1
|
Li JZ, Ramalingam N, Li S. Targeting epigenetic mechanisms in amyloid-β-mediated Alzheimer's pathophysiology: unveiling therapeutic potential. Neural Regen Res 2025; 20:54-66. [PMID: 38767476 PMCID: PMC11246147 DOI: 10.4103/nrr.nrr-d-23-01827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Accepted: 02/07/2024] [Indexed: 05/22/2024] Open
Abstract
Alzheimer's disease is a prominent chronic neurodegenerative condition characterized by a gradual decline in memory leading to dementia. Growing evidence suggests that Alzheimer's disease is associated with accumulating various amyloid-β oligomers in the brain, influenced by complex genetic and environmental factors. The memory and cognitive deficits observed during the prodromal and mild cognitive impairment phases of Alzheimer's disease are believed to primarily result from synaptic dysfunction. Throughout life, environmental factors can lead to enduring changes in gene expression and the emergence of brain disorders. These changes, known as epigenetic modifications, also play a crucial role in regulating the formation of synapses and their adaptability in response to neuronal activity. In this context, we highlight recent advances in understanding the roles played by key components of the epigenetic machinery, specifically DNA methylation, histone modification, and microRNAs, in the development of Alzheimer's disease, synaptic function, and activity-dependent synaptic plasticity. Moreover, we explore various strategies, including enriched environments, exposure to non-invasive brain stimulation, and the use of pharmacological agents, aimed at improving synaptic function and enhancing long-term potentiation, a process integral to epigenetic mechanisms. Lastly, we deliberate on the development of effective epigenetic agents and safe therapeutic approaches for managing Alzheimer's disease. We suggest that addressing Alzheimer's disease may require distinct tailored epigenetic drugs targeting different disease stages or pathways rather than relying on a single drug.
Collapse
Affiliation(s)
- Jennie Z Li
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | | | | |
Collapse
|
2
|
Che LQ, Qu ZZ, Mao ZF, Qiao Q, Zhou KP, Jia LJ, Wang WP. Low-frequency rTMS Plays a Neuroprotective role in Pilocarpine-induced Status Epilepticus Rat Models Through the AMPAR GluA1-STIM-Ca 2+ Pathway. Mol Neurobiol 2024:10.1007/s12035-024-04521-w. [PMID: 39384697 DOI: 10.1007/s12035-024-04521-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Accepted: 09/23/2024] [Indexed: 10/11/2024]
Abstract
Low-frequency repetitive transcranial magnetic stimulation (rTMS) refers to the stimulation of the brain using repetitive magnetic field pulses at a low frequency (≤ 1 Hz) to reduce seizures. Currently, the mechanism is not well understood. Male Sprague-Dawley rats underwent pilocarpine-induced status epilepticus (SE) and were then stimulated with low-frequency rTMS. An epilepsy cell model was then established by incubating rat hippocampal neurons with Mg2+-free extracellular fluids. The effects of the low-frequency rTMS on epileptogenesis and hippocampal neuron injury were evaluated using a video electroencephalogram (vEEG) and Nissl staining, and the expression of AMPAR GluA1 and STIM in the hippocampus and hippocampal neurons was assessed using western blot and immunofluorescence. Additionally, the intracellular Ca2+ concentration and reactive oxygen species (ROS) were measured using flow cytometry. Low-frequency rTMS attenuated spontaneous recurrent seizures in rats with epilepsy, with the SE group exhibiting a higher incidence (100%) and frequency (3.00 ± 0.18 times/day) than the SE + 0.3 (50% incidence, 0.06 ± 0.03 times/day), SE + 0.5 (0.20 ± 0.02 times/day) and SE + 1 Hz (1.02 ± 0.05 times/day) groups. Additionally, rTMS reduced the damage and apoptosis of hippocampal pyramidal neurons, increasing their numbers in the CA1 and CA3 regions. Furthermore, AMPAR GluA1 and STIM expression were upregulated in the hippocampus when using rTMS, reversing the downregulation caused by seizures. Immunofluorescence verified the increased fluorescence intensity of AMPAR GluA1 and STIM. Moreover, rTMS inhibited Ca2+ overload and ROS in epileptic neuron models. Low-frequency rTMS may exert neuroprotective effects through the AMPAR GluA1-STIM-Ca2+ pathway.
Collapse
Affiliation(s)
- Li-Qin Che
- Department of Neurology, The Second Hospital of Hebei Medical University, No.215 of West Heping Road, Xinhua District, Shijiazhuang, 050000, Hebei Province, China
| | - Zhen-Zhen Qu
- Department of Neurology, The Second Hospital of Hebei Medical University, No.215 of West Heping Road, Xinhua District, Shijiazhuang, 050000, Hebei Province, China
| | - Zhuo-Feng Mao
- Department of Neurology, The Second Hospital of Hebei Medical University, No.215 of West Heping Road, Xinhua District, Shijiazhuang, 050000, Hebei Province, China
| | - Qi Qiao
- Department of Neurology, The Second Hospital of Hebei Medical University, No.215 of West Heping Road, Xinhua District, Shijiazhuang, 050000, Hebei Province, China
| | - Kai-Ping Zhou
- Department of Neurology, The Second Hospital of Hebei Medical University, No.215 of West Heping Road, Xinhua District, Shijiazhuang, 050000, Hebei Province, China
| | - Li-Jing Jia
- Department of Neurology, The Second Hospital of Hebei Medical University, No.215 of West Heping Road, Xinhua District, Shijiazhuang, 050000, Hebei Province, China.
| | - Wei-Ping Wang
- Department of Neurology, The Second Hospital of Hebei Medical University, No.215 of West Heping Road, Xinhua District, Shijiazhuang, 050000, Hebei Province, China.
| |
Collapse
|
3
|
Tan XW, Gulwant Singh HK, Koh JZJ, Tan RSY, Tor PC. Personalised transcranial magnetic stimulation for treatment-resistant depression, depression with comorbid anxiety and negative symptoms of schizophrenia: a narrative review. Singapore Med J 2024; 65:544-551. [PMID: 39379030 DOI: 10.4103/singaporemedj.smj-2024-133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Accepted: 08/27/2024] [Indexed: 10/10/2024]
Abstract
ABSTRACT Transcranial magnetic stimulation (TMS) is a promising intervention for treatment-resistant psychiatric disorders. However, conventional TMS typically utilises a one-size-fits-all approach when determining stimulation targets. Recent retrospective brain circuit-based analyses using lesion network mapping have suggested that a left dorsal lateral prefrontal cortex target has a higher efficacy for alleviating depression symptoms, a dorsomedial prefrontal cortex target is more effective for anxiety symptoms, and a rostromedial prefrontal cortex target is effective for schizophrenia-associated psychiatric symptoms. Nonetheless, symptom-specific brain circuit targeting has not been tested prospectively. We conducted a narrative review of selected literature to investigate individualised targeting for TMS and discuss potential future directions to elucidate the efficacy of this approach.
Collapse
Affiliation(s)
- Xiao Wei Tan
- Department of Mood and Anxiety, Institute of Mental Health, Singapore
| | | | | | | | | |
Collapse
|
4
|
Lin J, Xing Q, Zhang C, Luo Y, Chen X, Xie Y, Wang Y. Advances in Repetitive Transcranial Magnetic Stimulation for the Treatment of Post-traumatic Stress Disorder. ALPHA PSYCHIATRY 2024; 25:440-448. [PMID: 39360295 PMCID: PMC11443297 DOI: 10.5152/alphapsychiatry.2024.241587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 04/29/2024] [Indexed: 10/04/2024]
Abstract
Post-traumatic stress disorder (PTSD) is a psychiatric disorder that develops and persists after an individual experiences a major traumatic or life-threatening event. While pharmacological treatment and psychological interventions can alleviate some symptoms, pharmacotherapy is time-consuming with low patient compliance, and psychological interventions are costly. Repetitive Transcranial Magnetic Stimulation (rTMS) is a safe and effective technique for treating PTSD, with advantages such as high compliance, low cost, and simplicity of implementation. It can even simultaneously improve depressive symptoms in some patients. Current research indicates that high-frequency rTMS shows better therapeutic effects compared to low-frequency rTMS, with no significant difference in the likelihood of adverse reactions between the two. Theta Burst Stimulation (TBS) exhibits similar efficacy to high-frequency rTMS, with shorter duration and significant improvement in depressive symptoms. However, it carries a slightly higher risk of adverse reactions compared to traditional high-frequency rTMS. Combining rTMS with psychological therapy appears to be more effective in improving PTSD symptoms, with early onset of effects and longer duration, albeit at higher cost and requiring individualized patient control. The most common adverse effect of treatment is headache, which can be improved by stopping treatment or using analgesics. Despite these encouraging data, several aspects remain unknown. Given the highly heterogeneous nature of PTSD, defining unique treatment methods for this patient population is quite challenging. There are also considerable differences between trials regarding stimulation parameters, therapeutic effects, and the role of combined psychological therapy, which future research needs to address.
