151
|
Chang YC, Peng YJ, Lee JY, Chang KT. Peripheral glia and neurons jointly regulate activity-induced synaptic remodeling at the Drosophila neuromuscular junction. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.27.600908. [PMID: 39005352 PMCID: PMC11244886 DOI: 10.1101/2024.06.27.600908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/16/2024]
Abstract
In the nervous system, reliable communication depends on the ability of neurons to adaptively remodel their synaptic structure and function in response to changes in neuronal activity. While neurons are the main drivers of synaptic plasticity, glial cells are increasingly recognized for their roles as active modulators. However, the underlying molecular mechanisms remain unclear. Here, using Drosophila neuromuscular junction as a model system for a tripartite synapse, we show that peripheral glial cells collaborate with neurons at the NMJ to regulate activity-induced synaptic remodeling, in part through a protein called shriveled (Shv). Shv is an activator of integrin signaling previously shown to be released by neurons during intense stimulation at the fly NMJ to regulate activity-induced synaptic remodeling. We demonstrate that Shv is also present in peripheral glia, and glial Shv is both necessary and sufficient for synaptic remodeling. However, unlike neuronal Shv, glial Shv does not activate integrin signaling at the NMJ. Instead, it regulates synaptic plasticity in two ways: 1) maintaining the extracellular balance of neuronal Shv proteins to regulate integrin signaling, and 2) controlling ambient extracellular glutamate concentration to regulate postsynaptic glutamate receptor abundance. Loss of glial cells showed the same phenotype as loss of Shv in glia. Together, these results reveal that neurons and glial cells homeostatically regulate extracellular Shv protein levels to control activity-induced synaptic remodeling. Additionally, peripheral glia maintains postsynaptic glutamate receptor abundance and contribute to activity-induced synaptic remodeling by regulating ambient glutamate concentration at the fly NMJ.
Collapse
|
152
|
Cai JW, Ye JT, Zhong YN, Zhang ZD, Zong H, Li LX, Han XE, Xu JL, Gao X, Lee ST, Wang SD. Solution-Processed Polymer Memcapacitors with Stimulus-Controlled and Evolvable Synaptic Functionalities: From Short-Term Plasticity to Long-Term Plasticity to Metaplasticity. ACS APPLIED MATERIALS & INTERFACES 2024; 16:47996-48004. [PMID: 39221579 DOI: 10.1021/acsami.4c09593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
In the vanguard of neuromorphic engineering, we develop a paradigm of biocompatible polymer memcapacitors using a seamless solution process, unleashing comprehensive synaptic capabilities depending on both the stimulation form and history. Like the human brain to learn and adapt, the memcapacitors exhibit analogue-type and evolvable capacitance shifts that mirror the complex flexibility of synaptic strengthening and weakening. With increasing frequency and intensity of the stimulation, the memcapacitors demonstrate an evolution from short-term plasticity (STP) to long-term plasticity (LTP), and even to metaplasticity (MP) at a higher level. A physical picture, featuring the stimulus-controlled spatiotemporal ion redistribution in the polymer, elaborates the origin of the memcapacitive prowess and resultant versatile synaptic plasticity. The distinctive MP behavior endows the memcapacitors with a dynamic learning rate (LR), which is utilized in an artificial neural network. The superiority of implementing a dynamic LR compared with conventional practices of using constant LR shines light on the potential of the memcapacitors to exploit organic neuromorphic computing hardware.
Collapse
Affiliation(s)
- Jia-Wei Cai
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou, Jiangsu 215123, P. R. China
| | - Jing-Ting Ye
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou, Jiangsu 215123, P. R. China
| | - Ya-Nan Zhong
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou, Jiangsu 215123, P. R. China
| | - Zhong-Da Zhang
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou, Jiangsu 215123, P. R. China
| | - Hao Zong
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou, Jiangsu 215123, P. R. China
| | - Li-Xing Li
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou, Jiangsu 215123, P. R. China
| | - Xue-Er Han
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou, Jiangsu 215123, P. R. China
| | - Jian-Long Xu
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou, Jiangsu 215123, P. R. China
| | - Xu Gao
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou, Jiangsu 215123, P. R. China
| | - Shuit-Tong Lee
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou, Jiangsu 215123, P. R. China
- Macao Institute of Materials Science and Engineering (MIMSE), MUST-SUDA Joint Research Center for Advanced Functional Materials, Macau University of Science and Technology, Taipa 999078, Macao, P. R. China
| | - Sui-Dong Wang
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou, Jiangsu 215123, P. R. China
- Macao Institute of Materials Science and Engineering (MIMSE), MUST-SUDA Joint Research Center for Advanced Functional Materials, Macau University of Science and Technology, Taipa 999078, Macao, P. R. China
| |
Collapse
|
153
|
Lee N, Pujar P, Hong S. Low-Cost, High-Efficiency Aluminum Zinc Oxide Synaptic Transistors: Blue LED Stimulation for Enhanced Neuromorphic Computing Applications. Biomimetics (Basel) 2024; 9:547. [PMID: 39329569 PMCID: PMC11430796 DOI: 10.3390/biomimetics9090547] [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: 07/31/2024] [Revised: 09/05/2024] [Accepted: 09/06/2024] [Indexed: 09/28/2024] Open
Abstract
Neuromorphic devices are electronic devices that mimic the information processing methods of neurons and synapses, enabling them to perform multiple tasks simultaneously with low power consumption and exhibit learning ability. However, their large-scale production and efficient operation remain a challenge. Herein, we fabricated an aluminum-doped zinc oxide (AZO) synaptic transistor via solution-based spin-coating. The transistor is characterized by low production costs and high performance. It demonstrates high responsiveness under UV laser illumination. In addition, it exhibits effective synaptic behaviors under blue LED illumination, indicating high-efficiency operation. The paired-pulse facilitation (PPF) index measured from optical stimulus modulation was 179.6%, indicating strong synaptic connectivity and effective neural communication and processing. Furthermore, by modulating the blue LED light pulse frequency, an excitatory postsynaptic current gain of 4.3 was achieved, demonstrating efficient neuromorphic functionality. This study shows that AZO synaptic transistors are promising candidates for artificial synaptic devices.
Collapse
Affiliation(s)
- Namgyu Lee
- Department of Physics, Gachon University, Seongnam 13120, Republic of Korea
| | - Pavan Pujar
- Department of Ceramic Engineering, Indian Institute of Technology (IIT-BHU), Varanasi 221005, Uttar Pradesh, India
| | - Seongin Hong
- Department of Physics, Gachon University, Seongnam 13120, Republic of Korea
- Department of Semiconductor Engineering, Gachon University, Seongnam 13120, Republic of Korea
| |
Collapse
|
154
|
Wen W, Wenjing Z, Xia X, Duan X, Zhang L, Duomao L, Zeyou Q, Wang S, Gao M, Liu C, Li H, Ma J. Efficacy of ketamine versus esketamine in the treatment of perioperative depression: A review. Pharmacol Biochem Behav 2024; 242:173773. [PMID: 38806116 DOI: 10.1016/j.pbb.2024.173773] [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: 10/31/2023] [Revised: 04/16/2024] [Accepted: 04/17/2024] [Indexed: 05/30/2024]
Abstract
Depression is a significant factor contributing to postoperative occurrences, and patients diagnosed with depression have a higher risk for postoperative complications. Studies on cardiovascular surgery extensively addresses this concern. Several studies report that people who undergo coronary artery bypass graft surgery have a 20% chance of developing postoperative depression. A retrospective analysis of medical records spanning 21 years, involving 817 patients, revealed that approximately 40% of individuals undergoing coronary artery bypass grafting (CABG) were at risk of perioperative depression. Patients endure prolonged suffering from illness because each attempt with standard antidepressants requires several weeks to be effective. In addition, multi-drug combination adjuvants or combination medication therapy may alleviate symptoms for some individuals, but they also increase the risk of side effects. Conventional antidepressants primarily modulate the monoamine system, whereas different therapies target the serotonin, norepinephrine, and dopamine systems. Esketamine is a fast-acting antidepressant with high efficacy. Esketamine is the S-enantiomer of ketamine, a derivative of phencyclidine developed in 1956. Esketamine exerts its effect by targeting the glutaminergic system the glutaminergic system. In this paper, we discuss the current depression treatment strategies with a focus on the pharmacology and mechanism of action of esketamine. In addition, studies reporting use of esketamine to treat perioperative depressive symptoms are reviwed, and the potential future applications of the drug are presented.
Collapse
Affiliation(s)
- Wen Wen
- Beijing Anzhen Hospital, Capital Medical University
| | - Zhao Wenjing
- Beijing Anzhen Hospital, Capital Medical University
| | - Xing Xia
- Beijing Anzhen Hospital, Capital Medical University
| | | | - Liang Zhang
- Beijing Anzhen Hospital, Capital Medical University
| | - Lin Duomao
- Beijing Anzhen Hospital, Capital Medical University
| | - Qi Zeyou
- Beijing Anzhen Hospital, Capital Medical University
| | - Sheng Wang
- Beijing Anzhen Hospital, Capital Medical University
| | - Mingxin Gao
- Beijing Anzhen Hospital, Capital Medical University
| | | | - Haiyang Li
- Beijing Anzhen Hospital, Capital Medical University.
| | - Jun Ma
- Beijing Anzhen Hospital, Capital Medical University.
| |
Collapse
|
155
|
Oliva HNP, Prudente TP, Nunes EJ, Cosgrove KP, Radhakrishnan R, Potenza MN, Angarita GA. Substance use and spine density: a systematic review and meta-analysis of preclinical studies. Mol Psychiatry 2024; 29:2873-2885. [PMID: 38561468 DOI: 10.1038/s41380-024-02519-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2023] [Revised: 02/28/2024] [Accepted: 03/04/2024] [Indexed: 04/04/2024]
Abstract
The elucidation of synaptic density changes provides valuable insights into the underlying brain mechanisms of substance use. In preclinical studies, synaptic density markers, like spine density, are altered by substances of abuse (e.g., alcohol, amphetamine, cannabis, cocaine, opioids, nicotine). These changes could be linked to phenomena including behavioral sensitization and drug self-administration in rodents. However, studies have produced heterogeneous results for spine density across substances and brain regions. Identifying patterns will inform translational studies given tools that now exist to measure in vivo synaptic density in humans. We performed a meta-analysis of preclinical studies to identify consistent findings across studies. PubMed, ScienceDirect, Scopus, and EBSCO were searched between September 2022 and September 2023, based on a protocol (PROSPERO: CRD42022354006). We screened 6083 publications and included 70 for meta-analysis. The meta-analysis revealed drug-specific patterns in spine density changes. Hippocampal spine density increased after amphetamine. Amphetamine, cocaine, and nicotine increased spine density in the nucleus accumbens. Alcohol and amphetamine increased, and cannabis reduced, spine density in the prefrontal cortex. There was no convergence of findings for morphine's effects. The effects of cocaine on the prefrontal cortex presented contrasting results compared to human studies, warranting further investigation. Publication bias was small for alcohol or morphine and substantial for the other substances. Heterogeneity was moderate-to-high across all substances. Nonetheless, these findings inform current translational efforts examining spine density in humans with substance use disorders.
Collapse
Affiliation(s)
- Henrique Nunes Pereira Oliva
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
- Clinical Neuroscience Research Unit, Connecticut Mental Health Center, New Haven, CT, USA
| | - Tiago Paiva Prudente
- Faculdade de Medicina, Universidade Federal de Goiás (UFG), Goiânia, Goiás, Brazil
| | - Eric J Nunes
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
- Yale Tobacco Center of Regulatory Science, Yale University School of Medicine, New Haven, CT, USA
| | - Kelly P Cosgrove
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | - Rajiv Radhakrishnan
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | - Marc N Potenza
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
- Clinical Neuroscience Research Unit, Connecticut Mental Health Center, New Haven, CT, USA
- Child Study Center, Yale University School of Medicine, New Haven, CT, USA
- Department of Neuroscience, Yale University, New Haven, CT, USA
- Connecticut Council on Problem Gambling, Wethersfield, CT, USA
- Wu Tsai Institute, Yale University, New Haven, CT, USA
| | - Gustavo A Angarita
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA.
- Clinical Neuroscience Research Unit, Connecticut Mental Health Center, New Haven, CT, USA.
| |
Collapse
|
156
|
Gupta A, Bohara VS, Siddegowda YB, Chaudhary N, Kumar S. Alpha-synuclein and RNA viruses: Exploring the neuronal nexus. Virology 2024; 597:110141. [PMID: 38917691 DOI: 10.1016/j.virol.2024.110141] [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: 12/28/2023] [Accepted: 06/12/2024] [Indexed: 06/27/2024]
Abstract
Alpha-synuclein (α-syn), known for its pivotal role in Parkinson's disease, has recently emerged as a significant player in neurotropic RNA virus infections. Upregulation of α-syn in various viral infections has been found to impact neuroprotective functions by regulating neurotransmitter synthesis, vesicle trafficking, and synaptic vesicle recycling. This review focuses on the multifaceted role of α-syn in controlling viral replication by modulating chemoattractant properties towards microglial cells, virus-induced ER stress signaling, anti-oxidative proteins expression. Furthermore, the text underlines the α-syn-mediated regulation of interferon-stimulated genes. The review may help suggest potential therapeutic avenues for mitigating the impact of RNA viruses on the central nervous system by exploiting α-syn neuroprotective biology.
Collapse
Affiliation(s)
- Anjali Gupta
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, 781039, Assam, India
| | - Vijay Singh Bohara
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, 781039, Assam, India
| | | | - Nitin Chaudhary
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, 781039, Assam, India
| | - Sachin Kumar
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, 781039, Assam, India.
| |
Collapse
|
157
|
He X, Hu XY, Yin XY, Wu XM, Liu QR, Shen JC. GluR2 can Drive Neuroinflammation and Cognitive Impairments Following Peripherally Repeated Lipopolysaccharide Exposures. Neurochem Res 2024; 49:2393-2407. [PMID: 38837093 DOI: 10.1007/s11064-024-04183-z] [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: 03/15/2024] [Revised: 05/06/2024] [Accepted: 05/22/2024] [Indexed: 06/06/2024]
Abstract
Neuroinflammation is being increasingly recognized as a vital factor in the development of various neurological and neuropsychiatric diseases. Lipopolysaccharides (LPS), an outer membrane component of gram-negative bacteria, can trigger innate immune responses, resulting in neuroinflammation and subsequent cognitive deficits. The expression of glutamate receptors (GluRs) on glial cells can induce glial activation. Therefore, we hypothesized that repeated LPS exposure can increase GluR levels, promoting microglial activation and ultimately affecting synaptic plasticity and cognitive function. In this study, C57/BL6 mice were repeatedly exposed to LPS to construct a neuroinflammation animal model. The levels of GluRs, inflammatory cytokines, ionized calcium-binding adaptor molecule 1, postsynaptic density protein 95, synaptophysin 38, NMDA receptor 2 A, and NMDA receptor 2B (GluN2B) were measured in the hippocampi. Furthermore, dendritic spine density in the CA1 hippocampal region was determined. Repeated LPS exposure induced cognitive impairments and microglial activation and increased GluR1 and GluR2 levels. This was accompanied by a significant decrease in GluN2B expression and dendritic spine density in the hippocampi. However, CFM-2, an α-amino-3- hydroxy-5-methyl-4-isoxazolepropionate receptor antagonist, reversed these anomalies. Furthermore, minocycline, a microglial inhibitor, reversed these anomalies and downregulated GluR2 but not GluR1 expression. In summary, we demonstrated that GluR2 plays an essential role in microglia-induced neuroinflammation, resulting in synaptic plasticity and cognitive impairment induced by repeated exposure to LPS.
