1
|
Klein B, Ciesielska A, Losada PM, Sato A, Shah-Morales S, Ford JB, Higashikubo B, Tager D, Urry A, Bombosch J, Chang WC, Andrews-Zwilling Y, Nejadnik B, Warraich Z, Paz JT. Modified human mesenchymal stromal/stem cells restore cortical excitability after focal ischemic stroke in rats. Mol Ther 2024:S1525-0016(24)00807-4. [PMID: 39668560 DOI: 10.1016/j.ymthe.2024.12.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 09/18/2024] [Accepted: 12/06/2024] [Indexed: 12/14/2024] Open
Abstract
Allogeneic modified bone marrow-derived human mesenchymal stromal/stem cells (hMSC-SB623 cells) are in clinical development for the treatment of chronic motor deficits after traumatic brain injury and cerebral ischemic stroke. However, their exact mechanisms of action remain unclear. Here, we investigated the effects of this cell therapy on cortical network excitability, brain tissue, and peripheral blood at a chronic stage after ischemic stroke in a rat model. One month after focal cortical ischemic stroke, hMSC-SB623 cells or the vehicle solution were injected into the peri-stroke cortex. Starting one week after treatment, cortical excitability was assessed ex vivo. hMSC-SB623 cell transplants reduced stroke-induced cortical hyperexcitability, restoring cortical excitability to control levels. The histology of brain tissue revealed an increase of factors relevant to neuroregeneration, and synaptic and cellular plasticity. Whole-blood RNA sequencing and serum protein analyses showed that intra-cortical hMSC-SB623 cell transplantation reversed effects of stroke on peripheral blood factors known to be involved in stroke pathophysiology. Our findings demonstrate that intra-cortical transplants of hMSC-SB623 cells correct stroke-induced circuit disruptions even at the chronic stage, suggesting broad usefulness as a therapeutic for neurological conditions with network hyperexcitability. Additionally, the transplanted cells exert far-reaching immunomodulatory effects whose therapeutic impact remains to be explored.
Collapse
Affiliation(s)
| | - Agnieszka Ciesielska
- Gladstone Institute of Neurological Disease, San Francisco, CA, USA; University of California, San Francisco, Department of Neurology, and the Kavli Institute for Fundamental Neuroscience, San Francisco, CA, USA
| | | | | | | | - Jeremy B Ford
- Gladstone Institute of Neurological Disease, San Francisco, CA, USA
| | | | - Dale Tager
- Gladstone Institute of Neurological Disease, San Francisco, CA, USA
| | - Alexander Urry
- Gladstone Institute of Neurological Disease, San Francisco, CA, USA
| | | | | | | | | | | | - Jeanne T Paz
- Gladstone Institute of Neurological Disease, San Francisco, CA, USA; University of California, San Francisco, Department of Neurology, and the Kavli Institute for Fundamental Neuroscience, San Francisco, CA, USA; University of California, San Francisco, Neurosciences Graduate Program, San Francisco, CA, USA.
| |
Collapse
|
2
|
Li M, Mo Y, Yu Q, Anayyat U, Yang H, Zhang F, Wei Y, Wang X. Rotating magnetic field improves cognitive and memory impairments in APP/PS1 mice by activating autophagy and inhibiting the PI3K/AKT/mTOR signaling pathway. Exp Neurol 2024; 383:115029. [PMID: 39461710 DOI: 10.1016/j.expneurol.2024.115029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2024] [Revised: 08/16/2024] [Accepted: 10/23/2024] [Indexed: 10/29/2024]
Abstract
Alzheimer's disease (AD) is a geriatric disorder that can be roughly classified into sporadic AD and hereditary AD. The latter is strongly associated with genetic factors, and its treatment poses greater challenges compared to sporadic AD. Rotating magnetic fields (RMF) is a non-invasive treatment known to have diverse biological effects, including the modulation of the central nervous system and aging. However, the impact of RMF on hereditary AD and its underlying mechanism remain unexplored. In this study, we exposed APP/PS1 mice to RMF (2 h/day, 0.2 T, 4 Hz) for a duration of 6 months. The results demonstrated that RMF treatment significantly ameliorated their cognitive and memory impairments, attenuated neuronal damage, and reduced amyloid deposition. Furthermore, RNA-sequencing analysis revealed a significant enrichment of autophagy-related genes and the PI3K/AKT-mTOR signaling pathway. Western blotting further confirmed that RMF activated autophagy and suppressed the phosphorylation of proteins associated with the PI3K/AKT/mTOR signaling pathway in APP/PS1 mice. These protective effects and the underlying mechanism were also observed in Aβ25-35-exposed HT22 cells. Collectively, our findings indicate that RMF improves cognitive and memory dysfunction in APP/PS1 mice by activating autophagy and inhibiting the PI3K/AKT/mTOR signaling pathway, thus highlighting the potential of RMF as a clinical treatment for hereditary AD.
Collapse
Affiliation(s)
- Mengqing Li
- School of Basic Medical Sciences, Shenzhen University, Shenzhen, Guangdong 518061, China
| | - Yaxian Mo
- Songgang People's Hospital, Shenzhen, Guangdong 518105, China
| | - Qinyao Yu
- School of Pharmacy, Shenzhen University, Shenzhen, Guangdong 518061, China
| | - Umer Anayyat
- School of Basic Medical Sciences, Shenzhen University, Shenzhen, Guangdong 518061, China
| | - Hua Yang
- School of Basic Medical Sciences, Shenzhen University, Shenzhen, Guangdong 518061, China
| | - Fen Zhang
- School of Basic Medical Sciences, Shenzhen University, Shenzhen, Guangdong 518061, China
| | - Yunpeng Wei
- Songgang People's Hospital, Shenzhen, Guangdong 518105, China.
| | - Xiaomei Wang
- School of Basic Medical Sciences, Shenzhen University, Shenzhen, Guangdong 518061, China; International Cancer Center, Shenzhen University Health Sciences Center, Shenzhen, Guangdong 518061, China.
| |
Collapse
|
3
|
Zhang S, Gao Z, Feng L, Li M. Prevention and Treatment Strategies for Alzheimer's Disease: Focusing on Microglia and Astrocytes in Neuroinflammation. J Inflamm Res 2024; 17:7235-7259. [PMID: 39421566 PMCID: PMC11484773 DOI: 10.2147/jir.s483412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Accepted: 10/02/2024] [Indexed: 10/19/2024] Open
Abstract
Alzheimer's disease (AD) is a fatal neurodegenerative disease characterized by its insidious onset and progressive development, making it the most common form of dementia. Despite its prevalence, the exact causes and mechanisms responsible for AD remain unclear. Recent studies have highlighted that inflammation in the central nervous system (CNS) plays a crucial role in both the initiation and progression of AD. Neuroinflammation, an immune response within the CNS triggered by glial cells in response to various stimuli, such as nerve injury, infection, toxins, or autoimmune reactions, has emerged as a significant factor alongside amyloid deposition and neurofibrillary tangles (NFTs) commonly associated with AD. This article aims to provide an overview of the most recent research regarding the involvement of neuroinflammation in AD, with a particular focus on elucidating the specific mechanisms involving microglia and astrocytes. By exploring these intricate processes, a new theoretical framework can be established to further probe the impact of neuroinflammation on the development and progression of AD. Through a deeper understanding of these underlying mechanisms, potential targets for therapeutic interventions and novel treatment strategies can be identified in the ongoing battle against AD.
Collapse
Affiliation(s)
- Shenghao Zhang
- Department of Neurology, The Third Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, Jilin Province, 130021, People’s Republic of China
| | - Zhejianyi Gao
- Department of Orthopaedics, Fushun Hospital of Chinese Medicine, Fushun, Liaoning Province, 113008, People’s Republic of China
| | - Lina Feng
- Department of Neurology, The Third Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, Jilin Province, 130021, People’s Republic of China
- Shandong Key Laboratory of TCM Multi-Targets Intervention and Disease Control, The Second Affiliated Hospital of Shandong First Medical University, Taian, Shandong Province, 271000, People’s Republic of China
| | - Mingquan Li
- Department of Neurology, The Third Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, Jilin Province, 130021, People’s Republic of China
| |
Collapse
|
4
|
Zhang L, Xiao Z, Su Z, Wang X, Tian H, Su M. Repetitive transcranial magnetic stimulation promotes motor function recovery in mice after spinal cord injury via regulation of the Cx43-autophagy loop. J Orthop Surg Res 2024; 19:387. [PMID: 38956661 PMCID: PMC11218133 DOI: 10.1186/s13018-024-04879-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 06/25/2024] [Indexed: 07/04/2024] Open
Abstract
Spinal cord injury (SCI) is a severe condition with an extremely high disability rate. It is mainly manifested as the loss of motor, sensory and autonomic nerve functions below the injury site. High-frequency transcranial magnetic stimulation, a recently developed neuromodulation method, can increase motor function in mice with spinal cord injury. This study aimed to explore the possible mechanism by which transcranial magnetic stimulation (TMS) restores motor function after SCI. A complete T8 transection model of the spinal cord was established in mice, and the mice were treated daily with 15 Hz high-frequency transcranial magnetic stimulation. The BMS was used to evaluate the motor function of the mice after SCI. Western blotting and immunofluorescence were used to detect the expression of Connexin43 (CX43) and autophagy-related proteins in vivo and in vitro, and correlation analysis was performed to study the relationships among autophagy, CX43 and motor function recovery after SCI in mice. Western blotting was used to observe the effect of magnetic stimulation on the expression of mTOR pathway members. In the control group, the expression of CX43 was significantly decreased, and the expression of microtubule-associated protein 1 A/1b light chain 3 (LC3II) and P62 was significantly increased after 4 weeks of spinal cord transection. After high-frequency magnetic stimulation, the level of CX43 decreased, and the levels of LC3II and P62 increased in primary astrocytes. The BMS of the magnetic stimulation group was greater than that of the control group. High-frequency magnetic stimulation can inhibit the expression of CX43, which negatively regulates autophagic flux. HF-rTMS increased the expression levels of mTOR, p-mTOR and p-S6. Our experiments showed that rTMS can restore hindlimb motor function in mice after spinal cord injury via regulation of the Cx43-autophagy loop and activation of the mTOR signalling pathway.
