1
|
Yao Y, Cui C, Shi Y, Lei J, Li T, Li M, Peng X, Yang X, Ren K, Yang J, Luo G, Du J, Chen S, Zhang P, Tian B. DRN-SNc serotonergic circuit drives stress-induced motor deficits and Parkinson's disease vulnerability. Neuropsychopharmacology 2025; 50:1051-1062. [PMID: 40097739 DOI: 10.1038/s41386-025-02080-9] [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: 09/23/2024] [Revised: 01/31/2025] [Accepted: 02/25/2025] [Indexed: 03/19/2025]
Abstract
Stress is a recognized risk factor for Parkinson's disease (PD), but the mechanisms by which stress exacerbates PD symptoms through the serotonergic system are not fully understood. This study investigates the role of serotonergic (5-HT) neurons in the dorsal raphe nucleus (DRN) in mediating stress-induced motor deficits and PD progression. Acute and chronic stress were induced in mice using an elevated platform (EP) and combined with MPTP administration to model early-stage PD. Acute EP stress caused transient motor deficits and significant activation of DRN5-HT neurons projecting to substantia nigra compacta (SNc) dopaminergic (DA) neurons. Manipulating the DRN-SNc pathway with optogenetics and chemogenetics confirmed its critical role in stress-induced motor deficits. Activation of the SNc 5-HT2C receptor with an agonist replicated these deficits, while receptor inhibition prevented them, underscoring its importance. Chronic EP stress worsened MPTP-induced deficits and caused significant SNcDA neurons loss, suggesting it accelerates PD progression. Prolonged chemogenetic inhibition of the DRN-SNc circuit mitigated chronic stress effects in MPTP-treated mice. These findings highlight the crucial role of the DRN-SNc serotonergic circuit and 5-HT2C receptors in stress-related motor deficits, suggesting potential targets for therapies aimed at treating both stress-related motor disorders and Parkinson's disease.
Collapse
Affiliation(s)
- Yibo Yao
- Department of Neurobiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
| | - Chi Cui
- Department of Neurobiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
| | - Yulong Shi
- Department of Neurobiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
| | - Jie Lei
- Department of Neurobiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
| | - Tongxia Li
- Department of Neurobiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
| | - Ming Li
- Department of Neurobiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
| | - Xiang Peng
- Department of Neurobiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
| | - Xueke Yang
- Department of Neurobiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
| | - Kun Ren
- Department of Neurobiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
| | - Jian Yang
- Department of Neurobiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
| | - Gangan Luo
- Department of Neurobiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
| | - Junsong Du
- Department of Neurobiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
| | - Sitong Chen
- Department of Neurobiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
| | - Pei Zhang
- Department of Neurobiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China.
- Institute for Brain Research, Huazhong University of Science and Technology, Wuhan, Hubei, PR China.
- Key Laboratory of Neurological Diseases, Ministry of Education, Wuhan, Hubei, PR China.
| | - Bo Tian
- School of Medicine, Wuhan University of Science and Technology, Wuhan, Hubei, PR China.
| |
Collapse
|
2
|
Cheong WH, Kim G, Lee Y, Kim EY, Jeon JB, Kim DH, Kim KM. A Neuransistor with Excitatory and Inhibitory Neuronal Behaviors for Liquid State Machine. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2419122. [PMID: 40195785 PMCID: PMC12177865 DOI: 10.1002/adma.202419122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Revised: 03/03/2025] [Indexed: 04/09/2025]
Abstract
A liquid state machine (LSM) is a spiking neural network model inspired by biological neural network dynamics designed to process time-varying inputs. In the LSM, maintaining a proper excitatory/inhibitory (E/I) balance among neurons is essential for ensuring network stability and generating rich temporal dynamics for accurate data processing. In this study, a "neuransistor" is proposed that implements the E/I neurons in a single device, allowing for the hardware implementation of the LSM. The device features a three-terminal transistor structure embodying TiO2- x/Al2O3 bi-layer, providing a two-dimensional electron electron gas (2DEG) channel at their interface. This device demonstrates hybrid excitatory and inhibitory dynamics with respect to the applied gate bias polarity, originating from the charge trapping/detrapping between the 2DEG and TiO2- x layers. Additionally, the three-terminal configuration allows masking capabilities by selecting terminal biases, realizing a reservoir behavior with superior reliability and durability. Its use in an LSM reservoir for time-series data prediction tasks using the Henon dataset and a chaotic equation solver for the Lorenz attractor is demonstrated. This benchmarking indicates that the LSM exhibits enhanced performance and efficiency compared to the conventional echo state network, underscoring its potential for advanced applications in reservoir computing.
