1
|
Wei X, Wang S, Zhang M, Yan Y, Wang Z, Wei W, Tuo H, Wang Z. Alterations of diffusion kurtosis measures in gait-related white matter in the "ON-OFF state" of Parkinson's disease. Chin Med J (Engl) 2025; 138:1094-1102. [PMID: 40012092 PMCID: PMC12068762 DOI: 10.1097/cm9.0000000000003486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Indexed: 02/28/2025] Open
Abstract
BACKGROUND Gait impairment is closely related to quality of life in patients with Parkinson's disease (PD). This study aimed to explore alterations in brain microstructure in PD patients and healthy controls (HCs) and to identify the correlation of gait impairment in the ON and OFF states of patients with PD, respectively. METHODS We enrolled 24 PD patients and 29 HCs from the Movement Disorders Program at Beijing Friendship Hospital Capital Medical University between 2019 and 2020. We acquired magnetic resonance imaging (MRI) scans and processed the diffusion kurtosis imaging (DKI) images. Preprocessing of diffusion-weighted data was performed with Mrtrix3 software, using a directional distribution function to track participants' main white matter fiber bundles. Demographic and clinical characteristics were recorded. Quantitative gait and clinical scales were used to assess the status of medication ON and OFF in PD patients. RESULTS The axial kurtosis (AK), mean kurtosis (MK), and radial kurtosis (RK) of five specific white matter fiber tracts, the bilateral corticospinal tract, left superior longitudinal fasciculus, left anterior thalamic radiation, forceps minor, and forceps major were significantly higher in PD patients compared to HCs. Additionally, the MK values were negatively correlated with Timed Up and Go Test (TUG) scores in both the ON and OFF in PD patients. Within the PD group, higher AK, MK, and RK values, whether the patients were ON or OFF, were associated with better gait performance (i.e., higher velocity and stride length). CONCLUSIONS PD exhibits characteristic regional patterns of white matter microstructural degradation. Correlations between objective gait parameters and DKI values suggest that dopamine-responsive gait function depends on preserved white matter microstructure. DKI-based Tract-Based Spatial Statistics (TBSS) analysis may serve as a tool for evaluating PD-related motor impairments (e.g., gait impairment) and could yield potential neuroimaging biomarkers.
Collapse
Affiliation(s)
- Xuan Wei
- Department of Radiology, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Shiya Wang
- Department of Neurology, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Mingkai Zhang
- Department of Neurology, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Ying Yan
- Department of Radiology, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Zheng Wang
- Department of Radiology, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Wei Wei
- Division of Science and Technology, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Houzhen Tuo
- Department of Neurology, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Zhenchang Wang
- Department of Radiology, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| |
Collapse
|
2
|
Meccariello R, Bellenchi GC, Pulcrano S, D’Addario SL, Tafuri D, Mercuri NB, Guatteo E. Neuronal dysfunction and gene modulation by non-coding RNA in Parkinson's disease and synucleinopathies. Front Cell Neurosci 2024; 17:1328269. [PMID: 38249528 PMCID: PMC10796818 DOI: 10.3389/fncel.2023.1328269] [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: 10/26/2023] [Accepted: 12/07/2023] [Indexed: 01/23/2024] Open
Abstract
Over the last few decades, emerging evidence suggests that non-coding RNAs (ncRNAs) including long-non-coding RNA (lncRNA), microRNA (miRNA) and circular-RNA (circRNA) contribute to the molecular events underlying progressive neuronal degeneration, and a plethora of ncRNAs have been identified significantly misregulated in many neurodegenerative diseases, including Parkinson's disease and synucleinopathy. Although a direct link between neuropathology and causative candidates has not been clearly established in many cases, the contribution of ncRNAs to the molecular processes leading to cellular dysfunction observed in neurodegenerative diseases has been addressed, suggesting that they may play a role in the pathophysiology of these diseases. Aim of the present Review is to overview and discuss recent literature focused on the role of RNA-based mechanisms involved in different aspects of neuronal pathology in Parkinson's disease and synucleinopathy models.
