1
|
Granado N, Mendieta L, Tizabi Y, Murer MG, Moratalla R. Attenuated neurotoxicity after repeated methamphetamine binges linked to dopamine transporter (DAT) decline. Neurobiol Dis 2025; 207:106839. [PMID: 39947439 DOI: 10.1016/j.nbd.2025.106839] [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: 10/15/2024] [Revised: 01/26/2025] [Accepted: 02/10/2025] [Indexed: 02/17/2025] Open
Abstract
Methamphetamine (METH) abuse increases worldwide. In addition to its acute life-threatening effects, METH is toxic for dopaminergic neurons, increasing the risk of developing Parkinson's disease. The impact of repeated METH binge consumption on dopaminergic and neurotoxicity markers remains unclear. We exposed mice to a repeated "binge-like" METH regime, consisting of three doses over a 6 h interval, repeated three times with 14-day intervals. After the first binge, spontaneous motor activity decreased markedly but remained normal after subsequent binges. Following the first binge, we observed a 25 % loss of nigral dopaminergic cell bodies and significant axon terminal damage as assessed through striatal silver staining, with minimal further degeneration after additional binges. Dopaminergic markers were substantially depleted after the first and second binges, despite partial recovery between binges, dropping to below 20 % of control levels. By one day after the third binge, tyrosine hydroxylase (TH) and vesicular monoamine transporter 2 (VMAT2) stabilized at 50-60 % of control levels, but the dopamine transporter (DAT) remained at only 25 %, showing less recovery. These changes were accompanied by an evolving neuroinflammation pattern, with a transient microglial surge after the first binge and persistent astroglial and temperature responses. Overall, our findings indicate partial recovery of dopaminergic markers and the development of tolerance to METH neurotoxicity. The robust reduction of DAT after the first binge may contribute to this tolerance to subsequence binges by limiting METH entry into neurons thereby mitigating its neurotoxic effects.
Collapse
Affiliation(s)
- Noelia Granado
- Instituto Cajal, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Liliana Mendieta
- Instituto Cajal, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Yousef Tizabi
- Department of Pharmacology, Howard University College of Medicine, Washington, DC, USA
| | - Mario Gustavo Murer
- Universidad de Buenos Aires, Facultad de Medicina, and CONICET, Instituto de Fisiología y Biofísica (IFIBIO), Buenos Aires, Argentina
| | - Rosario Moratalla
- Instituto Cajal, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain.
| |
Collapse
|
2
|
Lin X, Li Q, Pu M, Dong H, Zhang Q. Significance of nicotine and nicotinic acetylcholine receptors in Parkinson's disease. Front Aging Neurosci 2025; 17:1535310. [PMID: 40191787 PMCID: PMC11968747 DOI: 10.3389/fnagi.2025.1535310] [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: 11/27/2024] [Accepted: 03/06/2025] [Indexed: 04/09/2025] Open
Abstract
Parkinson's disease (PD) is a multifaceted neurodegenerative disorder characterized by the degeneration of dopaminergic neurons in the substantia nigra and the aggregation of α-synuclein. According to epidemiological data, PD is the second most prevalent neurodegenerative disorder after Alzheimer's disease (AD) and has emerged as a significant global health concern. This review examines the intricate pathological mechanisms and high-risk factors associated with PD, and discusses the challenges in its clinical diagnosis and treatment. We elucidate the relationship between smoking and the reduced risk of PD, highlighting the potential neuroprotective effects of nicotine present in tobacco. The interaction between nicotine and nicotinic acetylcholine receptors (nAChRs) is analyzed in detail, emphasizing their neuroprotective capabilities and underlying molecular mechanisms. Furthermore, we analyze the structural and functional diversity of nAChRs and their roles in the pathological progression of PD. Our review aims to elucidate the complex interplay of genetic, environmental, and biochemical factors in PD and to propose future research directions that may facilitate therapeutic development.
Collapse
Affiliation(s)
- Xia Lin
- Department of Neurology, First People's Hospital of Tianshui, Tianshui, Gansu, China
| | - Qian Li
- First Clinical Medical School, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
| | - Min Pu
- First Clinical Medical School, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
| | - Hao Dong
- First Clinical Medical School, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
| | - Qinghua Zhang
- Department of Neurology, First People's Hospital of Tianshui, Tianshui, Gansu, China
| |
Collapse
|
3
|
Xu FF, Liu Z, Fang XX, Cao BB, Huang Y, Peng YP, Qiu YH. Microglia-derived exosomal ciRS-7 mediates IL-17A effect of promoting neurodegeneration via miR-7 and SNCA targets in an experimental Parkinson's disease. Int Immunopharmacol 2025; 148:114089. [PMID: 39832458 DOI: 10.1016/j.intimp.2025.114089] [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: 10/16/2024] [Revised: 12/19/2024] [Accepted: 01/11/2025] [Indexed: 01/22/2025]
Abstract
Parkinson' s disease (PD) is a chronic neurodegenerative disorder characterized by progressive loss of dopaminergic neurons in the substantia nigra (SN). Our research has demonstrated that the levels of interleukin (IL)-17A are elevated in the SN of rodent models of PD, and that IL-17A accelerates neurodegeneration in PD depending on microglial activation. Furthermore, existing studies indicate that exosomes released by activated microglia may play a significant role as mediators of neurodegeneration in PD. Herein, we demonstrated that BV-2-derived exosomes were taken up by ventral mesencephalic (VM) dopaminergic neurons, and mediated IL-17A effect of promoting dopaminergic neuronal injury. IL-17A-treated BV-2-derived exosomes altered neuronal miR-7 and SNCA expression and promoted dopaminergic neuronal injury in vitro. Inhibiting BV-2 exosome formation and secretion by GW4869 alleviated dopaminergic neuronal injury. Silencing ciRS-7 in BV-2 altered neuronal miR-7 and SNCA expression and mitigated dopaminergic neuronal injury. Overexpression of ciRS-7 in VM neurons altered neuronal miR-7 and SNCA expression and promoted dopaminergic neuronal injury. Injection with exosomes derived from IL-17A-treated BV-2 altered ciRS-7, miR-7 and SNCA expression in SN in MPTP-intoxicated mice and promoted nigrostriatal dopaminergic neurodegeneration and motor impairment. However, injection with exosomes derived from IL-17A and ciRS-7-shRNA treated BV-2 attenuates the manifestations mentioned above. These findings suggest that microglia-derived exosomal ciRS-7 mediates IL-17A effect of promoting neurodegeneration via miR-7 and SNCA targets and may provide a new paradigm to study the pathology of PD.
Collapse
Affiliation(s)
- Fen-Fen Xu
- Department of Physiology, School of Medicine, and Co-innovation Center of Neuroregeneration, Nantong University, 19 Qixiu Road, Nantong 226001 China
| | - Zhan Liu
- Department of Physiology, School of Medicine, and Co-innovation Center of Neuroregeneration, Nantong University, 19 Qixiu Road, Nantong 226001 China
| | - Xiao-Xia Fang
- Department of Physiology, School of Medicine, and Co-innovation Center of Neuroregeneration, Nantong University, 19 Qixiu Road, Nantong 226001 China
| | - Bei-Bei Cao
- Department of Physiology, School of Medicine, and Co-innovation Center of Neuroregeneration, Nantong University, 19 Qixiu Road, Nantong 226001 China
| | - Yan Huang
- Department of Physiology, School of Medicine, and Co-innovation Center of Neuroregeneration, Nantong University, 19 Qixiu Road, Nantong 226001 China
| | - Yu-Ping Peng
- Department of Physiology, School of Medicine, and Co-innovation Center of Neuroregeneration, Nantong University, 19 Qixiu Road, Nantong 226001 China.
| | - Yi-Hua Qiu
- Department of Physiology, School of Medicine, and Co-innovation Center of Neuroregeneration, Nantong University, 19 Qixiu Road, Nantong 226001 China.
| |
Collapse
|
4
|
Abhilash PL, Bharti U, Rashmi SK, Philip M, Raju TR, Kutty BM, Sagar BKC, Alladi PA. Aging and MPTP Sensitivity Depend on Molecular and Ultrastructural Signatures of Astroglia and Microglia in Mice Substantia Nigra. Cell Mol Neurobiol 2025; 45:13. [PMID: 39833644 PMCID: PMC11753320 DOI: 10.1007/s10571-024-01528-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2024] [Accepted: 12/24/2024] [Indexed: 01/22/2025]
Abstract
Both astroglia and microglia show region-specific distribution in CNS and often maladapt to age-associated alterations within their niche. Studies on autopsied substantia nigra (SN) of Parkinson's disease (PD) patients and experimental models propose gliosis as a trigger for neuronal loss. Epidemiological studies propose an ethnic bias in PD prevalence, since Caucasians are more susceptible than non-whites. Similarly, different mice strains are variably sensitive to MPTP. We had earlier likened divergent MPTP sensitivity of C57BL/6 J and CD-1 mice with differential susceptibility to PD, based on the numbers of SN neurons. We examined whether the variability was incumbent to inter-strain differences in glial features of male C57BL/6 J and CD-1 mice. Stereological counts showed relatively more microglia and fewer astrocytes in the SN of normal C57BL/6 J mice, suggesting persistence of an immune-vigilant state. MPTP-induced microgliosis and astrogliosis in both strains suggest their involvement in pathogenesis. ELISA of pro-inflammatory cytokines in the ventral-midbrain revealed augmentation of TNF-α and IL-6 at middle age in both strains that reduced at old age, suggesting middle age as a critical, inflamm-aging-associated time point. TNF-α levels were high in C57BL/6 J, through aging and post-MPTP, while IL-6 and IL-1β were upregulated at old age. CD-1 had higher levels of anti-inflammatory cytokine TGF-β. MPTP challenge caused upregulation of enzymes MAO-A, MAO-B, and iNOS in both strains. Post-MPTP enhancement in fractalkine and hemeoxygenase-1 may be neuron-associated compensatory signals. Ultrastructural observations of elongated astroglial/microglial mitochondria vis-à-vis the shrunken ones in neurons suggest a scale-up of their functions with neurotoxic consequences. Thus, astroglia and microglia may modulate aging and PD susceptibility.
Collapse
Affiliation(s)
- P L Abhilash
- Department of Neurophysiology, National Institute of Mental Health and Neurosciences, Hosur Road, Bengaluru, 560029, India
| | - Upasna Bharti
- Department of Clinical Psychopharmacology and Neurotoxicology, National Institute of Mental Health and Neurosciences, Bengaluru, 560029, India
| | - Santhosh Kumar Rashmi
- Department of Neuropathology, National Institute of Mental Health and Neurosciences, Bengaluru, 560029, India
| | - Mariamma Philip
- Department of Biostatistics, National Institute of Mental Health and Neurosciences, Bengaluru, 560029, India
| | - T R Raju
- Department of Neurophysiology, National Institute of Mental Health and Neurosciences, Hosur Road, Bengaluru, 560029, India
| | - Bindu M Kutty
- Department of Neurophysiology, National Institute of Mental Health and Neurosciences, Hosur Road, Bengaluru, 560029, India
| | - B K Chandrasekhar Sagar
- Department of Biostatistics, National Institute of Mental Health and Neurosciences, Bengaluru, 560029, India
| | - Phalguni Anand Alladi
- Department of Neurophysiology, National Institute of Mental Health and Neurosciences, Hosur Road, Bengaluru, 560029, India.
- Department of Clinical Psychopharmacology and Neurotoxicology, National Institute of Mental Health and Neurosciences, Bengaluru, 560029, India.
- Department of Clinical Psychopharmacology and Neurotoxicology, National Institute of Mental Health and Neuro Sciences, Hosur Road, Bangalore, India.
| |
Collapse
|
5
|
Kuo PC, Weng WT, Scofield BA, Paraiso HC, Yu ICI, Yen JHJ. Ischemia-induced endogenous Nrf2/HO-1 axis activation modulates microglial polarization and restrains ischemic brain injury. Front Immunol 2024; 15:1440592. [PMID: 39469715 PMCID: PMC11513276 DOI: 10.3389/fimmu.2024.1440592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 09/18/2024] [Indexed: 10/30/2024] Open
Abstract
Cerebral ischemic stroke accounts for more than 80% of all stroke cases. During cerebral ischemia, reactive oxygen species produced in the ischemic brain induce oxidative stress and inflammatory responses. Nrf2 is a transcription factor responsible for regulating cellular redox balance through the induction of protective antioxidant and phase II detoxification responses. Although the induction of endogenous Nrf2/HO-1 axis activation has been observed in the ischemic brain, whether ischemia-induced endogenous Nrf2/HO-1 axis activation plays a role in modulating microglia (MG) phenotypes and restraining ischemic brain injury is not characterized and requires further exploration. To investigate that, we generated mice with Nrf2 knockdown specifically in MG to rigorously assess the role of endogenous Nrf2 activation in ischemic brain injury after stroke. Our results showed that MG-specific Nrf2 knockdown exacerbated ischemic brain injury after stroke. We found that Nrf2 knockdown altered MG phenotypes after stroke, in which increased frequency of inflammatory MG and decreased frequency of anti-inflammatory MG were detected in the ischemic brain. Moreover, we identified attenuated Nrf2/HO-1 axis activation led to increased CD68/IL-1β and suppressed CD206 expression in MG, resulting in aggravated inflammatory MG in MG-specific Nrf2 knockdown mice after stroke. Intriguingly, using type II diabetic preclinical models, we revealed that diabetic mice exhibited attenuated Nrf2/HO-1 axis activation in MG and exacerbated ischemic brain injury after stroke that phenocopy mice with MG-specific Nrf2 knockdown. Finally, the induction of exogenous Nrf2/HO-1 axis activation in MG through pharmacological approaches ameliorated ischemic brain injury in diabetic mice. In conclusion, our findings provide cellular and molecular insights demonstrating ischemia-induced endogenous Nrf2/HO-1 axis activation modulates MG phenotypes and restrains ischemic brain injury. These results further strengthen the therapeutic potential of targeting Nrf2/HO-1 axis in MG for the treatment of ischemic stroke and diabetic stroke.
Collapse
Affiliation(s)
- Ping-Chang Kuo
- Department of Microbiology and Immunology, Indiana University School of Medicine, Fort Wayne, IN, United States
| | - Wen-Tsan Weng
- Department of Microbiology and Immunology, Indiana University School of Medicine, Fort Wayne, IN, United States
| | - Barbara A. Scofield
- Department of Microbiology and Immunology, Indiana University School of Medicine, Fort Wayne, IN, United States
| | - Hallel C. Paraiso
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Fort Wayne, IN, United States
| | - I-Chen Ivorine Yu
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Fort Wayne, IN, United States
| | - Jui-Hung Jimmy Yen
- Department of Microbiology and Immunology, Indiana University School of Medicine, Fort Wayne, IN, United States
| |
Collapse
|
6
|
Li C, Wu J, Dong Q, Ma J, Gao H, Liu G, Chen Y, Ning J, Lv X, Zhang M, Zhong H, Zheng T, Liu Y, Peng Y, Qu Y, Gao X, Shi H, Sun C, Hui Y. The crosstalk between oxidative stress and DNA damage induces neural stem cell senescence by HO-1/PARP1 non-canonical pathway. Free Radic Biol Med 2024; 223:443-457. [PMID: 39047850 DOI: 10.1016/j.freeradbiomed.2024.07.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 07/19/2024] [Accepted: 07/21/2024] [Indexed: 07/27/2024]
Abstract
Neural stem cells play a crucial role in maintaining brain homeostasis. Neural stem cells senescence can lead to the decline of nerve repair and regeneration, causing brain aging and neurodegenerative diseases. However, the mechanism underlying neural stem cells senescence remains poorly understood. In this study, we report a novel HO-1/PARP1 non-canonical pathway highlighting how oxidative stress triggers the DNA damage response, ultimately leading to premature cellular senescence in neural stem cells. HO-1 acts as a sensor for oxidative stress, while PARP1 functions as a sensor for DNA damage. The simultaneous expression and molecular interaction of these two sensors can initiate a crosstalk of oxidative stress and DNA damage response processes, leading to the vicious cycle. The persistent activation of this pathway contributes to the senescence of neural stem cells, which in turn plays a crucial role in the progression of neurodegenerative diseases. Consequently, targeting this novel signaling pathway holds promise for the development of innovative therapeutic strategies and targets aimed at mitigating neural stem cells senescence-related disorders.