Collapse
Affiliation(s)
- Woon Hyung Cheong
- Applied Science Research InstituteKorea Advanced Institute of Science and Technology (KAIST)Daejeon34141Republic of Korea
| | - Geunyoung Kim
- Applied Science Research InstituteKorea Advanced Institute of Science and Technology (KAIST)Daejeon34141Republic of Korea
| | - Younghyun Lee
- Department of Materials Science and EngineeringKorea Advanced Institute of Science and Technology (KAIST)Daejeon34141Republic of Korea
| | - Eun Young Kim
- Department of Semiconductor TechnologyKorea Advanced Institute of Science and Technology (KAIST)Daejeon34141Republic of Korea
| | - Jae Bum Jeon
- Department of Materials Science and EngineeringKorea Advanced Institute of Science and Technology (KAIST)Daejeon34141Republic of Korea
| | - Do Hoon Kim
- Department of Materials Science and EngineeringKorea Advanced Institute of Science and Technology (KAIST)Daejeon34141Republic of Korea
| | - Kyung Min Kim
- Department of Materials Science and EngineeringKorea Advanced Institute of Science and Technology (KAIST)Daejeon34141Republic of Korea
- Department of Semiconductor TechnologyKorea Advanced Institute of Science and Technology (KAIST)Daejeon34141Republic of Korea
| |
Collapse
|
3
|
Zhong S, Su L, Xu M, Loke D, Yu B, Zhang Y, Zhao R. Recent Advances in Artificial Sensory Neurons: Biological Fundamentals, Devices, Applications, and Challenges. NANO-MICRO LETTERS 2024; 17:61. [PMID: 39537845 PMCID: PMC11561216 DOI: 10.1007/s40820-024-01550-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Accepted: 09/28/2024] [Indexed: 11/16/2024]
Abstract
Spike-based neural networks, which use spikes or action potentials to represent information, have gained a lot of attention because of their high energy efficiency and low power consumption. To fully leverage its advantages, converting the external analog signals to spikes is an essential prerequisite. Conventional approaches including analog-to-digital converters or ring oscillators, and sensors suffer from high power and area costs. Recent efforts are devoted to constructing artificial sensory neurons based on emerging devices inspired by the biological sensory system. They can simultaneously perform sensing and spike conversion, overcoming the deficiencies of traditional sensory systems. This review summarizes and benchmarks the recent progress of artificial sensory neurons. It starts with the presentation of various mechanisms of biological signal transduction, followed by the systematic introduction of the emerging devices employed for artificial sensory neurons. Furthermore, the implementations with different perceptual capabilities are briefly outlined and the key metrics and potential applications are also provided. Finally, we highlight the challenges and perspectives for the future development of artificial sensory neurons.
Collapse
Affiliation(s)
- Shuai Zhong
- Guangdong Institute of Intelligence Science and Technology, Hengqin, Zhuhai, 519031, People's Republic of China.
| | - Lirou Su
- Guangdong Institute of Intelligence Science and Technology, Hengqin, Zhuhai, 519031, People's Republic of China
| | - Mingkun Xu
- Guangdong Institute of Intelligence Science and Technology, Hengqin, Zhuhai, 519031, People's Republic of China
| | - Desmond Loke
- Department of Science, Mathematics and Technology, Singapore University of Technology and Design, Singapore, 487372, Singapore
| | - Bin Yu
- College of Integrated Circuits, Zhejiang University, Hangzhou, 3112000, People's Republic of China
- ZJU-Hangzhou Global Scientific and Technological Innovation Center, Hangzhou, 310027, People's Republic of China
| | - Yishu Zhang
- College of Integrated Circuits, Zhejiang University, Hangzhou, 3112000, People's Republic of China.
- ZJU-Hangzhou Global Scientific and Technological Innovation Center, Hangzhou, 310027, People's Republic of China.
| | - Rong Zhao
- Department of Precision Instruments, Tsinghua University, Beijing, 100084, People's Republic of China
- Center for Brain-Inspired Computing Research, Tsinghua University, Beijing, 100084, People's Republic of China
- IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing, 100084, People's Republic of China
| |
Collapse
|
4
|
McGregor R, Wu MF, Thannickal TC, Li S, Siegel JM. Opioid-induced neuroanatomical, microglial and behavioral changes are blocked by suvorexant without diminishing opioid analgesia. NATURE. MENTAL HEALTH 2024; 2:1018-1031. [PMID: 39989723 PMCID: PMC11845277 DOI: 10.1038/s44220-024-00278-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Accepted: 05/31/2024] [Indexed: 02/25/2025]
Abstract
Heroin use disorder in humans and chronic opioid administration to mice result in an increase in the number and a decrease in the size of detected hypocretin (Hcrt, or orexin) neurons. Chronic morphine administration to mice increases Hcrt axonal projections to the ventral tegmental area (VTA), the level of tyrosine hydroxylase (TH) in VTA and the number of detected TH+ cells in VTA, and activates VTA and hypothalamic microglia. Co-administration of morphine with the dual Hcrt receptor antagonist suvorexant prevents morphine-induced changes in the number and size of Hcrt neurons, the increase in Hcrt projections to the VTA and microglial activation in the VTA and hypothalamus. Co-administration of suvorexant with morphine also prevents morphine anticipatory behavior and reduces opioid withdrawal symptoms. However, suvorexant does not diminish morphine analgesia. Here we show that combined administration of opioids and suvorexant may reduce the addiction potential of opioid use for pain relief in humans while maintaining the analgesic effects of opioids.