Collapse
Affiliation(s)
- Rosaria Meccariello
- Department of Medical and Movement Sciences and Wellness, University of Naples Parthenope, Naples, Italy
| | - Gian Carlo Bellenchi
- Institute of Genetics and Biophysics, CNR, Naples, Italy
- Experimental Neurology Laboratory, Santa Lucia Foundation IRCCS, Rome, Italy
| | | | - Sebastian Luca D’Addario
- Experimental Neurology Laboratory, Santa Lucia Foundation IRCCS, Rome, Italy
- Computational and Translational Neuroscience Laboratory, Institute of Cognitive Sciences and Technologies, CNR, Rome, Italy
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, United States
| | - Domenico Tafuri
- Department of Medical and Movement Sciences and Wellness, University of Naples Parthenope, Naples, Italy
| | - Nicola B. Mercuri
- Experimental Neurology Laboratory, Santa Lucia Foundation IRCCS, Rome, Italy
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, United States
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Ezia Guatteo
- Department of Medical and Movement Sciences and Wellness, University of Naples Parthenope, Naples, Italy
- Experimental Neurology Laboratory, Santa Lucia Foundation IRCCS, Rome, Italy
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, United States
| |
Collapse
|
3
|
Huang S, Dong Y, Zhao J. The mean kurtosis (MK) is more sensitive diagnostic biomarker than fractional anisotropy (FA) for Parkinson's disease: A diagnostic performance study and meta-analysis. Medicine (Baltimore) 2022; 101:e31312. [PMID: 36397320 PMCID: PMC9666087 DOI: 10.1097/md.0000000000031312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND The mean kurtosis (MK) and fractional anisotropy (FA) in patients of Parkinson's disease (PD) are usually measured by diffusion kurtosis imaging (DKI) and diffusion tensor imaging (DTI), separately. METHODS In this study we perform a meta-analysis to discuss which noninvasive biomarker is more advantageous for PD, MK, or FA. Databases including Medline via PubMed, the Cochrane Central Register of Controlled Trials, Embase via OVID and China National Knowledge Infrastructure. Databases are searched up to December 31st, 2019. Four brain regions are identified for analysis based on data extracted from articles. RESULTS The articles contain 5 trials with 274 total PD patients and 189 healthy controls (HCs). The results show not only significantly higher MK values of putamen, caudate, globus pallidus in PD compared to that of HCs (weighted mean difference [WMD] = 0.06, 95% CI = 0.02-0.09, P = .002, WMD = 0.03, 95% CI = 0.01-0.067, P = .01, WMD = 0.18, 95% CI = 0.11-0.24, P < .00001), but also a significantly higher FA in caudate of PD compared to HCs (WMD = 0.02, 95% CI = 0.00-0.03, P = .006). CONCLUSION This indicates that the sharp difference detected between PD patients and HCs can be detected by DKI and DTI. By further discussing results, we found that MK could be more sensitive diagnostic biomarker than FA toward PD diagnosis.
Collapse
Affiliation(s)
- Songtao Huang
- Department of Radiology, Guang’an People’s Hospital, Guangan, Sichuan Province, P. R. China
| | - Yanchao Dong
- Department of Interventional Treatment, Qinhuangdao Municipal, Qinhuangdao, Hebei Province, P. R. China
| | - Jiaying Zhao
- Department of Internal Medicine, Guang’an People’s Hospital, Guangan, Sichuan Province, P. R. China
- * Correspondence: Jiaying Zhao, Department of Internal Medicine, Guang’an People’s Hospital, No. 1, Section 4, Binhe Road, Guangan 638500, Sichuan Province, P. R. China (e-mail: )
| |
Collapse
|
4
|
Dysregulated miRNAs as Biomarkers and Therapeutical Targets in Neurodegenerative Diseases. J Pers Med 2022; 12:jpm12050770. [PMID: 35629192 PMCID: PMC9143965 DOI: 10.3390/jpm12050770] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 05/06/2022] [Accepted: 05/07/2022] [Indexed: 12/20/2022] Open
Abstract
Alzheimer’s disease (AD), Parkinson’s disease (PD), and Amyotrophic Lateral Sclerosis (ALS) are representative neurodegenerative diseases (NDs) characterized by degeneration of selective neurons, as well as the lack of effective biomarkers and therapeutic treatments. In the last decade, microRNAs (miRNAs) have gained considerable interest in diagnostics and therapy of NDs, owing to their aberrant expression and their ability to target multiple molecules and pathways. Here, we provide an overview of dysregulated miRNAs in fluids (blood or cerebrospinal fluid) and nervous tissue of AD, PD, and ALS patients. By emphasizing those that are commonly dysregulated in these NDs, we highlight their potential role as biomarkers or therapeutical targets and describe the use of antisense oligonucleotides as miRNA therapies.
Collapse
|
5
|
Jiang L, Wang X. The miR-133b/brefeldin A-inhibited guanine nucleotide-exchange protein 1 (ARFGEF1) axis represses proliferation, invasion, and migration in cervical cancer cells. Bioengineered 2022; 13:3323-3332. [PMID: 35048795 PMCID: PMC8973932 DOI: 10.1080/21655979.2022.2027063] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2022] Open
Abstract
Cervical cancer is a common gynecological malignancy, and miR-133b is an abnormally expressed cervical cancer gene, which suggests that miR-133b may be involved in the occurrence and development of cervical cancer. However, the underlying mechanism is still unclear. miR-133b was overexpressed or silenced in the cervical cancer cell line C33A. Brefeldin A-inhibited guanine nucleotide-exchange protein 1 (ARFGEF1) was combined with overexpression of miR-133b in C33A cells. Cell Counting Kit-8, clone formation, and Transwell assays were performed to determine the influence of miR-133b and ARFGEF1 on clone formation, proliferation, migration, and invasion of C33A cells. The interaction between miR-133b and ARFGEF1 was verified using a luciferase reporter assay. Finally, the mRNA and protein expression of miR-133b and ARFGEF1 in the tumor and adjacent normal tissues of cervical cancer patients was detected by real-time quantitative PCR, Western blotting, and immunohistochemistry. The results indicated that miR-133b up-regulation suppressed the proliferation, invasion, migration, and clone formation abilities of C33A cells (P < 0.05). However, silence of miR-133b promoted the proliferation, invasion, and migration of C33A cells (P < 0.05). Clone formation ability of C33A cells was also elevated by miR-133b deficiency (P < 0.05). Moreover, miR-133b interacted with ARFGEF1 and repressed ARFGEF1 expression in C33A cells (P < 0.05). ARFGEF1 overexpression weakened miR-133b overexpression-mediated inhibition of proliferation, invasion, and migration of C33A cells (P < 0.05). miR-133b expression was decreased, and ARFGEF1 was up-regulated in tumor tissues of cervical cancer patients (P < 0.05). All results revealed that miR-133b suppresses cervical cancer progression by inhibiting proliferation, invasion, and migration of cervical cancer cells via targeting ARFGEF1. Thus, our study determined the mechanism of miR-133b in cervical cancer, and confirmed miR-133b/ARFGEF1 may become a potential therapeutic target for cervical cancer.