Collapse
Affiliation(s)
- Lechi Zhang
- Department of Rehabilitation Medicine, The Fourth Affiliated Hospital of Soochow University, Suzhou, China
- Institute of Rehabilitation, Soochow University, Suzhou, China
- Rehabilitation Medicine Center of Suzhou Hospital Affiliated to Nanjing Medical University, Suzhou, China
| | - Zhihang Xiao
- Department of Rehabilitation Medicine, The Fourth Affiliated Hospital of Soochow University, Suzhou, China
- Institute of Rehabilitation, Soochow University, Suzhou, China
| | - Zelin Su
- Department of Rehabilitation Medicine, The Fourth Affiliated Hospital of Soochow University, Suzhou, China
- Institute of Rehabilitation, Soochow University, Suzhou, China
| | - Xinlong Wang
- Department of Rehabilitation Medicine, The Fourth Affiliated Hospital of Soochow University, Suzhou, China
- Institute of Rehabilitation, Soochow University, Suzhou, China
- Rehabilitation Medicine Center of Suzhou Hospital Affiliated to Nanjing Medical University, Suzhou, China
| | - Huifang Tian
- Department of Rehabilitation Medicine, The Fourth Affiliated Hospital of Soochow University, Suzhou, China
- Institute of Rehabilitation, Soochow University, Suzhou, China
| | - Min Su
- Department of Rehabilitation Medicine, The Fourth Affiliated Hospital of Soochow University, Suzhou, China.
- Institute of Rehabilitation, Soochow University, Suzhou, China.
| |
Collapse
|
5
|
Wang X, Ding Q, Li Y, Li T, Li Y, Yin J, Zhuang W. Repetitive transcranial magnetic stimulation impacts the executive function of patients with vascular cognitive impairment: a systematic review and meta-analysis. Front Neurol 2024; 15:1374395. [PMID: 38962482 PMCID: PMC11220282 DOI: 10.3389/fneur.2024.1374395] [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: 01/24/2024] [Accepted: 06/07/2024] [Indexed: 07/05/2024] Open
Abstract
Objective Executive dysfunction is a core symptom of vascular cognitive impairment (VCI), which seriously affects patients' prognosis. This paper aims to investigate the effectiveness of rTMS on executive function in VCI. Methods The databases selected for this study included Pubmed, Embase, Cochrane Library, China National Knowledge Infrastructure (CNKI), Wanfang, China Science and Technology Journal Database (VIP), and China Biology Medicine Disc (CBM). The screening times were conducted from the time of library construction until August 23, 2023. The inclusion criteria for this meta-analysis were randomized controlled trials (RCTs) on rTMS for VCI, which include executive function scores. The primary metrics were executive subscale scores of the Cognitive Comprehensive Scale and total scores of the Executive Specificity Scale. The secondary metrics were subscale scores of the Executive Specificity Scale. The quality of each eligible study was assessed using the Cochrane Risk of Bias tool. Meta-analysis and bias analysis were performed using Stata (version 16.0) and RevMan (version 5.3). Results A total of 20 high-quality clinical RCTs with 1,049 samples were included in this paper. The findings from the primary outcomes revealed that within the rTMS group, there were significantly higher scores observed for the executive sub-item on the cognitive composite scale (SMD = 0.93, 95% CI = 0.77-1.08, p < 0.00001, I 2 = 14%) and the total score on the executive specific scale (SMD = 0.69, 95% CI = 0.44-0.94, p < 0.00001, I 2 = 0%) compared to the control group. As for the secondary outcome measures, as shown by the Trail Making Test-A (time) (MD = -35.75, 95% CI = -68.37 to -3.12, p = 0.03, I 2 = 55%), the Stroop-C card (time) (SMD = -0.46, 95% CI = -0.86 to -0.06, p = 0.02, I 2 = 0%) and the Stroop-C card (correct number) (SMD = 0.49, 95% CI = 0.04-0.94, p = 0.03, I 2 = 0%), the experimental group shorts time and enhances accuracy of executive task in comparison to the control group. Subgroup analysis of the main outcome demonstrated that intermittent theta burst stimulation (iTBS), higher frequency, lower intensity, longer duration, and combined comprehensive therapy exhibited superior efficacy. Conclusion rTMS is effective in the treatment of the executive function of VCI. The present study has some limitations, so multi-center, large-sample, objective indicators and parameters are needed to further explore in the future.Systematic review registration:https://www.crd.york.ac.uk/prospero/, CRD42023459669.
Collapse
Affiliation(s)
- Xu Wang
- School of Rehabilitation Medicine, Henan University of Chinese Medicine, Zhengzhou, China
| | - Qixin Ding
- School of Rehabilitation Medicine, Henan University of Chinese Medicine, Zhengzhou, China
| | - Yuefang Li
- School of Clinical Medicine, Henan University, Zhengzhou, China
| | - Tianshu Li
- Department of Rehabilitation Medicine, The First People’s Hospital of Zhengzhou, Zhengzhou, China
| | - Yakun Li
- Department of Rehabilitation, Henan Provincial People's Hospital, Zhengzhou, China
| | - Jialin Yin
- Department of Rehabilitation, Henan Provincial People's Hospital, Zhengzhou, China
| | - Weisheng Zhuang
- Department of Rehabilitation, School of Rehabilitation Medicine, Henan Provincial People's Hospital, Henan University of Chinese Medicine, Zhengzhou, China
| |
Collapse
|
6
|
Zheng J, Peng S, Cui L, Liu X, Li T, Zhao Z, Li Y, Hu Y, Zhang M, Xu L, Zhang J. Enriched environment attenuates hippocampal theta and gamma rhythms dysfunction in chronic cerebral hypoperfusion via improving imbalanced neural afferent levels. Front Cell Neurosci 2023; 17:985246. [PMID: 37265581 PMCID: PMC10231328 DOI: 10.3389/fncel.2023.985246] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2022] [Accepted: 02/27/2023] [Indexed: 06/03/2023] Open
Abstract
Chronic cerebral hypoperfusion (CCH) is increasingly recognized as a common cognitive impairment-causing mechanism. However, no clinically effective drugs to treat cognitive impairment due to CCH have been identified. An abnormal distribution of neural oscillations was found in the hippocampus of CCH rats. By releasing various neurotransmitters, distinct afferent fibers in the hippocampus influence neuronal oscillations in the hippocampus. Enriched environments (EE) are known to improve cognitive levels by modulating neurotransmitter homeostasis. Using EE as an intervention, we examined the levels of three classical neurotransmitters and the dynamics of neural oscillations in the hippocampus of the CCH rat model. The results showed that EE significantly improved the balance of three classical neurotransmitters (acetylcholine, glutamate, and GABA) in the hippocampus, enhanced the strength of theta and slow-gamma (SG) rhythms, and dramatically improved neural coupling across frequency bands in CCH rats. Furthermore, the expression of the three neurotransmitter vesicular transporters-vesicular acetylcholine transporters (VAChT) and vesicular GABA transporters (VGAT)-was significantly reduced in CCH rats, whereas the expression of vesicular glutamate transporter 1 (VGLUT1) was abnormally elevated. EE partially restored the expression of the three protein levels to maintain the balance of hippocampal afferent neurotransmitters. More importantly, causal mediation analysis showed EE increased the power of theta rhythm by increasing the level of VAChT and VGAT, which then enhanced the phase amplitude coupling of theta-SG and finally led to an improvement in the cognitive level of CCH. These findings shed light on the role of CCH in the disruption of hippocampal afferent neurotransmitter balance and neural oscillations. This study has implications for our knowledge of disease pathways.
Collapse
Affiliation(s)
- Jiaxin Zheng
- Department of Neurology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Sisi Peng
- Department of Neurology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Lingling Cui
- Department of Anesthesiology, Tongren Hospital of Wuhan University, Wuhan, China
| | - Xi Liu
- Department of Neurology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Tian Li
- Clinical Medical Research Center for Dementia and Cognitive Impairment in Hubei Province, Wuhan, China
| | - Zhenyu Zhao
- Clinical Medical Research Center for Dementia and Cognitive Impairment in Hubei Province, Wuhan, China
| | - Yaqing Li
- Clinical Medical Research Center for Dementia and Cognitive Impairment in Hubei Province, Wuhan, China
| | - Yuan Hu
- Department of Neurology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Miao Zhang
- Department of Neurology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Linling Xu
- Clinical Medical Research Center for Dementia and Cognitive Impairment in Hubei Province, Wuhan, China
| | - JunJian Zhang
- Department of Neurology, Zhongnan Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
7
|
Liu C, Shi R, Liu Y, Zhao X, Zhang X, Wang H, Wu L, Shang X. Low-frequency transcranial magnetic stimulation protects cognition in mice with chronic unpredictable mild stress through autophagy regulation. Behav Brain Res 2023; 444:114366. [PMID: 36854362 DOI: 10.1016/j.bbr.2023.114366] [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: 10/11/2022] [Revised: 01/29/2023] [Accepted: 02/25/2023] [Indexed: 02/27/2023]
Abstract
Although transcranial magnetic stimulation (TMS) has been approved for the treatment of major depression, few studies have analyzed the ability of low-frequency TMS (LF-TMS) to treat depressive symptoms. Our study confirmed that LF-TMS protects the cognitive function,which can play a certain reference role in the future clinical treatment. The effectiveness of high-frequency TMS therapy has been well documented. However, the use of low-frequency TMS (LF-TMS) in the treatment of depression is rarely reported. This study aims to evaluate whether LF-TMS can reverse depression-induced cognitive impairment. We created a mouse model of depression using the chronic unpredictable mild stress (CUMS) paradigm. Male C57BL/6J mice,6-8 weeks old,were randomly divided into four groups: a CON (control) group, a CUMS group, a CUMS+LF-TMS group, and a CUMS+LF-TMS+RAP (rapamycin) group. The CUMS was maintained for 28 days. LF-TMS (1 Hz) and Rap were administered for 28 days from the first day of CUMS. The mice in all groups except the control demonstrated evidence of anhedonia, anxiety, and cognitive decline on behavioral tests during the four weeks of CUMS.All the experiments were carried out under a 12-h light/dark cycle (lights on at 7 a.m.) except for the dark/light cycle reversal stimulation of CUMS. LF-TMS at 20 Mt, 1 Hz for 1 min alleviated damage to spatial cognition and synaptic plasticity in the hippocampus. Western blot analysis showed that LF-TMS reduced the down-regulation of autophagy signals in the CUMS+LF-TMS group, and enhanced the expression of synaptic plasticity-related factors, thereby improving the spatial cognitive impairment resulting from the CUMS. We concluded that LF-TMS can effectively relieve depressive behavior and cognitive dysfunction in mice subjected to CUMS by regulating autophagy signals and synaptic proteins.