Collapse
Affiliation(s)
- Ronald McGregor
- Neuropsychiatric Institute and Brain Research Institute, University of California, Los Angeles, Los Angeles, CA, USA
- VA Greater Los Angeles Healthcare System, Los Angeles, CA, USA
- These authors contributed equally: Ronald McGregor, Ming-Fung Wu
| | - Ming-Fung Wu
- Neuropsychiatric Institute and Brain Research Institute, University of California, Los Angeles, Los Angeles, CA, USA
- VA Greater Los Angeles Healthcare System, Los Angeles, CA, USA
- These authors contributed equally: Ronald McGregor, Ming-Fung Wu
| | - Thomas C. Thannickal
- Neuropsychiatric Institute and Brain Research Institute, University of California, Los Angeles, Los Angeles, CA, USA
- VA Greater Los Angeles Healthcare System, Los Angeles, CA, USA
| | - Songlin Li
- Neuropsychiatric Institute and Brain Research Institute, University of California, Los Angeles, Los Angeles, CA, USA
- VA Greater Los Angeles Healthcare System, Los Angeles, CA, USA
| | - Jerome M. Siegel
- Neuropsychiatric Institute and Brain Research Institute, University of California, Los Angeles, Los Angeles, CA, USA
- VA Greater Los Angeles Healthcare System, Los Angeles, CA, USA
| |
Collapse
|
5
|
Godavarthi SK, Li HQ, Pratelli M, Spitzer NC. Embryonic exposure to environmental factors drives transmitter switching in the neonatal mouse cortex causing autistic-like adult behavior. Proc Natl Acad Sci U S A 2024; 121:e2406928121. [PMID: 39178233 PMCID: PMC11363343 DOI: 10.1073/pnas.2406928121] [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: 04/06/2024] [Accepted: 07/23/2024] [Indexed: 08/25/2024] Open
Abstract
Autism spectrum disorders (ASD) can be caused by environmental factors. These factors act early in the development of the nervous system and induce stereotyped repetitive behaviors and diminished social interactions, among other outcomes. Little is known about how these behaviors are produced. In pregnant women, delivery of valproic acid (VPA) (to control seizure activity or stabilize mood) or immune activation by a virus increases the incidence of ASD in offspring. We found that either VPA or Poly Inosine:Cytosine (which mimics a viral infection), administered at mouse embryonic day 12.5, induced a neurotransmitter switch from GABA to glutamate in PV- and CCK-expressing interneurons in the medial prefrontal cortex by postnatal day 10. The switch was present for only a brief period during early postnatal development, observed in male and female mice at postnatal day 21 and reversed in both males and females by postnatal day 30. At postnatal day 90, male mice exhibited stereotyped repetitive behaviors and diminished social interaction while female mice exhibited only stereotyped repetitive behavior. Transfecting GAD1 in PV- and CCK-expressing interneurons at postnatal day 10, to reintroduce GABA expression, overrode the switch and prevented expression of autistic-like behavior. These findings point to an important role of neurotransmitter switching in mediating the environmental causes of autism.
Collapse
Affiliation(s)
- Swetha K. Godavarthi
- Neurobiology Department, University of California San Diego, La Jolla, CA92093
- Kavli Institute for Brain & Mind, University of California San Diego, La Jolla, CA92093
| | - Hui-quan Li
- Neurobiology Department, University of California San Diego, La Jolla, CA92093
- Kavli Institute for Brain & Mind, University of California San Diego, La Jolla, CA92093
| | - Marta Pratelli
- Neurobiology Department, University of California San Diego, La Jolla, CA92093
- Kavli Institute for Brain & Mind, University of California San Diego, La Jolla, CA92093
| | - Nicholas C. Spitzer
- Neurobiology Department, University of California San Diego, La Jolla, CA92093
- Kavli Institute for Brain & Mind, University of California San Diego, La Jolla, CA92093
| |
Collapse
|
6
|
Dawit H, Zhao Y, Wang J, Pei R. Advances in conductive hydrogels for neural recording and stimulation. Biomater Sci 2024; 12:2786-2800. [PMID: 38682423 DOI: 10.1039/d4bm00048j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/01/2024]
Abstract
The brain-computer interface (BCI) allows the human or animal brain to directly interact with the external environment through the neural interfaces, thus playing the role of monitoring, protecting, improving/restoring, enhancing, and replacing. Recording electrophysiological information such as brain neural signals is of great importance in health monitoring and disease diagnosis. According to the electrode position, it can be divided into non-implantable, semi-implantable, and implantable. Among them, implantable neural electrodes can obtain the highest-quality electrophysiological information, so they have the most promising application. However, due to the chemo-mechanical mismatch between devices and tissues, the adverse foreign body response and performance loss over time seriously restrict the development and application of implantable neural electrodes. Given the challenges, conductive hydrogel-based neural electrodes have recently attracted much attention, owing to many advantages such as good mechanical match with the native tissues, negligible foreign body response, and minimal signal attenuation. This review mainly focuses on the current development of conductive hydrogels as a biocompatible framework for neural tissue and conductivity-supporting substrates for the transmission of electrical signals of neural tissue to speed up electrical regeneration and their applications in neural sensing and recording as well as stimulation.
Collapse
Affiliation(s)
- Hewan Dawit
- School of Nano-Tech and Nano-Bionics, University of Science and Technology of China (USTC), Hefei 230026, PR China
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China.
| | - Yuewu Zhao
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China.
| | - Jine Wang
- College of Medicine and Nursing, Shandong Provincial Engineering Laboratory of Novel Pharmaceutical Excipients, Sustained and Controlled Release Preparations, Dezhou University, China.
- Jiangxi Institute of Nanotechnology, Nanchang, 330200, China
| | - Renjun Pei
- School of Nano-Tech and Nano-Bionics, University of Science and Technology of China (USTC), Hefei 230026, PR China
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China.