Collapse
Affiliation(s)
- Jingyi Lin
- Department of Rehabilitation Medicine, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
| | - Qijia Xing
- Department of Rehabilitation Medicine, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
| | - Chunyu Zhang
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Stem Cell Immunity and Regeneration Key Laboratory of Luzhou, Luzhou, Sichuan, China
| | - Yaomin Luo
- Department of Rehabilitation Medicine, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
| | - Xin Chen
- Department of Rehabilitation Medicine, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
| | - Yulei Xie
- Department of Rehabilitation Medicine, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
- School of Rehabilitation, Capital Medical University, Beijing, China
| | - Yinxu Wang
- Department of Rehabilitation Medicine, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
| |
Collapse
|
5
|
Sun Y, Xiao Z, Chen B, Zhao Y, Dai J. Advances in Material-Assisted Electromagnetic Neural Stimulation. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2400346. [PMID: 38594598 DOI: 10.1002/adma.202400346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 03/26/2024] [Indexed: 04/11/2024]
Abstract
Bioelectricity plays a crucial role in organisms, being closely connected to neural activity and physiological processes. Disruptions in the nervous system can lead to chaotic ionic currents at the injured site, causing disturbances in the local cellular microenvironment, impairing biological pathways, and resulting in a loss of neural functions. Electromagnetic stimulation has the ability to generate internal currents, which can be utilized to counter tissue damage and aid in the restoration of movement in paralyzed limbs. By incorporating implanted materials, electromagnetic stimulation can be targeted more accurately, thereby significantly improving the effectiveness and safety of such interventions. Currently, there have been significant advancements in the development of numerous promising electromagnetic stimulation strategies with diverse materials. This review provides a comprehensive summary of the fundamental theories, neural stimulation modulating materials, material application strategies, and pre-clinical therapeutic effects associated with electromagnetic stimulation for neural repair. It offers a thorough analysis of current techniques that employ materials to enhance electromagnetic stimulation, as well as potential therapeutic strategies for future applications.
Collapse
Affiliation(s)
- Yuting Sun
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Zhifeng Xiao
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Bing Chen
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Yannan Zhao
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Jianwu Dai
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
- Tianjin Key Laboratory of Biomedical Materials, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300192, China
| |
Collapse
|
6
|
Huang H, Zhu Y, Liao L, Gao S, Tao Y, Fang X, Lian Y, Gao C. The long-term effects of intermittent theta burst stimulation on Alzheimer's disease-type pathologies in APP/PS1 mice. Brain Res Bull 2023; 202:110735. [PMID: 37586425 DOI: 10.1016/j.brainresbull.2023.110735] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 07/31/2023] [Accepted: 08/13/2023] [Indexed: 08/18/2023]
Abstract
Intermittent theta burst stimulation (iTBS), an emerging and highly efficient paradigm of repetitive transcranial magnetic stimulation (rTMS), has been demonstrated to mitigate cognitive impairment in Alzheimer's disease. Previous clinical studies have shown that the cognitive improvement of iTBS could last several weeks after treatment. Nonetheless, it is largely uncertain how the long-term effects of iTBS treatment are sustained. To investigate whether iTBS has a long-term effect on AD-type pathologies, 6-month-old APP/PS1 mice are administrated with 30 consecutive days of iTBS treatment. After a 2-month interval, morphological alterations in the brain are examined by immunohistochemistry and immunofluorescence staining, while levels of associated proteins are assessed by Western blot at the age of 9 months. We find that iTBS treatment significantly diminishes Aβ burden in the cerebral cortex and hippocampus of APP/PS1 mice. Moreover, we observe that iTBS treatment inhibits the expression of BACE1 and elevates the level of IDE, suggesting that the reduction of Aβ load could be attributed to the inhibition of Aβ production and facilitation of Aβ degradation. Furthermore, iTBS treatment attenuates neuroinflammation, neuronal apoptosis, and synaptic loss in APP/PS1 mice. Collectively, these data indicate that 1 month of iTBS treatment ameliorates pathologies in the brain of AD mice for at least 2 months. We provide the novel evidence that iTBS may exert after-effects on AD-type pathologies via inhibition of Aβ production and facilitation of Aβ degradation.
Collapse
Affiliation(s)
- Hao Huang
- Department of Rehabilitation Medicine, Daping Hospital, Third Military Medical University, Chongqing 400042, China; Department of Rehabilitation Medicine, General Hospital of Southern Theatre Command of PLA, Guangzhou 510010, China
| | - Yang Zhu
- Department of Rehabilitation Medicine, Daping Hospital, Third Military Medical University, Chongqing 400042, China
| | - Lingyi Liao
- Department of Rehabilitation Medicine, Daping Hospital, Third Military Medical University, Chongqing 400042, China
| | - Shihao Gao
- Department of Rehabilitation Medicine, Daping Hospital, Third Military Medical University, Chongqing 400042, China
| | - Yong Tao
- Department of Rehabilitation Medicine, Daping Hospital, Third Military Medical University, Chongqing 400042, China
| | - Xiangqin Fang
- Department of Rehabilitation Medicine, Daping Hospital, Third Military Medical University, Chongqing 400042, China
| | - Yan Lian
- Department of Preventive Medicine, Daping Hospital, Third Military Medical University, Chongqing 400042, China.
| | - Changyue Gao
- Department of Rehabilitation Medicine, Daping Hospital, Third Military Medical University, Chongqing 400042, China.
| |
Collapse
|
7
|
Meneses-San Juan D, Lamas M, Ramírez-Rodríguez GB. Repetitive Transcranial Magnetic Stimulation Reduces Depressive-like Behaviors, Modifies Dendritic Plasticity, and Generates Global Epigenetic Changes in the Frontal Cortex and Hippocampus in a Rodent Model of Chronic Stress. Cells 2023; 12:2062. [PMID: 37626872 PMCID: PMC10453847 DOI: 10.3390/cells12162062] [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: 06/01/2023] [Revised: 07/19/2023] [Accepted: 07/24/2023] [Indexed: 08/27/2023] Open
Abstract
Depression is the most common affective disorder worldwide, accounting for 4.4% of the global population, a figure that could increase in the coming decades. In depression, there exists a reduction in the availability of dendritic spines in the frontal cortex (FC) and hippocampus (Hp). In addition, histone modification and DNA methylation are also dysregulated epigenetic mechanisms in depression. Repetitive transcranial magnetic stimulation (rTMS) is a technique that is used to treat depression. However, the epigenetic mechanisms of its therapeutic effect are still not known. Therefore, in this study, we evaluated the antidepressant effect of 5 Hz rTMS and examined its effect on dendritic remodeling, immunoreactivity of synapse proteins, histone modification, and DNA methylation in the FC and Hp in a model of chronic mild stress. Our data indicated that stress generated depressive-like behaviors and that rTMS reverses this effect, romotes the formation of dendritic spines, and favors the presynaptic connection in the FC and DG (dentate gyrus), in addition to increasing histone H3 trimethylation and DNA methylation. These results suggest that the antidepressant effect of rTMS is associated with dendritic remodeling, which is probably regulated by epigenetic mechanisms. These data are a first approximation of the impact of rTMS at the epigenetic level in the context of depression. Therefore, it is necessary to analyze in future studies as to which genes are regulated by these mechanisms, and how they are associated with the neuroplastic modifications promoted by rTMS.
Collapse
Affiliation(s)
- David Meneses-San Juan
- National Institute of Psychiatry “Ramón de la Fuente Muñiz”, Mexico City 14370, Mexico;
- Center of Research and Advanced Studies of the National Polytechnic Institute, Mexico City 07360, Mexico;
| | - Mónica Lamas
- Center of Research and Advanced Studies of the National Polytechnic Institute, Mexico City 07360, Mexico;
| | | |
Collapse
|
8
|
Zhou L, Jin Y, Wu D, Cun Y, Zhang C, Peng Y, Chen N, Yang X, Zhang S, Ning R, Kuang P, Wang Z, Zhang P. Current evidence, clinical applications, and future directions of transcranial magnetic stimulation as a treatment for ischemic stroke. Front Neurosci 2023; 17:1177283. [PMID: 37534033 PMCID: PMC10390744 DOI: 10.3389/fnins.2023.1177283] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Accepted: 06/28/2023] [Indexed: 08/04/2023] Open
Abstract
Transcranial magnetic stimulation (TMS) is a non-invasive brain neurostimulation technique that can be used as one of the adjunctive treatment techniques for neurological recovery after stroke. Animal studies have shown that TMS treatment of rats with middle cerebral artery occlusion (MCAO) model reduced cerebral infarct volume and improved neurological dysfunction in model rats. In addition, clinical case reports have also shown that TMS treatment has positive neuroprotective effects in stroke patients, improving a variety of post-stroke neurological deficits such as motor function, swallowing, cognitive function, speech function, central post-stroke pain, spasticity, and other post-stroke sequelae. However, even though numerous studies have shown a neuroprotective effect of TMS in stroke patients, its possible neuroprotective mechanism is not clear. Therefore, in this review, we describe the potential mechanisms of TMS to improve neurological function in terms of neurogenesis, angiogenesis, anti-inflammation, antioxidant, and anti-apoptosis, and provide insight into the current clinical application of TMS in multiple neurological dysfunctions in stroke. Finally, some of the current challenges faced by TMS are summarized and some suggestions for its future research directions are made.