Collapse
Affiliation(s)
- Cheng Li
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, PR China
| | - Jiajia Wu
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, PR China
| | - Qi Dong
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, PR China
| | - Jiajia Ma
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, PR China
| | - Huiqun Gao
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, PR China
| | - Guiyan Liu
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, PR China
| | - You Chen
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, PR China
| | - Jiaqi Ning
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, PR China
| | - Xuebing Lv
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, PR China
| | - Mingyang Zhang
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, PR China
| | - Haojie Zhong
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, PR China
| | - Tianhu Zheng
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, PR China
| | - Yuanli Liu
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, PR China
| | - Yahui Peng
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, PR China
| | - Yilin Qu
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, PR China
| | - Xu Gao
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, PR China; Basic Medical Institute of Heilongjiang Medical Science Academy, PR China; Translational Medicine Center of Northern China, PR China
| | - Huaizhang Shi
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Harbin, PR China.
| | - Chongran Sun
- Department of Neurosurgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, PR China.
| | - Yang Hui
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, PR China; Basic Medical Institute of Heilongjiang Medical Science Academy, PR China; Translational Medicine Center of Northern China, PR China.
| |
Collapse
|
7
|
Rose KN, Zorlu M, Fassini A, Lee H, Cai W, Xue X, Lin S, Kivisakk P, Schwarzschild MA, Chen X, Gomperts SN. Neuroprotection of low dose carbon monoxide in Parkinson's disease models commensurate with the reduced risk of Parkinson's among smokers. NPJ Parkinsons Dis 2024; 10:152. [PMID: 39174550 PMCID: PMC11341721 DOI: 10.1038/s41531-024-00763-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Accepted: 07/24/2024] [Indexed: 08/24/2024] Open
Abstract
Paradoxically, cigarette smoking is associated with a reduced risk of Parkinson's Disease (PD). This led us to hypothesize that carbon monoxide (CO) levels, which are constitutively but modestly elevated in smokers, might contribute to neuroprotection. Using rodent models of PD based on α-synuclein (αSyn) accumulation and oxidative stress, we show that low-dose CO mitigates neurodegeneration and reduces αSyn pathology. Oral CO administration activated signaling cascades mediated by heme oxygenase-1 (HO-1), which have been implicated in limiting oxidative stress, and in promoting αSyn degradation, thereby conferring neuroprotection. Consistent with the neuroprotective effect of smoking, HO-1 levels in cerebrospinal fluid were higher in human smokers compared to nonsmokers. Moreover, in PD brain samples, HO-1 levels were higher in neurons without αSyn pathology. Thus, CO in rodent PD models reduces pathology and increases oxidative stress responses, phenocopying possible protective effects of smoking evident in PD patients. These data highlight the potential for low-dose CO-modulated pathways to slow symptom onset and limit pathology in PD patients.
Collapse
Affiliation(s)
- K N Rose
- Department of Neurology, Massachusetts General Hospital, Boston, MA, 02114, USA
- Harvard Medical School, Boston, MA, 02115, USA
| | - M Zorlu
- Department of Neurology, Massachusetts General Hospital, Boston, MA, 02114, USA
| | - A Fassini
- Department of Neurology, Massachusetts General Hospital, Boston, MA, 02114, USA
- Harvard Medical School, Boston, MA, 02115, USA
| | - H Lee
- Department of Neurology, Massachusetts General Hospital, Boston, MA, 02114, USA
| | - W Cai
- Department of Neurology, Massachusetts General Hospital, Boston, MA, 02114, USA
| | - X Xue
- Department of Neurology, Massachusetts General Hospital, Boston, MA, 02114, USA
| | - S Lin
- Department of Neurology, Massachusetts General Hospital, Boston, MA, 02114, USA
| | - P Kivisakk
- Department of Neurology, Massachusetts General Hospital, Boston, MA, 02114, USA
- Harvard Medical School, Boston, MA, 02115, USA
| | - M A Schwarzschild
- Department of Neurology, Massachusetts General Hospital, Boston, MA, 02114, USA
- Harvard Medical School, Boston, MA, 02115, USA
| | - X Chen
- Department of Neurology, Massachusetts General Hospital, Boston, MA, 02114, USA
- Harvard Medical School, Boston, MA, 02115, USA
| | - S N Gomperts
- Department of Neurology, Massachusetts General Hospital, Boston, MA, 02114, USA.
- Harvard Medical School, Boston, MA, 02115, USA.
| |
Collapse
|
8
|
Rose KN, Zorlu M, Xue X, Fassini A, Cai W, Lin S, Webb P, Schwarzschild MA, Chen X, Gomperts SN. Neuroprotection of low dose carbon monoxide in Parkinson's disease models commensurate with the reduced risk of Parkinson's among smokers. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.05.27.542565. [PMID: 37398030 PMCID: PMC10312428 DOI: 10.1101/2023.05.27.542565] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
Paradoxically, cigarette smoking is associated with a reduced risk of Parkinson's disease (PD). This led us to hypothesize that carbon monoxide (CO) levels, which are constitutively but modestly elevated in smokers, might contribute to neuroprotection. Using rodent models of PD based on α-synuclein (αSyn) accumulation and oxidative stress, we show that low-dose CO mitigates neurodegeneration and reduces αSyn pathology. Oral CO administration activated signaling cascades mediated by heme oxygenase-1 (HO-1), which have been implicated in limiting oxidative stress, and in promoting αSyn degradation, thereby conferring neuroprotection. Consistent with a neuroprotective effect of smoking, HO-1 levels in cerebrospinal fluid were higher in human smokers compared to nonsmokers. Moreover, in PD brain samples, HO-1 levels were higher in neurons without αSyn pathology. Thus, CO in rodent PD models reduces pathology and increases oxidative stress responses, phenocopying possible protective effects of smoking evident in PD patients. These data highlight the potential for low-dose CO modulated pathways to slow symptom onset and limit pathology in PD patients.
Collapse
|
9
|
Rose KN, Schwarzschild MA, Gomperts SN. Clearing the Smoke: What Protects Smokers from Parkinson's Disease? Mov Disord 2024; 39:267-272. [PMID: 38226487 PMCID: PMC10923097 DOI: 10.1002/mds.29707] [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: 08/23/2023] [Revised: 11/22/2023] [Accepted: 12/15/2023] [Indexed: 01/17/2024] Open
Abstract
The link between smoking and a lower risk of Parkinson's disease (PD) is one of the strongest environmental or lifestyle associations in neuroepidemiology. Growing evidence supports the hypothesis that the association is based on a neuroprotective effect of smoking on PD, despite the plausible alternative that smoking serves as a marker for a proximal protective influence without itself conferring benefit. But how smoking could protect against neurodegeneration in PD is not well understood. Of several candidate molecules and mechanisms that have been nominated, nicotine has received the most attention. However, randomized controlled clinical trials of nicotine in PD have failed to demonstrate benefit on motor endpoints, including the NIC-PD study in which recently diagnosed participants were randomly assigned to placebo or nicotine treatment for 1 year. Given these results, the time is right to evaluate the neuroprotective potential of other molecules and biochemical cascades triggered by smoking. Here, we review the evidence supporting smoking's possible protective effect on PD, compounds in tobacco and smoke that might mediate such benefit, and non-causal classes of explanation, including reverse causation and the prospect of shared genetic determinants of smoking and PD resistance. The therapeutic potential of non-nicotine components of smoke is suggested by studies supporting multiple alternative mechanisms ranging from monoamine oxidase inhibitors to gut microbiome disruption to antioxidant response induction by chronic exposure to low levels of carbon monoxide. Rigorous investigation is warranted to evaluate this molecule and others for disease-preventing and disease-modifying activity in PD models and, if warranted, in clinical trials. © 2024 The Authors. Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Kenneth N. Rose
- Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | | | - Stephen N. Gomperts
- Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts, USA
| |
Collapse
|
10
|
Pan S, Hale AT, Lemieux ME, Raval DK, Garton TP, Sadler B, Mahaney KB, Strahle JM. Iron homeostasis and post-hemorrhagic hydrocephalus: a review. Front Neurol 2024; 14:1287559. [PMID: 38283681 PMCID: PMC10811254 DOI: 10.3389/fneur.2023.1287559] [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: 09/02/2023] [Accepted: 11/21/2023] [Indexed: 01/30/2024] Open
Abstract
Iron physiology is regulated by a complex interplay of extracellular transport systems, coordinated transcriptional responses, and iron efflux mechanisms. Dysregulation of iron metabolism can result in defects in myelination, neurotransmitter synthesis, and neuronal maturation. In neonates, germinal matrix-intraventricular hemorrhage (GMH-IVH) causes iron overload as a result of blood breakdown in the ventricles and brain parenchyma which can lead to post-hemorrhagic hydrocephalus (PHH). However, the precise mechanisms by which GMH-IVH results in PHH remain elusive. Understanding the molecular determinants of iron homeostasis in the developing brain may lead to improved therapies. This manuscript reviews the various roles iron has in brain development, characterizes our understanding of iron transport in the developing brain, and describes potential mechanisms by which iron overload may cause PHH and brain injury. We also review novel preclinical treatments for IVH that specifically target iron. Understanding iron handling within the brain and central nervous system may provide a basis for preventative, targeted treatments for iron-mediated pathogenesis of GMH-IVH and PHH.
Collapse
Affiliation(s)
- Shelei Pan
- Department of Neurosurgery, Washington University School of Medicine, Washington University in St. Louis, St. Louis, MO, United States
| | - Andrew T. Hale
- Department of Neurosurgery, University of Alabama at Birmingham School of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Mackenzie E. Lemieux
- Department of Neurosurgery, Washington University School of Medicine, Washington University in St. Louis, St. Louis, MO, United States
| | - Dhvanii K. Raval
- Department of Neurosurgery, Washington University School of Medicine, Washington University in St. Louis, St. Louis, MO, United States
| | - Thomas P. Garton
- Department of Neurology, Johns Hopkins University School of Medicine, Johns Hopkins University, Baltimore, MD, United States
| | - Brooke Sadler
- Department of Pediatrics, Washington University School of Medicine, Washington University in St. Louis, St. Louis, MO, United States
- Department of Hematology and Oncology, Washington University School of Medicine, Washington University in St. Louis, St. Louis, MO, United States
| | - Kelly B. Mahaney
- Department of Neurosurgery, Stanford University School of Medicine, Stanford University, Palo Alto, CA, United States
| | - Jennifer M. Strahle
- Department of Neurosurgery, Washington University School of Medicine, Washington University in St. Louis, St. Louis, MO, United States
- Department of Pediatrics, Washington University School of Medicine, Washington University in St. Louis, St. Louis, MO, United States
- Department of Orthopedic Surgery, Washington University School of Medicine, Washington University in St. Louis, St. Louis, MO, United States
| |
Collapse
|
11
|
Jayanti S, Dalla Verde C, Tiribelli C, Gazzin S. Inflammation, Dopaminergic Brain and Bilirubin. Int J Mol Sci 2023; 24:11478. [PMID: 37511235 PMCID: PMC10380707 DOI: 10.3390/ijms241411478] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 07/07/2023] [Accepted: 07/11/2023] [Indexed: 07/30/2023] Open
Abstract
Dopamine is a well-known neurotransmitter due to its involvement in Parkinson's disease (PD). Dopamine is not only involved in PD but also controls multiple mental and physical activities, such as the pleasure of food, friends and loved ones, music, art, mood, cognition, motivation, fear, affective disorders, addiction, attention deficit disorder, depression, and schizophrenia. Dopaminergic neurons (DOPAn) are susceptible to stressors, and inflammation is a recognized risk for neuronal malfunctioning and cell death in major neurodegenerative diseases. Less is known for non-neurodegenerative conditions. Among the endogenous defenses, bilirubin, a heme metabolite, has been shown to possess important anti-inflammatory activity and, most importantly, to prevent DOPAn demise in an ex vivo model of PD by acting on the tumor necrosis factor-alpha (TNFα). This review summarizes the evidence linking DOPAn, inflammation (when possible, specifically TNFα), and bilirubin as an anti-inflammatory in order to understand what is known, the gaps that need filling, and the hypotheses of anti-inflammatory strategies to preserve dopamine homeostasis with bilirubin included.
Collapse
Affiliation(s)
- Sri Jayanti
- Italian Liver Foundation, Liver Brain Unit “Rita Moretti”, Area Science Park, Bldg. Q, SS 14, Km 163,5, 34149 Trieste, Italy; (S.J.); (C.D.V.); (S.G.)
- Eijkman Research Centre for Molecular Biology, Research Organization for Health, National Research and Innovation Agency, Cibinong 16915, Indonesia
| | - Camilla Dalla Verde
- Italian Liver Foundation, Liver Brain Unit “Rita Moretti”, Area Science Park, Bldg. Q, SS 14, Km 163,5, 34149 Trieste, Italy; (S.J.); (C.D.V.); (S.G.)
| | - Claudio Tiribelli
- Italian Liver Foundation, Liver Brain Unit “Rita Moretti”, Area Science Park, Bldg. Q, SS 14, Km 163,5, 34149 Trieste, Italy; (S.J.); (C.D.V.); (S.G.)
| | - Silvia Gazzin
- Italian Liver Foundation, Liver Brain Unit “Rita Moretti”, Area Science Park, Bldg. Q, SS 14, Km 163,5, 34149 Trieste, Italy; (S.J.); (C.D.V.); (S.G.)
| |
Collapse
|
12
|
Şimşek H, Akaras N, Gür C, Küçükler S, Mehmet Kandemir F. Beneficial effects of Chrysin on Cadmium-induced nephrotoxicity in rats: Modulating the levels of Nrf2/HO-1, RAGE/NLRP3, and Caspase-3/Bax/Bcl-2 signaling pathways. Gene 2023; 875:147502. [PMID: 37224935 DOI: 10.1016/j.gene.2023.147502] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 05/12/2023] [Accepted: 05/18/2023] [Indexed: 05/26/2023]
Abstract
Cadmium (Cd) is a toxic heavy metal that targets the kidney directly in the body. Chrysin (CHR) is a natural flavonoid with many properties such as antioxidant, anti-inflammatory and anti-apoptotic. The current study discloses new evidence as regards of the curative effects of CHR on Cd-induced nephrotoxicity by regulating oxidative stress, apoptosis, autophagy, and inflammation. Cd was administered orally at a dose of 25 mg/kg body weight alone or in combination with orally administered CHR (25 and 50 mg/kg body weight) for 7 days. Biochemical, molecular, and histological methods were used to investigate inflammation, apoptosis, autophagy, and oxidant pathways in renal tissue. Renal function tests were also evaluated. Cd caused an increase in serum toxicity markers, lipid peroxidation and a decrease in the activities of antioxidant enzymes. Nrf-2 triggered inflammatory responses by suppressing HO-1 and NQO1 mRNA transcripts and increasing NF-κB, TNF-α, IL-1β and iNOS mRNA transcripts. Cd caused inflammasome by increasing RAGE and NLRP3 mRNA transcripts. In addition, Cd application caused apoptosis by increasing Bax, Apaf-1 and Caspase-3 mRNA transcripts and decreasing Bcl-2 mRNA transcript level. It caused autophagy by increasing the activity of Beclin-1 level. CHR treatment had the opposite effect on all these values and reduced the damage caused by all these signal pathways. Overall, the data of this study indicate that renal damage associated with Cd toxicity could be ameliorated by CHR administration.