| |
Collapse
|
7
|
Mitra R, Richhariya S, Hasan G. Orai-mediated calcium entry determines activity of central dopaminergic neurons by regulation of gene expression. eLife 2024; 12:RP88808. [PMID: 38289659 PMCID: PMC10945566 DOI: 10.7554/elife.88808] [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] [Indexed: 02/01/2024] Open
Abstract
Maturation and fine-tuning of neural circuits frequently require neuromodulatory signals that set the excitability threshold, neuronal connectivity, and synaptic strength. Here, we present a mechanistic study of how neuromodulator-stimulated intracellular Ca2+ signals, through the store-operated Ca2+ channel Orai, regulate intrinsic neuronal properties by control of developmental gene expression in flight-promoting central dopaminergic neurons (fpDANs). The fpDANs receive cholinergic inputs for release of dopamine at a central brain tripartite synapse that sustains flight (Sharma and Hasan, 2020). Cholinergic inputs act on the muscarinic acetylcholine receptor to stimulate intracellular Ca2+ release through the endoplasmic reticulum (ER) localised inositol 1,4,5-trisphosphate receptor followed by ER-store depletion and Orai-mediated store-operated Ca2+ entry (SOCE). Analysis of gene expression in fpDANs followed by genetic, cellular, and molecular studies identified Orai-mediated Ca2+ entry as a key regulator of excitability in fpDANs during circuit maturation. SOCE activates the transcription factor trithorax-like (Trl), which in turn drives expression of a set of genes, including Set2, that encodes a histone 3 lysine 36 methyltransferase (H3K36me3). Set2 function establishes a positive feedback loop, essential for receiving neuromodulatory cholinergic inputs and sustaining SOCE. Chromatin-modifying activity of Set2 changes the epigenetic status of fpDANs and drives expression of key ion channel and signalling genes that determine fpDAN activity. Loss of activity reduces the axonal arborisation of fpDANs within the MB lobe and prevents dopamine release required for the maintenance of long flight.
Collapse
Affiliation(s)
- Rishav Mitra
- National Centre for Biological Sciences, Tata Institute of Fundamental ResearchBangaloreIndia
| | - Shlesha Richhariya
- National Centre for Biological Sciences, Tata Institute of Fundamental ResearchBangaloreIndia
- Department of Biology, Brandeis UniversityWalthamUnited States
| | - Gaiti Hasan
- National Centre for Biological Sciences, Tata Institute of Fundamental ResearchBangaloreIndia
| |
Collapse
|
8
|
Hunter I, Coulson B, Pettini T, Davies JJ, Parkin J, Landgraf M, Baines RA. Balance of activity during a critical period tunes a developing network. eLife 2024; 12:RP91599. [PMID: 38193543 PMCID: PMC10945558 DOI: 10.7554/elife.91599] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2024] Open
Abstract
Developing neural circuits are influenced by activity and are especially sensitive to changes in activity during critical periods (CPs) of development. Changes occurring during a CP often become 'locked in' so that they affect the mature network. Indeed, several neurodevelopmental disorders have been linked to excessive activity during such periods. It is, therefore, important to identify those aspects of neural circuit development that are influenced by neural activity during a CP. In this study, we take advantage of the genetic tractability of Drosophila to show that activity perturbation during an embryonic CP permanently alters properties of the locomotor circuit. Specific changes we identify include increased synchronicity of motoneuron activity and greater strengthening of excitatory over inhibitory synaptic drive to motoneurons. These changes are sufficient to reduce network robustness, evidenced by increased sensitivity to induced seizure. We also show that we can rescue these changes when increased activity is mitigated by inhibition provided by mechanosensory neurons. Similarly, we demonstrate a dose-dependent relationship between inhibition experienced during the CP and the extent to which it is possible to rescue the hyperexcitable phenotype characteristic of the parabss mutation. This suggests that developing circuits must be exposed to a properly balanced sum of excitation and inhibition during the CP to achieve normal mature network function. Our results, therefore, provide novel insight into how activity during a CP shapes specific elements of a circuit, and how activity during this period is integrated to tune neural circuits to the environment in which they will likely function.
Collapse
Affiliation(s)
- Iain Hunter
- Division of Neuroscience, School of Biological Sciences, Faculty of Biology, Medicine and Health,University of ManchesterManchesterUnited Kingdom
| | - Bramwell Coulson
- Division of Neuroscience, School of Biological Sciences, Faculty of Biology, Medicine and Health,University of ManchesterManchesterUnited Kingdom
| | - Tom Pettini
- Department of Zoology, University of CambridgeCambridgeUnited Kingdom
| | - Jacob J Davies
- Division of Neuroscience, School of Biological Sciences, Faculty of Biology, Medicine and Health,University of ManchesterManchesterUnited Kingdom
| | - Jill Parkin
- Division of Neuroscience, School of Biological Sciences, Faculty of Biology, Medicine and Health,University of ManchesterManchesterUnited Kingdom
| | - Matthias Landgraf
- Department of Zoology, University of CambridgeCambridgeUnited Kingdom
| | - Richard A Baines
- Division of Neuroscience, School of Biological Sciences, Faculty of Biology, Medicine and Health,University of ManchesterManchesterUnited Kingdom
| |
Collapse
|
9
|
Hakeem IJ. Molecular docking analysis of acetylcholinesterase inhibitors for Alzheimer's disease management. Bioinformation 2023; 19:565-570. [PMID: 37886145 PMCID: PMC10599677 DOI: 10.6026/97320630019565] [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: 05/01/2023] [Revised: 05/31/2023] [Accepted: 05/31/2023] [Indexed: 10/28/2023] Open
Abstract
Alzheimer's disease (AD) is a neurological disease that is related to aging and is the leading cause of dementia globally. AD has a significant influence on cognitive functions, particularly memory, resulting in a variety of functional deficits. Given the increasing prevalence of AD, there is an urgent need for the development of effective therapeutic therapies. In a quest to uncover a holistic remedy for AD, a total of 41 bioactive compounds derived from three distinct medicinal plant sources were screened to evaluate their potential to inhibit the active sites of acetylcholinesterase (AChE). The insilico screening protocol included 24 licorice-derived compounds, 5 ginkgo biloba-derived compounds, and 11 ginseng-derived compounds. Two compounds (Ginkgolide A and Licorice glycoside D2) were observed to display greater binding energy (BE) relative to the control by interacting with crucial residues in the active site of AChE. Ginkgolide A and Licorice glycoside D2 exhibited BEs of -11.3 and -11.2 kcal/mol, respectively, whereas the control, Donepezil, demonstrated a BE of -10.8 kcal/mol. Further, these compounds exhibit favorable drug-likeness properties. This study suggests that further experimental investigations can be conducted on Ginkgolide A and Licorice glycoside D2 to explore their potential therapeutic applications for AD.
