1, hypoxia-inducible factor-1α, and heme oxidase-1 expression levels increased in the late phase of sepsis. These results were not noted in the WS group. These findings suggest that calcitriol treatment elicits a more-balanced glutathione redox status, alleviates liver ferroptosis, and enhances mitochondrial biogenesis-associated gene expressions. Weight reduction alone had minimal influences on liver ferroptosis and mitochondrial biogenesis in obese mice with sepsis.
Collapse
Affiliation(s)
- Ya-Ling Chen
- School of Nutrition and Health Sciences, College of Nutrition, Taipei Medical University, Taipei 11031, Taiwan; Nutrition Research Center, Taipei Medical University Hospital, Taipei 11031, Taiwan
| | - Jin-Ming Wu
- Department of Surgery, National Taiwan University Hospital and College of Medicine, National Taiwan University, Taipei 100, Taiwan
| | - Kuen-Yuan Chen
- Department of Surgery, National Taiwan University Hospital and College of Medicine, National Taiwan University, Taipei 100, Taiwan
| | - Ming-Hsun Wu
- Department of Surgery, National Taiwan University Hospital and College of Medicine, National Taiwan University, Taipei 100, Taiwan
| | - Po-Jen Yang
- Department of Surgery, National Taiwan University Hospital and College of Medicine, National Taiwan University, Taipei 100, Taiwan
| | - Po-Chu Lee
- Department of Surgery, National Taiwan University Hospital and College of Medicine, National Taiwan University, Taipei 100, Taiwan
| | - Po-Da Chen
- Department of Surgery, National Taiwan University Hospital and College of Medicine, National Taiwan University, Taipei 100, Taiwan
| | - Ting-Chun Kuo
- Department of Surgery, National Taiwan University Hospital and College of Medicine, National Taiwan University, Taipei 100, Taiwan
| | - Sung-Ling Yeh
- Department of Surgery, National Taiwan University Hospital and College of Medicine, National Taiwan University, Taipei 100, Taiwan
| | - Ming-Tsan Lin
- Department of Surgery, National Taiwan University Hospital and College of Medicine, National Taiwan University, Taipei 100, Taiwan.
| |
Collapse
|
8
|
Rabie MA, Ghoneim AT, Fahmy MI, El-Yamany MF, Sayed RH. Activation of alpha-7 nicotinic acetylcholine receptor by tropisetron mitigates 3-nitropropionic acid-induced Huntington's disease in rats: Role of PI3K/Akt and JAK2/NF-κB signaling pathways. Chem Biol Interact 2024; 393:110957. [PMID: 38513929 DOI: 10.1016/j.cbi.2024.110957] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 03/09/2024] [Accepted: 03/13/2024] [Indexed: 03/23/2024]
Abstract
Huntington's disease (HD) is an inheritable autosomal-dominant disorder that targets mainly the striatum. 3-Nitropropionic acid (3-NP) induces obvious deleterious behavioral, neurochemical, and histological effects similar to the symptoms of HD. Our study aimed to examine the neuroprotective activity of tropisetron, an alpha-7 neuronal nicotinic acetylcholine receptor (α-7nAChR) agonist, against neurotoxic events associated with 3-NP-induced HD in rats. Forty-eight rats were randomly allocated into four groups. Group I received normal saline, while Groups II, III and IV received 3-NP for 2 weeks. In addition, Group III and IV were treated with tropisetron 1 h after 3-NP administration. Meanwhile, Group IV received methyllycaconitine (MLA), an α-7nAChR antagonist, 30 min before tropisetron administration. Treatment with tropisetron improved motor deficits as confirmed by the behavioral tests and restored normal histopathological features of the striatum. Moreover, tropisetron showed an anti-oxidant activity via increasing the activities of SDH and HO-1 as well as Nrf2 expression along with reducing MDA level. Tropisetron also markedly upregulated the protein expression of p-PI3K and p-Akt which in turn hampered JAK2/NF-κB inflammatory cascade. In addition, tropisetron showed an anti-apoptotic activity through boosting the expression of Bcl-2 and reducing Bax expression and caspase-3 level. Interestingly, all the aforementioned effects of tropisetron were blocked by pre-administration of MLA, which confirms that such neuroprotective effects are mediated via activating of α-7nAChR. In conclusion, tropisetron showed a neuroprotective activity against 3-NP-induced HD via activating PI3K/Akt signaling and suppressing JAK2/NF-κB inflammatory axis. Thus, repositioning of tropisetron could represent a promising therapeutic strategy in management of HD.
Collapse
Affiliation(s)
- Mostafa A Rabie
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, 11562, Cairo, Egypt.
| | - Ahmed T Ghoneim
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Heliopolis University, Cairo, Egypt
| | - Mohamed I Fahmy
- Department of Pharmacology and Toxicology, College of Pharmaceutical sciences and drug manufacturing, Misr University for Science and Technology (MUST), 12585, Giza, Egypt
| | - Mohammed F El-Yamany
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, 11562, Cairo, Egypt
| | - Rabab H Sayed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, 11562, Cairo, Egypt; School of Pharmacy, Newgiza University, Giza, Egypt
| |
Collapse
|
9
|
Ciubuc-Batcu MT, Stapelberg NJC, Headrick JP, Renshaw GMC. A mitochondrial nexus in major depressive disorder: Integration with the psycho-immune-neuroendocrine network. Biochim Biophys Acta Mol Basis Dis 2024; 1870:166920. [PMID: 37913835 DOI: 10.1016/j.bbadis.2023.166920] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 10/06/2023] [Accepted: 10/09/2023] [Indexed: 11/03/2023]
Abstract
Nervous system processes, including cognition and affective state, fundamentally rely on mitochondria. Impaired mitochondrial function is evident in major depressive disorder (MDD), reflecting cumulative detrimental influences of both extrinsic and intrinsic stressors, genetic predisposition, and mutation. Glucocorticoid 'stress' pathways converge on mitochondria; oxidative and nitrosative stresses in MDD are largely mitochondrial in origin; both initiate cascades promoting mitochondrial DNA (mtDNA) damage with disruptions to mitochondrial biogenesis and tryptophan catabolism. Mitochondrial dysfunction facilitates proinflammatory dysbiosis while directly triggering immuno-inflammatory activation via released mtDNA, mitochondrial lipids and mitochondria associated membranes (MAMs), further disrupting mitochondrial function and mitochondrial quality control, promoting the accumulation of abnormal mitochondria (confirmed in autopsy studies). Established and putative mechanisms highlight a mitochondrial nexus within the psycho-immune neuroendocrine (PINE) network implicated in MDD. Whether lowering neuronal resilience and thresholds for disease, or linking mechanistic nodes within the MDD pathogenic network, impaired mitochondrial function emerges as an important risk, a functional biomarker, providing a therapeutic target in MDD. Several treatment modalities have been demonstrated to reset mitochondrial function, which could benefit those with MDD.
Collapse
Affiliation(s)
- M T Ciubuc-Batcu
- Griffith University School of Medicine and Dentistry, Australia; Gold Coast Health, Queensland, Australia
| | - N J C Stapelberg
- Bond University Faculty of Health Sciences and Medicine, Australia; Gold Coast Health, Queensland, Australia
| | - J P Headrick
- Griffith University School of Pharmacy and Medical Science, Australia
| | - G M C Renshaw
- Hypoxia and Ischemia Research Unit, Griffith University, School of Health Sciences and Social Work, Australia.
| |
Collapse
|
10
|
Moubarak MM, Pagano Zottola AC, Larrieu CM, Cuvellier S, Daubon T, Martin OCB. Exploring the multifaceted role of NRF2 in brain physiology and cancer: A comprehensive review. Neurooncol Adv 2024; 6:vdad160. [PMID: 38221979 PMCID: PMC10785770 DOI: 10.1093/noajnl/vdad160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2024] Open
Abstract
Chronic oxidative stress plays a critical role in the development of brain malignancies due to the high rate of brain oxygen utilization and concomitant production of reactive oxygen species. The nuclear factor-erythroid-2-related factor 2 (NRF2), a master regulator of antioxidant signaling, is a key factor in regulating brain physiology and the development of age-related neurodegenerative diseases. Also, NRF2 is known to exert a protective antioxidant effect against the onset of oxidative stress-induced diseases, including cancer, along with its pro-oncogenic activities through regulating various signaling pathways and downstream target genes. In glioblastoma (GB), grade 4 glioma, tumor resistance, and recurrence are caused by the glioblastoma stem cell population constituting a small bulk of the tumor core. The persistence and self-renewal capacity of these cell populations is enhanced by NRF2 expression in GB tissues. This review outlines NRF2's dual involvement in cancer and highlights its regulatory role in human brain physiology and diseases, in addition to the development of primary brain tumors and therapeutic potential, with a focus on GB.
Collapse
Affiliation(s)
- Maya M Moubarak
- University of Bordeaux, CNRS, IBGC, UMR 5095, Bordeaux, France
| | | | | | | | - Thomas Daubon
- University of Bordeaux, CNRS, IBGC, UMR 5095, Bordeaux, France
| | | |
Collapse
|
11
|
Li C, Han S, Zhu J, Cheng F. MiR-132-3p activation aggravates renal ischemia-reperfusion injury by targeting Sirt1/PGC1alpha axis. Cell Signal 2023; 110:110801. [PMID: 37433399 DOI: 10.1016/j.cellsig.2023.110801] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 06/13/2023] [Accepted: 07/07/2023] [Indexed: 07/13/2023]
Abstract
The pathogenesis of renal ischemic diseases remains unclear. In this study, we demonstrate the induction of microRNA-132-3p (miR-132-3p) in ischemic acute kidney injury (AKI) and cultured renal tubular cells under oxidative stress. miR-132-3p mimic increased apoptosis in renal tubular cells and enhanced ischemic AKI in mice, whereas miR-132-3p inhibition offered protective effects. We analyzed miR-132-3p target genes through bioinformatic analysis and Sirt1 was predicted as the target gene of miR-132-3p. Luciferase microRNA target reporter assay further verified Sirt1 as a direct target of miR-132-3p. In cultured tubular cells and mouse kidneys, IRI and H2O2 treatment repressed Sirt1 and PGC-1α/NRF2/HO-1 expression, whereas anti-miR-132-3p preserved Sirt1 and PGC-1α/NRF2/HO-1 expression. In renal tubular, Sirt1 inhibitor suppressed PGC1-1α/NRF2/HO-1 expression and aggravated tubular apoptosis. Together, the results suggest that miR-132-3p induction aggravates ischemic AKI and oxidative stress by repressing Sirt1 expression, and miR-132-3p inhibition offers renal protection and may be a potential therapeutic target.
Collapse
Affiliation(s)
- Chenglong Li
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China
| | - Shangting Han
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China; Department of Organ Transplantation, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China
| | - Jiefu Zhu
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China; Department of Organ Transplantation, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China.
| | - Fan Cheng
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China.
| |
Collapse
|
12
|
Hafez HA, Mahmoud SA, Alhmoud JF, Khafaga RH, Kamel MA, Shaker SA. The Intergeneration Long-Lasting Consequences of Pre-Conceptional Exposure to Sofosbuvir on the Ovarian Tissues of F1 Offspring: Experimental Study on Rats. Int J Mol Sci 2023; 24:13675. [PMID: 37761983 PMCID: PMC10531293 DOI: 10.3390/ijms241813675] [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: 07/31/2023] [Revised: 08/30/2023] [Accepted: 08/30/2023] [Indexed: 09/29/2023] Open
Abstract
Sofosbuvir (SOF), a nucleos(t)ide polymerase inhibitor, has been used during the past decade for mass treatment of viral hepatitis C in endemic countries like Egypt, increasing the exposure of women at childbearing age to SOF. This study investigated the long-lasting consequences of the pre-conceptional exposure of young female rats to SOF on the ovarian tissues of F1 offspring and explored the possible molecular mechanisms of these intergenerational effects at various levels. The study was conducted on young female rats that were divided into control group and SOF-exposed group at a dose of 4 mg/kg/day for three months. After that, pregnancy was induced in both groups by mating with healthy male rats. After delivery, the female neonates were followed for 4 months, and the ovarian tissues were collected to assess the studied parameters. Pre-conceptional exposure to SOF affected the ovarian functions of F1 offspring through modulation of estrogen receptors, ovarian Kiss1 and its receptor, increased lipid peroxidation marker, DNA oxidation marker, and redox-sensitive nuclear factor kappa B, and decreased nuclear erythroid-2-related factor 2, mitochondrial function, and biogenesis. SOF affected the ovarian function of the F1 offspring by inducing oxidative stress and inflammation, leading to the modulation of mitochondrial functions and biogenesis.