Collapse
Affiliation(s)
- Xue He
- Department of Anesthesiology, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, China
| | - Xiao-Yi Hu
- Department of Anesthesiology, the Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xiao-Yu Yin
- Department of Anesthesiology, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, China
| | - Xin-Miao Wu
- Department of Anesthesiology, the Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Qing-Ren Liu
- Department of Anesthesiology, Xishan People's Hospital of Wuxi City, Wuxi, China.
| | - Jin-Chun Shen
- Department of Anesthesiology, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, China.
| |
Collapse
|
158
|
Liu X, Sun C, Ye X, Zhu X, Hu C, Tan H, He S, Shao M, Li RW. Neuromorphic Nanoionics for Human-Machine Interaction: From Materials to Applications. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2311472. [PMID: 38421081 DOI: 10.1002/adma.202311472] [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: 10/31/2023] [Revised: 02/06/2024] [Indexed: 03/02/2024]
Abstract
Human-machine interaction (HMI) technology has undergone significant advancements in recent years, enabling seamless communication between humans and machines. Its expansion has extended into various emerging domains, including human healthcare, machine perception, and biointerfaces, thereby magnifying the demand for advanced intelligent technologies. Neuromorphic computing, a paradigm rooted in nanoionic devices that emulate the operations and architecture of the human brain, has emerged as a powerful tool for highly efficient information processing. This paper delivers a comprehensive review of recent developments in nanoionic device-based neuromorphic computing technologies and their pivotal role in shaping the next-generation of HMI. Through a detailed examination of fundamental mechanisms and behaviors, the paper explores the ability of nanoionic memristors and ion-gated transistors to emulate the intricate functions of neurons and synapses. Crucial performance metrics, such as reliability, energy efficiency, flexibility, and biocompatibility, are rigorously evaluated. Potential applications, challenges, and opportunities of using the neuromorphic computing technologies in emerging HMI technologies, are discussed and outlooked, shedding light on the fusion of humans with machines.
Collapse
Affiliation(s)
- Xuerong Liu
- CAS Key Laboratory of Magnetic Materials and Devices, and Zhejiang Province Key Laboratory of Magnetic Materials and Application Technology, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, 315201, China
- Zhejiang Province Key Laboratory of Magnetic Materials and Application Technology, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, 315201, China
- College of Materials Sciences and Opto-Electronic Technology, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Cui Sun
- CAS Key Laboratory of Magnetic Materials and Devices, and Zhejiang Province Key Laboratory of Magnetic Materials and Application Technology, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, 315201, China
- Zhejiang Province Key Laboratory of Magnetic Materials and Application Technology, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, 315201, China
| | - Xiaoyu Ye
- CAS Key Laboratory of Magnetic Materials and Devices, and Zhejiang Province Key Laboratory of Magnetic Materials and Application Technology, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, 315201, China
- Zhejiang Province Key Laboratory of Magnetic Materials and Application Technology, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, 315201, China
| | - Xiaojian Zhu
- CAS Key Laboratory of Magnetic Materials and Devices, and Zhejiang Province Key Laboratory of Magnetic Materials and Application Technology, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, 315201, China
- Zhejiang Province Key Laboratory of Magnetic Materials and Application Technology, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, 315201, China
| | - Cong Hu
- CAS Key Laboratory of Magnetic Materials and Devices, and Zhejiang Province Key Laboratory of Magnetic Materials and Application Technology, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, 315201, China
- Zhejiang Province Key Laboratory of Magnetic Materials and Application Technology, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, 315201, China
| | - Hongwei Tan
- Department of Applied Physics, Aalto University, Aalto, FI-00076, Finland
| | - Shang He
- CAS Key Laboratory of Magnetic Materials and Devices, and Zhejiang Province Key Laboratory of Magnetic Materials and Application Technology, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, 315201, China
- Zhejiang Province Key Laboratory of Magnetic Materials and Application Technology, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, 315201, China
| | - Mengjie Shao
- CAS Key Laboratory of Magnetic Materials and Devices, and Zhejiang Province Key Laboratory of Magnetic Materials and Application Technology, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, 315201, China
- Zhejiang Province Key Laboratory of Magnetic Materials and Application Technology, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, 315201, China
| | - Run-Wei Li
- CAS Key Laboratory of Magnetic Materials and Devices, and Zhejiang Province Key Laboratory of Magnetic Materials and Application Technology, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, 315201, China
- Zhejiang Province Key Laboratory of Magnetic Materials and Application Technology, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, 315201, China
| |
Collapse
|
159
|
Camargo L, Pacheco-Barrios K, Gianlorenço AC, Menacho M, Choi H, Song JJ, Fregni F. Evidence of bottom-up homeostatic modulation induced taVNS during emotional and Go/No-Go tasks. Exp Brain Res 2024; 242:2069-2081. [PMID: 38963558 DOI: 10.1007/s00221-024-06876-x] [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: 04/14/2024] [Accepted: 06/17/2024] [Indexed: 07/05/2024]
Abstract
Bilateral transcutaneous auricular vagus nerve stimulation (taVNS) - a non-invasive neuromodulation technique - has been investigated as a safe and feasible technique to treat many neuropsychiatric conditions. such as epilepsy, depression, anxiety, and chronic pain. Our aim is to investigate the effect of taVNS on neurophysiological processes during emotional and Go/No-Go tasks, and changes in frontal alpha asymmetry. We performed a randomized, double-blind, sham-controlled trial with 44 healthy individuals who were allocated into two groups (the active taVNS group and the sham taVNS group). Subjects received one session of taVNS (active or sham) for 60 min. QEEG was recorded before and after the interventions, and the subjects were assessed while exposed to emotional conditions with sad and happy facial expressions, followed by a Go/No-Go trial. The results demonstrated a significant increase in N2 amplitude in the No-Go condition for the active taVNS post-intervention compared to the sham taVNS after adjusting by handedness, mood, and fatigue levels (p = 0.046), significantly reduced ERD during sad conditions after treatment (p = 0.037), and increased frontal alpha asymmetry towards the right frontal hemisphere during the emotional task condition (p = 0.046). Finally, we observed an interesting neural signature in this study that suggests a bottom-up modulation from brainstem/subcortical to cortical areas as characterized by improved lateralization of alpha oscillations towards the frontal right hemisphere, and changes in ERP during emotional and Go/No-Go tasks that suggests a better subcortical response to the tasks. Such bottom-up effects may mediate some of the clinical effects of taVNS.
Collapse
Affiliation(s)
- Lucas Camargo
- Spaulding Neuromodulation Center, Center for Clinical Research Learning, Spaulding Rehabilitation Hospital, Harvard Medical School, 1575 Cambridge Street, Boston, MA, United States of America
| | - Kevin Pacheco-Barrios
- Spaulding Neuromodulation Center, Center for Clinical Research Learning, Spaulding Rehabilitation Hospital, Harvard Medical School, 1575 Cambridge Street, Boston, MA, United States of America
- Vicerrectorado de Investigación, Unidad de Investigación para la Generación y Síntesis de Evidencias en Salud, Universidad San Ignacio de Loyola, Lima, Peru
| | - Anna Carolyna Gianlorenço
- Spaulding Neuromodulation Center, Center for Clinical Research Learning, Spaulding Rehabilitation Hospital, Harvard Medical School, 1575 Cambridge Street, Boston, MA, United States of America
- Neurosciences Laboratory, Physical Therapy Department, Federal University of Sao Carlos, Sao Carlos, SP, Brazil
| | - Maryela Menacho
- Spaulding Neuromodulation Center, Center for Clinical Research Learning, Spaulding Rehabilitation Hospital, Harvard Medical School, 1575 Cambridge Street, Boston, MA, United States of America
- Neurosciences Laboratory, Physical Therapy Department, Federal University of Sao Carlos, Sao Carlos, SP, Brazil
| | - Hyuk Choi
- Department of Medical Sciences, Graduate School of Medicine, Korea University, Seoul, Republic of Korea
- Neurive Co., Ltd, Gimhae, Republic of Korea
| | - Jae-Jun Song
- Neurive Co., Ltd, Gimhae, Republic of Korea
- Department of Otorhinolaryngology-Head and Neck Surgery, Korea University Medical Center, Seoul, Republic of Korea
| | - Felipe Fregni
- Spaulding Neuromodulation Center, Center for Clinical Research Learning, Spaulding Rehabilitation Hospital, Harvard Medical School, 1575 Cambridge Street, Boston, MA, United States of America.
- Department of Otorhinolaryngology-Head and Neck Surgery, Korea University Medical Center, Seoul, Republic of Korea.
| |
Collapse
|
160
|
Fortulan R, Kheirabadi NR, Raeisi-Kheirabadi N, Nezamzadeh-Ejhieh A, Chiolerio A, Adamatzky A. Fractional-order memristive dynamics in colloidal graphitic carbon nitride systems. Phys Rev E 2024; 110:034607. [PMID: 39425438 DOI: 10.1103/physreve.110.034607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 08/20/2024] [Indexed: 10/21/2024]
Abstract
We report on the synthesis and characterization of a colloidal graphitic carbon nitride (g-C_{3}N_{4}) system exhibiting complex memfractance behavior. The g-C_{3}N_{4} colloid was prepared through thermal polymerization of urea, followed by dispersion in deionized water. X-ray diffraction and scanning electron microscopy confirmed the successful synthesis of g-C_{3}N_{4}. Electrical characterization revealed nonpinched hysteresis loops in current-voltage curves, indicative of memristive behavior with additional capacitive components. The device demonstrated stable resistive switching between high (∼50kΩ) and low (∼22kΩ) impedance states over 500 cycles, as well as synaptic plasticity-like conductance modulation. To capture these complex dynamics, we employed a generalized memfractance model that interpolates between memristive, memcapacitive, and second-order memristive elements. This model, employing fractional-order derivatives, accurately fitted the experimental data, revealing the device's memory effects. The emergence of memfractance in this colloidal system opens new avenues for neuromorphic computing and unconventional information processing architectures, leveraging the unique properties of liquid-state memory devices.
Collapse
|
161
|
Baghcheghi Y, Razazpour F, Seyedi F, Arefinia N, Hedayati-Moghadam M. Exploring the molecular mechanisms of PPARγ agonists in modulating memory impairment in neurodegenerative disorders. Mol Biol Rep 2024; 51:945. [PMID: 39215798 DOI: 10.1007/s11033-024-09850-6] [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: 05/06/2024] [Accepted: 08/08/2024] [Indexed: 09/04/2024]
Abstract
Neurodegenerative diseases are characterized by progressive memory impairment and cognitive decline. This review aims to unravel the molecular mechanisms involved in the enhancement of memory function and mitigation of memory impairment through the activation of PPARγ agonists in neurodegenerative diseases. The findings suggest that PPARγ agonists modulate various molecular pathways involved in memory formation and maintenance. Activation of PPARγ enhances synaptic plasticity, promotes neuroprotection, suppresses neuroinflammation, attenuates oxidative stress, and regulates amyloid-beta metabolism. The comprehensive understanding of these molecular mechanisms would facilitate the development of novel therapeutic approaches targeting PPARγ to improve memory function and ultimately to alleviate the burden of neurodegenerative diseases. Further research, including clinical trials, is warranted to explore the efficacy, safety, and optimal use of specific PPARγ agonists as potential therapeutic agents in the treatment of memory impairments associated with neurodegenerative diseases.
Collapse
Affiliation(s)
- Yousef Baghcheghi
- Bio Environmental Health Hazards Research Center, Jiroft University of Medical Sciences, Jiroft, Iran
- Student Research Committee, Jiroft University of Medical Sciences, Jiroft, Iran
| | - Fateme Razazpour
- Student Research Committee, Jiroft University of Medical Sciences, Jiroft, Iran
| | - Fatemeh Seyedi
- Department of Anatomical Sciences, School of Medicine, Jiroft University of Medical Sciences, Jiroft, Iran
| | - Nasir Arefinia
- Bio Environmental Health Hazards Research Center, Jiroft University of Medical Sciences, Jiroft, Iran
- Student Research Committee, Jiroft University of Medical Sciences, Jiroft, Iran
| | - Mahdiyeh Hedayati-Moghadam
- Student Research Committee, Jiroft University of Medical Sciences, Jiroft, Iran.
- Department of Physiology, School of Medicine, Jiroft University of Medical Sciences, Jiroft, Iran.
| |
Collapse
|
162
|
Hroudová J, Fišar Z. Alzheimer's disease approaches - Focusing on pathology, biomarkers and clinical trial candidates. Prog Neuropsychopharmacol Biol Psychiatry 2024; 134:111069. [PMID: 38917881 DOI: 10.1016/j.pnpbp.2024.111069] [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: 01/03/2024] [Revised: 06/19/2024] [Accepted: 06/19/2024] [Indexed: 06/27/2024]
Abstract
The strategy for the development of new drugs for Alzheimer's disease (AD) recognizes that an effective therapy requires early therapeutic intervention and a multifactorial approach that considers the individual initiators of AD development. Current knowledge of AD includes the understanding of pathophysiology, risk factors, biomarkers, and the evolving patterns of biomarker abnormalities. This knowledge is essential in identifying potential molecular targets for new drug development. This review summarizes promising AD drug candidates, many of which are currently in phase 2 or 3 clinical trials. New agents are classified according to the Common Alzheimer's Disease Research Ontology (CADRO). The main targets of new drugs for AD are processes related to amyloid beta and tau neurotoxicity, neurotransmission, inflammation, metabolism and bioenergetics, synaptic plasticity, and oxidative stress. These interventions are aimed at preventing disease onset and slowing or eliminating disease progression. The efficacy of pharmacotherapy may be enhanced by combining these drugs with other treatments, antioxidants, and dietary supplements. Ongoing research into AD pathophysiology, risk factors, biomarkers, and the dynamics of biomarker abnormalities may contribute to the understanding of AD and offer hope for effective therapeutic strategies in the near future.
Collapse
Affiliation(s)
- Jana Hroudová
- Department of Psychiatry, First Faculty of Medicine, Charles University and General University Hospital in Prague, Ke Karlovu 11, 120 00 Prague 2, Czech Republic.
| | - Zdeněk Fišar
- Department of Psychiatry, First Faculty of Medicine, Charles University and General University Hospital in Prague, Ke Karlovu 11, 120 00 Prague 2, Czech Republic
| |
Collapse
|
163
|
Demchenko I, Rampersad S, Datta A, Horn A, Churchill NW, Kennedy SH, Krishnan S, Rueda A, Schweizer TA, Griffiths JD, Boyden ES, Santarnecchi E, Bhat V. Target engagement of the subgenual anterior cingulate cortex with transcranial temporal interference stimulation in major depressive disorder: a protocol for a randomized sham-controlled trial. Front Neurosci 2024; 18:1390250. [PMID: 39268031 PMCID: PMC11390435 DOI: 10.3389/fnins.2024.1390250] [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: 02/23/2024] [Accepted: 08/06/2024] [Indexed: 09/15/2024] Open
Abstract
Background Transcranial temporal interference stimulation (tTIS) is a new, emerging neurostimulation technology that utilizes two or more electric fields at specific frequencies to modulate the oscillations of neurons at a desired spatial location in the brain. The physics of tTIS offers the advantage of modulating deep brain structures in a non-invasive fashion and with minimal stimulation of the overlying cortex outside of a selected target. As such, tTIS can be effectively employed in the context of therapeutics for the psychiatric disease of disrupted brain connectivity, such as major depressive disorder (MDD). The subgenual anterior cingulate cortex (sgACC), a key brain center that regulates human emotions and influences negative emotional states, is a plausible target for tTIS in MDD based on reports of its successful neuromodulation with invasive deep brain stimulation. Methods This pilot, single-site, double-blind, randomized, sham-controlled interventional clinical trial will be conducted at St. Michael's Hospital - Unity Health Toronto in Toronto, ON, Canada. The primary objective is to demonstrate target engagement of the sgACC with 130 Hz tTIS using resting-state magnetic resonance imaging (MRI) techniques. The secondary objective is to estimate the therapeutic potential of tTIS for MDD by evaluating the change in clinical characteristics of participants and electrophysiological outcomes and providing feasibility and tolerability estimates for a large-scale efficacy trial. Thirty participants (18-65 years) with unipolar, non-psychotic MDD will be recruited and randomized to receive 10 sessions of 130 Hz tTIS or sham stimulation (n = 15 per arm). The trial includes a pre- vs. post-treatment 3T MRI scan of the brain, clinical evaluation, and electroencephalography (EEG) acquisition at rest and during the auditory mismatch negativity (MMN) paradigm. Discussion This study is one of the first-ever clinical trials among patients with psychiatric disorders examining the therapeutic potential of repetitive tTIS and its neurobiological mechanisms. Data obtained from this trial will be used to optimize the tTIS approach and design a large-scale efficacy trial. Research in this area has the potential to provide a novel treatment option for individuals with MDD and circuitry-related disorders and may contribute to the process of obtaining regulatory approval for therapeutic applications of tTIS. Clinical Trial Registration ClinicalTrials.gov, identifier NCT05295888.
