1
|
De Paolis ML, Loffredo G, Krashia P, La Barbera L, Nobili A, Cauzzi E, Babicola L, Di Segni M, Coccurello R, Puglisi-Allegra S, Latagliata EC, D'Amelio M. Repetitive prefrontal tDCS activates VTA dopaminergic neurons, resulting in attenuation of Alzheimer's Disease-like deficits in Tg2576 mice. Alzheimers Res Ther 2025; 17:94. [PMID: 40301905 PMCID: PMC12039073 DOI: 10.1186/s13195-025-01736-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2025] [Accepted: 04/07/2025] [Indexed: 05/01/2025]
Abstract
BACKGROUND Emerging evidence implicates early dysfunction of dopaminergic neurons in the Ventral Tegmental Area (VTA) as a key contributor to Alzheimer's Disease (AD) pathophysiology. Specifically, the VTA dopaminergic neurodegeneration and the consequent reduction of dopamine (DA) in mesocorticolimbic targets are associated with the onset of cognitive impairments and neuropsychiatric-like manifestations in AD animal models. Moreover, decreased midbrain volume and functional VTA disconnection are identified as predictors of accelerated progression from Mild Cognitive Impairment to AD-dementia in clinical populations. Given these findings, interventions capable of directly modulating VTA activity and augmenting DA release, despite the ongoing neurodegeneration, may hold therapeutic potential for mitigating DA-related deficits in AD. This study aims at evaluating the therapeutic potential of prefrontal transcranial Direct Current Stimulation (tDCS) in the Tg2576 mouse model of AD, exhibiting early VTA dopaminergic neurodegeneration. METHODS Repeated tDCS was applied to assess its ability to activate VTA DA neurons. We also evaluated tDCS effects on synaptic plasticity, cognitive and non-cognitive behaviours and AD-related pathology. Hippocampal DA release and Nucleus Accumbens (NAc) DA transporter (DAT) expression were measured. With immunohistochemistry we examined microglial density and morphological complexity at different disease stages. Additionally, intracellular amyloid-β (Aβ) levels and plaque burden were evaluated to determine the impact of tDCS on AD pathology. RESULTS Prefrontal tDCS enhanced the activity of VTA dopaminergic neurons, leading to increased hippocampal DA release and higher DAT levels in the NAc. The enhanced DA outflow is associated with restored CA3-CA1 synaptic plasticity and improvements in recognition memory and motivational behaviours. tDCS reduced microglial numbers and morphological complexity in Tg2576 mice at both pre-plaque stage (7-months) and at an advanced stage characterized by plaque accumulation (12-months). Notably, tDCS also decreased Aβ plaque burden, although no changes in intracellular Aβ levels were observed in younger Tg2576 mice. CONCLUSIONS These findings highlight the multifaceted therapeutic potential of prefrontal tDCS in targeting key AD pathophysiological hallmarks, including dopaminergic dysfunction, synaptic impairments, neuroinflammation and plaque deposition. As a non-invasive neuromodulatory approach, prefrontal tDCS emerges as a promising early intervention strategy to complement existing AD treatments, with the potential to improve patient outcomes and quality of life.
Collapse
Affiliation(s)
- Maria Luisa De Paolis
- Department of Medicine and Surgery, Università Campus Bio-Medico Di Roma, Via Alvaro del Portillo, 21, 00128, Rome, Italy
| | - Gilda Loffredo
- Department of Medicine and Surgery, Università Campus Bio-Medico Di Roma, Via Alvaro del Portillo, 21, 00128, Rome, Italy
| | - Paraskevi Krashia
- Department of Sciences and Technologies for Sustainable Development and One Health, Università Campus Bio-Medico Di Roma, Via Alvaro del Portillo, 21, 00128, Rome, Italy
- Department of Experimental Neurosciences, IRCCS Santa Lucia Foundation, Via del Fosso Di Fiorano, 64, 00143, Rome, Italy
| | - Livia La Barbera
- Department of Medicine and Surgery, Università Campus Bio-Medico Di Roma, Via Alvaro del Portillo, 21, 00128, Rome, Italy
- Department of Experimental Neurosciences, IRCCS Santa Lucia Foundation, Via del Fosso Di Fiorano, 64, 00143, Rome, Italy
| | - Annalisa Nobili
- Department of Medicine and Surgery, Università Campus Bio-Medico Di Roma, Via Alvaro del Portillo, 21, 00128, Rome, Italy
- Department of Experimental Neurosciences, IRCCS Santa Lucia Foundation, Via del Fosso Di Fiorano, 64, 00143, Rome, Italy
| | - Emma Cauzzi
- Department of Medicine and Surgery, Università Campus Bio-Medico Di Roma, Via Alvaro del Portillo, 21, 00128, Rome, Italy
| | - Lucy Babicola
- Department of Experimental Neurosciences, IRCCS Santa Lucia Foundation, Via del Fosso Di Fiorano, 64, 00143, Rome, Italy
- Department of Psychology, Sapienza University of Rome, P.Le Aldo Moro, 5, 00185, Rome, Italy
| | - Matteo Di Segni
- Child Psychopathology Unit, IRCCS Eugenio Medea, Via Don Luigi Monza, 20, 23842, Bosisio Parini, Italy
| | - Roberto Coccurello
- Department of Experimental Neurosciences, IRCCS Santa Lucia Foundation, Via del Fosso Di Fiorano, 64, 00143, Rome, Italy
- National Research Council (CNR), Institute for Complex System (ISC), Via Dei Taurini, 19, 00185, Rome, Italy
| | - Stefano Puglisi-Allegra
- Istituto Di Ricovero E Cura a Carattere Scientifico (IRCCS) Neuromed, Via Atinense, 18, 86077, Pozzilli, Italy
| | - Emanuele Claudio Latagliata
- Department of Experimental Neurosciences, IRCCS Santa Lucia Foundation, Via del Fosso Di Fiorano, 64, 00143, Rome, Italy.
- Department of Psychology, International Telematic University Uninettuno, Corso Vittorio Emanuele II, 39, 00186, Rome, Italy.
| | - Marcello D'Amelio
- Department of Medicine and Surgery, Università Campus Bio-Medico Di Roma, Via Alvaro del Portillo, 21, 00128, Rome, Italy.
- Department of Experimental Neurosciences, IRCCS Santa Lucia Foundation, Via del Fosso Di Fiorano, 64, 00143, Rome, Italy.
| |
Collapse
|
2
|
Javanmardi S, Moradpour F, Veisi M, Omidian N, Kavyannejad R. Effects of a mitochondrial calcium uniporter and P-selectin inhibitors on neural injury induced by global cerebral ischemia-reperfusion in male rats. Metab Brain Dis 2025; 40:150. [PMID: 40085331 DOI: 10.1007/s11011-025-01570-5] [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: 10/20/2024] [Accepted: 03/03/2025] [Indexed: 03/16/2025]
Abstract
Neural injury following ischemia-reperfusion (I/R) is induced by multiple pathophysiological pathways. This study aimed to use mitochondrial calcium channel and p-selectin inhibitors to weaken these pathways. One hundred and two rats were randomly divided into six groups. In the sham group, cerebral I/R induction and drug intervention were not performed. In the I/R group, cerebral I/R induction was induced. In the RR + FCN group, animals received only ruthenium red (RR) and fucoidan (FCN) intraperitoneally without I/R induction. In the I/R + RR group, animals received RR during the cerebral I/R period. In the I/R + FCN group, FCN was administered along with cerebral I/R. In the I/R +(RR + FCN) group, animals exposed to cerebral I/R received a combination of RR and FCN simultaneously. The shuttle box and new object tests were used to assess learning and memory. The superoxide dismutase (SOD), malondialdehyde (MDA), interleukin-1 beta (IL-1β), and tumor necrosis factor-alpha (TNF-α) levels in the hippocampus were measured. Neuronal death in the hippocampal CA1 area was assessed via hematoxylin-eosin staining. FCN and RR significantly decreased the tissue MDA, IL-1β, TNF-α levels while increased the SOD level. These inhibitors significantly reduced learning disorders and cerebral edema following I/R. The rate of neuronal death was significantly lower in each of the receiving RR and FCN groups. This study revealed that the use of FCN and RR significantly attenuated the pathways associated with oxidative stress and inflammation as well as neuronal death following cerebral I/R, thereby reducing learning and memory impairments. The effects of neuroprotection were further determined when two inhibitors were used simultaneously. HIGHLIGHTS: Cerebral ischemia-reperfusion is associated with many neurological, sensory and motor defects. Multiple pathways of neural pathophysiology are activated during cerebral ischemia-reperfusion. The Administration of ruthenium and fucoidan weakens inflammatory pathways, oxidative stress, and learning dysfunctions caused by cerebral ischemia and reperfusion. Stronger Neuroprotective effects were observed during the simultaneous administration of ruthenium and fucoidan.
Collapse
Affiliation(s)
- Setareh Javanmardi
- Department of Anatomical Sciences, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Farshad Moradpour
- Department of Physiology, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Mojgan Veisi
- Department of Physiology, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Neda Omidian
- Department of Physiology, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Rasoul Kavyannejad
- Department of Physiology, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| |
Collapse
|
3
|
Piccoli M, Barbato L, Maiorana NV, Mingione A, Raimondo F, Ghirimoldi M, Cirillo F, Schiepati M, Salerno D, Anastasia L, Albi E, Manfredi M, Bocci T, Priori A, Signorelli P. Direct Current Stimulation (DCS) Modulates Lipid Metabolism and Intercellular Vesicular Trafficking in SHSY-5Y Cell Line: Implications for Parkinson's Disease. J Neurochem 2025; 169:e70014. [PMID: 39930930 PMCID: PMC11811683 DOI: 10.1111/jnc.70014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 12/12/2024] [Accepted: 01/13/2025] [Indexed: 02/14/2025]
Abstract
The modulation of the brain's electrical activity for therapeutic purposes has recently gained attention, supported by the promising results obtained through the non-invasive application of transcranial direct current stimulation (tDCS) in the treatment of neurodegenerative and neurological diseases. To optimize therapeutic efficacy, it is crucial to investigate the cellular and molecular effects of tDCS. This will help to identify important biomarkers, predict patient's response and develop personalized treatments. In this study, we applied direct current stimulation (DCS) to a neural cell line, using mild currents over short periods of time (0.5 mA, 20 min), with 24-h intervals. We observed that DCS induced changes in the cellular lipidome, with transient effects observed after a single stimulation (lasting 24 h) and more significant, long-lasting effects (up to 72 h) after repeated stimulation cycles. In neural cells, multiple DCS treatment modulated structural membrane lipids (PE, PS, PI), downregulated glycerol lipids with ether-linked fatty acids and pro-inflammatory lipids (ceramides and lyso-glycerophospholipids) (p ≤ 0.005). Multiple DCS sessions altered transcriptional activity by decreasing the expression of inflammatory cytokines (TNF-α, p ≤ 0.05; IL-1β, p ≤ 0.01), while increasing the expression of neuroprotective factors such as heme oxygenase-1 (p ≤ 0.0001) and brain-derived neurotrophic factor (p ≤ 0.05), as well as proteins involved in vesicular transport (SNARE, sorting nexins and seipin and α-synuclein; p ≤ 0.05). In addition, DCS enhanced the release of extracellular vesicles, with repeated stimulations significantly increasing the release of exosomes threefold. In conclusion, while a single electrical stimulation induces transient metabolic changes with limited phenotypic effects, repeated applications induce a broader and deeper modulation of lipid species. This may lead to a neuroprotective and neuroplasticity-focussed transcriptional profile, potentially supporting the therapeutic effects of tDCS at the cellular and molecular level in patients..
Collapse
Affiliation(s)
- Marco Piccoli
- Institute for Molecular and Translational Cardiology (IMTC)IRCCS Policlinico San DonatoMilanItaly
- School of MedicineUniversity Vita‐Salute San RaffaeleMilanItaly
| | - Luisa Barbato
- Biochemistry LaboratoryIRCCS Policlinico San DonatoMilanItaly
| | | | - Alessandra Mingione
- “Aldo Ravelli” Research Centre, Department of Health SciencesUniversity of MilanMilanItaly
| | | | - Marco Ghirimoldi
- Biological Mass Spectrometry Lab, Department of Translational MedicineUniversity of Piemonte OrientaleNovaraItaly
| | - Federica Cirillo
- Institute for Molecular and Translational Cardiology (IMTC)IRCCS Policlinico San DonatoMilanItaly
| | - Mattia Schiepati
- “Aldo Ravelli” Research Centre, Department of Health SciencesUniversity of MilanMilanItaly
| | - Domenico Salerno
- School of Medicine and Surgery BioNanoMedicine Center NANOMIBUniversity of Milan‐BicoccaMonzaItaly
| | - Luigi Anastasia
- Institute for Molecular and Translational Cardiology (IMTC)IRCCS Policlinico San DonatoMilanItaly
- School of MedicineUniversity Vita‐Salute San RaffaeleMilanItaly
| | - Elisabetta Albi
- Department of Pharmaceutical Sciences, Interno Orto BotanicoUniversity of PerugiaPerugiaItaly
| | - Marcello Manfredi
- Institute for Molecular and Translational Cardiology (IMTC)IRCCS Policlinico San DonatoMilanItaly
- Biological Mass Spectrometry Lab, Department of Translational MedicineUniversity of Piemonte OrientaleNovaraItaly
- Center for Translational Research Autoimmune Diseases and Allergic DiseasesUniversity of Piemonte OrientaleNovaraItaly
| | - Tommaso Bocci
- “Aldo Ravelli” Research Centre, Department of Health SciencesUniversity of MilanMilanItaly
| | - Alberto Priori
- “Aldo Ravelli” Research Centre, Department of Health SciencesUniversity of MilanMilanItaly
| | - Paola Signorelli
- Biochemistry LaboratoryIRCCS Policlinico San DonatoMilanItaly
- “Aldo Ravelli” Research Centre, Department of Health SciencesUniversity of MilanMilanItaly
| |
Collapse
|
4
|
Jin WJ, Zhu XX, Luo KT, Wang S, Li JA, Qian LF, Xu GX. Enhancement of Cognitive Function in Rats with Vascular Dementia Through Modulation of the Nrf2/GPx4 Signaling Pathway by High-Frequency Repetitive Transcranial Magnetic Stimulation. Physiol Res 2024; 73:857-868. [PMID: 39560194 PMCID: PMC11629951 DOI: 10.33549/physiolres.935330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Accepted: 05/16/2024] [Indexed: 12/13/2024] Open
Abstract
Repetitive transcranial magnetic stimulation (rTMS) represents a non-invasive therapeutic modality acknowledged for augmenting neurological function recovery following stroke. Nonetheless, uncertainties remain regarding its efficacy in promoting cognitive function recovery in patients diagnosed with vascular dementia (VD). In this study, VD was experimentally induced in a rat model utilizing the bilateral common carotid artery occlusion method. Following a recuperation period of seven days, rats were subjected to high-frequency repetitive transcranial magnetic stimulation (HF-rTMS) at a frequency of 10 Hz. Cognitive function was assessed utilizing the Morris water maze test, and the levels of IL-6, TNF-alpha, SOD, GSH, MDA, and Fe2+ in cerebral tissue were quantitatively analyzed through enzyme-linked immunosorbent assay. Moreover, the gene and protein expressions of nuclear factor erythroid 2-related factor 2 (Nrf2) and glutathione peroxidase 4 (GPx4) were meticulously investigated via quantitative polymerase chain reaction (qPCR) and Western blotting techniques. The use of HF-rTMS notably augmented cognitive function in rats with VD, concomitantly reducing neuroinflammation, oxidative stress, and ferroptosis within the brain. The group subjected to HF-rTMS demonstrated an increase in the levels of both proteins and genes associated with Nrf2 and GPx4, in comparison to the VD group. These results highlight the potential of HF-rTMS treatment in enhancing cognitive function in rats diagnosed with VD through the modulation of the Nrf2/GPx4 signaling pathway. This modulation, in turn, mitigates processes linked with neuroinflammation, oxidative stress, and ferroptosis. Nevertheless, additional studies are essential to comprehensively elucidate the underlying mechanisms and clinical implications of HF-rTMS treatment in the treatment of VD.