Collapse
Affiliation(s)
- Hala A. Hafez
- Department of Biochemistry, Medical Research Institute, Alexandria University, Alexandria 21561, Egypt; (S.A.M.); (M.A.K.); (S.A.S.)
| | - Shimaa A. Mahmoud
- Department of Biochemistry, Medical Research Institute, Alexandria University, Alexandria 21561, Egypt; (S.A.M.); (M.A.K.); (S.A.S.)
| | - Jehad F. Alhmoud
- Department of Medical Laboratory Sciences, Jordan University of Science and Technology, Irbid 22110, Jordan;
| | - Rana H.M. Khafaga
- Department of Biochemistry, Medical Research Institute, Alexandria University, Alexandria 21561, Egypt; (S.A.M.); (M.A.K.); (S.A.S.)
| | - Maher A. Kamel
- Department of Biochemistry, Medical Research Institute, Alexandria University, Alexandria 21561, Egypt; (S.A.M.); (M.A.K.); (S.A.S.)
| | - Sara A. Shaker
- Department of Biochemistry, Medical Research Institute, Alexandria University, Alexandria 21561, Egypt; (S.A.M.); (M.A.K.); (S.A.S.)
| |
Collapse
|
13
|
Liu J, Lu W, Yan D, Guo J, Zhou L, Shi B, Su X. Mitochondrial respiratory complex I deficiency inhibits brown adipogenesis by limiting heme regulation of histone demethylation. Mitochondrion 2023; 72:22-32. [PMID: 37451354 DOI: 10.1016/j.mito.2023.07.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 06/13/2023] [Accepted: 07/11/2023] [Indexed: 07/18/2023]
Abstract
Mitochondrial functions play a crucial role in determining the metabolic and thermogenic status of brown adipocytes. Increasing evidence reveals that the mitochondrial oxidative phosphorylation (OXPHOS) system plays an important role in brown adipogenesis, but the mechanistic insights are limited. Herein, we explored the potential metabolic mechanisms leading to OXPHOS regulation of brown adipogenesis in pharmacological and genetic models of mitochondrial respiratory complex I deficiency. OXPHOS deficiency inhibits brown adipogenesis through disruption of the brown adipogenic transcription circuit without affecting ATP levels. Neither blockage of calcium signaling nor antioxidant treatment can rescue the suppressed brown adipogenesis. Metabolomics analysis revealed a decrease in levels of tricarboxylic acid cycle intermediates and heme. Heme supplementation specifically enhances respiratory complex I activity without affecting complex II and partially reverses the inhibited brown adipogenesis by OXPHOS deficiency. Moreover, the regulation of brown adipogenesis by the OXPHOS-heme axis may be due to the suppressed histone methylation status by increasing histone demethylation. In summary, our findings identified a heme-sensing retrograde signaling pathway that connects mitochondrial OXPHOS to the regulation of brown adipocyte differentiation and metabolic functions.
Collapse
Affiliation(s)
- Jingjing Liu
- Department of Biochemistry and Molecular Biology, Suzhou Medical College of Soochow University, Suzhou 215123, China
| | - Wen Lu
- Department of Endocrinology, The First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Dongyue Yan
- Department of Biochemistry and Molecular Biology, Suzhou Medical College of Soochow University, Suzhou 215123, China
| | - Junyuan Guo
- Department of Biochemistry and Molecular Biology, Suzhou Medical College of Soochow University, Suzhou 215123, China
| | - Li Zhou
- Department of Nutrition, The First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Bimin Shi
- Department of Endocrinology, The First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Xiong Su
- Department of Biochemistry and Molecular Biology, Suzhou Medical College of Soochow University, Suzhou 215123, China.
| |
Collapse
|
14
|
You YL, Choi HS. Dibenzoylmethane ameliorates adiposity-mediated neuroinflammatory response and inflammation-mediated neuronal cell death in mouse microglia and neuronal cells. Food Sci Biotechnol 2023; 32:1123-1132. [PMID: 37215256 PMCID: PMC10195951 DOI: 10.1007/s10068-023-01245-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Revised: 12/21/2022] [Accepted: 01/02/2023] [Indexed: 01/18/2023] Open
Abstract
Dibenzoylmethane (DBM), a licorice-derived component, has numerous health benefits. The current study aimed to investigate the effect of DBM on adiposity-induced neuroinflammatory/oxidative response and microglial activation-induced neuronal cell damage. For this research, BV2 and HT22 cells were cultured using adipcyte- and microglia-conditioned media, respectively. DBM effectively suppressed lipopolysaccharide-induced productions in inducible nitric oxide synthase and cyclooxygenase2. Interleukin (IL)-6, monocyte chemoattractant protein-1, IL-1β, and tumor necrosis factor-α levels were also downregulated by DBM. In adipocyte-conditioned medium (ACM)-cultured BV2 cells, DBM effectively decreased ACM-induced generation of nitric oxide, reactive oxygen species, and inflammatory cytokines by activating nuclear factor erythroid 2-related factor 2/heme oxygenase-1 signaling and reducing nuclear factor kappa-light-chain-enhancer of activated B cells. In BV2-conditioned medium (BVM)-cultured neuron cells, DBM recovered the BVM-induced reduction of neuronal cell viability, thereby regulating B-cell lymphoma 2 (BCL2), BCL2-associated X (BAX), and cleaved caspase-3 protein expression. Taken together, DBM suppressed adiposity-induced inflammation/oxidative responses and inflammation-induced neuronal cell death.
Collapse
Affiliation(s)
- Ye-Lim You
- Department of Food Nutrition, Sangmyung University, Hongjimun 2-gil 20, Jongno-gu, Seoul, 03016 Republic of Korea
| | - Hyeon-Son Choi
- Department of Food Nutrition, Sangmyung University, Hongjimun 2-gil 20, Jongno-gu, Seoul, 03016 Republic of Korea
| |
Collapse
|
15
|
Wang YY, Lin SY, Chang CY, Wu CC, Chen WY, Huang WC, Liao SL, Wang WY, Chen CJ. α7 nicotinic acetylcholine receptor agonist improved brain injury and impaired glucose metabolism in a rat model of ischemic stroke. Metab Brain Dis 2023; 38:1249-1259. [PMID: 36662413 DOI: 10.1007/s11011-023-01167-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 01/13/2023] [Indexed: 01/21/2023]
Abstract
Vagus nerve stimulation through the action of acetylcholine can modulate inflammatory responses and metabolism. α7 Nicotinic Acetylcholine Receptor (α7nAChR) is a key component in the biological functions of acetylcholine. To further explore the health benefits of vagus nerve stimulation, this study aimed to investigate whether α7nAChR agonists offer beneficial effects against poststroke inflammatory and metabolic changes and to identify the underlying mechanisms in a rat model of stroke established by permanent cerebral ischemia. We found evidence showing that pretreatment with α7nAChR agonist, GTS-21, improved poststroke brain infarction size, impaired motor coordination, brain apoptotic caspase 3 activation, dysregulated glucose metabolism, and glutathione reduction. In ischemic cortical tissues and gastrocnemius muscles with GTS-21 pretreatment, macrophages/microglia M1 polarization-associated Tumor Necrosis Factor-α (TNF-α) mRNA, Cluster of Differentiation 68 (CD68) protein, and Inducible Nitric Oxide Synthase (iNOS) protein expression were reduced, while expression of anti-inflammatory cytokine IL-4 mRNA, and levels of M2 polarization-associated CD163 mRNA and protein were increased. In the gastrocnemius muscles, stroke rats showed a reduction in both glutathione content and Akt Serine 473 phosphorylation, as well as an elevation in Insulin Receptor Substrate-1 Serine 307 phosphorylation and Dynamin-Related Protein 1 Serine 616 phosphorylation. GTS-21 reversed poststroke changes in the gastrocnemius muscles. Overall, our findings, provide further evidence supporting the neuroprotective benefits of α7nAChR agonists, and indicate that they may potentially exert anti-inflammatory and metabolic effects peripherally in the skeletal muscle in an acute ischemic stroke animal model.
Collapse
Affiliation(s)
- Ya-Yu Wang
- Department of Family Medicine, Taichung Veterans General Hospital, 407, Taichung City, Taiwan
| | - Shih-Yi Lin
- Center for Geriatrics and Gerontology, Taichung Veterans General Hospital, 407, Taichung City, Taiwan
- Institute of Clinical Medicine, National Yang Ming Chiao Tung University, 112, Taipei City, Taiwan
| | - Cheng-Yi Chang
- Department of Surgery, Feng Yuan Hospital, 420, Taichung City, Taiwan
- Department of Veterinary Medicine, National Chung Hsing University, 402, Taichung City, Taiwan
| | - Chih-Cheng Wu
- Department of Anesthesiology, Taichung Veterans General Hospital, 407, Taichung City, Taiwan
| | - Wen-Ying Chen
- Department of Veterinary Medicine, National Chung Hsing University, 402, Taichung City, Taiwan
| | - Wei-Chi Huang
- Department of Veterinary Medicine, National Chung Hsing University, 402, Taichung City, Taiwan
| | - Su-Lan Liao
- Department of Medical Research, Taichung Veterans General Hospital, No. 1650, Sec. 4, Taiwan Boulevard, 407, Taichung City, Taiwan
| | - Wen-Yi Wang
- Department of Nursing, Hung Kuang University, 433, Taichung City, Taiwan
| | - Chun-Jung Chen
- Department of Medical Research, Taichung Veterans General Hospital, No. 1650, Sec. 4, Taiwan Boulevard, 407, Taichung City, Taiwan.
- Department of Medical Laboratory Science and Biotechnology, China Medical University, 404, Taichung City, Taiwan.
| |
Collapse
|
16
|
Babaei-Abraki S, Karamali F, Nasr-Esfahani MH. Ferroptosis: The functions of Nrf2 in human embryonic stem cells. Cell Signal 2023; 106:110654. [PMID: 36906163 DOI: 10.1016/j.cellsig.2023.110654] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 02/14/2023] [Accepted: 03/07/2023] [Indexed: 03/12/2023]
Abstract
Human embryonic stem cells (hESCs) have the capacity of self-renewal as well as differentiation towards three germ layer derivatives which makes them as a source of therapeutic application. hESCs are tremendously prone to cell death after dissociation into single cells. Therefore, it technically hinders their applications. Our recent study has revealed that hESCs can be prone to ferroptosis which differs from those in earlier explorations reporting that cellular detachment results in a process cited as anoikis. Ferroptosis occurs via increasing intracellular iron. Therefore, this form of programmed cell death is distinct from other cell deaths in terms of biochemistry, morphology, and genetics. Ferroptosis is found by excessive iron which plays an important part role in reactive oxygen species (ROS) generation through the Fenton reaction as a cofactor. Many genes are related to ferroptosis under the control of nuclear factor erythroid 2-related factor 2 (Nrf2) which is a transcription factor regulating the expression of genes to protect cells from oxidative stress. Nrf2 was demonstrated to take a perilous role in the suppression of ferroptosis by regulating the iron, antioxidant defense enzymes, usage, and restoration of glutathione, thioredoxin, and NADPH. Mitochondrial function is another target of Nrf2 to control cell homeostasis through the modulation of ROS production. In this review, we will give a succinct overview of lipid peroxidation and discuss the major players in the ferroptotic cascade. Additionally, we discussed the important role of the Nrf2 signaling pathway in mediating lipid peroxidation and ferroptosis, with a focus on known Nrf2 target genes that inhibit these processes and their possible role in hESCs.
Collapse
Affiliation(s)
- Shahnaz Babaei-Abraki
- Department of Animal Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran.
| | - Fereshteh Karamali
- Department of Animal Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran.
| | - Mohammad Hossein Nasr-Esfahani
- Department of Animal Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran.
| |
Collapse
|
17
|
Inhibition of Microglial GSK3β Activity Is Common to Different Kinds of Antidepressants: A Proposal for an In Vitro Screen to Detect Novel Antidepressant Principles. Biomedicines 2023; 11:biomedicines11030806. [PMID: 36979785 PMCID: PMC10045655 DOI: 10.3390/biomedicines11030806] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 02/17/2023] [Accepted: 03/04/2023] [Indexed: 03/09/2023] Open
Abstract
Depression is a major public health concern. Unfortunately, the present antidepressants often are insufficiently effective, whilst the discovery of more effective antidepressants has been extremely sluggish. The objective of this review was to combine the literature on depression with the pharmacology of antidepressant compounds, in order to formulate a conceivable pathophysiological process, allowing proposals how to accelerate the discovery process. Risk factors for depression initiate an infection-like inflammation in the brain that involves activation microglial Toll-like receptors and glycogen synthase kinase-3β (GSK3β). GSK3β activity alters the balance between two competing transcription factors, the pro-inflammatory/pro-oxidative transcription factor NFκB and the neuroprotective, anti-inflammatory and anti-oxidative transcription factor NRF2. The antidepressant activity of tricyclic antidepressants is assumed to involve activation of GS-coupled microglial receptors, raising intracellular cAMP levels and activation of protein kinase A (PKA). PKA and similar kinases inhibit the enzyme activity of GSK3β. Experimental antidepressant principles, including cannabinoid receptor-2 activation, opioid μ receptor agonists, 5HT2 agonists, valproate, ketamine and electrical stimulation of the Vagus nerve, all activate microglial pathways that result in GSK3β-inhibition. An in vitro screen for NRF2-activation in microglial cells with TLR-activated GSK3β activity, might therefore lead to the detection of totally novel antidepressant principles with, hopefully, an improved therapeutic efficacy.