Collapse
Affiliation(s)
- Hasan Şimşek
- Department of Physiology, Faculty of Medicine, Aksaray University, Aksaray, TÜRKİYE.
| | - Nurhan Akaras
- Department of Histology and Embryology, Faculty of Medicine, Aksaray University, Aksaray, TÜRKİYE
| | - Cihan Gür
- Department of Veterinary Biochemistry, Faculty of Veterinary, Atatürk University, Erzurum, TÜRKİYE
| | - Sefa Küçükler
- Department of Veterinary Biochemistry, Faculty of Veterinary, Atatürk University, Erzurum, TÜRKİYE
| | - Fatih Mehmet Kandemir
- Department of Medical Biochemistry, Faculty of Medicine, Aksaray University, Aksaray, TÜRKİYE
| |
Collapse
|
13
|
Jayanti S, Moretti R, Tiribelli C, Gazzin S. Bilirubin Prevents the TH + Dopaminergic Neuron Loss in a Parkinson's Disease Model by Acting on TNF-α. Int J Mol Sci 2022; 23:14276. [PMID: 36430754 PMCID: PMC9693357 DOI: 10.3390/ijms232214276] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 11/09/2022] [Accepted: 11/16/2022] [Indexed: 11/19/2022] Open
Abstract
Parkinson's disease (PD), the fastest-growing movement disorder, is still challenged by the unavailability of disease-modifying therapy. Mildly elevated levels of unconjugated bilirubin (UCB, PubChem CID 5280352) have been shown to be protective against several extra-CNS diseases, and the effect is attributed to its well-known anti-oxidant and anti-inflammatory capability. We explored the neuroprotective effect of low concentrations of UCB (from 0.5 to 4 µM) in our PD model based on organotypic brain cultures of substantia nigra (OBCs-SN) challenged with a low dose of rotenone (Rot). UCB at 0.5 and 1 µM fully protects against the loss of TH+ (dopaminergic) neurons (DOPAn). The alteration in oxidative stress is involved in TH+ positive neuron demise induced by Rot, but is not the key player in UCB-conferred protection. On the contrary, inflammation, specifically tumor necrosis factor alpha (TNF-α), was found to be the key to UCB protection against DOPAn sufferance. Further work will be needed to introduce the use of UCB into clinical settings, but determining that TNF-α plays a key role in PD may be crucial in designing therapeutic options.
Collapse
Affiliation(s)
- Sri Jayanti
- The Liver-Brain Unit “Rita-Moretti”, Fondazione Italiana Fegato-Onlus, Bldg. Q, AREA Science Park, ss14, Km 163.5, Basovizza, 34149 Trieste, Italy
- Faculty of Medicine, University of Hasanuddin, Makassar 90245, Indonesia
- Molecular Biomedicine Ph.D. Program, Department of Life Sciences, University of Trieste, 34127 Trieste, Italy
| | - Rita Moretti
- Neurology Clinic, Department of Medical, Surgical and Health Sciences, University of Trieste, 34139 Trieste, Italy
| | - Claudio Tiribelli
- The Liver-Brain Unit “Rita-Moretti”, Fondazione Italiana Fegato-Onlus, Bldg. Q, AREA Science Park, ss14, Km 163.5, Basovizza, 34149 Trieste, Italy
| | - Silvia Gazzin
- The Liver-Brain Unit “Rita-Moretti”, Fondazione Italiana Fegato-Onlus, Bldg. Q, AREA Science Park, ss14, Km 163.5, Basovizza, 34149 Trieste, Italy
| |
Collapse
|
14
|
Huang SS, Su HH, Chien SY, Chung HY, Luo ST, Chu YT, Wang YH, MacDonald IJ, Lee HH, Chen YH. Activation of peripheral TRPM8 mitigates ischemic stroke by topically applied menthol. J Neuroinflammation 2022; 19:192. [PMID: 35897101 PMCID: PMC9327358 DOI: 10.1186/s12974-022-02553-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Accepted: 07/08/2022] [Indexed: 11/12/2022] Open
Abstract
Background No reports exist as to neuroprotective effects associated with topical activation of transient receptor potential melastatin 8 (TRPM8), a noted cold receptor. In the present study, we identified whether activating peripheral TRPM8 can be an adjuvant therapy for ischemic stroke.
Methods Menthol, an agonist of TRPM8, was applied orally or topically to all paws or back of the mouse after middle cerebral artery occlusion (MCAO). We used Trpm8 gene knockout (Trpm8−/−) mice or TRPM8 antagonist and lidocaine to validate the roles of TRPM8 and peripheral nerve conduction in menthol against ischemic stroke. Results Application of menthol 16% to paw derma attenuated infarct volumes and ameliorated sensorimotor deficits in stroke mice induced by MCAO. The benefits of topically applied menthol were associated with reductions in oxidative stress, neuroinflammation and infiltration of monocytes and macrophages in ischemic brains. Antagonizing TRPM8 or Trpm8 knockout dulls the neuroprotective effects of topically application of menthol against MCAO. Immunohistochemistry analyses revealed significantly higher TRPM8 expression in skin tissue samples obtained from the paws compared with skin from the backs, which was reflected by significantly smaller infarct lesion volumes and better sensorimotor function in mice treated with menthol on the paws compared with the back. Blocking conduction of peripheral nerve in the four paws reversed the neuroprotective effects of topical menthol administrated to paws. On the other hand, oral menthol dosing did not assist with recovery from MCAO in our study. Conclusion Our results suggested that activation of peripheral TRPM8 expressed in the derma tissue of limbs with sufficient concentration of menthol is beneficial to stroke recovery. Topical application of menthol on hands and feet could be a novel and simple-to-use therapeutic strategy for stroke patients. Supplementary Information The online version contains supplementary material available at 10.1186/s12974-022-02553-4.
Collapse
Affiliation(s)
- Shiang-Suo Huang
- Department of Pharmacology, Chung Shan Medical University, Taichung, 40201, Taiwan.,School of Medicine, Institute of Medicine, Chung Shan Medical University, Taichung, 40201, Taiwan.,Department of Pharmacy, Chung Shan Medical University Hospital, Taichung, 40201, Taiwan
| | - Hsing-Hui Su
- Department of Pharmacology, Chung Shan Medical University, Taichung, 40201, Taiwan.,Graduate Institute of Acupuncture Science, China Medical University, Taichung, 40402, Taiwan
| | - Szu-Yu Chien
- Graduate Institute of Acupuncture Science, China Medical University, Taichung, 40402, Taiwan
| | - Hsin-Yi Chung
- Graduate Institute of Acupuncture Science, China Medical University, Taichung, 40402, Taiwan
| | - Sih-Ting Luo
- Graduate Institute of Acupuncture Science, China Medical University, Taichung, 40402, Taiwan
| | - Yu-Ting Chu
- Graduate Institute of Acupuncture Science, China Medical University, Taichung, 40402, Taiwan
| | - Yi-Hsin Wang
- Department of Pharmacology, Chung Shan Medical University, Taichung, 40201, Taiwan
| | - Iona J MacDonald
- Graduate Institute of Acupuncture Science, China Medical University, Taichung, 40402, Taiwan
| | - Hsun-Hua Lee
- Department of Neurology, Shuang Ho Hospital, Taipei Medical University, New Taipei City, 23561, Taiwan. .,Dizziness and Balance Disorder Center, Shuang Ho Hospital, Taipei Medical University, New Taipei City, 23561, Taiwan. .,Department of Neurology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, 11031, Taiwan. .,Department of Neurology, Taipei Medical University Hospital, Taipei Medical University, Taipei, 11031, Taiwan.
| | - Yi-Hung Chen
- Graduate Institute of Acupuncture Science, China Medical University, Taichung, 40402, Taiwan. .,Chinese Medicine Research Center, China Medical University, Taichung, 40402, Taiwan. .,Department of Computer Science and Information Engineering, Asia University, Wufeng, Taichung, 41354, Taiwan.
| |
Collapse
|
15
|
Yttrium chloride-induced cytotoxicity and DNA damage response via ROS generation and inhibition of Nrf2/PPARγ pathways in H9c2 cardiomyocytes. Arch Toxicol 2022; 96:767-781. [PMID: 35088107 DOI: 10.1007/s00204-022-03225-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Accepted: 01/12/2022] [Indexed: 12/30/2022]
Abstract
Increasing exploration of rare-earth elements (REEs) has resulted in a high REEs' exposure risk. Owing to their persistence and accumulation of REEs in the environment, their adverse effects have caused widespread concern. However, limited toxicological data are available for the adverse effects of yttrium (Y) and its underlying mechanisms of action. In the present study, H9c2 cardiomyocytes were used in vitro model to investigate the cardiotoxicity of yttrium chloride (YCl3). Results show that YCl3 treatment resulted in reactive oxygen species (ROS) overproduction, decrease in ∆Ψm, and DNA damage. Mechanistically, we detected expression levels of protein in response to cellular DNA damage and antioxidative defense. Results indicated that the phosphorylation of histone H2AX remarkably increased in a dose-dependent manner. At a high YCl3-exposure concentration (120 μM), specific DNA damage sensors ATM/ATR-Chk1/Chk2 were significantly decreased. The protein levels of key antioxidant genes Nrf2/PPARγ/HO-1 were also remarkably inhabited. Additionally, the antioxidant N-acetyl-L-cysteine (NAC) pretreatment promoted the activation of antioxidative defense Nrf2/PPARγ signaling pathways, and prevented the production of cellular ROS, thus protecting the DNA from cleavage. Altogether, our findings suggest that YCl3 can induce DNA damage through causing intracellular ROS overproduction and inhibition of antioxidative defense, leading to cytotoxicity in H9c2 cardiomyocytes.
Collapse
|
16
|
Characterization of the Inducible and Slow-Releasing Hydrogen Sulfide and Persulfide Donor P*: Insights into Hydrogen Sulfide Signaling. Antioxidants (Basel) 2021; 10:antiox10071049. [PMID: 34209813 PMCID: PMC8300844 DOI: 10.3390/antiox10071049] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 06/21/2021] [Accepted: 06/23/2021] [Indexed: 12/04/2022] Open
Abstract
Hydrogen sulfide (H2S) is an important mediator of inflammatory processes. However, controversial findings also exist, and its underlying molecular mechanisms are largely unknown. Recently, the byproducts of H2S, per-/polysulfides, emerged as biological mediators themselves, highlighting the complex chemistry of H2S. In this study, we characterized the biological effects of P*, a slow-releasing H2S and persulfide donor. To differentiate between H2S and polysulfide-derived effects, we decomposed P* into polysulfides. P* was further compared to the commonly used fast-releasing H2S donor sodium hydrogen sulfide (NaHS). The effects on oxidative stress and interleukin-6 (IL-6) expression were assessed in ATDC5 cells using superoxide measurement, qPCR, ELISA, and Western blotting. The findings on IL-6 expression were corroborated in primary chondrocytes from osteoarthritis patients. In ATDC5 cells, P* not only induced the expression of the antioxidant enzyme heme oxygenase-1 via per-/polysulfides, but also induced activation of Akt and p38 MAPK. NaHS and P* significantly impaired menadione-induced superoxide production. P* reduced IL-6 levels in both ATDC5 cells and primary chondrocytes dependent on H2S release. Taken together, P* provides a valuable research tool for the investigation of H2S and per-/polysulfide signaling. These data demonstrate the importance of not only H2S, but also per-/polysulfides as bioactive signaling molecules with potent anti-inflammatory and, in particular, antioxidant properties.
Collapse
|
17
|
Jayanti S, Moretti R, Tiribelli C, Gazzin S. Bilirubin: A Promising Therapy for Parkinson's Disease. Int J Mol Sci 2021; 22:6223. [PMID: 34207581 PMCID: PMC8228391 DOI: 10.3390/ijms22126223] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 05/28/2021] [Accepted: 05/30/2021] [Indexed: 02/07/2023] Open
Abstract
Following the increase in life expectancy, the prevalence of Parkinson's disease (PD) as the most common movement disorder is expected to rise. Despite the incredibly huge efforts in research to find the definitive biomarker, to date, the diagnosis of PD still relies mainly upon clinical symptoms. A wide range of treatments is available for PD, mainly alleviating the clinical symptoms. However, none of these current therapies can stop or even slow down the disease evolution. Hence, disease-modifying treatment is still a paramount unmet medical need. On the other side, bilirubin and its enzymatic machinery and precursors have offered potential benefits by targeting multiple mechanisms in chronic diseases, including PD. Nevertheless, only limited discussions are available in the context of neurological conditions, particularly in PD. Therefore, in this review, we profoundly discuss this topic to understand bilirubin's therapeutical potential in PD.
Collapse
Affiliation(s)
- Sri Jayanti
- Fondazione Italiana Fegato-Onlus, Bldg. Q, AREA Science Park, ss14, Km 163.5, Basovizza, 34149 Trieste, Italy; (C.T.); (S.G.)
- Faculty of Medicine, University of Hasanuddin, Makassar 90245, Indonesia
- Molecular Biomedicine Ph.D. Program, University of Trieste, 34127 Trieste, Italy
| | - Rita Moretti
- Neurology Clinic, Department of Medical, Surgical, and Health Sciences, University of Trieste, 34139 Trieste, Italy;
| | - Claudio Tiribelli
- Fondazione Italiana Fegato-Onlus, Bldg. Q, AREA Science Park, ss14, Km 163.5, Basovizza, 34149 Trieste, Italy; (C.T.); (S.G.)
| | - Silvia Gazzin
- Fondazione Italiana Fegato-Onlus, Bldg. Q, AREA Science Park, ss14, Km 163.5, Basovizza, 34149 Trieste, Italy; (C.T.); (S.G.)
| |
Collapse
|
18
|
Xiong L, Huang J, Gao Y, Gao Y, Wu C, He S, Zou L, Yang D, Han Y, Yuan Q, Zheng Z, Hu G. Sodium arsenite induces spatial learning and memory impairment associated with oxidative stress and activates the Nrf2/PPARγ pathway against oxidative injury in mice hippocampus. Toxicol Res (Camb) 2021; 10:277-283. [PMID: 33884178 PMCID: PMC8045596 DOI: 10.1093/toxres/tfab007] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 11/06/2020] [Accepted: 01/11/2021] [Indexed: 11/14/2022] Open
Abstract
Arsenic (As) is a ubiquitous environmental and industrial toxin with known correlates of oxidative stress and cognitive deficits in the brain. Nuclear factor erythroid 2-related factor 2 (Nrf2) is a transcriptional factor that represents a central cellular antioxidant defense mechanism and transcribes many antioxidant genes. Peroxisome proliferator-activated receptor-gamma (PPARγ) is a well-known nuclear receptor to regulate lipid metabolism in many tissues, and it has been also associated with the control of oxidative stress, neuronal death, neurogenesis and differentiation. The role of Nrf2 and PPARγ in As-induced neurotoxicity is still debated. The present study was designed to investigate the neurobehavioral toxic effect of sub-chronic and middle-dose sodium arsenite exposure in mice hippocampus, as well as the response of Nrf2/PPARγ expression and influence on protein expression levels of their downstream antioxidant genes. Our results showed that mice treated with intraperitoneal injection of sodium arsenite (50 mg/kg body wt.) twice a week for 7 weeks resulted in increased generation of reactive oxygen species and impairment of spatial cognitive function. The present study also found a positive association between Nrf2/PPARγ expression in hippocampus of mice, and activation of antioxidant defenses by the evidently upregulated expression of their downstream genes, including superoxide dismutase, heme oxygenase-1 and glutathione peroxidase-3. Therefore, our findings were helpful for further understanding the role of Nrf2/PPARγ feedback loop in As-induced neurobehavioral toxicity.