Collapse
Affiliation(s)
- Ilya Demchenko
- Interventional Psychiatry Program, St. Michael's Hospital - Unity Health Toronto, Toronto, ON, Canada
- Institute of Medical Science, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Institute for Biomedical Engineering, Science and Technology (iBEST), Keenan Research Centre for Biomedical Science, St. Michael's Hospital - Unity Health Toronto, Toronto, ON, Canada
| | - Sumientra Rampersad
- Department of Physics, University of Massachusetts Boston, Boston, MA, United States
- Department of Electrical and Computer Engineering, Northeastern University, Boston, MA, United States
| | - Abhishek Datta
- Research and Development, Soterix Medical, Inc., Woodbridge, NJ, United States
- Department of Biomedical Engineering, City College of New York, New York, NY, United States
| | - Andreas Horn
- Department of Neurology, Center for Brain Circuit Therapeutics, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, United States
- Department of Neurosurgery & Center for NeuroTechnology and NeuroRecovery (CNTR), Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
- Movement Disorder and Neuromodulation Unit, Department of Neurology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt - Universität zu Berlin, Berlin, Germany
| | - Nathan W Churchill
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital - Unity Health Toronto, Toronto, ON, Canada
- Neuroscience Research Program, St. Michael's Hospital - Unity Health Toronto, Toronto, ON, Canada
| | - Sidney H Kennedy
- Institute of Medical Science, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital - Unity Health Toronto, Toronto, ON, Canada
- Neuroscience Research Program, St. Michael's Hospital - Unity Health Toronto, Toronto, ON, Canada
- Department of Psychiatry, Temerty Faculty of Medicine, Toronto, ON, Canada
| | - Sridhar Krishnan
- Institute for Biomedical Engineering, Science and Technology (iBEST), Keenan Research Centre for Biomedical Science, St. Michael's Hospital - Unity Health Toronto, Toronto, ON, Canada
- Department of Electrical, Computer, and Biomedical Engineering, Toronto Metropolitan University, Toronto, ON, Canada
| | - Alice Rueda
- Interventional Psychiatry Program, St. Michael's Hospital - Unity Health Toronto, Toronto, ON, Canada
- Institute for Biomedical Engineering, Science and Technology (iBEST), Keenan Research Centre for Biomedical Science, St. Michael's Hospital - Unity Health Toronto, Toronto, ON, Canada
| | - Tom A Schweizer
- Institute of Medical Science, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Institute for Biomedical Engineering, Science and Technology (iBEST), Keenan Research Centre for Biomedical Science, St. Michael's Hospital - Unity Health Toronto, Toronto, ON, Canada
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital - Unity Health Toronto, Toronto, ON, Canada
- Neuroscience Research Program, St. Michael's Hospital - Unity Health Toronto, Toronto, ON, Canada
- Division of Neurosurgery, Department of Surgery, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - John D Griffiths
- Institute of Medical Science, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Department of Psychiatry, Temerty Faculty of Medicine, Toronto, ON, Canada
- Krembil Centre for Neuroinformatics, Centre for Addiction and Mental Health (CAMH), Toronto, ON, Canada
| | - Edward S Boyden
- Department of Brain and Cognitive Sciences, Media Arts and Sciences, and Biological Engineering, McGovern Institute for Brain Research and Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, United States
- Howard Hughes Medical Institute, Chevy Chase, MD, United States
| | - Emiliano Santarnecchi
- Precision Neuroscience and Neuromodulation Program, Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - Venkat Bhat
- Interventional Psychiatry Program, St. Michael's Hospital - Unity Health Toronto, Toronto, ON, Canada
- Institute of Medical Science, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Institute for Biomedical Engineering, Science and Technology (iBEST), Keenan Research Centre for Biomedical Science, St. Michael's Hospital - Unity Health Toronto, Toronto, ON, Canada
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital - Unity Health Toronto, Toronto, ON, Canada
- Neuroscience Research Program, St. Michael's Hospital - Unity Health Toronto, Toronto, ON, Canada
- Department of Psychiatry, Temerty Faculty of Medicine, Toronto, ON, Canada
| |
Collapse
|
164
|
Patton MH, Thomas KT, Bayazitov IT, Newman KD, Kurtz NB, Robinson CG, Ramirez CA, Trevisan AJ, Bikoff JB, Peters ST, Pruett-Miller SM, Jiang Y, Schild AB, Nityanandam A, Zakharenko SS. Synaptic plasticity in human thalamocortical assembloids. Cell Rep 2024; 43:114503. [PMID: 39018245 PMCID: PMC11407288 DOI: 10.1016/j.celrep.2024.114503] [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: 03/11/2024] [Revised: 04/23/2024] [Accepted: 06/27/2024] [Indexed: 07/19/2024] Open
Abstract
Synaptic plasticities, such as long-term potentiation (LTP) and depression (LTD), tune synaptic efficacy and are essential for learning and memory. Current studies of synaptic plasticity in humans are limited by a lack of adequate human models. Here, we modeled the thalamocortical system by fusing human induced pluripotent stem cell-derived thalamic and cortical organoids. Single-nucleus RNA sequencing revealed that >80% of cells in thalamic organoids were glutamatergic neurons. When fused to form thalamocortical assembloids, thalamic and cortical organoids formed reciprocal long-range axonal projections and reciprocal synapses detectable by light and electron microscopy, respectively. Using whole-cell patch-clamp electrophysiology and two-photon imaging, we characterized glutamatergic synaptic transmission. Thalamocortical and corticothalamic synapses displayed short-term plasticity analogous to that in animal models. LTP and LTD were reliably induced at both synapses; however, their mechanisms differed from those previously described in rodents. Thus, thalamocortical assembloids provide a model system for exploring synaptic plasticity in human circuits.
Collapse
Affiliation(s)
- Mary H Patton
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Kristen T Thomas
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Ildar T Bayazitov
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Kyle D Newman
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Nathaniel B Kurtz
- Cell and Tissue Imaging Center, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Camenzind G Robinson
- Cell and Tissue Imaging Center, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Cody A Ramirez
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Alexandra J Trevisan
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Jay B Bikoff
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Samuel T Peters
- Center for Advanced Genome Engineering, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Shondra M Pruett-Miller
- Center for Advanced Genome Engineering, St. Jude Children's Research Hospital, Memphis, TN 38105, USA; Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Yanbo Jiang
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Andrew B Schild
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Anjana Nityanandam
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Stanislav S Zakharenko
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA.
| |
Collapse
|
165
|
Yin Y, Sun T, Wang L, Li L, Guo P, Liu X, Xiong L, Zu G, Huang J. In-Sensor Organic Electrochemical Transistor for the Multimode Neuromorphic Olfactory System. ACS Sens 2024; 9:4277-4285. [PMID: 39099107 DOI: 10.1021/acssensors.4c01423] [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] [Indexed: 08/06/2024]
Abstract
The olfactory system is one of the six basic sensory nervous systems. Developing artificial olfactory systems is challenging due to the complexity of chemical information decoding and memory. Conventional chemical sensors can convert chemical signals into electric signals to decode gas information but they lack memory functions. Additional storage and processing units would significantly increase the complexity and power consumption of the devices, especially for portable and wearable devices. Here, an olfactory-inspired in-sensor organic electrochemical transistor (OI-OECT) is proposed, with the integrated functions of chemical information decoding, tunable memory level, and selectivity of vapor sensing. The ion-gel electrolyte endows the OI-OECT with the function of tunable memory levels and a low operating voltage. Typical synaptic behaviors, including inhibitory postsynaptic current and paired-pulse facilitations, are successfully achieved. Importantly, the gas memory level can be effectively modulated by the gate voltages (0 and -1 V), which realized the transformation of volatile and nonvolatile memory. Furthermore, benefiting from the recognition of multiple gases and ability to detect cumulative damage caused by gases, the OI-OECT is demonstrated for early warning system targeting leakage detection of two gases (NH3 and H2S). This work achieves the integrated functions of chemical gas information decode, tunable gas memory level, and selectivity of gas in a single device, which provides a promising pathway for the development of future artificial olfactory systems.
Collapse
Affiliation(s)
- Yifeng Yin
- School of Materials Science and Engineering, Tongji University, Shanghai 201804, P. R. China
| | - Tongrui Sun
- School of Materials Science and Engineering, Tongji University, Shanghai 201804, P. R. China
| | - Lu Wang
- School of Materials Science and Engineering, Tongji University, Shanghai 201804, P. R. China
| | - Li Li
- School of Materials Science and Engineering, Tongji University, Shanghai 201804, P. R. China
| | - Pu Guo
- School of Materials Science and Engineering, Tongji University, Shanghai 201804, P. R. China
| | - Xu Liu
- School of Materials Science and Engineering, Tongji University, Shanghai 201804, P. R. China
| | - Lize Xiong
- Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Shanghai Fourth People's Hospital Affiliated to Tongji University, Tongji University, Shanghai 200434, P. R. China
| | - Guoqing Zu
- School of Materials Science and Engineering, Tongji University, Shanghai 201804, P. R. China
| | - Jia Huang
- School of Materials Science and Engineering, Tongji University, Shanghai 201804, P. R. China
- Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Shanghai Fourth People's Hospital Affiliated to Tongji University, Tongji University, Shanghai 200434, P. R. China
| |
Collapse
|
166
|
Marcatti M, Tumurbaatar B, Borghi M, Guptarak J, Zhang WR, Krishnan B, Kayed R, Fracassi A, Taglialatela G. Inhibition of Calcineurin with FK506 Reduces Tau Levels and Attenuates Synaptic Impairment Driven by Tau Oligomers in the Hippocampus of Male Mouse Models. Int J Mol Sci 2024; 25:9092. [PMID: 39201779 PMCID: PMC11354963 DOI: 10.3390/ijms25169092] [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: 07/11/2024] [Revised: 08/13/2024] [Accepted: 08/15/2024] [Indexed: 09/03/2024] Open
Abstract
Alzheimer's disease (AD) is the most common age-associated neurodegenerative disorder, characterized by progressive cognitive decline, memory impairment, and structural brain changes, primarily involving Aβ plaques and neurofibrillary tangles of hyperphosphorylated tau protein. Recent research highlights the significance of smaller Aβ and Tau oligomeric aggregates (AβO and TauO, respectively) in synaptic dysfunction and disease progression. Calcineurin (CaN), a key calcium/calmodulin-dependent player in regulating synaptic function in the central nervous system (CNS) is implicated in mediating detrimental effects of AβO on synapses and memory function in AD. This study aims to investigate the specific impact of CaN on both exogenous and endogenous TauO through the acute and chronic inhibition of CaN. We previously demonstrated the protective effect against AD of the immunosuppressant CaN inhibitor, FK506, but its influence on TauO remains unclear. In this study, we explored the short-term effects of acute CaN inhibition on TauO phosphorylation and TauO-induced memory deficits and synaptic dysfunction. Mice received FK506 post-TauO intracerebroventricular injection and TauO levels and phosphorylation were assessed, examining their impact on CaN and GSK-3β. The study investigated FK506 preventive/reversal effects on TauO-induced clustering of CaN and GSK-3β. Memory and synaptic function in TauO-injected mice were evaluated with/without FK506. Chronic FK506 treatment in 3xTgAD mice explored its influence on CaN, Aβ, and Tau levels. This study underscores the significant influence of CaN inhibition on TauO and associated AD pathology, suggesting therapeutic potential in targeting CaN for addressing various aspects of AD onset and progression. These findings provide valuable insights for potential interventions in AD, emphasizing the need for further exploration of CaN-targeted strategies.
Collapse
|
167
|
Islam R, Shi Y, de Oliveira Silva GV, Sachdev M, Miao GX. Volatile and Nonvolatile Programmable Iontronic Memristor with Lithium Imbued TiO x for Neuromorphic Computing Applications. ACS NANO 2024; 18:22045-22054. [PMID: 39110089 PMCID: PMC11342358 DOI: 10.1021/acsnano.4c05137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 07/13/2024] [Accepted: 07/24/2024] [Indexed: 08/21/2024]
Abstract
We demonstrate a lithium (Li) imbued TiOx iontronic device that exhibits synapse-like short-term plasticity behavior without requiring a forming process beforehand or a compliance current during switching. A solid-state electrolyte lithium phosphorus oxynitride (LiPON) behaves as the ion source, and the embedding and releasing of Li ions inside the cathodic like TiOx renders volatile conductance responses from the device and offers a natural platform for hardware simulating neuron functionalities. Besides, these devices possess high uniformity and great endurance as no conductive filaments are present. Different short-term pulse-based phenomena, including paired pulse facilitation, post-tetanic potentiation, and spike rate-dependent plasticity, were observed with self-relaxation characteristics. Based on the voltage excitation period, the time scale of the volatile memory can be tuned. Temperature measurement reveals the ion displacement-induced conductance channels become frozen below 220 K. In addition, the volatile analog devices can be configured into nonvolatile memory units with multibit storage capabilities after an electroforming process. Therefore, on the same platform, we can configure volatile units as nonlinear dynamic reservoirs for performing neuromorphic training and the nonvolatile units as the weight storage layer. We proceed to use voice recognition as an example with the tunable time constant relationship and obtain 94.4% accuracy with a minimal training data set. Thus, this iontronic platform can effectively process and update temporal information for reservoir and neuromorphic computing paradigms.
Collapse
Affiliation(s)
- Rabiul Islam
- Department
of Electrical and Computer Engineering, University of Waterloo, Waterloo N2L 3G1, Ontario, Canada
- Institute
for Quantum Computing, University of Waterloo, Waterloo N2L 3G1, Ontario, Canada
| | - Yu Shi
- Department
of Electrical and Computer Engineering, University of Waterloo, Waterloo N2L 3G1, Ontario, Canada
- Institute
for Quantum Computing, University of Waterloo, Waterloo N2L 3G1, Ontario, Canada
| | - Gabriel Vinicius de Oliveira Silva
- Department
of Electrical and Computer Engineering, University of Waterloo, Waterloo N2L 3G1, Ontario, Canada
- Institute
for Quantum Computing, University of Waterloo, Waterloo N2L 3G1, Ontario, Canada
| | - Manoj Sachdev
- Department
of Electrical and Computer Engineering, University of Waterloo, Waterloo N2L 3G1, Ontario, Canada
| | - Guo-Xing Miao
- Department
of Electrical and Computer Engineering, University of Waterloo, Waterloo N2L 3G1, Ontario, Canada
- Institute
for Quantum Computing, University of Waterloo, Waterloo N2L 3G1, Ontario, Canada
| |
Collapse
|
168
|
López-García S, López-Merino E, Fernández-Rodrigo A, Zamorano-González P, Gutiérrez-Eisman S, Jiménez-Sánchez R, Esteban JA. PI3K couples long-term synaptic potentiation with cofilin recruitment and actin polymerization in dendritic spines via its regulatory subunit p85α. Cell Mol Life Sci 2024; 81:358. [PMID: 39158722 PMCID: PMC11335278 DOI: 10.1007/s00018-024-05394-x] [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: 01/09/2024] [Revised: 07/16/2024] [Accepted: 08/02/2024] [Indexed: 08/20/2024]
Abstract
Long-term synaptic plasticity is typically associated with morphological changes in synaptic connections. However, the molecular mechanisms coupling functional and structural aspects of synaptic plasticity are still poorly defined. The catalytic activity of type I phosphoinositide-3-kinase (PI3K) is required for specific forms of synaptic plasticity, such as NMDA receptor-dependent long-term potentiation (LTP) and mGluR-dependent long-term depression (LTD). On the other hand, PI3K signaling has been linked to neuronal growth and synapse formation. Consequently, PI3Ks are promising candidates to coordinate changes in synaptic strength with structural remodeling of synapses. To investigate this issue, we targeted individual regulatory subunits of type I PI3Ks in hippocampal neurons and employed a combination of electrophysiological, biochemical and imaging techniques to assess their role in synaptic plasticity. We found that a particular regulatory isoform, p85α, is selectively required for LTP. This specificity is based on its BH domain, which engages the small GTPases Rac1 and Cdc42, critical regulators of the actin cytoskeleton. Moreover, cofilin, a key regulator of actin dynamics that accumulates in dendritic spines after LTP induction, failed to do so in the absence of p85α or when its BH domain was overexpressed as a dominant negative construct. Finally, in agreement with this convergence on actin regulatory mechanisms, the presence of p85α in the PI3K complex determined the extent of actin polymerization in dendritic spines during LTP. Therefore, this study reveals a molecular mechanism linking structural and functional synaptic plasticity through the coordinate action of PI3K catalytic activity and a specific isoform of the regulatory subunits.