Collapse
Affiliation(s)
- Chuan Liu
- School of Psychology and Mental Health, North China University of Science and Technology, 21 Bohai Road, Tang'shan 063210, Hebei Province, PR China; College of Basic Medical Sciences, North China University of Science and Technology, 21 Bohai Road, Tang'shan 063210, Hebei Province, PR China
| | - Ruidie Shi
- School of Psychology and Mental Health, North China University of Science and Technology, 21 Bohai Road, Tang'shan 063210, Hebei Province, PR China
| | - Yuting Liu
- School of Psychology and Mental Health, North China University of Science and Technology, 21 Bohai Road, Tang'shan 063210, Hebei Province, PR China
| | - Xiangwei Zhao
- School of Psychology and Mental Health, North China University of Science and Technology, 21 Bohai Road, Tang'shan 063210, Hebei Province, PR China
| | - Xiujun Zhang
- School of Psychology and Mental Health, North China University of Science and Technology, 21 Bohai Road, Tang'shan 063210, Hebei Province, PR China.
| | - Haitao Wang
- School of Psychology and Mental Health, North China University of Science and Technology, 21 Bohai Road, Tang'shan 063210, Hebei Province, PR China; College of Basic Medical Sciences, North China University of Science and Technology, 21 Bohai Road, Tang'shan 063210, Hebei Province, PR China.
| | - Lei Wu
- School of Psychology and Mental Health, North China University of Science and Technology, 21 Bohai Road, Tang'shan 063210, Hebei Province, PR China.
| | - Xueliang Shang
- School of Psychology and Mental Health, North China University of Science and Technology, 21 Bohai Road, Tang'shan 063210, Hebei Province, PR China.
| |
Collapse
|
8
|
Neubauer AC, Wood G. Intelligenzsteigerung durch Neuroenhancement? PSYCHOLOGISCHE RUNDSCHAU 2022. [DOI: 10.1026/0033-3042/a000599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
Zusammenfassung. Die menschliche Intelligenz gehört zu den bestuntersuchten psychologischen Merkmalen, in denen interindividuelle Differenzen bestehen. Die mehr als 100jährige Forschungsgeschichte hat einen hoch belastbaren Wissensstand hervorgebracht; dieser umfasst die Definition, die Psychometrie, die (ontogenetische) Entwicklung, die Struktur, die Vorhersagekraft für real-life-Variablen, das Wissen über elementar-kognitive, verhaltensgenetische und neurobiologische Grundlagen der Intelligenz, u.v.m. Jüngst steht zudem die Frage des ‚enhancements‘ der Intelligenz im Fokus, eine Frage, die nicht zuletzt durch die aktuelle philosophische Strömung des Transhumanismus stark an Bedeutung gewinnt. Der Transhumanismus nimmt eine substanzielle Erhöhung (enhancement) von Fähigkeiten und anderen (auch) psychologischen Eigenschaften des Menschen ins Zentrum und postuliert, dass ein soziokultureller Fortschritt – und letztlich das Überlegen des Homo Sapiens und unseres Planeten – erst durch technologischen Fortschritt ermöglicht werde. Viele Transhumanisten stellen eine substanzielle Steigerung der Intelligenz in den Vordergrund, die primär durch (neuro–)technologische und pharmakologische Maßnahmen zu bewerkstelligen seien. Diese Debatten sind jedoch oft gekennzeichnet durch übertrieben optimistische Annahmen der Möglichkeiten moderner neurowissenschaftlicher Methoden bei gleichzeitiger Vernachlässigung der potenziellen negativen Folgen für das Individuum, für die Gesellschaften und insgesamt für unsere Spezies. Im gegenständlichen Überblicksbeitrag werden behaviorale, neuroelektrische und pharmakologische Methoden im Hinblick auf ihr aktuelles Potenzial einer Steigerung der individuellen Intelligenz analysiert. Die zwischenzeitlich zu diesen Fragen vorliegenden experimentellen Studien, sowie verfügbare Metaanalysen lassen allerdings den Schluss zu, dass bislang keine der gegenwärtig verfügbaren Methoden das Potenzial haben, die individuelle Intelligenz substanziell zu steigern. Und selbst falls solche möglicherweise in absehbarer Zeit zur Verfügung stünden, müssen zuvor sowohl individuelle als auch gesellschaftliche (negative) Konsequenzen einer kritischen Analyse unterzogen werden. Diese sind Gegenstand einer abschließenden Diskussion.
Collapse
|
9
|
Repetitive transcranial magnetic stimulation (rTMS) for multiple neurological conditions in rodent animal models: A systematic review. Neurochem Int 2022; 157:105356. [DOI: 10.1016/j.neuint.2022.105356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 03/31/2022] [Accepted: 04/28/2022] [Indexed: 12/09/2022]
|
10
|
Moghaddam MH, Hatari S, Shahidi AMEJ, Nikpour F, Omran HS, Fathi M, Vakili K, Abdollahifar MA, Tizro M, Eskandari N, Raoofi A, Ebrahimi V, Aliaghaei A. Human olfactory epithelium-derived stem cells ameliorate histopathological deficits and improve behavioral functions in a rat model of cerebellar ataxia. J Chem Neuroanat 2022; 120:102071. [PMID: 35051594 DOI: 10.1016/j.jchemneu.2022.102071] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 12/18/2021] [Accepted: 01/15/2022] [Indexed: 12/11/2022]
Abstract
Cell replacement therapy (CRT) is one of the most effective approaches used to alleviate symptoms of neurodegenerative syndromes such as cerebellar ataxia (CA). Human olfactory epithelium mesenchymal stem cells (OE-MSCs) have been recognized as a promising candidate for CRT, due to their distinctive features including immunomodulatory properties and ease of accessible compared to other types of MSCs. Hence, the main goal of our study was to explore the impacts of OE-MSCs transplantation on behavioral, structural, and histological deficiencies in a rat model of CA. After obtained an informed consent from volunteers, OE-MSCs were obtained from their nasal cavity. Then, OE-MSCs were characterized by the positive expression of CD73, CD90, and CD105 as MSCs as well as nestin and vimentin as primitive neuroectodermal stem cells markers. Then, the animals were randomized into three control, 3-acetylpyridine (3-AP) treated, and 3-AP + cell groups. In both experimental groups, the rats received intraperitoneal injection of 3-AP (75 mg/kg), followed by the implantation of OE-MSCs into the cerebellum of 3-AP + cell group. The impact of engrafted OE-MSCs on motor coordination and performance along with biochemical, immunohistochemical, and stereological changes in the cerebellum of the rat models of CA were investigated. According to our findings, the administration of 3-AP decreased the cerebellar GSH concentration. The injection of 3-AP also altered the morphological characteristics of the cerebellar Golgi cells. On the other hand, OE-MSCs transplantation improved motor coordination in CA. Besides, the implantation of OE-MSCs reduced caspase-3 expression and microglia proliferation in the cerebellum upon 3-AP administration. Finally, the transplant of OE-MSCs protected Purkinje cells against 3-AP toxicity. In sum, the present study revealed considerable advantages of OE-MSCs in managing CA animal model.
Collapse
Affiliation(s)
- Meysam Hassani Moghaddam
- Brain Mapping Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Department of Anatomical Sciences, Faculty of Medicine, AJA University of Medical Sciences, Tehran, Iran
| | - Saba Hatari
- Brain Mapping Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Amir Mahdi Emam Jome Shahidi
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fatemeh Nikpour
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hossein Salehi Omran
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mobina Fathi
- Student Research Committee, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Kimia Vakili
- Student Research Committee, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Amin Abdollahifar
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mahdi Tizro
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Neda Eskandari
- Brain Mapping Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Amir Raoofi
- Cellular and Molecular Research Center, Department of Anatomy, Faculty of Medicine, Sabzevar University of Medical Sciences, Sabzevar, Iran
| | - Vahid Ebrahimi
- Department of Anatomy and Cell Biology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Applied Biomedical Research Center, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Abbas Aliaghaei
- Brain Mapping Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
11
|
Tan L, Liu X, Dou H, Hou Y. Characteristics and regulation of mesenchymal stem cell plasticity by the microenvironment — specific factors involved in the regulation of MSC plasticity. Genes Dis 2022; 9:296-309. [PMID: 35224147 PMCID: PMC8843883 DOI: 10.1016/j.gendis.2020.10.006] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 10/05/2020] [Accepted: 10/22/2020] [Indexed: 02/07/2023] Open
Abstract
Mesenchymal stem cells (MSCs), multipotent stromal cells, have attracted extensive attention in the field of regenerative medicine and cell therapy due to the capacity of self-renewal, multilineage differentiation, and immune regulation. MSCs have different cellular effects in different diseases, and even have markedly different curative effects with different tissue sources, indicating the plasticity of MSCs. The phenotypes, secreted factors, and proliferative, migratory, differentiating, and immunomodulatory effects of MSCs depend on certain mediators present in their microenvironment. Understanding microenvironmental factors and their internal mechanisms in MSC responses may help in subsequent prediction and improvement of clinical benefits. This review highlighted the recent advances in MSC plasticity in the physiological and pathological microenvironment and multiple microenvironmental factors regulating MSC plasticity. It also highlighted some progress in the underlying molecular mechanisms of MSC remodeling in the microenvironment. It might provide references for the improvement in vitro culture of MSCs, clinical application, and in vivo induction.