Collapse
|
18
|
Guan X, Wu P, Cao B, Liu X, Chen X, Zhang W, Zhang Y, Guan Z, Wang Y. PGC-1α-siRNA suppresses inflammation in substantia nigra of PD mice by inhibiting microglia. Int J Neurosci 2023; 133:269-277. [PMID: 33784949 DOI: 10.1080/00207454.2021.1910257] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Background and purpose: Parkinson's disease is a common degenerative disease of the central nervous system with complex pathogenesis. More and more studies have found that inflammatory response promotes the occurrence and development of the disease, in which the activation of microglia plays an important role. PGC-1α (peroxisome proliferator activated receptor-γ coactivator-1α) is the main factor in mitochondrial biogenetic, and is closely related to the inflammatory response. Our immunofluorescence test results showed that PGC-1α and microglia (Iba1) have double-labeled phenomenon. The expression of microglia in the MPTP (1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine) group increased, and PGC-1α/Iba1 double label increased. To test whether lowering the expression of PGC-1α can reduce the activation of microglia and protect the substantia nigra dopaminergic neurons, we constructed PGC-1α interference lentivirus.Methods: Immunofluorescence, western blot, and ELISA were used to detect microglial phenotype.Results: The results showed that PGC-1α interfering with lentivirus can transfect microglial cells in substantia nigra, and the PGC-1α protein level decreased in substantia nigra accordingly; TH protein expression had no statistical difference compared with MPTP group; PGC-1α interfering lentivirus reduced microglia number and activation, and at the same time the expression of iNOS and Arg1 significantly reduced compared with MPTP group. The IL-6 expression in blood detected using ELISA was significantly reduced compared with MPTP group.Conclusion: PGC-1α downregulation inhibited microglia activity, and both M1 and M2 microglial activities are reduced.
Collapse
Affiliation(s)
- Xin Guan
- Department of Physiology, College of Life Sciences, Hebei Normal University, Shijiazhuang, Hebei, PR China
| | - Pengyue Wu
- Department of Physiology, College of Life Sciences, Hebei Normal University, Shijiazhuang, Hebei, PR China
| | - Bing Cao
- Department of Physiology, College of Life Sciences, Hebei Normal University, Shijiazhuang, Hebei, PR China
| | - Xiaoting Liu
- Department of Physiology, College of Life Sciences, Hebei Normal University, Shijiazhuang, Hebei, PR China
| | - Xi Chen
- Department of Physiology, College of Life Sciences, Hebei Normal University, Shijiazhuang, Hebei, PR China
| | - Wenpei Zhang
- Department of Physiology, College of Life Sciences, Hebei Normal University, Shijiazhuang, Hebei, PR China
| | - Yanqiu Zhang
- Department of Physiology, College of Life Sciences, Hebei Normal University, Shijiazhuang, Hebei, PR China
| | - Zhenlong Guan
- Department of Physiology, College of Life Sciences, Hebei Normal University, Shijiazhuang, Hebei, PR China
| | - Yanqin Wang
- Department of Physiology, College of Life Sciences, Hebei Normal University, Shijiazhuang, Hebei, PR China
| |
Collapse
|
19
|
Deng S, Zhang Y, Xin Y, Hu X. Vagus nerve stimulation attenuates acute kidney injury induced by hepatic ischemia/reperfusion injury in rats. Sci Rep 2022; 12:21662. [PMID: 36522408 PMCID: PMC9755310 DOI: 10.1038/s41598-022-26231-w] [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: 08/07/2022] [Accepted: 12/12/2022] [Indexed: 12/23/2022] Open
Abstract
Hepatic ischemia/reperfusion (I/R) injury, caused by limited blood supply and subsequent blood supply, is a causative factor resulting in morbidity and mortality during liver transplantation and liver resection. Hepatic I/R injury frequently contributes to remote organ injury, such as kidney, lung, and heart. It has been demonstrated that vagus nerve stimulation (VNS) is effective in remote organ injury after I/R injury. Here, our aim is to investigate the potential action of VNS on hepatic I/R injury-induced acute kidney injury (AKI) and explore its underlying mechanisms. To test this hypothesis, male Sprague-Dawley rats were randomly assigned into three experimental groups: Sham group (sham operation, n = 6); I/R group (hepatic I/R with sham VNS, n = 6); and VNS group (hepatic I/R with VNS, n = 6). VNS was performed during the entire hepatic I/R process. Our results showed that throughout the hepatic I/R process, VNS significantly regulated the expression levels of various iconic factors and greatly enhanced the protein expression levels of nuclear factor erythroid 2-related factor 2 (Nrf2) and hemeoxygenase-1 (HO-1) in the kidneys. These findings suggested that VNS may ameliorate hepatic I/R injury-induced AKI by suppressing inflammation, oxidative stress, and apoptosis probably through activating the Nrf2/HO-1 signaling pathway.
Collapse
Affiliation(s)
- Simin Deng
- grid.216417.70000 0001 0379 7164Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, 139 Middle Renmin Road, Changsha, 410011 Hunan China
| | - Yifeng Zhang
- grid.216417.70000 0001 0379 7164Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, 139 Middle Renmin Road, Changsha, 410011 Hunan China
| | - Ying Xin
- grid.216417.70000 0001 0379 7164Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, 139 Middle Renmin Road, Changsha, 410011 Hunan China
| | - Xinqun Hu
- grid.216417.70000 0001 0379 7164Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, 139 Middle Renmin Road, Changsha, 410011 Hunan China
| |
Collapse
|
20
|
Esteras N, Abramov AY. Nrf2 as a regulator of mitochondrial function: Energy metabolism and beyond. Free Radic Biol Med 2022; 189:136-153. [PMID: 35918014 DOI: 10.1016/j.freeradbiomed.2022.07.013] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 06/20/2022] [Accepted: 07/19/2022] [Indexed: 12/14/2022]
Abstract
Mitochondria are unique and essential organelles that mediate many vital cellular processes including energy metabolism and cell death. The transcription factor Nrf2 (NF-E2 p45-related factor 2) has emerged in the last few years as an important modulator of multiple aspects of mitochondrial function. Well-known for controlling cellular redox homeostasis, the cytoprotective effects of Nrf2 extend beyond its ability to regulate a diverse network of antioxidant and detoxification enzymes. Here, we review the role of Nrf2 in the regulation of mitochondrial function and structure. We focus on Nrf2 involvement in promoting mitochondrial quality control and regulation of basic aspects of mitochondrial function, including energy production, reactive oxygen species generation, calcium signalling, and cell death induction. Given the importance of mitochondria in the development of multiple diseases, these findings reinforce the pharmacological activation of Nrf2 as an attractive strategy to counteract mitochondrial dysfunction.
Collapse
Affiliation(s)
- Noemí Esteras
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, Queen Square, London, UK.
| | - Andrey Y Abramov
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, Queen Square, London, UK.
| |
Collapse
|
21
|
Deng S, Zhang Y, Xin Y, Hu X. Vagus Nerve Stimulation Attenuates Acute Kidney Injury Induced by Hepatic Ischemia/Reperfusion Injury by Suppressing Inflammation, Oxidative Stress, and Apoptosis in Rats.. [DOI: 10.21203/rs.3.rs-1937916/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Abstract
Hepatic ischemia reperfusion (I/R) injury, caused by limited blood supply and subsequent blood supply, is a causative factor resulting in morbidity and mortality during liver transplantation (LT) and liver resection. Hepatic I/R injury frequently contributes to remote organ injury, such as kidney, lung, and heart. It has been demonstrated that vagus nerve stimulation (VNS) is effective in remote organ injury after ischemia reperfusion injury. Here, our aim is to investigate the potential action of VNS on hepatic I/R injury-induced acute kidney injury (AKI) and explore its underlying mechanisms. To test this hypothesis, male Sprague-Dawley rats were randomly assigned into three experimental groups: Sham group (sham operation, n=6); I/R group (hepatic I/R with sham VNS, n=6); and VNS group (hepatic I/R with VNS, n=6). VNS was performed during the entire hepatic I/R process. Our results showed that throughout the hepatic I/R process, VNS significantly reduced inflammation, oxidative stress, and apoptosis, and greatly enhanced the protein expression levels of nuclear factor erythroid 2-related factor 2 (Nrf2) and hemeoxygenase-1 (HO-1) in the kidneys. These findings suggest that VNS may ameliorate hepatic I/R injury-induced AKI by suppressing inflammation, oxidative stress, and apoptosis probably through activating the Nrf2/HO-1 signaling pathway.
Collapse
Affiliation(s)
- Simin Deng
- Second Xiangya Hospital of Central South University
| | - Yifeng Zhang
- Second Xiangya Hospital of Central South University
| | - Ying Xin
- Second Xiangya Hospital of Central South University
| | - Xinqun Hu
- Second Xiangya Hospital of Central South University
| |
Collapse
|
22
|
Rupprecht A, Theisen U, Wendt F, Frank M, Hinz B. The Combination of Δ9-Tetrahydrocannabinol and Cannabidiol Suppresses Mitochondrial Respiration of Human Glioblastoma Cells via Downregulation of Specific Respiratory Chain Proteins. Cancers (Basel) 2022; 14:cancers14133129. [PMID: 35804909 PMCID: PMC9265124 DOI: 10.3390/cancers14133129] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 05/25/2022] [Accepted: 06/01/2022] [Indexed: 11/21/2022] Open
Abstract
Simple Summary Cannabidiol (CBD) is a phytocannabinoid from Cannabis sativa L. that exhibits no psychoactivity and, like the psychoactive cannabinoid Δ9-tetrahydrocannabinol (THC), shows anticancer effects in preclinical cell and animal models. Previous studies have indicated a stronger cancer-targeting effect when THC and CBD are combined. Here, we investigated how the combination of THC and CBD in a 1:1 ratio affects glioblastoma cell survival. The compounds were found to synergistically enhance cell death, which was attributed to mitochondrial damage and disruption of energy metabolism. A detailed look at the mitochondrial electron transfer chain showed that THC/CBD selectively decreased certain subunits of complexes I and IV. These data highlight the fundamental changes in cellular energy metabolism when cancer cells are exposed to a mixture of cannabinoids and underscore the potential of combining cannabinoids in cancer treatment. Abstract Phytocannabinoids represent a promising approach in glioblastoma therapy. Previous work has shown that a combined treatment of glioblastoma cells with submaximal effective concentrations of psychoactive Δ9-tetrahydrocannabinol (THC) and non-psychoactive cannabidiol (CBD) greatly increases cell death. In the present work, the glioblastoma cell lines U251MG and U138MG were used to investigate whether the combination of THC and CBD in a 1:1 ratio is associated with a disruption of cellular energy metabolism, and whether this is caused by affecting mitochondrial respiration. Here, the combined administration of THC and CBD (2.5 µM each) led to an inhibition of oxygen consumption rate and energy metabolism. These effects were accompanied by morphological changes to the mitochondria, a release of mitochondrial cytochrome c into the cytosol and a marked reduction in subunits of electron transport chain complexes I (NDUFA9, NDUFB8) and IV (COX2, COX4). Experiments with receptor antagonists and inhibitors showed that the degradation of NDUFA9 occurred independently of the activation of the cannabinoid receptors CB1, CB2 and TRPV1 and of usual degradation processes mediated via autophagy or the proteasomal system. In summary, the results describe a previously unknown mitochondria-targeting mechanism behind the toxic effect of THC and CBD on glioblastoma cells that should be considered in future cancer therapy, especially in combination strategies with other chemotherapeutics.
Collapse
Affiliation(s)
- Anne Rupprecht
- Institute of Pharmacology and Toxicology, Rostock University Medical Centre, 18057 Rostock, Germany; (A.R.); (U.T.); (F.W.)
| | - Ulrike Theisen
- Institute of Pharmacology and Toxicology, Rostock University Medical Centre, 18057 Rostock, Germany; (A.R.); (U.T.); (F.W.)
| | - Franziska Wendt
- Institute of Pharmacology and Toxicology, Rostock University Medical Centre, 18057 Rostock, Germany; (A.R.); (U.T.); (F.W.)
| | - Marcus Frank
- Electron Microscopy Centre, Rostock University Medical Centre, 18057 Rostock, Germany;
- Department Life, Light and Matter, University of Rostock, 18059 Rostock, Germany
| | - Burkhard Hinz
- Institute of Pharmacology and Toxicology, Rostock University Medical Centre, 18057 Rostock, Germany; (A.R.); (U.T.); (F.W.)