Collapse
Affiliation(s)
- Israa J Hakeem
- Department of Biochemistry, College of Science, University of Jeddah, Jeddah, Saudi Arabia
| |
Collapse
|
10
|
Suárez R, Bluett T, McCullough MH, Avitan L, Black DA, Paolino A, Fenlon LR, Goodhill GJ, Richards LJ. Cortical activity emerges in region-specific patterns during early brain development. Proc Natl Acad Sci U S A 2023; 120:e2208654120. [PMID: 37216522 PMCID: PMC10235933 DOI: 10.1073/pnas.2208654120] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2022] [Accepted: 04/17/2023] [Indexed: 05/24/2023] Open
Abstract
The development of precise neural circuits in the brain requires spontaneous patterns of neural activity prior to functional maturation. In the rodent cerebral cortex, patchwork and wave patterns of activity develop in somatosensory and visual regions, respectively, and are present at birth. However, whether such activity patterns occur in noneutherian mammals, as well as when and how they arise during development, remain open questions relevant for understanding brain formation in health and disease. Since the onset of patterned cortical activity is challenging to study prenatally in eutherians, here we offer an approach in a minimally invasive manner using marsupial dunnarts, whose cortex forms postnatally. We discovered similar patchwork and travelling waves in the dunnart somatosensory and visual cortices at stage 27 (equivalent to newborn mice) and examined earlier stages of development to determine the onset of these patterns and how they first emerge. We observed that these patterns of activity emerge in a region-specific and sequential manner, becoming evident as early as stage 24 in somatosensory and stage 25 in visual cortices (equivalent to embryonic day 16 and 17, respectively, in mice), as cortical layers establish and thalamic axons innervate the cortex. In addition to sculpting synaptic connections of existing circuits, evolutionarily conserved patterns of neural activity could therefore help regulate other early events in cortical development.
Collapse
Affiliation(s)
- Rodrigo Suárez
- The University of Queensland, Queensland Brain Institute, BrisbaneQLD4072, Australia
- The University of Queensland, School of Biomedical Sciences, BrisbaneQLD4072, Australia
| | - Tobias Bluett
- The University of Queensland, Queensland Brain Institute, BrisbaneQLD4072, Australia
| | - Michael H. McCullough
- The University of Queensland, Queensland Brain Institute, BrisbaneQLD4072, Australia
| | - Lilach Avitan
- The University of Queensland, Queensland Brain Institute, BrisbaneQLD4072, Australia
| | - Dylan A. Black
- The University of Queensland, Queensland Brain Institute, BrisbaneQLD4072, Australia
- The University of Queensland, School of Biomedical Sciences, BrisbaneQLD4072, Australia
| | - Annalisa Paolino
- The University of Queensland, Queensland Brain Institute, BrisbaneQLD4072, Australia
- The University of Queensland, School of Biomedical Sciences, BrisbaneQLD4072, Australia
| | - Laura R. Fenlon
- The University of Queensland, Queensland Brain Institute, BrisbaneQLD4072, Australia
- The University of Queensland, School of Biomedical Sciences, BrisbaneQLD4072, Australia
| | - Geoffrey J. Goodhill
- The University of Queensland, Queensland Brain Institute, BrisbaneQLD4072, Australia
- The University of Queensland, School of Mathematics and Physics, BrisbaneQLD4072, Australia
| | - Linda J. Richards
- The University of Queensland, Queensland Brain Institute, BrisbaneQLD4072, Australia
- The University of Queensland, School of Biomedical Sciences, BrisbaneQLD4072, Australia
| |
Collapse
|
11
|
Tian H, Wang C, Chen Y, Zheng L, Jing H, Xu L, Wang X, Liu Y, Hao J. Optically modulated ionic conductivity in a hydrogel for emulating synaptic functions. SCIENCE ADVANCES 2023; 9:eadd6950. [PMID: 36791203 PMCID: PMC9931204 DOI: 10.1126/sciadv.add6950] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Accepted: 01/11/2023] [Indexed: 06/18/2023]
Abstract
Ion-conductive hydrogels, with ions as signal carriers, have become promising candidates to construct functional ionotronics for sensing, actuating, and robotics engineering. However, rational modulation of ionic migration to mimic biological information processing, including learning and memory, remains challenging to be realized in hydrogel materials. Here, we develop a hybrid hydrogel with optically modulated ionic conductivity to emulate the functions of a biological synapse. Through a responsive supramolecular approach, optical stimuli can trigger the release of mobile ions for tuning the conductivity of the hydrogel, which is analogous to the modulation of synaptic plasticity. As a proof of concept, this hydrogel can be used as an information processing unit to perceive different optical stimuli and regulate the grasping motion of a robotic hand, performing logical motion feedback with "learning-experience" function. Our ionic hydrogel provides a valuable strategy toward developing bioinspired ionotronic systems and pushes forward the functional applications of hydrogel materials.