Collapse
Affiliation(s)
- Sergio López-García
- Department of Molecular Neuropathology, Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Madrid, Spain
| | - Esperanza López-Merino
- Department of Molecular Neuropathology, Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Madrid, Spain
| | - Alba Fernández-Rodrigo
- Department of Molecular Neuropathology, Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Madrid, Spain
| | - Pablo Zamorano-González
- Department of Molecular Neuropathology, Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Madrid, Spain
- Current address: Universidad de Málaga, Málaga, Spain
| | - Silvia Gutiérrez-Eisman
- Department of Molecular Neuropathology, Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Madrid, Spain
| | - Raquel Jiménez-Sánchez
- Department of Molecular Neuropathology, Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Madrid, Spain
| | - José A Esteban
- Department of Molecular Neuropathology, Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Madrid, Spain.
| |
Collapse
|
169
|
Miranda-Riestra A, Cercós MG, Trueta C, Oikawa-Sala J, Argueta J, Constantino-Jonapa LA, Cruz-Garduño R, Benítez-King G, Estrada-Reyes R. Participation of Ca 2+-Calmodulin-Dependent Protein Kinase II in the Antidepressant-Like Effects of Melatonin. Mol Pharmacol 2024; 106:107-116. [PMID: 39079719 DOI: 10.1124/molpharm.124.000890] [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: 02/08/2024] [Accepted: 06/26/2024] [Indexed: 08/18/2024] Open
Abstract
Melatonin (N-acetyl-5-methoxytryptamine) is an indoleamine secreted by the pineal gland during the dark phase of the photoperiod. Its main function is the synchronization of different body rhythms with the dark-light cycle. Research on melatonin has significantly advanced since its discovery and we now know that it has considerable significance in various physiological processes, including immunity, aging, and reproduction. Moreover, in recent years evidence of the pharmacological possibilities of melatonin has increased. Indoleamine, on the other hand, has antidepressant-like effects in rodents, which may be mediated by the activation of calcium-calmodulin-dependent kinase II (CaMKII) and are also related to the regulation of neuroplasticity processes, including neurogenesis, synaptic maintenance, and long-term potentiation. Remarkably, patients with major depression show decreased levels of circulating melatonin in plasma. This review presents evidence of the antidepressant-like effects of melatonin in preclinical models and the participation of CaMKII in these actions. CaMKII's role in cognition and memory processes, which are altered in depressive states, are part of the review, and the effects of melatonin in these processes are also reviewed. Furthermore, participation of CaMKII on structural and synaptic plasticity and the effects of melatonin are also described. Finally, the advantages of using melatonin in combination with other antidepressants such as ketamine for neuroplasticity are described. Evidence supports that CaMKII is activated by melatonin and downstream melatonin receptors and may be the common effector in the synergistic effects of melatonin with other antidepressants. SIGNIFICANCE STATEMENT: This review compiled evidence supporting that melatonin causes antidepressant-like effects in mice through calmodulin kinase II stimulation of downstream melatonin receptors as well as the participation of this enzyme in neuroplasticity, memory, and cognition. Finally, we describe evidence about the effectiveness of antidepressant-like effects of melatonin in combination with ketamine.
Collapse
Affiliation(s)
- Armida Miranda-Riestra
- Laboratorio de Neurofarmacología (A.M.-R., J.O.-S., J.A., L.A.C.-J., G.B.-K.), Departamento de Neurofisiología, Dirección de Investigaciones en Neurociencias (M.G.C., C.T., R.C.-G.), and Laboratorio de Fitofarmacología, Dirección de Investigaciones en Neurociencias (R.E.-R.), Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Mexico City, Mexico
| | - Montserrat G Cercós
- Laboratorio de Neurofarmacología (A.M.-R., J.O.-S., J.A., L.A.C.-J., G.B.-K.), Departamento de Neurofisiología, Dirección de Investigaciones en Neurociencias (M.G.C., C.T., R.C.-G.), and Laboratorio de Fitofarmacología, Dirección de Investigaciones en Neurociencias (R.E.-R.), Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Mexico City, Mexico
| | - Citlali Trueta
- Laboratorio de Neurofarmacología (A.M.-R., J.O.-S., J.A., L.A.C.-J., G.B.-K.), Departamento de Neurofisiología, Dirección de Investigaciones en Neurociencias (M.G.C., C.T., R.C.-G.), and Laboratorio de Fitofarmacología, Dirección de Investigaciones en Neurociencias (R.E.-R.), Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Mexico City, Mexico
| | - Julián Oikawa-Sala
- Laboratorio de Neurofarmacología (A.M.-R., J.O.-S., J.A., L.A.C.-J., G.B.-K.), Departamento de Neurofisiología, Dirección de Investigaciones en Neurociencias (M.G.C., C.T., R.C.-G.), and Laboratorio de Fitofarmacología, Dirección de Investigaciones en Neurociencias (R.E.-R.), Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Mexico City, Mexico
| | - Jesús Argueta
- Laboratorio de Neurofarmacología (A.M.-R., J.O.-S., J.A., L.A.C.-J., G.B.-K.), Departamento de Neurofisiología, Dirección de Investigaciones en Neurociencias (M.G.C., C.T., R.C.-G.), and Laboratorio de Fitofarmacología, Dirección de Investigaciones en Neurociencias (R.E.-R.), Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Mexico City, Mexico
| | - Luis A Constantino-Jonapa
- Laboratorio de Neurofarmacología (A.M.-R., J.O.-S., J.A., L.A.C.-J., G.B.-K.), Departamento de Neurofisiología, Dirección de Investigaciones en Neurociencias (M.G.C., C.T., R.C.-G.), and Laboratorio de Fitofarmacología, Dirección de Investigaciones en Neurociencias (R.E.-R.), Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Mexico City, Mexico
| | - Ricardo Cruz-Garduño
- Laboratorio de Neurofarmacología (A.M.-R., J.O.-S., J.A., L.A.C.-J., G.B.-K.), Departamento de Neurofisiología, Dirección de Investigaciones en Neurociencias (M.G.C., C.T., R.C.-G.), and Laboratorio de Fitofarmacología, Dirección de Investigaciones en Neurociencias (R.E.-R.), Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Mexico City, Mexico
| | - Gloria Benítez-King
- Laboratorio de Neurofarmacología (A.M.-R., J.O.-S., J.A., L.A.C.-J., G.B.-K.), Departamento de Neurofisiología, Dirección de Investigaciones en Neurociencias (M.G.C., C.T., R.C.-G.), and Laboratorio de Fitofarmacología, Dirección de Investigaciones en Neurociencias (R.E.-R.), Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Mexico City, Mexico
| | - Rosa Estrada-Reyes
- Laboratorio de Neurofarmacología (A.M.-R., J.O.-S., J.A., L.A.C.-J., G.B.-K.), Departamento de Neurofisiología, Dirección de Investigaciones en Neurociencias (M.G.C., C.T., R.C.-G.), and Laboratorio de Fitofarmacología, Dirección de Investigaciones en Neurociencias (R.E.-R.), Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Mexico City, Mexico
| |
Collapse
|
170
|
Ju D, Kim S. Volatile tin oxide memristor for neuromorphic computing. iScience 2024; 27:110479. [PMID: 39129832 PMCID: PMC11315111 DOI: 10.1016/j.isci.2024.110479] [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: 05/03/2024] [Revised: 07/03/2024] [Accepted: 07/06/2024] [Indexed: 08/13/2024] Open
Abstract
The rise of neuromorphic systems has addressed the shortcomings of current computing architectures, especially regarding energy efficiency and scalability. These systems use cutting-edge technologies such as Pt/SnOx/TiN memristors, which efficiently mimic synaptic behavior and provide potential solutions to modern computing challenges. Moreover, their unipolar resistive switching ability enables precise modulation of the synaptic weights, facilitating energy-efficient parallel processing that is similar to biological synapses. Additionally, memristors' spike-rate-dependent plasticity enhances the adaptability of neural circuits, offering promising applications in intelligent computing. Integrating memristors into edge computing architectures further highlights their importance in tackling the security and efficiency issues associated with conventional cloud computing models.
Collapse
Affiliation(s)
- Dongyeol Ju
- Division of Electronics and Electrical Engineering, Dongguk University, Seoul 04620, Republic of Korea
| | - Sungjun Kim
- Division of Electronics and Electrical Engineering, Dongguk University, Seoul 04620, Republic of Korea
| |
Collapse
|
171
|
Wong VC, Houlihan PR, Liu H, Walpita D, DeSantis MC, Liu Z, O'Shea EK. Plasticity-induced actin polymerization in the dendritic shaft regulates intracellular AMPA receptor trafficking. eLife 2024; 13:e80622. [PMID: 39146380 PMCID: PMC11326776 DOI: 10.7554/elife.80622] [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: 05/27/2022] [Accepted: 07/09/2024] [Indexed: 08/17/2024] Open
Abstract
AMPA-type receptors (AMPARs) are rapidly inserted into synapses undergoing plasticity to increase synaptic transmission, but it is not fully understood if and how AMPAR-containing vesicles are selectively trafficked to these synapses. Here, we developed a strategy to label AMPAR GluA1 subunits expressed from their endogenous loci in cultured rat hippocampal neurons and characterized the motion of GluA1-containing vesicles using single-particle tracking and mathematical modeling. We find that GluA1-containing vesicles are confined and concentrated near sites of stimulation-induced structural plasticity. We show that confinement is mediated by actin polymerization, which hinders the active transport of GluA1-containing vesicles along the length of the dendritic shaft by modulating the rheological properties of the cytoplasm. Actin polymerization also facilitates myosin-mediated transport of GluA1-containing vesicles to exocytic sites. We conclude that neurons utilize F-actin to increase vesicular GluA1 reservoirs and promote exocytosis proximal to the sites of synaptic activity.
Collapse
Affiliation(s)
- Victor C Wong
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, United States
| | - Patrick R Houlihan
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, United States
| | - Hui Liu
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, United States
| | - Deepika Walpita
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, United States
| | - Michael C DeSantis
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, United States
| | - Zhe Liu
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, United States
| | - Erin K O'Shea
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, United States
| |
Collapse
|
172
|
Weilenmann C, Ziogas AN, Zellweger T, Portner K, Mladenović M, Kaniselvan M, Moraitis T, Luisier M, Emboras A. Single neuromorphic memristor closely emulates multiple synaptic mechanisms for energy efficient neural networks. Nat Commun 2024; 15:6898. [PMID: 39138160 PMCID: PMC11322324 DOI: 10.1038/s41467-024-51093-3] [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: 01/30/2024] [Accepted: 07/27/2024] [Indexed: 08/15/2024] Open
Abstract
Biological neural networks do not only include long-term memory and weight multiplication capabilities, as commonly assumed in artificial neural networks, but also more complex functions such as short-term memory, short-term plasticity, and meta-plasticity - all collocated within each synapse. Here, we demonstrate memristive nano-devices based on SrTiO3 that inherently emulate all these synaptic functions. These memristors operate in a non-filamentary, low conductance regime, which enables stable and energy efficient operation. They can act as multi-functional hardware synapses in a class of bio-inspired deep neural networks (DNN) that make use of both long- and short-term synaptic dynamics and are capable of meta-learning or learning-to-learn. The resulting bio-inspired DNN is then trained to play the video game Atari Pong, a complex reinforcement learning task in a dynamic environment. Our analysis shows that the energy consumption of the DNN with multi-functional memristive synapses decreases by about two orders of magnitude as compared to a pure GPU implementation. Based on this finding, we infer that memristive devices with a better emulation of the synaptic functionalities do not only broaden the applicability of neuromorphic computing, but could also improve the performance and energy costs of certain artificial intelligence applications.
Collapse
Affiliation(s)
| | | | - Till Zellweger
- Integrated Systems Laboratory, ETH Zurich, Zurich, Switzerland
| | - Kevin Portner
- Integrated Systems Laboratory, ETH Zurich, Zurich, Switzerland
| | | | | | | | - Mathieu Luisier
- Integrated Systems Laboratory, ETH Zurich, Zurich, Switzerland
| | | |
Collapse
|
173
|
Smith G, McCoy K, Di Prisco GV, Kuklish A, Grant E, Bhat M, Patel S, Mackie K, Atwood B, Kalinovsky A. Deletion of endocannabinoid synthesizing enzyme DAGLα from cerebellar Purkinje cells decreases social preference and elevates anxiety. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.08.607068. [PMID: 39211155 PMCID: PMC11361171 DOI: 10.1101/2024.08.08.607068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
The endocannabinoid (eCB) signaling system is robustly expressed in the cerebellum starting from the embryonic developmental stages to adulthood. There it plays a key role in regulating cerebellar synaptic plasticity and excitability, suggesting that impaired eCB signaling will lead to deficits in cerebellar adjustments of ongoing behaviors and cerebellar learning. Indeed, human mutations in DAGLα are associated with neurodevelopmental disorders. In this study, we show that selective deletion of the eCB synthesizing enzyme diacylglycerol lipase alpha (Daglα) from mouse cerebellar Purkinje cells (PCs) alters motor and social behaviors, disrupts short-term synaptic plasticity in both excitatory and inhibitory synapses, and reduces Purkinje cell activity during social exploration. Our results provide the first evidence for cerebellar-specific eCB regulation of social behaviors and implicate eCB regulation of synaptic plasticity and PC activity as the neural substrates contributing to these deficits. Abstract Figure
Collapse
|
174
|
Vallejo-Mancero B, Madrenas J, Zapata M. Real-time execution of SNN models with synaptic plasticity for handwritten digit recognition on SIMD hardware. Front Neurosci 2024; 18:1425861. [PMID: 39165339 PMCID: PMC11333227 DOI: 10.3389/fnins.2024.1425861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 07/22/2024] [Indexed: 08/22/2024] Open
Abstract
Recent advancements in neuromorphic computing have led to the development of hardware architectures inspired by Spiking Neural Networks (SNNs) to emulate the efficiency and parallel processing capabilities of the human brain. This work focuses on testing the HEENS architecture, specifically designed for high parallel processing and biological realism in SNN emulation, implemented on a ZYNQ family FPGA. The study applies this architecture to the classification of digits using the well-known MNIST database. The image resolutions were adjusted to match HEENS' processing capacity. Results were compared with existing work, demonstrating HEENS' performance comparable to other solutions. This study highlights the importance of balancing accuracy and efficiency in the execution of applications. HEENS offers a flexible solution for SNN emulation, allowing for the implementation of programmable neural and synaptic models. It encourages the exploration of novel algorithms and network architectures, providing an alternative for real-time processing with efficient energy consumption.
Collapse
Affiliation(s)
| | - Jordi Madrenas
- Department of Electronic Engineering, Universitat Politècnica de Catalunya, Barcelona, Spain
| | - Mireya Zapata
- Centro de Investigación en Mecatrónica y Sistemas Interactivos—MIST, Universidad Indoamérica, Quito, Ecuador
| |
Collapse
|
175
|
Bitar L, Puig B, Oertner TG, Dénes Á, Magnus T. Changes in Neuroimmunological Synapses During Cerebral Ischemia. Transl Stroke Res 2024:10.1007/s12975-024-01286-1. [PMID: 39103660 DOI: 10.1007/s12975-024-01286-1] [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: 03/28/2024] [Revised: 06/24/2024] [Accepted: 07/25/2024] [Indexed: 08/07/2024]
Abstract
The direct interplay between the immune and nervous systems is now well established. Within the brain, these interactions take place between neurons and resident glial cells, i.e., microglia and astrocytes, or infiltrating immune cells, influenced by systemic factors. A special form of physical cell-cell interactions is the so-called "neuroimmunological (NI) synapse." There is compelling evidence that the same signaling pathways that regulate inflammatory responses to injury or ischemia also play potent roles in brain development, plasticity, and function. Proper synaptic wiring is as important during development as it is during disease states, as it is necessary for activity-dependent refinement of neuronal circuits. Since the process of forming synaptic connections in the brain is highly dynamic, with constant changes in strength and connectivity, the immune component is perfectly suited for the regulatory task as it is in constant turnover. Many cellular and molecular players in this interaction remain to be uncovered, especially in pathological states. In this review, we discuss and propose possible communication hubs between components of the adaptive and innate immune systems and the synaptic element in ischemic stroke pathology.