Collapse
Affiliation(s)
- Lingling Jiang
- Department of Gynaecology and Obstetrics, First People's Hospital of Wenling, Wenling, Zhejing, China
| | - Xuexin Wang
- Department of Gynaecology and Obstetrics, First People's Hospital of Wenling, Wenling, Zhejing, China
| |
Collapse
|
6
|
Daniel NH, Aravind A, Thakur P. Are ion channels potential therapeutic targets for Parkinson's disease? Neurotoxicology 2021; 87:243-257. [PMID: 34699791 DOI: 10.1016/j.neuro.2021.10.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 10/15/2021] [Accepted: 10/21/2021] [Indexed: 01/31/2023]
Abstract
Parkinson's disease (PD) is primarily associated with the progressive neurodegeneration of the dopaminergic neurons in the substantia nigra region of the brain. The resulting motor symptoms are managed with the help of dopamine replacement therapies. However, these therapeutics do not prevent the neurodegeneration underlying the disease and therefore lose their effectiveness in managing disease symptoms over time. Thus, there is an urgent need to develop newer therapeutics for the benefit of patients. The release of dopamine and the firing activity of substantia nigra neurons is regulated by several ion channels that act in concert. Dysregulations of these channels cause the aberrant movement of various ions in the intracellular milieu. This eventually leads to disruption of intracellular signalling cascades, alterations in cellular homeostasis, and bioenergetic deficits. Therefore, ion channels play a central role in driving the high vulnerability of dopaminergic neurons to degenerate during PD. Targeting ion channels offers an attractive mechanistic strategy to combat the process of neurodegeneration. In this review, we highlight the evidence pointing to the role of various ion channels in driving the PD processes. In addition, we also discuss the various drugs or compounds that target the ion channels and have shown neuroprotective potential in the in-vitro and in-vivo models of PD. We also discuss the current clinical status of various drugs targeting the ion channels in the context of PD.
Collapse
Affiliation(s)
- Neha Hanna Daniel
- School of Biology, Indian Institute of Science Education and Research (IISER)-Thiruvananthapuram, Kerala, 695551, India
| | - Ananya Aravind
- School of Biology, Indian Institute of Science Education and Research (IISER)-Thiruvananthapuram, Kerala, 695551, India
| | - Poonam Thakur
- School of Biology, Indian Institute of Science Education and Research (IISER)-Thiruvananthapuram, Kerala, 695551, India.
| |
Collapse
|
7
|
Zhang Y, Liu J, Xie C, Wu P. Overexpression of miR-133b protects against isoflurane-induced learning and memory impairment. Exp Ther Med 2021; 22:1207. [PMID: 34584552 DOI: 10.3892/etm.2021.10641] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Accepted: 11/17/2020] [Indexed: 12/21/2022] Open
Abstract
A number of microRNAs (miRs) have been identified as being involved in the regulation of anesthesia-induced cognitive impairment. The aim of the present study was to investigated the role and potential mechanism of miR-133b in isoflurane-induced learning and memory impairment. An animal model of isoflurane exposure was established using neonatal Sprague-Dawley rats. The rats were trained for Morris water maze (MWM) testing to assess their spatial learning and memory ability. Reverse transcription-quantitative polymerase chain reaction was used for the measurement of miR-133b expression in hippocampal tissues and primary hippocampal neuron cultures. Cell viability was assessed using a Cell Counting Kit-8 assay, and flow cytometric analysis was used to determine the rate of apoptosis. The MWM test results indicated that during the training period, the time required to locate the platform was significantly increased for rats exposed to isoflurane, and this increased time was reduced by the overexpression of miR-133b. The results of a probe trial indicated that isoflurane exposure increased escape latency and decreased the time spent in the platform area for isoflurane-treated rats; however, these effects were reversed by the injection of miR-133b agomir. The in vitro experiments demonstrated that the overexpression of miR-133b attenuated the reduction of neuronal cell viability induced by isoflurane, and inhibited the isoflurane-induced apoptosis of hippocampal neurons. In conclusion, the present study revealed that the overexpression of miR-133b attenuated isoflurane-induced learning and memory impairment in rats. Furthermore, miR-133b overexpression promoted the viability of hippocampal neurons and their resistance to apoptosis when exposed to isoflurane.