Collapse
Affiliation(s)
- Huasheng Tian
- Key Laboratory of Colloid and Interface Chemistry of the Ministry of Education, School of Chemistry and Chemical Engineering, Shandong University, Jinan, Shandong, China
- Suzhou Research Institute, Shandong University, Suzhou, Jiangsu, China
| | - Chen Wang
- Key Laboratory of Colloid and Interface Chemistry of the Ministry of Education, School of Chemistry and Chemical Engineering, Shandong University, Jinan, Shandong, China
| | - Yuwei Chen
- Key Laboratory of Colloid and Interface Chemistry of the Ministry of Education, School of Chemistry and Chemical Engineering, Shandong University, Jinan, Shandong, China
| | - Liping Zheng
- Key Laboratory of Colloid and Interface Chemistry of the Ministry of Education, School of Chemistry and Chemical Engineering, Shandong University, Jinan, Shandong, China
| | - Houchao Jing
- Key Laboratory of Colloid and Interface Chemistry of the Ministry of Education, School of Chemistry and Chemical Engineering, Shandong University, Jinan, Shandong, China
- Suzhou Research Institute, Shandong University, Suzhou, Jiangsu, China
| | - Lin Xu
- Key Laboratory of Colloid and Interface Chemistry of the Ministry of Education, School of Chemistry and Chemical Engineering, Shandong University, Jinan, Shandong, China
- Suzhou Research Institute, Shandong University, Suzhou, Jiangsu, China
| | - Xuanqi Wang
- School of Software, Shandong University, Jinan, Shandong, China
| | - Yaqing Liu
- Key Laboratory of Colloid and Interface Chemistry of the Ministry of Education, School of Chemistry and Chemical Engineering, Shandong University, Jinan, Shandong, China
- Suzhou Research Institute, Shandong University, Suzhou, Jiangsu, China
| | - Jingcheng Hao
- Key Laboratory of Colloid and Interface Chemistry of the Ministry of Education, School of Chemistry and Chemical Engineering, Shandong University, Jinan, Shandong, China
| |
Collapse
|
12
|
Ncube D, Tallafuss A, Serafin J, Bruckner J, Farnsworth DR, Miller AC, Eisen JS, Washbourne P. A conserved transcriptional fingerprint of multi-neurotransmitter neurons necessary for social behavior. BMC Genomics 2022; 23:675. [PMID: 36175871 PMCID: PMC9523972 DOI: 10.1186/s12864-022-08879-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 09/02/2022] [Indexed: 11/11/2022] Open
Abstract
Background An essential determinant of a neuron’s functionality is its neurotransmitter phenotype. We previously identified a defined subpopulation of cholinergic neurons required for social orienting behavior in zebrafish. Results We transcriptionally profiled these neurons and discovered that they are capable of synthesizing both acetylcholine and GABA. We also established a constellation of transcription factors and neurotransmitter markers that can be used as a “transcriptomic fingerprint” to recognize a homologous neuronal population in another vertebrate. Conclusion Our results suggest that this transcriptomic fingerprint and the cholinergic-GABAergic neuronal subtype that it defines are evolutionarily conserved. Supplementary Information The online version contains supplementary material available at 10.1186/s12864-022-08879-w.
Collapse
Affiliation(s)
- Denver Ncube
- Institute of Neuroscience, 1254 University of Oregon, Eugene, OR, 97403, USA
| | - Alexandra Tallafuss
- Institute of Neuroscience, 1254 University of Oregon, Eugene, OR, 97403, USA
| | - Jen Serafin
- Institute of Neuroscience, 1254 University of Oregon, Eugene, OR, 97403, USA
| | - Joseph Bruckner
- Institute of Neuroscience, 1254 University of Oregon, Eugene, OR, 97403, USA
| | - Dylan R Farnsworth
- Institute of Neuroscience, 1254 University of Oregon, Eugene, OR, 97403, USA
| | - Adam C Miller
- Institute of Neuroscience, 1254 University of Oregon, Eugene, OR, 97403, USA
| | - Judith S Eisen
- Institute of Neuroscience, 1254 University of Oregon, Eugene, OR, 97403, USA
| | - Philip Washbourne
- Institute of Neuroscience, 1254 University of Oregon, Eugene, OR, 97403, USA.
| |
Collapse
|
13
|
Salceda R. Glycine neurotransmission: Its role in development. Front Neurosci 2022; 16:947563. [PMID: 36188468 PMCID: PMC9525178 DOI: 10.3389/fnins.2022.947563] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 08/30/2022] [Indexed: 11/13/2022] Open
Abstract
The accurate function of the central nervous system (CNS) depends of the consonance of multiple genetic programs and external signals during the ontogenesis. A variety of molecules including neurotransmitters, have been implied in the regulation of proliferation, survival, and cell-fate of neurons and glial cells. Among these, neurotransmitters may play a central role since functional ligand-gated ionic channel receptors have been described before the establishment of synapses. This review argues on the function of glycine during development, and show evidence indicating it regulates morphogenetic events by means of their transporters and receptors, emphasizing the role of glycinergic activity in the balance of excitatory and inhibitory signals during development. Understanding the mechanisms involved in these processes would help us to know the etiology of cognitive dysfunctions and lead to improve brain repair strategies.