Collapse
Affiliation(s)
- Lynn Bitar
- Neurology Department, Experimental Research in Stroke and Inflammation (ERSI) Group, University Medical Center Hamburg-Eppendorf (UKE), Martinistraße, 52, Hamburg, 20246, Germany
| | - Berta Puig
- Neurology Department, Experimental Research in Stroke and Inflammation (ERSI) Group, University Medical Center Hamburg-Eppendorf (UKE), Martinistraße, 52, Hamburg, 20246, Germany
| | - Thomas G Oertner
- Institute for Synaptic Physiology, Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ádám Dénes
- "Momentum" Laboratory of Neuroimmunology, Institute of Experimental Medicine, Budapest, Hungary
| | - Tim Magnus
- Neurology Department, Experimental Research in Stroke and Inflammation (ERSI) Group, University Medical Center Hamburg-Eppendorf (UKE), Martinistraße, 52, Hamburg, 20246, Germany.
| |
Collapse
|
176
|
Azarfarin M, Ghadiri T, Dadkhah M, Sahab-Negah S. The interaction between cannabinoids and long-term synaptic plasticity: A survey on memory formation and underlying mechanisms. Cell Biochem Funct 2024; 42:e4100. [PMID: 39090824 DOI: 10.1002/cbf.4100] [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: 04/12/2024] [Revised: 07/05/2024] [Accepted: 07/12/2024] [Indexed: 08/04/2024]
Abstract
Synaptic plasticity, including long-term potentiation (LTP) and long-term depression (LTD), is an essential phenomenon in memory formation as well as maintenance along with many other cognitive functions, such as those needed for coping with external stimuli. Synaptic plasticity consists of gradual changes in the biochemistry and morphology of pre- and postsynaptic neurons, particularly in the hippocampus. Consuming marijuana as a primary source of exocannabinoids immediately impairs attention and working memory-related tasks. Evidence regarding the effects of cannabinoids on LTP and memory is contradictory. While cannabinoids can affect a variety of specific cannabinoid receptors (CBRs) and nonspecific receptors throughout the body and brain, they exert miscellaneous systemic and local cerebral effects. Given the increasing use of cannabis, mainly among the young population, plus its potential adverse long-term effects on learning and memory processes, it could be a future global health challenge. Indeed, the impact of cannabinoids on memory is multifactorial and depends on the dosage, timing, formula, and route of consumption, plus the background complex interaction of the endocannabinoids system with other cerebral networks. Herein, we review how exogenously administrated organic cannabinoids, CBRs agonists or antagonists, and endocannabinoids can affect LTP and synaptic plasticity through various receptors in interaction with other cerebral pathways and primary neurotransmitters.
Collapse
Affiliation(s)
- Maryam Azarfarin
- Department of Neuroscience,Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
- Neurosciences Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Tahereh Ghadiri
- Department of Neuroscience,Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Masoomeh Dadkhah
- Neurosciences Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sajad Sahab-Negah
- Pharmaceutical Sciences Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
- Department of Neuroscience, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
177
|
Sayehmiri F, Motamedi F, Batool Z, Naderi N, Shaerzadeh F, Zoghi A, Rezaei O, Khodagholi F, Pourbadie HG. Mitochondrial plasticity and synaptic plasticity crosstalk; in health and Alzheimer's disease. CNS Neurosci Ther 2024; 30:e14897. [PMID: 39097920 PMCID: PMC11298206 DOI: 10.1111/cns.14897] [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: 01/05/2024] [Revised: 06/19/2024] [Accepted: 07/18/2024] [Indexed: 08/06/2024] Open
Abstract
Synaptic plasticity is believed to underlie the cellular and molecular basis of memory formation. Mitochondria are one of the main organelles involved in metabolism and energy maintenance as plastic organelles that change morphologically and functionally in response to cellular needs and regulate synaptic function and plasticity through multiple mechanisms, including ATP generation, calcium homeostasis, and biogenesis. An increased neuronal activity enhances synaptic efficiency, during which mitochondria's spatial distribution and morphology change significantly. These organelles build up in the pre-and postsynaptic zones to produce ATP, which is necessary for several synaptic processes like neurotransmitter release and recycling. Mitochondria also regulate calcium homeostasis by buffering intracellular calcium, which ensures proper synaptic activity. Furthermore, mitochondria in the presynaptic terminal have distinct morphological properties compared to dendritic or postsynaptic mitochondria. This specialization enables precise control of synaptic activity and plasticity. Mitochondrial dysfunction has been linked to synaptic failure in many neurodegenerative disorders, like Alzheimer's disease (AD). In AD, malfunctioning mitochondria cause delays in synaptic vesicle release and recycling, ionic gradient imbalances, and mostly synaptic failure. This review emphasizes mitochondrial plasticity's contribution to synaptic function. It also explores the profound effect of mitochondrial malfunction on neurodegenerative disorders, focusing on AD, and provides an overview of how they sustain cellular health under normal conditions and how their malfunction contributes to neurodegenerative diseases, highlighting their potential as a therapeutic target for such conditions.
Collapse
Affiliation(s)
- Fatemeh Sayehmiri
- Neuroscience Research Center, Faculty of MedicineShahid Beheshti University of Medical SciencesTehranIran
| | - Fereshteh Motamedi
- Neuroscience Research Center, Faculty of MedicineShahid Beheshti University of Medical SciencesTehranIran
- Faculty of MedicineShahid Beheshti University of Medical SciencesTehranIran
| | - Zehra Batool
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological SciencesUniversity of KarachiKarachiPakistan
| | - Nima Naderi
- Department of Pharmacology and Toxicology, Faculty of PharmacyShahid Beheshti University of Medical SciencesTehranIran
| | | | - Anahita Zoghi
- Department of Neurology, Loghman Hakim HospitalShahid Beheshti University of Medical SciencesTehranIran
| | - Omidvar Rezaei
- Skull Base Research CenterLoghman Hakim Hospital, Shahid Beheshti University of Medical SciencesTehranIran
| | - Fariba Khodagholi
- Neuroscience Research Center, Faculty of MedicineShahid Beheshti University of Medical SciencesTehranIran
| | | |
Collapse
|
178
|
Herbet G, Duffau H, Mandonnet E. Predictors of cognition after glioma surgery: connectotomy, structure-function phenotype, plasticity. Brain 2024; 147:2621-2635. [PMID: 38573324 DOI: 10.1093/brain/awae093] [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: 12/02/2023] [Revised: 02/19/2024] [Accepted: 03/09/2024] [Indexed: 04/05/2024] Open
Abstract
Determining preoperatively the maximal extent of resection that would preserve cognitive functions is the core challenge of brain tumour surgery. Over the past decade, the methodological framework to achieve this goal has been thoroughly renewed: the population-level topographically-focused voxel-based lesion-symptom mapping has been progressively overshadowed by machine learning (ML) algorithmics, in which the problem is framed as predicting cognitive outcomes in a patient-specific manner from a typically large set of variables. However, the choice of these predictors is of utmost importance, as they should be both informative and parsimonious. In this perspective, we first introduce the concept of connectotomy: instead of parameterizing resection topography through the status (intact/resected) of a huge number of voxels (or parcels) paving the whole brain in the Cartesian 3D-space, the connectotomy models the resection in the connectivity space, by computing a handful number of networks disconnection indices, measuring how the structural connectivity sustaining each network of interest was hit by the resection. This connectivity-informed reduction of dimensionality is a necessary step for efficiently implementing ML tools, given the relatively small number of patient-examples in available training datasets. We further argue that two other major sources of interindividual variability must be considered to improve the accuracy with which outcomes are predicted: the underlying structure-function phenotype and neuroplasticity, for which we provide an in-depth review and propose new ways of determining relevant predictors. We finally discuss the benefits of our approach for precision surgery of glioma.
Collapse
Affiliation(s)
- Guillaume Herbet
- Department of Neurosurgery, Gui de Chauliac Hospital, Montpellier 34090, France
- Praxiling lab, UMR5267 CNRS & Paul Valéry University, Montpellier 34090, France
- Department of Medicine, University of Montpellier, Montpellier 34090, France
- Institut Universitaire de France, Paris 75000, France
| | - Hugues Duffau
- Department of Neurosurgery, Gui de Chauliac Hospital, Montpellier 34090, France
- Department of Medicine, University of Montpellier, Montpellier 34090, France
- Team 'Plasticity of Central Nervous System, Stem Cells and Glial Tumors', U1191 Laboratory, Institute of Functional Genomics, National Institute for Health and Medical Research (INSERM), University of Montpellier, Montpellier 34000, France
| | - Emmanuel Mandonnet
- Department of Neurosurgery, Lariboisière Hospital, AP-HP, Paris 75010, France
- Frontlab, CNRS UMR 7225, INSERM U1127, Paris Brain Institute (ICM), Paris 75013, France
- Université de Paris Cité, UFR de médecine, Paris 75005, France
| |
Collapse
|
179
|
Stavgiannoudaki I, Goulielmaki E, Garinis GA. Broken strands, broken minds: Exploring the nexus of DNA damage and neurodegeneration. DNA Repair (Amst) 2024; 140:103699. [PMID: 38852477 DOI: 10.1016/j.dnarep.2024.103699] [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: 12/15/2023] [Revised: 05/15/2024] [Accepted: 05/28/2024] [Indexed: 06/11/2024]
Abstract
Neurodegenerative disorders are primarily characterized by neuron loss progressively leading to cognitive decline and the manifestation of incurable and debilitating conditions, such as Alzheimer's, Parkinson's, and Huntington's diseases. Loss of genome maintenance causally contributes to age-related neurodegeneration, as exemplified by the premature appearance of neurodegenerative features in a growing family of human syndromes and mice harbouring inborn defects in DNA repair. Here, we discuss the relevance of persistent DNA damage, key DNA repair mechanisms and compromised genome integrity in age-related neurodegeneration highlighting the significance of investigating these connections to pave the way for the development of rationalized intervention strategies aimed at delaying the onset of neurodegenerative disorders and promoting healthy aging.
Collapse
Affiliation(s)
- Ioanna Stavgiannoudaki
- Institute of Molecular Biology and Biotechnology (IMBB), Foundation for Research and Technology-Hellas, Crete, Heraklion, Greece; Department of Biology, University of Crete, Crete, Heraklion, Greece
| | - Evi Goulielmaki
- Institute of Molecular Biology and Biotechnology (IMBB), Foundation for Research and Technology-Hellas, Crete, Heraklion, Greece
| | - George A Garinis
- Institute of Molecular Biology and Biotechnology (IMBB), Foundation for Research and Technology-Hellas, Crete, Heraklion, Greece; Department of Biology, University of Crete, Crete, Heraklion, Greece.
| |
Collapse
|
180
|
Donders Z, Skorupska IJ, Willems E, Mussen F, Broeckhoven JV, Carlier A, Schepers M, Vanmierlo T. Beyond PDE4 inhibition: A comprehensive review on downstream cAMP signaling in the central nervous system. Biomed Pharmacother 2024; 177:117009. [PMID: 38908196 DOI: 10.1016/j.biopha.2024.117009] [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: 03/28/2024] [Revised: 05/27/2024] [Accepted: 06/17/2024] [Indexed: 06/24/2024] Open
Abstract
Cyclic adenosine monophosphate (cAMP) is a key second messenger that regulates signal transduction pathways pivotal for numerous biological functions. Intracellular cAMP levels are spatiotemporally regulated by their hydrolyzing enzymes called phosphodiesterases (PDEs). It has been shown that increased cAMP levels in the central nervous system (CNS) promote neuroplasticity, neurotransmission, neuronal survival, and myelination while suppressing neuroinflammation. Thus, elevating cAMP levels through PDE inhibition provides a therapeutic approach for multiple CNS disorders, including multiple sclerosis, stroke, spinal cord injury, amyotrophic lateral sclerosis, traumatic brain injury, and Alzheimer's disease. In particular, inhibition of the cAMP-specific PDE4 subfamily is widely studied because of its high expression in the CNS. So far, the clinical translation of full PDE4 inhibitors has been hampered because of dose-limiting side effects. Hence, focusing on signaling cascades downstream activated upon PDE4 inhibition presents a promising strategy, offering novel and pharmacologically safe targets for treating CNS disorders. Yet, the underlying downstream signaling pathways activated upon PDE(4) inhibition remain partially elusive. This review provides a comprehensive overview of the existing knowledge regarding downstream mediators of cAMP signaling induced by PDE4 inhibition or cAMP stimulators. Furthermore, we highlight existing gaps and future perspectives that may incentivize additional downstream research concerning PDE(4) inhibition, thereby providing novel therapeutic approaches for CNS disorders.
Collapse
Affiliation(s)
- Zoë Donders
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht 6229ER, the Netherlands; Department of Neuroscience, Biomedical Research Institute, Faculty of Medicine and Life Sciences, Hasselt University, Hasselt 3500, Belgium
| | - Iga Joanna Skorupska
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht 6229ER, the Netherlands; Department of Neuroscience, Biomedical Research Institute, Faculty of Medicine and Life Sciences, Hasselt University, Hasselt 3500, Belgium; Department of Cell Biology-Inspired Tissue Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht 6629ER, the Netherlands
| | - Emily Willems
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht 6229ER, the Netherlands; Department of Neuroscience, Biomedical Research Institute, Faculty of Medicine and Life Sciences, Hasselt University, Hasselt 3500, Belgium
| | - Femke Mussen
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht 6229ER, the Netherlands; Department of Neuroscience, Biomedical Research Institute, Faculty of Medicine and Life Sciences, Hasselt University, Hasselt 3500, Belgium; Department of Immunology and Infection, Biomedical Research Institute, Faculty of Medicine and Life Sciences, Hasselt University, Hasselt 3500, Belgium
| | - Jana Van Broeckhoven
- Department of Immunology and Infection, Biomedical Research Institute, Faculty of Medicine and Life Sciences, Hasselt University, Hasselt 3500, Belgium; University MS Centre (UMSC) Hasselt - Pelt, Belgium
| | - Aurélie Carlier
- Department of Cell Biology-Inspired Tissue Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht 6629ER, the Netherlands
| | - Melissa Schepers
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht 6229ER, the Netherlands; Department of Neuroscience, Biomedical Research Institute, Faculty of Medicine and Life Sciences, Hasselt University, Hasselt 3500, Belgium; University MS Centre (UMSC) Hasselt - Pelt, Belgium
| | - Tim Vanmierlo
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht 6229ER, the Netherlands; Department of Neuroscience, Biomedical Research Institute, Faculty of Medicine and Life Sciences, Hasselt University, Hasselt 3500, Belgium; University MS Centre (UMSC) Hasselt - Pelt, Belgium.
| |
Collapse
|
181
|
Chujan S, Cholpraipimolrat W, Satayavivad J. Integrated Transcriptomics and Network Analysis Identified Altered Neural Mechanisms in Frontal Aging Brain-Associated Alzheimer's Disease. Biochem Genet 2024; 62:2382-2398. [PMID: 37934339 DOI: 10.1007/s10528-023-10549-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Accepted: 10/10/2023] [Indexed: 11/08/2023]
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disease. The late stage of AD typically develops after 60 years of age and AD pathogenesis can be detected predominately in the frontal lobe, which is responsible for memory. Multiple alterations in cellular mechanisms have been associated with AD, but there is no clear information on AD pathogenesis during brain aging. This study aimed to explore the differentially expressed genes (DEGs) in the frontal lobe of aging brains and to identify shared crucial mechanisms in the aging brain linked to AD pathogenesis. Three datasets were downloaded from the Gene Expression Omnibus (GEO). Biological function analysis was performed by DAVID and KEGG databases. An AD patient's cohort (GSE150696) was collected for verification of the enriched pathway. The results demonstrated that multiple neurochemical synapsis and regulation of the cytoskeleton are linked to AD pathogenesis during aging. Taken together, this study contributes to our further understanding of neural alterations during aging in AD that could be used to develop therapeutics for early intervention to prevent or slow progression.
Collapse
Affiliation(s)
- Suthipong Chujan
- Laboratory of Pharmacology, Chulabhorn Research Institute, Bangkok, 10210, Thailand
- Center of Excellence on Environmental Health and Toxicology (EHT), OPS, MHESI, Bangkok, 10400, Thailand
| | | | - Jutamaad Satayavivad
- Laboratory of Pharmacology, Chulabhorn Research Institute, Bangkok, 10210, Thailand.