Collapse
Affiliation(s)
- Yu Zhang
- Department of Anesthesiology, Jining No. 1 People's Hospital, Jining, Shandong 272000, P.R. China
| | - Jinyong Liu
- Department of Anesthesiology, Jining No. 1 People's Hospital, Jining, Shandong 272000, P.R. China
| | - Cuili Xie
- Department of Anesthesiology, Jining No. 1 People's Hospital, Jining, Shandong 272000, P.R. China
| | - Pingping Wu
- Clinical Laboratory, Jining No. 1 People's Hospital, Jining, Shandong 272000, P.R. China
| |
Collapse
|
8
|
Srivastava P, Kane A, Harrison C, Levin M. A Meta-Analysis of Bioelectric Data in Cancer, Embryogenesis, and Regeneration. Bioelectricity 2021; 3:42-67. [PMID: 34476377 DOI: 10.1089/bioe.2019.0034] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Developmental bioelectricity is the study of the endogenous role of bioelectrical signaling in all cell types. Resting potentials and other aspects of ionic cell physiology are known to be important regulatory parameters in embryogenesis, regeneration, and cancer. However, relevant quantitative measurement and genetic phenotyping data are distributed throughout wide-ranging literature, hampering experimental design and hypothesis generation. Here, we analyze published studies on bioelectrics and transcriptomic and genomic/phenotypic databases to provide a novel synthesis of what is known in three important aspects of bioelectrics research. First, we provide a comprehensive list of channelopathies-ion channel and pump gene mutations-in a range of important model systems with developmental patterning phenotypes, illustrating the breadth of channel types, tissues, and phyla (including man) in which bioelectric signaling is a critical endogenous aspect of embryogenesis. Second, we perform a novel bioinformatic analysis of transcriptomic data during regeneration in diverse taxa that reveals an electrogenic protein to be the one common factor specifically expressed in regeneration blastemas across Kingdoms. Finally, we analyze data on distinct Vmem signatures in normal and cancer cells, revealing a specific bioelectrical signature corresponding to some types of malignancies. These analyses shed light on fundamental questions in developmental bioelectricity and suggest new avenues for research in this exciting field.
Collapse
Affiliation(s)
- Pranjal Srivastava
- Rye High School, Rye, New York, USA; Current Affiliation: College of Chemistry, University of California, Berkeley, Berkeley, California, USA
| | - Anna Kane
- Department of Biology, Allen Discovery Center, Tufts University, Medford, Massachusetts, USA
| | - Christina Harrison
- Department of Biology, Allen Discovery Center, Tufts University, Medford, Massachusetts, USA
| | - Michael Levin
- Department of Biology, Allen Discovery Center, Tufts University, Medford, Massachusetts, USA
| |
Collapse
|
9
|
Song N, Zhu H, Xu R, Liu J, Fang Y, Zhang J, Ding J, Hu G, Lu M. Induced Expression of kir6.2 in A1 Astrocytes Propagates Inflammatory Neurodegeneration via Drp1-dependent Mitochondrial Fission. Front Pharmacol 2021; 11:618992. [PMID: 33584303 PMCID: PMC7876245 DOI: 10.3389/fphar.2020.618992] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Accepted: 12/24/2020] [Indexed: 12/20/2022] Open
Abstract
Glia-mediated inflammatory processes are crucial in the pathogenesis of Parkinson’s disease (PD). As the most abundant cells of the brain and active participants in neuroinflammatory responses, astrocytes largely propagate inflammatory signals and amplify neuronal loss. Hence, intensive control of astrocytic activation is necessary to prevent neurodegeneration. In this study, we report that the astrocytic kir6.2, as a abnormal response after inflammatory stimuli, promotes the reactivity of A1 neurotoxic astrocytes. Using kir6.2 knockout (KO) mice, we find reversal effects of kir6.2 deficiency on A1-like astrocyte activation and death of dopaminergic neurons in lipopolysaccharide (LPS)-induced mouse models for PD. Further in vitro experiments show that aberrant kir6.2 expression induced by inflammatory irritants in astrocytes mediates the dynamin-related protein 1 (Drp1)-dependent excessive mitochondrial fragmentation and results in mitochondrial malfunctions. By deleting kir6.2, astrocytic activation is reduced and astrocytes-derived neuronal injury is prevented. We therefore conclude that astrocytic kir6.2 can potentially elucidate the pathology of PD and promote the development of therapeutic strategies for PD.