Collapse
Affiliation(s)
- W-J Jin
- Department of Rehabilitation Medicine, Nanjing Medical University, Nanjing, China; Rehabilitation Medicine Center, Zhejiang Chinese Medical University Affiliated Jiaxing TCM Hospital, Jiaxing, China.
| | | | | | | | | | | | | |
Collapse
|
5
|
Khan WU, Salman M, Ali M, Majid H, Yar MS, Akhtar M, Parvez S, Najmi AK. Neuroprotective Effects of Sulforaphane in a rat model of Alzheimer's Disease induced by Aβ (1-42) peptides. Neurochem Int 2024; 179:105839. [PMID: 39173832 DOI: 10.1016/j.neuint.2024.105839] [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: 05/31/2024] [Revised: 08/13/2024] [Accepted: 08/19/2024] [Indexed: 08/24/2024]
Abstract
The intricate nature of Alzheimer's disease (AD) has presented significant hurdles in the development of effective interventions. Sulforaphane (SFN) is of interest due to its antioxidative, anti-inflammatory, and neuroprotective properties, which could address various aspects of AD pathology. This study explores the potential of SFN in a rat model of AD induced by Aβ (1-42) peptides. AD symptoms were triggered in rats by injecting Aβ (1-42) peptides directly into their cerebral ventricles. SFN (10 mg/kg and 20 mg/kg), Trigonelline (10 mg/kg), and Pioglitazone (10 mg/kg) were administered in Aβ (1-42) treated animals. Behavioral assessments were performed using the Novel Object Recognition tests. Various biochemical parameters, such as soluble Aβ (1-42), IRS-S312, GSK-3β, TNF-α, acetylcholinesterase, nitrite levels, lipid peroxidation, and reduced glutathione activity, were quantified using ELISA kits and spectrophotometric assays. Histopathological analyses included Hematoxylin and Eosin, Crystal Violet, Congo red, and IRS-1 Immunohistochemistry staining. Quantification was performed to assess neuronal loss and Aβ plaque burden. The novelty of this study lies in its comprehensive evaluation of SFN's impact on multiple AD-related pathways at dual doses. The Novel Object Recognition test revealed that SFN, especially at higher doses, improved memory deficits induced by Aβ (1-42). Biochemically, SFN reduced hippocampal Aβ levels, IRS-S312, GSK-3β, TNF-α, and acetylcholinesterase activity, while increasing glutathione levels, all in a dose-dependent manner. Histopathological analyses further confirmed SFN's protective role against Aβ-induced neuronal damage, amyloidosis, and changes in insulin signaling. These results highlight SFN's potential as a multifaceted therapeutic agent for AD, offering a promising avenue for treatment due to its antioxidative, anti-inflammatory, and neuroprotective properties. The inclusion of combination treatments with Trigonelline and Pioglitazone alongside SFN offers insights into potential synergistic effects, which could pave the way for developing combination therapies for AD.
Collapse
Affiliation(s)
- Wasi Uzzaman Khan
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India
| | - Mohd Salman
- Department of Medical Elementology and Toxicology, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi, 110062, India
| | - Mubashshir Ali
- Department of Medical Elementology and Toxicology, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi, 110062, India
| | - Haya Majid
- Department of Translational and Clinical Research, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi, 110062, India
| | - M Shahar Yar
- Department of Pharmaceutical Chemistry, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India
| | - Mohd Akhtar
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India
| | - Suhel Parvez
- Department of Medical Elementology and Toxicology, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi, 110062, India
| | - Abul Kalam Najmi
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India.
| |
Collapse
|
6
|
Wei H, Wang X, Zhong H, Kong X, Zhu J, Li B. Artesunate improves learning and memory impairment in rats with vascular cognitive impairment by down-regulating the level of autophagy in cerebral cortex neurons. Heliyon 2024; 10:e33068. [PMID: 38948049 PMCID: PMC11211894 DOI: 10.1016/j.heliyon.2024.e33068] [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: 02/23/2024] [Revised: 05/15/2024] [Accepted: 06/13/2024] [Indexed: 07/02/2024] Open
Abstract
Background Vascular cognitive impairment (VCI) is the second leading cause of dementia. Cognitive impairment is a common consequence of VCI. However, there is no effective treatment for VCI and the underlying mechanism of its pathogenesis remains unclear. This study to investigate whether artesunate (ART) can improve the learning and memory function in rats with VCI by down-regulating he level of autophagy in cerebral cortex neurons. Methods The models for VCI were the rat bilateral common carotid artery occlusion (BACCO), which were randomized into three groups including the sham operation group (Sham), model + vehicle group (Model) and model + ART group (ART). Then the animal behaviors were recorded, as well as staining the results of cortical neurons. Western blot was performed to determine the protein expressions of LC3BⅡ/Ⅰ, p-AMPK, p-mTOR, and Beclin-1. Results Behavioral outcomes and the protein expressions in Model group were supposedly affected by the induction of autophagy in cerebral cortex neurons. Compared to the Model group, ART improved memory impairment in VCI rats. And the expression of LC3BⅡ/Ⅰ, p-AMPK/AMPK, Beclin-1 is significant decreased in the ART group, while significant increases of p-mTOR/mTOR were showed. These results suggest that ART improved learning and memory impairment in VCI rats by down-regulating the level of autophagy in cerebral cortex neurons. Conclusion The results suggest that autophagy occurs in cerebral cortex neurons in rats with VCI. It is speculated that ART can improve learning and memory impairment in VCI rats by down-regulating the level of autophagy in cerebral cortex neurons.
Collapse
Affiliation(s)
- Honqiao Wei
- School of Basic Medical Sciences, Guangxi Medical University, Nanning, 530022, China
| | - Xiaokun Wang
- Research Center for Clinical Medicine, Jinshan Hospital Affiliated to Fudan University, Shanghai, 201508, China
| | - Hequan Zhong
- Research Center for Clinical Medicine, Jinshan Hospital Affiliated to Fudan University, Shanghai, 201508, China
| | - Xiangyu Kong
- Research Center for Clinical Medicine, Jinshan Hospital Affiliated to Fudan University, Shanghai, 201508, China
| | - Jie Zhu
- Department of Rehabilitation, Jinshan Hospital, Fudan University, Shanghai, 201508, China
| | - Bing Li
- Research Center for Clinical Medicine, Jinshan Hospital Affiliated to Fudan University, Shanghai, 201508, China
| |
Collapse
|
7
|
Zhang B, Deng L, Liu X, Hu Y, Wang W, Li M, Xu T, Pang L, Lv M. Transcranial direct current stimulation combined with swimming exercise improves the learning and memory abilities of vascular dementia rats by regulating microglia through miR-223-3p/PRMT8. Neurol Res 2024; 46:525-537. [PMID: 38563325 DOI: 10.1080/01616412.2024.2337517] [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: 09/23/2023] [Accepted: 03/26/2024] [Indexed: 04/04/2024]
Abstract
BACKGROUND Vascular dementia (VD) is the second most common type of dementia worldwide. Previous studies have proven that transcranial direct current stimulation (tDCS) has potential applications in relieving cognitive impairment in VD animal models. The purpose of this study was to probe the mechanism by which tDCS combined with swimming exercise improves the learning and memory abilities of VD model rats. METHOD The VD rat model was induced using the permanent bilateral common carotid artery occlusion (2-VO) method; tDCS was applied to the rats and then they took part in swimming exercises. Rat memory, platform crossing time, and platform crossing frequency were analyzed via a water maze experiment. Nerve damage in the cortex and hippocampal CA1 area of the rats was observed using Nissl staining. Western blotting, immunohistochemistry, immunofluorescence staining and reverse transcription quantitative polymerase chain reaction (RT - qPCR) were used to determine the expression of related proteins and genes. The levels of oxidative stress were detected by kits. RESULTS We demonstrated that VD model rats treated with tDCS combined with swimming exercise exhibited significant improvement in memory, and VD model rats exhibited significantly reduced neuronal loss in the hippocampus, and reduced microglial activation and M1 polarization. tDCS combined with swimming exercise protects VD model rats from oxidative stress through the miR-223-3p/protein arginine methyltransferase 8 (PRMT8) axis and inhibits the activation of the TLR4/NF-κB signaling pathway. CONCLUSION Our results suggest that tDCS combined with swimming exercise improved the learning and memory ability of VD model rats by regulating the expression of PRMT8 through miR-223-3p to affect microglial activation and M1 polarization.
Collapse
Affiliation(s)
- Bingxue Zhang
- Rehabilitation Medicine, Qujing No.1 Hospital, Qujing, Yunnan, China
| | - Li Deng
- Rehabilitation Medicine, Qujing No.1 Hospital, Qujing, Yunnan, China
| | - Xiaodan Liu
- Rehabilitation Medicine, Qujing No.1 Hospital, Qujing, Yunnan, China
| | - Yao Hu
- Rehabilitation Medicine, Qujing No.1 Hospital, Qujing, Yunnan, China
| | - Wenyi Wang
- Rehabilitation Medicine, Qujing No.1 Hospital, Qujing, Yunnan, China
| | - Minghua Li
- Department of Neurology, Luoping County People's Hospital, Luoping, Yunnan, China
| | - Ting Xu
- Rehabilitation Medicine, Qujing No.1 Hospital, Qujing, Yunnan, China
| | - Li Pang
- Rehabilitation Medicine, Qujing No.1 Hospital, Qujing, Yunnan, China
| | - Meifen Lv
- Rehabilitation Medicine, Qujing No.1 Hospital, Qujing, Yunnan, China
| |
Collapse
|
8
|
Pei J, Zhang C, Zhang X, Zhao Z, Zhang X, Yuan Y. Low-intensity transcranial ultrasound stimulation improves memory in vascular dementia by enhancing neuronal activity and promoting spine formation. Neuroimage 2024; 291:120584. [PMID: 38522806 DOI: 10.1016/j.neuroimage.2024.120584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 03/01/2024] [Accepted: 03/21/2024] [Indexed: 03/26/2024] Open
Abstract
Memory is closely associated with neuronal activity and dendritic spine formation. Low-intensity transcranial ultrasound stimulation (TUS) improves the memory of individuals with vascular dementia (VD). However, it is unclear whether neuronal activity and dendritic spine formation under ultrasound stimulation are involved in memory improvement in VD. In this study, we found that seven days of TUS improved memory in VD model while simultaneously increasing pyramidal neuron activity, promoting dendritic spine formation, and reducing dendritic spine elimination. These effects lasted for 7 days but disappeared on 14 d after TUS. Neuronal activity and dendritic spine formation strongly corresponded to improvements in memory behavior over time. In addition, we also found that the memory, neuronal activity and dendritic spine of VD mice cannot be restored again by TUS of 7 days after 28 d. Collectively, these findings suggest that TUS increases neuronal activity and promotes dendritic spine formation and is thus important for improving memory in patients with VD.
Collapse
Affiliation(s)
- Jiamin Pei
- School of Electrical Engineering, Yanshan University, No.438 Hebei Street, Qinhuangdao 066004, China; Key Laboratory of Intelligent Rehabilitation and Neuromodulation of Hebei Province, Yanshan University, No.438 Hebei Street, Qinhuangdao 066004, China
| | - Cong Zhang
- Department of Neurology, Hebei Key Laboratory of Vascular Homeostasis and Hebei Collaborative Innovation Center for Cardio-cerebrovascular Disease, The Second Hospital of Hebei Medical University, No.215 Heping Road, Shijiazhuang 050000, China
| | - Xiao Zhang
- Department of Neurology, Hebei Key Laboratory of Vascular Homeostasis and Hebei Collaborative Innovation Center for Cardio-cerebrovascular Disease, The Second Hospital of Hebei Medical University, No.215 Heping Road, Shijiazhuang 050000, China
| | - Zhe Zhao
- School of Electrical Engineering, Yanshan University, No.438 Hebei Street, Qinhuangdao 066004, China; Key Laboratory of Intelligent Rehabilitation and Neuromodulation of Hebei Province, Yanshan University, No.438 Hebei Street, Qinhuangdao 066004, China
| | - Xiangjian Zhang
- Department of Neurology, Hebei Key Laboratory of Vascular Homeostasis and Hebei Collaborative Innovation Center for Cardio-cerebrovascular Disease, The Second Hospital of Hebei Medical University, No.215 Heping Road, Shijiazhuang 050000, China.
| | - Yi Yuan
- School of Electrical Engineering, Yanshan University, No.438 Hebei Street, Qinhuangdao 066004, China; Key Laboratory of Intelligent Rehabilitation and Neuromodulation of Hebei Province, Yanshan University, No.438 Hebei Street, Qinhuangdao 066004, China.
| |
Collapse
|
9
|
Seok JW, Kim G, Kim JU. Comparative efficacy of seven nonpharmacological interventions on global cognition in older adults with and without mild cognitive impairment: a network meta-analysis of randomized controlled trials. Sci Rep 2024; 14:8402. [PMID: 38600212 PMCID: PMC11006946 DOI: 10.1038/s41598-024-58232-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Accepted: 03/26/2024] [Indexed: 04/12/2024] Open
Abstract
To maintain current cognitive function and access greater cognitive reserves, nonpharmacological interventions may be a viable alternative for older adults with or without cognitive impairment. This study aimed to compare different nonpharmacological interventions for enhancing global cognition, including mind-body exercise, physical exercise, non-invasive brain stimulation, cognitive training intervention (CTI), acutherapy (ACU), meditation, and music therapy, by applying a network meta-analysis (NMA). Sixty-one randomized controlled trials evaluating the efficacy of interventions on global cognition in older adults with or without mild cognitive decline were selected. An NMA was conducted to compare the efficacy of different nonpharmacological interventions. The NMA revealed that mind-body exercise (standardized mean difference, 1.384; 95% confidence interval, 0.777-1.992); ACU (1.283; 0.478-2.088); meditation (0.910; 0.097-1.724); non-invasive brain stimulation (1.242; 0.254-2.230); CTI (1.269; 0.736-1.802); and physical exercise (0.977; 0.212-1.742), showed positive effects compared to passive controls. There were no significant differences between the efficacies of other interventions. Nonpharmacological interventions may potentially enhance and maintain global cognition through various pathways, such as memorizing movements and enhancing brain plasticity by reducing stress in the older adult population. Additional studies are needed to clarify the impact of other variables, including intervention methods or psychological variables.