- Center of Excellence on Environmental Health and Toxicology (EHT), OPS, MHESI, Bangkok, 10400, Thailand.
| |
Collapse
|
182
|
Kim H, Kornman PT, Kweon J, Wassermann EM, Wright DL, Li J, Brown JC. Combined effects of pharmacological interventions and intermittent theta-burst stimulation on motor sequence learning. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.24.604878. [PMID: 39211172 PMCID: PMC11361068 DOI: 10.1101/2024.07.24.604878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Drugs that modulate N-methyl-D-aspartate (NMDA) or γ-Aminobutyric acid type A (GABA A ) receptors can shed light on their role in synaptic plasticity mechanisms underlying the effects of non-invasive brain stimulation. However, research on the combined effects of these drugs and exogenous stimulation on motor learning is limited. This study aimed to investigate the effects of pharmacological interventions combined with intermittent theta-burst stimulation (iTBS) on human motor learning. Nine right-handed healthy subjects (mean age ± SD: 31.56 ± 12.96 years; 6 females) participated in this double-blind crossover study. All participants were assigned to four drug conditions in a randomized order: (1) D-cycloserine (partial NMDA receptor agonist), (2) D-cycloserine + dextromethorphan (NMDA receptor agonist + antagonist), (3) lorazepam (GABA A receptor agonist), and (4) placebo (identical microcrystalline cellulose capsule). After drug intake, participants practiced the 12-item keyboard sequential task as a baseline measure. Two hours after drug intake, iTBS was administered at the primary motor cortex. Following iTBS, the retention test was performed in the same manner as the baseline measure. Our findings revealed that lorazepam combined with iTBS impaired motor learning during the retention test. Future studies are still needed for a better understanding of the mechanisms through which TMS may influence human motor learning.
Collapse
|
183
|
Costacurta JC, Bhandarkar S, Zoltowski DM, Linderman SW. Structured flexibility in recurrent neural networks via neuromodulation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.26.605315. [PMID: 39091788 PMCID: PMC11291173 DOI: 10.1101/2024.07.26.605315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/04/2024]
Abstract
The goal of theoretical neuroscience is to develop models that help us better understand biological intelligence. Such models range broadly in complexity and biological detail. For example, task-optimized recurrent neural networks (RNNs) have generated hypotheses about how the brain may perform various computations, but these models typically assume a fixed weight matrix representing the synaptic connectivity between neurons. From decades of neuroscience research, we know that synaptic weights are constantly changing, controlled in part by chemicals such as neuromodulators. In this work we explore the computational implications of synaptic gain scaling, a form of neuromodulation, using task-optimized low-rank RNNs. In our neuromodulated RNN (NM-RNN) model, a neuromodulatory subnetwork outputs a low-dimensional neuromodulatory signal that dynamically scales the low-rank recurrent weights of an output-generating RNN. In empirical experiments, we find that the structured flexibility in the NM-RNN allows it to both train and generalize with a higher degree of accuracy than low-rank RNNs on a set of canonical tasks. Additionally, via theoretical analyses we show how neuromodulatory gain scaling endows networks with gating mechanisms commonly found in artificial RNNs. We end by analyzing the low-rank dynamics of trained NM-RNNs, to show how task computations are distributed.
Collapse
Affiliation(s)
- Julia C Costacurta
- Wu Tsai Neurosciences Institute, Stanford, CA, USA
- Department of Electrical Engineering, Stanford, CA, USA
| | | | - David M Zoltowski
- Wu Tsai Neurosciences Institute, Stanford, CA, USA
- Department of Statistics, Stanford University, Stanford, CA, USA
| | - Scott W Linderman
- Wu Tsai Neurosciences Institute, Stanford, CA, USA
- Department of Statistics, Stanford University, Stanford, CA, USA
| |
Collapse
|
184
|
Yu L, Liu Y, Xia J, Feng S, Chen F. KCNH5 deletion increases autism susceptibility by regulating neuronal growth through Akt/mTOR signaling pathway. Behav Brain Res 2024; 470:115069. [PMID: 38797494 DOI: 10.1016/j.bbr.2024.115069] [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: 03/28/2024] [Revised: 05/15/2024] [Accepted: 05/23/2024] [Indexed: 05/29/2024]
Abstract
Recent clinical studies have highlighted mutations in the voltage-gated potassium channel Kv10.2 encoded by the KCNH5 gene among individuals with autism spectrum disorder (ASD). Our preliminary study found that Kv10.2 was decreased in the hippocampus of valproic acid (VPA) - induced ASD rats. Nevertheless, it is currently unclear how KCNH5 regulates autism-like features, or becomes a new target for autism treatment. We employed KCNH5 knockout (KCNH5-/-) rats and VPA - induced ASD rats in this study. Then, we used behavioral assessments, combined with electrophysiological recordings and hippocampal brain slice, to elucidate the impact of KCNH5 deletion and environmental factors on neural development and function in rats. We found that KCNH5-/- rats showed early developmental delay, neuronal overdevelopment, and abnormal electroencephalogram (EEG) signals, but did not exhibit autism-like behavior. KCNH5-/- rats exposed to VPA (KCNH5-/--VPA) exhibit even more severe autism-like behaviors and abnormal neuronal development. The absence of KCNH5 excessively enhances the activity of the Protein Kinase B (Akt)/Mechanistic Target of Rapamycin (mTOR) signaling pathway in the hippocampus of rats after exposure to VPA. Overall, our findings underscore the deficiency of KCNH5 increases the susceptibility to autism under environmental exposures, suggesting its potential utility as a target for screening and diagnosis in ASD.
Collapse
Affiliation(s)
- Lele Yu
- School of Life Sciences, Shanghai University, No. 99 Shangda Road, Shanghai 200444, PR China.
| | - Yamei Liu
- School of Life Sciences, Shanghai University, No. 99 Shangda Road, Shanghai 200444, PR China.
| | - Junyu Xia
- School of Life Sciences, Shanghai University, No. 99 Shangda Road, Shanghai 200444, PR China.
| | - Shini Feng
- School of Life Sciences, Shanghai University, No. 99 Shangda Road, Shanghai 200444, PR China.
| | - Fuxue Chen
- School of Life Sciences, Shanghai University, No. 99 Shangda Road, Shanghai 200444, PR China.
| |
Collapse
|
185
|
Dingil AE, Pribyl O. Understanding state-of-the-art situation of transport planning strategies in earthquake-prone areas by using AI-supported literature review methodology. Heliyon 2024; 10:e33645. [PMID: 39040344 PMCID: PMC11261024 DOI: 10.1016/j.heliyon.2024.e33645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 06/13/2024] [Accepted: 06/25/2024] [Indexed: 07/24/2024] Open
Abstract
Aim This review aims to explore earthquake-based transport strategies in seismic areas, providing state-of-the-art insights into the components necessary to guide urban planners and policymakers in their decision-making processes. Outputs The review provides a variety of methodologies and approaches employed for the reinforcement planning and emergency demand management to analyze and evaluate the impact of seismic events on transportation systems, in turn to develop strategies for preparedness, mitigation, response, and recovery phases. The selection of the appropriate approach depends on factors such as the specific transport system, urbanization level and type, built environment, and critical components involved. Originality and value Besides providing a distinctive illustration of the integration of transportation and seismic literature as a valuable consolidated resource, this article introduces a novel methodology named ALARM for conducting state-of-the-art reviews on any topic, incorporating AI through the utilization of large language models (LLMs) built upon transformer deep neural networks, along with indexing data structures (in this study mainly OPEN-AI DAVINCI-003 model and vector-storing index). Hence, it is of paramount significance as the first instance of implementing LLMs within academic review standards. This paves the way for the potential integration of AI and human collaboration to become a standard practice under enhanced criteria for comprehending and analyzing specific information.
Collapse
Affiliation(s)
- Ali Enes Dingil
- LAMbDA Lab. (λ), Faculty of Transportation Sciences, Czech Technical University, 110 00 Prague, Czech Republic
| | - Ondrej Pribyl
- LAMbDA Lab. (λ), Faculty of Transportation Sciences, Czech Technical University, 110 00 Prague, Czech Republic
| |
Collapse
|
186
|
Canonica T, Kidd EJ, Gibbins D, Lana-Elola E, Fisher EMC, Tybulewicz VLJ, Good M. Dissecting the contribution of human chromosome 21 syntenic regions to recognition memory processes in adult and aged mouse models of Down syndrome. Front Behav Neurosci 2024; 18:1428146. [PMID: 39050700 PMCID: PMC11266108 DOI: 10.3389/fnbeh.2024.1428146] [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: 05/05/2024] [Accepted: 06/24/2024] [Indexed: 07/27/2024] Open
Abstract
Background Trisomy of human chromosome 21 (Hsa21) results in a constellation of features known as Down syndrome (DS), the most common genetic form of intellectual disability. Hsa21 is orthologous to three regions in the mouse genome on mouse chromosome 16 (Mmu16), Mmu17 and Mmu10. We investigated genotype-phenotype relationships by assessing the contribution of these three regions to memory function and age-dependent cognitive decline, using three mouse models of DS, Dp1Tyb, Dp(17)3Yey, Dp(10)2Yey, that carry an extra copy of the Hsa21-orthologues on Mmu16, Mmu17 and Mmu10, respectively. Hypothesis Prior research on cognitive function in DS mouse models has largely focused on models with an extra copy of the Mmu16 region and relatively little is known about the effects of increased copy number on Mmu17 and Mmu10 on cognition and how this interacts with the effects of aging. As aging is is a critical contributor to cognitive and psychiatric changes in DS, we hypothesised that ageing would differentially impact memory function in Dp1Tyb, Dp(17)3Yey, and Dp(10)2Yey, models of DS. Methods Young (12-13 months and old (18-20 months mice Dp1Tyb, Dp(17)3Yey and Dp(10)2Yey mice were tested on a battery of object recognition memory test that assessed object novelty detection, novel location detection and associative object-in place memory. Following behavioral testing, hippocampal and frontal cortical tissue was analysed for expression of glutamatergic receptor proteins using standard immunoblot techniques. Results Young (12-13 months and old (18-20 months mice Dp1Tyb, Dp(17)3Yey and Dp(10)2Yey mice were tested on a battery of object recognition memory test that assessed object novelty detection, novel location detection and associative object-in place memory. Following behavioral testing, hippocampal and frontal cortical tissue was analysed for expression of glutamatergic receptor proteins using standard immunoblot techniques. Conclusion Our results show that distinct Hsa21-orthologous regions contribute differentially to cognitive dysfunction in DS mouse models and that aging interacts with triplication of Hsa21-orthologous genes on Mmu10.
Collapse
Affiliation(s)
- Tara Canonica
- School of Psychology, Cardiff University, Cardiff, United Kingdom
| | - Emma J. Kidd
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Cardiff, United Kingdom
| | | | | | - Elizabeth M. C. Fisher
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London, United Kingdom
| | | | - Mark Good
- School of Psychology, Cardiff University, Cardiff, United Kingdom
| |
Collapse
|
187
|
Morales-Vicente DA, Tahira AC, Woellner-Santos D, Amaral MS, Berzoti-Coelho MG, Verjovski-Almeida S. The Human Developing Cerebral Cortex Is Characterized by an Elevated De Novo Expression of Long Noncoding RNAs in Excitatory Neurons. Mol Biol Evol 2024; 41:msae123. [PMID: 38913688 PMCID: PMC11221658 DOI: 10.1093/molbev/msae123] [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: 11/23/2023] [Revised: 06/12/2024] [Accepted: 06/14/2024] [Indexed: 06/26/2024] Open
Abstract
The outstanding human cognitive capacities are computed in the cerebral cortex, a mammalian-specific brain region and the place of massive biological innovation. Long noncoding RNAs have emerged as gene regulatory elements with higher evolutionary turnover than mRNAs. The many long noncoding RNAs identified in neural tissues make them candidates for molecular sources of cerebral cortex evolution and disease. Here, we characterized the genomic and cellular shifts that occurred during the evolution of the long noncoding RNA repertoire expressed in the developing cerebral cortex and explored putative roles for these long noncoding RNAs in the evolution of the human brain. Using transcriptomics and comparative genomics, we comprehensively annotated the cortical transcriptomes of humans, rhesus macaques, mice, and chickens and classified human cortical long noncoding RNAs into evolutionary groups as a function of their predicted minimal ages. Long noncoding RNA evolutionary groups showed differences in expression levels, splicing efficiencies, transposable element contents, genomic distributions, and transcription factor binding to their promoters. Furthermore, older long noncoding RNAs showed preferential expression in germinative zones, outer radial glial cells, and cortical inhibitory (GABAergic) neurons. In comparison, younger long noncoding RNAs showed preferential expression in cortical excitatory (glutamatergic) neurons, were enriched in primate and human-specific gene co-expression modules, and were dysregulated in neurodevelopmental disorders. These results suggest different evolutionary routes for older and younger cortical long noncoding RNAs, highlighting old long noncoding RNAs as a possible source of molecular evolution of conserved developmental programs; conversely, we propose that the de novo expression of primate- and human-specific young long noncoding RNAs is a putative source of molecular evolution and dysfunction of cortical excitatory neurons, warranting further investigation.
Collapse
Affiliation(s)
- David A Morales-Vicente
- Laboratório de Ciclo Celular, Instituto Butantan, São Paulo, Brazil
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, Brazil
| | - Ana C Tahira
- Laboratório de Ciclo Celular, Instituto Butantan, São Paulo, Brazil
| | - Daisy Woellner-Santos
- Laboratório de Ciclo Celular, Instituto Butantan, São Paulo, Brazil
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, Brazil
| | - Murilo S Amaral
- Laboratório de Ciclo Celular, Instituto Butantan, São Paulo, Brazil
| | - Maria G Berzoti-Coelho
- Laboratório de Ciclo Celular, Instituto Butantan, São Paulo, Brazil
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, Brazil
| | - Sergio Verjovski-Almeida
- Laboratório de Ciclo Celular, Instituto Butantan, São Paulo, Brazil
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, Brazil
| |
Collapse
|
188
|
Abdel-Hay N, Kabirova M, Yaka R. A discrete subpopulation of PFC-LHb neurons govern cocaine place preference. Transl Psychiatry 2024; 14:269. [PMID: 38956048 PMCID: PMC11220025 DOI: 10.1038/s41398-024-02988-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 06/19/2024] [Accepted: 06/26/2024] [Indexed: 07/04/2024] Open
Abstract
Addiction is a complex behavioral disorder characterized by compulsive drug-seeking and drug use despite harmful consequences. The prefrontal cortex (PFC) plays a crucial role in cocaine addiction, involving decision-making, impulse control, memory, and emotional regulation. The PFC interacts with the brain's reward system, including the ventral tegmental area (VTA) and nucleus accumbens (NAc). The PFC also projects to the lateral habenula (LHb), a brain region critical for encoding negative reward and regulating the reward system. In the current study, we examined the role of PFC-LHb projections in regulating cocaine reward-related behaviors. We found that optogenetic stimulation of the PFC-LHb circuit during cocaine conditioning abolished cocaine preference without causing aversion. In addition, increased c-fos expression in LHb neurons was observed in animals that received optic stimulation during cocaine conditioning, supporting the circuit's involvement in cocaine preference regulation. Molecular analysis in animals that received optic stimulation revealed that cocaine-induced alterations in the expression of GluA1 subunit of AMPA receptor was normalized to saline levels in a region-specific manner. Moreover, GluA1 serine phosphorylation on S845 and S831 were differentially altered in LHb and VTA but not in the PFC. Together these findings highlight the critical role of the PFC-LHb circuit in controlling cocaine reward-related behaviors and shed light on the underlying mechanisms. Understanding this circuit's function may provide valuable insights into addiction and contribute to developing targeted treatments for substance use disorders.