- Correspondence:
| |
Collapse
|
23
|
Choi YK, Kim YM. Beneficial and Detrimental Roles of Heme Oxygenase-1 in the Neurovascular System. Int J Mol Sci 2022; 23:ijms23137041. [PMID: 35806040 PMCID: PMC9266949 DOI: 10.3390/ijms23137041] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 06/15/2022] [Accepted: 06/21/2022] [Indexed: 02/04/2023] Open
Abstract
Heme oxygenase (HO) has both beneficial and detrimental effects via its metabolites, including carbon monoxide (CO), biliverdin or bilirubin, and ferrous iron. HO-1 is an inducible form of HO that is upregulated by oxidative stress, nitric oxide, CO, and hypoxia, whereas HO-2 is a constitutive form that regulates vascular tone and homeostasis. In brains injured by trauma, ischemia-reperfusion, or Alzheimer’s disease (AD), the long-term expression of HO-1 can be detected, which can lead to cytotoxic ferroptosis via iron accumulation. In contrast, the transient induction of HO-1 in the peri-injured region may have regenerative potential (e.g., angiogenesis, neurogenesis, and mitochondrial biogenesis) and neurovascular protective effects through the CO-mediated signaling pathway, the antioxidant properties of bilirubin, and the iron-mediated ferritin synthesis. In this review, we discuss the dual roles of HO-1 and its metabolites in various neurovascular diseases, including age-related macular degeneration, ischemia-reperfusion injury, traumatic brain injury, Gilbert’s syndrome, and AD.
Collapse
Affiliation(s)
- Yoon Kyung Choi
- Bio/Molecular Informatics Center, Department of Bioscience and Biotechnology, Konkuk University, Seoul 05029, Korea
- Correspondence: (Y.K.C.); (Y.-M.K.); Tel.: +82-2-450-0558 (Y.K.C.); +82-33-250-8831 (Y.-M.K.)
| | - Young-Myeong Kim
- Department of Molecular and Cellular Biochemistry, School of Medicine, Kangwon National University, Chuncheon 24341, Korea
- Correspondence: (Y.K.C.); (Y.-M.K.); Tel.: +82-2-450-0558 (Y.K.C.); +82-33-250-8831 (Y.-M.K.)
| |
Collapse
|
24
|
Rahimian R, Belliveau C, Chen R, Mechawar N. Microglial Inflammatory-Metabolic Pathways and Their Potential Therapeutic Implication in Major Depressive Disorder. Front Psychiatry 2022; 13:871997. [PMID: 35782423 PMCID: PMC9245023 DOI: 10.3389/fpsyt.2022.871997] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Accepted: 05/23/2022] [Indexed: 12/19/2022] Open
Abstract
Increasing evidence supports the notion that neuroinflammation plays a critical role in the etiology of major depressive disorder (MDD), at least in a subset of patients. By virtue of their capacity to transform into reactive states in response to inflammatory insults, microglia, the brain's resident immune cells, play a pivotal role in the induction of neuroinflammation. Experimental studies have demonstrated the ability of microglia to recognize pathogens or damaged cells, leading to the activation of a cytotoxic response that exacerbates damage to brain cells. However, microglia display a wide range of responses to injury and may also promote resolution stages of inflammation and tissue regeneration. MDD has been associated with chronic priming of microglia. Recent studies suggest that altered microglial morphology and function, caused either by intense inflammatory activation or by senescence, may contribute to depression and associated impairments in neuroplasticity. In this context, modifying microglia phenotype by tuning inflammatory pathways might have important translational relevance to harness neuroinflammation in MDD. Interestingly, it was recently shown that different microglial phenotypes are associated with distinct metabolic pathways and analysis of the underlying molecular mechanisms points to an instrumental role for energy metabolism in shaping microglial functions. Here, we review various canonical pro-inflammatory, anti-inflammatory and metabolic pathways in microglia that may provide new therapeutic opportunities to control neuroinflammation in brain disorders, with a strong focus on MDD.
Collapse
Affiliation(s)
- Reza Rahimian
- Douglas Mental Health University Institute, McGill Group for Suicide Studies, Verdun, QC, Canada
| | - Claudia Belliveau
- Douglas Mental Health University Institute, McGill Group for Suicide Studies, Verdun, QC, Canada
- Integrated Program in Neuroscience, McGill University, Montreal, QC, Canada
| | - Rebecca Chen
- Douglas Mental Health University Institute, McGill Group for Suicide Studies, Verdun, QC, Canada
- Integrated Program in Neuroscience, McGill University, Montreal, QC, Canada
| | - Naguib Mechawar
- Douglas Mental Health University Institute, McGill Group for Suicide Studies, Verdun, QC, Canada
- Integrated Program in Neuroscience, McGill University, Montreal, QC, Canada
- Department of Psychiatry, McGill University, Montreal, QC, Canada
| |
Collapse
|
25
|
Reducing Chemotherapy-Induced DNA Damage via nAChR-Mediated Redox Reprograming-A New Mechanism for SCLC Chemoresistance Boosted by Nicotine. Cancers (Basel) 2022; 14:cancers14092272. [PMID: 35565402 PMCID: PMC9100082 DOI: 10.3390/cancers14092272] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 04/27/2022] [Accepted: 04/29/2022] [Indexed: 02/01/2023] Open
Abstract
Up to 60% of patients with small cell lung cancer (SCLC) continue to smoke, which is associated with worse clinical outcomes. Platinum-based chemotherapies, in combination with topoisomerase inhibitors, are first-line therapies for SCLC, with rapid chemoresistance as a major barrier. We provided evidence in this study that nicotine and its major metabolite, cotinine, at physiologically relevant concentrations, reduced the efficacy of platinum-based chemotherapies and facilitated chemoresistance in SCLC cells. Mechanistically, nicotine or cotinine reduced chemotherapy-induced DNA damage by modulating cellular redox processes, with nAChRs as the upstream targets. Surprisingly, cisplatin treatment alone also increased the levels of nAChRs in SCLC cells, which served as a self-defense mechanism against platinum-based therapies. These discoveries were confirmed in long-term in vitro and in vivo studies. Collectively, our results depicted a novel and clinically important mechanism of chemoresistance in SCLC treatment: nicotine exposure significantly compromises the efficacy of platinum-based chemotherapies in SCLC treatment by reducing therapy-induced DNA damage and accelerating chemoresistance acquisition. The results also emphasized the urgent need for tobacco cessation and the control of NRT use for SCLC management.
Collapse
|
26
|
Mancuso C. The brain heme oxygenase/biliverdin reductase system as a target in drug research and development. Expert Opin Ther Targets 2022; 26:361-374. [PMID: 35285395 DOI: 10.1080/14728222.2022.2052848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION The heme oxygenase/biliverdin reductase (HO/BVR) system is involved in heme metabolism. The inducible isoform of HO (HO-1) and BVR both exert cytoprotective effects by enhancing cell stress response. In this context, some xenobiotics, which target HO-1, including herbal products, behave as neuroprotectants in several experimental models of neurodegeneration. Despite this, no drug having either HO-1 or BVR as a main target is currently available. AREAS COVERED After a description of the brain HO/BVR system, the paper analyzes the main classes of drugs acting on the nervous system, with HO as second-level target, and their neuroprotective potential. Finally, the difficulties that exist for the development of drugs acting on HO/BVR and the possible ways to overcome these hurdles are examined. EXPERT OPINION Although the limited clinical evidence has restricted the translational research on the HO/BVR system, mainly because of the dual nature of its by-products, there has been growing interest in the therapeutic potential of these enzymes. Scientists should boost the translational research on the HO/BVR system which could be supported by the significant evidence provided by preclinical studies.
Collapse
Affiliation(s)
- Cesare Mancuso
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy.,Department of Healthcare Surveillance and Bioethics, Section of Pharmacology, Università Cattolica del Sacro Cuore, Rome, Italy
| |
Collapse
|
27
|
Namgung U, Kim KJ, Jo BG, Park JM. Vagus nerve stimulation modulates hippocampal inflammation caused by continuous stress in rats. J Neuroinflammation 2022; 19:33. [PMID: 35109857 PMCID: PMC8812005 DOI: 10.1186/s12974-022-02396-z] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2021] [Accepted: 01/20/2022] [Indexed: 12/12/2022] Open
Abstract
Background Previous studies have shown that vagus nerve stimulation (VNS) can attenuate inflammatory responses in peripheral tissues and also improve some neurological disorders and cognitive function in the brain. However, it is not clear how VNS is involved in neuropathological processes in brain tissues. Here, we investigated the regulatory effects of VNS on the production of proinflammatory cytokines in the hippocampus of an animal model of continuous stress (CS). Methods CS was induced by placing rats in cages immersed with water, and acute or chronic electrical stimulation was applied to the cervical vagus nerve of CS animals. Protein levels in the gastric and hippocampal tissues were measured by western blotting and protein signals analyzed by immunofluorescence staining. von Frey test and forced swimming test were performed to assess pain sensitivity and depressive-like behavior in rats, respectively. Results Levels of TNF-α, IL-1β, and IL-6 in the gastric and hippocampal tissues were significantly increased in CS animals compared to the untreated control and downregulated by acute VNS (aVNS). Iba-1-labeled microglial cells in the hippocampus of CS animals revealed morphological features of activated inflammatory cells and then changed to a normal shape by VNS. VNS elevated hippocampal expression of α7 nicotinic acetylcholine receptors (α7 nAChR) in CS animals, and pharmacological blockade of α7 nAChR increased the production of TNF-α, IL-1β, and IL-6, thus suppressing cholinergic anti-inflammatory activity that was mediated by VNS. Chronic VNS (cVNS) down-regulated the hippocampal production of active form of caspase 3 and 5-HT1A receptors and also decreased levels of TNF-α, IL-1β, and IL-6 in the gastric and hippocampal tissues of CS animals. Pain sensitivity and depressive-like behavior, which were increased by CS, were improved by cVNS. Conclusions Our data suggest that VNS may be involved in modulating pathophysiological processes caused by CS in the brain.
Collapse
Affiliation(s)
- Uk Namgung
- Department of Oriental Medicine, Institute of Bioscience and Integrative Medicine, Daejeon University, Daehak-ro 62, Daejeon, 34520, South Korea.
| | - Ki-Joong Kim
- Department of Oriental Medicine, Institute of Bioscience and Integrative Medicine, Daejeon University, Daehak-ro 62, Daejeon, 34520, South Korea
| | - Byung-Gon Jo
- Department of Oriental Medicine, Institute of Bioscience and Integrative Medicine, Daejeon University, Daehak-ro 62, Daejeon, 34520, South Korea
| | - Jong Min Park
- Department of Oriental Medicine, Institute of Bioscience and Integrative Medicine, Daejeon University, Daehak-ro 62, Daejeon, 34520, South Korea
| |
Collapse
|
28
|
Cao K, Xiang J, Dong YT, Xu Y, Guan ZZ. Activation of α7 Nicotinic Acetylcholine Receptor by its Selective Agonist Improved Learning and Memory of Amyloid Precursor Protein/Presenilin 1 (APP/PS1) Mice via the Nrf2/HO-1 Pathway. Med Sci Monit 2022; 28:e933978. [PMID: 34980874 PMCID: PMC8742434 DOI: 10.12659/msm.933978] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Accepted: 10/24/2021] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND To reveal the mechanism underlying the effect of alpha7 nicotinic acetylcholine receptor (nAChR) on neurodegeneration in Alzheimer disease (AD), the influence of the receptor on recognition in APP/PS1 mice was evaluated by using its selective agonist (PNU-282987). MATERIAL AND METHODS APP/PS1 and wild-type (WT) mice were treated with PNU or saline, respectively, for 7 days at the ages of 6 and 10 months. RESULTS Morris water maze analysis showed that both at 6 and 10 months of age, PNU treatment enhanced the learning and memory of APP/PS1 mice. However, PNU treatment did not alter the number of senile plaques. Furthermore, a higher protein expression of Nrf2/HO-1, ADAM10, SYP, and SNAP-25, and a lower level of oxidative stress, were observed in the hippocampus of APP/PS1 mice treated with PNU compared with the control group. CONCLUSIONS The results indicated that the activation of alpha7 nAChR by PNU improved the learning and memory of mice carrying the APP/PS1 mutation, regulated the levels of enzymes that mediate APP metabolization to reduce ß-amyloid peptide damage, and decreased the level of oxidative stress and maintained synaptic plasticity, in which the mechanism might be enhancement of the Nrf2/HO-1 pathway.