Collapse
Affiliation(s)
- Liang Xiong
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases of Ministry of Education, Gannan Medical University, Number 1 Yixueyuan Road, Ganzhou 341000, China
- School of Public Health and Health Management, Gannan Medical University, Number 1 Yixueyuan Road, Ganzhou 341000, Jiangxi, China
| | - Jinyu Huang
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases of Ministry of Education, Gannan Medical University, Number 1 Yixueyuan Road, Ganzhou 341000, China
- Department of Anatomy, School of Basic Medicine Sciences, Gannan Medical University, Number 1 Yixueyuan Road, Ganzhou 341000, Jiangxi, China
| | - Ying Gao
- Department of rehabilitation medicine, School of Rehabilitation, Gannan Medical University, Number 1 Yixueyuan Road, Ganzhou 341000, Jiangxi, China
| | - Yanfang Gao
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases of Ministry of Education, Gannan Medical University, Number 1 Yixueyuan Road, Ganzhou 341000, China
- School of Public Health and Health Management, Gannan Medical University, Number 1 Yixueyuan Road, Ganzhou 341000, Jiangxi, China
| | - Chunmei Wu
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases of Ministry of Education, Gannan Medical University, Number 1 Yixueyuan Road, Ganzhou 341000, China
- School of Public Health and Health Management, Gannan Medical University, Number 1 Yixueyuan Road, Ganzhou 341000, Jiangxi, China
| | - Shengfa He
- School of Public Health and Health Management, Gannan Medical University, Number 1 Yixueyuan Road, Ganzhou 341000, Jiangxi, China
| | - Lijun Zou
- School of Public Health and Health Management, Gannan Medical University, Number 1 Yixueyuan Road, Ganzhou 341000, Jiangxi, China
| | - Dongmei Yang
- School of Public Health and Health Management, Gannan Medical University, Number 1 Yixueyuan Road, Ganzhou 341000, Jiangxi, China
| | - Yuhao Han
- School of Public Health and Health Management, Gannan Medical University, Number 1 Yixueyuan Road, Ganzhou 341000, Jiangxi, China
| | - Qiong Yuan
- School of Public Health and Health Management, Gannan Medical University, Number 1 Yixueyuan Road, Ganzhou 341000, Jiangxi, China
| | - Zuobing Zheng
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases of Ministry of Education, Gannan Medical University, Number 1 Yixueyuan Road, Ganzhou 341000, China
| | - Gonghua Hu
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases of Ministry of Education, Gannan Medical University, Number 1 Yixueyuan Road, Ganzhou 341000, China
- School of Public Health and Health Management, Gannan Medical University, Number 1 Yixueyuan Road, Ganzhou 341000, Jiangxi, China
| |
Collapse
|
19
|
Ayaydin H, Akaltun İ, Koyuncu İ, Çelİk H, Kİrmİt A, Takatak H. High KEAP1, NRF2 and Low HO-1 Serum Levels in Children with Autism. ACTA ACUST UNITED AC 2020; 57:274-279. [PMID: 33354117 DOI: 10.29399/npa.24862] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2019] [Accepted: 05/13/2020] [Indexed: 12/27/2022]
Abstract
Introduction The purpose of our study was to investigate heme oxygenase-1 (HO-1), nuclear factor erythroid-2-related factor 2 (NRF2), and kelch-like ECH-associated protein 1 (KEAP1) levels in children with autism spectrum disorder (ASD) and to reveal their association with the severity of autism. Methods This study measured serum HO-1, KEAP1, and NRF2 levels in 43 patients with ASD (aged 3-12 years) and in 41 age- and gender-matched healthy controls. ASD severity was rated using the Childhood Autism Rating Scale (CARS). HO-1, KEAP1, and NRF2 levels were determined in the biochemistry laboratory using the ELISA technique. Results HO-1 levels were significantly lower in patients aged 3-12 years compared to controls aged 3-12, while KEAP1 and NRF2 levels were significantly higher (p=0.020, p<0.001, and p=0.017, respectively). No correlation was determined between ASD severity on the basis of total CARS scores and HO-1, KEAP1 or NRF2 (p>0.05). Conclusion This study suggests that oxidative stress is higher in children with ASD and that HO-1 levels are insufficient to achieve oxidative balance.
Collapse
Affiliation(s)
- Hamza Ayaydin
- Harran University Faculty of Medicine, Department of Child and Adolescent Psychiatry, Şanlıurfa, Turkey
| | - İsmail Akaltun
- Gaziantep Dr. Ersin Arslan Training and Research Hospital, Department of Child and Adolescent Psychiatry, Gaziantep, Turkey
| | - İsmail Koyuncu
- Harran University Faculty of Medicine, Department of Biochemistry, Şanlıurfa, Turkey
| | - Hakim Çelİk
- Harran University Faculty of Medicine, Department of Physiology, Şanlıurfa, Turkey
| | - Adnan Kİrmİt
- Harran University Faculty of Medicine, Department of Biochemistry, Şanlıurfa, Turkey
| | - Hatice Takatak
- Harran University Faculty of Medicine, Department of Child and Adolescent Psychiatry, Şanlıurfa, Turkey
| |
Collapse
|
20
|
Hahn D, Shin SH, Bae JS. Natural Antioxidant and Anti-Inflammatory Compounds in Foodstuff or Medicinal Herbs Inducing Heme Oxygenase-1 Expression. Antioxidants (Basel) 2020; 9:E1191. [PMID: 33260980 PMCID: PMC7761319 DOI: 10.3390/antiox9121191] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 11/20/2020] [Accepted: 11/24/2020] [Indexed: 02/06/2023] Open
Abstract
Heme oxygenase-1 (HO-1) is an inducible antioxidant enzyme that catalyzes heme group degradation. Decreased level of HO-1 is correlated with disease progression, and HO-1 induction suppresses development of metabolic and neurological disorders. Natural compounds with antioxidant activities have emerged as a rich source of HO-1 inducers with marginal toxicity. Here we discuss the therapeutic role of HO-1 in obesity, hypertension, atherosclerosis, Parkinson's disease and hepatic fibrosis, and present important signaling pathway components that lead to HO-1 expression. We provide an updated, comprehensive list of natural HO-1 inducers in foodstuff and medicinal herbs categorized by their chemical structures. Based on the continued research in HO-1 signaling pathways and rapid development of their natural inducers, HO-1 may serve as a preventive and therapeutic target for metabolic and neurological disorders.
Collapse
Affiliation(s)
- Dongyup Hahn
- School of Food Science and Biotechnology, College of Agriculture and Life Sciences, Kyungpook National University, Daegu 41566, Korea;
- Department of Integrative Biology, Kyungpook National University, Daegu 41566, Korea
| | - Seung Ho Shin
- Department of Food and Nutrition, Institute of Agriculture and Life Science, Gyeongsang National University, Jinju 52828, Korea;
| | - Jong-Sup Bae
- College of Pharmacy, CMRI, Research Institute of Pharmaceutical Sciences, BK21 Plus KNU Multi-Omics based Creative Drug Research Team, Kyungpook National University, Daegu 41566, Korea
| |
Collapse
|
21
|
Ali AM, Kunugi H. Apitherapy for Parkinson's Disease: A Focus on the Effects of Propolis and Royal Jelly. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:1727142. [PMID: 33123309 PMCID: PMC7586183 DOI: 10.1155/2020/1727142] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 08/27/2020] [Accepted: 10/05/2020] [Indexed: 02/08/2023]
Abstract
The vast increase of world's aging populations is associated with increased risk of age-related neurodegenerative diseases such as Parkinson's disease (PD). PD is a widespread disorder characterized by progressive loss of dopaminergic neurons in the substantia nigra, which encompasses a wide range of debilitating motor, emotional, cognitive, and physical symptoms. PD threatens the quality of life of millions of patients and their families. Additionally, public welfare and healthcare systems are burdened with its high cost of care. Available treatments provide only a symptomatic relief and produce a trail of noxious side effects, which increase noncompliance. Hence, researchers have recently focused on the use of nutraceuticals as safe adjunctive treatments of PD to limit its progress and associated damages in affected groups. Propolis is a common product of the beehive, which possesses a large number of therapeutic properties. Royal jelly (RJ) is a bee product that is fed to bee queens during their entire life, and it contributes to their high physical fitness, fertility, and long lifespan. Evidence suggests that propolis and RJ can promote health by preventing the occurrence of age-related debilitating diseases. Therefore, they have been used to treat various serious disorders such as diabetes mellitus, cardiovascular diseases, and cancer. Some evolving studies used these bee products to treat PD in animal models. However, a clear understanding of the collective effect of propolis and RJ as well as their mechanism of action in PD is lacking. This review evaluates the available literature for the effects of propolis and RJ on PD. Whenever possible, it elaborates on the underlying mechanisms through which they function in this disorder and offers insights for fruitful use of bee products in future clinical trials.
Collapse
Affiliation(s)
- Amira Mohammed Ali
- Department of Mental Disorder Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo, Japan
- Department of Psychiatric Nursing and Mental Health, Faculty of Nursing, Alexandria University, Alexandria, Egypt
| | - Hiroshi Kunugi
- Department of Mental Disorder Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo, Japan
- Department of Psychiatry, Teikyo University School of Medicine, Tokyo, Japan
| |
Collapse
|
22
|
Prasad EM, Hung SY. Behavioral Tests in Neurotoxin-Induced Animal Models of Parkinson's Disease. Antioxidants (Basel) 2020; 9:E1007. [PMID: 33081318 PMCID: PMC7602991 DOI: 10.3390/antiox9101007] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 09/21/2020] [Accepted: 09/22/2020] [Indexed: 02/06/2023] Open
Abstract
Currently, neurodegenerative diseases are a major cause of disability around the world. Parkinson's disease (PD) is the second-leading cause of neurodegenerative disorder after Alzheimer's disease. In PD, continuous loss of dopaminergic neurons in the substantia nigra causes dopamine depletion in the striatum, promotes the primary motor symptoms of resting tremor, bradykinesia, muscle rigidity, and postural instability. The risk factors of PD comprise environmental toxins, drugs, pesticides, brain microtrauma, focal cerebrovascular injury, aging, and hereditary defects. The pathologic features of PD include impaired protein homeostasis, mitochondrial dysfunction, nitric oxide, and neuroinflammation, but the interaction of these factors contributing to PD is not fully understood. In neurotoxin-induced PD models, neurotoxins, for instance, 6-hydroxydopamine (6-OHDA), 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP), 1-Methyl-4-phenylpyridinium (MPP+), paraquat, rotenone, and permethrin mainly impair the mitochondrial respiratory chain, activate microglia, and generate reactive oxygen species to induce autooxidation and dopaminergic neuronal apoptosis. Since no current treatment can cure PD, using a suitable PD animal model to evaluate PD motor symptoms' treatment efficacy and identify therapeutic targets and drugs are still needed. Hence, the present review focuses on the latest scientific developments in different neurotoxin-induced PD animal models with their mechanisms of pathogenesis and evaluation methods of PD motor symptoms.
Collapse
Affiliation(s)
- E. Maruthi Prasad
- Graduate Institute of Acupuncture Science, College of Chinese Medicine, China Medical University, No.91, Hsueh-Shih Road, Taichung 40402, Taiwan;
| | - Shih-Ya Hung
- Graduate Institute of Acupuncture Science, College of Chinese Medicine, China Medical University, No.91, Hsueh-Shih Road, Taichung 40402, Taiwan;
- Department of Medical Research, China Medical University Hospital, No. 2, Yude Road, Taichung 40447, Taiwan
| |
Collapse
|
23
|
Casares L, Unciti-Broceta JD, Prados ME, Caprioglio D, Mattoteia D, Higgins M, Apendino G, Dinkova-Kostova AT, Muñoz E, de la Vega L. Isomeric O-methyl cannabidiolquinones with dual BACH1/NRF2 activity. Redox Biol 2020; 37:101689. [PMID: 32863231 PMCID: PMC7476313 DOI: 10.1016/j.redox.2020.101689] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 07/31/2020] [Accepted: 08/17/2020] [Indexed: 01/04/2023] Open
Abstract
Oxidative stress and inflammation in the brain are two key hallmarks of neurodegenerative diseases (NDs) such as Alzheimer's, Parkinson's, Huntington's and multiple sclerosis. The axis NRF2-BACH1 has anti-inflammatory and anti-oxidant properties that could be exploited pharmacologically to obtain neuroprotective effects. Activation of NRF2 or inhibition of BACH1 are, individually, promising therapeutic approaches for NDs. Compounds with dual activity as NRF2 activators and BACH1 inhibitors, could therefore potentially provide a more robust antioxidant and anti-inflammatory effects, with an overall better neuroprotective outcome. The phytocannabinoid cannabidiol (CBD) inhibits BACH1 but lacks significant NRF2 activating properties. Based on this scaffold, we have developed a novel CBD derivative that is highly effective at both inhibiting BACH1 and activating NRF2. This new CBD derivative provides neuroprotection in cell models of relevance to Huntington's disease, setting the basis for further developments in vivo.
Collapse
Affiliation(s)
- Laura Casares
- Jacqui Wood Cancer Centre, Division of Cellular Medicine, School of Medicine, University of Dundee, UK
| | | | | | - Diego Caprioglio
- Dipartimento di Scienze Del Farmaco, Università Del Piemonte Orientale, Novara, Italy
| | - Daiana Mattoteia
- Dipartimento di Scienze Del Farmaco, Università Del Piemonte Orientale, Novara, Italy
| | - Maureen Higgins
- Jacqui Wood Cancer Centre, Division of Cellular Medicine, School of Medicine, University of Dundee, UK
| | - Giovanni Apendino
- Dipartimento di Scienze Del Farmaco, Università Del Piemonte Orientale, Novara, Italy
| | - Albena T Dinkova-Kostova
- Jacqui Wood Cancer Centre, Division of Cellular Medicine, School of Medicine, University of Dundee, UK
| | - Eduardo Muñoz
- Instituto Maimónides de Investigación Biomédica de Córdoba, Spain; Departamento de Biología Celular, Fisiología e Inmunología, Universidad de Córdoba, Spain; Hospital Universitario Reina Sofía, Córdoba, Spain
| | - Laureano de la Vega
- Jacqui Wood Cancer Centre, Division of Cellular Medicine, School of Medicine, University of Dundee, UK.
| |
Collapse
|
24
|
Jayanti S, Vítek L, Tiribelli C, Gazzin S. The Role of Bilirubin and the Other "Yellow Players" in Neurodegenerative Diseases. Antioxidants (Basel) 2020; 9:900. [PMID: 32971784 PMCID: PMC7555389 DOI: 10.3390/antiox9090900] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 09/16/2020] [Accepted: 09/20/2020] [Indexed: 02/07/2023] Open
Abstract
Bilirubin is a yellow endogenous derivate of the heme catabolism. Since the 1980s, it has been recognized as one of the most potent antioxidants in nature, able to counteract 10,000× higher intracellular concentrations of H2O2. In the recent years, not only bilirubin, but also its precursor biliverdin, and the enzymes involved in their productions (namely heme oxygenase and biliverdin reductase; altogether the "yellow players"-YPs) have been recognized playing a protective role in diseases characterized by a chronic prooxidant status. Based on that, there is an ongoing effort in inducing their activity as a therapeutic option. Nevertheless, the understanding of their specific contributions to pathological conditions of the central nervous system (CNS) and their role in these diseases are limited. In this review, we will focus on the most recent evidence linking the role of the YPs specifically to neurodegenerative and neurological conditions. Both the protective, as well as potentially worsening effects of the YP's activity will be discussed.