Collapse
|
12
|
Mesenchymal Stem Cells: Therapeutic Mechanisms for Stroke. Int J Mol Sci 2022; 23:ijms23052550. [PMID: 35269692 PMCID: PMC8910569 DOI: 10.3390/ijms23052550] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Revised: 02/20/2022] [Accepted: 02/23/2022] [Indexed: 12/12/2022] Open
Abstract
Due to aging of the world’s population, stroke has become increasingly prevalent, leading to a rise in socioeconomic burden. In the recent past, stroke research and treatment have become key scientific issues that need urgent solutions, with a sharp focus on stem cell transplantation, which is known to treat neurodegenerative diseases related to traumatic brain injuries, such as stroke. Indeed, stem cell therapy has brought hope to many stroke patients, both in animal and clinical trials. Mesenchymal stem cells (MSCs) are most commonly utilized in biological medical research, due to their pluripotency and universality. MSCs are often obtained from adipose tissue and bone marrow, and transplanted via intravenous injection. Therefore, this review will discuss the therapeutic mechanisms of MSCs and extracellular vehicles (EVs) secreted by MSCs for stroke, such as in attenuating inflammation through immunomodulation, releasing trophic factors to promote therapeutic effects, inducing angiogenesis, promoting neurogenesis, reducing the infarct volume, and replacing damaged cells.
Collapse
|
13
|
Akbari S, Hooshmandi E, Bayat M, Borhani Haghighi A, Salehi MS, Pandamooz S, Yousefi Nejad A, Haghani M. The neuroprotective properties and therapeutic potential of epidermal neural crest stem cells transplantation in a rat model of vascular dementia. Brain Res 2021; 1776:147750. [PMID: 34896332 DOI: 10.1016/j.brainres.2021.147750] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 11/17/2021] [Accepted: 12/06/2021] [Indexed: 01/04/2023]
Abstract
INTRODUCTION The incidence rate of senile dementia is rising, and there is no definite cure for it yet. Cell therapy, as a new investigational approach, has shown promising results. Hair bulges with abundant easily accessible neural stem cells permit autologous implantation in irreversible neurodegenerative disorders. METHODS Fifty rats were randomly divided into 5 groups of control, sham-operation, two-common carotid vessel-occlusion rats that received vehicle (2VO + V), 2VO rats that received 1 × 106 epidermal stem cells (2VO + ESC1), and 2VO rats that received 2.5 × 106 epidermal stem cells (2VO + ESC2) in 300 µl PBS intravenously on days 4, 9, and 14 after surgery. The epidermal neural crest stem cells (EPI-NCSCs) were isolated from hair follicles of rat whiskers. The open-field, passive avoidance, and Morris water maze were used as behavioral tests. The basal-synaptic transmission, long-term potentiation (LTP), and short-term synaptic plasticity were evaluated by field-potential recording of the CA1 hippocampal area. RESULTS 30 days after the first transplantation in the 2VO + ESC1 group, functional recovery was prominent in anxiety and fear memory compared to the 2VO + ESC2 group, while LTP induction was recovered in both groups of grafted animals without improvement in basal synaptic transmission. These positive recoveries may be related to the release of different neurotrophic factors from grafted cells that can stimulate endogenous neurogenesis and synaptic plasticity. CONCLUSIONS Our results showed that EPI-NCSCs implantation could rescue LTP and cognitive disability in 2VO rats, while transplantation of 1 million cells showed better performance relative to 2.5 million cells.
Collapse
Affiliation(s)
- Somayeh Akbari
- Department of Physiology, The Medical School, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Etrat Hooshmandi
- Clinical Neurology Research Centre, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mahnaz Bayat
- Clinical Neurology Research Centre, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | - Mohammad Saied Salehi
- Clinical Neurology Research Centre, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Sareh Pandamooz
- Stem Cells Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Amirhossein Yousefi Nejad
- Department of Veterinary Medicine, Faculty of Veterinary Medicine. Islamic Azad University of Kazeroon, Shiraz, Iran
| | - Masoud Haghani
- Department of Physiology, The Medical School, Shiraz University of Medical Sciences, Shiraz, Iran; Histomorphometry and Stereology Research Centre, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
14
|
Di Lazzaro V, Bella R, Benussi A, Bologna M, Borroni B, Capone F, Chen KHS, Chen R, Chistyakov AV, Classen J, Kiernan MC, Koch G, Lanza G, Lefaucheur JP, Matsumoto H, Nguyen JP, Orth M, Pascual-Leone A, Rektorova I, Simko P, Taylor JP, Tremblay S, Ugawa Y, Dubbioso R, Ranieri F. Diagnostic contribution and therapeutic perspectives of transcranial magnetic stimulation in dementia. Clin Neurophysiol 2021; 132:2568-2607. [PMID: 34482205 DOI: 10.1016/j.clinph.2021.05.035] [Citation(s) in RCA: 86] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 04/22/2021] [Accepted: 05/28/2021] [Indexed: 02/07/2023]
Abstract
Transcranial magnetic stimulation (TMS) is a powerful tool to probe in vivo brain circuits, as it allows to assess several cortical properties such asexcitability, plasticity and connectivity in humans. In the last 20 years, TMS has been applied to patients with dementia, enabling the identification of potential markers of thepathophysiology and predictors of cognitive decline; moreover, applied repetitively, TMS holds promise as a potential therapeutic intervention. The objective of this paper is to present a comprehensive review of studies that have employed TMS in dementia and to discuss potential clinical applications, from the diagnosis to the treatment. To provide a technical and theoretical framework, we first present an overview of the basic physiological mechanisms of the application of TMS to assess cortical excitability, excitation and inhibition balance, mechanisms of plasticity and cortico-cortical connectivity in the human brain. We then review the insights gained by TMS techniques into the pathophysiology and predictors of progression and response to treatment in dementias, including Alzheimer's disease (AD)-related dementias and secondary dementias. We show that while a single TMS measure offers low specificity, the use of a panel of measures and/or neurophysiological index can support the clinical diagnosis and predict progression. In the last part of the article, we discuss the therapeutic uses of TMS. So far, only repetitive TMS (rTMS) over the left dorsolateral prefrontal cortex and multisite rTMS associated with cognitive training have been shown to be, respectively, possibly (Level C of evidence) and probably (Level B of evidence) effective to improve cognition, apathy, memory, and language in AD patients, especially at a mild/early stage of the disease. The clinical use of this type of treatment warrants the combination of brain imaging techniques and/or electrophysiological tools to elucidate neurobiological effects of neurostimulation and to optimally tailor rTMS treatment protocols in individual patients or specific patient subgroups with dementia or mild cognitive impairment.
Collapse
Affiliation(s)
- Vincenzo Di Lazzaro
- Unit of Neurology, Neurophysiology, Neurobiology, Department of Medicine, Università Campus Bio-Medico di Roma, Rome, Italy.