Collapse
Affiliation(s)
- Ji-Woo Seok
- Digital Health Research Division, Korea Institute of Oriental Medicine, 1672, Yuseong-daero, Yuseong-gu, Daejeon, 34054, Republic of Korea
| | - Gahye Kim
- Digital Health Research Division, Korea Institute of Oriental Medicine, 1672, Yuseong-daero, Yuseong-gu, Daejeon, 34054, Republic of Korea
| | - Jaeuk U Kim
- Digital Health Research Division, Korea Institute of Oriental Medicine, 1672, Yuseong-daero, Yuseong-gu, Daejeon, 34054, Republic of Korea.
- KM Convergence Science, University of Science and Technology, Daejeon, South Korea.
| |
Collapse
|
10
|
Yang J, Qi Y, Zhu B, Lin S. A Novel Tetrapeptide Ala-Phe-Phe-Pro (AFFP) Derived from Antarctic Krill Prevents Scopolamine-Induced Memory Disorder by Balancing Lipid Metabolism of Mice Hippocampus. Nutrients 2024; 16:1019. [PMID: 38613052 PMCID: PMC11013912 DOI: 10.3390/nu16071019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 03/20/2024] [Accepted: 03/27/2024] [Indexed: 04/14/2024] Open
Abstract
Memory impairment is a serious problem with organismal aging and increased social pressure. The tetrapeptide Ala-Phe-Phe-Pro (AFFP) is a synthetic analogue of Antarctic krill derived from the memory-improving Antarctic krill peptide Ser-Ser-Asp-Ala-Phe-Phe-Pro-Phe-Arg (SSDAFFPFR) after digestion and absorption. The objective of this research was to assess the neuroprotective effects of AFFP by reducing oxidative stress and controlling lipid metabolism in the brains of mice with memory impairment caused by scopolamine. The 1H Nuclear magnetic resonance spectroscopy results showed that AFFP had three active hydrogen sites that could contribute to its antioxidant properties. The findings from in vivo tests demonstrated that AFFP greatly enhanced the mice's behavioral performance in the passive avoidance, novel object recognition, and eight-arm maze experiments. AFFP reduced oxidative stress by enhancing superoxide dismutase activity and malondialdehyde levels in mice serum, thereby decreasing reactive oxygen species level in the mice hippocampus. In addition, AFFP increased the unsaturated lipid content to balance the unsaturated lipid level against the neurotoxicity of the mice hippocampus. Our findings suggest that AFFP emerges as a potential dietary intervention for the prevention of memory impairment disorders.
Collapse
Affiliation(s)
- Jingqi Yang
- SKL of Marine Food Processing & Safety Control, School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, China; (J.Y.); (Y.Q.)
- Engineering Research Center of Food, Dalian Polytechnic University, Dalian 116034, China
| | - Yan Qi
- SKL of Marine Food Processing & Safety Control, School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, China; (J.Y.); (Y.Q.)
- Engineering Research Center of Food, Dalian Polytechnic University, Dalian 116034, China
| | - Beiwei Zhu
- National Engineering Research Center of Seafood, Dalian 116034, China
| | - Songyi Lin
- SKL of Marine Food Processing & Safety Control, School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, China; (J.Y.); (Y.Q.)
- Engineering Research Center of Food, Dalian Polytechnic University, Dalian 116034, China
- Engineering Research Center of Special Dietary Food, The Education Department of Liaoning Province, Dalian 116034, China
| |
Collapse
|
11
|
Marino N, Bedeschi M, Vaccari ME, Cambiaghi M, Tesei A. Glitches in the brain: the dangerous relationship between radiotherapy and brain fog. Front Cell Neurosci 2024; 18:1328361. [PMID: 38515789 PMCID: PMC10956129 DOI: 10.3389/fncel.2024.1328361] [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: 02/22/2024] [Indexed: 03/23/2024] Open
Abstract
Up to approximately 70% of cancer survivors report persistent deficits in memory, attention, speed of information processing, multi-tasking, and mental health functioning, a series of symptoms known as "brain fog." The severity and duration of such effects can vary depending on age, cancer type, and treatment regimens. In particular, every year, hundreds of thousands of patients worldwide undergo radiotherapy (RT) for primary brain tumors and brain metastases originating from extracranial tumors. Besides its potential benefits in the control of tumor progression, recent studies indicate that RT reprograms the brain tumor microenvironment inducing increased activation of microglia and astrocytes and a consequent general condition of neuroinflammation that in case it becomes chronic could lead to a cognitive decline. Furthermore, radiation can induce endothelium reticulum (ER) stress directly or indirectly by generating reactive oxygen species (ROS) activating compensatory survival signaling pathways in the RT-surviving fraction of healthy neuronal and glial cells. In particular, the anomalous accumulation of misfolding proteins in neuronal cells exposed to radiation as a consequence of excessive activation of unfolded protein response (UPR) could pave the way to neurodegenerative disorders. Moreover, exposure of cells to ionizing radiation was also shown to affect the normal proteasome activity, slowing the degradation rate of misfolded proteins, and further exacerbating ER-stress conditions. This compromises several neuronal functions, with neuronal accumulation of ubiquitinated proteins with a consequent switch from proteasome to immunoproteasome that increases neuroinflammation, a crucial risk factor for neurodegeneration. The etiology of brain fog remains elusive and can arise not only during treatment but can also persist for an extended period after the end of RT. In this review, we will focus on the molecular pathways triggered by radiation therapy affecting cognitive functions and potentially at the origin of so-called "brain fog" symptomatology, with the aim to define novel therapeutic strategies to preserve healthy brain tissue from cognitive decline.
Collapse
Affiliation(s)
- Noemi Marino
- Bioscience Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Martina Bedeschi
- Bioscience Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Melania Elettra Vaccari
- Bioscience Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Marco Cambiaghi
- Bioscience Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Anna Tesei
- Bioscience Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| |
Collapse
|
12
|
Wu F, Zhang J, Wang Q, Liu W, Zhang X, Ning F, Cui M, Qin L, Zhao G, Liu D, Lv S, Xu Y. Identification of immune-associated genes in vascular dementia by integrated bioinformatics and inflammatory infiltrates. Heliyon 2024; 10:e26304. [PMID: 38384571 PMCID: PMC10879030 DOI: 10.1016/j.heliyon.2024.e26304] [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: 12/11/2023] [Revised: 02/04/2024] [Accepted: 02/09/2024] [Indexed: 02/23/2024] Open
Abstract
OBJECTIVE Dysregulation of the immune system plays a vital role in the pathological process of vascular dementia, and this study aims to spot critical biomarkers and immune infiltrations in vascular dementia employing a bioinformatics approach. METHODS We acquired gene expression profiles from the Gene Expression Database. The gene expression data were analyzed using the bioinformatics method to identify candidate immune-related central genes for the diagnosis of vascular dementia. and the diagnostic value of nomograms and Receiver Operating Characteristic (ROC) curves were evaluated. We also examined the role of the VaD hub genes. Using the database and potential therapeutic drugs, we predicted the miRNA and lncRNA controlling the Hub genes. Immune cell infiltration was initiated to examine immune cell dysregulation in vascular dementia. RESULTS 1321 immune genes were included in the combined immune dataset, and 2816 DEGs were examined in GSE122063. Twenty potential genes were found using differential gene analysis and co-expression network analysis. PPI network design and functional enrichment analysis were also done using the immune system as the main subject. To create the nomogram for evaluating the diagnostic value, four potential core genes were chosen by machine learning. All four putative center genes and nomograms have a solid diagnostic value (AUC ranged from 0.81 to 0.92). Their high confidence level became unquestionable by validating each of the four biomarkers using a different dataset. According to GeneMANIA and GSEA enrichment investigations, the pathophysiology of VaD is strongly related to inflammatory responses, drug reactions, and central nervous system degeneration. The data and Hub genes were used to construct a ceRNA network that includes three miRNAs, 90 lncRNA, and potential VaD therapeutics. Immune cells with varying dysregulation were also found. CONCLUSION Using bioinformatic techniques, our research identified four immune-related candidate core genes (HMOX1, EBI3, CYBB, and CCR5). Our study confirms the role of these Hub genes in the onset and progression of VaD at the level of immune infiltration. It predicts potential RNA regulatory pathways control VaD progression, which may provide ideas for treating clinical disease.
Collapse
Affiliation(s)
- Fangchao Wu
- Department of Rehabilitation Medicine, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China
| | - Junling Zhang
- Shandong Medicine Technician College, Taian 271000, China
| | - Qian Wang
- Department of Central Laboratory, The Affiliated Taian City Central Hospital of Qingdao University, Taian, 271000, China
| | - Wenxin Liu
- Department of Rehabilitation, The Second Affiliated Hospital of Shandong First Medical University, Taian, 271000, China
| | - Xinlei Zhang
- Department of Rehabilitation, The Second Affiliated Hospital of Shandong First Medical University, Taian, 271000, China
| | - Fangli Ning
- Department of Rehabilitation, The Second Affiliated Hospital of Shandong First Medical University, Taian, 271000, China
| | - Mengmeng Cui
- Department of Rehabilitation, The Second Affiliated Hospital of Shandong First Medical University, Taian, 271000, China
| | - Lei Qin
- Department of Rehabilitation, The Second Affiliated Hospital of Shandong First Medical University, Taian, 271000, China
| | - Guohua Zhao
- Department of Rehabilitation, The Second Affiliated Hospital of Shandong First Medical University, Taian, 271000, China
| | - Di Liu
- Department of Neurology, Dongping County People's Hospital, Taian, 271000, China
| | - Shi Lv
- Department of Rehabilitation, The Second Affiliated Hospital of Shandong First Medical University, Taian, 271000, China
| | - Yuzhen Xu
- Department of Rehabilitation, The Second Affiliated Hospital of Shandong First Medical University, Taian, 271000, China
| |
Collapse
|
13
|
Zhu J, Du J, Kou W, Liu C, Fan J, Zhu Z, Deng L, Guan L, Wang Y, Yu A. Probucol protects against brain damage caused by intra-neural pyroptosis in rats with vascular dementia through inhibition of the Syk/Ros pathway. Aging (Albany NY) 2024; 16:4363-4377. [PMID: 38441564 PMCID: PMC10968675 DOI: 10.18632/aging.205593] [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: 05/31/2023] [Accepted: 01/19/2024] [Indexed: 03/22/2024]
Abstract
BACKGROUND Neuronal injury in chronic cerebral hypoperfusion (CCH) is the main pathogenic factor of vascular dementia (VD). Clinically, there isn't a drug specifically for VD; instead, the majority of medications used to treat Alzheimer's disease (AD) are also used to treat VD. Based on the proven anti-inflammatory and antioxidant effects of Probucol, we hypothesized that it may have therapeutic effects on VD, but more research is required to determine its exact mechanism of action. METHODS In vivo experiment: We used SD rats and most commonly used bilateral carotid artery occlusion (2-VO) in VD for modeling. After successful modeling, SD rats were given Probucol 3.5 mg/kg/day for 8 weeks to evaluate the therapeutic effect. In vitro experiment: BV-2 microglia of rats were cultured and divided into Control group and Probucol group. Each group was treated with hypoxia-hypoglycemia, hypoxia-hypoglycemia hydrogen peroxide and hypoxia-hypoglycemia hydrogen peroxide Syk inhibitor respectively. RESULTS The results of immunofluorescence and Western blot showed that Probucol could significantly improve the cognitive impairment induced by CCH, and the neuronal damage was also attenuated. On the one hand, the underlying mechanism of Probucol was to reduce oxidative stress and cell apoptosis of hippocampal neurons by inhibiting the expression of phosphorylated spleen tyrosine kinase (P-Syk); On the other hand, it exerted a protective effect by reducing NLRP3-dependent cell pyroptosis and inhibiting neuroinflammation induced by microglia activation. CONCLUSION Probucol could reduce oxidative stress and cell apoptosis by inhibiting the Syk/ROS signaling pathway, thereby improving CCH-induced cognitive impairment in vitro and in vivo.
Collapse
Affiliation(s)
- Jingyi Zhu
- Graduate School, Hebei North University, Zhangjiakou, Hebei, China
| | - Jing Du
- Department of Neurology, The First Affiliated Hospital of Hebei North University, Zhangjiakou, Hebei, China
| | - Wenhui Kou
- Department of Neurology, The First Affiliated Hospital of Hebei North University, Zhangjiakou, Hebei, China
| | - Chenling Liu
- Graduate School, Hebei North University, Zhangjiakou, Hebei, China
| | - Jianchun Fan
- Graduate School, Hebei North University, Zhangjiakou, Hebei, China
| | - Ziyan Zhu
- Graduate School, Hebei North University, Zhangjiakou, Hebei, China
| | - Lexiu Deng
- Graduate School, Hebei North University, Zhangjiakou, Hebei, China
| | - Lingling Guan
- Graduate School, Hebei North University, Zhangjiakou, Hebei, China
| | - Yuandi Wang
- Graduate School, Hebei North University, Zhangjiakou, Hebei, China
| | - Aimei Yu
- Department of Neurology, The First Affiliated Hospital of Hebei North University, Zhangjiakou, Hebei, China
| |
Collapse
|
14
|
Wang S, Du SH, Wang XQ, Lu JY. Mechanisms of transcranial direct current stimulation (tDCS) for pain in patients with fibromyalgia syndrome. Front Mol Neurosci 2024; 17:1269636. [PMID: 38356687 PMCID: PMC10865494 DOI: 10.3389/fnmol.2024.1269636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Accepted: 01/10/2024] [Indexed: 02/16/2024] Open
Abstract
Fibromyalgia syndrome (FMS) is a recurrent pain condition that can be challenging to treat. Transcranial direct current stimulation (tDCS) has become a promising non-invasive therapeutic option in alleviating FMS pain, but the mechanisms underlying its effectiveness are not yet fully understood. In this article, we discuss the most current research investigating the analgesic effects of tDCS on FMS and discuss the potential mechanisms. TDCS may exert its analgesic effects by influencing neuronal activity in the brain, altering cortical excitability, changing regional cerebral blood flow, modulating neurotransmission and neuroinflammation, and inducing neuroplasticity. Overall, evidence points to tDCS as a potentially safe and efficient pain relief choice for FMS by multiple underlying mechanisms. This article provides a thorough overview of our ongoing knowledge regarding the mechanisms underlying tDCS and emphasizes the possibility of further studies to improve the clinical utility of tDCS as a pain management tool.