Collapse
Affiliation(s)
- Sri Jayanti
- Fondazione Italiana Fegato-Onlus, Bldg. Q, AREA Science Park, ss14, Km 163.5, Basovizza, 34149 Trieste, Italy; (S.J.); (C.T.)
- Faculty of Medicine, Universitas Hasanuddin, Makassar 90245, Indonesia
- Molecular Biomedicine Ph.D. Program, University of Trieste, 34127 Trieste, Italy
| | - Libor Vítek
- Institute of Medical Biochemistry and Laboratory Diagnostics, and 4th Department of Internal Medicine, Faculty General Hospital and 1st Faculty of Medicine, Charles University, 12000 Prague, Czech Republic;
| | - Claudio Tiribelli
- Fondazione Italiana Fegato-Onlus, Bldg. Q, AREA Science Park, ss14, Km 163.5, Basovizza, 34149 Trieste, Italy; (S.J.); (C.T.)
| | - Silvia Gazzin
- Fondazione Italiana Fegato-Onlus, Bldg. Q, AREA Science Park, ss14, Km 163.5, Basovizza, 34149 Trieste, Italy; (S.J.); (C.T.)
| |
Collapse
|
25
|
El-Ghaiesh SH, Bahr HI, Ibrahiem AT, Ghorab D, Alomar SY, Farag NE, Zaitone SA. Metformin Protects From Rotenone-Induced Nigrostriatal Neuronal Death in Adult Mice by Activating AMPK-FOXO3 Signaling and Mitigation of Angiogenesis. Front Mol Neurosci 2020; 13:84. [PMID: 32625061 PMCID: PMC7314970 DOI: 10.3389/fnmol.2020.00084] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Accepted: 04/24/2020] [Indexed: 12/12/2022] Open
Abstract
Parkinson’s disease (PD) is a neurodegenerative disease that affects substantia nigra dopamine neurons. Many studies have documented the role of oxidative stress and angiogenesis in the pathogenesis of PD. Metformin (MTF) is an antidiabetic medication and AMP-activated protein kinase (AMPK) regulator that has shown antioxidant and antiangiogenic properties in many disorders. The aim of this study is to investigate the neuroprotective effect of MTF in a mouse model of rotenone-prompted PD with a highlight on its influence on the AMPK/forkhead box transcription factor O3 (FOXO3) pathway and striatal angiogenesis. In the running study, PD was induced in mice using repeated doses of rotenone and concomitantly treated with MTF 100 or 200 mg/kg/day for 18 days. Rotarod and pole tests were used to examine the animals’ motor functionality. After that, animals were sacrificed, and brains were isolated and processed for immunohistochemical investigations or biochemical analyses. Oxidant stress and angiogenic markers were measured, including reduced glutathione, malondialdehyde, the nuclear factor erythroid 2–related factor 2 (Nrf2), hemoxygenase-1, thioredoxin, AMPK, FOXO3, and vascular endothelial growth factor (VEGF). Results indicated that MTF improved animals’ motor function, improved striatal glutathione, Nrf2, hemoxygenase-1, and thioredoxin. Furthermore, MTF upregulated AMPK-FOXO3 proteins and reduced VEGF and cleaved caspase 3. MTF also increased the number of tyrosine hydroxylase (TH)–stained neurons in the substantia nigra neurons and in striatal neuronal terminals. This study is the first to highlight that the neuroprotective role of MTF is mediated through activation of AMPK-FOXO3 signaling and inhibition of the proangiogenic factor, VEGF. Further studies are warranted to confirm this mechanism in other models of PD and neurodegenerative diseases.
Collapse
Affiliation(s)
- Sabah H El-Ghaiesh
- Department of Pharmacology, Faculty of Medicine, University of Tabuk, Tabuk, Saudi Arabia.,Department of Pharmacology, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Hoda I Bahr
- Department of Biochemistry, Faculty of Veterinary Medicine, Suez Canal University, Ismailia, Egypt
| | - Afaf T Ibrahiem
- Department of Pathology, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Doaa Ghorab
- Department of Pathology, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Suliman Y Alomar
- Doping Research Chair, Department of Zoology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Noha E Farag
- Department of Physiology, Faculty of Medicine, Suez Canal University, Ismailia, Egypt.,Department of Physiology, College of Medicine, Taif University, Taif, Saudi Arabia
| | - Sawsan A Zaitone
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Suez Canal University, Ismailia, Egypt.,Department of Pharmacology and Toxicology, Faculty of Pharmacy, University of Tabuk, Tabuk, Saudi Arabia
| |
Collapse
|
26
|
Wang D, Zhu B, Liu X, Han Q, Ge W, Zhang W, Lu Y, Wu Q, Shi L. Daphnetin Ameliorates Experimental Autoimmune Encephalomyelitis Through Regulating Heme Oxygenase-1. Neurochem Res 2020; 45:872-881. [PMID: 31950453 DOI: 10.1007/s11064-020-02960-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2019] [Revised: 12/14/2019] [Accepted: 01/09/2020] [Indexed: 01/17/2023]
Abstract
To assess the potential role of daphnetin, a clinically used anti-inflammatory agent, on the development of the inflammatory and neurodegenerative disease, we investigated its immune regulatory function in a murine model of experimental autoimmune encephalomyelitis (EAE). Significantly, lower levels of pro-inflammatory cytokines including interleukin (IL)-17, interferon-γ, Il6, Il12a, and Il23a were observed in brains of daphnetin-treated EAE mice, compared with those in control littermates. We also confirmed that daphnetin suppressed the production of IL-1β, IL-6, and tumor necrosis factor-α in lipopolysaccharide-stimulated mouse BV2 microglial cells. Mechanistically, heme oxygenase-1 (HO-1), a canonical anti-oxidant and anti-inflammatory factor, was found to be substantially induced by daphnetin treatment in BV2 cells. Also, a significantly higher level of HO-1, accompanied by a decreased level of malondialdehyde, was observed in daphnetin-treated EAE mice. More importantly, the deletion of HO-1 in BV2 microglia largely abrogated daphnetin-mediated inhibition of the inflammatory response. Together, our data demonstrate that daphnetin has an anti-inflammatory and neuroprotective role during the pathogenesis of EAE, which is partially at least, dependent on its regulation of HO-1.
Collapse
Affiliation(s)
- Dan Wang
- Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, 310018, China
| | - Bo Zhu
- Department of Immunology and Medical Microbiology, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Xiaoyi Liu
- Department of Immunology and Medical Microbiology, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Qin Han
- Department of Immunology and Medical Microbiology, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Weihong Ge
- College of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Wenping Zhang
- Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, 310018, China
| | - Yin Lu
- College of Pharmaceutical Science, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Qinan Wu
- College of Pharmaceutical Science, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Liyun Shi
- Department of Immunology and Medical Microbiology, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| |
Collapse
|
27
|
Regenerative Potential of Carbon Monoxide in Adult Neural Circuits of the Central Nervous System. Int J Mol Sci 2020; 21:ijms21072273. [PMID: 32218342 PMCID: PMC7177523 DOI: 10.3390/ijms21072273] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 03/17/2020] [Accepted: 03/23/2020] [Indexed: 01/04/2023] Open
Abstract
Regeneration of adult neural circuits after an injury is limited in the central nervous system (CNS). Heme oxygenase (HO) is an enzyme that produces HO metabolites, such as carbon monoxide (CO), biliverdin and iron by heme degradation. CO may act as a biological signal transduction effector in CNS regeneration by stimulating neuronal intrinsic and extrinsic mechanisms as well as mitochondrial biogenesis. CO may give directions by which the injured neurovascular system switches into regeneration mode by stimulating endogenous neural stem cells and endothelial cells to produce neurons and vessels capable of replacing injured neurons and vessels in the CNS. The present review discusses the regenerative potential of CO in acute and chronic neuroinflammatory diseases of the CNS, such as stroke, traumatic brain injury, multiple sclerosis and Alzheimer’s disease and the role of signaling pathways and neurotrophic factors. CO-mediated facilitation of cellular communications may boost regeneration, consequently forming functional adult neural circuits in CNS injury.
Collapse
|
28
|
The Nrf2/HO-1 Axis as Targets for Flavanones: Neuroprotection by Pinocembrin, Naringenin, and Eriodictyol. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:4724920. [PMID: 31814878 PMCID: PMC6878820 DOI: 10.1155/2019/4724920] [Citation(s) in RCA: 90] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Revised: 09/20/2019] [Accepted: 09/25/2019] [Indexed: 12/16/2022]
Abstract
Flavanones are a group of flavonoids that derive from their immediate chalcone precursors through the action of chalcone isomerase enzymes. The Aromatic A and B rings, C4-keto group, and the 15-carbon flavonoid skeleton are all evident in flavanones, but a notable absence of C2-C3 double bond and a lack of oxygenation at C-3 position of the C-ring makes them distinctively different from other groups such as flavonols (e.g., quercetin). On the basis of oxygenation level in the B ring, flavanones can vary from each other as exemplified by pinocembrin (no oxygenation), naringenin (4′-hydroxyl), or eriodictyol (3′,4′-dihydroxyl substitution). These groups are generally weaker free radical scavengers as compared to quercetin and derivatives though eriodictyol has a better free radical scavenging profile within the group due to the presence of the catechol functional moiety. In this communication, their antioxidant potential through the induction of antioxidant defenses is scrutinized. These compounds as exemplified by pinocembrin could induce the nuclear factor erythroid 2-related factor 2- (Nrf2-) heme oxygenase-1 (HO-1) axis leading to amelioration of oxidative stress in cellular and animal models. Their neuroprotective effect through such mechanism is discussed.
Collapse
|
29
|
Ammal Kaidery N, Ahuja M, Thomas B. Crosstalk between Nrf2 signaling and mitochondrial function in Parkinson's disease. Mol Cell Neurosci 2019; 101:103413. [PMID: 31644952 DOI: 10.1016/j.mcn.2019.103413] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 09/10/2019] [Accepted: 09/20/2019] [Indexed: 12/12/2022] Open
Abstract
Search for a definitive cure for neurodegenerative disorders like Parkinson's disease (PD) has met with little success. Mitochondrial dysfunction and elevated oxidative stress precede characteristic loss of dopamine-producing neurons from the midbrain in PD. The majority of PD cases are classified as sporadic (sPD) with an unknown etiology, whereas mutations in a handful of genes cause monogenic form called familial (fPD). Both sPD and fPD is characterized by proteinopathy and mitochondrial dysfunction leading to increased oxidative stress. These pathophysiological mechanisms create a vicious cycle feeding into each other, ultimately tipping the neurons to its demise. Effect of iron accumulation and dopamine oxidation adds an additional dimension to mitochondrial oxidative stress and apoptotic pathways affected. Nrf2 is a redox-sensitive transcription factor which regulates basal as well as inducible expression of antioxidant enzymes and proteins involved in xenobiotic detoxification. Recent advances, however, shows a multifaceted role for Nrf2 in the regulation of genes connected with inflammatory response, metabolic pathways, protein homeostasis, iron management, and mitochondrial bioenergetics. Here we review the role of mitochondria and oxidative stress in the PD etiology and the potential crosstalk between Nrf2 signaling and mitochondrial function in PD. We also make a case for the development of therapeutics that safely activates Nrf2 pathway in halting the progression of neurodegeneration in PD patients.
Collapse
Affiliation(s)
- Navneet Ammal Kaidery
- Darby Research Institute, Medical University of South Carolina, Charleston, SC 29425, United States of America; Department of Pediatrics, Medical University of South Carolina, Charleston, SC 29425, United States of America
| | - Manuj Ahuja
- Darby Research Institute, Medical University of South Carolina, Charleston, SC 29425, United States of America; Department of Pediatrics, Medical University of South Carolina, Charleston, SC 29425, United States of America
| | - Bobby Thomas
- Darby Research Institute, Medical University of South Carolina, Charleston, SC 29425, United States of America; Department of Pediatrics, Medical University of South Carolina, Charleston, SC 29425, United States of America; Department of Neuroscience, Medical University of South Carolina, Charleston, SC 29425, United States of America; Department of Drug Discovery, Medical University of South Carolina, Charleston, SC 29425, United States of America.
| |
Collapse
|
30
|
Liu Z, Qiu AW, Huang Y, Yang Y, Chen JN, Gu TT, Cao BB, Qiu YH, Peng YP. IL-17A exacerbates neuroinflammation and neurodegeneration by activating microglia in rodent models of Parkinson's disease. Brain Behav Immun 2019; 81:630-645. [PMID: 31351185 DOI: 10.1016/j.bbi.2019.07.026] [Citation(s) in RCA: 114] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Revised: 07/17/2019] [Accepted: 07/23/2019] [Indexed: 12/26/2022] Open
Abstract
Neuroinflammation has been involved in pathogenesis of Parkinson's disease (PD), a chronic neurodegenerative disease characterized neuropathologically by progressive dopaminergic neuronal loss in the substantia nigra (SN). We recently have shown that helper T (Th)17 cells facilitate dopaminergic neuronal loss in vitro. Herein, we demonstrated that interleukin (IL)-17A, a proinflammatory cytokine produced mainly by Th17 cells, contributed to PD pathogenesis depending on microglia. Mouse and rat models for PD were prepared by intraperitoneal injection of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) or striatal injection of 1-methyl-4-phenylpyridinium (MPP+), respectively. Both in MPTP-treated mice and MPP+-treated rats, blood-brain barrier (BBB) was disrupted and IL-17A level increased in the SN but not in cortex. Effector T (Teff) cells that were adoptively transferred via tail veins infiltrated into the brain of PD mice but not into that of normal mice. The Teff cell transfer aggravated nigrostriatal dopaminergic neurodegeneration, microglial activation and motor impairment. Contrarily, IL-17A deficiency alleviated BBB disruption, dopaminergic neurodegeneration, microglial activation and motor impairment. Anti-IL-17A-neutralizing antibody that was injected into lateral cerebral ventricle in PD rats ameliorated the manifestations mentioned above. IL-17A activated microglia but did not directly affect dopaminergic neuronal survival in vitro. IL-17A exacerbated dopaminergic neuronal loss only in the presence of microglia, and silencing IL-17A receptor gene in microglia abolished the IL-17A effect. IL-17A-treated microglial medium that contained higher concentration of tumor necrosis factor (TNF)-α facilitated dopaminergic neuronal death. Further, TNF-α-neutralizing antibody attenuated MPP+-induced neurotoxicity. The findings suggest that IL-17A accelerates neurodegeneration in PD depending on microglial activation and at least partly TNF-α release.