Collapse
Affiliation(s)
- Nur Abdel-Hay
- Institute for Drug Research (IDR), School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Marina Kabirova
- Institute for Drug Research (IDR), School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Rami Yaka
- Institute for Drug Research (IDR), School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel.
| |
Collapse
|
189
|
Merces L, Ferro LMM, Nawaz A, Sonar P. Advanced Neuromorphic Applications Enabled by Synaptic Ion-Gating Vertical Transistors. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2305611. [PMID: 38757653 PMCID: PMC11251569 DOI: 10.1002/advs.202305611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 12/07/2023] [Indexed: 05/18/2024]
Abstract
Bioinspired synaptic devices have shown great potential in artificial intelligence and neuromorphic electronics. Low energy consumption, multi-modal sensing and recording, and multifunctional integration are critical aspects limiting their applications. Recently, a new synaptic device architecture, the ion-gating vertical transistor (IGVT), has been successfully realized and timely applied to perform brain-like perception, such as artificial vision, touch, taste, and hearing. In this short time, IGVTs have already achieved faster data processing speeds and more promising memory capabilities than many conventional neuromorphic devices, even while operating at lower voltages and consuming less power. This work focuses on the cutting-edge progress of IGVT technology, from outstanding fabrication strategies to the design and realization of low-voltage multi-sensing IGVTs for artificial-synapse applications. The fundamental concepts of artificial synaptic IGVTs, such as signal processing, transduction, plasticity, and multi-stimulus perception are discussed comprehensively. The contribution draws special attention to the development and optimization of multi-modal flexible sensor technologies and presents a roadmap for future high-end theoretical and experimental advancements in neuromorphic research that are mostly achievable by the synaptic IGVTs.
Collapse
Affiliation(s)
- Leandro Merces
- Research Center for MaterialsArchitectures, and Integration of Nanomembranes (MAIN)Chemnitz University of Technology09126ChemnitzGermany
| | - Letícia Mariê Minatogau Ferro
- Research Center for MaterialsArchitectures, and Integration of Nanomembranes (MAIN)Chemnitz University of Technology09126ChemnitzGermany
| | - Ali Nawaz
- Center for Sensors and DevicesBruno Kessler Foundation (FBK)Trento38123Italy
| | - Prashant Sonar
- School of Chemistry and PhysicsQueensland University of Technology (QUT)BrisbaneQLD4000Australia
- Centre for Materials ScienceQueensland University of Technology2 George StreetBrisbaneQLD4000Australia
| |
Collapse
|
190
|
Chen J, Wei X, Wu X, Zhang Q, Xia G, Xia H, Shang H, Lin S. Disorder of neuroplasticity aggravates cognitive impairment via neuroinflammation associated with intestinal flora dysbiosis in chronic heart failure. Aging (Albany NY) 2024; 16:10882-10904. [PMID: 38968172 PMCID: PMC11272129 DOI: 10.18632/aging.205960] [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: 12/12/2023] [Accepted: 03/28/2024] [Indexed: 07/07/2024]
Abstract
BACKGROUND Chronic heart failure (CHF) impairs cognitive function, yet its effects on brain structure and underlying mechanisms remain elusive. This study aims to explore the mechanisms behind cognitive impairment. METHODS CHF models in rats were induced by ligation of the left anterior descending coronary artery. Cardiac function was analyzed by cardiac ultrasound and hemodynamics. ELISA, immunofluorescence, Western blot, Golgi staining and transmission electron microscopy were performed on hippocampal tissues. The alterations of intestinal flora under the morbid state were investigated via 16S rRNA sequencing. The connection between neuroinflammation and synapses is confirmed by a co-culture system of BV2 microglia and HT22 cells in vitro. Results: CHF rats exhibited deteriorated cognitive behaviors. CHF induced neuronal structural disruption, loss of Nissl bodies, and synaptic damage, exhibiting alterations in multiple parameters. CHF rats showed increased hippocampal levels of inflammatory cytokines and activated microglia and astrocytes. Furthermore, the study highlights dysregulated PDE4-dependent cAMP signaling and intestinal flora dysbiosis, closely associated with neuroinflammation, and altered synaptic proteins. In vitro, microglial neuroinflammation impaired synaptic plasticity via PDE4-dependent cAMP signaling. CONCLUSIONS Neuroinflammation worsens CHF-related cognitive impairment through neuroplasticity disorder, tied to intestinal flora dysbiosis. PDE4 emerges as a potential therapeutic target. These findings provide insightful perspectives on the heart-gut-brain axis.
Collapse
Affiliation(s)
- Jie Chen
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100700, China
| | - Xiaohong Wei
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100700, China
| | - Xuefen Wu
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100700, China
| | - Qian Zhang
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100700, China
| | - Guiyang Xia
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100700, China
| | - Huan Xia
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100700, China
| | - Hongcai Shang
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100700, China
| | - Sheng Lin
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100700, China
| |
Collapse
|
191
|
Pizzella A, Penna E, Abate N, Frenna E, Canafoglia L, Ragona F, Russo R, Chambery A, Perrone-Capano C, Cappello S, Crispino M, Di Giaimo R. Pathological Deficit of Cystatin B Impairs Synaptic Plasticity in EPM1 Human Cerebral Organoids. Mol Neurobiol 2024; 61:4318-4334. [PMID: 38087165 PMCID: PMC11236866 DOI: 10.1007/s12035-023-03812-y] [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: 09/19/2023] [Accepted: 11/17/2023] [Indexed: 07/11/2024]
Abstract
Cystatin B (CSTB) is a small protease inhibitor protein being involved in cell proliferation and neuronal differentiation. Loss-of-function mutations in CSTB gene cause progressive myoclonic epilepsy 1 (EPM1). We previously demonstrated that CSTB is locally synthesized in synaptic nerve terminals from rat brain and secreted into the media, indicating its role in synaptic plasticity. In this work, we have further investigated the involvement of CSTB in synaptic plasticity, using synaptosomes from human cerebral organoids (hCOs) as well as from rodents' brain. Our data demonstrate that CSTB is released from synaptosomes in two ways: (i) as a soluble protein and (ii) in extracellular vesicles-mediated pathway. Synaptosomes isolated from hCOs are enriched in pre-synaptic proteins and contain CSTB at all developmental stages analyzed. CSTB presence in the synaptic territories was also confirmed by immunostaining on human neurons in vitro. To investigate if the depletion of CSTB affects synaptic plasticity, we characterized the synaptosomes from EPM1 hCOs. We found that the levels of presynaptic proteins and of an initiation factor linked to local protein synthesis were both reduced in EPM1 hCOs and that the extracellular vesicles trafficking pathway was impaired. Moreover, EPM1 neurons displayed anomalous morphology with longer and more branched neurites bearing higher number of intersections and nodes, suggesting connectivity alterations. In conclusion, our data strengthen the idea that CSTB plays a critical role in the synapse physiology and reveal that pathologically low levels of CSTB may affect synaptic plasticity, leading to synaptopathy and altered neuronal morphology.
Collapse
Affiliation(s)
- Amelia Pizzella
- Department of Biology, University of Naples Federico II, Naples, Italy
- Department of Developmental Neurobiology, Max Planck Institute of Psychiatry, Munich, Germany
| | - Eduardo Penna
- Department of Biology, University of Naples Federico II, Naples, Italy
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA, 91766, USA
| | - Natalia Abate
- Department of Biology, University of Naples Federico II, Naples, Italy
| | - Elisa Frenna
- Department of Biology, University of Naples Federico II, Naples, Italy
- Department of Developmental Neurobiology, Max Planck Institute of Psychiatry, Munich, Germany
| | - Laura Canafoglia
- Integrated Diagnostics for Epilepsy, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Francesca Ragona
- Department of Pediatric Neuroscience, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Rosita Russo
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, University of Campania "Luigi Vanvitelli, Caserta, Italy
| | - Angela Chambery
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, University of Campania "Luigi Vanvitelli, Caserta, Italy
| | | | - Silvia Cappello
- Department of Developmental Neurobiology, Max Planck Institute of Psychiatry, Munich, Germany
- Biomedical Center, Ludwig Maximilian University of Munich, Munich, Germany
| | - Marianna Crispino
- Department of Biology, University of Naples Federico II, Naples, Italy.
| | - Rossella Di Giaimo
- Department of Biology, University of Naples Federico II, Naples, Italy.
- Department of Developmental Neurobiology, Max Planck Institute of Psychiatry, Munich, Germany.
| |
Collapse
|
192
|
Koenig A, Lewis M, Wald J, Li S, Varoglu M, Dai J, Sankoh A, Paumier K, Doherty J, Quirk M. Dalzanemdor (SAGE-718), a novel, investigational N-methyl-D-aspartate receptor positive allosteric modulator: Safety, tolerability, and clinical pharmacology in randomized dose-finding studies in healthy participants and an open-label study in participants with Huntington's disease. Clin Transl Sci 2024; 17:e13852. [PMID: 38988035 PMCID: PMC11236904 DOI: 10.1111/cts.13852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 04/26/2024] [Accepted: 05/07/2024] [Indexed: 07/12/2024] Open
Abstract
N-methyl-D-aspartate receptor (NMDAR)-positive allosteric modulators (PAMs) represent a potential therapeutic strategy for cognitive impairment in disorders associated with NMDAR hypofunction, including Huntington's disease (HD) and Alzheimer's disease. Dalzanemdor (SAGE-718) is a novel, investigational NMDAR PAM being evaluated for the potential treatment of cognitive impairment in these disorders. We report first-in-human, phase I, double-blind, dose-finding studies to assess the safety, tolerability, and clinical pharmacology of dalzanemdor. A single-ascending dose study (dalzanemdor 0.35, 0.75, 1.5, or 3.0 mg vs. placebo) was conducted in healthy participants and included food effects. A multiple-ascending dose study (14 days) was conducted in healthy participants (dalzanemdor 0.5 or 1.0 mg vs. placebo) and HD participants (open-label dalzanemdor 1.0 mg) and included exploratory pharmacodynamics on cognitive performance. Dalzanemdor was generally well tolerated with no adverse events leading to discontinuation. Dalzanemdor exhibited pharmacokinetic parameters appropriate for once-daily dosing. Following single and multiple doses in healthy participants, median terminal half-life was 8-118 h, and the median time to reach maximum plasma concentration was 4-7 h. Exposures were dose-proportional after single dose (6-46 ng/mL) and more than dose-proportional after multiple doses (6-41 ng/mL). With multiple dosing, a steady state was achieved after 11 days in healthy participants and 13 days in HD participants. Dalzanemdor exposure decreased slightly with food. In HD participants, results suggest that dalzanemdor may improve cognitive performance on tests of executive function. These results support continued clinical development of dalzanemdor for the potential treatment of cognitive impairment in disorders of NMDAR hypofunction.
Collapse
Affiliation(s)
| | | | - Jeff Wald
- Sage Therapeutics, IncCambridgeMassachusettsUSA
| | - Sigui Li
- Sage Therapeutics, IncCambridgeMassachusettsUSA
| | | | - Jing Dai
- Sage Therapeutics, IncCambridgeMassachusettsUSA
| | | | | | | | - Mike Quirk
- Sage Therapeutics, IncCambridgeMassachusettsUSA
| |
Collapse
|
193
|
Xu X, Li F, Liu C, Wang Y, Yang Z, Xie G, Zhang T. Low-frequency repetitive transcranial magnetic stimulation alleviates abnormal behavior in valproic acid rat model of autism through rescuing synaptic plasticity and inhibiting neuroinflammation. Pharmacol Biochem Behav 2024; 240:173788. [PMID: 38734150 DOI: 10.1016/j.pbb.2024.173788] [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: 04/08/2024] [Revised: 05/06/2024] [Accepted: 05/08/2024] [Indexed: 05/13/2024]
Abstract
Autism is a complex neurodevelopmental disorder with no effective treatment available currently. Repetitive transcranial magnetic stimulation (rTMS) is emerging as a promising neuromodulation technique to treat autism. However, the mechanism how rTMS works remains unclear, which restrict the clinical application of magnetic stimulation in the autism treatment. In this study, we investigated the effect of low-frequency rTMS on the autistic-like symptoms and explored if this neuroprotective effect was associated with synaptic plasticity and neuroinflammation in the hippocampus. A rat model of autism was established by intraperitoneal injection of valproic acid (VPA) in pregnant rats and male offspring were treated with 1 Hz rTMS daily for two weeks continuously. Behavior tests were performed to identify behavioral abnormality. Synaptic plasticity was measured by in vivo electrophysiological recording and Golgi-Cox staining. Synapse and inflammation associated proteins were detected by immunofluorescence and Western blot analyses. Results showed prenatal VPA-exposed rats exhibited autistic-like and anxiety-like behaviors, and cognitive impairment. Synaptic plasticity deficits and the abnormality expression of synapse-associated proteins were found in the hippocampus of prenatal VPA-exposed rats. Prenatal VPA exposure increased the level of inflammation cytokines and promoted the excessive activation of microglia. rTMS significantly alleviated the prenatal VPA-induced abnormalities including behavioral and synaptic plasticity deficits, and excessive neuroinflammation. TMS maybe a potential strategy for autism therapy via rescuing synaptic plasticity and inhibiting neuroinflammation.
Collapse
Affiliation(s)
- Xinxin Xu
- State Key Laboratory of Reliability and Intelligence of Electrical Equipment, School of Health Sciences and Biomedical Engineering, Hebei University of Technology, 300130 Tianjin, China; College of Life Sciences and Key Laboratory of Bioactive Materials Ministry of Education, Nankai University, 300071 Tianjin, China
| | - Fangjuan Li
- College of Life Sciences and Key Laboratory of Bioactive Materials Ministry of Education, Nankai University, 300071 Tianjin, China
| | - Chunhua Liu
- School of Medicine, State Key Laboratory of Medicinal Chemical Biology, Nankai University, 300071 Tianjin, China
| | - Yue Wang
- School of Medicine, State Key Laboratory of Medicinal Chemical Biology, Nankai University, 300071 Tianjin, China
| | - Zhuo Yang
- School of Medicine, State Key Laboratory of Medicinal Chemical Biology, Nankai University, 300071 Tianjin, China
| | - Guoming Xie
- Ningbo Medical Center Lihuili Hospital, 315040 Ningbo, Zhejiang, China.
| | - Tao Zhang
- College of Life Sciences and Key Laboratory of Bioactive Materials Ministry of Education, Nankai University, 300071 Tianjin, China.
| |
Collapse
|
194
|
Chauhan K, Neiman AB, Tass PA. Synaptic reorganization of synchronized neuronal networks with synaptic weight and structural plasticity. PLoS Comput Biol 2024; 20:e1012261. [PMID: 38980898 PMCID: PMC11259284 DOI: 10.1371/journal.pcbi.1012261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 07/19/2024] [Accepted: 06/20/2024] [Indexed: 07/11/2024] Open
Abstract
Abnormally strong neural synchronization may impair brain function, as observed in several brain disorders. We computationally study how neuronal dynamics, synaptic weights, and network structure co-emerge, in particular, during (de)synchronization processes and how they are affected by external perturbation. To investigate the impact of different types of plasticity mechanisms, we combine a network of excitatory integrate-and-fire neurons with different synaptic weight and/or structural plasticity mechanisms: (i) only spike-timing-dependent plasticity (STDP), (ii) only homeostatic structural plasticity (hSP), i.e., without weight-dependent pruning and without STDP, (iii) a combination of STDP and hSP, i.e., without weight-dependent pruning, and (iv) a combination of STDP and structural plasticity (SP) that includes hSP and weight-dependent pruning. To accommodate the diverse time scales of neuronal firing, STDP, and SP, we introduce a simple stochastic SP model, enabling detailed numerical analyses. With tools from network theory, we reveal that structural reorganization may remarkably enhance the network's level of synchrony. When weaker contacts are preferentially eliminated by weight-dependent pruning, synchrony is achieved with significantly sparser connections than in randomly structured networks in the STDP-only model. In particular, the strengthening of contacts from neurons with higher natural firing rates to those with lower rates and the weakening of contacts in the opposite direction, followed by selective removal of weak contacts, allows for strong synchrony with fewer connections. This activity-led network reorganization results in the emergence of degree-frequency, degree-degree correlations, and a mixture of degree assortativity. We compare the stimulation-induced desynchronization of synchronized states in the STDP-only model (i) with the desynchronization of models (iii) and (iv). The latter require stimuli of significantly higher intensity to achieve long-term desynchronization. These findings may inform future pre-clinical and clinical studies with invasive or non-invasive stimulus modalities aiming at inducing long-lasting relief of symptoms, e.g., in Parkinson's disease.