Collapse
Affiliation(s)
- Shan Wang
- Department of Health School, Shanghai Normal University Tianhua College, Shanghai, China
| | - Shu-Hao Du
- Department of Sport Rehabilitation, Shanghai University of Sport, Shanghai, China
| | - Xue-Qiang Wang
- Rehabilitation Medicine Center, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- School of Rehabilitation Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, China
- Department of Rehabilitation Medicine, Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
| | - Jun-Yan Lu
- Rehabilitation Medicine Center, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- School of Rehabilitation Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, China
- Department of Rehabilitation Medicine, Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
| |
Collapse
|
15
|
Chmiel J, Rybakowski F, Leszek J. Effect of Transcranial Direct Current Stimulation (tDCS) on Depression in Parkinson's Disease-A Narrative Review. J Clin Med 2024; 13:699. [PMID: 38337395 PMCID: PMC10856764 DOI: 10.3390/jcm13030699] [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: 01/06/2024] [Revised: 01/22/2024] [Accepted: 01/24/2024] [Indexed: 02/12/2024] Open
Abstract
INTRODUCTION Depression is the most prevalent comorbid neuropsychiatric condition in individuals with Parkinson's disease (PD), and its underlying mechanisms are not yet fully understood. Current treatment methods are characterised by moderate effectiveness and possible side effects, prompting the search for new non-invasive and safe treatment methods. METHODS This narrative review explores the use of transcranial direct current stimulation (tDCS) in the treatment of depression in PD, based on neuropsychological measures. Searches were conducted in the PubMed/Medline, Research Gate, and Cochrane databases. RESULTS Nine relevant studies were identified, where depression scores served as either primary or secondary outcomes. Stimulation protocols displayed heterogeneity, especially concerning choice of stimulation site. Patient samples were also heterogeneous. The majority of the studies incorporated anodal stimulation targeting the left dorsolateral prefrontal cortex (DLPFC). The results revealed a reduction in depression scores among PD patients following tDCS. Potential mechanisms through which tDCS may alleviate depression in PD were discussed and recommendations for future research were made. CONCLUSIONS Preliminary evidence suggests that tDCS applied anodally to the left DLPFC reduces depression scores in people with PD; however, due to the heterogeneity of the studies analysed, the use of tDCS in this field should be approached with caution and warrants further validation and confirmation.
Collapse
Affiliation(s)
- James Chmiel
- Institute of Neurofeedback and tDCS Poland, 70-393 Szczecin, Poland
| | - Filip Rybakowski
- Department and Clinic of Psychiatry, Poznan University of Medical Sciences, 61-701 Poznań, Poland
| | - Jerzy Leszek
- Department and Clinic of Psychiatry, Wrocław Medical University, 54-235 Wrocław, Poland
| |
Collapse
|
16
|
Zarifkar AH, Zarifkar A, Safaei S. Different paradigms of transcranial electrical stimulation induce structural changes in the CA1 region of the hippocampus in a rat model of Alzheimer's disease. Neurosci Lett 2024; 818:137570. [PMID: 38000774 DOI: 10.1016/j.neulet.2023.137570] [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: 10/07/2023] [Revised: 11/15/2023] [Accepted: 11/21/2023] [Indexed: 11/26/2023]
Abstract
One of the prominent sign of Alzheimer's disease (AD) is structural changes in the hippocampus. Recently, the new methods used to treat this disease is transcranial electrical stimulation (tES). This study evaluated the effect of four primary standards of tES, including tDCS, tACS, tRNS, and tPCS on beta-amyloid 25-35 (Aβ25-35)-induced structural changes in the CA1 region of hippocampus in male rats. For this purpose, rats weighing 250-275 g were selected, the cannula was embedded reciprocally into the hippocampi. Aβ25-35 (5 μg/ 2.5 ml/ day) was infused reciprocally for four continuous days.Then, animals were then given tES for 6 days.Subsequently, structural changes in the hippocampal CA1 were evaluated using the stereological method. Aβ25-35 resulted in loss of neurons (P < 0.01) and decreased hippocampal volume (P < 0.05). However, the administration of tES paradigms prevented these changes. The results proposed that through the improvement of hippocampal cell number and volume, tES paradigms can retain efficiency in remediating structural impairments in AD. From this, it can be concluded that other tES paradigms besides tDCS can also be considered for the treatment of AD.
Collapse
Affiliation(s)
- Amir Hossein Zarifkar
- Department of Physiology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran; Cellular and Molecular Biology Research Center, Larestan University of Medical Sciences, Larestan, Iran.
| | - Asadollah Zarifkar
- Department of Physiology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Sepideh Safaei
- Gerash Amir-al-Momenin Medical and Educational Center, Gerash University of Medical Sciences, Gerash, Iran
| |
Collapse
|
17
|
Chen Y, Cheng W, Tang J, Xiao B, Kuang M, Xu L, Liu H. Mitigating cognitive impairment in aging mice: Exploring the therapeutic potential of ischelium. Biochem Biophys Res Commun 2023; 687:149173. [PMID: 37944469 DOI: 10.1016/j.bbrc.2023.149173] [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: 10/18/2023] [Accepted: 10/26/2023] [Indexed: 11/12/2023]
Abstract
To deeply explore the intervention effects of ischelium on the cognitive memory decline in naturally aging mice and its potential mechanisms, we randomly divided mice into four groups: young control group (C), elderly group (M), ischelium low-dose group (L), and ischelium high-dose group (H). The experiment lasted for 12 weeks. We employed the Y-maze test, open field test, and conditioned fear test to evaluate the memory functions of each group. Through HE staining and electron microscopy, we observed morphological changes in the mouse hippocampus. RT-PCR was used to detect changes in the expression of factors related to cognitive function in the hippocampus of elderly mice. We analyzed the changes in the Nrf2/HO-1 pathway and the inflammatory factors IL-1β and TNF-α using elisa. Additionally, we examined the enzymatic activities of SOD, CAT, GSH-Px, and MDA in the hippocampus and analyzed the compositional changes of gut microbiota in mice using 16S technology. Our results indicate that ischelium effectively ameliorates cognitive impairments in elderly mice.
Collapse
Affiliation(s)
- Yanhong Chen
- Laboratory Animal Center of Zhejiang University, Zhejiang, China
| | - Wei Cheng
- Hangzhou Kang Ming Information Technology Co., Ltd, Zhejiang, China
| | - Jia Tang
- Hangzhou Kang Ming Information Technology Co., Ltd, Zhejiang, China
| | - Boneng Xiao
- Laboratory Animal Center of Zhejiang University, Zhejiang, China
| | - Ming Kuang
- Hangzhou Kang Ming Information Technology Co., Ltd, Zhejiang, China
| | - Liaoyi Xu
- Department of Integrative Biology, The University of Texas at Austin, Texas, USA
| | - Hongying Liu
- Hangzhou Kang Ming Information Technology Co., Ltd, Zhejiang, China.
| |
Collapse
|
18
|
Zin LEF, Vizuete AFK, Callai EMM, Catarina LS, Fróes F, Moreira AP, de Oliveira Marques C, Leal MB, Ponzoni D, Puricelli E, da Silva Torres IL, Gonçalves CA, Quevedo AS. Astroglial Alterations in the Hippocampus of Rats Submitted to a Single Trans-Cranial Direct Current Stimulation Trial. Neurochem Res 2023; 48:3447-3456. [PMID: 37464227 DOI: 10.1007/s11064-023-03990-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 06/14/2023] [Accepted: 07/07/2023] [Indexed: 07/20/2023]
Abstract
Evidence indicates that transcranial direct current stimulation (tDCS) provides therapeutic benefits in different situations, such as epilepsy, depression, inflammatory and neuropathic pain. Despite the increasing use of tDCS, its cellular and molecular basis remains unknown. Astrocytes display a close functional and structural relationship with neurons and have been identified as mediators of neuroprotection in tDCS. Considering the importance of hippocampal glutamatergic neurotransmission in nociceptive pathways, we decided to investigate short-term changes in the hippocampal astrocytes of rats subjected to tDCS, evaluating specific cellular markers (GFAP and S100B), as well as markers of astroglial activity; glutamate uptake, glutamine synthesis by glutamine synthetase (GS) and glutathione content. Data clearly show that a single session of tDCS increases the pain threshold elicited by mechanical and thermal stimuli, as evaluated by von Frey and hot plate tests, respectively. These changes involve inflammatory and astroglial neurochemical changes in the hippocampus, based on specific changes in cell markers, such as S100B and GS. Alterations in S100B were also observed in the cerebrospinal fluid of tDCS animals and, most importantly, specific functional changes (increased glutamate uptake and increased GS activity) were detected in hippocampal astrocytes. These findings contribute to a better understanding of tDCS as a therapeutic strategy for nervous disorders and reinforce the importance of astrocytes as therapeutic targets.
Collapse
Affiliation(s)
- Lisandra Eda Fusinato Zin
- Biochemistry Graduate Program, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, 2600, 90035-003, Brazil
- Atitus Educação, Campus Santa Teresinha, Passo Fundo, Brazil
| | - Adriana Fernanda K Vizuete
- Biochemistry Graduate Program, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, 2600, 90035-003, Brazil
| | | | | | - Fernanda Fróes
- Biochemistry Graduate Program, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, 2600, 90035-003, Brazil
| | - Ana Paula Moreira
- Biochemistry Graduate Program, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, 2600, 90035-003, Brazil
| | | | - Miriara B Leal
- Biochemistry Graduate Program, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, 2600, 90035-003, Brazil
| | - Deise Ponzoni
- Dentistry Graduate Program, UFRGS, Porto Alegre, Brazil
| | | | | | - Carlos-Alberto Gonçalves
- Biochemistry Graduate Program, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, 2600, 90035-003, Brazil.
- Neuroscience Graduate Program, UFRGS, Porto Alegre, Brazil.
| | - Alexandre Silva Quevedo
- Biochemistry Graduate Program, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, 2600, 90035-003, Brazil
- Dentistry Graduate Program, UFRGS, Porto Alegre, Brazil
- Neuroscience Graduate Program, UFRGS, Porto Alegre, Brazil
| |
Collapse
|
19
|
Yamada Y, Sumiyoshi T. Preclinical Evidence for the Mechanisms of Transcranial Direct Current Stimulation in the Treatment of Psychiatric Disorders; A Systematic Review. Clin EEG Neurosci 2023; 54:601-610. [PMID: 34898301 PMCID: PMC10625720 DOI: 10.1177/15500594211066151] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 11/15/2021] [Accepted: 11/21/2021] [Indexed: 11/15/2022]
Abstract
Backgrounds. Transcranial direct current stimulation (tDCS) is a non-invasive brain stimulation technique for the treatment of several psychiatric disorders, eg, mood disorders and schizophrenia. Although tDCS provides a promising approach, its neurobiological mechanisms remain to be explored. Objectives. To provide a systematic review of animal studies, and consider how tDCS ameliorates psychiatric conditions. Methods. A literature search was conducted on English articles identified by PubMed. We defined the inclusion criteria as follows: (1) articles published from the original data; (2) experimental studies in animals; (3) studies delivering direct current transcranially, ie, positioning electrodes onto the skull. Results. 138 papers met the inclusion criteria. 62 papers deal with model animals without any dysfunctions, followed by 52 papers for neurological disorder models, and 12 for psychiatric disorder models. The most studied category of functional areas is neurocognition, followed by motor functions and pain. These studies overall suggest the role for the late long-term potentiation (LTP) via anodal stimulation in the therapeutic effects of tDCS. Conclusions. tDCS Anodal stimulation may provide a novel therapeutic strategy to particularly enhance neurocognition in psychiatric disorders. Its mechanisms are likely to involve facilitation of the late LTP.
Collapse
Affiliation(s)
- Yuji Yamada
- Department of Psychiatry, National Center Hospital, National Center Hospital, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Tomiki Sumiyoshi
- Department of Preventive Intervention for Psychiatric Disorders, National Institute of Mental Health, National Center of Neurology and Psychiatry, Tokyo, Japan
| |
Collapse
|
20
|
Ji Y, Ni X, Zheng K, Jiang Y, Ren C, Zhu H, Xiao M, Wang T. Synergistic effects of aerobic exercise and transcranial direct current stimulation on executive function and biomarkers in healthy young adults. Brain Res Bull 2023; 202:110747. [PMID: 37611879 DOI: 10.1016/j.brainresbull.2023.110747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2023] [Revised: 08/09/2023] [Accepted: 08/19/2023] [Indexed: 08/25/2023]
Abstract
OBJECTIVE This research explored the combined effects of transcranial direct current stimulation (tDCS) and aerobic exercise (AE) on executive function and specific serum biomarkers in healthy adults. METHODS Sixty healthy young adults were randomly assigned into tDCS+AE, tDCS only, or AE only groups. Interventions were carried out for 20 days. Executive functions were evaluated using tasks such as the 2,3-back task, the spatial working memory task, the Stroop test, T test, and hexagonal obstacle jump task. Serum biomarkers, including brain-derived neurotrophic factor (BDNF), malondialdehyde (MDA), superoxide dismutase (SOD), glutamate, glutathione peroxidase 4 (GPX4) and iron ion, were analyzed pre- and post-intervention. RESULTS The tDCS+AE group showed superior enhancements in executive function, evidenced by improved accuracy rates in 2,3-back tasks, better performance in the staircase task, and reduced reaction times in the incongruent reaction time of the Stroop task compared to other groups. Importantly, we found substantial changes in serum biomarkers: increased levels of BDNF and SOD, and decreased levels of MDA and glutamate in the tDCS+AE group. These changes were significantly different when compared with the tDCS and AE only groups. Notably, these alterations in serum biomarkers were correlated with improvements in executive function tasks, thus offering a potential physiological basis for the cognitive improvements witnessed. CONCLUSION The combined tDCS and AE intervention effectively improved executive function in healthy young adults, with the improvements linked to changes in key serum biomarkers. The results emphasize the potential of combined tDCS and AE interventions in engaging multiple physiological pathways to enhance executive function.