Collapse
Affiliation(s)
- Zhan Liu
- Department of Physiology, School of Medicine, and Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province 226001, China
| | - Ao-Wang Qiu
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province 210029, China
| | - Yan Huang
- Department of Physiology, School of Medicine, and Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province 226001, China
| | - Ya Yang
- Department of Physiology, School of Medicine, and Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province 226001, China
| | - Jin-Na Chen
- Department of Physiology, School of Medicine, and Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province 226001, China
| | - Ting-Ting Gu
- Department of Physiology, School of Medicine, and Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province 226001, China
| | - Bei-Bei Cao
- Department of Physiology, School of Medicine, and Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province 226001, China
| | - Yi-Hua Qiu
- Department of Physiology, School of Medicine, and Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province 226001, China.
| | - Yu-Ping Peng
- Department of Physiology, School of Medicine, and Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province 226001, China.
| |
Collapse
|
31
|
Dal Ben M, Bongiovanni R, Tuniz S, Fioriti E, Tiribelli C, Moretti R, Gazzin S. Earliest Mechanisms of Dopaminergic Neurons Sufferance in a Novel Slow Progressing Ex Vivo Model of Parkinson Disease in Rat Organotypic Cultures of Substantia Nigra. Int J Mol Sci 2019; 20:2224. [PMID: 31064126 PMCID: PMC6539377 DOI: 10.3390/ijms20092224] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Revised: 04/30/2019] [Accepted: 05/03/2019] [Indexed: 12/17/2022] Open
Abstract
The current treatments of Parkinson disease (PD) are ineffective mainly due to the poor understanding of the early events causing the decline of dopaminergic neurons (DOPAn). To overcome this problem, slow progressively degenerating models of PD allowing the study of the pre-clinical phase are crucial. We recreated in a short ex vivo time scale (96 h) all the features of human PD (needing dozens of years) by challenging organotypic culture of rat substantia nigra with low doses of rotenone. Thus, taking advantage of the existent knowledge, the model was used to perform a time-dependent comparative study of the principal possible causative molecular mechanisms undergoing DOPAn demise. Alteration in the redox state and inflammation started at 3 h, preceding the reduction in DOPAn number (pre-diagnosis phase). The number of DOPAn declined to levels compatible with diagnosis only at 12 h. The decline was accompanied by a persistent inflammation and redox imbalance. Significant microglia activation, apoptosis, a reduction in dopamine vesicle transporters, and the ubiquitination of misfolded protein clearance pathways were late (96 h, consequential) events. The work suggests inflammation and redox imbalance as simultaneous early mechanisms undergoing DOPAn sufferance, to be targeted for a causative treatment aimed to stop/delay PD.
Collapse
Affiliation(s)
- Matteo Dal Ben
- Department of Medical, Surgical, and Health Sciences, University of Trieste, 34100 Trieste, Italy.
- Fondazione Italiana Fegato, AREA Science Park, 34149 Trieste, Italy.
| | | | - Simone Tuniz
- Fondazione Italiana Fegato, AREA Science Park, 34149 Trieste, Italy.
| | - Emanuela Fioriti
- Fondazione Italiana Fegato, AREA Science Park, 34149 Trieste, Italy.
| | - Claudio Tiribelli
- Fondazione Italiana Fegato, AREA Science Park, 34149 Trieste, Italy.
| | - Rita Moretti
- Neurology Clinic, Department of Medical, Surgical, and Health Sciences, University of Trieste, 34100 Trieste, Italy.
| | - Silvia Gazzin
- Fondazione Italiana Fegato, AREA Science Park, 34149 Trieste, Italy.
| |
Collapse
|
32
|
Ifhar LS, Ene HM, Ben-Shachar D. Impaired heme metabolism in schizophrenia-derived cell lines and in a rat model of the disorder: Possible involvement of mitochondrial complex I. Eur Neuropsychopharmacol 2019; 29:577-589. [PMID: 30948194 DOI: 10.1016/j.euroneuro.2019.03.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2018] [Revised: 03/18/2019] [Accepted: 03/22/2019] [Indexed: 02/07/2023]
Abstract
Accumulating data point to heme involvement in neuropsychiatric disorders. Heme plays a role in major cellular processes such as signal transduction, protein complex assembly and regulation of transcription and translation. Its synthesis involves the mitochondria, which dysfunction, specifically that of the complex I (Co-I) of the electron transport chain is involved in the pathophysiology of schizophrenia (SZ). Here we aimed to demonstrate that deficits in Co-I affect heme metabolism. We show a significant decrease in heme levels in Co-I deficient SZ-derived EBV transformed lymphocytes (lymphoblastoid cell lines - LCLs) as compared to healthy subjects-derived cells (n = 9/cohort). Moreover, protein levels assessed by immunoblotting and mRNA levels assessed by qRT-PCR of heme catabolic enzyme, heme Oxygenase 1 (HO-1), and protein levels of heme downstream target phosphorylated eukaryotic initiation factor 2-alpha (Peif2a/eif2a) were significantly elevated in SZ-derived cells. In contrast, protein and mRNA levels of heme synthesis rate limiting enzyme aminolevulinic acid synthase-1 (ALAS1) were unchanged in SZ derived LCLs. In addition, inhibition of Co-I by rotenone in healthy subjects-derived LCLs (n = 4/cohort) exhibited an initial increase followed by a later decrease in heme levels. These findings were associated with opposite changes in heme's downstream target and HO-1 level, similar to our findings in SZ-derived cells. We also show a brain region specific pattern of impairment in Co-I subunits and in HO-1 and PeIF2α/eIF2α in the Poly-IC rat model of SZ (n = 6/cohort). Our results provide evidence for a link between CoI and heme metabolism both in-vitro and in-vivo suggesting its contribution to SZ pathophysiology.
Collapse
Affiliation(s)
- Lee S Ifhar
- Laboratory of Psychobiology, Department of Psychiatry, Rambam Health Care Campus, B. Rappaport Faculty of Medicine, Rappaport Family Institute for Research in Medical Sciences, Technion IIT, POB 9649, Haifa 31096 Israel
| | - Hila M Ene
- Laboratory of Psychobiology, Department of Psychiatry, Rambam Health Care Campus, B. Rappaport Faculty of Medicine, Rappaport Family Institute for Research in Medical Sciences, Technion IIT, POB 9649, Haifa 31096 Israel
| | - Dorit Ben-Shachar
- Laboratory of Psychobiology, Department of Psychiatry, Rambam Health Care Campus, B. Rappaport Faculty of Medicine, Rappaport Family Institute for Research in Medical Sciences, Technion IIT, POB 9649, Haifa 31096 Israel.
| |
Collapse
|
33
|
Armagan G, Sevgili E, Gürkan FT, Köse FA, Bilgiç T, Dagcı T, Saso L. Regulation of the Nrf2 Pathway by Glycogen Synthase Kinase-3β in MPP⁺-Induced Cell Damage. Molecules 2019; 24:molecules24071377. [PMID: 30965670 PMCID: PMC6480928 DOI: 10.3390/molecules24071377] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Revised: 04/05/2019] [Accepted: 04/07/2019] [Indexed: 01/14/2023] Open
Abstract
Recently, nuclear translocation and stability of nuclear factor erythroid 2 (NF-E2)-related factor 2 (Nrf2) have gained increasing attention in the prevention of oxidative stress. The present study was aimed to evaluate the regulatory role of glycogen synthase kinase-3β (GSK-3β) inhibition by tideglusib through the Nrf2 pathway in a cellular damage model. Gene silencing (siRNA-mediated) was performed to examine the responses of Nrf2-target genes (i.e., heme oxygenase-1, NAD(P)H:quinone oxidoreductase1) to siRNA depletion of Nrf2 in MPP⁺-induced dopaminergic cell death. Nrf2 and its downstream regulated genes/proteins were analyzed using Real-time PCR and Western Blotting techniques, respectively. Moreover, free radical production, the changes in mitochondrial membrane potential, total glutathione, and glutathione-S-transferase were examined. The possible contribution of peroxisome proliferator-activated receptor gamma (PPARγ) to tideglusib-mediated neuroprotection was evaluated. The number of viable cells and mitochondrial membrane potential were increased following GSK-3β enzyme inhibition against MPP⁺. HO-1, NQO1 mRNA/protein expressions and Nrf2 nuclear translocation significantly triggered by tideglusib. Moreover, the neuroprotection by tideglusib was not observed in the presence of siRNA Nrf2. Our study supports the idea that GSK-3β enzyme inhibition may modulate the Nrf2/ARE pathway in cellular damage and the inhibitory role of tideglusib on GSK-3β along with PPARγ activation may be responsible for neuroprotection.
Collapse
Affiliation(s)
- Güliz Armagan
- Department of Biochemistry, Faculty of Pharmacy, Ege University, 35100 Bornova, Izmir, Turkey.
| | - Elvin Sevgili
- Department of Biochemistry, Faculty of Pharmacy, Ege University, 35100 Bornova, Izmir, Turkey.
| | - Fulya Tuzcu Gürkan
- Department of Physiology, School of Medicine, Ege University, 35100 Bornova, Izmir, Turkey.
| | - Fadime Aydın Köse
- Department of Biochemistry, Faculty of Pharmacy, Ege University, 35100 Bornova, Izmir, Turkey.
| | - Tuğçe Bilgiç
- Department of Physiology, School of Medicine, Ege University, 35100 Bornova, Izmir, Turkey.
| | - Taner Dagcı
- Department of Physiology, School of Medicine, Ege University, 35100 Bornova, Izmir, Turkey.
| | - Luciano Saso
- Department of Physiology and Pharmacology "Vittorio Erspamer", Sapienza University of Rome, 00185 Rome, Italy.
| |
Collapse
|
34
|
CRISPR/Cas9 Editing of Glia Maturation Factor Regulates Mitochondrial Dynamics by Attenuation of the NRF2/HO-1 Dependent Ferritin Activation in Glial Cells. J Neuroimmune Pharmacol 2019; 14:537-550. [PMID: 30810907 DOI: 10.1007/s11481-019-09833-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Accepted: 01/08/2019] [Indexed: 12/26/2022]
Abstract
Microglial cells are brain specific professional phagocytic immune cells that play a crucial role in the inflammation- mediated neurodegeneration especially in Parkinson's disease (PD) and Alzheimer's disease. Glia maturation factor (GMF) is a neuroinflammatory protein abundantly expressed in the brain. We have previously shown that GMF expression is significantly upregulated in the substantia nigra (SN) of PD brains. However, its possible role in PD progression is still not fully understood. The Clustered-Regularly Interspaced Short Palindromic Repeats (CRISPR)-CRISPR Associated (Cas) protein9 (CRISPR/Cas9) system is a simple, rapid and often extremely efficient gene editing tool at desired loci, enabling complete gene knockout or homology directed repair. In this study, we examined the effect of GMF editing by using the CRISPR/Cas9 technique in BV2 microglial cells (hereafter referred to as BV2-G) on oxidative stress and nuclear factor erythroid 2-related factor 2 (NRF2)/Hemeoxygenase1 (HO-1)-dependent ferritin activation after treatment with (1-methyl-4-phenylpyridinium) MPP+. Knockout of GMF in BV2-G cells significantly attenuated oxidative stress via reduced ROS production and calcium flux. Furthermore, deficiency of GMF significantly reduced nuclear translocation of NRF2, which modulates HO-1 and ferritin activation, cyclooxygenase 2 (COX2) and nitric oxide synthase 2 (NOS2) expression in BV2 microglial cells. Lack of GMF significantly improved CD11b and CD68 positive microglial cells as compared with untreated cells. Our results also suggest that pharmacological and genetic intervention targeting GMF may represent a promising and a novel therapeutic strategy in controlling Parkinsonism by regulating microglial functions. Targeted regulation of GMF possibly mediates protein aggregation in microglial homeostasis associated with PD progression through regulation of iron metabolism by modulating NRF2-HO1 and ferritin expression.
Collapse
|
35
|
The sinister face of heme oxygenase-1 in brain aging and disease. Prog Neurobiol 2019; 172:40-70. [DOI: 10.1016/j.pneurobio.2018.06.008] [Citation(s) in RCA: 160] [Impact Index Per Article: 26.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Revised: 06/19/2018] [Accepted: 06/30/2018] [Indexed: 11/23/2022]
|
36
|
Lee H, Choi YK. Regenerative Effects of Heme Oxygenase Metabolites on Neuroinflammatory Diseases. Int J Mol Sci 2018; 20:ijms20010078. [PMID: 30585210 PMCID: PMC6337166 DOI: 10.3390/ijms20010078] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Revised: 12/20/2018] [Accepted: 12/21/2018] [Indexed: 12/13/2022] Open
Abstract
Heme oxygenase (HO) catabolizes heme to produce HO metabolites, such as carbon monoxide (CO) and bilirubin (BR), which have gained recognition as biological signal transduction effectors. The neurovascular unit refers to a highly evolved network among endothelial cells, pericytes, astrocytes, microglia, neurons, and neural stem cells in the central nervous system (CNS). Proper communication and functional circuitry in these diverse cell types is essential for effective CNS homeostasis. Neuroinflammation is associated with the vascular pathogenesis of many CNS disorders. CNS injury elicits responses from activated glia (e.g., astrocytes, oligodendrocytes, and microglia) and from damaged perivascular cells (e.g., pericytes and endothelial cells). Most brain lesions cause extensive proliferation and growth of existing glial cells around the site of injury, leading to reactions causing glial scarring, which may act as a major barrier to neuronal regrowth in the CNS. In addition, damaged perivascular cells lead to the breakdown of the blood-neural barrier, and an increase in immune activation, activated glia, and neuroinflammation. The present review discusses the regenerative role of HO metabolites, such as CO and BR, in various vascular diseases of the CNS such as stroke, traumatic brain injury, diabetic retinopathy, and Alzheimer's disease, and the role of several other signaling molecules.
Collapse
Affiliation(s)
- Huiju Lee
- Department of Integrative Bioscience and Biotechnology, Konkuk University, Seoul 05029, Korea.
| | - Yoon Kyung Choi
- Department of Integrative Bioscience and Biotechnology, Konkuk University, Seoul 05029, Korea.
| |
Collapse
|
37
|
Feng JH, Hu XL, Lv XY, Wang BL, Lin J, Zhang XQ, Ye WC, Xiong F, Wang H. Synthesis and biological evaluation of clovamide analogues with catechol functionality as potent Parkinson's disease agents in vitro and in vivo. Bioorg Med Chem Lett 2018; 29:302-312. [PMID: 30470490 DOI: 10.1016/j.bmcl.2018.11.030] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Revised: 10/23/2018] [Accepted: 11/14/2018] [Indexed: 10/27/2022]
Affiliation(s)
- Jia-Hao Feng
- State Key Laboratory of Natural Medicines, Department of TCMs Pharmaceuticals, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, People's Republic of China
| | - Xiao-Long Hu
- State Key Laboratory of Natural Medicines, Department of TCMs Pharmaceuticals, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, People's Republic of China
| | - Xian-Yu Lv
- State Key Laboratory of Natural Medicines, Department of TCMs Pharmaceuticals, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, People's Republic of China
| | - Bao-Lin Wang
- State Key Laboratory of Natural Medicines, Department of TCMs Pharmaceuticals, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, People's Republic of China
| | - Jun Lin
- State Key Laboratory of Natural Medicines, Department of TCMs Pharmaceuticals, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, People's Republic of China
| | - Xiao-Qi Zhang
- Institute of Traditional Chinese Medicine & Natural Products, Jinan University, Guangzhou 510632, People's Republic of China
| | - Wen-Cai Ye
- Institute of Traditional Chinese Medicine & Natural Products, Jinan University, Guangzhou 510632, People's Republic of China
| | - Fei Xiong
- State Key Laboratory of Bioelectronics, Jiangsu Laboratory for Biomaterials and Devices, Southeast University, Nanjing 210009, People's Republic of China.
| | - Hao Wang
- State Key Laboratory of Natural Medicines, Department of TCMs Pharmaceuticals, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, People's Republic of China.
| |
Collapse
|
38
|
Heme Oxygenase 1 in the Nervous System: Does It Favor Neuronal Cell Survival or Induce Neurodegeneration? Int J Mol Sci 2018; 19:ijms19082260. [PMID: 30071692 PMCID: PMC6121636 DOI: 10.3390/ijms19082260] [Citation(s) in RCA: 116] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Revised: 07/25/2018] [Accepted: 07/30/2018] [Indexed: 02/08/2023] Open
Abstract
Heme oxygenase 1 (HO-1) up-regulation is recognized as a pivotal mechanism of cell adaptation to stress. Under control of different transcription factors but with a prominent role played by Nrf2, HO-1 induction is crucial also in nervous system response to damage. However, several lines of evidence have highlighted that HO-1 expression is associated to neuronal damage and neurodegeneration especially in Alzheimer’s and Parkinson’s diseases. In this review, we summarize the current literature regarding the role of HO-1 in nervous system pointing out different molecular mechanisms possibly responsible for HO-1 up-regulation in nervous system homeostasis and neurodegeneration.