Collapse
|
14
|
Molecular Mechanism and Regulation of Autophagy and Its Potential Role in Epilepsy. Cells 2022; 11:cells11172621. [PMID: 36078029 PMCID: PMC9455075 DOI: 10.3390/cells11172621] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 08/14/2022] [Accepted: 08/22/2022] [Indexed: 01/18/2023] Open
Abstract
Autophagy is an evolutionally conserved degradation mechanism for maintaining cell homeostasis whereby cytoplasmic components are wrapped in autophagosomes and subsequently delivered to lysosomes for degradation. This process requires the concerted actions of multiple autophagy-related proteins and accessory regulators. In neurons, autophagy is dynamically regulated in different compartments including soma, axons, and dendrites. It determines the turnover of selected materials in a spatiotemporal control manner, which facilitates the formation of specialized neuronal functions. It is not surprising, therefore, that dysfunctional autophagy occurs in epilepsy, mainly caused by an imbalance between excitation and inhibition in the brain. In recent years, much attention has been focused on how autophagy may cause the development of epilepsy. In this article, we overview the historical landmarks and distinct types of autophagy, recent progress in the core machinery and regulation of autophagy, and biological roles of autophagy in homeostatic maintenance of neuronal structures and functions, with a particular focus on synaptic plasticity. We also discuss the relevance of autophagy mechanisms to the pathophysiology of epileptogenesis.
Collapse
|
15
|
Li G, Shi Y, Zhang L, Yang C, Wan T, Lv H, Jian W, Li J, Li M. Efficacy of acupuncture in animal models of vascular dementia: A systematic review and network meta-analysis. Front Aging Neurosci 2022; 14:952181. [PMID: 36062145 PMCID: PMC9434110 DOI: 10.3389/fnagi.2022.952181] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 07/14/2022] [Indexed: 12/09/2022] Open
Abstract
Background and purpose Acupuncture is widely used in clinical practice for the treatment of vascular diseases. However, the protocol, efficacy, and mechanism of acupuncture in animal models of vascular dementia are still controversial. Based on the above problems, we initiated this comprehensive study. Methods To analyze the literatures included in this study, 4 databases were searched and the SYRCLE's Risk of bias tool was employed. To perform the subgroup analysis of different acupuncture methods and the Review Manager 5.3 was applied. Meanwhile, the pairwise and network meta-analysis were conducted using Addis 1.16.8. The outcomes included escape latency, number of crossings, time spent in the target quadrant, and swimming speed. Results Forty-two studies with a total of 1,486 animals were included in this meta-analysis. According to the results from subgroup analysis, GV20 + ST36 (Baihui + bilateral Zusanli) combined with 14-day manual acupuncture can obtain best improvement of the rats cognitive function among all acupuncture regimens (MD: -23.41; 95%CI: -26.66, -20.15; I2 = 0%; P < 0.001). The heterogeneity of other acupuncture treatments was significantly higher than that of GV20 + ST36, because the treatment courses were not uniform. Pair-wise and network comparisons are highly consistent. The major results of the network meta-analysis were as follows, In comparison to the impaired group, the acupuncture group showed significantly reduced escape latency (MD: -25.87; 95%CI: -30.75, -21.12), increased number of original platform crossings (MD: 2.63; 95%CI: 1.94, 3.34) and time spent in the target quadrant (MD: 7.88; 95%CI: 4.25, 11.44). The overall results of the network meta-analysis are as follows: the normal and sham-operated groups performed the best, followed by medicine and acupuncture, while no effect was found in the impaired group treated with non-acupoint and palliative. Conclusions Acupuncture significantly improves cognitive function in rats with vascular dementia. Compared to other acupuncture plans, (GV20 + ST36, MA) and 14 -day manual acupuncture can be used to obtain better results. The main mechanism of acupuncture in the treatment of vascular dementia is reduced oxidative stress, neuronal inflammation, and apoptosis, as well as the increased synaptic plasticity and neurotransmitters. Systematic review registration https://inplasy.com/inplasy-2021-11-0036/, identifier: INPLASY2021110036.
Collapse
Affiliation(s)
- Guangyao Li
- Medical College of Acupuncture Moxibustion and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, China
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yuling Shi
- The Second Affiliated Hospital, Guangzhou University of Chinese Medicine (Guangdong Hospital of Traditional Chinese Medicine), Guangzhou, China
| | - Lu Zhang
- Medical College of Acupuncture Moxibustion and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Chuanghui Yang
- Medical College of Acupuncture Moxibustion and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Ting Wan
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Hang Lv
- Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Wenxuan Jian
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jinghu Li
- Department of Massage, The Third Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Min Li
- Medical College of Acupuncture Moxibustion and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
16
|
High-frequency stimulation of the subthalamic nucleus induces a sustained inhibition of serotonergic system via loss of cell phenotype. Sci Rep 2022; 12:14011. [PMID: 35978112 PMCID: PMC9385659 DOI: 10.1038/s41598-022-18294-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 08/09/2022] [Indexed: 11/08/2022] Open
Abstract
Deep brain stimulation (DBS) of the subthalamic nucleus (STN) has become a standard treatment for Parkinson's disease (PD). However, in a considerable number of patients debilitating psychiatric side-effects occur. Recent research has revealed that external stimuli can alter the neurotransmitters' homeostasis in neurons, which is known as "neurotransmitter respecification". Herein, we addressed if neurotransmitter respecification could be a mechanism by which DBS suppresses the serotonergic function in the dorsal raphe nucleus (DRN) leading to mood changes. We infused transgenic 5-HT-Cre (ePET-Cre) mice with AAV viruses to achieve targeted expression of eYFP and the genetically encoded calcium indicator GCaMP6s in the DRN prior to methyl-4phenyl-1,2,3,6-tetrahydropyridine (MPTP) treatment. Mice received bilateral DBS electrodes in the STN and an optic fiber in the DRN for calcium photometry. MPTP-treated mice demonstrated behavioral and histological PD phenotype, whereas all STN-DBS animals exhibited an increased immobility time in the forced swim test, reduced calcium activity, and loss of tryptophan hydroxylase-2 expression in the DRN. Given the prominent role of calcium transients in mediating neurotransmitter respecification, these results suggest a loss of serotonergic phenotype in the DRN following STN-DBS. These findings indicate that loss of serotonergic cell phenotype may underlie the unwanted depressive symptoms following STN-DBS.