Collapse
Affiliation(s)
- Nanshan Song
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing, China
| | - Hong Zhu
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing, China
| | - Rong Xu
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing, China
| | - Jiaqi Liu
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing, China
| | - Yinquan Fang
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing, China
| | - Jing Zhang
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing, China
| | - Jianhua Ding
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing, China
| | - Gang Hu
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing, China.,Department of Pharmacology, Nanjing University of Chinese Medicine, Nanjing, China
| | - Ming Lu
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing, China.,Neuroprotective Drug Discovery Key Laboratory, Department of Pharmacology, Nanjing Medical University, Nanjing, China
| |
Collapse
|
10
|
Danbaran GR, Aslani S, Sharafkandi N, Hemmatzadeh M, Hosseinzadeh R, Azizi G, Jadidi-Niaragh F, Babaie F, Mohammadi H. How microRNAs affect the PD-L1 and its synthetic pathway in cancer. Int Immunopharmacol 2020; 84:106594. [PMID: 32416456 DOI: 10.1016/j.intimp.2020.106594] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 04/27/2020] [Accepted: 05/08/2020] [Indexed: 12/17/2022]
Abstract
Programmed cell death-ligand 1 (PD-L1) is a glycoprotein that is expressed on the cell surface of both hematopoietic and nonhematopoietic cells. PD-L1 play a role in the immune tolerance and protect self-tissues from immune system attack. Dysfunction of this molecule has been highlighted in the pathogenesis of tumors, autoimmunity, and infectious disorders. MicroRNAs (miRNAs) are endogenous molecules that are classified as small non-coding RNA with approximately 20-22 nucleotides (nt) length. The function of miRNAs is based on complementary interactions with target mRNA via matching completely or incompletely. The result of this function is decay of the target mRNA or preventing mRNA translation. In the past decades, several miRNAs have been discovered which play an important role in the regulation of PD-L1 in various malignancies. In this review, we discuss the effect of miRNAs on PD-L1 expression and consider the effect of miRNAs on the synthetic pathway of PD-L1, especially during cancers.
Collapse
Affiliation(s)
| | - Saeed Aslani
- Department of Medical Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Nadia Sharafkandi
- Student Research Committee, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Maryam Hemmatzadeh
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ramin Hosseinzadeh
- Department of Medical Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Gholamreza Azizi
- Non-Communicable Diseases Research Center, Alborz University of Medical Sciences, Karaj, Iran
| | - Farhad Jadidi-Niaragh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Farhad Babaie
- Cellular and Molecular Research Center, Urmia University of Medical Sciences, Urmia, Iran
| | - Hamed Mohammadi
- Non-Communicable Diseases Research Center, Alborz University of Medical Sciences, Karaj, Iran; Department of Immunology, School of Medicine, Alborz University of Medical Sciences, Karaj, Iran.
| |
Collapse
|
11
|
Zhang L, Zheng Y, Xie J, Shi L. Potassium channels and their emerging role in parkinson's disease. Brain Res Bull 2020; 160:1-7. [PMID: 32305406 DOI: 10.1016/j.brainresbull.2020.04.004] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Revised: 03/21/2020] [Accepted: 04/05/2020] [Indexed: 12/21/2022]
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disorder, which is associated with a selective loss of dopaminergic neurons in the substantia nigra (SN) and a reduction of dopamine in the striatum. Recently, ion channel dysfunction has been considered a reason for the pathogenesis of PD. Potassium (K+) channels are widespread in the central nervous system, and play key roles in modulating cellular excitability, synaptic transmission, and neurotransmitter release. Based on recent studies and data, we propose that K+ channels may be new therapeutic targets for PD that slow the progressive loss of dopaminergic neurons and attenuate motor and non-motor symptoms. In this review, we mainly focus on: delayed rectifier, inwardly rectifying, and double-pore K+ channels. We summarize the expression and function of these channels in PD-related brain regions. We also discuss the effects of pharmacological blockade or activation of K+ channels in the progression and treatment of PD.
Collapse
Affiliation(s)
- Linlin Zhang
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, School of Basic Medicine, Qingdao University, Qingdao, 266071, China; Institute of Brain Science and Disease, Qingdao University, Qingdao, 266071, China
| | - Yanan Zheng
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, School of Basic Medicine, Qingdao University, Qingdao, 266071, China; Institute of Brain Science and Disease, Qingdao University, Qingdao, 266071, China
| | - Junxia Xie
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, School of Basic Medicine, Qingdao University, Qingdao, 266071, China; Institute of Brain Science and Disease, Qingdao University, Qingdao, 266071, China
| | - Limin Shi
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, School of Basic Medicine, Qingdao University, Qingdao, 266071, China; Institute of Brain Science and Disease, Qingdao University, Qingdao, 266071, China.
| |
Collapse
|
12
|
Gizak A, Duda P, Pielka E, McCubrey JA, Rakus D. GSK3 and miRNA in neural tissue: From brain development to neurodegenerative diseases. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2020; 1867:118696. [PMID: 32165184 DOI: 10.1016/j.bbamcr.2020.118696] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Revised: 02/28/2020] [Accepted: 03/08/2020] [Indexed: 12/12/2022]
Abstract
MicroRNAs (miRs) are small RNAs modulating gene expression and creating intricate regulatory networks that are dysregulated in many pathological states, including neurodegenerative disorders. In silico analyses denote a multifunctional kinase glycogen synthase kinase-3 (GSK3) as a putative target of numerous miRs identified in neural tissue. GSK3 is engaged in almost all aspects of neuronal development and functioning. Moreover, there is an autoregulatory feedback between GSK3 and miRNAs as the kinase can influence biogenesis of miRs. Members of the miR-GSK3 axes might thus represent convenient therapeutic targets in neuropathologies that display its abnormal regulation. This review summarizes the present knowledge about direct interactions of GSK3 and miRs in brain, and their putative roles in pathogenesis of neurodegenerative and neuropsychiatric disorders. This article is part of a Special Issue entitled: GSK-3 and related kinases in cancer, neurological and other disorders edited by James McCubrey, Agnieszka Gizak and Dariusz Rakus.