Collapse
Affiliation(s)
- Kun Cao
- Department of Pathology, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, PR China
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, PR China
| | - Jie Xiang
- Department of Pathology, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, PR China
- Key Laboratory of Endemic and Ethnic Diseases (Guizhou Medical University) of the Ministry of Education, Guiyang, Guizhou, PR China
| | - Yang-Ting Dong
- Key Laboratory of Endemic and Ethnic Diseases (Guizhou Medical University) of the Ministry of Education, Guiyang, Guizhou, PR China
- Provincial Key Laboratory of Medical Molecular Biology, Guiyang, Guizhou, PR China
| | - Yi Xu
- Department of Pathology, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, PR China
- Key Laboratory of Endemic and Ethnic Diseases (Guizhou Medical University) of the Ministry of Education, Guiyang, Guizhou, PR China
| | - Zhi-Zhong Guan
- Department of Pathology, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, PR China
- Key Laboratory of Endemic and Ethnic Diseases (Guizhou Medical University) of the Ministry of Education, Guiyang, Guizhou, PR China
- Provincial Key Laboratory of Medical Molecular Biology, Guiyang, Guizhou, PR China
| |
Collapse
|
29
|
Lycium barbarum Polysaccharides Promotes Mitochondrial Biogenesis and Energy Balance in a NAFLD Cell Model. Chin J Integr Med 2021; 28:975-982. [PMID: 34874519 DOI: 10.1007/s11655-021-3309-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/17/2021] [Indexed: 10/19/2022]
Abstract
OBJECTIVE To explore the protective effect and underlying mechanism of Lycium barbarum polysaccharides (LBP) in a non-alcoholic fatty liver disease (NAFLD) cell model. METHODS Normal human hepatocyte LO2 cells were treated with 1 mmol/L free fatty acids (FFA) mixture for 24 h to induce NAFLD cell model. Cells were divided into 5 groups, including control, model, low-, medium- and high dose LBP (30,100 and 300 µg/mL) groups. The monosaccharide components of LBP were analyzed with high performance liquid chromatography. Effects of LBP on cell viability and intracellular lipid accumulation were assessed by cell counting Kit-8 assay and oil red O staining, respectively. Triglyceride (TG), alanine aminotransferase (ALT), aspartate aminotransferase (AST), adenosine triphosphate (ATP) and oxidative stress indicators were evaluated. Energy balance and mitochondrial biogenesis related mRNA and proteins were determined by quantitative real-time polymerase chain reaction and Western blot, respectively. RESULTS Heteropolysaccharides with mannose and glucose are the main components of LBP. LBP treatment significantly decreased intracellular lipid accumulation as well as TG, ALT, AST and malondialdehyde levels (P<0.05 or P<0.01), increased the levels of superoxide dismutase, phospholipid hydroperoxide glutathione peroxidase, catalase, and ATP in NAFLD cell model (P<0.05). Meanwhile, the expression of uncoupling protein 2 was down-regulated and peroxisome proliferator-activated receptor gamma coactivator-1α/nuclear respiratory factor 1/mitochondrial transcription factor A pathway was up-regulated (P<0.05). CONCLUSION LBP promotes mitochondrial biogenesis and improves energy balance in NAFLD cell model.
Collapse
|
30
|
Singh NK, Garabadu D. Quercetin Exhibits α7nAChR/Nrf2/HO-1-Mediated Neuroprotection Against STZ-Induced Mitochondrial Toxicity and Cognitive Impairments in Experimental Rodents. Neurotox Res 2021; 39:1859-1879. [PMID: 34554409 DOI: 10.1007/s12640-021-00410-5] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 08/28/2021] [Accepted: 08/30/2021] [Indexed: 01/10/2023]
Abstract
The objective of the present study was to investigate the α7nAChR-mediated Nrf2-dependant protective activity against streptozotocin (STZ)-induced brain mitochondrial toxicity in Alzheimer's disease (AD)-like rats. STZ (3 mg/kg) was injected through an intracerebroventricular route to induce AD-like dementia. Repeated Quercetin (50 mg/kg, i.p.) administration attenuated cognitive impairments in the STZ-challenged animals during Morris water-maze and Y-maze tests. Quercetin significantly mitigated the STZ-induced increase in cholinergic dysfunction, such as the increase in acetylcholinesterase activity, decrease in acetylcholine level, and activity of choline acetyltransferase, and increase in amyloid-beta aggregation and mitochondrial toxicity in respect of mitochondrial bioenergetics, integrity, and oxidative stress in memory-challenged rat hippocampus, prefrontal cortex and, amygdala. Further, Quercetin significantly attenuated STZ-induced reduction in the α7nAChRs and HO-1 expression levels in the selected rat brain regions. On the contrary, trigonelline (10 mg/kg, i.p.) and methyllycaconitine (2 mg/kg; i.p.) abolished the neuroprotective effects of Quercetin against STZ-induced behavioral, molecular, and biochemical alterations in the AD-like animals. Hence, Quercetin exhibits α7nAChR/Nrf2/HO-1-mediated neuroprotection against STZ-challenged AD-like animals. Thus, Quercetin could be considered as a potential therapeutic option in the management of AD.
Collapse
Affiliation(s)
- Niraj Kumar Singh
- Division of Pharmacology, Institute of Pharmaceutical Research, GLA University, Mathura, 281406, India
| | - Debapriya Garabadu
- Division of Pharmacology, Institute of Pharmaceutical Research, GLA University, Mathura, 281406, India.
- Department of Pharmacology, School of Health Sciences, Central University of Punjab, Bathinda, 151401, India.
| |
Collapse
|
31
|
Wang C, Tang T, Wang Y, Nie X, Li K. Simvastatin affects the PPARα signaling pathway and causes oxidative stress and embryonic development interference in Mugilogobius abei. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2021; 239:105951. [PMID: 34467877 DOI: 10.1016/j.aquatox.2021.105951] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/27/2021] [Revised: 08/15/2021] [Accepted: 08/18/2021] [Indexed: 06/13/2023]
Abstract
Simvastatin (SV) is a common hypolipidemic drug in clinical medicine that can reduce endogenous cholesterol biosynthesis by inhibiting hydroxyl-methyl-glutaryl coenzyme A reductase. SV took a large market share in the lipid-lowering drugs and it is frequently detected in various water bodies due to its increasing consumption in past years. In the present investigation, we selected a native fish species in the Pearl River Basin in China, Mugilogobius abei (M. abei), to study the effects of SV on non-target aquatic organisms. Results showed that a significant decrease in the volume of adipocytes under SV exposure were observed on oil red O section, and the expression of HMG-CoAR decreased significantly. The mRNA and protein expression of PPARα were significantly up-regulated, the expressions of other genes related to lipid metabolism were up-regulated to varying degrees as well. There was a positive correlation between the concentrations of SV and the protein expressions of plasma phospholipid transfer protein (PLTP) and cholesterolester transfer protein (CETP). In addition, the frozen sections showed that SV led to ROS accumulation in liver in a time and concentration dependent manner. The mRNA and protein expressions of Nrf2 were significantly up-regulated after 24 hours of SV exposure. Some biomarkers associated with antioxidant such as Trx2, TrxR and MDA content were positively correlated with the exposure concentration and time, while the content of GSH decreased sharply. It is noteworthy that the environmentally relevant concentration (0.5 μg/L) of SV exposure caused delayed embryonic development and deformations, decreased hatching rates. We conclude that SV promotes fat metabolism, gives rise to oxidative stress and has significant toxicity on embryo development in M. abei.
Collapse
Affiliation(s)
- Chao Wang
- Department of Ecology/Hydrobiology Research Institute, Jinan University, Guangzhou 510632, China
| | - Tianli Tang
- Department of Ecology/Hydrobiology Research Institute, Jinan University, Guangzhou 510632, China
| | - Yimeng Wang
- Department of Ecology/Hydrobiology Research Institute, Jinan University, Guangzhou 510632, China
| | - Xiangping Nie
- Department of Ecology/Hydrobiology Research Institute, Jinan University, Guangzhou 510632, China; Key Laboratory of Eutrophication and Red Tide Prevention of Guangdong Higher Education Institutes, Jinan University, Guangzhou 510632, China
| | - Kaibin Li
- Pearl River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou 510380, China
| |
Collapse
|
32
|
Webster SE, Sklar NC, Spitsbergen JB, Stanchfield ML, Webster MK, Linn DM, Otteson DC, Linn CL. Stimulation of α7 nAChR leads to regeneration of damaged neurons in adult mammalian retinal disease models. Exp Eye Res 2021; 210:108717. [PMID: 34348130 PMCID: PMC8459670 DOI: 10.1016/j.exer.2021.108717] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 07/10/2021] [Accepted: 07/30/2021] [Indexed: 12/13/2022]
Abstract
The adult mammal lacks the ability to regenerate neurons lost to retinal damage or disease in a meaningful capacity. However, previous studies from this laboratory have demonstrated that PNU-282987, an α7 nicotinic acetylcholine receptor agonist, elicits a robust neurogenic response in the adult murine retina. With eye drop application of PNU-282987, Müller glia cells re-enter the cell cycle and produce progenitor-like cells that can differentiate into various types of retinal neurons. In this study, we analyzed the regenerative capability of PNU-282987 in two retinal disease models and identified the source of newly regenerated neurons. Wild-type mice and mice with a transgenic Müller-glia lineage tracer were manipulated to mimic loss of retinal cells associated with glaucoma or photoreceptor degeneration. Following treatment with PNU-282987, the regenerative response of retinal neurons was quantified and characterized. After onset of photoreceptor degeneration, PNU-282987 was able to successfully regenerate both rod and cone photoreceptors. Quantification of this response demonstrated significant regeneration, restoring photoreceptors to near wild-type density. In mice that had glaucoma-like conditions induced, PNU-282987 treatment led to a significant increase in retinal ganglion cells. Retrograde labeling of optic nerve axon fibers demonstrated that newly regenerated axons projected into the optic nerve. Lineage tracing analysis demonstrated that these new neurons were derived from Müller glia. These results demonstrate that PNU-282987 can induce retinal regeneration in adult mice following onset of retinal damage. The ability of PNU-282987 to regenerate retinal neurons in a robust manner offers a new direction for developing novel and potentially transformative treatments to combat neurodegenerative disease.
Collapse
Affiliation(s)
- Sarah E Webster
- Western Michigan University, Department of Biological Sciences, Kalamazoo, MI, United States
| | - Nathan C Sklar
- Western Michigan University Homer Stryker M.D. School of Medicine, Kalamazoo, MI, United States
| | - Jake B Spitsbergen
- Western Michigan University, Department of Biological Sciences, Kalamazoo, MI, United States
| | - Megan L Stanchfield
- Western Michigan University, Department of Biological Sciences, Kalamazoo, MI, United States
| | - Mark K Webster
- Western Michigan University, Department of Biological Sciences, Kalamazoo, MI, United States
| | - David M Linn
- Grand Valley State University, Department of Biomedical Sciences, Allendale, MI, United States
| | - Deborah C Otteson
- University of Houston College of Optometry, Houston, TX, United States
| | - Cindy L Linn
- Western Michigan University, Department of Biological Sciences, Kalamazoo, MI, United States.
| |
Collapse
|
33
|
P P, Justin A, Ananda Kumar TD, Chinaswamy M, Kumar BRP. Glitazones Activate PGC-1α Signaling via PPAR-γ: A Promising Strategy for Antiparkinsonism Therapeutics. ACS Chem Neurosci 2021; 12:2261-2272. [PMID: 34125534 DOI: 10.1021/acschemneuro.1c00085] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Understanding various aspects of Parkinson's disease (PD) by researchers could lead to a better understanding of the disease and provide treatment alternatives that could significantly improve the quality of life of patients suffering from neurodegenerative disorders. Significant progress has been made in recent years toward this goal, but there is yet no available treatment with confirmed neuroprotective effects. Recent studies have shown the potential of PPARγ agonists, which are the ligand activated transcriptional factor of the nuclear hormone superfamily, as therapeutic targets for various neurodegenerative disorders. The activation of central PGC-1α mediates the potential role against neurogenerative diseases like PD, Huntington's disease, Alzheimer's disease, and amyotrophic lateral sclerosis. Further understanding the mechanism of neurodegeneration and the role of glitazones in the activation of PGC-1α signaling could lead to a novel therapeutic interventions against PD. Keeping this aspect in focus, the present review highlights the pathogenic mechanism of PD and the role of glitazones in the activation of PGC-1α via PPARγ for the treatment of neurodegenerative disorders.