Collapse
|
39
|
Shefa U, Kim D, Kim MS, Jeong NY, Jung J. Roles of Gasotransmitters in Synaptic Plasticity and Neuropsychiatric Conditions. Neural Plast 2018; 2018:1824713. [PMID: 29853837 PMCID: PMC5960547 DOI: 10.1155/2018/1824713] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Revised: 02/25/2018] [Accepted: 03/11/2018] [Indexed: 12/22/2022] Open
Abstract
Synaptic plasticity is important for maintaining normal neuronal activity and proper neuronal functioning in the nervous system. It is crucial for regulating synaptic transmission or electrical signal transduction to neuronal networks, for sharing essential information among neurons, and for maintaining homeostasis in the body. Moreover, changes in synaptic or neural plasticity are associated with many neuropsychiatric conditions, such as schizophrenia (SCZ), bipolar disorder (BP), major depressive disorder (MDD), and Alzheimer's disease (AD). The improper maintenance of neural plasticity causes incorrect neurotransmitter transmission, which can also cause neuropsychiatric conditions. Gas neurotransmitters (gasotransmitters), such as hydrogen sulfide (H2S), nitric oxide (NO), and carbon monoxide (CO), play roles in maintaining synaptic plasticity and in helping to restore such plasticity in the neuronal architecture in the central nervous system (CNS). Indeed, the upregulation or downregulation of these gasotransmitters may cause neuropsychiatric conditions, and their amelioration may restore synaptic plasticity and proper neuronal functioning and thereby improve such conditions. Understanding the specific molecular mechanisms underpinning these effects can help identify ways to treat these neuropsychiatric conditions.
Collapse
Affiliation(s)
- Ulfuara Shefa
- Department of Biomedical Science, Graduate School, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea
| | - Dokyoung Kim
- Department of Biomedical Science, Graduate School, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea
- Department of Anatomy and Neurobiology, College of Medicine, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea
| | - Min-Sik Kim
- Department of Applied Chemistry, College of Applied Science, Kyung Hee University, Deogyeong-daero, Giheung-gu, Yongin-si, Gyeonggi-do 17104, Republic of Korea
| | - Na Young Jeong
- Department of Anatomy and Cell Biology, College of Medicine, Dong-A University, 32 Daesingongwon-ro, Seo-gu, Busan 49201, Republic of Korea
| | - Junyang Jung
- Department of Biomedical Science, Graduate School, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea
- Department of Anatomy and Neurobiology, College of Medicine, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea
- East-West Medical Research Institute, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, 13 Seoul 02447, Republic of Korea
| |
Collapse
|
40
|
Waza AA, Hamid Z, Ali S, Bhat SA, Bhat MA. A review on heme oxygenase-1 induction: is it a necessary evil. Inflamm Res 2018; 67:579-588. [PMID: 29693710 DOI: 10.1007/s00011-018-1151-x] [Citation(s) in RCA: 185] [Impact Index Per Article: 26.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2018] [Revised: 04/08/2018] [Accepted: 04/16/2018] [Indexed: 02/07/2023] Open
Abstract
Heme oxygenase-1 (HO-1) is considered to be the main protein in diseases arising as a result of oxidative and inflammatory insults. Tremendous research has been carried out on HO-1 since years, pertaining its cytoprotective effect against oxidative injury and other cellular stresses. HO-1, by regulating intracellular levels of pro-oxidant heme, or by other benefits of its by-products such as carbon monoxide (CO) and biliverdin (BV) had become an important candidate protein to be up-regulated to combat diverse stressful events. Although the beneficial effects of HO-1 induction have been reported in a number of cells and tissues, a growing body of evidence indicates that this increased HO-1 expression may lead to the progression of several diseases such as neurodegeneration, carcinogenesis. But it is not clear, what accounts for the increased expression of HO-1 in cells and tissues. The observed friendly role of HO-1 in a wide range of stress conditions since times is now doubtful. Therefore, more studies are needed to elucidate the exact role of HO-1 in various stressful events. Being more concise, elucidating the effect of HO-1 up-regulation on critical genes involved in particular diseases such as cancer will help to a larger extent to comprehend the exact role of HO-1. This review will assist in understanding the dual role (protective and detrimental) of HO-1 and the signaling pathway involved and will help in unraveling the doubtful role of HO-1 induction.
Collapse
Affiliation(s)
- Ajaz Ahmad Waza
- Centre of Research for Development (CORD), University of Kashmir, Srinagar, Jammu and Kashmir, India.
| | - Zeenat Hamid
- Department of Biotechnology, University of Kashmir, Srinagar, Jammu and Kashmir, India
| | - Sajad Ali
- Centre of Research for Development (CORD), University of Kashmir, Srinagar, Jammu and Kashmir, India
| | - Shabir Ahmad Bhat
- Centre of Research for Development (CORD), University of Kashmir, Srinagar, Jammu and Kashmir, India
| | | |
Collapse
|
41
|
Son HJ, Han SH, Lee JA, Shin EJ, Hwang O. Potential repositioning of exemestane as a neuroprotective agent for Parkinson's disease. Free Radic Res 2018; 51:633-645. [PMID: 28770670 DOI: 10.1080/10715762.2017.1353688] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder characterised by selective degeneration of the nigral dopaminergic neurons, and neuroinflammation and oxidative stress are believed to be involved in its pathogenesis. In the present study, we provide data that the synthetic steroid exemestane, which is currently being used to treat breast cancer, may be useful for PD therapy. In BV-2 microglial cells, exemestane activated the transcription factor Nrf2 and induced expression of the Nrf2-dependent genes that encode the antioxidant enzymes NAD(P)H: quinone oxidoreductase 1, haem oxygenase-1, and glutamylcysteine ligase. It also downregulated gene expression of inducible nitric oxide (NO) synthase, lowered the levels of NO and reactive oxygen species, interleukin-1β and tumour necrosis factor-α in lipopolysaccharide-activated microglial cells. In CATH.a dopaminergic neuronal cells, exemestane also induced the same set of Nrf2-dependent antioxidant enzyme genes and provided neuroprotection against oxidative damage. In vivo, the drug protected the nigral dopaminergic neurons, decreased microglial activation, and prevented motor deficits in C57Bl/6 male mice that had been administered with the dopaminergic neurotoxin MPTP. Taken together, the results suggested a utility of repositioning exemestane towards disease-modifying therapy for PD.
Collapse
Affiliation(s)
- Hyo Jin Son
- a Department of Biochemistry and Molecular Biology , University of Ulsan College of Medicine , Seoul , South Korea
| | - Se Hee Han
- a Department of Biochemistry and Molecular Biology , University of Ulsan College of Medicine , Seoul , South Korea
| | - Ji Ae Lee
- a Department of Biochemistry and Molecular Biology , University of Ulsan College of Medicine , Seoul , South Korea
| | - Eun Jung Shin
- a Department of Biochemistry and Molecular Biology , University of Ulsan College of Medicine , Seoul , South Korea
| | - Onyou Hwang
- a Department of Biochemistry and Molecular Biology , University of Ulsan College of Medicine , Seoul , South Korea
| |
Collapse
|
42
|
Gill AJ, Garza R, Ambegaokar SS, Gelman BB, Kolson DL. Heme oxygenase-1 promoter region (GT)n polymorphism associates with increased neuroimmune activation and risk for encephalitis in HIV infection. J Neuroinflammation 2018; 15:70. [PMID: 29510721 PMCID: PMC5838989 DOI: 10.1186/s12974-018-1102-z] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Accepted: 02/21/2018] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Heme oxygenase-1 (HO-1) is a critical cytoprotective enzyme that limits oxidative stress, inflammation, and cellular injury within the central nervous system (CNS) and other tissues. We previously demonstrated that HO-1 protein expression is decreased within the brains of HIV+ subjects and that this HO-1 reduction correlates with CNS immune activation and neurocognitive dysfunction. To define a potential CNS protective role for HO-1 against HIV, we analyzed a well-characterized HIV autopsy cohort for two common HO-1 promoter region polymorphisms that are implicated in regulating HO-1 promoter transcriptional activity, a (GT)n dinucleotide repeat polymorphism and a single nucleotide polymorphism (A(-413)T). Shorter HO-1 (GT)n repeats and the 'A' SNP allele associate with higher HO-1 promoter activity. METHODS Brain dorsolateral prefrontal cortex tissue samples from an autopsy cohort of HIV-, HIV+, and HIV encephalitis (HIVE) subjects (n = 554) were analyzed as follows: HO-1 (GT)n polymorphism allele lengths were determined by PCR and capillary electrophoresis, A(-413)T SNP alleles were determined by PCR with allele specific probes, and RNA expression of selected neuroimmune markers was analyzed by quantitative PCR. RESULTS HIV+ subjects with shorter HO-1 (GT)n alleles had a significantly lower risk of HIVE; however, shorter HO-1 (GT)n alleles did not correlate with CNS or peripheral viral loads. In HIV+ subjects without HIVE, shorter HO-1 (GT)n alleles associated significantly with lower expression of brain type I interferon response markers (MX1, ISG15, and IRF1) and T-lymphocyte activation markers (CD38 and GZMB). No significant correlations were found between the HO-1 (GT)n repeat length and brain expression of macrophage markers (CD163, CD68), endothelial markers (PECAM1, VWF), the T-lymphocyte marker CD8A, or the B-lymphocyte maker CD19. Finally, we found no significant associations between the A(-413)T SNP and HIVE diagnosis, HIV viral loads, or any neuroimmune markers. CONCLUSION Our data suggest that an individual's HO-1 promoter region (GT)n polymorphism allele repeat length exerts unique modifying risk effects on HIV-induced CNS neuroinflammation and associated neuropathogenesis. Shorter HO-1 (GT)n alleles increase HO-1 promoter activity, which could provide neuroprotection through decreased neuroimmune activation. Therapeutic strategies that induce HO-1 expression could decrease HIV-associated CNS neuroinflammation and decrease the risk for development of HIV neurological disease.
Collapse
Affiliation(s)
- Alexander J. Gill
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, 415 Curie Boulevard, 280C Clinical Research Building, Philadelphia, PA 19104 USA
| | - Rolando Garza
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, 415 Curie Boulevard, 280C Clinical Research Building, Philadelphia, PA 19104 USA
| | - Surendra S. Ambegaokar
- Department of Botany & Microbiology, Robbins Program in Neuroscience, Ohio Wesleyan University, Delaware, OH 43016 USA
| | - Benjamin B. Gelman
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555 USA
| | - Dennis L. Kolson
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, 415 Curie Boulevard, 280C Clinical Research Building, Philadelphia, PA 19104 USA
| |
Collapse
|
43
|
Dreyer-Andersen N, Almeida AS, Jensen P, Kamand M, Okarmus J, Rosenberg T, Friis SD, Martínez Serrano A, Blaabjerg M, Kristensen BW, Skrydstrup T, Gramsbergen JB, Vieira HLA, Meyer M. Intermittent, low dose carbon monoxide exposure enhances survival and dopaminergic differentiation of human neural stem cells. PLoS One 2018; 13:e0191207. [PMID: 29338033 PMCID: PMC5770048 DOI: 10.1371/journal.pone.0191207] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Accepted: 12/30/2017] [Indexed: 12/18/2022] Open
Abstract
Exploratory studies using human fetal tissue have suggested that intrastriatal transplantation of dopaminergic neurons may become a future treatment for patients with Parkinson's disease. However, the use of human fetal tissue is compromised by ethical, regulatory and practical concerns. Human stem cells constitute an alternative source of cells for transplantation in Parkinson's disease, but efficient protocols for controlled dopaminergic differentiation need to be developed. Short-term, low-level carbon monoxide (CO) exposure has been shown to affect signaling in several tissues, resulting in both protection and generation of reactive oxygen species. The present study investigated the effect of CO produced by a novel CO-releasing molecule on dopaminergic differentiation of human neural stem cells. Short-term exposure to 25 ppm CO at days 0 and 4 significantly increased the relative content of β-tubulin III-immunoreactive immature neurons and tyrosine hydroxylase expressing catecholaminergic neurons, as assessed 6 days after differentiation. Also the number of microtubule associated protein 2-positive mature neurons had increased significantly. Moreover, the content of apoptotic cells (Caspase3) was reduced, whereas the expression of a cell proliferation marker (Ki67) was left unchanged. Increased expression of hypoxia inducible factor-1α and production of reactive oxygen species (ROS) in cultures exposed to CO may suggest a mechanism involving mitochondrial alterations and generation of ROS. In conclusion, the present procedure using controlled, short-term CO exposure allows efficient dopaminergic differentiation of human neural stem cells at low cost and may as such be useful for derivation of cells for experimental studies and future development of donor cells for transplantation in Parkinson's disease.
Collapse
Affiliation(s)
- Nanna Dreyer-Andersen
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Ana Sofia Almeida
- Instituto de Biologia Experimental e Tecnológica (IBET), Oeiras, Portugal
- Instituto de Tecnologia Química e Biológica (ITQB), Oeiras, Portugal
- CEDOC, NOVA Medical School/Faculdade de Ciência Médicas, Universidade Nova de Lisboa, Lisboa, Portugal
| | - Pia Jensen
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Morad Kamand
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Justyna Okarmus
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Tine Rosenberg
- Department of Pathology, Odense University Hospital, Denmark & Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Stig Düring Friis
- Center for Insoluble Protein Structures (inSPIN), Department of Chemistry, Aarhus University, Aarhus, Denmark
| | - Alberto Martínez Serrano
- Department of Molecular Biology and Center of Molecular Biology Severo Ochoa, University Autonoma Madrid-C.S.I.C Campus Cantoblanco, Madrid, Spain
| | - Morten Blaabjerg
- Department of Neurology, Zealand University Hospital, Roskilde, Denmark
| | - Bjarne Winther Kristensen
- Department of Pathology, Odense University Hospital, Denmark & Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Troels Skrydstrup
- Center for Insoluble Protein Structures (inSPIN), Department of Chemistry, Aarhus University, Aarhus, Denmark
| | - Jan Bert Gramsbergen
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Helena L. A. Vieira
- Instituto de Biologia Experimental e Tecnológica (IBET), Oeiras, Portugal
- CEDOC, NOVA Medical School/Faculdade de Ciência Médicas, Universidade Nova de Lisboa, Lisboa, Portugal
| | - Morten Meyer
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
- Department of Neurology, Zealand University Hospital, Roskilde, Denmark
| |
Collapse
|
44
|
Saghazadeh A, Ferrari CC, Rezaei N. Deciphering variability in the role of interleukin-1β in Parkinson's disease. Rev Neurosci 2018; 27:635-50. [PMID: 27166719 DOI: 10.1515/revneuro-2015-0059] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2015] [Accepted: 04/01/2016] [Indexed: 12/16/2022]
Abstract
Although the role of inflammation in neurodegeneration has been well acknowledged, less is known on the issue of each cytokine in specific neurodegenerative diseases. In this review, we will present evidence elucidating that interleukin-1β (IL-1β) has a multi-faceted character in pathogenesis of Parkinson's disease, which is a progressive neurodegenerative disorder. Increased levels of IL-1β were found in PD patients. Besides, PD symptoms were observed in IL-1β wild-type, but not deficient, animals. These lines of evidence suggest that IL-1β may contribute to the initiation or progression of PD. On the other hand, some studies reported decreased levels of IL-1β in PD patients. Also, genetic studies provided evidence suggesting that IL-1β may protect individuals against PD. Presumably, the broad range of IL-1β role is due to its interaction with both upstream and downstream mediators. Differences in IL-1β levels could be because of glia population (i.e. microglia and astrocytes), mitogen-activated protein kinase and nuclear factor κ light-chain-enhancer of activated B cells signaling pathways, and several mediators (including cyclooxygenase, neurotrophic factors, reactive oxygen species, caspases, heme oxygenase-1, and matrix metalloproteinases). Although far from practice at this point, unraveling theoretical therapeutic targets based on the up-down IL-1β neuroweb could facilitate the development of strategies that are likely to be used for pharmaceutical designs of anti-neurodegenerative drugs of the future.