Collapse
Affiliation(s)
- Li Zhou
- Key Laboratory of Acupuncture and Massage for Treatment of Encephalopathy, College of Acupuncture, Tuina and Rehabilitation, Yunnan University of Traditional Chinese Medicine, Kunming, China
| | - Yaju Jin
- Key Laboratory of Acupuncture and Massage for Treatment of Encephalopathy, College of Acupuncture, Tuina and Rehabilitation, Yunnan University of Traditional Chinese Medicine, Kunming, China
| | - Danli Wu
- Key Laboratory of Acupuncture and Massage for Treatment of Encephalopathy, College of Acupuncture, Tuina and Rehabilitation, Yunnan University of Traditional Chinese Medicine, Kunming, China
| | - Yongdan Cun
- Key Laboratory of Acupuncture and Massage for Treatment of Encephalopathy, College of Acupuncture, Tuina and Rehabilitation, Yunnan University of Traditional Chinese Medicine, Kunming, China
| | - Chengcai Zhang
- Key Laboratory of Acupuncture and Massage for Treatment of Encephalopathy, College of Acupuncture, Tuina and Rehabilitation, Yunnan University of Traditional Chinese Medicine, Kunming, China
| | - Yicheng Peng
- Key Laboratory of Acupuncture and Massage for Treatment of Encephalopathy, College of Acupuncture, Tuina and Rehabilitation, Yunnan University of Traditional Chinese Medicine, Kunming, China
| | - Na Chen
- Key Laboratory of Acupuncture and Massage for Treatment of Encephalopathy, College of Acupuncture, Tuina and Rehabilitation, Yunnan University of Traditional Chinese Medicine, Kunming, China
| | - Xichen Yang
- Key Laboratory of Acupuncture and Massage for Treatment of Encephalopathy, College of Acupuncture, Tuina and Rehabilitation, Yunnan University of Traditional Chinese Medicine, Kunming, China
| | - Simei Zhang
- Key Laboratory of Acupuncture and Massage for Treatment of Encephalopathy, College of Acupuncture, Tuina and Rehabilitation, Yunnan University of Traditional Chinese Medicine, Kunming, China
| | - Rong Ning
- Key Laboratory of Acupuncture and Massage for Treatment of Encephalopathy, College of Acupuncture, Tuina and Rehabilitation, Yunnan University of Traditional Chinese Medicine, Kunming, China
| | - Peng Kuang
- Key Laboratory of Acupuncture and Massage for Treatment of Encephalopathy, College of Acupuncture, Tuina and Rehabilitation, Yunnan University of Traditional Chinese Medicine, Kunming, China
| | - Zuhong Wang
- Kunming Municipal Hospital of Traditional Chinese Medicine, Kunming, Yunnan, China
| | - Pengyue Zhang
- Key Laboratory of Acupuncture and Massage for Treatment of Encephalopathy, College of Acupuncture, Tuina and Rehabilitation, Yunnan University of Traditional Chinese Medicine, Kunming, China
| |
Collapse
|
9
|
Ramírez-Rodríguez GB, Meneses San-Juan D, Rico-Becerra AI, González-Olvera JJ, Reyes-Galindo V. Repetitive transcranial magnetic stimulation and fluoxetine reverse depressive-like behavior but with differential effects on Olig2-positive cells in chronically stressed mice. Neuropharmacology 2023; 236:109567. [PMID: 37209812 DOI: 10.1016/j.neuropharm.2023.109567] [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: 12/21/2022] [Revised: 04/26/2023] [Accepted: 04/29/2023] [Indexed: 05/22/2023]
Abstract
Depression is a mood disorder coursing with several behavioral, cellular, and neurochemical alterations. The negative impact of chronic stress may precipitate this neuropsychiatric disorder. Interestingly, downregulation of oligodendrocyte-related genes, abnormal myelin structure, and reduced numbers and density of oligodendrocytes in the limbic system have been identified in patients diagnosed with depression, but also in rodents exposed to chronic mild stress (CMS). Several reports have emphasized the importance of pharmacological or stimulation-related strategies in influencing oligodendrocytes in the hippocampal neurogenic niche. Repetitive transcranial magnetic stimulation (rTMS) has gained attention as an intervention to revert depression. Here, we hypothesized that 5 Hz (Hz) of rTMS or Fluoxetine (Flx) would revert depressive-like behaviors by influencing oligodendrocytes and revert neurogenic alterations caused by CMS in female Swiss Webster mice. Our results showed that 5 Hz rTMS or Flx revert depressive-like behavior. Only rTMS influenced oligodendrocytes by increasing the number of Olig2-positive cells in the hilus of the dentate gyrus and the prefrontal cortex. However, both strategies exerted effects on some events of the hippocampal neurogenic processes, such as cell proliferation (Ki67-positive cells), survival (CldU-positive cells), and intermediate stages (doublecortin-positive cells) along the dorsal-ventral axis of this region. Interestingly, the combination of rTMS-Flx exerted antidepressant-like effects, but the increased number of Olig2-positive cells observed in mice treated only with rTMS was canceled. However, rTMS-Flx exerted a synergistic effect by increasing the number of Ki67-positive cells. It also increased the number of CldU- and doublecortin-positive cells in the dentate gyrus. Our results demonstrate that 5 Hz rTMS has beneficial effects, as it reverted depressive-like behavior by increasing the number of Olig2-positive cells and reverting the decrement in hippocampal neurogenesis in CMS-exposed mice. Nevertheless, the effects of rTMS on other glial cells require further investigation.
Collapse
Affiliation(s)
- Gerardo Bernabé Ramírez-Rodríguez
- Laboratorio de Neurogénesis, Subdirección de Investigaciones Clínicas, Instituto Nacional de Psiquiatría "Ramón de la Fuente Muñiz", Calzada México-Xochimilco 101, Alcaldía Tlalpan, C.P, 14370, Ciudad de México, Mexico.
| | - David Meneses San-Juan
- Laboratorio de Neurogénesis, Subdirección de Investigaciones Clínicas, Instituto Nacional de Psiquiatría "Ramón de la Fuente Muñiz", Calzada México-Xochimilco 101, Alcaldía Tlalpan, C.P, 14370, Ciudad de México, Mexico
| | - Allan Irasek Rico-Becerra
- Laboratorio de Neurogénesis, Subdirección de Investigaciones Clínicas, Instituto Nacional de Psiquiatría "Ramón de la Fuente Muñiz", Calzada México-Xochimilco 101, Alcaldía Tlalpan, C.P, 14370, Ciudad de México, Mexico; Licenciatura en Neurociencias, Facultad de Medicina. Universidad Nacional Autónoma de México. Circuito Interior, Avenida Universidad 3000, Ciudad Universitaria, Alcaldía Coyoacán, C.P, 04510, Ciudad de México, Mexico
| | - Jorge Julio González-Olvera
- Subdirección de Investigaciones Clínicas, Instituto Nacional de Psiquiatría "Ramón de la Fuente Muñiz", Calzada México-Xochimilco 101. Alcaldía Tlalpan, C.P, 14370, Ciudad de México, Mexico
| | - Verónica Reyes-Galindo
- Instituto de Ecología. Universidad Nacional Autónoma de México. Circuito Interior, Avenida Universidad 3000, Ciudad Universitaria. Alcaldía Coyoacán, C.P, 04510, Ciudad de México, Mexico
| |
Collapse
|
10
|
Genetic profile for dopamine signaling predicts brain functional reactivity to repetitive transcranial magnetic stimulation. Eur Arch Psychiatry Clin Neurosci 2023; 273:99-111. [PMID: 35951113 DOI: 10.1007/s00406-022-01436-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Accepted: 05/15/2022] [Indexed: 11/03/2022]
Abstract
Research integrating molecular and imaging data provides important insights into how the genetic profile associated with dopamine signaling influences inter-individual differences in brain functions. However, the effects of genetic variations in dopamine signaling on the heterogeneity of brain changes induced by repetitive transcranial magnetic stimulation (rTMS) still remain unclear. The current study examined the composite effects of genetic variations in dopamine-related genes on rTMS-induced brain responses in terms of the functional network connectivity and working memory performance. Healthy individuals (n = 30) participated in a randomized, double-blind, sham-controlled study with a crossover design of five consecutive days where active rTMS or sham stimulation sessions were administered over the left dorsolateral prefrontal cortex (DLPFC) of the brain. Participants were mostly women (n = 29) and genotyped for polymorphisms in the catechol-O-methyltransferase and D2 dopamine receptor genes and categorized according to their genetic composite scores: high vs. low dopamine signaling groups. Pre- and post-intervention data of resting-state functional magnetic resonance imaging and working memory performance were obtained from 27 individuals with active rTMS and 30 with sham stimulation sessions. The mean functional connectivity within the resting-state networks centered on the DLPFC increased in the high dopamine signaling group. Working memory performance also improved with rTMS in the high dopamine signaling group compared to that in the low dopamine signaling group. The present results suggest that genetic predisposition to higher dopamine signaling may be a promising neurobiological predictor for rTMS effects on cognitive enhancement.Trial registration: ClinicalTrials.gov (NCT02932085).
Collapse
|
11
|
Dinakaran D, Sreeraj VS, Venkatasubramanian G. Dengue and Psychiatry: Manifestations, Mechanisms, and Management Options. Indian J Psychol Med 2022; 44:429-435. [PMID: 36157026 PMCID: PMC9460008 DOI: 10.1177/02537176211022571] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Dengue is an arboviral infection endemic in tropical countries. Neurological sequelae to dengue infection are not uncommon, and psychiatric manifestations are increasingly reported. This narrative review aims to present the varied manifestations, postulated mechanisms, and the available treatment options for psychiatric morbidity associated with dengue. The evidence available from eight observational studies is summarized in this review. Depression and anxiety are noted to be prevalent during both the acute and convalescent stages of the infection. The presence of encephalopathy and other neurological conditions is not a prerequisite for developing psychiatric disorders. However, treatment options to manage such psychiatric manifestations were not specified in the observational studies. Anecdotal evidence from case reports is outlined. Special attention is paid to the role of epigenetic modifications following dengue infections and the role of histone deacetylase inhibitors in the management. DNA methylation inhibitors such as valproic acid play a significant role in reversing stress-, viral-, or drug-induced epigenetic modifications.
Collapse
Affiliation(s)
- Damodharan Dinakaran
- Dept. of Psychiatry, National Institute of Mental Health And Neurosciences (NIMHANS), Bengaluru, Karnataka, India
| | - Vanteemar S Sreeraj
- Dept. of Psychiatry, National Institute of Mental Health And Neurosciences (NIMHANS), Bengaluru, Karnataka, India
| | - Ganesan Venkatasubramanian
- Dept. of Psychiatry, National Institute of Mental Health And Neurosciences (NIMHANS), Bengaluru, Karnataka, India
| |
Collapse
|
12
|
Fecteau S. Influencing Human Behavior with Noninvasive Brain Stimulation: Direct Human Brain Manipulation Revisited. Neuroscientist 2022; 29:317-331. [PMID: 35057668 PMCID: PMC10159214 DOI: 10.1177/10738584211067744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The use of tools to perturb brain activity can generate important insights into brain physiology and offer valuable therapeutic approaches for brain disorders. Furthermore, the potential of such tools to enhance normal behavior has become increasingly recognized, and this has led to the development of various noninvasive technologies that provides a broader access to the human brain. While providing a brief survey of brain manipulation procedures used in the past decades, this review aims at stimulating an informed discussion on the use of these new technologies to investigate the human. It highlights the importance to revisit the past use of this unique armamentarium and proceed to a detailed analysis of its present state, especially in regard to human behavioral regulation.