Collapse
Affiliation(s)
- Agnieszka Gizak
- Department of Molecular Physiology and Neurobiology, University of Wrocław, Wrocław 50-137, Poland.
| | - Przemysław Duda
- Department of Molecular Physiology and Neurobiology, University of Wrocław, Wrocław 50-137, Poland
| | - Ewa Pielka
- Department of Molecular Physiology and Neurobiology, University of Wrocław, Wrocław 50-137, Poland
| | - James A McCubrey
- Department of Microbiology and Immunology, Brody School of Medicine at East Carolina University Greenville, NC 27858, USA
| | - Dariusz Rakus
- Department of Molecular Physiology and Neurobiology, University of Wrocław, Wrocław 50-137, Poland
| |
Collapse
|
13
|
Pei G, Xu L, Huang W, Yin J. RETRACTED: The protective role of microRNA-133b in restricting hippocampal neurons apoptosis and inflammatory injury in rats with depression by suppressing CTGF. Int Immunopharmacol 2020; 78:106076. [PMID: 31830619 DOI: 10.1016/j.intimp.2019.106076] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Revised: 11/14/2019] [Accepted: 11/21/2019] [Indexed: 02/03/2023]
Abstract
This article has been retracted: please see Elsevier Policy on Article Withdrawal (http://www.elsevier.com/locate/withdrawalpolicy). This article has been retracted at the request of the Editor-in-Chief. Concern was raised about the integrity of the images in Figures 5, 6A and 8A, which appear to contain suspected duplications, as detailed here: https://pubpeer.com/publications/773D824533241B2186D16AA3FFCB3F and here: https://docs.google.com/spreadsheets/d/1r0MyIYpagBc58BRF9c3luWNlCX8VUvUuPyYYXzxWvgY/edit#gid=262337249. Additional suspected image duplications were detected within Figure 7A. Our analysis suggested these image anomalies represent either direct duplications of the entire image, or contain several repeated features between or within an image. The journal requested the corresponding author comment on these concerns and provide the raw data. The authors did not respond to this request and therefore the Editor-in-Chief decided to retract the article.
Collapse
Affiliation(s)
- Gaoyou Pei
- Intervention Department, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450014, China
| | - Liguo Xu
- Intervention Department, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450014, China
| | - Wenhao Huang
- Intervention Department, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450014, China
| | - Jianjun Yin
- Health Examination Department, Qingdao Hiser Medica, Qingdao 266000, Shandong, China.
| |
Collapse
|
14
|
Palasz E, Niewiadomski W, Gasiorowska A, Wysocka A, Stepniewska A, Niewiadomska G. Exercise-Induced Neuroprotection and Recovery of Motor Function in Animal Models of Parkinson's Disease. Front Neurol 2019; 10:1143. [PMID: 31736859 PMCID: PMC6838750 DOI: 10.3389/fneur.2019.01143] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Accepted: 10/11/2019] [Indexed: 12/19/2022] Open
Abstract
Parkinson's disease (PD) is manifested by progressive motor, autonomic, and cognitive disturbances. Dopamine (DA) synthesizing neurons in the substantia nigra (SN) degenerate, causing a decline in DA level in the striatum that leads to the characteristic movement disorders. A disease-modifying therapy to arrest PD progression remains unattainable with current pharmacotherapies, most of which cause severe side effects and lose their efficacy with time. For this reason, there is a need to seek new therapies supporting the pharmacological treatment of PD. Motor therapy is recommended for pharmacologically treated PD patients as it alleviates the symptoms. Molecular mechanisms behind the beneficial effects of motor therapy are unknown, nor is it known whether such therapy may be neuroprotective in PD patients. Due to obvious limitations, human studies are unlikely to answer these questions; therefore, the use of animal models of PD seems indispensable. Motor therapy in animal models of PD characterized by the loss of dopaminergic neurons has neuroprotective and neuroregenerative effects, and the completeness of neuronal protection may depend on (i) degree of neuronal loss, (ii) duration and intensity of exercise, and (iii) time elapsed between insult and commencing of training. As the physical activity is neuroprotective for dopaminergic neurons, the question arises what is the mechanism of this protective action. A current hypothesis assumes a central role of neurotrophic factors in the neuroprotection of dopaminergic neurons, even though it is still not clear whether increased DA level in the nigrostriatal axis results from neurogenesis of dopaminergic neurons in the SN, recovery of the phenotype of dopaminergic neurons, increased sprouting of the residual dopaminergic axons in the striatum, or generation of local striatal neurons from inhibitory interneurons. In the present review, we discuss studies describing the influence of physical exercise on the PD-like changes manifested in animal models of the disease and focus our interest on the current state of knowledge on the mechanism of neuroprotection induced by physical activity as a supportive therapy in PD.