Collapse
Affiliation(s)
- Prabitha P
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru, Karnataka 570 015, India
| | - Antony Justin
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty, Nilgiris, Tamilnadu 643 001, India
| | - T. Durai Ananda Kumar
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru, Karnataka 570 015, India
| | - Mithuna Chinaswamy
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru, Karnataka 570 015, India
| | - B. R. Prashantha Kumar
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru, Karnataka 570 015, India
| |
Collapse
|
34
|
Abstract
Mutations in the genes coding for tryptophan-hydrolase-2 and the scaffold protein FKBP5 are associated with an increased risk of suicide. The mutation in both cases enhances the enzymatic activity of glycogen synthase kinase-3 (GSK3). Conversely, anti-suicidal medications, such as lithium, clozapine, and ketamine, indirectly inhibit the activity of GSK3. When GSK3 is active, it promotes the metabolic removal of the transcription factor NRF2 (nuclear factor erythroid 2-related factor-2), which suppresses the transcription of multiple genes that encode anti-oxidative and anti-inflammatory proteins. Notably, several suicide-biomarkers bear witness to an ongoing inflammatory process. Moreover, alterations in serum lipid levels measured in suicidal individuals are mirrored by data obtained in mice with genetic deletion of the NRF2 gene. Inflammation is presumably causally related to both dysphoria and anger, two factors relevant for suicide ideation and attempt. Preventing the catabolism of NRF2 could be a strategy to obtain novel suicide-prophylactic medications. Possible candidates are minocycline and nicotinic-α7 agonists. The antibiotic minocycline indirectly activates NRF2-transcriptional activity, whereas the activation of nicotinic-α7 receptors indirectly inhibits GSK3.
Collapse
|
35
|
Mazzoccoli G, Kvetnoy I, Mironova E, Yablonskiy P, Sokolovich E, Krylova J, Carbone A, Anderson G, Polyakova V. The melatonergic pathway and its interactions in modulating respiratory system disorders. Biomed Pharmacother 2021; 137:111397. [PMID: 33761613 DOI: 10.1016/j.biopha.2021.111397] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 02/09/2021] [Accepted: 02/10/2021] [Indexed: 02/08/2023] Open
Abstract
Melatonin is a key intracellular neuroimmune-endocrine regulator and coordinator of multiple complex and interrelated biological processes. The main functions of melatonin include the regulation of neuroendocrine and antioxidant system activity, blood pressure, rhythms of the sleep-wake cycle, the retardation of ageing processes, as well as reseting and optimizing mitochondria and thereby the cells of the immune system. Melatonin and its agonists have therefore been mooted as a treatment option across a wide array of medical disorders. This article reviews the role of melatonin in the regulation of respiratory system functions under normal and pathological conditions. Melatonin can normalize the structural and functional organization of damaged lung tissues, by a number of mechanisms, including the regulation of signaling molecules, oxidant status, lipid raft function, optimized mitochondrial function and reseting of the immune response over the circadian rhythm. Consequently, melatonin has potential clinical utility for bronchial asthma, chronic obstructive pulmonary disease, lung cancer, lung vascular diseases, as well as pulmonary and viral infections. The integration of melatonin's effects with the alpha 7 nicotinic receptor and the aryl hydrocarbon receptor in the regulation of mitochondrial function are proposed as a wider framework for understanding the role of melatonin across a wide array of diverse pulmonary disorders.
Collapse
Affiliation(s)
- Gianluigi Mazzoccoli
- Department of Medical Sciences, Division of Internal Medicine and Chronobiology Laboratory, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo 71013, Italy.
| | - Igor Kvetnoy
- Saint Petersburg Institute of Phthisiopulmonology, Lygovsky Ave. 2-4, Saint Petersburg 191036, Russian Federation; Department of Pathology, Saint Petersburg State University, University Embankment, 7/9, Saint Petersburg 199034, Russian Federation
| | - Ekaterina Mironova
- Saint Petersburg Institute of Bioregulation and Gerontology, Dynamo Ave., 3, Saint Petersburg 197110, Russian Federation
| | - Petr Yablonskiy
- Saint Petersburg Institute of Phthisiopulmonology, Lygovsky Ave. 2-4, Saint Petersburg 191036, Russian Federation
| | - Evgenii Sokolovich
- Saint Petersburg Institute of Phthisiopulmonology, Lygovsky Ave. 2-4, Saint Petersburg 191036, Russian Federation
| | - Julia Krylova
- Saint Petersburg Institute of Phthisiopulmonology, Lygovsky Ave. 2-4, Saint Petersburg 191036, Russian Federation; Pavlov First Saint Petersburg State Medical University, Lev Tolstoy str. 6-8, Saint Petersburg 197022, Russian Federation
| | - Annalucia Carbone
- Department of Medical Sciences, Division of Internal Medicine and Chronobiology Laboratory, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo 71013, Italy
| | | | - Victoria Polyakova
- Saint Petersburg Institute of Phthisiopulmonology, Lygovsky Ave. 2-4, Saint Petersburg 191036, Russian Federation; St. Petersburg State Pediatric Medical University, Litovskaia str. 2, Saint-Petersburg 194100, Russian Federation
| |
Collapse
|
36
|
Changing Functional Signatures of Microglia along the Axis of Brain Aging. Int J Mol Sci 2021; 22:ijms22031091. [PMID: 33499206 PMCID: PMC7865559 DOI: 10.3390/ijms22031091] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Revised: 01/19/2021] [Accepted: 01/20/2021] [Indexed: 12/19/2022] Open
Abstract
Microglia, the innate immune cells of the brain, are commonly perceived as resident macrophages of the central nervous system (CNS). This definition, however, requires further specification, as under healthy homeostatic conditions, neither morphological nor functional properties of microglia mirror those of classical macrophages. Indeed, microglia adapt exceptionally well to their microenvironment, becoming a legitimate member of the cellular brain architecture. The ramified or surveillant microglia in the young adult brain are characterized by specific morphology (small cell body and long, thin motile processes) and physiology (a unique pattern of Ca2+ signaling, responsiveness to various neurotransmitters and hormones, in addition to classic “immune” stimuli). Their numerous physiological functions far exceed and complement their immune capabilities. As the brain ages, the respective changes in the microglial microenvironment impact the functional properties of microglia, triggering further rounds of adaptation. In this review, we discuss the recent data showing how functional properties of microglia adapt to age-related changes in brain parenchyma in a sex-specific manner, with a specific focus on early changes occurring at middle age as well as some strategies counteracting the aging of microglia.
Collapse
|
37
|
Anderson G, Maes M. Mitochondria and immunity in chronic fatigue syndrome. Prog Neuropsychopharmacol Biol Psychiatry 2020; 103:109976. [PMID: 32470498 DOI: 10.1016/j.pnpbp.2020.109976] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2020] [Accepted: 05/19/2020] [Indexed: 02/07/2023]
Abstract
It is widely accepted that the pathophysiology and treatment of myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS) could be considerably improved. The heterogeneity of ME/CFS and the confusion over its classification have undoubtedly contributed to this, although this would seem a consequence of the complexity of the array of ME/CFS presentations and high levels of diverse comorbidities. This article reviews the biological underpinnings of ME/CFS presentations, including the interacting roles of the gut microbiome/permeability, endogenous opioidergic system, immune cell mitochondria, autonomic nervous system, microRNA-155, viral infection/re-awakening and leptin as well as melatonin and the circadian rhythm. This details not only relevant pathophysiological processes and treatment options, but also highlights future research directions. Due to the complexity of interacting systems in ME/CFS pathophysiology, clarification as to its biological underpinnings is likely to considerably contribute to the understanding and treatment of other complex and poorly managed conditions, including fibromyalgia, depression, migraine, and dementia. The gut and immune cell mitochondria are proposed to be two important hubs that interact with the circadian rhythm in driving ME/CFS pathophysiology.
Collapse
Affiliation(s)
- G Anderson
- CRC Scotland & London, Eccleston Square, London, UK.
| | - M Maes
- Dept Psychiatry, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand; Dept Psychiatry, Medical University Plovdiv, Plovdiv, Bulgaria.; IMPACT Research Center, Deakin University, Geelong, Australia
| |
Collapse
|
38
|
Osama A, Zhang J, Yao J, Yao X, Fang J. Nrf2: a dark horse in Alzheimer's disease treatment. Ageing Res Rev 2020; 64:101206. [PMID: 33144124 DOI: 10.1016/j.arr.2020.101206] [Citation(s) in RCA: 150] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2020] [Revised: 10/23/2020] [Accepted: 10/27/2020] [Indexed: 12/12/2022]
Abstract
Alzheimer's disease (AD), an age-dependent neurodegenerative disorder, is the main cause of dementia. Common hallmarks of AD include the amyloid β-peptide (Aβ) aggregation, high levels of hyperphosphorylated tau protein (p-tau) and failure in redox homeostasis. To date, all proposed drugs affecting Aβ and/or p-tau have been failed in clinical trials. A decline in the expression of the transcription factor Nrf2 (nuclear factor-erythroid 2-p45 derived factor 2) and its driven genes (NQO1, HO-1, and GCLC), and alteration of the Nrf2-related pathways have been observed in AD brains. Nrf2 plays a critical role in maintaining cellular redox homeostasis and regulating inflammation response. Nrf2 activation also provides cytoprotection against increasing pathologies including neurodegenerative diseases. These lines of evidence imply that Nrf2 activation may be a novel AD treatment option. Interestingly, recent studies have also demonstrated that Nrf2 interferes with several key pathogenic processes in AD including Aβ and p-tau pathways. The current review aims to provide insights into the role of Nrf2 in AD. Also, we discuss the progress and challenges regarding the Nrf2 activators for AD treatment.
Collapse
Affiliation(s)
- Alsiddig Osama
- State Key Laboratory of Applied Organic Chemistry and College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, 730000, China
| | - Junmin Zhang
- State Key Laboratory of Applied Organic Chemistry and College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, 730000, China; State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Avenida Wai Long, Taipa, Macau, China
| | - Juan Yao
- School of pharmacy, Gansu University of Chinese Medicine, Lanzhou, 730000, China
| | - Xiaojun Yao
- State Key Laboratory of Applied Organic Chemistry and College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, 730000, China; State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Avenida Wai Long, Taipa, Macau, China.
| | - Jianguo Fang
- State Key Laboratory of Applied Organic Chemistry and College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, 730000, China.
| |
Collapse
|
39
|
Qu Z, Sun J, Zhang W, Yu J, Zhuang C. Transcription factor NRF2 as a promising therapeutic target for Alzheimer's disease. Free Radic Biol Med 2020; 159:87-102. [PMID: 32730855 DOI: 10.1016/j.freeradbiomed.2020.06.028] [Citation(s) in RCA: 78] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 06/15/2020] [Accepted: 06/17/2020] [Indexed: 12/12/2022]
Abstract
Oxidative stress is considered as one of the pathogenesis of Alzheimer's disease (AD) and plays an important role in the occurrence and development of AD. Nuclear factor erythroid 2 related factor 2 (NRF2) is a key regulatory of oxidative stress defence. There is growing evidence indicating the relationship between NRF2 and AD. NRF2 activation mitigates multiple pathogenic processes involved in AD by upregulating antioxidative defense, inhibiting neuroinflammation, improving mitochondrial function, maintaining proteostasis, and inhibiting ferroptosis. In addition, several NRF2 activators are currently being evaluated as AD therapeutic agents in clinical trials. Thus, targeting NRF2 has been the focus of a new strategy for prevention and treatment of AD. In this review, the role of NRF2 in AD and the NRF2 activators advanced into clinical and preclinical studies will be summarized.
Collapse
Affiliation(s)
- Zhuo Qu
- School of Pharmacy, Key Laboratory of Hui Ethnic Medicine Modernization, Ministry of Education, Ningxia Medical University, 1160 Shengli Street, Yinchuan, 750004, China
| | - Jiachen Sun
- School of Biotechnology and Food Science, Tianjin University of Commerce, 409 Guangrong Road, Tianjin, 300134, China
| | - Wannian Zhang
- School of Pharmacy, Key Laboratory of Hui Ethnic Medicine Modernization, Ministry of Education, Ningxia Medical University, 1160 Shengli Street, Yinchuan, 750004, China; School of Pharmacy, Second Military Medical University, 325 Guohe Road, Shanghai, 200433, China
| | - Jianqiang Yu
- School of Pharmacy, Key Laboratory of Hui Ethnic Medicine Modernization, Ministry of Education, Ningxia Medical University, 1160 Shengli Street, Yinchuan, 750004, China.
| | - Chunlin Zhuang
- School of Pharmacy, Key Laboratory of Hui Ethnic Medicine Modernization, Ministry of Education, Ningxia Medical University, 1160 Shengli Street, Yinchuan, 750004, China; School of Pharmacy, Second Military Medical University, 325 Guohe Road, Shanghai, 200433, China.
| |
Collapse
|
40
|
Mitochondrial biogenesis in organismal senescence and neurodegeneration. Mech Ageing Dev 2020; 191:111345. [DOI: 10.1016/j.mad.2020.111345] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 08/17/2020] [Accepted: 08/27/2020] [Indexed: 12/19/2022]
|
41
|
Bento-Pereira C, Dinkova-Kostova AT. Activation of transcription factor Nrf2 to counteract mitochondrial dysfunction in Parkinson's disease. Med Res Rev 2020; 41:785-802. [PMID: 32681666 DOI: 10.1002/med.21714] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 06/29/2020] [Accepted: 07/06/2020] [Indexed: 12/20/2022]
Abstract
Parkinson's disease (PD) is a progressive neurodegenerative disorder, for which no disease-modifying therapies are available to date. Although understanding of the precise aetiology of PD is incomplete, it is clear that age, genetic predisposition and environmental stressors increase the risk. At the cellular level, oxidative stress, chronic neuroinflammation, mitochondrial dysfunction and aberrant protein aggregation have been implicated as contributing factors. These detrimental processes are counteracted by elaborate networks of cellular defence mechanisms, one of which is orchestrated by transcription factor nuclear factor-erythroid 2 p45-related factor 2 (Nrf2; gene name NFE2L2). A wealth of preclinical evidence suggests that Nrf2 activation is beneficial in cellular and animal models of PD. In this review, we summarise the current understanding of mitochondrial dysfunction in PD, the role of Nrf2 in mitochondrial function and explore the potential of Nrf2 as a therapeutic target for mitochondrial dysfunction in PD.