Collapse
|
13
|
Heath AM, Brewer M, Yesavage J, McNerney MW. Improved object recognition memory using post-encoding repetitive transcranial magnetic stimulation. Brain Stimul 2022; 15:78-86. [PMID: 34785386 PMCID: PMC10612530 DOI: 10.1016/j.brs.2021.11.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 11/01/2021] [Accepted: 11/09/2021] [Indexed: 01/02/2023] Open
Abstract
BACKGROUND Brain stimulation is known to affect canonical pathways and proteins involved in memory. However, there are conflicting results on the ability of brain stimulation to improve to memory, which may be due to variations in timing of stimulation. HYPOTHESIS We hypothesized that repetitive transcranial magnetic stimulation (rTMS) given following a learning task and within the time period before retrieval could help improve memory. METHODS We implanted male B6129SF2/J mice (n = 32) with a cranial attachment to secure the rTMS coil so that the mice could be given consistent stimulation to the frontal area whilst freely moving. Mice then underwent the object recognition test sampling phase and given treatment +3, +24, +48 h following the test. Treatment consisted of 10 min 10 Hz rTMS stimulation (TMS, n = 10), sham treatment (SHAM, n = 11) or a control group which did not do the behavior test or receive rTMS (CONTROL n = 11). At +72 h mice were tested for their exploration of the novel vs familiar object. RESULTS At 72-h's, only the mice which received rTMS had greater exploration of the novel object than the familiar object. We further show that promoting synaptic GluR2 and maintaining synaptic connections in the perirhinal cortex and hippocampal CA1 are important for this effect. In addition, we found evidence that these changes were linked to CAMKII and CREB pathways in hippocampal neurons. CONCLUSION By linking the known biological effects of rTMS to memory pathways we provide evidence that rTMS is effective in improving memory when given during the consolidation and maintenance phases.
Collapse
Affiliation(s)
- A M Heath
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA, 94305, USA; Department of Veterans Affairs, Sierra-Pacific Mental Illness Research Educational and Clinical Center, Palo Alto, CA, 94304, USA.
| | - M Brewer
- Stanford University, Stanford, CA, 94305, USA
| | - J Yesavage
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA, 94305, USA; Department of Veterans Affairs, Sierra-Pacific Mental Illness Research Educational and Clinical Center, Palo Alto, CA, 94304, USA
| | - M W McNerney
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA, 94305, USA; Department of Veterans Affairs, Sierra-Pacific Mental Illness Research Educational and Clinical Center, Palo Alto, CA, 94304, USA
| |
Collapse
|
14
|
Hou Y, Zhao J, Yang D, Xuan R, Xie R, Wang M, Mo H, Liang L, Wang W, Wu S, Wang Y, Liu X. LF-rTMS ameliorates social dysfunction of FMR1 -/- mice via modulating Akt/GSK-3β signaling. Biochem Biophys Res Commun 2021; 550:22-29. [PMID: 33677132 DOI: 10.1016/j.bbrc.2021.02.086] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Accepted: 02/19/2021] [Indexed: 12/29/2022]
Abstract
Autism spectrum disorders (ASD) are a group of neurological disorders which affect approximately 1% of children around the world. Social dysfunction is one of the two core syndromes of ASD, and still lacks effective treatment. Transcranial magnetic stimulation (TMS) is a noninvasive and safe procedure that uses magnetic fields to modulate neural activity. Whether it were effective in modulating social function remains unclear. By using 3-chamber test, ultrasonic vocalization recording and Western-blotting, we demonstrated that FMR1 (fragile X mental retardation protein) mutant mice, a model of ASD, exhibited obvious defects in social preference and ultrasonic communication. In addition, we detected increase of p-Akt (S473) and p-GSK-3β (S9), and decrease of p-PSD-95 (T19) in the anterior cingulate cortex (ACC) of FMR1-/- mice. Treating FMR1-/- mice with 1 Hz repetitive TMS (rTMS) exerted a long lasting effect in improving both the ultrasonic communication and social preference, as well as restoring the levels of Akt/GSK-3β activity and spine density in the FMR1-/-ACC. Our data, for the first time, demonstrated a beneficial effect of low frequency rTMS (LF-rTMS) on the social function of FMR1-/- mice and an involvement of Akt/GSK-3β signaling in this process, indicating LF-rTMS as a potential therapeutic strategy for ASD patients.
Collapse
Affiliation(s)
- Yilin Hou
- Department of Military Psychology, Fourth Military Medical University, Xi'an, Shaanxi, 710032, PR China
| | - Jiqian Zhao
- Department of Neurobiology and Institute of Neurosciences, School of Basic Medicine, Fourth Military Medical University, Xi'an, Shaanxi, 710032, PR China
| | - Dingding Yang
- Department of Neurobiology and Institute of Neurosciences, School of Basic Medicine, Fourth Military Medical University, Xi'an, Shaanxi, 710032, PR China
| | - Runkang Xuan
- Department of Neurobiology and Institute of Neurosciences, School of Basic Medicine, Fourth Military Medical University, Xi'an, Shaanxi, 710032, PR China
| | - Rougang Xie
- Department of Neurobiology and Institute of Neurosciences, School of Basic Medicine, Fourth Military Medical University, Xi'an, Shaanxi, 710032, PR China
| | - Mengmeng Wang
- Department of Neurobiology and Institute of Neurosciences, School of Basic Medicine, Fourth Military Medical University, Xi'an, Shaanxi, 710032, PR China
| | - Huiming Mo
- Department of Physiology, Medicine College of Yanan University, Yan'an, 716000, PR China
| | - Lirong Liang
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Engineering Research, Center for Dental Materials and Advanced Manufacture, Department of Anethesiology, School of Stomatology, Fourth Military Medical University, Xi'an, Shaanxi, 710032, PR China
| | - Wenting Wang
- Department of Neurobiology and Institute of Neurosciences, School of Basic Medicine, Fourth Military Medical University, Xi'an, Shaanxi, 710032, PR China
| | - Shengxi Wu
- Department of Neurobiology and Institute of Neurosciences, School of Basic Medicine, Fourth Military Medical University, Xi'an, Shaanxi, 710032, PR China
| | - Yazhou Wang
- Department of Neurobiology and Institute of Neurosciences, School of Basic Medicine, Fourth Military Medical University, Xi'an, Shaanxi, 710032, PR China.
| | - Xufeng Liu
- Department of Military Psychology, Fourth Military Medical University, Xi'an, Shaanxi, 710032, PR China.
| |
Collapse
|
15
|
Choung JS, Kim JM, Ko MH, Cho DS, Kim M. Therapeutic efficacy of repetitive transcranial magnetic stimulation in an animal model of Alzheimer's disease. Sci Rep 2021; 11:437. [PMID: 33432077 PMCID: PMC7801521 DOI: 10.1038/s41598-020-80147-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Accepted: 12/14/2020] [Indexed: 12/02/2022] Open
Abstract
Previous studies on repetitive transcranial magnetic stimulation (rTMS) suggested potential neurorestorative properties in Alzheimer's disease (AD). This study aimed to investigate therapeutic effects of rTMS on an AD mouse model at high and low frequencies. The subject mice were allocated into the AD model group (AD induced by intracerebroventricular amyloid beta 42 oligomer [Aβ42] injection) and the saline-injected control group. Each group was subdivided according to rTMS treatment: high frequency (20 Hz), low frequency (1 Hz), and not rTMS-treated. Behavioural assessments with Y-maze test and novel object recognition task were performed; the results indicated cognition recovery by both the frequencies of rTMS after treatment in the AD model (Ps < 0.01). Tendency of further effects by high frequency compared to low frequency rTMS was also shown in Y-maze test. Neurotransmitter assay showed increment in dopamine concentration and upregulation of dopamine-receptor 4 (DR4) by rTMS in AD mice with higher response by high frequency stimulation (Ps < 0.05). Only high-frequency rTMS induced an elevation of brain-derived neurotrophic factor (BDNF) levels and enhanced the expression of Nestin and NeuN in the brain tissue (Ps < 0.05). Under in vitro conditions, Aβ42 incubated mouse hippocampal cell showed an increase in dopamine levels and BDNF by application of high-frequency rTMS treatment. In conclusion, rTMS might have a potential therapeutic effect on AD, and it seems to be related with dopaminergic activation. High frequency of stimulation seems to induce higher efficacy than that induced by low frequency, with elevated expressions of DR4 gene and neurogenic proteins.
Collapse
Affiliation(s)
- Jin Seung Choung
- Rehabilitation and Regeneration Research Center, CHA University, Seongnam, Republic of Korea
| | - Jong Moon Kim
- Rehabilitation and Regeneration Research Center, CHA University, Seongnam, Republic of Korea
- Department of Rehabilitation Medicine, CHA Bundang Medical Center, CHA University, 59 Yatap-ro, Bundang-gu, Seongnam, Gyeonggi-do, 13496, Republic of Korea
| | - Myoung-Hwan Ko
- Department of Physical Medicine and Rehabilitation, Jeonbuk National University Medical School, Jeonju, Republic of Korea
| | - Dong Sik Cho
- R&D Center, Remed Co., Ltd., Seongnam, Republic of Korea
| | - MinYoung Kim
- Rehabilitation and Regeneration Research Center, CHA University, Seongnam, Republic of Korea.