Collapse
Affiliation(s)
- Yingying Ji
- Wuxi Mental Health Center, Nanjing Medical University, Wuxi, Jiangsu, China
| | - Xuemei Ni
- Wuxi Mental Health Center, Nanjing Medical University, Wuxi, Jiangsu, China
| | - Kai Zheng
- Wuxi Mental Health Center, Nanjing Medical University, Wuxi, Jiangsu, China
| | - Ying Jiang
- Wuxi Mental Health Center, Nanjing Medical University, Wuxi, Jiangsu, China
| | - Caili Ren
- Wuxi Mental Health Center, Nanjing Medical University, Wuxi, Jiangsu, China
| | - Haohao Zhu
- Wuxi Mental Health Center, Nanjing Medical University, Wuxi, Jiangsu, China.
| | - Ming Xiao
- Jiangsu Province Key Laboratory of Neurodegeneration, Center for Global Health, Nanjing Medical University, Nanjing, Jiangsu, China.
| | - Tong Wang
- Rehabilitation Medicine Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China.
| |
Collapse
|
21
|
Centeno Crespo P, Anderson Meira Martins L, Camacho Dos Reis C, Fernandes Medeiros L, Leal Scarabelot V, Duzzo Gamaro G, Sandrielly Pereira Soares M, Maria Spanevello R, Moro Stefanello F, Cristina Custódio De Souza I. Transcranial direct current stimulation effects in the pain threshold and in oxidative stress parameters of neuropathic pain rats. Neurosci Lett 2023; 803:137179. [PMID: 36914044 DOI: 10.1016/j.neulet.2023.137179] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Accepted: 03/08/2023] [Indexed: 03/13/2023]
Abstract
Transcranial direct current stimulation (tDCS) can modulate cortical excitability and relieve neuropathic pain (NP), but the role of several biomarkers in this process is not well understood. This study aimed to analyze the effects of tDCS on biochemical parameters in rats with neuropathic pain (NP) induced by chronic constriction injury (CCI) of the right sciatic nerve. Eighty-eight male 60-day-old Wistar rats were divided into nine groups: control (C), control-electrode off (CEoff), control-tDCS (C-tDCS), sham-lesion (SL), sham-lesion electrode off (SLEoff), sham-lesion (SL-tDCS), lesion (L), lesion electrode off (LEoff), and lesion-tDCS (L-tDCS). After NP establishment, 20-minute bimodal tDCS for 8 consecutive days was applied to the rats. Fourteen days after the induction of NP, rats developed mechanical hyperalgesia with a decreased threshold, and at the end of treatment, an increase in the pain threshold was observed in NP rats. In addition, NP rats had increased levels of reactive species (RS) in the prefrontal cortex, while superoxide dismutase (SOD) activity was decreased in NP rats. In the spinal cord, nitrite levels and glutathione-S-transferase (GST) activity decreased in the L-tDCS group, and it was observed that increased levels in total sulfhydryl content for neuropathic pain rats were reversed by tDCS. In serum analyses, the neuropathic pain model increased the levels of RS and thiobarbituric acid-reactive substances (TBARS) and decreased the activity of butyrylcholinesterase (BuChE). In conclusion, bimodal tDCS increased total sulfhydryl content in the spinal cord of rats with neuropathic pain, positively modulating this parameter.
Collapse
Affiliation(s)
- Priscila Centeno Crespo
- Postgraduate Program in Bioprospecting and Biochemistry, Universidade Federal de Pelotas (UFPel), Pelotas, RS 96010-900, Brazil; Laboratory of Cell Neuromodulation: Basic Research, Biology Institute, Department of Morphology, UFPel, Pelotas, RS 96030-000, Brazil
| | | | - Clara Camacho Dos Reis
- Laboratory of Cell Neuromodulation: Basic Research, Biology Institute, Department of Morphology, UFPel, Pelotas, RS 96030-000, Brazil
| | - Liciane Fernandes Medeiros
- Laboratory of Pain Pharmacology and Neuromodulation: Preclinical Researches, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS 90050-170, Brazil; Postgraduate Program in Health and Human Development, Unilasalle, Canoas, RS 92010-000, Brazil
| | - Vanessa Leal Scarabelot
- Laboratory of Pain Pharmacology and Neuromodulation: Preclinical Researches, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS 90050-170, Brazil; Postgraduate Program in Medicine, Medical Sciences, Medicine School, Universidade Federal do Rio Grande do Sul (UFRGS) Porto Alegre, RS 90035-003, Brazil
| | - Giovana Duzzo Gamaro
- Laboratory of Cell Neuromodulation: Basic Research, Biology Institute, Department of Morphology, UFPel, Pelotas, RS 96030-000, Brazil
| | - Mayara Sandrielly Pereira Soares
- Postgraduate Program in Bioprospecting and Biochemistry, Universidade Federal de Pelotas (UFPel), Pelotas, RS 96010-900, Brazil; Laboratory of Biomarkers, Center of Chemical, Pharmaceutical and Food Sciences, UFPel, Campus Universitário Capão do Leão s/n, Pelotas, RS, Brazil
| | - Roselia Maria Spanevello
- Postgraduate Program in Bioprospecting and Biochemistry, Universidade Federal de Pelotas (UFPel), Pelotas, RS 96010-900, Brazil
| | - Francieli Moro Stefanello
- Postgraduate Program in Bioprospecting and Biochemistry, Universidade Federal de Pelotas (UFPel), Pelotas, RS 96010-900, Brazil; Postgraduate Program in Medicine, Medical Sciences, Medicine School, Universidade Federal do Rio Grande do Sul (UFRGS) Porto Alegre, RS 90035-003, Brazil
| | - Izabel Cristina Custódio De Souza
- Postgraduate Program in Bioprospecting and Biochemistry, Universidade Federal de Pelotas (UFPel), Pelotas, RS 96010-900, Brazil; Laboratory of Cell Neuromodulation: Basic Research, Biology Institute, Department of Morphology, UFPel, Pelotas, RS 96030-000, Brazil.
| |
Collapse
|
22
|
Guo B, Zhang M, Hao W, Wang Y, Zhang T, Liu C. Neuroinflammation mechanisms of neuromodulation therapies for anxiety and depression. Transl Psychiatry 2023; 13:5. [PMID: 36624089 PMCID: PMC9829236 DOI: 10.1038/s41398-022-02297-y] [Citation(s) in RCA: 102] [Impact Index Per Article: 51.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2022] [Revised: 12/19/2022] [Accepted: 12/21/2022] [Indexed: 01/11/2023] Open
Abstract
Mood disorders are associated with elevated inflammation, and the reduction of symptoms after multiple treatments is often accompanied by pro-inflammation restoration. A variety of neuromodulation techniques that regulate regional brain activities have been used to treat refractory mood disorders. However, their efficacy varies from person to person and lack reliable indicator. This review summarizes clinical and animal studies on inflammation in neural circuits related to anxiety and depression and the evidence that neuromodulation therapies regulate neuroinflammation in the treatment of neurological diseases. Neuromodulation therapies, including transcranial magnetic stimulation (TMS), transcranial electrical stimulation (TES), electroconvulsive therapy (ECT), photobiomodulation (PBM), transcranial ultrasound stimulation (TUS), deep brain stimulation (DBS), and vagus nerve stimulation (VNS), all have been reported to attenuate neuroinflammation and reduce the release of pro-inflammatory factors, which may be one of the reasons for mood improvement. This review provides a better understanding of the effective mechanism of neuromodulation therapies and indicates that inflammatory biomarkers may serve as a reference for the assessment of pathological conditions and treatment options in anxiety and depression.
Collapse
Affiliation(s)
- Bingqi Guo
- grid.413259.80000 0004 0632 3337Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, 100053 China ,grid.24696.3f0000 0004 0369 153XBeijing Key Laboratory of Neuromodulation, Beijing, 100053 China
| | - Mengyao Zhang
- grid.413259.80000 0004 0632 3337Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, 100053 China ,grid.24696.3f0000 0004 0369 153XBeijing Key Laboratory of Neuromodulation, Beijing, 100053 China
| | - Wensi Hao
- grid.413259.80000 0004 0632 3337Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, 100053 China ,grid.24696.3f0000 0004 0369 153XBeijing Key Laboratory of Neuromodulation, Beijing, 100053 China
| | - Yuping Wang
- grid.413259.80000 0004 0632 3337Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, 100053 China ,grid.24696.3f0000 0004 0369 153XBeijing Key Laboratory of Neuromodulation, Beijing, 100053 China ,grid.24696.3f0000 0004 0369 153XInstitute of sleep and consciousness disorders, Center of Epilepsy, Beijing Institute for Brain Disorders, Capital Medical University, Beijing, 100069 China
| | - Tingting Zhang
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China. .,Beijing Key Laboratory of Neuromodulation, Beijing, 100053, China.
| | - Chunyan Liu
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China. .,Beijing Key Laboratory of Neuromodulation, Beijing, 100053, China.
| |
Collapse
|
23
|
Wu C, Yang L, Feng S, Zhu L, Yang L, Liu TCY, Duan R. Therapeutic non-invasive brain treatments in Alzheimer's disease: recent advances and challenges. Inflamm Regen 2022; 42:31. [PMID: 36184623 PMCID: PMC9527145 DOI: 10.1186/s41232-022-00216-8] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Accepted: 06/13/2022] [Indexed: 11/10/2022] Open
Abstract
Alzheimer's disease (AD) is one of the major neurodegenerative diseases and the most common form of dementia. Characterized by the loss of learning, memory, problem-solving, language, and other thinking abilities, AD exerts a detrimental effect on both patients' and families' quality of life. Although there have been significant advances in understanding the mechanism underlying the pathogenesis and progression of AD, there is no cure for AD. The failure of numerous molecular targeted pharmacologic clinical trials leads to an emerging research shift toward non-invasive therapies, especially multiple targeted non-invasive treatments. In this paper, we reviewed the advances of the most widely studied non-invasive therapies, including photobiomodulation (PBM), transcranial magnetic stimulation (TMS), transcranial direct current stimulation (tDCS), and exercise therapy. Firstly, we reviewed the pathological changes of AD and the challenges for AD studies. We then introduced these non-invasive therapies and discussed the factors that may affect the effects of these therapies. Additionally, we review the effects of these therapies and the possible mechanisms underlying these effects. Finally, we summarized the challenges of the non-invasive treatments in future AD studies and clinical applications. We concluded that it would be critical to understand the exact underlying mechanisms and find the optimal treatment parameters to improve the translational value of these non-invasive therapies. Moreover, the combined use of non-invasive treatments is also a promising research direction for future studies and sheds light on the future treatment or prevention of AD.
Collapse
Affiliation(s)
- Chongyun Wu
- Laboratory of Regenerative Medicine in Sports Science, School of Physical Education and Sports Science, South China Normal University, Guangzhou, 510006, China
| | - Luoman Yang
- Department of Anesthesiology, Peking University Third Hospital (PUTH), Beijing, 100083, China
| | - Shu Feng
- Laboratory of Regenerative Medicine in Sports Science, School of Physical Education and Sports Science, South China Normal University, Guangzhou, 510006, China
| | - Ling Zhu
- Laboratory of Regenerative Medicine in Sports Science, School of Physical Education and Sports Science, South China Normal University, Guangzhou, 510006, China
| | - Luodan Yang
- Department of Neurology, Louisiana State University Health Sciences Center, 1501 Kings Highway, Shreveport, LA, 71103, USA.
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA, 30912, USA.
| | - Timon Cheng-Yi Liu
- Laboratory of Regenerative Medicine in Sports Science, School of Physical Education and Sports Science, South China Normal University, Guangzhou, 510006, China.
| | - Rui Duan
- Laboratory of Regenerative Medicine in Sports Science, School of Physical Education and Sports Science, South China Normal University, Guangzhou, 510006, China.
| |
Collapse
|
24
|
Xia H, Wang H, Li Y, Luo Y, Liu Y, Wang F. Study on the Function of miR-134 on Cognitive Function of Vascular Dementia (VD) Rats and Mechanism About Oxidative Stress and Autophagy and Cofilin 2 Level. J BIOMATER TISS ENG 2022. [DOI: 10.1166/jbt.2022.3153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
To discuss the mechanism of miR-134 in improving cognitive function of VD rats through regulating the oxidative stress and autophagy and reducing the expression of Cofilin 2. VD rats was established. They were disposed with miR-134 antagonist. The cerebral regulatory capacity was observed
through ethology. The pathological change in CAI area of hippocampus and cerebral cortex was observed with HE staining method. The regulation of miR-134 targeting downstream was analyzed through bioinformatics. The presentation level of SOD, GSH, ROS and MDA was detected. The expression of
LC1/LC-3 and p62 was detected with Western Blot assay. There was visible activated microglial cells and gliocyte proliferation in VD rat’s model. The myelination was weakened. They were improved notably through the treatment with miR-134 antagonist. The expression of MDA and ROS could
be restrained by miR-134 antagonist through reducing the expression of Cofilin 2. The expression of SOD and GSH could be increased and oxidative stress could be reduced. The level of autophagy could be decreased. The cognitive function of VD rats could be improved by miR-134 antagonist through
regulating the oxidative stress and autophagy and reducing presentation of Cofilin 2.