Collapse
Affiliation(s)
- Ewelina Palasz
- Neurobiology Center, Nencki Institute of Experimental Biology, Polish Academy of Science, Warsaw, Poland
| | - Wiktor Niewiadomski
- Department of Applied Physiology, Mossakowski Medical Research Centre, Polish Academy of Sciences, Warsaw, Poland
| | - Anna Gasiorowska
- Neurobiology Center, Nencki Institute of Experimental Biology, Polish Academy of Science, Warsaw, Poland.,Department of Applied Physiology, Mossakowski Medical Research Centre, Polish Academy of Sciences, Warsaw, Poland
| | - Adrianna Wysocka
- Neurobiology Center, Nencki Institute of Experimental Biology, Polish Academy of Science, Warsaw, Poland
| | - Anna Stepniewska
- Department of Applied Physiology, Mossakowski Medical Research Centre, Polish Academy of Sciences, Warsaw, Poland
| | - Grazyna Niewiadomska
- Neurobiology Center, Nencki Institute of Experimental Biology, Polish Academy of Science, Warsaw, Poland
| |
Collapse
|
15
|
Hu ZL, Sun T, Lu M, Ding JH, Du RH, Hu G. Kir6.1/K-ATP channel on astrocytes protects against dopaminergic neurodegeneration in the MPTP mouse model of Parkinson's disease via promoting mitophagy. Brain Behav Immun 2019; 81:509-522. [PMID: 31288070 DOI: 10.1016/j.bbi.2019.07.009] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Revised: 06/24/2019] [Accepted: 07/05/2019] [Indexed: 12/17/2022] Open
Abstract
ATP-sensitive potassium (K-ATP) channels, coupling cell metabolism to cell membrane potential, are involved in brain diseases, including Parkinson's disease (PD). Kir6.1, a pore-forming subunit of K-ATP channel, is prominently expressed in astrocytes and participates in regulating its function. However, the precise role of astrocytic Kir6.1-contaning K-ATP channel (Kir6.1/K-ATP) in PD is not well characterized. In this study, astrocytic Kir6.1 knockout (KO) mice were used to examine the effect of astrocytic Kir6.1/K-ATP channel on dopaminergic (DA) neurodegeneration triggered by the neurotoxin 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine. Here, we found that astrocytic Kir6.1 KO mice showed more DA neuron loss in substantia nigra compacta (SNc), lower level of dopamine in the striatum, and more severe motor dysfunction than controls. Interestingly, this companied by increased neuroinflammation and decreased autophagy level in SNc in vivo and astrocytes in vitro. Mechanistically, astrocytic Kir6.1 KO inhibited mitophagy which resulted in an increase in the accumulation of damaged mitochondria, production of reactive oxygen species and neuroinflammation in astrocytes. Restoration of astrocytic mitophagy rescued the deleterious effects of astrocytic Kir6.1 ablation on mitochondrial dysfunction, inflammation and DA neuron death. Collectively, our findings reveal that astrocytic Kir6.1/K-ATP channel protects against DA neurodegeneration in PD via promoting mitophagy and suggest that astrocytic Kir6.1/K-ATP channel may be a promising therapeutic target for PD.
Collapse
Affiliation(s)
- Zhao-Li Hu
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, 101 Longmian Avenue, Nanjing, Jiangsu 211166, PR China
| | - Ting Sun
- Department of Pharmacology, Nanjing University of Chinese Medicine, 138 Xianlin Avenue, Nanjing, Jiangsu 210023, PR China
| | - Ming Lu
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, 101 Longmian Avenue, Nanjing, Jiangsu 211166, PR China
| | - Jian-Hua Ding
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, 101 Longmian Avenue, Nanjing, Jiangsu 211166, PR China
| | - Ren-Hong Du
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, 101 Longmian Avenue, Nanjing, Jiangsu 211166, PR China.
| | - Gang Hu
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, 101 Longmian Avenue, Nanjing, Jiangsu 211166, PR China; Department of Pharmacology, Nanjing University of Chinese Medicine, 138 Xianlin Avenue, Nanjing, Jiangsu 210023, PR China.