- Department of Rehabilitation Medicine, CHA Bundang Medical Center, CHA University, 59 Yatap-ro, Bundang-gu, Seongnam, Gyeonggi-do, 13496, Republic of Korea.
| |
Collapse
|
16
|
Clarke D, Beros J, Bates KA, Harvey AR, Tang AD, Rodger J. Low intensity repetitive magnetic stimulation reduces expression of genes related to inflammation and calcium signalling in cultured mouse cortical astrocytes. Brain Stimul 2020; 14:183-191. [PMID: 33359601 DOI: 10.1016/j.brs.2020.12.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 12/17/2020] [Accepted: 12/18/2020] [Indexed: 12/26/2022] Open
Abstract
Repetitive transcranial magnetic stimulation (rTMS) is a form of non-invasive brain stimulation frequently used to induce neuroplasticity in the brain. Even at low intensities, rTMS has been shown to modulate aspects of neuronal plasticity such as motor learning and structural reorganisation of neural tissue. However, the impact of low intensity rTMS on glial cells such as astrocytes remains largely unknown. This study investigated changes in RNA (qPCR array: 125 selected genes) and protein levels (immunofluorescence) in cultured mouse astrocytes following a single session of low intensity repetitive magnetic stimulation (LI-rMS - 18 mT). Purified neonatal cortical astrocyte cultures were stimulated with either 1Hz (600 pulses), 10Hz (600 or 6000 pulses) or sham (0 pulses) LI-rMS, followed by RNA extraction at 5 h post-stimulation, or fixation at either 5 or 24-h post-stimulation. LI-rMS resulted in a two-to-four-fold downregulation of mRNA transcripts related to calcium signalling (Stim1 and Orai3), inflammatory molecules (Icam1) and neural plasticity (Ncam1). 10Hz reduced expression of Stim1, Orai3, Kcnmb4, and Ncam1 mRNA, whereas 1Hz reduced expression of Icam1 mRNA and signalling-related genes. Protein levels followed a similar pattern for 10Hz rMS, with a significant reduction of STIM1, ORAI3, KCNMB4, and NCAM1 protein compared to sham, but 1Hz increased STIM1 and ORAI3 protein levels relative to sham. These findings demonstrate the ability of 1Hz and 10Hz LI-rMS to modulate specific aspects of astrocytic phenotype, potentially contributing to the known effects of low intensity rTMS on excitability and neuroplasticity.
Collapse
Affiliation(s)
- Darren Clarke
- Experimental and Regenerative Neuroscience, School of Biological Sciences, The University of Western Australia, Nedlands, WA, 6009, Australia; Perron Institute for Neurological and Translational Science, Nedlands, WA, 6009, Australia.
| | - Jamie Beros
- Experimental and Regenerative Neuroscience, School of Biological Sciences, The University of Western Australia, Nedlands, WA, 6009, Australia; Perron Institute for Neurological and Translational Science, Nedlands, WA, 6009, Australia
| | - Kristyn A Bates
- Experimental and Regenerative Neuroscience, School of Biological Sciences, The University of Western Australia, Nedlands, WA, 6009, Australia
| | - Alan R Harvey
- Perron Institute for Neurological and Translational Science, Nedlands, WA, 6009, Australia; School of Human Sciences, The University of Western Australia, Nedlands, WA, 6009, Australia
| | - Alexander D Tang
- Experimental and Regenerative Neuroscience, School of Biological Sciences, The University of Western Australia, Nedlands, WA, 6009, Australia; Perron Institute for Neurological and Translational Science, Nedlands, WA, 6009, Australia
| | - Jennifer Rodger
- Experimental and Regenerative Neuroscience, School of Biological Sciences, The University of Western Australia, Nedlands, WA, 6009, Australia; Perron Institute for Neurological and Translational Science, Nedlands, WA, 6009, Australia
| |
Collapse
|
17
|
Moretti J, Poh EZ, Bolland SJ, Harvey AR, Albrecht MA, Rodger J. Concurrent LI-rTMS induces changes in c-Fos expression but not behavior during a progressive ratio task with adult ephrin-A2A5 -/- mice. Behav Brain Res 2020; 400:113011. [PMID: 33181182 DOI: 10.1016/j.bbr.2020.113011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 10/04/2020] [Accepted: 11/05/2020] [Indexed: 12/28/2022]
Abstract
Changes within the dopaminergic system induced by repetitive transcranial magnetic stimulation (rTMS) may contribute to its therapeutic effects; however, dopamine-related behavioral effects of rTMS have not been widely investigated. We recently showed that ephrin-A2A5-/- mice completed significantly fewer trials in a visual task than wildtype mice, and that concurrent low-intensity (LI-) rTMS during the task could partially rescue the abnormal behavior [Poh et al. 2018, eNeuro, vol. 5]. Here, we investigated whether the behavioral differences in ephrin-A2A5-/- mice are due to abnormal motivation, primarily a dopamine-modulated behavior, and whether LI-rTMS would increase motivation. Ephrin-A2A5-/- and wildtype mice underwent 14 daily sessions of progressive ratio (PR) tasks and received either sham or LI-rTMS during the first 10 min. Ephrin-A2A5-/- mice responded more than wildtype comparisons, and LI-rTMS did not influence task performance for either strain. Therefore concurrent stimulation does not influence motivation in a PR task. However, ephrin-A2A5-/- mice did have abnormal performance in the PR tasks after a change in the PR schedule which suggests perseverative behavior. We stained for c-Fos in the prelimbic area (PrL), ventral tegmental area and nucleus accumbens (NAc) core and shell to examine neuronal activity from the final PR session. Sham ephrin-A2A5-/- mice had lower c-Fos expression in the PrL and NAc vs. wildtype mice. Ephrin-A2A5-/- mice that received LI-rTMS showed c-Fos expression closer to wildtype levels in the NAc. Combined with high PR performance, ephrin-A2A5-/- mice show an abnormal shift to habitual responding and LI-rTMS may attenuate this shift.
Collapse
Affiliation(s)
- Jessica Moretti
- School of Biological Sciences, The University of Western Australia, Perth, WA, Australia; School of Human Sciences, The University of Western Australia, Perth, WA, Australia; Perron Institute for Neurological and Translational Science, Perth, WA, Australia
| | - Eugenia Z Poh
- School of Biological Sciences, The University of Western Australia, Perth, WA, Australia; School of Human Sciences, The University of Western Australia, Perth, WA, Australia; Perron Institute for Neurological and Translational Science, Perth, WA, Australia
| | - Samuel J Bolland
- School of Biological Sciences, The University of Western Australia, Perth, WA, Australia; Perron Institute for Neurological and Translational Science, Perth, WA, Australia
| | - Alan R Harvey
- School of Human Sciences, The University of Western Australia, Perth, WA, Australia; Perron Institute for Neurological and Translational Science, Perth, WA, Australia
| | | | - Jennifer Rodger
- School of Biological Sciences, The University of Western Australia, Perth, WA, Australia; Perron Institute for Neurological and Translational Science, Perth, WA, Australia.
| |
Collapse
|
18
|
Facchin L, Schöne C, Mensen A, Bandarabadi M, Pilotto F, Saxena S, Libourel PA, Bassetti CLA, Adamantidis AR. Slow Waves Promote Sleep-Dependent Plasticity and Functional Recovery after Stroke. J Neurosci 2020; 40:8637-8651. [PMID: 33087472 PMCID: PMC7643301 DOI: 10.1523/jneurosci.0373-20.2020] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 09/15/2020] [Accepted: 09/24/2020] [Indexed: 01/13/2023] Open
Abstract
Functional recovery after stroke is associated with a remapping of neural circuits. This reorganization is often associated with low-frequency, high-amplitude oscillations in the peri-infarct zone in both rodents and humans. These oscillations are reminiscent of sleep slow waves (SW) and suggestive of a role for sleep in brain plasticity that occur during stroke recovery; however, direct evidence is missing. Using a stroke model in male mice, we showed that stroke was followed by a transient increase in NREM sleep accompanied by reduced amplitude and slope of ipsilateral NREM sleep SW. We next used 5 ms optical activation of Channelrhodopsin 2-expressing pyramidal neurons, or 200 ms silencing of Archeorhodopsin T-expressing pyramidal neurons, to generate local cortical UP, or DOWN, states, respectively, both sharing similarities with spontaneous NREM SW in freely moving mice. Importantly, we found that single optogenetically evoked SW (SWopto) in the peri-infarct zone, randomly distributed during sleep, significantly improved fine motor movements of the limb corresponding to the sensorimotor stroke lesion site compared with spontaneous recovery and control conditions, while motor strength remained unchanged. In contrast, SWopto during wakefulness had no effect. Furthermore, chronic SWopto during sleep were associated with local axonal sprouting as revealed by the increase of anatomic presynaptic and postsynaptic markers in the peri-infarct zone and corresponding contralesional areas to cortical circuit reorganization during stroke recovery. These results support a role for sleep SW in cortical circuit plasticity and sensorimotor recovery after stroke and provide a clinically relevant framework for rehabilitation strategies using neuromodulation during sleep.SIGNIFICANCE STATEMENT Brain stroke is one of the leading causes of death and major disabilities in the elderly worldwide. A better understanding of the pathophysiological mechanisms underlying spontaneous brain plasticity after stroke, together with an optimization of rehabilitative strategies, are essential to improve stroke treatments. Here, we investigate the role of optogenetically induced sleep slow waves in an animal model of ischemic stroke and identify sleep as a window for poststroke intervention that promotes neuroplasticity and facilitates sensorimotor recovery.