Collapse
Affiliation(s)
- Claudia Bento-Pereira
- Division of Cellular Medicine, School of Medicine, Jacqui Wood Cancer Centre, University of Dundee, Dundee, Scotland, UK
| | - Albena T Dinkova-Kostova
- Division of Cellular Medicine, School of Medicine, Jacqui Wood Cancer Centre, University of Dundee, Dundee, Scotland, UK.,Departments of Medicine and Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
42
|
Mitochondrial Transplantation Attenuates Brain Dysfunction in Sepsis by Driving Microglial M2 Polarization. Mol Neurobiol 2020; 57:3875-3890. [PMID: 32613465 DOI: 10.1007/s12035-020-01994-3] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Accepted: 06/22/2020] [Indexed: 12/14/2022]
Abstract
Activation of microglia and mitochondrial dysfunction are two major contributors to the pathogenesis of sepsis-associated brain dysfunction. Mitochondrial dysfunction can alter the immunological profile of microglia favoring to a pro-inflammatory phenotype. Mitochondrial transplantation, as an emerging mitochondria-targeted therapy, possesses considerable therapeutic potential in various central nervous system injuries or diseases. However, the effects of mitochondrial transplantation on microglial polarization and neuroprotection after sepsis remain unclear. In this study, lipopolysaccharide (LPS)/interferon-γ (IFN-γ) and interleukin-4 (IL-4)/interleukin-13 (IL-13) were used to induce different phenotypes of BV2 microglial cells. We observed that mitochondrial content and function were enhanced in IL-4-/IL-13-stimulated microglia. In vitro, mitochondria treatment conferred neuroprotection by enhancing microglial polarization from the M1 phenotype to the M2 phenotype and suppressing microglial-derived inflammatory cytokine release. Furthermore, microglial phenotypes and behavior tests were assessed after mice were subjected to sepsis by cecal ligation and puncture (CLP) followed by intracerebroventricular injection of exogenous functional mitochondria. We found that mitochondrial transplantation induced microglial M2 rather than M1 response 24 h after sepsis. Mitochondrial transplantation improved behavioral deficits by increasing the latency time in inhibitory avoidance test and decreasing the number of crossing and rearing in the test session of open field test 10 days after CLP onset. These findings indicate that mitochondrial transplantation promotes the phenotypic conversion of microglia and improves cognitive impairment in sepsis survivors, supporting the potential use of exogenous mitochondrial transplantation therapy that may be a potential therapeutic opportunity for sepsis-associated brain dysfunction.
Collapse
|
43
|
Zeng XX, Deng J, Xiang J, Dong YT, Cao K, Liu XH, Chen D, Ran LY, Yang Y, Guan ZZ. Protections against toxicity in the brains of rat with chronic fluorosis and primary neurons exposed to fluoride by resveratrol involves nicotinic acetylcholine receptors. J Trace Elem Med Biol 2020; 60:126475. [PMID: 32142957 DOI: 10.1016/j.jtemb.2020.126475] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 01/28/2020] [Accepted: 02/08/2020] [Indexed: 01/08/2023]
Abstract
Protection of Resveratrol (RSV) against the neurotoxicity induced by high level of fluoride was investigated. Sprague-Dawley (SD) rats and their offspring, as well as cultures of primary neurons were divided randomly into four groups: untreated (control); treated with 50 mg RSV/kg/ (once daily by gavage) or (20 M in the cultured medium); exposed to 50 ppm F- in drinking water or 4 mmol/l in the cultured medium; and exposed to fluoride then RSV as above. The adult rats were treated for 7 months and the offspring sacrificed at 28 days of age; the cultured neurons for 48 h. For general characterization, dental fluorosis was assessed and the fluoride content of the urine measured (by fluoride-electrode) in the rates and the survival of cultured neurons monitored with the CCK-8 test. The spatial learning and memory of rats were assessed with the Morris water maze test. The levels of α7 and α4 nicotinic acetylcholine receptors (nAChRs) were quantified by Western blotting; and the activities of superoxide dismutase (SOD) and catalase (CAT), and the levels of malondialdehyde (MDA) and H2O2 assayed biochemically. The results showed that chronic fluorosis resulted in the impaired learning and memory in rats and their offspring, and more oxidative stress in both rat brains and cultured neurons, which may be associated the lower levels of α7 and α4 nAChR subunits. Interestingly, RSV attenuated all of these toxic effects by fluorosis, indicating that protection against the neurotoxicity of fluoride by RSV might be in mechanism involved enhancing the expressions of these nAChRs.
Collapse
Affiliation(s)
- Xiao-Xiao Zeng
- Departments of Pathology at Guizhou Medical University and the Affiliated Hospital of Guizhou Medical University, PR China; Key Laboratory of Endemic and Ethnic Diseases, (Guizhou Medical University) of the Ministry of Education, PR China; Provincial Key Laboratory of Medical Molecular Biology, Guiyang, 550004, Guizhou, PR China
| | - Jie Deng
- Key Laboratory of Endemic and Ethnic Diseases, (Guizhou Medical University) of the Ministry of Education, PR China; Provincial Key Laboratory of Medical Molecular Biology, Guiyang, 550004, Guizhou, PR China
| | - Jie Xiang
- Departments of Pathology at Guizhou Medical University and the Affiliated Hospital of Guizhou Medical University, PR China; Key Laboratory of Endemic and Ethnic Diseases, (Guizhou Medical University) of the Ministry of Education, PR China
| | - Yang-Ting Dong
- Key Laboratory of Endemic and Ethnic Diseases, (Guizhou Medical University) of the Ministry of Education, PR China; Provincial Key Laboratory of Medical Molecular Biology, Guiyang, 550004, Guizhou, PR China
| | - Kun Cao
- Departments of Pathology at Guizhou Medical University and the Affiliated Hospital of Guizhou Medical University, PR China; Key Laboratory of Endemic and Ethnic Diseases, (Guizhou Medical University) of the Ministry of Education, PR China
| | - Xian-Hong Liu
- Key Laboratory of Endemic and Ethnic Diseases, (Guizhou Medical University) of the Ministry of Education, PR China; Provincial Key Laboratory of Medical Molecular Biology, Guiyang, 550004, Guizhou, PR China
| | - Dan Chen
- Departments of Pathology at Guizhou Medical University and the Affiliated Hospital of Guizhou Medical University, PR China; Key Laboratory of Endemic and Ethnic Diseases, (Guizhou Medical University) of the Ministry of Education, PR China
| | - Long-Yan Ran
- Departments of Pathology at Guizhou Medical University and the Affiliated Hospital of Guizhou Medical University, PR China; Key Laboratory of Endemic and Ethnic Diseases, (Guizhou Medical University) of the Ministry of Education, PR China
| | - Ye Yang
- Key Laboratory of Endemic and Ethnic Diseases, (Guizhou Medical University) of the Ministry of Education, PR China; Provincial Key Laboratory of Medical Molecular Biology, Guiyang, 550004, Guizhou, PR China
| | - Zhi-Zhong Guan
- Departments of Pathology at Guizhou Medical University and the Affiliated Hospital of Guizhou Medical University, PR China; Key Laboratory of Endemic and Ethnic Diseases, (Guizhou Medical University) of the Ministry of Education, PR China; Provincial Key Laboratory of Medical Molecular Biology, Guiyang, 550004, Guizhou, PR China.
| |
Collapse
|
44
|
Song X, Long D. Nrf2 and Ferroptosis: A New Research Direction for Neurodegenerative Diseases. Front Neurosci 2020; 14:267. [PMID: 32372896 PMCID: PMC7186402 DOI: 10.3389/fnins.2020.00267] [Citation(s) in RCA: 371] [Impact Index Per Article: 74.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Accepted: 03/09/2020] [Indexed: 12/19/2022] Open
Abstract
Ferroptosis is a kind of regulated cell death (RCD) caused by the redox state disorder of intracellular microenvironment controlled by glutathione (GSH) peroxidase 4 (GPX4), which is inhibited by iron chelators and lipophilic antioxidants. In addition to classical regulatory mechanisms, new regulatory factors for ferroptosis have been discovered in recent years, such as the P53 pathway, the activating transcription factor (ATF)3/4 pathway, Beclin 1 (BECN1) pathway, and some non-coding RNA. Ferroptosis is closely related to cancer treatment, neurodegenerative diseases, ischemia–reperfusion of organ, neurotoxicity, and others, in particular, in the field of neurodegenerative diseases treatment has aroused people’s interest. The nuclear factor E2 related factor 2 (Nrf2/NFE2L2) has been proved to play a key role in neurodegenerative disease treatment and ferroptosis regulation. Ferroptosis promotes the progression of neurodegenerative diseases, while the expression of Nrf2 and its target genes (Ho-1, Nqo-1, and Trx) has been declined with aging; therefore, there is still insufficient evidence for ferroptosis and Nrf2 regulatory networks in the field of neurodegenerative diseases. In this review, we will provide a brief overview of ferroptosis regulatory mechanisms, as well as an emphasis on the mechanism of Nrf2 regulating ferroptosis. We also highlight the role of ferroptosis and Nrf2 during the process of neurodegenerative diseases and investigate a theoretical basis for further research on the relationship between Nrf2 and ferroptosis in the process of neurodegenerative diseases treatment.
Collapse
Affiliation(s)
- Xiaohua Song
- School of Public Health, University of South China, Hengyang, China
| | - Dingxin Long
- School of Public Health, University of South China, Hengyang, China
| |
Collapse
|
45
|
Su YC, Huang YF, Wu YW, Chen HF, Wu YH, Hsu CC, Hsu YC, Lee JC. MicroRNA-155 inhibits dengue virus replication by inducing heme oxygenase-1-mediated antiviral interferon responses. FASEB J 2020; 34:7283-7294. [PMID: 32277848 DOI: 10.1096/fj.201902878r] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Revised: 02/03/2020] [Accepted: 02/17/2020] [Indexed: 12/21/2022]
Abstract
MicroRNAs (miRNAs) have been reported to directly alter the virus life cycle and virus-host interactions, and so are considered promising molecules for controlling virus infection. In the present study, we observed that miR-155 time-dependently downregulated upon dengue virus (DENV) infection. In contrast, exogenous overexpression of miR-155 appeared to limit viral replication in vitro, suggesting that the low levels of miR-155 would be beneficial for DENV replication. In vivo, overexpression of miR-155 protected ICR suckling mice from the life-threatening effects of DENV infection and reduced virus propagation. Further investigation revealed that the anti-DENV activity of miR-155 was due to target Bach1, resulting in the induction of the heme oxygenase-1 (HO-1)-mediated inhibition of DENV NS2B/NS3 protease activity, ultimately leading to induction of antiviral interferon responses, including interferon-induced protein kinase R (PKR), 2'-5'-oligoadenylate synthetase 1 (OAS1), OAS2, and OAS3 expression, against DENV replication. Collectively, our results provide a promising new strategy to manage DENV infection by modulation of miR-155 expression.