Collapse
Affiliation(s)
- Haimiao Xia
- The First Department of Neurology, The First Affiliated Hospital of Qiqihar Medical College, Qiqihar City, 161000, Heilongjiang Province, China
| | - Haipeng Wang
- The Third Department of Neurology, The First Affiliated Hospital of Qiqihar Medical College, Qiqihar City, 161000, Heilongjiang Province, China
| | - Yue Li
- The First Department of Neurology, The First Affiliated Hospital of Qiqihar Medical College, Qiqihar City, 161000, Heilongjiang Province, China
| | - Ye Luo
- The First Department of Neurology, The First Affiliated Hospital of Qiqihar Medical College, Qiqihar City, 161000, Heilongjiang Province, China
| | - Yuxiang Liu
- The First Department of Neurology, The First Affiliated Hospital of Qiqihar Medical College, Qiqihar City, 161000, Heilongjiang Province, China
| | - Feng Wang
- The First Department of Neurology, The First Affiliated Hospital of Qiqihar Medical College, Qiqihar City, 161000, Heilongjiang Province, China
| |
Collapse
|
25
|
Jangwan NS, Ashraf GM, Ram V, Singh V, Alghamdi BS, Abuzenadah AM, Singh MF. Brain augmentation and neuroscience technologies: current applications, challenges, ethics and future prospects. Front Syst Neurosci 2022; 16:1000495. [PMID: 36211589 PMCID: PMC9538357 DOI: 10.3389/fnsys.2022.1000495] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Accepted: 08/31/2022] [Indexed: 12/02/2022] Open
Abstract
Ever since the dawn of antiquity, people have strived to improve their cognitive abilities. From the advent of the wheel to the development of artificial intelligence, technology has had a profound leverage on civilization. Cognitive enhancement or augmentation of brain functions has become a trending topic both in academic and public debates in improving physical and mental abilities. The last years have seen a plethora of suggestions for boosting cognitive functions and biochemical, physical, and behavioral strategies are being explored in the field of cognitive enhancement. Despite expansion of behavioral and biochemical approaches, various physical strategies are known to boost mental abilities in diseased and healthy individuals. Clinical applications of neuroscience technologies offer alternatives to pharmaceutical approaches and devices for diseases that have been fatal, so far. Importantly, the distinctive aspect of these technologies, which shapes their existing and anticipated participation in brain augmentations, is used to compare and contrast them. As a preview of the next two decades of progress in brain augmentation, this article presents a plausible estimation of the many neuroscience technologies, their virtues, demerits, and applications. The review also focuses on the ethical implications and challenges linked to modern neuroscientific technology. There are times when it looks as if ethics discussions are more concerned with the hypothetical than with the factual. We conclude by providing recommendations for potential future studies and development areas, taking into account future advancements in neuroscience innovation for brain enhancement, analyzing historical patterns, considering neuroethics and looking at other related forecasts.
Collapse
Affiliation(s)
- Nitish Singh Jangwan
- Department of Pharmacology, School of Pharmaceutical Sciences and Technology, Sardar Bhagwan Singh University, Balawala, India
| | - Ghulam Md Ashraf
- Pre-Clinical Research Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Veerma Ram
- Department of Pharmacology, School of Pharmaceutical Sciences and Technology, Sardar Bhagwan Singh University, Balawala, India
| | - Vinod Singh
- Prabha Harji Lal College of Pharmacy and Paraclinical Sciences, University of Jammu, Jammu, India
| | - Badrah S. Alghamdi
- Pre-Clinical Research Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Physiology, Neuroscience Unit, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Adel Mohammad Abuzenadah
- Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Mamta F. Singh
- Department of Pharmacology, School of Pharmaceutical Sciences and Technology, Sardar Bhagwan Singh University, Balawala, India
| |
Collapse
|
26
|
Zhao K, Zeng L, Cai Z, Liu M, Sun T, Li Z, Liu R. RNA sequencing-based identification of the regulatory mechanism of microRNAs, transcription factors, and corresponding target genes involved in vascular dementia. Front Neurosci 2022; 16:917489. [PMID: 36203804 PMCID: PMC9531238 DOI: 10.3389/fnins.2022.917489] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 08/22/2022] [Indexed: 11/13/2022] Open
Abstract
Vascular dementia (VaD) is the second most common form of dementia with uncertain mechanisms and no effective treatments. microRNAs (miRNAs) and transcription factors (TFs) are considered regulatory factors of genes involved in many diseases. Therefore, this work investigated the aberrantly expressed miRNAs, TFs, corresponding target genes, and their co-regulatory networks in the cortex of rats with bilateral common carotid artery occlusion (2VO) to uncover the potential mechanism and biomarkers of VaD. Differentially expressed genes (DEGs), miRNAs (DEMs), and TFs (DETFs) were identified using RNA sequencing, and their interaction networks were constructed using Cytoscape. The results showed that rats with 2VO had declined cognitive abilities and neuronal loss in the cortex than sham rats. DEGs, DEMs, and DETFs were discriminated between rats with 2VO and sham rats in the cortex, as shown by the 13 aberrantly expressed miRNAs, 805 mRNAs, and 63 TFs. The miRNA-TF-target gene network was constructed, showing 523 nodes and 7237 edges. Five miRNAs (miR-5132-5p, miR-764-3p, miR-223-3p, miR-145-5p, and miR-122-5p), ten TFs (Mxi1, Nfatc4, Rxrg, Zfp523, Foxj2, Nkx6-1, Klf4, Klf5, Csrnp1, and Prdm6), and seven target genes (Serpine1, Nedd4l, Pxn, Col1a1, Plec, Trip12, and Tpm1) were chosen as the significant nodes to construct feed-forward loops (FFLs). Gene Ontology and pathway enrichment analysis revealed that these miRNA and TF-associated genes are mostly involved in the PI3K/Akt, neuroactive ligand–receptor interaction, calcium signaling, and Wnt signaling pathways, along with central locations around the cell membrane. They exert functions such as growth factor binding, integrin binding, and extracellular matrix structural constituent, with representative biological processes like vasculature development, cell–substrate adhesion, cellular response to growth factor stimulus, and synaptic transmission. Furthermore, the expression of three miRNAs (miR-145-5p, miR-122-5p, and miR-5132-5p), six TFs (Csrnp1, Klf4, Nfatc4, Rxrg, Foxj2, and Klf5), and five mRNAs (Serpine1, Plec, Nedd4l, Trip12, and Tpm1) were significantly changed in rats with VaD, in line with the outcome of RNA sequencing. In the potential FFL, miR-145-5p directly bound Csrnp1 and decreased its mRNA expression. These results might help the understanding of the underlying regulatory mechanisms of miRNA-TF-genes, providing potential therapeutic targets in VaD.
Collapse
|
27
|
Guidetti M, Arlotti M, Bocci T, Bianchi AM, Parazzini M, Ferrucci R, Priori A. Electric Fields Induced in the Brain by Transcranial Electric Stimulation: A Review of In Vivo Recordings. Biomedicines 2022; 10:biomedicines10102333. [PMID: 36289595 PMCID: PMC9598743 DOI: 10.3390/biomedicines10102333] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 09/10/2022] [Accepted: 09/14/2022] [Indexed: 01/12/2023] Open
Abstract
Transcranial electrical stimulation (tES) techniques, such as direct current stimulation (tDCS) and transcranial alternating current stimulation (tACS), cause neurophysiological and behavioral modifications as responses to the electric field are induced in the brain. Estimations of such electric fields are based mainly on computational studies, and in vivo measurements have been used to expand the current knowledge. Here, we review the current tDCS- and tACS-induced electric fields estimations as they are recorded in humans and non-human primates using intracerebral electrodes. Direct currents and alternating currents were applied with heterogeneous protocols, and the recording procedures were characterized by a tentative methodology. However, for the clinical stimulation protocols, an injected current seems to reach the brain, even at deep structures. The stimulation parameters (e.g., intensity, frequency and phase), the electrodes’ positions and personal anatomy determine whether the intensities might be high enough to affect both neuronal and non-neuronal cell activity, also deep brain structures.
Collapse
Affiliation(s)
- Matteo Guidetti
- Aldo Ravelli Research Center for Neurotechnology and Experimental Neurotherapeutics, Department of Health Sciences, University of Milan, Via Antonio di Rudinì 8, 20142 Milan, Italy
- Department of Electronics, Information and Bioengineering, Politecnico di Milano, Piazza Leonardo da Vinci 32, 20133 Milan, Italy
| | | | - Tommaso Bocci
- Aldo Ravelli Research Center for Neurotechnology and Experimental Neurotherapeutics, Department of Health Sciences, University of Milan, Via Antonio di Rudinì 8, 20142 Milan, Italy
- III Neurology Clinic, ASST-Santi Paolo e Carlo University Hospital, 20142 Milan, Italy
| | - Anna Maria Bianchi
- Department of Electronics, Information and Bioengineering, Politecnico di Milano, Piazza Leonardo da Vinci 32, 20133 Milan, Italy
| | - Marta Parazzini
- Istituto di Elettronica e di Ingegneria dell’Informazione e delle Telecomunicazioni (IEIIT), Consiglio Nazionale delle Ricerche (CNR), 20133 Milan, Italy
| | - Roberta Ferrucci
- Aldo Ravelli Research Center for Neurotechnology and Experimental Neurotherapeutics, Department of Health Sciences, University of Milan, Via Antonio di Rudinì 8, 20142 Milan, Italy
- III Neurology Clinic, ASST-Santi Paolo e Carlo University Hospital, 20142 Milan, Italy
| | - Alberto Priori
- Aldo Ravelli Research Center for Neurotechnology and Experimental Neurotherapeutics, Department of Health Sciences, University of Milan, Via Antonio di Rudinì 8, 20142 Milan, Italy
- III Neurology Clinic, ASST-Santi Paolo e Carlo University Hospital, 20142 Milan, Italy
- Correspondence:
| |
Collapse
|
28
|
Zhang J, Zhang Y, Liu Y, Niu X. Naringenin Attenuates Cognitive Impairment in a Rat Model of Vascular Dementia by Inhibiting Hippocampal Oxidative Stress and Inflammatory Response and Promoting N-Methyl-D-Aspartate Receptor Signaling Pathway. Neurochem Res 2022; 47:3402-3413. [PMID: 36028734 DOI: 10.1007/s11064-022-03696-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Revised: 07/11/2022] [Accepted: 07/14/2022] [Indexed: 10/15/2022]
Abstract
Vascular dementia (VaD) is the second most common form of dementia globally, yet there are no efficient treatments. Naringenin, a natural flavonoid, exerts antioxidative, anti-inflammatory, and neuroprotective properties; however, its potential effect on VaD remain unclear. Herein, the purpose of present study was to elucidate whether naringenin attenuates cognitive dysfunction in VaD via inhibiting hippocampal oxidative stress and inflammatory response, and promoting N-methyl-D-aspartate receptors (NMDARs) signaling pathway. A rat model of VaD was established by permanent bilateral common carotid artery occlusion [2-vessel occlusion (2VO)]. Behavioral performance analyses results revealed that administration of naringenin improves cognitive impairment in rats with VaD according to the new object recognition test and the Morris water maze test. In addition, naringenin attenuated hippocampal oxidative stress by reducing reactive oxygen species generation, decreasing malondialdehyde content and recombinant reactive oxygen species modulator 1 (Romo-1) expression, and increasing superoxide dismutase and glutathione peroxidase activities in the hippocampus of VaD rats. Moreover, naringenin decreased the proinflammatory cytokines (IL-1β, IL-6, and TNF-α) levels and increased the anti-inflammatory cytokines (IL-10 and IL-4) levels in the hippocampus of 2VO surgery-treated rats, attenuating hippocampal inflammatory response during VaD. Furthermore, naringenin promoted synaptophysin (SYP), postsynaptic density protein 95 (PSD95), N-methyl-Daspartic acid receptor 1 (NR1) and N-methyl-D-aspartate receptor subunit 2B (NR2B) expressions levels in hippocampus of VaD rats. Collectively, these findings indicated that naringenin mitigates cognitive impairment in VaD rats partly via inhibiting hippocampal oxidative stress and inflammatory response and restoring NMDARs signaling pathway.
Collapse
Affiliation(s)
- Jin Zhang
- Department of Neurology, The First Hospital of Shanxi Medical University, No. 85 Jiefang South Road, Yingze District, Taiyuan, 030000, Shanxi, People's Republic of China
| | - Yu Zhang
- Department of Neurology, Shanxi Hospital of Integrated Traditional and Western Medicine, Taiyuan, 030000, Shanxi, People's Republic of China
| | - Yan Liu
- Department of Neurology, The First Hospital of Shanxi Medical University, No. 85 Jiefang South Road, Yingze District, Taiyuan, 030000, Shanxi, People's Republic of China
| | - Xiaoyuan Niu
- Department of Neurology, The First Hospital of Shanxi Medical University, No. 85 Jiefang South Road, Yingze District, Taiyuan, 030000, Shanxi, People's Republic of China.
| |
Collapse
|
29
|
Jenkins TA. Metabolic Syndrome and Vascular-Associated Cognitive Impairment: a Focus on Preclinical Investigations. Curr Diab Rep 2022; 22:333-340. [PMID: 35737273 PMCID: PMC9314301 DOI: 10.1007/s11892-022-01475-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/22/2022] [Indexed: 11/03/2022]
Abstract
PURPOSE OF REVIEW Metabolic syndrome is associated with an increased risk of vascular cognitive impairment or, in the more extreme, vascular dementia. Animal models are used to investigate the relationship between pathology and behaviour. This review summarizes the latest understanding of the role of the hippocampus and prefrontal cortex in vascular cognitive impairment, the influence of inflammation in this association while also commenting on some of the latest interventions proposed. RECENT FINDINGS Models of vascular cognitive impairment and vascular dementia, whether they develop from an infarct or non-infarct base, demonstrate increased neuroinflammation, reduced neuronal function and deficits in prefrontal and hippocampal-associated cognitive domains. Promising new research shows agents and environmental interventions that inhibit central oxidative stress and inflammation can reverse both pathology and cognitive dysfunction. While preclinical studies suggest that reversal of deficits in vascular cognitive impairment models is possible, replication in patients still needs to be demonstrated.
Collapse
Affiliation(s)
- Trisha A Jenkins
- Human Biosciences, School of Health and Biomedical Sciences, STEM College, RMIT University, Plenty Road, Bundoora, VIC, 3083, Australia.
| |
Collapse
|
30
|
Kaviannejad R, Karimian SM, Riahi E, Ashabi G. Using dual polarities of transcranial direct current stimulation in global cerebral ischemia and its following reperfusion period attenuates neuronal injury. Metab Brain Dis 2022; 37:1503-1516. [PMID: 35499797 DOI: 10.1007/s11011-022-00985-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 04/10/2022] [Indexed: 10/18/2022]
Abstract
Multiple neuronal injury pathways are activated during cerebral ischemia and reperfusion (I/R). This study was designed to decrease potential neuronal injuries by using both transcranial direct current stimulation (tDCS) polarities in cerebral ischemia and its following reperfusion period. Ninety rats were randomly divided into six groups. In the sham group, rats were intact. In the I/R group, global cerebral I/R was only induced. In the I/R + c-tDCS and I/R + a-tDCS groups, cathodal and anodal currents were applied, respectively. In the I/R + c/a-tDCS, cathodal current was used in the cerebral ischemia and anodal in the reperfusion. In the I/R + a/c-tDCS group, cathodal and anodal currents were applied in the I/R, respectively. Hippocampal tissue was used to determine the levels of IL-1β, TNF-α, NOS, SOD, MDA, and NMDAR. Hot plate and open field tests evaluated sensory and locomotor performances. The cerebral edema was also measured. Histological assessment was assessed by H/E and Nissl staining of the hippocampal CA1 region. All tDCS modes significantly decreased IL-1β and TNF-α levels, especially in the c/a-tDCS. All tDCS caused a significant decrease in MDA and NOS levels while increasing SOD activity compared to the I/R group, especially in the c/a-tDCS mode. In the c-tDCS and a/c-tDCS groups, the NMDAR level was significantly decreased. The c/a-tDCS group improved sensory and locomotor performances more than other groups receiving tDCS. Furthermore, the least neuronal death was observed in the c/a-tDCS mode. Using two different polarities of tDCS could induce more neuroprotective versus pathophysiological pathways in cerebral I/R, especially in c/a-tDCS mode. HIGHLIGHTS: Multiple pathways of neuronal injury are activated in cerebral ischemia and reperfusion (I/R). Using tDCS could modulate neuroinflammation and oxidative stress pathways in global cerebral I/R. Using c/a-tDCS mode during cerebral I/R causes more neuroprotective effects against neuronal injuries of cerebral I/R.