Collapse
Affiliation(s)
- Laura Facchin
- Centre for Experimental Neurology, Department of Neurology, Inselspital University Hospital, University of Bern, 3010, Bern, Switzerland
| | - Cornelia Schöne
- Centre for Experimental Neurology, Department of Neurology, Inselspital University Hospital, University of Bern, 3010, Bern, Switzerland
| | - Armand Mensen
- Department of Neurology, Inselspital University Hospital, University of Bern, Bern, 3010, Switzerland
| | - Mojtaba Bandarabadi
- Centre for Experimental Neurology, Department of Neurology, Inselspital University Hospital, University of Bern, 3010, Bern, Switzerland
| | - Federica Pilotto
- Centre for Experimental Neurology, Department of Neurology, Inselspital University Hospital, University of Bern, 3010, Bern, Switzerland
- Department for BioMedical Research, University of Bern, Bern, 3010, Switzerland
| | - Smita Saxena
- Centre for Experimental Neurology, Department of Neurology, Inselspital University Hospital, University of Bern, 3010, Bern, Switzerland
- Department for BioMedical Research, University of Bern, Bern, 3010, Switzerland
| | - Paul Antoine Libourel
- Centre de Recherche en Neurosciences de Lyon, University of Lyon, Bron, 69500, France
| | - Claudio L A Bassetti
- Centre for Experimental Neurology, Department of Neurology, Inselspital University Hospital, University of Bern, 3010, Bern, Switzerland
- Department of Neurology, Inselspital University Hospital, University of Bern, Bern, 3010, Switzerland
| | - Antoine R Adamantidis
- Centre for Experimental Neurology, Department of Neurology, Inselspital University Hospital, University of Bern, 3010, Bern, Switzerland
- Department of Neurology, Inselspital University Hospital, University of Bern, Bern, 3010, Switzerland
- Department for BioMedical Research, University of Bern, Bern, 3010, Switzerland
| |
Collapse
|
19
|
Frey J, Najib U, Lilly C, Adcock A. Novel TMS for Stroke and Depression (NoTSAD): Accelerated Repetitive Transcranial Magnetic Stimulation as a Safe and Effective Treatment for Post-stroke Depression. Front Neurol 2020; 11:788. [PMID: 32849235 PMCID: PMC7431489 DOI: 10.3389/fneur.2020.00788] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Accepted: 06/25/2020] [Indexed: 11/13/2022] Open
Abstract
Background: Post-stroke depression (PSD) affects up to 50% of stroke survivors, reducing quality of life, and increasing adverse outcomes. Conventional therapies to treat PSD may not be effective for some patients. Repetitive transcranial magnetic stimulation (rTMS) is well-established as an effective treatment for Major Depressive Disorder (MDD) and some small trials have shown that rTMS may be effective for chronic PSD; however, no trials have evaluated an accelerated rTMS protocol in a subacute stroke population. We hypothesized that an accelerated rTMS protocol will be a safe and viable option to treat PSD symptoms. Methods: Patients (N = 6) with radiographic evidence of ischemic stroke within the last 2 weeks to 6 months with Hamilton Depression Rating Scale (HAMD-17) scores >7 were recruited for an open label study using an accelerated rTMS protocol as follows: High-frequency (20-Hz) rTMS at 110% resting motor threshold (RMT) was applied to the left dorsolateral prefrontal cortex (DLPFC) during five sessions per day over four consecutive days for a total of 20 sessions. Safety assessment and adverse events were documented based on the patients' responses following each day of stimulation. Before and after the 4-days neurostimulation protocol, outcome measures were obtained for the HAMD, modified Rankin Scale (mRS), functional independence measures (FIM), and National Institutes of Health Stroke Scales (NIHSS). These same measures were obtained at 3-months follow up. Results: HAMD significantly decreased (Wilcoxon p = 0.03) from M = 15.5 (2.81)-4.17 (0.98) following rTMS, a difference which persisted at the 3-months follow-up (p = 0.03). No statistically significant difference in FIM, mRS, or NIHSS were observed. No significant adverse events related to the treatment were observed and patients tolerated the stimulation protocol well overall. Conclusions: This pilot study indicates that an accelerated rTMS protocol is a safe and viable option, and may be an effective alternative or adjunctive therapy for patients suffering from PSD. Future randomized, controlled studies are needed to confirm these preliminary findings. Clinical Trial Registration: https://clinicaltrials.gov/ct2/show/NCT04093843.
Collapse
Affiliation(s)
- Jessica Frey
- Department of Neurology, West Virginia University, Morgantown, WV, United States
| | - Umer Najib
- Department of Neurology, West Virginia University, Morgantown, WV, United States
| | - Christa Lilly
- Department of Biostatistics, West Virginia University, Morgantown, WV, United States
| | - Amelia Adcock
- Department of Neurology, West Virginia University, Morgantown, WV, United States
| |
Collapse
|
20
|
Sasegbon A, Cheng I, Zhang M, Hamdy S. Advances in the Use of Neuromodulation for Neurogenic Dysphagia: Mechanisms and Therapeutic Application of Pharyngeal Electrical Stimulation, Transcranial Magnetic Stimulation, and Transcranial Direct Current Stimulation. AMERICAN JOURNAL OF SPEECH-LANGUAGE PATHOLOGY 2020; 29:1044-1064. [PMID: 32650660 DOI: 10.1044/2020_ajslp-19-00073] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Purpose The swallowing motor system and, specifically, its cortical substrates appear to have certain unique properties that make it highly susceptible to brain plasticity, both driven and following injury. Furthermore, neurogenic dysphagia is a common complication of brain disease, associated with poor outcomes, and yet treatment options remain limited. Therefore, translating the physiology of neurostimulation into clinical populations becomes imperative. In this review, we describe therapeutic application of neuroplasticity in the human swallowing motor system by initially examining the role of pharyngeal electrical stimulation from a mechanistic perspective and then reporting on clinical studies using this approach. Thereafter, we explore the application of noninvasive brain stimulation, which has previously been used to treat nervous system disorders such as depression, pain modulation, and cognitive impairment. Transcranial brain stimulations, in particular, transcranial magnetic stimulation and transcranial direct current stimulation, have been utilized by a number of investigators for rehabilitation in early-stage clinical trials, including dysphagia after neurological disease. In this review, we assess its usefulness in neurogenic dysphagia. Conclusion Early studies indicate these emerging neurostimulatory techniques hold future therapeutic promise. However, both a greater number of and larger clinical trials are required to provide evidence delineating their efficacy and scope of application.
Collapse
Affiliation(s)
- Ayodele Sasegbon
- Faculty of Biology, Medicine, and Health, University of Manchester, Salford, United Kingdom
| | - Ivy Cheng
- Faculty of Biology, Medicine, and Health, University of Manchester, Salford, United Kingdom
| | - Mengqing Zhang
- Faculty of Biology, Medicine, and Health, University of Manchester, Salford, United Kingdom
| | - Shaheen Hamdy
- Faculty of Biology, Medicine, and Health, University of Manchester, Salford, United Kingdom
| |
Collapse
|
21
|
Moretti J, Poh EZ, Rodger J. rTMS-Induced Changes in Glutamatergic and Dopaminergic Systems: Relevance to Cocaine and Methamphetamine Use Disorders. Front Neurosci 2020; 14:137. [PMID: 32210744 PMCID: PMC7068681 DOI: 10.3389/fnins.2020.00137] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Accepted: 02/03/2020] [Indexed: 12/12/2022] Open
Abstract
Cocaine use disorder and methamphetamine use disorder are chronic, relapsing disorders with no US Food and Drug Administration-approved interventions. Repetitive transcranial magnetic stimulation (rTMS) is a non-invasive brain stimulation tool that has been increasingly investigated as a possible therapeutic intervention for substance use disorders. rTMS may have the ability to induce beneficial neuroplasticity in abnormal circuits and networks in individuals with addiction. The aim of this review is to highlight the rationale and potential for rTMS to treat cocaine and methamphetamine dependence: we synthesize the outcomes of studies in healthy humans and animal models to identify and understand the neurobiological mechanisms of rTMS that seem most involved in addiction, focusing on the dopaminergic and glutamatergic systems. rTMS-induced changes to neurotransmitter systems include alterations to striatal dopamine release and metabolite levels, as well as to glutamate transporter and receptor expression, which may be relevant for ameliorating the aberrant plasticity observed in individuals with substance use disorders. We also discuss the clinical studies that have used rTMS in humans with cocaine and methamphetamine use disorders. Many such studies suggest changes in network connectivity following acute rTMS, which may underpin reduced craving following chronic rTMS. We suggest several possible future directions for research relating to the therapeutic potential of rTMS in addiction that would help fill current gaps in the literature. Such research would apply rTMS to animal models of addiction, developing a translational pipeline that would guide evidence-based rTMS treatment of cocaine and methamphetamine use disorder.
Collapse
Affiliation(s)
- Jessica Moretti
- Experimental and Regenerative Neurosciences, School of Biological Sciences, The University of Western Australia, Crawley, WA, Australia.,School of Human Sciences, The University of Western Australia, Crawley, WA, Australia.,Brain Plasticity Group, Perron Institute for Neurological and Translational Science, Nedlands, WA, Australia
| | - Eugenia Z Poh
- Experimental and Regenerative Neurosciences, School of Biological Sciences, The University of Western Australia, Crawley, WA, Australia.,School of Human Sciences, The University of Western Australia, Crawley, WA, Australia.,Brain Plasticity Group, Perron Institute for Neurological and Translational Science, Nedlands, WA, Australia
| | - Jennifer Rodger
- Experimental and Regenerative Neurosciences, School of Biological Sciences, The University of Western Australia, Crawley, WA, Australia.,Brain Plasticity Group, Perron Institute for Neurological and Translational Science, Nedlands, WA, Australia
| |
Collapse
|
22
|
Chou YH, Ton That V, Sundman M. A systematic review and meta-analysis of rTMS effects on cognitive enhancement in mild cognitive impairment and Alzheimer's disease. Neurobiol Aging 2020; 86:1-10. [PMID: 31783330 PMCID: PMC6995441 DOI: 10.1016/j.neurobiolaging.2019.08.020] [Citation(s) in RCA: 160] [Impact Index Per Article: 40.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2019] [Revised: 08/06/2019] [Accepted: 08/21/2019] [Indexed: 12/23/2022]
Abstract
Repetitive transcranial magnetic stimulation (rTMS), a noninvasive brain stimulation technique, has emerged as a promising treatment for mild cognitive impairment (MCI) and Alzheimer's disease (AD). Currently, however, the effectiveness of this therapy is unclear because of the low statistical power and heterogeneity of previous trials. The purpose of the meta-analysis was to systematically characterize the effectiveness of various combinations of rTMS parameters on different cognitive domains in patients with MCI and AD. Thirteen studies comprising 293 patients with MCI or AD were included in this analysis. Random-effects analysis revealed an overall medium-to-large effect size (0.77) favoring active rTMS over sham rTMS in the improvement of cognitive functions. Subgroup analyses revealed that (1) high-frequency rTMS over the left dorsolateral prefrontal cortex and low-frequency rTMS at the right dorsolateral prefrontal cortex significantly improved memory functions; (2) high-frequency rTMS targeting the right inferior frontal gyrus significantly enhanced executive performance; and (3) the effects of 5-30 consecutive rTMS sessions could last for 4-12 weeks. Potential mechanisms of rTMS effects on cognitive functions are discussed.