Collapse
Affiliation(s)
- Yu-Chieh Su
- Division of Hematology-Oncology, Department of Internal Medicine, E-Da Hospital, Kaohsiung, Taiwan.,School of Medicine, I-Shou University, Kaohsiung, Taiwan
| | - Yi-Fang Huang
- Department of Biotechnology, College of Life Science, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Yu-Wen Wu
- Department of Biotechnology, College of Life Science, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Hui-Feng Chen
- Department of Biotechnology, College of Life Science, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Yu-Hsuan Wu
- Department of Biotechnology, College of Life Science, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chia-Chun Hsu
- Department of Biotechnology, College of Life Science, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Yao-Chin Hsu
- Department of Chinese medicine, Chi Mei Medical Center, Tainan, Taiwan
| | - Jin-Ching Lee
- Department of Biotechnology, College of Life Science, Kaohsiung Medical University, Kaohsiung, Taiwan.,Drug Development and Value Creation Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan.,Graduate Institute of Medicine, College of Medicine and Graduate Institute of Natural Products, College of Pharmacy, Kaohsiung Medical University, Kaohsiung, Taiwan.,Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan.,Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung, Taiwan
| |
Collapse
|
46
|
The Role of Reactive Oxygen Species in the Life Cycle of the Mitochondrion. Int J Mol Sci 2020; 21:ijms21062173. [PMID: 32245255 PMCID: PMC7139706 DOI: 10.3390/ijms21062173] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 03/12/2020] [Accepted: 03/19/2020] [Indexed: 02/07/2023] Open
Abstract
Currently, it is known that, in living systems, free radicals and other reactive oxygen and nitrogen species play a double role, because they can cause oxidative damage and tissue dysfunction and serve as molecular signals activating stress responses that are beneficial to the organism. It is also known that mitochondria, because of their capacity to produce free radicals, play a major role in tissue oxidative damage and dysfunction and provide protection against excessive tissue dysfunction through several mechanisms, including the stimulation of permeability transition pore opening. This process leads to mitoptosis and mitophagy, two sequential processes that are a universal route of elimination of dysfunctional mitochondria and is essential to protect cells from the harm due to mitochondrial disordered metabolism. To date, there is significant evidence not only that the above processes are induced by enhanced reactive oxygen species (ROS) production, but also that such production is involved in the other phases of the mitochondrial life cycle. Accumulating evidence also suggests that these effects are mediated through the regulation of the expression and the activity of proteins that are engaged in processes such as genesis, fission, fusion, and removal of mitochondria. This review provides an account of the developments of the knowledge on the dynamics of the mitochondrial population, examining the mechanisms governing their genesis, life, and death, and elucidating the role played by free radicals in such processes.
Collapse
|
47
|
Transcriptional activation of antioxidant gene expression by Nrf2 protects against mitochondrial dysfunction and neuronal death associated with acute and chronic neurodegeneration. Exp Neurol 2020; 328:113247. [PMID: 32061629 DOI: 10.1016/j.expneurol.2020.113247] [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: 12/05/2019] [Revised: 02/04/2020] [Accepted: 02/11/2020] [Indexed: 02/07/2023]
Abstract
Mitochondria are both a primary source of reactive oxygen species (ROS) and a sensitive target of oxidative stress; damage to mitochondria can result in bioenergetic dysfunction and both necrotic and apoptotic cell death. These relationships between mitochondria and cell death are particularly strong in both acute and chronic neurodegenerative disorders. ROS levels are affected by both the production of superoxide and its toxic metabolites and by antioxidant defense mechanisms. Mitochondrial antioxidant activities include superoxide dismutase 2, glutathione peroxidase and reductase, and intramitochondrial glutathione. When intracellular conditions disrupt the homeostatic balance between ROS production and detoxification, a net increase in ROS and an oxidized shift in cellular redox state ensues. Cells respond to this imbalance by increasing the expression of genes that code for proteins that protect against oxidative stress and inhibit cytotoxic oxidation of proteins, DNA, and lipids. If, however, the genomic response to mitochondrial oxidative stress is insufficient to maintain homeostasis, mitochondrial bioenergetic dysfunction and release of pro-apoptotic mitochondrial proteins into the cytosol initiate a variety of cell death pathways, ultimately resulting in potentially lethal damage to vital organs, including the brain. Nuclear factor erythroid 2-related factor 2 (Nrf2) is a translational activating protein that enters the nucleus in response to oxidative stress, resulting in increased expression of numerous cytoprotective genes, including genes coding for mitochondrial and non-mitochondrial antioxidant proteins. Many experimental and some FDA-approved drugs promote this process. Since mitochondria are targets of ROS, it follows that protection against mitochondrial oxidative stress by the Nrf2 pathway of gene expression contributes to neuroprotection by these drugs. This document reviews the evidence that Nrf2 activation increases mitochondrial antioxidants, thereby protecting mitochondria from dysfunction and protecting neural cells from damage and death. New experimental results are provided demonstrating that post-ischemic administration of the Nrf2 activator sulforaphane protects against hippocampal neuronal death and neurologic injury in a clinically-relevant animal model of cardiac arrest and resuscitation.
Collapse
|
48
|
Montecinos-Oliva C, Arrázola MS, Jara C, Tapia-Rojas C, Inestrosa NC. Hormetic-Like Effects of L-Homocysteine on Synaptic Structure, Function, and Aβ Aggregation. Pharmaceuticals (Basel) 2020; 13:ph13020024. [PMID: 32024240 PMCID: PMC7168909 DOI: 10.3390/ph13020024] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2019] [Revised: 01/13/2020] [Accepted: 01/28/2020] [Indexed: 02/06/2023] Open
Abstract
Alzheimer’s Disease (AD) is the primary cause of dementia among the elderly population. Elevated plasma levels of homocysteine (HCy), an amino acid derived from methionine metabolism, are considered a risk factor and biomarker of AD and other types of dementia. An increase in HCy is mostly a consequence of high methionine and/or low vitamin B intake in the diet. Here, we studied the effects of physiological and pathophysiological HCy concentrations on oxidative stress, synaptic protein levels, and synaptic activity in mice hippocampal slices. We also studied the in vitro effects of HCy on the aggregation kinetics of Aβ40. We found that physiological cerebrospinal concentrations of HCy (0.5 µM) induce an increase in synaptic proteins, whereas higher doses of HCy (30–100 µM) decrease their levels, thereby increasing oxidative stress and causing excitatory transmission hyperactivity, which are all considered to be neurotoxic effects. We also observed that normal cerebrospinal concentrations of HCy slow the aggregation kinetic of Aβ40, whereas high concentrations accelerate its aggregation. Finally, we studied the effects of HCy and HCy + Aβ42 over long-term potentiation. Altogether, by studying an ample range of effects under different HCy concentrations, we report, for the first time, that HCy can exert beneficial or toxic effects over neurons, evidencing a hormetic-like effect. Therefore, we further encourage the use of HCy as a biomarker and modifiable risk factor with therapeutic use against AD and other types of dementia.
Collapse
Affiliation(s)
- Carla Montecinos-Oliva
- Centro de Envejecimiento y Regeneración (CARE); Departamento de Biología Celular y Molecular; Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago 8331150, Chile
| | - Macarena S Arrázola
- Centro de Envejecimiento y Regeneración (CARE); Departamento de Biología Celular y Molecular; Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago 8331150, Chile
- Center for Integrative Biology, Faculty of Sciences, Universidad Mayor de Chile, Santiago 8580745, Chile
| | - Claudia Jara
- Laboratory of Neurobiology of Aging, Centro de Biología Celular y Biomedicina (CEBICEM), Facultad de Medicina y Ciencia, Universidad San Sebastián, Santiago 7510156, Chile
| | - Cheril Tapia-Rojas
- Laboratory of Neurobiology of Aging, Centro de Biología Celular y Biomedicina (CEBICEM), Facultad de Medicina y Ciencia, Universidad San Sebastián, Santiago 7510156, Chile
| | - Nibaldo C Inestrosa
- Centro de Envejecimiento y Regeneración (CARE); Departamento de Biología Celular y Molecular; Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago 8331150, Chile
- Centro de Excelencia en Biomedicina de Magallanes (CEBIMA), Universidad de Magallanes, Punta Arenas 6213515, Chile
| |
Collapse
|
49
|
Nielsen BE, Bermudez I, Bouzat C. Flavonoids as positive allosteric modulators of α7 nicotinic receptors. Neuropharmacology 2019; 160:107794. [PMID: 31560909 DOI: 10.1016/j.neuropharm.2019.107794] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Revised: 08/09/2019] [Accepted: 09/23/2019] [Indexed: 12/11/2022]
Abstract
The use of positive allosteric modulators (PAM) of α7 nicotinic receptors is a promising therapy for neurodegenerative, inflammatory and cognitive disorders. Flavonoids are polyphenolic compounds showing neuroprotective, anti-inflammatory and pro-cognitive actions. Besides their well-known antioxidant activity, flavonoids trigger intracellular pathways and interact with receptors, including α7. To reveal how the beneficial actions of flavonoids are linked to α7 function, we evaluated the effects of three representative flavonoids -genistein, quercetin and the neoflavonoid 5,7-dihydroxy-4-phenylcoumarin- on whole-cell and single-channel currents. All flavonoids increase the maximal currents elicited by acetylcholine with minimal effects on desensitization and do not reactivate desensitized receptors, a behaviour consistent with type I PAMs. At the single-channel level, they increase the duration of the open state and produce activation in long-duration episodes with a rank order of efficacy of genistein > quercetin ≥ neoflavonoid. By using mutant and chimeric α7 receptors, we demonstrated that flavonoids share transmembrane structural determinants with other PAMs. The α7-PAM activity of flavonoids results in decreased cell levels of reactive oxygen species. Thus, allosteric potentiation of α7 may be an additional mechanism underlying neuroprotective actions of flavonoids, which may be used as scaffolds for designing new therapeutic agents.
Collapse
Affiliation(s)
- Beatriz Elizabeth Nielsen
- Instituto de Investigaciones Bioquímicas de Bahía Blanca, Departamento de Biología, Bioquímica y Farmacia, Universidad Nacional del Sur-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Bahía Blanca 8000, Argentina
| | - Isabel Bermudez
- Department of Medical and Biological Sciences, Oxford Brookes University, Oxford, OX3 0BP, United Kingdom
| | - Cecilia Bouzat
- Instituto de Investigaciones Bioquímicas de Bahía Blanca, Departamento de Biología, Bioquímica y Farmacia, Universidad Nacional del Sur-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Bahía Blanca 8000, Argentina.
| |
Collapse
|
50
|
Fontecha‐Barriuso M, Martín‐Sánchez D, Martinez‐Moreno JM, Carrasco S, Ruiz‐Andrés O, Monsalve M, Sanchez‐Ramos C, Gómez MJ, Ruiz‐Ortega M, Sánchez‐Niño MD, Cannata‐Ortiz P, Cabello R, Gonzalez‐Enguita C, Ortiz A, Sanz AB. PGC‐1α deficiency causes spontaneous kidney inflammation and increases the severity of nephrotoxic AKI. J Pathol 2019; 249:65-78. [DOI: 10.1002/path.5282] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Revised: 03/15/2019] [Accepted: 04/10/2019] [Indexed: 01/05/2023]
Affiliation(s)
- Miguel Fontecha‐Barriuso
- Deparment of Nephrology, Research Institute‐Fundacion Jimenez Diaz Autonoma University Madrid Spain
- REDINREN Madrid Spain
| | - Diego Martín‐Sánchez
- Deparment of Nephrology, Research Institute‐Fundacion Jimenez Diaz Autonoma University Madrid Spain
- REDINREN Madrid Spain
| | - Julio M Martinez‐Moreno
- Deparment of Nephrology, Research Institute‐Fundacion Jimenez Diaz Autonoma University Madrid Spain
| | - Susana Carrasco
- Deparment of Nephrology, Research Institute‐Fundacion Jimenez Diaz Autonoma University Madrid Spain
- REDINREN Madrid Spain
| | - Olga Ruiz‐Andrés
- Deparment of Nephrology, Research Institute‐Fundacion Jimenez Diaz Autonoma University Madrid Spain
- REDINREN Madrid Spain
| | - Maria Monsalve
- Department of Metabolism and Cell Signaling, Instituto de Investigaciones Biomédicas ‘Alberto Sols’ (CSIC‐UAM) Madrid Spain
| | - Cristina Sanchez‐Ramos
- Department of Metabolism and Cell Signaling, Instituto de Investigaciones Biomédicas ‘Alberto Sols’ (CSIC‐UAM) Madrid Spain
| | - Manuel J Gómez
- Bioinformatics Unit, Centro Nacional de Investigaciones Cardiovasculares (CNIC) Madrid Spain
| | - Marta Ruiz‐Ortega
- Deparment of Nephrology, Research Institute‐Fundacion Jimenez Diaz Autonoma University Madrid Spain
- REDINREN Madrid Spain
- School of Medicine UAM Madrid Spain
| | - Maria D Sánchez‐Niño
- Deparment of Nephrology, Research Institute‐Fundacion Jimenez Diaz Autonoma University Madrid Spain
- REDINREN Madrid Spain
| | - Pablo Cannata‐Ortiz
- Department of Pathology, Research Institute – Fundación Jiménez Díaz, School of Medicine UAM Madrid Spain
| | - Ramiro Cabello
- Deparment of Nephrology, Research Institute‐Fundacion Jimenez Diaz Autonoma University Madrid Spain
| | - Carmen Gonzalez‐Enguita
- Deparment of Nephrology, Research Institute‐Fundacion Jimenez Diaz Autonoma University Madrid Spain
| | - Alberto Ortiz
- Deparment of Nephrology, Research Institute‐Fundacion Jimenez Diaz Autonoma University Madrid Spain
- REDINREN Madrid Spain
- School of Medicine UAM Madrid Spain
- IRSIN Madrid Spain
| | - Ana B Sanz
- Deparment of Nephrology, Research Institute‐Fundacion Jimenez Diaz Autonoma University Madrid Spain
- REDINREN Madrid Spain
| |
Collapse
|