Collapse
Affiliation(s)
- Rasoul Kaviannejad
- Department of Physiology, School of Medicine, Tehran University of Medical Sciences, PourSina St., 1417613151, Tehran, Iran
| | - Seyed Morteza Karimian
- Department of Physiology, School of Medicine, Tehran University of Medical Sciences, PourSina St., 1417613151, Tehran, Iran.
| | - Esmail Riahi
- Department of Physiology, School of Medicine, Tehran University of Medical Sciences, PourSina St., 1417613151, Tehran, Iran
| | - Ghorbangol Ashabi
- Department of Physiology, School of Medicine, Tehran University of Medical Sciences, PourSina St., 1417613151, Tehran, Iran
| |
Collapse
|
31
|
Kaviannejad R, Karimian SM, Riahi E, Ashabi G. The neuroprotective effects of transcranial direct current stimulation on global cerebral ischemia and reperfusion via modulating apoptotic pathways. Brain Res Bull 2022; 186:70-78. [PMID: 35654262 DOI: 10.1016/j.brainresbull.2022.05.013] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 05/25/2022] [Accepted: 05/26/2022] [Indexed: 11/30/2022]
Abstract
BACKGROUND Cerebral ischemia-reperfusion, subsequent hyperthermia, and hyperglycemia lead to neural damage. This study aimed to investigate the effects of using cathodal and/or anodal transcranial direct current stimulation (tDCS) in different stages of ischemia-reperfusion on apoptosis and controlling hyperthermia and hyperglycemia. MATERIALS AND METHODS A total of 78 male Wistar rats were randomly assigned into six groups (n=13), including sham, ischemia/reperfusion (I/R), anodal-tDCS (a-tDCS), cathodal-tDCS (c-tDCS), anodal/cathodal-tDCS (a/c-tDCS), and cathodal/anodal-tDCS (c/a-tDCS) groups. Global cerebral I/R was induced in all of the groups except for sham group. In a-tDCS and c-tDCS groups, the rats received anodal and cathodal currents in both I/R stages, respectively. In a/c-tDCS group, the rats received anodal current during the ischemia and cathodal current during the reperfusion. The c/a-tDCS group received the currents in the reverse order. The current intensity of 400µA was applied in ischemia phase (15min) and reperfusion phase (30min, twice a day). Body temperature and plasma blood sugar were measured daily. Rats were also tested for novel object recognition and passive avoidance memory. The apoptosis of hippocampal tissue was evaluated by measuring Bax, Bcl-2, Caspase-3, and TUNEL staining. RESULTS All tDCS significantly reduced hyperthermia and hyperglycemia, as well as Bax and Caspase-3 levels, it also increased Bcl-2 expression. The preliminary results from c/a-tDCS mode could improve the expression of apoptotic markers, memory function, hyperthermia, and hyperglycemia control and reduce DNA fragmentation compared to other stimulatory therapies. CONCLUSION All tDCS modes could save neurons by suppressing apoptotic and enhancing anti-apoptotic pathways, especially in the c/a tDCS mode.
Collapse
Affiliation(s)
- Rasoul Kaviannejad
- Department of Physiology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran; Department of Anesthesiology, School of Allied Medical Sciences, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| | - Seyed Morteza Karimian
- Department of Physiology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| | - Esmail Riahi
- Department of Physiology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| | - Ghorbangol Ashabi
- Department of Physiology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
32
|
Shu J, Wei W, Zhang L. Identification of Molecular Signatures and Candidate Drugs in Vascular Dementia by Bioinformatics Analyses. Front Mol Neurosci 2022; 15:751044. [PMID: 35221911 PMCID: PMC8873373 DOI: 10.3389/fnmol.2022.751044] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 01/17/2022] [Indexed: 01/30/2023] Open
Abstract
Vascular dementia (VaD) is considered to be the second most common form of dementia after Alzheimer’s disease, and no specific drugs have been approved for VaD treatment. We aimed to identify shared transcriptomic signatures between the frontal cortex and temporal cortex in VaD by bioinformatics analyses. Gene ontology and pathway enrichment analyses, protein–protein interaction (PPI) and hub gene identification, hub gene–transcription factor interaction, hub gene–microRNA interaction, and hub gene–drug interaction analyses were performed. We identified 159 overlapping differentially expressed genes (DEGs) between the frontal cortex and temporal cortex that were enriched mainly in inflammation and innate immunity, synapse pruning, regeneration, positive regulation of angiogenesis, response to nutrient levels, and positive regulation of the digestive system process. We identified 10 hub genes in the PPI network (GNG13, CD163, C1QA, TLR2, SST, C1QB, ITGB2, CCR5, CRH, and TAC1), four central regulatory transcription factors (FOXC1, CREB1, GATA2, and HINFP), and four microRNAs (miR-27a-3p, miR-146a-5p, miR-335-5p, and miR-129-2-3p). Hub gene–drug interaction analysis found four drugs (maraviroc, cenicriviroc, PF-04634817, and efalizumab) that could be potential drugs for VaD treatment. Together, our results may contribute to understanding the underlying mechanisms in VaD and provide potential targets and drugs for therapeutic intervention.
Collapse
|
33
|
Deng Y, Zhang X, Chen F, Huang J, Zhang D, Luo J. HO-1 mediated by PI3K/Akt/Nrf2 signaling pathway is involved in (-)-epigallocatechin-3-gallate-rescueing impaired cognitive function induced by chronic cerebral hypoperfusion in rat model. Exp Aging Res 2022; 48:428-443. [DOI: 10.1080/0361073x.2021.2011689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Affiliation(s)
- Yu Deng
- Department of Geratology, Chongqing Mental Health Center, Chongqing, China
| | - Xiong Zhang
- Neuroscience Research Center, Chongqing Medical University, Chongqing, China
| | - Fei Chen
- Department of Geratology, Chongqing Mental Health Center, Chongqing, China
| | - Jie Huang
- Department of Geratology, Chongqing Mental Health Center, Chongqing, China
| | - Daijiang Zhang
- Department of Geratology, Chongqing Mental Health Center, Chongqing, China
| | - Jie Luo
- Department of Geratology, Chongqing Mental Health Center, Chongqing, China
| |
Collapse
|
34
|
A Single Immediate Use of the Cathodal Transcranial Direct Current Stimulation Induces Neuroprotection of Hippocampal Region Against Global Cerebral Ischemia. J Stroke Cerebrovasc Dis 2022; 31:106241. [PMID: 34983004 DOI: 10.1016/j.jstrokecerebrovasdis.2021.106241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Revised: 11/17/2021] [Accepted: 11/21/2021] [Indexed: 11/20/2022] Open
Abstract
OBJECTIVES Global cerebral ischemia (CI) causes severe neuronal injury, mainly in the hippocampal CA1 region. This study aimed to investigate an immediate using transcranial direct current stimulation (tDCS) in reducing neuronal injury induced by CI. MATERIALS AND METHODS The 32 Wistar male rats were randomly divided into four groups (n=8 per group). In the ischemia group (I), CI was induced via the 4-vessel occlusion model. In the sham group (Sh), rats did not receive any intervention. In the ischemia+cathodal group (I+c/tDCS), the cathodal current was applied during CI. In the ischemia+anodal group (I+a/tDCS), the anodal current was applied. The current intensity of 400 μA was applied for 15-min during the ischemia. Hippocampal tissue was used to assess levels of NMDAR, IL-1β, TNF-α, MDA, SOD, NOS, and apoptosis markers. Histological assessment and TUNEL staining were performed in CA1 hippocampal region. RESULTS The c/tDCS significantly decreased the levels of IL-1β and TNF-α than the I and a/tDCS groups. The c/tDCS significantly reduced MDA and NOS levels, while increasing the level of SOD than the I and a/tDCS. The c/tDCS caused a significant decrease in NMDAR level than the a/tDCS. Using c/tDCS significantly reduced the Bax and Caspase-3 expressions, while increasing the Bcl-2 expression than the I group. In the c/tDCS group, DNA fragmentation and neuronal death were significantly lower than the I and a/tDCS groups. CONCLUSION Using cathodal a direct current could attenuate primary pathophysiological pathways induced by CI, and it eventually reduced neurons death and apoptosis in the CA1 hippocampal region.
Collapse
|
35
|
Liu CS, Herrmann N, Song BX, Ba J, Gallagher D, Oh PI, Marzolini S, Rajji TK, Charles J, Papneja P, Rapoport MJ, Andreazza AC, Vieira D, Kiss A, Lanctôt KL. Exercise priming with transcranial direct current stimulation: a study protocol for a randomized, parallel-design, sham-controlled trial in mild cognitive impairment and Alzheimer's disease. BMC Geriatr 2021; 21:677. [PMID: 34863115 PMCID: PMC8645072 DOI: 10.1186/s12877-021-02636-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 11/16/2021] [Indexed: 12/14/2022] Open
Abstract
Background Transcranial direct current stimulation (tDCS) is a non-invasive type of brain stimulation that uses electrical currents to modulate neuronal activity. A small number of studies have investigated the effects of tDCS on cognition in patients with Mild Cognitive Impairment (MCI) and Alzheimer’s disease (AD), and have demonstrated variable effects. Emerging evidence suggests that tDCS is most effective when applied to active brain circuits. Aerobic exercise is known to increase cortical excitability and improve brain network connectivity. Exercise may therefore be an effective, yet previously unexplored primer for tDCS to improve cognition in MCI and mild AD. Methods Participants with MCI or AD will be randomized to receive 10 sessions over 2 weeks of either exercise primed tDCS, exercise primed sham tDCS, or tDCS alone in a blinded, parallel-design trial. Those randomized to an exercise intervention will receive individualized 30-min aerobic exercise prescriptions to achieve a moderate-intensity dosage, equivalent to the ventilatory anaerobic threshold determined by cardiopulmonary assessment, to sufficiently increase cortical excitability. The tDCS protocol consists of 20 min sessions at 2 mA, 5 times per week for 2 weeks applied through 35 cm2 bitemporal electrodes. Our primary aim is to assess the efficacy of exercise primed tDCS for improving global cognition using the Montreal Cognitive Assessment (MoCA). Our secondary aims are to evaluate the efficacy of exercise primed tDCS for improving specific cognitive domains using various cognitive tests (n-back, Word Recall and Word Recognition Tasks from the Alzheimer’s Disease Assessment Scale-Cognitive subscale) and neuropsychiatric symptoms (Neuropsychiatric Inventory). We will also explore whether exercise primed tDCS is associated with an increase in markers of neurogenesis, oxidative stress and angiogenesis, and if changes in these markers are correlated with cognitive improvement. Discussion We describe a novel clinical trial to investigate the effects of exercise priming before tDCS in patients with MCI or mild AD. This proof-of-concept study may identify a previously unexplored, non-invasive, non-pharmacological combination intervention that improves cognitive symptoms in patients. Findings from this study may also identify potential mechanistic actions of tDCS in MCI and mild AD. Trial registration Clinicaltrials.gov, NCT03670615. Registered on September 13, 2018.
Collapse
Affiliation(s)
- Celina S Liu
- Department of Pharmacology & Toxicology, University of Toronto, 1 King's College Circle, Toronto, ON, M5S 3K1, Canada.,Neuropsychopharmacology Research Group, Hurvitz Brain Sciences Program, Sunnybrook Research Institute, 2075 Bayview Avenue, Toronto, ON, M4N 3M5, Canada
| | - Nathan Herrmann
- Neuropsychopharmacology Research Group, Hurvitz Brain Sciences Program, Sunnybrook Research Institute, 2075 Bayview Avenue, Toronto, ON, M4N 3M5, Canada.,Department of Psychiatry, University of Toronto, Toronto, ON, Canada.,Department of Psychiatry, Division of Geriatric Psychiatry, Sunnybrook Health Sciences Centre, 2075 Bayview Avenue, Toronto, Ontario, M4N 3M5, Canada
| | - Bing Xin Song
- Department of Pharmacology & Toxicology, University of Toronto, 1 King's College Circle, Toronto, ON, M5S 3K1, Canada.,Neuropsychopharmacology Research Group, Hurvitz Brain Sciences Program, Sunnybrook Research Institute, 2075 Bayview Avenue, Toronto, ON, M4N 3M5, Canada
| | - Joycelyn Ba
- Neuropsychopharmacology Research Group, Hurvitz Brain Sciences Program, Sunnybrook Research Institute, 2075 Bayview Avenue, Toronto, ON, M4N 3M5, Canada.,Department of Biology, Faculty of Science, The University of Western Ontario, London, ON, Canada
| | - Damien Gallagher
- Neuropsychopharmacology Research Group, Hurvitz Brain Sciences Program, Sunnybrook Research Institute, 2075 Bayview Avenue, Toronto, ON, M4N 3M5, Canada.,Department of Psychiatry, University of Toronto, Toronto, ON, Canada.,Department of Psychiatry, Division of Geriatric Psychiatry, Sunnybrook Health Sciences Centre, 2075 Bayview Avenue, Toronto, Ontario, M4N 3M5, Canada
| | - Paul I Oh
- Cardiovascular Prevention and Rehabilitation Program, KITE - Toronto Rehabilitation Institute, University Health Network, 347 Rumsey Road, Toronto, ON, M5G 1R7, Canada
| | - Susan Marzolini
- Cardiovascular Prevention and Rehabilitation Program, KITE - Toronto Rehabilitation Institute, University Health Network, 347 Rumsey Road, Toronto, ON, M5G 1R7, Canada
| | - Tarek K Rajji
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada.,Adult Neurodevelopment and Geriatric Psychiatry Division, Centre for Addiction & Mental Health, 80 Workman Way, Toronto, ON, M6J 1H4, Canada.,Toronto Dementia Research Alliance, University of Toronto, Toronto, ON, Canada
| | - Jocelyn Charles
- Family & Community Medicine, Sunnybrook Health Sciences Centre, 2075 Bayview Avenue, Toronto, ON, M4N 3M5, Canada
| | - Purti Papneja
- Family & Community Medicine, Sunnybrook Health Sciences Centre, 2075 Bayview Avenue, Toronto, ON, M4N 3M5, Canada
| | - Mark J Rapoport
- Neuropsychopharmacology Research Group, Hurvitz Brain Sciences Program, Sunnybrook Research Institute, 2075 Bayview Avenue, Toronto, ON, M4N 3M5, Canada.,Department of Psychiatry, University of Toronto, Toronto, ON, Canada.,Department of Psychiatry, Division of Geriatric Psychiatry, Sunnybrook Health Sciences Centre, 2075 Bayview Avenue, Toronto, Ontario, M4N 3M5, Canada
| | - Ana C Andreazza
- Department of Pharmacology & Toxicology, University of Toronto, 1 King's College Circle, Toronto, ON, M5S 3K1, Canada
| | - Danielle Vieira
- Neuropsychopharmacology Research Group, Hurvitz Brain Sciences Program, Sunnybrook Research Institute, 2075 Bayview Avenue, Toronto, ON, M4N 3M5, Canada
| | - Alex Kiss
- Institute for Clinical Evaluative Sciences, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| | - Krista L Lanctôt
- Department of Pharmacology & Toxicology, University of Toronto, 1 King's College Circle, Toronto, ON, M5S 3K1, Canada. .,Neuropsychopharmacology Research Group, Hurvitz Brain Sciences Program, Sunnybrook Research Institute, 2075 Bayview Avenue, Toronto, ON, M4N 3M5, Canada. .,Department of Psychiatry, University of Toronto, Toronto, ON, Canada. .,Department of Psychiatry, Division of Geriatric Psychiatry, Sunnybrook Health Sciences Centre, 2075 Bayview Avenue, Toronto, Ontario, M4N 3M5, Canada. .,Cardiovascular Prevention and Rehabilitation Program, KITE - Toronto Rehabilitation Institute, University Health Network, 347 Rumsey Road, Toronto, ON, M5G 1R7, Canada.