Collapse
Affiliation(s)
- Ying-Hui Chou
- Department of Psychology, Brain Imaging and TMS Laboratory, University of Arizona, Tucson, USA; Everlyn F McKnight Brain Institute, Arizona Center on Aging, and BIO5 Institute, University of Arizona, Tucson, USA.
| | - Viet Ton That
- Department of Psychology, Brain Imaging and TMS Laboratory, University of Arizona, Tucson, USA
| | - Mark Sundman
- Department of Psychology, Brain Imaging and TMS Laboratory, University of Arizona, Tucson, USA
| |
Collapse
|
23
|
Chou YH, Ton That V, Sundman M. A systematic review and meta-analysis of rTMS effects on cognitive enhancement in mild cognitive impairment and Alzheimer's disease. Neurobiol Aging 2020; 86:1-10. [PMID: 31783330 DOI: 10.1016/j.neurobiolaging.201908.020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2019] [Revised: 08/06/2019] [Accepted: 08/21/2019] [Indexed: 05/26/2023]
Abstract
Repetitive transcranial magnetic stimulation (rTMS), a noninvasive brain stimulation technique, has emerged as a promising treatment for mild cognitive impairment (MCI) and Alzheimer's disease (AD). Currently, however, the effectiveness of this therapy is unclear because of the low statistical power and heterogeneity of previous trials. The purpose of the meta-analysis was to systematically characterize the effectiveness of various combinations of rTMS parameters on different cognitive domains in patients with MCI and AD. Thirteen studies comprising 293 patients with MCI or AD were included in this analysis. Random-effects analysis revealed an overall medium-to-large effect size (0.77) favoring active rTMS over sham rTMS in the improvement of cognitive functions. Subgroup analyses revealed that (1) high-frequency rTMS over the left dorsolateral prefrontal cortex and low-frequency rTMS at the right dorsolateral prefrontal cortex significantly improved memory functions; (2) high-frequency rTMS targeting the right inferior frontal gyrus significantly enhanced executive performance; and (3) the effects of 5-30 consecutive rTMS sessions could last for 4-12 weeks. Potential mechanisms of rTMS effects on cognitive functions are discussed.
Collapse
Affiliation(s)
- Ying-Hui Chou
- Department of Psychology, Brain Imaging and TMS Laboratory, University of Arizona, Tucson, USA; Everlyn F McKnight Brain Institute, Arizona Center on Aging, and BIO5 Institute, University of Arizona, Tucson, USA.
| | - Viet Ton That
- Department of Psychology, Brain Imaging and TMS Laboratory, University of Arizona, Tucson, USA
| | - Mark Sundman
- Department of Psychology, Brain Imaging and TMS Laboratory, University of Arizona, Tucson, USA
| |
Collapse
|
24
|
Weiler M, Stieger KC, Long JM, Rapp PR. Transcranial Magnetic Stimulation in Alzheimer's Disease: Are We Ready? eNeuro 2020; 7:ENEURO.0235-19.2019. [PMID: 31848209 PMCID: PMC6948923 DOI: 10.1523/eneuro.0235-19.2019] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Revised: 12/09/2019] [Accepted: 12/11/2019] [Indexed: 02/08/2023] Open
Abstract
Transcranial magnetic stimulation (TMS) is among a growing family of noninvasive brain stimulation techniques being developed to treat multiple neurocognitive disorders, including Alzheimer's disease (AD). Although small clinical trials in AD have reported positive effects on cognitive outcome measures, significant knowledge gaps remain, and little attention has been directed at examining the potential influence of TMS on AD pathogenesis. Our review briefly outlines some of the proposed neurobiological mechanisms of TMS benefits in AD, with particular emphasis on the modulatory effects on excitatory/inhibitory balance. On the basis of converging evidence from multiple fields, we caution that TMS therapeutic protocols established in young adults may have unexpected detrimental effects in older individuals or in the brain compromised by AD pathology. Our review surveys clinical studies of TMS in AD alongside basic research as a guide for moving this important area of work forward toward effective treatment development.
Collapse
Affiliation(s)
- Marina Weiler
- Neurocognitive Aging Section, Laboratory of Behavioral Neuroscience, National Institute on Aging, Intramural Research Program, Baltimore, Maryland 21224
| | - Kevin C Stieger
- Neurocognitive Aging Section, Laboratory of Behavioral Neuroscience, National Institute on Aging, Intramural Research Program, Baltimore, Maryland 21224
| | - Jeffrey M Long
- Neurocognitive Aging Section, Laboratory of Behavioral Neuroscience, National Institute on Aging, Intramural Research Program, Baltimore, Maryland 21224
| | - Peter R Rapp
- Neurocognitive Aging Section, Laboratory of Behavioral Neuroscience, National Institute on Aging, Intramural Research Program, Baltimore, Maryland 21224
| |
Collapse
|
25
|
Madeo G, Bonci A. Rewiring the Addicted Brain: Circuits-Based Treatment for Addiction. COLD SPRING HARBOR SYMPOSIA ON QUANTITATIVE BIOLOGY 2019; 83:173-184. [PMID: 31097615 DOI: 10.1101/sqb.2018.83.038158] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
The advent of the noninvasive brain stimulation (NIBS) technique has paved the way for neural circuit-based treatments for addiction. Recently, evidence from both preclinical and clinical studies has evaluated the use of transcranial magnetic stimulation (TMS) as a safe and cost-effective therapeutic tool for substance use disorders (SUDs). Indeed, repetitive TMS impacts on neural activity inducing short- and long-term effects involving neuroplasticity mechanisms locally within the target area of stimulation and the network level throughout the brain. Here, we provide an integrated view of evidence highlighting the mechanisms of TMS-induced effects on modulating the maladaptive brain circuitry of addiction. We then review the preclinical and clinical findings suggesting rTMS as an effective interventional tool for the treatment of SUDs.
Collapse
Affiliation(s)
- Graziella Madeo
- Novella Fronda Foundation, Human Science and Brain Research Piazza Castello, 16-35141 Padua, Italy.,Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Baltimore, Maryland 21224, USA
| | - Antonello Bonci
- Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Baltimore, Maryland 21224, USA.,Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287, USA.,Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287, USA
| |
Collapse
|
26
|
Hack LM, Fries GR, Eyre HA, Bousman CA, Singh AB, Quevedo J, John VP, Baune BT, Dunlop BW. Moving pharmacoepigenetics tools for depression toward clinical use. J Affect Disord 2019; 249:336-346. [PMID: 30802699 PMCID: PMC6763314 DOI: 10.1016/j.jad.2019.02.009] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 02/01/2019] [Accepted: 02/05/2019] [Indexed: 12/13/2022]
Abstract
BACKGROUND Major depressive disorder (MDD) is a leading cause of disability worldwide, and over half of patients do not achieve symptom remission following an initial antidepressant course. Despite evidence implicating a strong genetic basis for the pathophysiology of MDD, there are no adequately validated biomarkers of treatment response routinely used in clinical practice. Pharmacoepigenetics is an emerging field that has the potential to combine both genetic and environmental information into treatment selection and further the goal of precision psychiatry. However, this field is in its infancy compared to the more established pharmacogenetics approaches. METHODS We prepared a narrative review using literature searches of studies in English pertaining to pharmacoepigenetics and treatment of depressive disorders conducted in PubMed, Google Scholar, PsychINFO, and Ovid Medicine from inception through January 2019. We reviewed studies of DNA methylation and histone modifications in both humans and animal models of depression. RESULTS Emerging evidence from human and animal work suggests a key role for epigenetic marks, including DNA methylation and histone modifications, in the prediction of antidepressant response. The challenges of heterogeneity of patient characteristics and loci studied as well as lack of replication that have impacted the field of pharmacogenetics also pose challenges to the development of pharmacoepigenetic tools. Additionally, given the tissue specific nature of epigenetic marks as well as their susceptibility to change in response to environmental factors and aging, pharmacoepigenetic tools face additional challenges to their development. LIMITATIONS This is a narrative and not systematic review of the literature on the pharmacoepigenetics of antidepressant response. We highlight key studies pertaining to pharmacoepigenetics and treatment of depressive disorders in humans and depressive-like behaviors in animal models, regardless of sample size or methodology. While we discuss DNA methylation and histone modifications, we do not cover microRNAs, which have been reviewed elsewhere recently. CONCLUSIONS Utilization of genome-wide approaches and reproducible epigenetic assays, careful selection of the tissue assessed, and integration of genetic and clinical information into pharmacoepigenetic tools will improve the likelihood of developing clinically useful tests.