| | - Rita Bella
- Department of Medical and Surgical Sciences and Advanced Technologies, Section of Neurosciences, University of Catania, Catania, Italy
| | - Alberto Benussi
- Centre for Neurodegenerative Disorders, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Matteo Bologna
- Department of Human Neurosciences, Sapienza University of Rome, Italy; IRCCS Neuromed, Pozzilli, IS, Italy
| | - Barbara Borroni
- Centre for Neurodegenerative Disorders, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Fioravante Capone
- Unit of Neurology, Neurophysiology, Neurobiology, Department of Medicine, Università Campus Bio-Medico di Roma, Rome, Italy
| | - Kai-Hsiang S Chen
- Department of Neurology, National Taiwan University Hospital Hsin-Chu Branch, Hsin-Chu, Taiwan
| | - Robert Chen
- Division of Neurology, Department of Medicine, University of Toronto, Toronto, Canada; Division of Brain, Imaging& Behaviour, Krembil Brain Institute, Toronto, Canada
| | | | - Joseph Classen
- Department of Neurology, University Hospital Leipzig, Leipzig University Medical Center, Germany
| | - Matthew C Kiernan
- Department of Neurology, Royal Prince Alfred Hospital, Brain and Mind Centre, University of Sydney, Sydney, New South Wales, Australia
| | - Giacomo Koch
- Non Invasive Brain Stimulation Unit/Department of Behavioral and Clinical Neurology, Santa Lucia Foundation IRCCS, Rome, Italy; Department of Neuroscience and Rehabilitation, University of Ferrara, Ferrara, Italy
| | - Giuseppe Lanza
- Department of Surgery and Medical-Surgical Specialties, University of Catania, Catania, Italy; Department of Neurology IC, Oasi Research Institute-IRCCS, Troina, Italy
| | - Jean-Pascal Lefaucheur
- ENT Team, EA4391, Faculty of Medicine, Paris Est Créteil University, Créteil, France; Clinical Neurophysiology Unit, Department of Physiology, Henri Mondor Hospital, Assistance Publique - Hôpitaux de Paris, Créteil, France
| | | | - Jean-Paul Nguyen
- Pain Center, clinique Bretéché, groupe ELSAN, Multidisciplinary Pain, Palliative and Supportive care Center, UIC 22/CAT2 and Laboratoire de Thérapeutique (EA3826), University Hospital, Nantes, France
| | - Michael Orth
- University Hospital of Old Age Psychiatry and Psychotherapy, University of Bern, Bern, Switzerland; Swiss Huntington's Disease Centre, Siloah, Bern, Switzerland
| | - Alvaro Pascual-Leone
- Hinda and Arthur Marcus Institute for Aging Research, Center for Memory Health, Hebrew SeniorLife, USA; Department of Neurology, Harvard Medical School, Boston, MA, USA; Guttmann Brain Health Institute, Universitat Autonoma Barcelona, Spain
| | - Irena Rektorova
- Applied Neuroscience Research Group, Central European Institute of Technology, Masaryk University (CEITEC MU), Brno, Czech Republic; Department of Neurology, St. Anne's University Hospital and Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Patrik Simko
- Applied Neuroscience Research Group, Central European Institute of Technology, Masaryk University (CEITEC MU), Brno, Czech Republic; Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - John-Paul Taylor
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Sara Tremblay
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, ON, Canada; Royal Ottawa Institute of Mental Health Research, Ottawa, ON, Canada
| | - Yoshikazu Ugawa
- Department of Human Neurophysiology, School of Medicine, Fukushima Medical University, Fukushima, Japan
| | - Raffaele Dubbioso
- Department of Neurosciences, Reproductive Sciences and Odontostomatology, University of Naples "Federico II", Naples, Italy
| | - Federico Ranieri
- Unit of Neurology, Department of Neuroscience, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| |
Collapse
|
15
|
Xu R, He Q, Wang Y, Yang Y, Guo ZN. Therapeutic Potential of Remote Ischemic Conditioning in Vascular Cognitive Impairment. Front Cell Neurosci 2021; 15:706759. [PMID: 34413726 PMCID: PMC8370253 DOI: 10.3389/fncel.2021.706759] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Accepted: 06/29/2021] [Indexed: 12/21/2022] Open
Abstract
Vascular cognitive impairment (VCI) is a heterogeneous disease caused by a variety of cerebrovascular diseases. Patients with VCI often present with slower cognitive processing speed and poor executive function, which affects their independence in daily life, thus increasing social burden. Remote ischemic conditioning (RIC) is a non-invasive and efficient intervention that triggers endogenous protective mechanisms to generate neuroprotection. Over the past decades, evidence from basic and clinical research has shown that RIC is promising for the treatment of VCI. To further our understanding of RIC and improve the management of VCI, we summarize the evidence on the therapeutic potential of RIC in relation to the risk factors and pathobiologies of VCI, including reducing the risk of recurrent stroke, decreasing high blood pressure, improving cerebral blood flow, restoring white matter integrity, protecting the neurovascular unit, attenuating oxidative stress, and inhibiting the inflammatory response.
Collapse
Affiliation(s)
- Rui Xu
- Department of Neurology, Stroke Center & Clinical Trial and Research Center for Stroke, The First Hospital of Jilin University, Changchun, China.,China National Comprehensive Stroke Center, Changchun, China.,Jilin Provincial Key Laboratory of Cerebrovascular Disease, Changchun, China
| | - Qianyan He
- Department of Neurology, Stroke Center & Clinical Trial and Research Center for Stroke, The First Hospital of Jilin University, Changchun, China.,China National Comprehensive Stroke Center, Changchun, China.,Jilin Provincial Key Laboratory of Cerebrovascular Disease, Changchun, China
| | - Yan Wang
- Department of Neurology, Stroke Center & Clinical Trial and Research Center for Stroke, The First Hospital of Jilin University, Changchun, China.,China National Comprehensive Stroke Center, Changchun, China.,Jilin Provincial Key Laboratory of Cerebrovascular Disease, Changchun, China
| | - Yi Yang
- Department of Neurology, Stroke Center & Clinical Trial and Research Center for Stroke, The First Hospital of Jilin University, Changchun, China.,China National Comprehensive Stroke Center, Changchun, China.,Jilin Provincial Key Laboratory of Cerebrovascular Disease, Changchun, China
| | - Zhen-Ni Guo
- Department of Neurology, Stroke Center & Clinical Trial and Research Center for Stroke, The First Hospital of Jilin University, Changchun, China.,China National Comprehensive Stroke Center, Changchun, China.,Jilin Provincial Key Laboratory of Cerebrovascular Disease, Changchun, China
| |
Collapse
|
16
|
Antczak J, Rusin G, Słowik A. Transcranial Magnetic Stimulation as a Diagnostic and Therapeutic Tool in Various Types of Dementia. J Clin Med 2021; 10:jcm10132875. [PMID: 34203558 PMCID: PMC8267667 DOI: 10.3390/jcm10132875] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Revised: 06/23/2021] [Accepted: 06/25/2021] [Indexed: 02/03/2023] Open
Abstract
Dementia is recognized as a healthcare and social burden and remains challenging in terms of proper diagnosis and treatment. Transcranial magnetic stimulation (TMS) is a diagnostic and therapeutic tool in various neurological diseases that noninvasively investigates cortical excitability and connectivity and can induce brain plasticity. This article reviews findings on TMS in common dementia types as well as therapeutic results. Alzheimer’s disease (AD) is characterized by increased cortical excitability and reduced cortical inhibition, especially as mediated by cholinergic neurons and as documented by impairment of short latency inhibition (SAI). In vascular dementia, excitability is also increased. SAI may have various outcomes, which probably reflects its frequent overlap with AD. Dementia with Lewy bodies (DLB) is associated with SAI decrease. Motor cortical excitability is usually normal, reflecting the lack of corticospinal tract involvement. DLB and other dementia types are also characterized by impairment of short interval intracortical inhibition. In frontotemporal dementia, cortical excitability is increased, but SAI is normal. Repetitive transcranial magnetic stimulation has the potential to improve cognitive function. It has been extensively studied in AD, showing promising results after multisite stimulation. TMS with electroencephalography recording opens new possibilities for improving diagnostic accuracy; however, more studies are needed to support the existing data.
Collapse
|
17
|
El-Derany MO, Noureldein MH. Bone marrow mesenchymal stem cells and their derived exosomes resolve doxorubicin-induced chemobrain: critical role of their miRNA cargo. Stem Cell Res Ther 2021; 12:322. [PMID: 34090498 PMCID: PMC8180158 DOI: 10.1186/s13287-021-02384-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Accepted: 05/13/2021] [Indexed: 02/06/2023] Open
Abstract
Background Doxorubicin (DOX), a widely used chemotherapeutic agent, can cause neurodegeneration in the brain, which leads to a condition known as chemobrain. In fact, chemobrain is a deteriorating condition which adversely affects the lives of cancer survivors. This study aimed to examine the potential therapeutic effects of bone marrow mesenchymal stem cells (BMSCs) and their derived exosomes (BMSCs-Exo) in DOX-induced chemobrain in rat models. Methods Chemobrain was induced by exposing rats to DOX (2 mg/kg, i.p) once weekly for 4 consecutive weeks. After 48 h of the last DOX dose, a subset of rats was supplied with either an intravenous injection of BMSCs (1 × 106) or a single dose of 150 μg of BMSCs-Exo. Behavioral tests were conducted 7 days post injection. Rats were sacrificed after 14 days from BMSCs or BMSCs-Exo injection. Results BMSCs and BMSCs-Exo successfully restored DOX-induced cognitive and behavioral distortion. These actions were mediated via decreasing hippocampal neurodegeneration and neural demyelination through upregulating neural myelination factors (myelin%, Olig2, Opalin expression), neurotropic growth factors (BDNF, FGF-2), synaptic factors (synaptophysin), and fractalkine receptor expression (Cx3cr1). Halting neurodegeneration in DOX-induced chemobrain was achieved through epigenetic induction of key factors in Wnt/β-catenin and hedgehog signaling pathways mediated primarily by the most abundant secreted exosomal miRNAs (miR-21-5p, miR-125b-5p, miR-199a-3p, miR-24-3p, let-7a-5p). Moreover, BMSCs and BMSCs-Exo significantly abrogate the inflammatory state (IL-6, TNF-α), apoptotic state (BAX/Bcl2), astrocyte, and microglia activation (GFAP, IBA-1) in DOX-induced chemobrain with a significant increase in the antioxidant mediators (GSH, GPx, SOD activity). Conclusions BMSCs and their derived exosomes offer neuroprotection against DOX-induced chemobrain via genetic and epigenetic abrogation of hippocampal neurodegeneration through modulating Wnt/β-catenin and hedgehog signaling pathways and through reducing inflammatory, apoptotic, and oxidative stress state. Graphical abstract Proposed mechanisms of the protective effects of bone marrow stem cells (BMSCs) and their exosomes (BMSCs-Exo) in doxorubicin (DOX)-induced chemobrain. Blue arrows: induce. Red arrows: inhibit.
![]() Supplementary Information The online version contains supplementary material available at 10.1186/s13287-021-02384-9.