| |
Collapse
|
16
|
Reddy AP, Ravichandran J, Carkaci-Salli N. Neural regeneration therapies for Alzheimer's and Parkinson's disease-related disorders. Biochim Biophys Acta Mol Basis Dis 2019; 1866:165506. [PMID: 31276770 DOI: 10.1016/j.bbadis.2019.06.020] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Revised: 06/24/2019] [Accepted: 06/26/2019] [Indexed: 12/20/2022]
Abstract
Neurodegenerative diseases are devastating mental illnesses without a cure. Alzheimer's disease (AD) characterized by memory loss, multiple cognitive impairments, and changes in personality and behavior. Although tremendous progress has made in understanding the basic biology in disease processes in AD and PD, we still do not have early detectable biomarkers for these diseases. Just in the United States alone, federal and nonfederal funding agencies have spent billions of dollars on clinical trials aimed at finding drugs, but we still do not have a drug or an agent that can slow the AD or PD disease process. One primary reason for this disappointing result may be that the clinical trials enroll patients with AD or PD at advances stages. Although many drugs and agents are tested preclinical and are promising, in human clinical trials, they are mostly ineffective in slowing disease progression. One therapy that has been promising is 'stem cell therapy' based on cell culture and pre-clinical studies. In the few clinical studies that have investigated therapies in clinical trials with AD and PD patients at stage I. The therapies, such as stem cell transplantation - appear to delay the symptoms in AD and PD. The purpose of this article is to describe clinical trials using 1) stem cell transplantation methods in AD and PD mouse models and 2) regenerative medicine in AD and PD mouse models, and 3) the current status of investigating preclinical stem cell transplantation in patients with AD and PD.
Collapse
Affiliation(s)
- Arubala P Reddy
- Pharmacology & Neuroscience Department, Texas Tech University Health Sciences Center, 3601 4th Street, MS 9424, Lubbock, TX 79430, United States.
| | - Janani Ravichandran
- Texas Tech University Health Sciences Center El Paso, 5001 El Paso Drive, El Paso, TX 79905, United States.
| | - Nurgul Carkaci-Salli
- Department of Pharmacology, Penn State College of Medicine, 500 University Drive, Hershey, PA 17033.
| |
Collapse
|
17
|
He HW, Zhang YL, Yu BQ, Ye G, You W, So KF, Li X. Soluble Nogo receptor 1 fusion protein protects neural progenitor cells in rats with ischemic stroke. Neural Regen Res 2019; 14:1755-1764. [PMID: 31169193 PMCID: PMC6585563 DOI: 10.4103/1673-5374.257531] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Soluble Nogo66 receptor-Fc protein (sNgR-Fc) enhances axonal regeneration following central nervous system injury. However, the underlying mechanisms remain unclear. In this study, we investigated the effects of sNgR-Fc on the proliferation and differentiation of neural progenitor cells. The photothrombotic cortical injury model of ischemic stroke was produced in the parietal cortex of Sprague-Dawley rats. The rats with photothrombotic cortical injury were randomized to receive infusion of 400 μg/kg sNgR-Fc (sNgR-Fc group) or an equal volume of phosphate-buffered saline (photothrombotic cortical injury group) into the lateral ventricle for 3 days. The effects of sNgR-Fc on the proliferation and differentiation of endogenous neural progenitor cells were examined using BrdU staining. Neurological function was evaluated with the Morris water maze test. To further examine the effects of sNgR-Fc treatment on neural progenitor cells, photothrombotic cortical injury was produced in another group of rats that received transplantation of neural progenitor cells from the hippocampus of embryonic Sprague-Dawley rats. The animals were then given an infusion of phosphate-buffered saline (neural progenitor cells group) or sNgR-Fc (sNgR-Fc + neural progenitor cells group) into the lateral ventricle for 3 days. sNgR-Fc enhanced the proliferation of cultured neural progenitor cells in vitro as well as that of endogenous neural progenitor cells in vivo, compared with phosphate-buffered saline, and it also induced the differentiation of neural progenitor cells into neurons. Compared with the photothrombotic cortical injury group, escape latency in the Morris water maze and neurological severity score were greatly reduced, and distance traveled in the target quadrant was considerably increased in the sNgR-Fc group, indicating a substantial improvement in neurological function. Furthermore, compared with phosphate-buffered saline infusion, sNgR-Fc infusion strikingly improved the survival and differentiation of grafted neural progenitor cells. Our findings show that sNgR-Fc regulates neural progenitor cell proliferation, migration and differentiation. Therefore, sNgR-Fc is a potential novel therapy for stroke and neurodegenerative diseases, The protocols were approved by the Committee on the Use of Live Animals in Teaching and Research of the University of Hong Kong (approval No. 4560-17) in November, 2015.
Collapse
Affiliation(s)
- Hai-Wei He
- School of Medicine, South China University of Technology; Department of Emergency Medicine, Department of Emergency and Critical Care Medicine, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong Province, China
| | - Yue-Lin Zhang
- Department of Emergency Medicine, Department of Emergency and Critical Care Medicine, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong Province, China
| | - Bao-Qi Yu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Key Laboratory of Remodelling- related Cardiovascular Diseases, Ministry of Education, Beijing, China
| | - Gen Ye
- Department of Emergency Medicine, Department of Emergency and Critical Care Medicine, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong Province, China
| | - Wei You
- Department of Emergency Medicine, Department of Emergency and Critical Care Medicine, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong Province, China
| | - Kwok-Fai So
- School of Biomedical Sciences, The State Key Laboratory of Brain and Cognitive sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Xin Li
- School of Medicine, South China University of Technology; Department of Emergency Medicine, Department of Emergency and Critical Care Medicine, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong Province, China
| |
Collapse
|