Collapse
Affiliation(s)
- Laura M Hack
- Department of Psychiatry and Behavioral Sciences, School of Medicine, Emory University, Atlanta, GA, USA; Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, 401 Quarry Road, Palo Alto, CA 94305, USA; Sierra Pacific Mental Illness Research Education and Clinical Centers, VA Palo Alto Health Care System, Palo Alto, CA, USA.
| | - Gabriel R Fries
- Department of Psychiatry and Behavioral Sciences, McGovern Medical School, University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
| | - Harris A Eyre
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, 401 Quarry Road, Palo Alto, CA 94305, USA; Innovation Institute, Texas Medical Center, Houston, TX, USA; IMPACT SRC, School of Medicine, Deakin University, Geelong, Victoria, Australia; Department of Psychiatry, University of Melbourne, Melbourne, Victoria, Australia
| | - Chad A Bousman
- Departments of Medical Genetics, Psychiatry, Physiology & Pharmacology, University of Calgary, Calgary, AB, Canada
| | - Ajeet B Singh
- IMPACT SRC, School of Medicine, Deakin University, Geelong, Victoria, Australia
| | - Joao Quevedo
- Department of Psychiatry and Behavioral Sciences, McGovern Medical School, University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
| | - Vineeth P John
- Department of Psychiatry and Behavioral Sciences, McGovern Medical School, University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
| | - Bernhard T Baune
- Department of Psychiatry, University of Melbourne, Melbourne, Victoria, Australia
| | - Boadie W Dunlop
- Department of Psychiatry and Behavioral Sciences, School of Medicine, Emory University, Atlanta, GA, USA
| |
Collapse
|
27
|
Repetitive transcranial magnetic stimulation: Re-wiring the alcoholic human brain. Alcohol 2019; 74:113-124. [PMID: 30420113 DOI: 10.1016/j.alcohol.2018.05.011] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2018] [Revised: 05/15/2018] [Accepted: 05/28/2018] [Indexed: 12/28/2022]
Abstract
Alcohol use disorders (AUDs) are one of the leading causes of mortality and morbidity worldwide. In spite of significant advances in understanding the neural underpinnings of AUDs, therapeutic options remain limited. Recent studies have highlighted the potential of repetitive transcranial magnetic stimulation (rTMS) as an innovative, safe, and cost-effective treatment for AUDs. Here, we summarize the fundamental principles of rTMS and its putative mechanisms of action via neurocircuitries related to alcohol addiction. We will also discuss advantages and limitations of rTMS, and argue that Hebbian plasticity and connectivity changes, as well as state-dependency, play a role in shaping some of the long-term effects of rTMS. Visual imaging studies will be linked to recent clinical pilot studies describing the effect of rTMS on alcohol craving and intake, pinpointing new advances, and highlighting conceptual gaps to be filled by future controlled studies.
Collapse
|
28
|
Li Y, Li L, Pan W. Repetitive Transcranial Magnetic Stimulation (rTMS) Modulates Hippocampal Structural Synaptic Plasticity in Rats. Physiol Res 2018; 68:99-105. [DOI: 10.33549/physiolres.933772] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Repetitive transcranial magnetic stimulation (rTMS) was shown to have therapeutic potential for some neurological and psychiatric disorders. Previous studies reported that low-frequency rTMS (</=1 Hz) affected synaptic plasticity in rats, however, there were few investigations to examine the possible effects of rTMS on structural synaptic plasticity changes in rats, which included the effects on synaptic morphology in the hippocampus, synaptic protein markers and Ca(2+)/calmodulin-dependent protein II (CaMKII). Sprague-Dawley rats were subject to 500 pulses of 0.5 Hz rTMS for 15 days, or sham stimulation. After last stimulation, transmission electron microscope (TEM) and real-time PCR were used to determine the effects of rTMS on synaptic plasticity. Results showed that rTMS could cause the change of structural synaptic plasticity, increase the expression of synaptic protein markers: synaptophysin (SYN) and increase the expression of CaMKII, relative to normal rats. suggesting a modulatory effect of chronic rTMS on synaptic plasticity that may be attributed to the increased expression of CaMKII in rats.
Collapse
Affiliation(s)
- Yue Li
- Beijing Key Laboratory of Bioelectromagnetics, Institute of Electrical Engineering, Chinese Academy of Sciences, Beijing, China
| | - Lulu Li
- Beijing Key Laboratory of Bioelectromagnetics, Institute of Electrical Engineering, Chinese Academy of Sciences, Beijing, China
| | - Weidong Pan
- Beijing Key Laboratory of Bioelectromagnetics, Institute of Electrical Engineering, Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
29
|
Therapeutic Effects of Repetitive Transcranial Magnetic Stimulation (rTMS) on Neuroinflammation and Neuroplasticity in Patients with Parkinson’s Disease: a Placebo-Controlled Study. Bull Exp Biol Med 2018; 165:195-199. [DOI: 10.1007/s10517-018-4128-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Indexed: 11/26/2022]
|
30
|
|
31
|
Consales C, Merla C, Marino C, Benassi B. The epigenetic component of the brain response to electromagnetic stimulation in Parkinson's Disease patients: A literature overview. Bioelectromagnetics 2017; 39:3-14. [PMID: 28990199 DOI: 10.1002/bem.22083] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Accepted: 08/20/2017] [Indexed: 12/12/2022]
Abstract
Modulations of epigenetic machinery, namely DNA methylation pattern, histone modification, and non-coding RNAs expression, have been recently included among the key determinants contributing to Parkinson's Disease (PD) aetiopathogenesis and response to therapy. Along this line of reasoning, a set of experimental findings are highlighting the epigenetic-based response to electromagnetic (EM) therapies used to alleviate PD symptomatology, mainly Deep Brain Stimulation (DBS) and Transcranial Magnetic Stimulation (TMS). Notwithstanding the proven efficacy of EM therapies, the precise molecular mechanisms underlying the brain response to these types of stimulations are still far from being elucidated. In this review we provide an overview of the epigenetic changes triggered by DBS and TMS in both PD patients and neurons from different experimental animal models. Furthermore, we also propose a critical overview of the exposure modalities currently applied, in order to evaluate the technical robustness and dosimetric control of the stimulation, which are key issues to be carefully assessed when new molecular findings emerge from experimental studies. Bioelectromagnetics. 39:3-14, 2018. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Claudia Consales
- ENEA-Italian National Agency for New Technologies, Energy and Sustainable Economic Development, Rome, Italy
| | - Caterina Merla
- ENEA-Italian National Agency for New Technologies, Energy and Sustainable Economic Development, Rome, Italy.,CNRS, Gustave Roussy, University of Paris-Sud, Université Paris-Saclay, Villejuif, France
| | - Carmela Marino
- ENEA-Italian National Agency for New Technologies, Energy and Sustainable Economic Development, Rome, Italy
| | - Barbara Benassi
- ENEA-Italian National Agency for New Technologies, Energy and Sustainable Economic Development, Rome, Italy
| |
Collapse
|
32
|
Diana M, Raij T, Melis M, Nummenmaa A, Leggio L, Bonci A. Rehabilitating the addicted brain with transcranial magnetic stimulation. Nat Rev Neurosci 2017; 18:685-693. [PMID: 28951609 DOI: 10.1038/nrn.2017.113] [Citation(s) in RCA: 149] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Substance use disorders (SUDs) are one of the leading causes of morbidity and mortality worldwide. In spite of considerable advances in understanding the neural underpinnings of SUDs, therapeutic options remain limited. Recent studies have highlighted the potential of transcranial magnetic stimulation (TMS) as an innovative, safe and cost-effective treatment for some SUDs. Repetitive TMS (rTMS) influences neural activity in the short and long term by mechanisms involving neuroplasticity both locally, under the stimulating coil, and at the network level, throughout the brain. The long-term neurophysiological changes induced by rTMS have the potential to affect behaviours relating to drug craving, intake and relapse. Here, we review TMS mechanisms and evidence that rTMS is opening new avenues in addiction treatments.
Collapse
Affiliation(s)
- Marco Diana
- 'G. Minardi' Laboratory for Cognitive Neuroscience, Department of Chemistry and Pharmacy, University of Sassari, 07100 Sassari, Italy
| | - Tommi Raij
- Shirley Ryan AbilityLab, Center for Brain Stimulation, the Department of Physical Medicine and Rehabilitation and the Department of Neurobiology, Northwestern University, Chicago, Illinois 60611, USA
| | - Miriam Melis
- Department of Biomedical Sciences, Division of Neuroscience and Clinical Pharmacology, University of Cagliari, 09042 Monserrato, Italy
| | - Aapo Nummenmaa
- Massachusetts General Hospital (MGH)/Massachusetts Institute of Technology (MIT)/Harvard Medical School (HMS) Athinoula A. Martinos Center for Biomedical Imaging, Harvard Medical School, Boston, Massachusetts 02129, USA
| | - Lorenzo Leggio
- Section on Clinical Psychoneuroendocrinology and Neuropsychopharmacology, US National Institute on Alcohol Abuse and Alcoholism Division of Intramural Clinical and Biological Research (NIAAA DICBR) and US National Institute on Drug Abuse Intramural Research Program (NIDA IRP), NIH (National Institutes of Health), Bethesda, Maryland 20892, USA; and at the Center for Alcohol and Addiction Studies, Brown University, Providence, Rhode Island 02912, USA
| | - Antonello Bonci
- US National Institute on Drug Abuse Intramural Research Program (NIDA IRP); and at the Departments of Neuroscience and Psychiatry, Johns Hopkins University, Baltimore, Maryland 21224, USA
| |
Collapse
|
33
|
Continuous theta burst stimulation facilitates the clearance efficiency of the glymphatic pathway in a mouse model of sleep deprivation. Neurosci Lett 2017; 653:189-194. [DOI: 10.1016/j.neulet.2017.05.064] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Revised: 05/23/2017] [Accepted: 05/29/2017] [Indexed: 01/06/2023]
|
34
|
Cirillo G, Di Pino G, Capone F, Ranieri F, Florio L, Todisco V, Tedeschi G, Funke K, Di Lazzaro V. Neurobiological after-effects of non-invasive brain stimulation. Brain Stimul 2017; 10:1-18. [DOI: 10.1016/j.brs.2016.11.009] [Citation(s) in RCA: 196] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Revised: 11/14/2016] [Accepted: 11/15/2016] [Indexed: 01/05/2023] Open
|