Collapse
|
45
|
De M, Ghosh S, Sen T, Shadab M, Banerjee I, Basu S, Ali N. A Novel Therapeutic Strategy for Cancer Using Phosphatidylserine Targeting Stearylamine-Bearing Cationic Liposomes. MOLECULAR THERAPY. NUCLEIC ACIDS 2017; 10:9-27. [PMID: 29499959 PMCID: PMC5723379 DOI: 10.1016/j.omtn.2017.10.019] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Accepted: 10/26/2017] [Indexed: 02/07/2023]
Abstract
There is a pressing need for a ubiquitously expressed antigen or receptor on the tumor surface for successful mitigation of the deleterious side effects of chemotherapy. Phosphatidylserine (PS), normally constrained to the intracellular surface, is exposed on the external surface of tumors and most tumorigenic cell lines. Here we report that a novel PS-targeting liposome, phosphatidylcholine-stearylamine (PC-SA), induced apoptosis and showed potent anticancer effects as a single agent against a majority of cancer cell lines. We experimentally proved that this was due to a strong affinity for and direct interaction of these liposomes with PS. Complexation of the chemotherapeutic drugs doxorubicin and camptothecin in these vesicles demonstrated a manyfold enhancement in the efficacies of the drugs both in vitro and across three advanced tumor models without any signs of toxicity. Both free and drug-loaded liposomes were maximally confined to the tumor site with low tissue concentration. These data indicate that PC-SA is a unique and promising liposome that, alone and as a combination therapy, has anticancer potential across a wide range of cancer types.
Collapse
Affiliation(s)
- Manjarika De
- Infectious Diseases and Immunology Division, Indian Institute of Chemical Biology, Kolkata, West Bengal, India
| | - Sneha Ghosh
- Infectious Diseases and Immunology Division, Indian Institute of Chemical Biology, Kolkata, West Bengal, India
| | - Triparna Sen
- Infectious Diseases and Immunology Division, Indian Institute of Chemical Biology, Kolkata, West Bengal, India
| | - Md Shadab
- Infectious Diseases and Immunology Division, Indian Institute of Chemical Biology, Kolkata, West Bengal, India
| | - Indranil Banerjee
- Infectious Diseases and Immunology Division, Indian Institute of Chemical Biology, Kolkata, West Bengal, India
| | - Santanu Basu
- Department of Oncology, ESI Hospital, Kolkata, West Bengal, India
| | - Nahid Ali
- Infectious Diseases and Immunology Division, Indian Institute of Chemical Biology, Kolkata, West Bengal, India.
| |
Collapse
|
46
|
Zhao L, Feng Y, Shi A, Zhang L, Guo S, Wan M. Neuroprotective Effect of Low-Intensity Pulsed Ultrasound Against MPP +-Induced Neurotoxicity in PC12 Cells: Involvement of K2P Channels and Stretch-Activated Ion Channels. ULTRASOUND IN MEDICINE & BIOLOGY 2017; 43:1986-1999. [PMID: 28583325 DOI: 10.1016/j.ultrasmedbio.2017.04.020] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Revised: 04/18/2017] [Accepted: 04/20/2017] [Indexed: 06/07/2023]
Abstract
Parkinson's disease is the second most common neurodegenerative disease. It is characterized by the loss of dopaminergic neurons in the substantia nigra pars compacta. 1-Methyl-4-phenylpyridinium (MPP+) is a dopaminergic neuronal toxin that is widely used in constructing Parkinson's disease models in vitro. Low-intensity pulsed ultrasound (LIPUS) is a non-invasive therapeutic approach that has neuromodulation and neuroprotective effects in the central neural system; however, whether LIPUS can provide protection for dopaminergic neurons against MPP+-induced neurocytotoxicity remains unknown. In this study, we found that pre-treatment with LIPUS (1 MHz, 50 mW/cm2, 20% duty cycle and 100-Hz pulse repetition frequency, 10 min) inhibited MPP+-induced neurotoxicity and mitochondrial dysfunction in PC12 cells. LIPUS decreased MPP+-induced oxidative stress by modulating antioxidant proteins, including thioredoxin-1 and heme oxygenase-1, and prevented neurocytotoxicity via the phosphoinositide 3-kinase (PI3K)-Akt and ERK1/2 pathways. Furthermore, these beneficial effects were attributed to the activation of K2P channels and stretch-activated ion channels by LIPUS. These data indicate that LIPUS protects neuronal cells from MPP+-induced cell death through the K2P channel- and stretch-activated ion channel-mediated downstream pathways. The data also suggest that LIPUS could be a promising therapeutic method in halting or retarding the degeneration of dopaminergic neurons in Parkinson's disease in a non-invasive manner.
Collapse
Affiliation(s)
- Lu Zhao
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, Department of Biomedical Engineering, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, China
| | - Yi Feng
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, Department of Biomedical Engineering, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, China.
| | - Aiwei Shi
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, Department of Biomedical Engineering, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, China
| | - Lei Zhang
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, Department of Biomedical Engineering, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, China
| | - Shifang Guo
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, Department of Biomedical Engineering, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, China
| | - Mingxi Wan
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, Department of Biomedical Engineering, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
47
|
Lin JG, Chen CJ, Yang HB, Chen YH, Hung SY. Electroacupuncture Promotes Recovery of Motor Function and Reduces Dopaminergic Neuron Degeneration in Rodent Models of Parkinson's Disease. Int J Mol Sci 2017; 18:ijms18091846. [PMID: 28837077 PMCID: PMC5618495 DOI: 10.3390/ijms18091846] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Revised: 08/17/2017] [Accepted: 08/18/2017] [Indexed: 12/12/2022] Open
Abstract
Parkinson’s disease (PD) is a common neurodegenerative disease. The pathological hallmark of PD is a progressive loss of dopaminergic neurons in the substantia nigra (SN) pars compacta in the brain, ultimately resulting in severe striatal dopamine deficiency and the development of primary motor symptoms (e.g., resting tremor, bradykinesia) in PD. Acupuncture has long been used in traditional Chinese medicine to treat PD for the control of tremor and pain. Accumulating evidence has shown that using electroacupuncture (EA) as a complementary therapy ameliorates motor symptoms of PD. However, the most appropriate timing for EA intervention and its effect on dopamine neuronal protection remain unclear. Thus, this study used the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-lesioned mouse model (systemic-lesioned by intraperitoneal injection) and the 1-methyl-4-phenylpyridinium (MPP+)-lesioned rat model (unilateral-lesioned by intra-SN infusion) of PD, to explore the therapeutic effects and mechanisms of EA at the GB34 (Yanglingquan) and LR3 (Taichong) acupoints. We found that EA increased the latency to fall from the accelerating rotarod and improved striatal dopamine levels in the MPTP studies. In the MPP+ studies, EA inhibited apomorphine induced rotational behavior and locomotor activity, and demonstrated neuroprotective effects via the activation of survival pathways of Akt and brain-derived neurotrophic factor (BDNF) in the SN region. In conclusion, we observed that EA treatment reduces motor symptoms of PD and dopaminergic neurodegeneration in rodent models, whether EA is given as a pretreatment or after the initiation of disease symptoms. The results indicate that EA treatment may be an effective therapy for patients with PD.
Collapse
Affiliation(s)
- Jaung-Geng Lin
- School of Chinese Medicine, China Medical University, Taichung 40402, Taiwan.
| | - Chao-Jung Chen
- Graduate Institute of Integrated Medicine, College of Chinese Medicine, China Medical University, Taichung 40402, Taiwan.
| | - Han-Bin Yang
- Graduate Institute of Acupuncture Science, China Medical University, Taichung 40402, Taiwan.
| | - Yi-Hung Chen
- Graduate Institute of Acupuncture Science, China Medical University, Taichung 40402, Taiwan.
- Department of Photonics and Communication Engineering, Asia University, Taichung 41354, Taiwan.
- Research Center for Chinese Medicine and Acupuncture, China Medical University, Taichung 40402, Taiwan.
| | - Shih-Ya Hung
- Graduate Institute of Acupuncture Science, China Medical University, Taichung 40402, Taiwan.
- Division of Colorectal Surgery, China Medical University Hospital, Taichung 40447, Taiwan.
| |
Collapse
|
48
|
Tran NQV, Nguyen AN, Takabe K, Yamagata Z, Miyake K. Pre-treatment with amitriptyline causes epigenetic up-regulation of neuroprotection-associated genes and has anti-apoptotic effects in mouse neuronal cells. Neurotoxicol Teratol 2017; 62:1-12. [PMID: 28511916 DOI: 10.1016/j.ntt.2017.05.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Revised: 05/09/2017] [Accepted: 05/11/2017] [Indexed: 12/12/2022]
Abstract
Antidepressants, such as imipramine and fluoxetine, are known to alter gene expression patterns by inducing changes in the epigenetic status of neuronal cells. There is also some evidence for the anti-apoptotic effect of various groups of antidepressants; however, this effect is complicated and cell-type dependent. Antidepressants of the tricyclic group, in particular amitriptyline, have been suggested to be beneficial in the treatment of neurodegenerative disorders. We examined whether amitriptyline exerts an anti-apoptotic effect via epigenetic mechanisms. Using DNA microarray, we analyzed global gene expression in mouse primary cultured neocortical neurons after treatment with amitriptyline and imipramine. The neuroprotection-associated genes, activating transcription factor 3 (Atf3) and heme oxygenase 1 (Hmox1), were up-regulated at both mRNA and protein levels by treatment with amitriptyline. Quantitative chromatin immunoprecipitation assay revealed that amitriptyline increased enrichments of trimethylation of histone H3 lysine 4 in the promoter regions of Atf3 and Hmox1 and acetylation of histone H3 lysine 9 in the promoter regions of Atf3, which indicate an active epigenetic status. Amitriptyline pre-treatment attenuated 1-methyl-4-phenylpyridinium ion (MPP+)- or amyloid β peptide 1-42 (Aβ1-42)-induced neuronal cell death and inhibited the activation of extracellular signal-regulated kinase 1 and 2 (ERK1/2). We found that Atf3 and Hmox1 were also up-regulated after Aβ1-42 treatment, and were further increased when pre-treated with amitriptyline. Interestingly, the highest up-regulation of Atf3 and Hmox1, at least at mRNA level, was observed after co-treatment with Aβ1-42 and amitriptyline, together with the loss of the neuroprotective effect. These findings suggest preconditioning and neuroprotective effects of amitriptyline; however, further investigations are needed for clarifying the contribution of epigenetic up-regulation of Atf3 and Hmox1 genes.
Collapse
Affiliation(s)
- Nguyen Quoc Vuong Tran
- Department of Health Sciences, Graduate School of Interdisciplinary Research, University of Yamanashi, 1110, Shimokato, Chuo, Yamanashi 409-3898, Japan
| | - An Nghia Nguyen
- Department of Health Sciences, Graduate School of Interdisciplinary Research, University of Yamanashi, 1110, Shimokato, Chuo, Yamanashi 409-3898, Japan
| | - Kyoko Takabe
- Department of Health Sciences, Graduate School of Interdisciplinary Research, University of Yamanashi, 1110, Shimokato, Chuo, Yamanashi 409-3898, Japan
| | - Zentaro Yamagata
- Department of Health Sciences, Graduate School of Interdisciplinary Research, University of Yamanashi, 1110, Shimokato, Chuo, Yamanashi 409-3898, Japan
| | - Kunio Miyake
- Department of Health Sciences, Graduate School of Interdisciplinary Research, University of Yamanashi, 1110, Shimokato, Chuo, Yamanashi 409-3898, Japan.
| |
Collapse
|
49
|
Anti-oxidative effects of 4-hydroxybenzyl alcohol in astrocytes confer protective effects in autocrine and paracrine manners. PLoS One 2017; 12:e0177322. [PMID: 28489907 PMCID: PMC5425201 DOI: 10.1371/journal.pone.0177322] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Accepted: 04/25/2017] [Indexed: 12/26/2022] Open
Abstract
4-Hydroxybenzyl alcohol (4-HBA) is an important phenolic constituent of Gastrodia elata Blume (GEB), a traditional herbal medicine used in East Asia. Many activities have been reported to underlie the beneficial effects of 4-HBA in the brain, and in particular, its anti-inflammatory, anti-oxidative, and anti-zinc-toxic effects have been implicated in the postischemic brain. Here, the authors investigated the anti-oxidative effect of 4-HBA on astrocytes and sought to identify the underlying molecular mechanisms involved. 4-HBA dose-dependently suppressed H2O2-induced astrocyte cell death. More specifically, pre-incubation of C6 cells (an astrocyte cell line) with 100 μM 4-HBA for 6 hrs increased survival when cells were treated with H2O2 (100 μM, 1 hr) from 54.2±0.7% to 85.9±1.5%. In addition, 4-HBA was found to up-regulate and activate Nrf2, and subsequently, to induce the expressions of several anti-oxidative genes, such as, HO-1, NQO1, and GCLM. Notably, HO-1 was induced by 3.4-fold in 4-HBA-treated C6 cells, and siRNA-mediated HO-1 knockdown demonstrated that Nrf2 activation and HO-1 induction were responsible for the observed cytoprotective effect of 4-HBA. ERK and Akt signaling pathways were activated by 4-HBA in C6 cells, suggesting their involvements in protective effect of 4-HBA. In addition, 4-HBA-conditioned astrocyte culture medium was found to have neuroprotective effects on primary neuronal cultures or fresh C6 cells exposed to oxidative stress, and these effects seemed to be mediated by glial cell line-derived neurotrophic factor (GDNF) and vascular endothelial growth factor (VEGF), which both accumulated in 4-HBA-treated astrocyte culture media. Thus, the 4-HBA-mediated activation of Nrf2 and induction of HO-1 in astrocytes were found to act via autocrine and paracrine mechanisms to confer protective effects. Furthermore, given the pleiotropic effects of 4-HBA with respect to its targeting of various brain cell types and functions, it would appear that 4-HBA has therapeutic potential for the prevention and amelioration of various brain diseases.
Collapse
|
50
|
Kuang L, Cao X, Lu Z. Baicalein Protects against Rotenone-Induced Neurotoxicity through Induction of Autophagy. Biol Pharm Bull 2017; 40:1537-1543. [DOI: 10.1248/bpb.b17-00392] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Lianghong Kuang
- Department of Neurology, Renmin Hospital of Wuhan University
| | - Xiongbin Cao
- Department of Neurology, Renmin Hospital of Wuhan University
| | - Zuneng Lu
- Department of Neurology, Renmin Hospital of Wuhan University
| |
Collapse
|