| |
Collapse
|
36
|
Stuckey SM, Ong LK, Collins-Praino LE, Turner RJ. Neuroinflammation as a Key Driver of Secondary Neurodegeneration Following Stroke? Int J Mol Sci 2021; 22:ijms222313101. [PMID: 34884906 PMCID: PMC8658328 DOI: 10.3390/ijms222313101] [Citation(s) in RCA: 80] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 11/25/2021] [Accepted: 12/01/2021] [Indexed: 01/13/2023] Open
Abstract
Ischaemic stroke involves the rapid onset of focal neurological dysfunction, most commonly due to an arterial blockage in a specific region of the brain. Stroke is a leading cause of death and common cause of disability, with over 17 million people worldwide suffering from a stroke each year. It is now well-documented that neuroinflammation and immune mediators play a key role in acute and long-term neuronal tissue damage and healing, not only in the infarct core but also in distal regions. Importantly, in these distal regions, termed sites of secondary neurodegeneration (SND), spikes in neuroinflammation may be seen sometime after the initial stroke onset, but prior to the presence of the neuronal tissue damage within these regions. However, it is key to acknowledge that, despite the mounting information describing neuroinflammation following ischaemic stroke, the exact mechanisms whereby inflammatory cells and their mediators drive stroke-induced neuroinflammation are still not fully understood. As a result, current anti-inflammatory treatments have failed to show efficacy in clinical trials. In this review we discuss the complexities of post-stroke neuroinflammation, specifically how it affects neuronal tissue and post-stroke outcome acutely, chronically, and in sites of SND. We then discuss current and previously assessed anti-inflammatory therapies, with a particular focus on how failed anti-inflammatories may be repurposed to target SND-associated neuroinflammation.
Collapse
Affiliation(s)
- Shannon M. Stuckey
- Discipline of Anatomy and Pathology, School of Biomedicine, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide 5005, Australia; (S.M.S.); (L.E.C.-P.)
| | - Lin Kooi Ong
- School of Pharmacy, Monash University Malaysia, Subang Jaya 47500, Malaysia;
- School of Biomedical Sciences and Pharmacy and the Priority Research Centre for Stroke and Brain Injury, The University of Newcastle, Callaghan 2308, Australia
| | - Lyndsey E. Collins-Praino
- Discipline of Anatomy and Pathology, School of Biomedicine, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide 5005, Australia; (S.M.S.); (L.E.C.-P.)
| | - Renée J. Turner
- Discipline of Anatomy and Pathology, School of Biomedicine, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide 5005, Australia; (S.M.S.); (L.E.C.-P.)
- Correspondence: ; Tel.: +61-8-8313-3114
| |
Collapse
|
37
|
Yan N, Xu Z, Qu C, Zhang J. Dimethyl fumarate improves cognitive deficits in chronic cerebral hypoperfusion rats by alleviating inflammation, oxidative stress, and ferroptosis via NRF2/ARE/NF-κB signal pathway. Int Immunopharmacol 2021; 98:107844. [PMID: 34153667 DOI: 10.1016/j.intimp.2021.107844] [Citation(s) in RCA: 127] [Impact Index Per Article: 31.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 05/30/2021] [Accepted: 05/31/2021] [Indexed: 12/12/2022]
Abstract
Cerebrovascular disease and its risk factors cause persistent decrease of cerebral blood flow, chronic cerebral hypoperfusion (CCH) is the major foundation of vascular cognitive impairment (VCI). The hippocampus is extremely vulnerable to cerebral ischemia and hypoxia. Oxidative stress and neuroinflammation injury are important pathophysiological mechanisms of this process, which is closely related to hippocampal neurons damage and loss. Dimethyl fumarate (DMF), an FDA-approved therapeutic for multiple sclerosis (MS), plays a protective role in multiple neurological disorders. Studies have shown that DMF exerts anti-inflammatory and antioxidant effects via the NRF2/ARE/NF-κB signaling pathway. Thus, this study aimed to evaluate the neuroprotective effect of DMF in the CCH rat model. Ferroptosis, a novel defined iron-dependent cell death form, were found to be strongly associated with the pathophysiology of CCH. Emerging evidences have shown that inhibition of ferroptosis by targeting NRF2 exerted neuroprotective effect in neurodegeneration diseases. We also investigated whether DMF can alleviate cognitive deficits through inhibition of ferroptosis by the NRF2 signaling pathway in this study. DMF was intragastric for consecutive five weeks (100 mg/kg/day). Then behavior test and histological, molecular, and biochemical analysis were performed. We found that DMF treatment significantly improved cognitive deficits and partially reversed hippocampus neuronal damage and loss caused by CCH. And DMF treatment decreased hippocampus IL-1β, TNF-α, and IL-6 pro-inflammatory cytokines concentration, and mediated the NF-κB signaling pathway. And DMF also alleviated hippocampus oxidative stress through reducing MDA, and increasing GSH and SOD levels, which are also closely associated with ferroptosis. Besides, DMF treatment reduced the expression of PTGS2, and increased the expression of FTH1 and xCT, and the iron content is also reduced, which were the important features related to ferroptosis. Furthermore, DMF activated the NRF2/ARE signaling pathway and upregulated the expression of HO-1, NQO1 and GPX4. These outcomes indicated that DMF can improve cognitive impairment in rats with CCH, possibly through alleviating neuroinflammation, oxidative stress damage and inhibiting ferroptosis of hippocampal neurons. Overall, our results provide new evidence for the neuroprotective role of DMF.
Collapse
Affiliation(s)
- Nao Yan
- Department of Neurology, Zhongnan Hospital of Wuhan University, No. 169, Donghu Road, Wuhan 430071, Hubei, China; Hubei Clinical Research Center for Dementias and Cognitive Impairments, Zhongnan Hospital of Wuhan University, No. 169, Donghu Road, Wuhan 430071, Hubei, China
| | - Zhipeng Xu
- Department of Neurology, Zhongnan Hospital of Wuhan University, No. 169, Donghu Road, Wuhan 430071, Hubei, China; Hubei Clinical Research Center for Dementias and Cognitive Impairments, Zhongnan Hospital of Wuhan University, No. 169, Donghu Road, Wuhan 430071, Hubei, China
| | - Changhua Qu
- Department of Neurology, Zhongnan Hospital of Wuhan University, No. 169, Donghu Road, Wuhan 430071, Hubei, China; Hubei Clinical Research Center for Dementias and Cognitive Impairments, Zhongnan Hospital of Wuhan University, No. 169, Donghu Road, Wuhan 430071, Hubei, China
| | - JunJian Zhang
- Department of Neurology, Zhongnan Hospital of Wuhan University, No. 169, Donghu Road, Wuhan 430071, Hubei, China; Hubei Clinical Research Center for Dementias and Cognitive Impairments, Zhongnan Hospital of Wuhan University, No. 169, Donghu Road, Wuhan 430071, Hubei, China.
| |
Collapse
|
38
|
Zheng Y, Zhang J, Zhao Y, Zhang Y, Zhang X, Guan J, Liu Y, Fu J. Curcumin protects against cognitive impairments in a rat model of chronic cerebral hypoperfusion combined with diabetes mellitus by suppressing neuroinflammation, apoptosis, and pyroptosis. Int Immunopharmacol 2021; 93:107422. [PMID: 33548579 DOI: 10.1016/j.intimp.2021.107422] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 01/12/2021] [Accepted: 01/19/2021] [Indexed: 12/17/2022]
Abstract
BACKGROUND Chronic cerebral hypoperfusion (CCH) is regarded as a high-risk factor for cognitive decline in vascular dementia (VaD). We have previously shown that diabetes mellitus (DM) synergistically promotes CCH-induced cognitive dysfunction via exacerbating neuroinflammation. Furthermore, curcumin has been shown to exhibit anti-inflammatory and neuroprotective activities. However, the effects of curcumin on CCH-induced cognitive impairments in DM have remained unknown. METHODS Rats were fed with a high-fat diet (HFD) and injected with low-dose streptozotocin (STZ), followed by bilateral common carotid artery occlusion (BCCAO), to model DM and CCH in vivo. After BCCAO, curcumin (50 mg/kg) was administered intraperitoneally every two days for eight weeks to evaluate its therapeutic effects. Additionally, mouse BV2 microglial cells were exposed to hypoxia and high glucose to model CCH and DM pathologies in vitro. RESULTS Curcumin treatment significantly improved DM/CCH-induced cognitive deficits and attenuated neuronal cell death. Molecular analysis revealed that curcumin exerted protective effects via suppressing neuroinflammation induced by microglial activation, regulating the triggering receptor expressed on myeloid cells 2 (TREM2)/toll-like receptor 4 (TLR4)/nuclear factor-κB (NF-κB) pathway, alleviating apoptosis, and reducing nod-like receptor protein 3 (NLRP3)-dependent pyroptosis. CONCLUSIONS Taken together, our findings suggest that curcumin represents a promising therapy for DM/CCH-induced cognitive impairments.
Collapse
Affiliation(s)
- Yaling Zheng
- Department of Neurology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai 200233, China
| | - Jiawei Zhang
- Department of Neurology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai 200233, China
| | - Yao Zhao
- Department of Neurology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai 200233, China
| | - Yaxuan Zhang
- Department of Neurology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai 200233, China
| | - Xiaojie Zhang
- Department of Neurology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai 200233, China
| | - Jian Guan
- Shanghai Key Laboratory of Sleep Disordered Breathing, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Yu Liu
- Department of Medicine, Shanghai Eighth People's Hospital, Shanghai 200235, China
| | - Jianliang Fu
- Department of Neurology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai 200233, China; Shanghai Key Laboratory of Sleep Disordered Breathing, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China.
| |
Collapse
|
39
|
Direct current stimulation enhances neuronal alpha-synuclein degradation in vitro. Sci Rep 2021; 11:2197. [PMID: 33500442 PMCID: PMC7838399 DOI: 10.1038/s41598-021-81693-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Accepted: 01/08/2021] [Indexed: 12/20/2022] Open
Abstract
Despite transcranial Direct Current Stimulation (DCS) is currently proposed as a symptomatic treatment in Parkinson's disease, the intracellular and molecular mechanisms elicited by this technique are still unknown, and its disease-modifying potential unexplored. Aim of this study was to elucidate the on-line and off-line effects of DCS on the expression, aggregation and degradation of alpha-synuclein (asyn) in a human neuroblastoma cell line under basal conditions and in presence of pharmachologically-induced increased asyn levels. Following DCS, gene and protein expression of asyn and its main autophagic catabolic pathways were assessed by real-time PCR and Western blot, extracellular asyn levels by Dot blot. We found that, under standard conditions, DCS increased monomeric and reduced oligomeric asyn forms, with a concomitant down-regulation of both macroautophagy and chaperone-mediated autophagy. Differently, in presence of rotenone-induced increased asyn, DCS efficiently counteracted asyn accumulation, not acting on its gene transcription, but potentiating its degradation. DCS also reduced intracellular and extracellular asyn levels, increased following lysosomal inhibition, independently from autophagic degradation, suggesting that other mechanisms are also involved. Collectively, these findings suggest that DCS exerts on-line and off-line effects on the expression, aggregation and autophagic degradation of asyn, indicating a till unknown neuroprotective role of tDCS.
Collapse
|
40
|
Wu D, Xu X, Sun N, Li D, Zhu B, Lin S. AGLPM and QMDDQ peptides exert a synergistic action on memory improvement against scopolamine-induced amnesiac mice. Food Funct 2020; 11:10925-10935. [PMID: 33242042 DOI: 10.1039/d0fo02570d] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
This study aimed to explore the synergistic action of pentapeptides Gln-Met-Asp-Asp-Gln (QMDDQ) and Ala-Gly-Leu-Pro-Met (AGLPM) on memory improvement against scopolamine-induced impairment in mice compared to those of either peptide alone. In behavioral tests, the codelivery of QMDDQ and AGLPM was superior to the individual supplements of either peptide alone not only in enhancing the memory ability at training trials but also in recovering the memory impairment in scopolamine-induced amnesiac mice in test trials. Furthermore, combination treatment with QMDDQ and AGLPM could significantly reduce the acetylcholinesterase (AChE) level and increase the acetylcholine (ACh) level in the hippocampus, and noticeably improve the pathological morphology of the neuron cells in hippocampal regions CA1 and CA2 and dentate gyrus (DG). The findings indicated that the combination treatment with QMDDQ and AGLPM could improve the memory function by regulating the cholinergic system.
Collapse
Affiliation(s)
- Dan Wu
- National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, P.R. China.
| | | | | | | | | | | |
Collapse
|