Collapse
Affiliation(s)
- Marwa O El-Derany
- Department of Biochemistry, Faculty of Pharmacy, Ain Shams University, Cairo, 11566, Egypt.
| | - Mohamed H Noureldein
- Department of Biochemistry, Faculty of Pharmacy, Ain Shams University, Cairo, 11566, Egypt.,Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut, Lebanon.,American University of Beirut Diabetes Program, Beirut, Lebanon
| |
Collapse
|
18
|
Andrzejewska A, Dabrowska S, Lukomska B, Janowski M. Mesenchymal Stem Cells for Neurological Disorders. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:2002944. [PMID: 33854883 PMCID: PMC8024997 DOI: 10.1002/advs.202002944] [Citation(s) in RCA: 172] [Impact Index Per Article: 43.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/02/2020] [Revised: 11/23/2020] [Indexed: 05/13/2023]
Abstract
Neurological disorders are becoming a growing burden as society ages, and there is a compelling need to address this spiraling problem. Stem cell-based regenerative medicine is becoming an increasingly attractive approach to designing therapies for such disorders. The unique characteristics of mesenchymal stem cells (MSCs) make them among the most sought after cell sources. Researchers have extensively studied the modulatory properties of MSCs and their engineering, labeling, and delivery methods to the brain. The first part of this review provides an overview of studies on the application of MSCs to various neurological diseases, including stroke, traumatic brain injury, spinal cord injury, multiple sclerosis, amyotrophic lateral sclerosis, Alzheimer's disease, Huntington's disease, Parkinson's disease, and other less frequently studied clinical entities. In the second part, stem cell delivery to the brain is focused. This fundamental but still understudied problem needs to be overcome to apply stem cells to brain diseases successfully. Here the value of cell engineering is also emphasized to facilitate MSC diapedesis, migration, and homing to brain areas affected by the disease to implement precision medicine paradigms into stem cell-based therapies.
Collapse
Affiliation(s)
- Anna Andrzejewska
- NeuroRepair DepartmentMossakowski Medical Research CentrePASWarsaw02‐106Poland
| | - Sylwia Dabrowska
- NeuroRepair DepartmentMossakowski Medical Research CentrePASWarsaw02‐106Poland
| | - Barbara Lukomska
- NeuroRepair DepartmentMossakowski Medical Research CentrePASWarsaw02‐106Poland
| | - Miroslaw Janowski
- NeuroRepair DepartmentMossakowski Medical Research CentrePASWarsaw02‐106Poland
- Center for Advanced Imaging ResearchDepartment of Diagnostic Radiology and Nuclear MedicineUniversity of Maryland Marlene and Stewart Greenebaum Comprehensive Cancer CenterUniversity of MarylandBaltimoreMD21201‐1595USA
- Tumor Immunology and Immunotherapy ProgramUniversity of Maryland Marlene and Stewart Greenebaum Comprehensive Cancer CenterUniversity of MarylandBaltimoreMD21201‐1595USA
| |
Collapse
|
19
|
Feng S, Wang S, Sun S, Su H, Zhang L. Effects of combination treatment with transcranial magnetic stimulation and bone marrow mesenchymal stem cell transplantation or Raf inhibition on spinal cord injury in rats. Mol Med Rep 2021; 23:294. [PMID: 33649786 PMCID: PMC7930933 DOI: 10.3892/mmr.2021.11934] [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: 09/02/2020] [Accepted: 01/07/2021] [Indexed: 11/07/2022] Open
Abstract
Spinal cord injury (SCI) remains a global challenge due to limited treatment strategies. Transcranial magnetic stimulation (TMS), bone marrow mesenchymal stem cell (BMSC) transplantation and downregulation of Raf/MEK/ERK signaling effectively improve SCI. The combination of BMSCs and TMS displays synergistic effects on vascular dementia. However, whether TMS displays a synergistic effect when combined with BMSC transplantation or Raf inhibitor (RafI) therapy for the treatment of SCI is not completely understood. The present study aimed to compare the therapeutic effect of monotherapy and combination therapy on SCI. In the present study, 8‑week‑old female Sprague Dawley rats were used to establish a model of SCI using the weight‑drop method followed by treatment with monotherapy (TMS, BMSCs or RafI) or combination therapy (TMS+BMSCs or TMS+RafI). The effect of monotherapy and combination therapy on locomotor function, pathological alterations, neuronal apoptosis and expression of axonal regeneration‑associated factors and Raf/MEK/ERK signaling‑associated proteins in the spinal cord was analyzed by Basso, Beattie and Bresnahan (BBB) scoring, hematoxylin and eosin staining, TUNEL‑neuronal nuclei (NeuN) staining and immunofluorescence or western blotting, respectively. The results demonstrated that compared with untreated SCI model rats, monotherapy significantly enhanced locomotor functional recovery, as evidenced by higher BBB scores, and slightly alleviated histopathological lesions of the spinal cord in SCI model rats. Furthermore, monotherapy markedly suppressed neuronal apoptosis and promoted axonal regeneration, as well as inhibiting astroglial activation in SCI model rats. The aforementioned results were demonstrated by significantly decreased numbers of apoptotic neurons, markedly decreased expression levels of glial fibrillary acidic protein (GFAP), significantly increased numbers of NeuN+ cells, markedly increased expression levels of growth‑associated protein 43 (GAP‑43) and significantly upregulated nerve growth factor (NGF) and brain derived neurotrophic factor (BDNF) expression levels in monotherapy groups (excluding the RafI monotherapy group) compared with untreated SCI model rats. In addition, monotherapy markedly suppressed activation of the Raf/MEK/ERK signaling pathway, as evidenced by significantly reduced p‑Raf/Raf, p‑MEK/MEK and p‑ERK/ERK protein expression levels in monotherapy groups (excluding the BMSC monotherapy group) compared with untreated SCI model rats. Notably, combination therapy further alleviated SCI‑induced spinal cord lesions and neuronal apoptosis, increased GAP‑43, NGF and BDNF expression levels, downregulated GFAP expression levels and inhibited activation of the Raf/MEK/ERK signaling pathway in SCI model rats compared with the corresponding monotherapy groups. Therefore, it was hypothesized that compared with monotherapy, combination therapy displayed an improved therapeutic effect on SCI by further suppressing Raf/MEK/ERK signaling. The results of the present study provided an important basis for the clinical application of combination therapy.
Collapse
Affiliation(s)
- Sining Feng
- Department of Rehabilitation, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| | - Shuai Wang
- Department of Rehabilitation, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| | - Shi Sun
- Department of Rehabilitation, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| | - Hao Su
- Department of Rehabilitation, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| | - Lixin Zhang
- Department of Rehabilitation, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| |
Collapse
|
20
|
Evaluation and Treatment of Vascular Cognitive Impairment by Transcranial Magnetic Stimulation. Neural Plast 2020. [PMID: 33193753 DOI: 10.1155/2020/8820881.] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The exact relationship between cognitive functioning, cortical excitability, and synaptic plasticity in dementia is not completely understood. Vascular cognitive impairment (VCI) is deemed to be the most common cognitive disorder in the elderly since it encompasses any degree of vascular-based cognitive decline. In different cognitive disorders, including VCI, transcranial magnetic stimulation (TMS) can be exploited as a noninvasive tool able to evaluate in vivo the cortical excitability, the propension to undergo neural plastic phenomena, and the underlying transmission pathways. Overall, TMS in VCI revealed enhanced cortical excitability and synaptic plasticity that seem to correlate with the disease process and progression. In some patients, such plasticity may be considered as an adaptive response to disease progression, thus allowing the preservation of motor programming and execution. Recent findings also point out the possibility to employ TMS to predict cognitive deterioration in the so-called "brains at risk" for dementia, which may be those patients who benefit more of disease-modifying drugs and rehabilitative or neuromodulatory approaches, such as those based on repetitive TMS (rTMS). Finally, TMS can be exploited to select the responders to specific drugs in the attempt to maximize the response and to restore maladaptive plasticity. While no single TMS index owns enough specificity, a panel of TMS-derived measures can support VCI diagnosis and identify early markers of progression into dementia. This work reviews all TMS and rTMS studies on VCI. The aim is to evaluate how cortical excitability, plasticity, and connectivity interact in the pathophysiology of the impairment and to provide a translational perspective towards novel treatments of these patients. Current pitfalls and limitations of both studies and techniques are also discussed, together with possible solutions and future research agenda.
Collapse
|
21
|
Kim J, Lee Y, Lee S, Kim K, Song M, Lee J. Mesenchymal Stem Cell Therapy and Alzheimer's Disease: Current Status and Future Perspectives. J Alzheimers Dis 2020; 77:1-14. [PMID: 32741816 DOI: 10.3233/jad-200219] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Alzheimer's disease (AD) is the most common progressive neurodegenerative disease worldwide, but its cause remains unclear. Although a few drugs can provide temporary and partial relief of symptoms in some patients, no curative treatment is available. Therefore, attention has been focused on research using stem cells to treat AD. Among stem cells, mesenchymal stem cells (MSCs) have been used to treat the related pathologies in animal models of AD, and other neurodegenerative disease. This review describes latest research trends on the use of MSC-based therapies in AD and its action of mechanism. MSCs have several beneficial effects. They would be specified as the reduction of neuroinflammation, the elimination of amyloid-β, neurofibrillary tangles, and abnormal protein degradation, the promotion of autophagy-associated and blood-brain barrier recoveries, the upregulation of acetylcholine levels, improved cognition, and the recovery of mitochondrial transport. Therefore, this review describes the latest research trends in MSC-based therapy for AD by demonstrating the importance of MSC-based therapy and understanding of its mechanisms in AD and discusses the limitations and perspectives of stem cell therapy in AD.