Collapse
Affiliation(s)
- Kanishk Chauhan
- Department of Physics and Astronomy, Ohio University, Athens, Ohio, United States of America
- Neuroscience Program, Ohio University, Athens, Ohio, United States of America
| | - Alexander B. Neiman
- Department of Physics and Astronomy, Ohio University, Athens, Ohio, United States of America
- Neuroscience Program, Ohio University, Athens, Ohio, United States of America
| | - Peter A. Tass
- Department of Neurosurgery, Stanford University, Stanford, California, United States of America
| |
Collapse
|
195
|
Papassotiropoulos A, de Quervain DJF. Tweaking synaptic plasticity: Deciphering the role of WWC1 in memory opens new therapeutic horizons. Sci Signal 2024; 17:eadp5354. [PMID: 38917220 DOI: 10.1126/scisignal.adp5354] [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: 04/29/2024] [Accepted: 05/31/2024] [Indexed: 06/27/2024]
Abstract
WWC1 is a scaffolding protein in the evolutionarily conserved Hippo signaling network and is genetically linked to human memory and synaptic plasticity. In the archives of Science Signaling, Stepan et al. demonstrate the translational potential of modulating WWC1 through pharmacological inhibition of Hippo-pathway kinases to enhance cognition.
Collapse
Affiliation(s)
- Andreas Papassotiropoulos
- Division of Molecular Neuroscience, Department of Biomedicine, University of Basel, CH-4055 Basel, Switzerland
- Research Cluster Molecular and Cognitive Neurosciences, University of Basel, CH-4055 Basel, Switzerland
- Psychiatric University Clinics, University of Basel, CH-4055 Basel, Switzerland
| | - Dominique J-F de Quervain
- Research Cluster Molecular and Cognitive Neurosciences, University of Basel, CH-4055 Basel, Switzerland
- Psychiatric University Clinics, University of Basel, CH-4055 Basel, Switzerland
- Division of Cognitive Neuroscience, Department of Biomedicine, University of Basel, CH-4055 Basel, Switzerland
| |
Collapse
|
196
|
Wadan AHS, Raza ML, Moradikor N. Synaptic modulation by coffee compounds: Insights into neural plasticity. PROGRESS IN BRAIN RESEARCH 2024; 289:181-191. [PMID: 39168580 DOI: 10.1016/bs.pbr.2024.06.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/23/2024]
Abstract
The physiological structure and functioning of the brain are determined by activity-dependent processes and affected by "synapse plasticity." Because chemical transmitters target and regulate synapses, exogenous chemical stimulants and transmitters can alter their physiological functions by interacting with synaptic surface receptors or chemical modulators. Caffeine, a commonly used pharmacologic substance, can target and alter synapses. It impact various biological, chemical, and metabolic processes related to synaptic function. This chapter investigates how caffeine affects fluctuations in structure and function in the hippocampus formation and neocortical structure, regions known for their high synaptic plasticity profile. Specifically, caffeine modulates various synaptic receptors and channel activities by mobilizing intracellular calcium, inhibiting phosphodiesterase, and blocking adenosine and GABA cellular receptors. These caffeine-induced pathways and functions allow neurons to generate plastic modulations in synaptic actions such as efficient and morphological transmission. Moreover, at a network level, caffeine can stimulate neural oscillators in the cortex, resulting in repetitive signals that strengthen long-range communication between cortical areas reliant on N-methyl-d-aspartate receptors. This suggests that caffeine could facilitate the reorganization of cortical network functions through its effects on synaptic mobilization.
Collapse
Affiliation(s)
- Al-Hassan Soliman Wadan
- Sinai University, Faculty of Dentistry, Arish, North Sinai, Egypt; Sinai University Research Center (SURC), Sinai University, Sinai Governorate, Egypt.
| | - Muhammad Liaquat Raza
- Department of Infection Prevention & Control, Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia; King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia; King Abdullah International Medical Research Center (KAIMRC), Riyadh, Saudi Arabia
| | - Nasrollah Moradikor
- International Center for Neuroscience Research, Institute for Intelligent Research, Tbilisi, Georgia
| |
Collapse
|
197
|
Balloff C, Janßen LK, Hartmann CJ, Meuth SG, Schnitzler A, Penner IK, Albrecht P. Predictive value of synaptic plasticity for functional decline in patients with multiple sclerosis. Front Neurol 2024; 15:1410673. [PMID: 38974686 PMCID: PMC11224454 DOI: 10.3389/fneur.2024.1410673] [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: 04/01/2024] [Accepted: 06/10/2024] [Indexed: 07/09/2024] Open
Abstract
Background Previous research suggested that quadripulse (QPS)-induced synaptic plasticity is associated with both cognitive and motor function in patients with multiple sclerosis (MS) and does not appear to be reduced compared to healthy controls (HCs). Objective This study aimed to explore the relationship between the degree of QPS-induced plasticity and clinically significant decline in motor and cognitive functions over time. We hypothesized that MS patients experiencing functional decline would exhibit lower levels of baseline plasticity compared to those without decline. Methods QPS-induced plasticity was evaluated in 80 MS patients (56 with relapsing-remitting MS and 24 with progressive MS), and 69 age-, sex-, and education-matched HCs. Cognitive and motor functions, as well as overall disability status were evaluated annually over a median follow-up period of 2 years. Clinically meaningful change thresholds were predefined for each outcome measure. Linear mixed-effects models, Cox proportional hazard models, logistic regression, and receiver-operating characteristic analysis were applied to analyse the relationship between baseline plasticity and clinical progression in the symbol digit modalities test, brief visuospatial memory test revised (BVMT-R), nine-hole peg test (NHPT), timed 25-foot walk test, and expanded disability status scale. Results Overall, the patient cohort showed no clinically relevant change in any functional outcome over time. Variability in performance was observed across time points in both patients and HCs. MS patients who experienced clinically relevant decline in manual dexterity and/or visuospatial learning and memory had significantly lower levels of synaptic plasticity at baseline compared to those without such decline (NHPT: β = -0.25, p = 0.02; BVMT-R: β = -0.50, p = 0.005). Receiver-operating characteristic analysis underscored the predictive utility of baseline synaptic plasticity in discerning between patients experiencing functional decline and those maintaining stability only for visuospatial learning and memory (area under the curve = 0.85). Conclusion Our study suggests that QPS-induced plasticity could be linked to clinically relevant functional decline in patients with MS. However, to solidify these findings, longer follow-up periods are warranted, especially in cohorts with higher prevalences of functional decline. Additionally, the variability in cognitive performance in both patients with MS and HCs underscores the importance of conducting further research on reliable change based on neuropsychological tests.
Collapse
Affiliation(s)
- Carolin Balloff
- Department of Neurology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University, Düsseldorf, Germany
- Department of Neurology, Kliniken Maria Hilf GmbH, Mönchengladbach, Germany
| | - Lisa Kathleen Janßen
- Department of Neurology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University, Düsseldorf, Germany
| | - Christian Johannes Hartmann
- Department of Neurology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University, Düsseldorf, Germany
- Institute of Clinical Neuroscience and Medical Psychology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University, Düsseldorf, Germany
| | - Sven Günther Meuth
- Department of Neurology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University, Düsseldorf, Germany
| | - Alfons Schnitzler
- Department of Neurology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University, Düsseldorf, Germany
- Institute of Clinical Neuroscience and Medical Psychology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University, Düsseldorf, Germany
| | - Iris-Katharina Penner
- Department of Neurology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University, Düsseldorf, Germany
- Department of Neurology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Philipp Albrecht
- Department of Neurology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University, Düsseldorf, Germany
- Department of Neurology, Kliniken Maria Hilf GmbH, Mönchengladbach, Germany
| |
Collapse
|
198
|
Lai XC, Tang Z, Fang J, Feng L, Yao DJ, Zhang L, Jiang YP, Liu QX, Tang XG, Zhou YC, Shang J, Zhong GK, Gao J. An adjustable multistage resistance switching behavior of a photoelectric artificial synaptic device with a ferroelectric diode effect for neuromorphic computing. MATERIALS HORIZONS 2024; 11:2886-2897. [PMID: 38563639 DOI: 10.1039/d4mh00064a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Neuromorphic computing, which mimics biological neural networks, is widely regarded as the optimal solution for addressing the limitations of traditional von Neumann computing architecture. In this work, an adjustable multistage resistance switching ferroelectric Bi2FeCrO6 diode artificial synaptic device was fabricated using a sol-gel method with a simple process. The device exhibits nonlinearity in its electrical characteristics, demonstrating tunable multistage resistance switching behavior and a strong ferroelectric diode effect through the manipulation of ferroelectric polarization. One of its salient advantages resides in its capacity to dynamically regulate its polarization state in response to an external electric field, thereby facilitating the fine-tuning of synaptic connection strength while maintaining synaptic stability. The device is capable of accurately simulating the fundamental properties of biological synapses, including long/short-term plasticity, paired-pulse facilitation, and spike-timing-dependent plasticity. Additionally, the device exhibits a distinctive photoelectric response and is capable of inducing synaptic plasticity by light signal activation. The utilization of a femtosecond laser for the scrutiny of carrier transport mechanisms imparts profound insights into the intricate dynamics governing the optical memory effect. Furthermore, utilizing a convolutional neural network (CNN) architecture, the recognition accuracy of the MNIST and fashion MNIST datasets was improved to 95.6% and 78%, respectively, through the implementation of improved random adaptive algorithms. These findings present a new opportunity for utilizing Bi2FeCrO6 materials in the development of artificial synapses for neuromorphic computation.
Collapse
Affiliation(s)
- Xi-Cai Lai
- School of Physics and Optoelectronic Engineering, Guangdong University of Technology, Guangzhou Higher Education Mega Center, Guangzhou, 510006, P. R. China.
| | - Zhenhua Tang
- School of Physics and Optoelectronic Engineering, Guangdong University of Technology, Guangzhou Higher Education Mega Center, Guangzhou, 510006, P. R. China.
| | - Junlin Fang
- School of Physics and Optoelectronic Engineering, Guangdong University of Technology, Guangzhou Higher Education Mega Center, Guangzhou, 510006, P. R. China.
| | - Leyan Feng
- School of Physics and Optoelectronic Engineering, Guangdong University of Technology, Guangzhou Higher Education Mega Center, Guangzhou, 510006, P. R. China.
| | - Di-Jie Yao
- School of Physics and Optoelectronic Engineering, Guangdong University of Technology, Guangzhou Higher Education Mega Center, Guangzhou, 510006, P. R. China.
| | - Li Zhang
- School of Physics and Optoelectronic Engineering, Guangdong University of Technology, Guangzhou Higher Education Mega Center, Guangzhou, 510006, P. R. China.
| | - Yan-Ping Jiang
- School of Physics and Optoelectronic Engineering, Guangdong University of Technology, Guangzhou Higher Education Mega Center, Guangzhou, 510006, P. R. China.
| | - Qiu-Xiang Liu
- School of Physics and Optoelectronic Engineering, Guangdong University of Technology, Guangzhou Higher Education Mega Center, Guangzhou, 510006, P. R. China.
| | - Xin-Gui Tang
- School of Physics and Optoelectronic Engineering, Guangdong University of Technology, Guangzhou Higher Education Mega Center, Guangzhou, 510006, P. R. China.
| | - Yi-Chun Zhou
- School of Advanced Materials and Nanotechnology, Xidian University, Xian 710126, China
| | - Jie Shang
- Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo 315201, China
| | - Gao-Kuo Zhong
- Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Ju Gao
- Department of Physics, The University of Hong Kong, Hong Kong 999077, P. R. China
| |
Collapse
|
199
|
Zhang W, Zhang X, Lei M, Zhang D, Qin G, Zhou J, Ji L, Chen L. Dopamine D2 Receptor Activation Blocks GluA2/ROS Positive Feedback Loop to Alienate Chronic-Migraine-Associated Pain Sensitization. Antioxidants (Basel) 2024; 13:725. [PMID: 38929165 PMCID: PMC11201052 DOI: 10.3390/antiox13060725] [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: 04/24/2024] [Revised: 05/29/2024] [Accepted: 06/10/2024] [Indexed: 06/28/2024] Open
Abstract
Chronic migraine is a disabling disorder without effective therapeutic medicine. AMPA receptors have been proven to be essential to pathological pain and headaches, but the related regulatory mechanisms in chronic migraine have not yet been explored. In this study, we found that the level of surface GluA2 was reduced in chronic migraine rats. Tat-GluR23Y (a GluA2 endocytosis inhibitor) reduced calcium inward flow and weakened synaptic structures, thus alleviating migraine-like pain sensitization. In addition, the inhibition of GluA2 endocytosis reduced the calcium influx and alleviated mitochondrial calcium overload and ROS generation in primary neurons. Furthermore, our results showed that ROS can induce allodynia and GluA2 endocytosis in rats, thus promoting migraine-like pain sensitization. In our previous study, the dopamine D2 receptor was identified as a potential target in the treatment of chronic migraine, and here we found that dopamine D2 receptor activation suppressed chronic-migraine-related pain sensitization through blocking the GluA2/ROS positive feedback loop in vivo and in vitro. Additionally, ligustrazine, a core component of ligusticum chuanxiong, was shown to target the dopamine D2 receptor, thereby alleviating ROS production and abnormal nociception in CM rats. This study provides valuable insight into the treatment of chronic migraine.
Collapse
Affiliation(s)
- Wei Zhang
- Laboratory Research Center, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; (W.Z.); (G.Q.)
| | - Xiaoyan Zhang
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400042, China; (X.Z.); (J.Z.)
| | - Ming Lei
- Laboratory Research Center, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; (W.Z.); (G.Q.)
| | - Dunke Zhang
- Laboratory Research Center, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; (W.Z.); (G.Q.)
| | - Guangcheng Qin
- Laboratory Research Center, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; (W.Z.); (G.Q.)
| | - Jiying Zhou
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400042, China; (X.Z.); (J.Z.)
| | - Lichun Ji
- Department of Respiration, The Thirteenth People’s Hospital of Chongqing, Chongqing 400016, China
| | - Lixue Chen
- Laboratory Research Center, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; (W.Z.); (G.Q.)
| |
Collapse
|
200
|
Cagnetta R, Lacaille JC, Sonenberg N. Exploration of new space elicits phosphorylation of GluA1(Ser831) and S6K and expression of Arc in the hippocampus in vivo as in long-term potentiation. Mol Brain 2024; 17:35. [PMID: 38858726 PMCID: PMC11165848 DOI: 10.1186/s13041-024-01100-x] [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: 01/17/2024] [Accepted: 05/14/2024] [Indexed: 06/12/2024] Open
Abstract
The brain responds to experience through modulation of synaptic transmission, that is synaptic plasticity. An increase in the strength of synaptic transmission is manifested as long-term potentiation (LTP), while a decrease in the strength of synaptic transmission is expressed as long-term depression (LTD). Most of the studies of synaptic plasticity have been carried out by induction via electrophysiological stimulation. It is largely unknown in which behavioural tasks such synaptic plasticity occurs. Moreover, some stimuli can induce both LTP and LTD, thus making it difficult to separately study the different forms of synaptic plasticity. Two studies have shown that an aversive memory task - inhibitory avoidance learning and contextual fear conditioning - physiologically and selectively induce LTP and an LTP-like molecular change, respectively, in the hippocampus in vivo. Here, we show that a non-aversive behavioural task - exploration of new space - physiologically and selectively elicits a biochemical change in the hippocampus that is a hallmark of LTP. Specifically, we found that exploration of new space induces an increase in the phosphorylation of GluA1(Ser831), without affecting the phosphorylation of GluA1(Ser845), which are biomarkers of early-LTP and not NMDAR-mediated LTD. We also show that exploration of new space engenders the phosphorylation of the translational regulator S6K and the expression of Arc, which are features of electrophysiologically-induced late-LTP in the hippocampus. Therefore, our results show that exploration of new space is a novel non-aversive behavioural paradigm that elicits molecular changes in vivo that are analogous to those occurring during early- and late-LTP, but not during NMDAR-mediated LTD.
Collapse
Affiliation(s)
- Roberta Cagnetta
- Department of Cell Biology and Program in Neuroscience, Harvard Medical School, Boston, MA, 02115, USA.
- Department of Biochemistry and Goodman Cancer Institute, McGill University, Montreal, QC, H3A 1A3, Canada.
| | - Jean-Claude Lacaille
- Department of Neurosciences, Center for Interdisciplinary Research On Brain and Learning, Research Group On Neural Signaling and Circuitry, University of Montreal, Montreal, QC, H3T1J4, Canada
| | - Nahum Sonenberg
- Department of Biochemistry and Goodman Cancer Institute, McGill University, Montreal, QC, H3A 1A3, Canada.
| |
Collapse
|