Collapse
Affiliation(s)
- Jieun Kim
- Department of Pharmacy, College of Pharmacy, Pusan National University, Busan, Republic of Korea
| | - Yujeong Lee
- Department of Pharmacy, College of Pharmacy, Pusan National University, Busan, Republic of Korea.,Cognitive Science Research Group, Korea Brain Research Institute, Daegu, Republic of Korea
| | - Seulah Lee
- Department of Pharmacy, College of Pharmacy, Pusan National University, Busan, Republic of Korea
| | - Kipom Kim
- Brain Research Core Facilities, Korea Brain Research Institute, Daegu, Republic of Korea
| | - Minjung Song
- Metabolic Research Laboratories and Medical Research Council Metabolic Diseases Unit, Wellcome Trust - Medical Research Council Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Jaewon Lee
- Department of Pharmacy, College of Pharmacy, Pusan National University, Busan, Republic of Korea
| |
Collapse
|
22
|
Vinciguerra L, Lanza G, Puglisi V, Fisicaro F, Pennisi M, Bella R, Cantone M. Update on the Neurobiology of Vascular Cognitive Impairment: From Lab to Clinic. Int J Mol Sci 2020; 21:E2977. [PMID: 32340195 PMCID: PMC7215552 DOI: 10.3390/ijms21082977] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2020] [Revised: 04/20/2020] [Accepted: 04/21/2020] [Indexed: 02/07/2023] Open
Abstract
In the last years, there has been a significant growth in the literature exploring the pathophysiology of vascular cognitive impairment (VCI). As an "umbrella term" encompassing any degree of vascular-related cognitive decline, VCI is deemed to be the most common cognitive disorder in the elderly, with a significant impact on social and healthcare expenses. Interestingly, some of the molecular, biochemical, and electrophysiological abnormalities detected in VCI seem to correlate with disease process and progression, eventually promoting an adaptive plasticity in some patients and a maladaptive, dysfunctional response in others. However, the exact relationships between vascular lesion, cognition, and neuroplasticity are not completely understood. Recent findings point out also the possibility to identify a panel of markers able to predict cognitive deterioration in the so-called "brain at risk" for vascular or mixed dementia. This will be of pivotal importance when designing trials of disease-modifying drugs or non-pharmacological approaches, including non-invasive neuromodulatory techniques. Taken together, these advances could make VCI a potentially preventable cause of both vascular and degenerative dementia in late life. This review provides a timely update on the recent serological, cerebrospinal fluid, histopathological, imaging, and neurophysiological studies on this "cutting-edge" topic, including the limitations, future perspectives and translational implications in the diagnosis and management of VCI patients.
Collapse
Affiliation(s)
- Luisa Vinciguerra
- Department of Neurology and Stroke Unit, ASST Cremona, 26100 Cremona, Italy; (L.V.); (V.P.)
| | - Giuseppe Lanza
- Department of Surgery and Medical-Surgical Specialties, University of Catania, 95123 Catania, Italy
- Department of Neurology IC, Oasi Research Institute – IRCCS, 94018 Troina, Italy
| | - Valentina Puglisi
- Department of Neurology and Stroke Unit, ASST Cremona, 26100 Cremona, Italy; (L.V.); (V.P.)
| | - Francesco Fisicaro
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (F.F.); (M.P.)
| | - Manuela Pennisi
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (F.F.); (M.P.)
| | - Rita Bella
- Department of Medical and Surgical Sciences and Advanced Technologies, University of Catania, 95123 Catania, Italy;
| | - Mariagiovanna Cantone
- Department of Neurology, Sant’Elia Hospital, ASP Caltanissetta, 93100 Caltanissetta, Italy;
| |
Collapse
|
23
|
Update on the Neurobiology of Vascular Cognitive Impairment: From Lab to Clinic. Int J Mol Sci 2020. [PMID: 32340195 DOI: 10.3390/ijms21082977.] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
In the last years, there has been a significant growth in the literature exploring the pathophysiology of vascular cognitive impairment (VCI). As an "umbrella term" encompassing any degree of vascular-related cognitive decline, VCI is deemed to be the most common cognitive disorder in the elderly, with a significant impact on social and healthcare expenses. Interestingly, some of the molecular, biochemical, and electrophysiological abnormalities detected in VCI seem to correlate with disease process and progression, eventually promoting an adaptive plasticity in some patients and a maladaptive, dysfunctional response in others. However, the exact relationships between vascular lesion, cognition, and neuroplasticity are not completely understood. Recent findings point out also the possibility to identify a panel of markers able to predict cognitive deterioration in the so-called "brain at risk" for vascular or mixed dementia. This will be of pivotal importance when designing trials of disease-modifying drugs or non-pharmacological approaches, including non-invasive neuromodulatory techniques. Taken together, these advances could make VCI a potentially preventable cause of both vascular and degenerative dementia in late life. This review provides a timely update on the recent serological, cerebrospinal fluid, histopathological, imaging, and neurophysiological studies on this "cutting-edge" topic, including the limitations, future perspectives and translational implications in the diagnosis and management of VCI patients.
Collapse
|
24
|
Stem cells and vascular dementia: from basic science to the clinic. Cell Tissue Bank 2020; 21:349-360. [PMID: 32248316 DOI: 10.1007/s10561-020-09829-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Accepted: 03/23/2020] [Indexed: 02/07/2023]
Abstract
Vascular dementia (VD) is the second most common cause of dementia following Alzheimer's disease (AD). The major symptoms of VD including memory loss, language deficits and impairment of executive functions. Its specific etiology and pathogenesis remain unknown. Currently, treatment options of VD are still limited. The therapeutic strategies aim to control the vascular risk factors and improve the cognitive function. In recent years, cell therapy for neurodegenerative diseases has attracted a great deal of attention. Evidence suggested that stem cell transplantation could improve the symptoms of cerebral infarction and AD. Therefore, it may serve as a potential therapy for VD. We summarized the latest research results both in vitro and in vivo. Further, the clinical trial of stem cell transplantation in VD patients was also reviewed. Finally, the limitations and future directions of cell therapy in VD treatment were discussed.
Collapse
|
25
|
Cantone M, Lanza G, Fisicaro F, Pennisi M, Bella R, Di Lazzaro V, Di Pino G. Evaluation and Treatment of Vascular Cognitive Impairment by Transcranial Magnetic Stimulation. Neural Plast 2020; 2020:8820881. [PMID: 33193753 PMCID: PMC7641667 DOI: 10.1155/2020/8820881] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 09/23/2020] [Accepted: 10/12/2020] [Indexed: 02/07/2023] Open
Abstract
The exact relationship between cognitive functioning, cortical excitability, and synaptic plasticity in dementia is not completely understood. Vascular cognitive impairment (VCI) is deemed to be the most common cognitive disorder in the elderly since it encompasses any degree of vascular-based cognitive decline. In different cognitive disorders, including VCI, transcranial magnetic stimulation (TMS) can be exploited as a noninvasive tool able to evaluate in vivo the cortical excitability, the propension to undergo neural plastic phenomena, and the underlying transmission pathways. Overall, TMS in VCI revealed enhanced cortical excitability and synaptic plasticity that seem to correlate with the disease process and progression. In some patients, such plasticity may be considered as an adaptive response to disease progression, thus allowing the preservation of motor programming and execution. Recent findings also point out the possibility to employ TMS to predict cognitive deterioration in the so-called "brains at risk" for dementia, which may be those patients who benefit more of disease-modifying drugs and rehabilitative or neuromodulatory approaches, such as those based on repetitive TMS (rTMS). Finally, TMS can be exploited to select the responders to specific drugs in the attempt to maximize the response and to restore maladaptive plasticity. While no single TMS index owns enough specificity, a panel of TMS-derived measures can support VCI diagnosis and identify early markers of progression into dementia. This work reviews all TMS and rTMS studies on VCI. The aim is to evaluate how cortical excitability, plasticity, and connectivity interact in the pathophysiology of the impairment and to provide a translational perspective towards novel treatments of these patients. Current pitfalls and limitations of both studies and techniques are also discussed, together with possible solutions and future research agenda.
Collapse
Affiliation(s)
- Mariagiovanna Cantone
- 1Department of Neurology, Sant'Elia Hospital, ASP Caltanissetta, Caltanissetta 93100, Italy
| | - Giuseppe Lanza
- 2Department of Surgery and Medical-Surgical Specialties, University of Catania, Catania 95123, Italy
- 3Department of Neurology IC, Oasi Research Institute–IRCCS, Troina 94108, Italy
| | - Francesco Fisicaro
- 4Department of Biomedical and Biotechnological Sciences, University of Catania, Catania 95123, Italy
| | - Manuela Pennisi
- 4Department of Biomedical and Biotechnological Sciences, University of Catania, Catania 95123, Italy
| | - Rita Bella
- 5Department of Medical and Surgical Sciences and Advanced Technologies, University of Catania, Catania 95123, Italy
| | - Vincenzo Di Lazzaro
- 6Unit of Neurology, Neurophysiology, Neurobiology, Department of Medicine, Università Campus Bio-Medico di Roma, Rome 00128, Italy
| | - Giovanni Di Pino
- 7Research Unit of Neurophysiology and Neuroengineering of Human-Technology Interaction (NeXTlab), Università Campus Bio-Medico di Roma, Rome 00128, Italy
| |
Collapse
|
26
|
Wang X, Zhou X, Bao J, Chen Z, Tang J, Gong X, Ni J, Fang Q, Liu Y, Su M. High-Frequency Repetitive Transcranial Magnetic Stimulation Mediates Autophagy Flux in Human Bone Mesenchymal Stromal Cells via NMDA Receptor-Ca 2+-Extracellular Signal-Regulated Kinase-Mammalian Target of Rapamycin Signaling. Front Neurosci 2019; 13:1225. [PMID: 31798406 PMCID: PMC6878833 DOI: 10.3389/fnins.2019.01225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Accepted: 10/29/2019] [Indexed: 11/13/2022] Open
Abstract
Aim Repetitive transcranial magnetic stimulation (rTMS) is a non-invasive and safe technique for treatment of central and peripheral nerve injury. In recent years, this technique has been widely used in clinic, and an increasing number of studies have reported its mechanisms. In this study, we investigated the mechanisms of rTMS-mediated autophagy flux in human bone mesenchymal stromal cells (BMSCs). Methods A frequency of 50 Hz was employed. Cells were divided into five groups: (1) normal, (2) sham, (3) 0.5 T, (4) 1.0 T, and (5) 1.5 T. Cells were stimulated for 20 min/day. The levels of p62, LC3-II/I, phosphorylated extracellular signal-regulated kinase (p-ERK), ERK, phosphorylated-AKT (p-AKT), AKT, phosphorylated mammalian target of rapamycin (p-mTOR), mTOR, phosphorylated protein kinase A (p-PKA), PKA, phosphorylated epidermal growth factor receptor (p-EGFR), EGFR, Nanog, Oct4, Sox2, and NMDA receptor (NMDAR1) were investigated by western blotting. Intracellular calcium (Ca2+) levels were quantified by flow cytometry. p62 and LC3 expression was also assessed by immunofluorescence analysis. Results In the 0.5 T group, rTMS increased the expression of LC3-II/I, p-ERK/ERK, and NMDAR1 and decreased the levels of p62 and p-mTOR/mTOR than in the normal group. The ratio of p-AKT/AKT, p-PKA/PKA, and p-EGFR/EGFR and the expression of Nanog, Oct4, and Sox2 remained unchanged. Immunofluorescence analysis revealed colocalization of p62 with LC3 puncta, and flow cytometry analysis displayed that Ca2+ levels were elevated. However, in the 1.0 and 1.5 T groups, no changes in the expression of these autophagy markers were observed. Conclusion In the 0.5 T group, high-frequency rTMS can induce autophagy through NMDAR–Ca2+–ERK–mTOR signaling in BMSCs. In the 1.0 and 1.5 T groups, autophagy is not activated.
Collapse
Affiliation(s)
- Xinlong Wang
- Department of Physical Medicine and Rehabilitation, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Xing Zhou
- Department of Physical Medicine and Rehabilitation, The First Affiliated Hospital of Soochow University, Suzhou, China.,Department of Children's Health Care Center, Wuxi Children's Hospital, Wuxi, China
| | - Jie Bao
- Sport Rehabilitation Center of Physical and Education School, Soochow University, Suzhou, China
| | - Zhiguo Chen
- Department of Neurology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Jingzhao Tang
- Department of Physical Medicine and Rehabilitation, The First Affiliated Hospital of Soochow University, Suzhou, China.,Department of Medical Rehabilitation, Community Health Service Center of Yangming Street, Wuxi, China
| | - Xueyang Gong
- Department of Physical Medicine and Rehabilitation, The First Affiliated Hospital of Soochow University, Suzhou, China.,Department of Cardiopulmonary Rehabilitation, Wuxi Tongren Rehabilitation Hospital, Wuxi, China
| | - Jing Ni
- Department of Physical Medicine and Rehabilitation, The First Affiliated Hospital of Soochow University, Suzhou, China.,Department of Geriatric Rehabilitation, Jiangsu Rongjun Hospital, Wuxi, China
| | - Qi Fang
- Department of Neurology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Yaobo Liu
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, China
| | - Min Su
- Department of Physical Medicine and Rehabilitation, The First Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
27
|
Özmert E, Arslan U. Management of Deep Retinal Capillary Ischemia by Electromagnetic Stimulation and Platelet-Rich Plasma: Preliminary Clinical Results. Adv Ther 2019; 36:2273-2286. [PMID: 31385285 DOI: 10.1007/s12325-019-01040-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2019] [Indexed: 12/22/2022]
Abstract
INTRODUCTION To investigate the efficacy of retinal electromagnetic stimulation and sub-tenon autologous platelet-rich plasma in the treatment of deep retinal capillary ischemia. METHODS The study included 28 eyes of 17 patients aged 15-76 years (mean 37.9 years) who had deep retinal capillary ischemia. Patients who had acute-onset paracentral scotoma in the last 1 month were included in the study between January 2018 and January 2019. The diagnosis of deep retinal capillary ischemia was based on clinical history and typical findings of optical coherence tomography angiography. The eyes were divided into three groups: group 1 (n = 7 eyes) received electromagnetic stimulation alone; group 2 (n = 7 eyes) received electromagnetic stimulation and sub-tenon autologous platelet-rich plasma injection; group 3 had no intervention and served as a control group (n = 14 eyes). The patients underwent ten sessions of electromagnetic stimulation in groups 1 and 2. Sub-tenon autologous platelet-rich plasma injection was performed immediately after the first, fifth, and tenth sessions of electromagnetic stimulation in group 2. The deep retinal capillary density and best corrected visual acuity changes were investigated before and after treatment at the first month. RESULTS The mean deep retinal capillary density was 52.0% before electromagnetic stimulation and 56.1% after ten sessions of application in group 1; this improvement was statistically significant (p = 0.01). In the combined treatment group (group 2), the mean deep retinal capillary density was 46.9% before the treatment and 56.5% after the treatment; this increase was also statistically significant (p = 0.01). Statistically significant best corrected visual acuity improvement (p = 0.01) could be achieved only in group 2. The combined treatment was significantly superior (p < 0.01) to treatment with only electromagnetic stimulation regarding best corrected visual acuity and deep retinal capillary density. In the control group (group 3), there was no statistically significant change (p = 0.09) in the mean deep retinal capillary density and best corrected visual acuity. CONCLUSION Treatment of the underlying cause is a priority in the treatment of deep retinal capillary ischemia. However, in the acute period, local ischemia treatment is necessary to prevent permanent retinal damage and scotomas. In mild cases, only electromagnetic stimulation, which is non-invasive and easy to use, might have a beneficial effect on deep retinal capillary density. In more severe cases, sub-tenon fresh autologous platelet-rich plasma injection together with electromagnetic stimulation may be more effective in the treatment of local ischemia of the retina in order to augment the response. FUNDING The Rapid Service Fees were funded by the Ankara University Tecnopolis Institute. CLINICAL TRIAL REGISTRATION titck.gov.tr identifier, 2018-136.
Collapse
Affiliation(s)
- Emin Özmert
- Department of Ophthalmology, Faculty of Medicine, Ankara University, Ankara, Turkey
| | - Umut Arslan
- Ankara University Technopolis, Ankara, Turkey.
| |
Collapse
|
28
|
Hare JM, Beerman I. Regenerative Medicine and the Biology of Aging. J Gerontol A Biol Sci Med Sci 2019; 74:1339-1340. [PMID: 31125054 DOI: 10.1093/gerona/glz132] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Indexed: 11/13/2022] Open
Affiliation(s)
- Joshua M Hare
- Interdisciplinary Stem Cell Institute, Department of Medicine, University of Miami, Miller School of Medicine, Florida
| | - Isabel Beerman
- Translational Gerontology Branch, National Institute on Aging, NIH, Baltimore, Maryland
| |
Collapse
|
29
|
Liang Y. Emerging Concepts and Functions of Autophagy as a Regulator of Synaptic Components and Plasticity. Cells 2019; 8:cells8010034. [PMID: 30634508 PMCID: PMC6357011 DOI: 10.3390/cells8010034] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Revised: 12/23/2018] [Accepted: 01/03/2019] [Indexed: 12/15/2022] Open
Abstract
Protein homeostasis (proteostasis) is crucial to the maintenance of neuronal integrity and function. As the contact sites between neurons, synapses rely heavily on precisely regulated protein-protein interactions to support synaptic transmission and plasticity processes. Autophagy is an effective degradative pathway that can digest cellular components and maintain cellular proteostasis. Perturbations of autophagy have been implicated in aging and neurodegeneration due to a failure to remove damaged proteins and defective organelles. Recent evidence has demonstrated that autophagosome formation is prominent at synaptic terminals and neuronal autophagy is regulated in a compartment-specific fashion. Moreover, synaptic components including synaptic proteins and vesicles, postsynaptic receptors and synaptic mitochondria are known to be degraded by autophagy, thereby contributing to the remodeling of synapses. Indeed, emerging studies indicate that modulation of autophagy may be required for different forms of synaptic plasticity and memory formation. In this review, I will discuss our current understanding of the important role of neuronal/synaptic autophagy in maintaining neuronal function by degrading synaptic components and try to propose a conceptual framework of how the degradation of synaptic components via autophagy might impact synaptic function and contribute to synaptic plasticity.
Collapse
Affiliation(s)
- YongTian Liang
- Neurogenetik, Institut für Biologie, Freie Universität Berlin, 14195 Berlin, Germany.
- NeuroCure, Cluster of Excellence, Charité Universitätsmedizin, 10117 Berlin, Germany.
| |
Collapse
|
30
|
Attenuating Spinal Cord Injury by Conditioned Medium from Bone Marrow Mesenchymal Stem Cells. J Clin Med 2018; 8:jcm8010023. [PMID: 30585207 PMCID: PMC6352201 DOI: 10.3390/jcm8010023] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2018] [Revised: 12/20/2018] [Accepted: 12/21/2018] [Indexed: 12/15/2022] Open
Abstract
Spinal cord injury (SCI) is a devastating neurological condition and might even result in death. However, current treatments are not sufficient to repair such damage. Bone marrow mesenchymal stem cells (BM-MSC) are ideal transplantable cells which have been shown to modulate the injury cascade of SCI mostly through paracrine effects. The present study investigates whether systemic administration of conditioned medium from MSCs (MSCcm) has the potential to be efficacious as an alternative to cell-based therapy for SCI. In neuron-glial cultures, MSC coculture effectively promoted neuronal connection and reduced oxygen glucose deprivation-induced cell damage. The protection was elicited even if neuron-glial culture was used to expose MSCcm, suggesting the effects possibly from released fractions of MSC. In vivo, intravenous administration of MSCcm to SCI rats significantly improved behavioral recovery from spinal cord injury, and there were increased densities of axons in the lesion site of MSCcm-treated rats compared to SCI rats. At early days postinjury, MSCcm treatment upregulated the protein levels of Olig 2 and HSP70 and also increased autophage-related proteins in the injured spinal cords. Together, these findings suggest that MSCcm treatment promotes spinal cord repair and functional recovery, possibly via activation of autophagy and enhancement of survival-related proteins.
Collapse
|