Collapse
|
17
|
Chen Q, Shen W, Sun H, Zhang H, Liu C, Chen Z, Yu L, Cai X, Ke J, Li L, Zhang L, Fang Q. The effect of coupled inhibitory-facilitatory repetitive transcranial magnetic stimulation on shaping early reorganization of the motor network after stroke. Brain Res 2022; 1790:147959. [PMID: 35654120 DOI: 10.1016/j.brainres.2022.147959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 05/25/2022] [Accepted: 05/27/2022] [Indexed: 11/29/2022]
Abstract
Neural plasticity is a major factor driving cortical reorganization after stroke. This study aimed to evaluate functional connectivity (FC) changes in the cortical motor network after coupled inhibitory-facilitatory repetitive transcranial magnetic stimulation (rTMS) treatment and to assess the correlation between FC changes and functional recovery, further characterizing the neural mechanisms underlying the beneficial effects of rTMS. We randomly divided 63 patients with acute stroke into four groups: (1) Group A received coupled inhibitory-facilitatory rTMS [1 Hz over the contralesional primary motor cortex (M1) and 10 Hz over ipsilesional M1]; (2) Group B received a contralesional sham stimulation and ipsilesional 10 Hz stimulation; (3) Group C received a contralesional 1 Hz rTMS and ipsilesional sham stimulation; and (4) Group D received bilateral sham stimulation only. Standardized rehabilitation therapy was performed immediately after rTMS, and each group was treated with their respective treatment modalities for 4 weeks. Twenty-four hours before and after the intervention, participants underwent resting-state functional magnetic resonance imaging. Additional functional assessments were conducted at baseline, after treatment, and at the 3 month follow-up. The rTMS treatment significantly changed the FCs of intra- and inter-hemispheric cortical motor networks in the rTMS groups (A and B) compared with the sham group (Group D). This effect was more pronounced in Group A, which displayed a changed FC between the contralesional postcentral gyrus and contralesional superior parietal gyrus, between the contralesional precentral gyrus and contralesional postcentral gyrus, and between the ipsilesional postcentral gyrus and contralesional superior parietal gyrus, when compared with Groups B and C. Importantly, FC changes were significantly correlated with improvement of motor function. In the early stages of ischemic stroke, coupled rTMS was more conducive to motor recovery by modulating the FCs of intra-hemispheric and inter-hemispheric motor networks. Our results suggested that FC changes were related to motor function recovery for early-stage cerebral stroke patients treated with coupled rTMS. These findings could help to understand the mechanism of coupled rTMS and further the use of this therapy as an adjunct rehabilitation technique in motor recovery.
Collapse
Affiliation(s)
- Qingmei Chen
- Department of Physical Medicine &Rehabilitation, The First Affiliated Hospital of Soochow University, Suzhou, 215000, Jiangsu Province, China; Department of Neurology, The First Affiliated Hospital of Soochow University, Suzhou, 215000, Jiangsu Province, China
| | - Wenjun Shen
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Haiwei Sun
- Department of Emergency Medicine, The Second Affiliated Hospital of Soochow University, Suzhou, 215000, Jiangsu Province, China
| | - Hanjun Zhang
- Department of Physical Medicine &Rehabilitation, The First Affiliated Hospital of Soochow University, Suzhou, 215000, Jiangsu Province, China
| | - Chuandao Liu
- Department of Physical Medicine &Rehabilitation, The First Affiliated Hospital of Soochow University, Suzhou, 215000, Jiangsu Province, China
| | - Zhiguo Chen
- Department of Neurology, The First Affiliated Hospital of Soochow University, Suzhou, 215000, Jiangsu Province, China
| | - Liqiang Yu
- Department of Neurology, The First Affiliated Hospital of Soochow University, Suzhou, 215000, Jiangsu Province, China
| | - Xiuying Cai
- Department of Neurology, The First Affiliated Hospital of Soochow University, Suzhou, 215000, Jiangsu Province, China
| | - Jun Ke
- Department of Radiology, The First Affiliated Hospital of Soochow University, Suzhou, 215000, Jiangsu Province, China
| | - Li Li
- Department of Physical Medicine &Rehabilitation, The First Affiliated Hospital of Soochow University, Suzhou, 215000, Jiangsu Province, China.
| | - Lichi Zhang
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200030, China.
| | - Qi Fang
- Department of Neurology, The First Affiliated Hospital of Soochow University, Suzhou, 215000, Jiangsu Province, China.
| |
Collapse
|
18
|
Pumo GM, Kitazawa T, Rijli FM. Epigenetic and Transcriptional Regulation of Spontaneous and Sensory Activity Dependent Programs During Neuronal Circuit Development. Front Neural Circuits 2022; 16:911023. [PMID: 35664458 PMCID: PMC9158562 DOI: 10.3389/fncir.2022.911023] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 04/28/2022] [Indexed: 11/13/2022] Open
Abstract
Spontaneous activity generated before the onset of sensory transduction has a key role in wiring developing sensory circuits. From axonal targeting, to synapse formation and elimination, to the balanced integration of neurons into developing circuits, this type of activity is implicated in a variety of cellular processes. However, little is known about its molecular mechanisms of action, especially at the level of genome regulation. Conversely, sensory experience-dependent activity implements well-characterized transcriptional and epigenetic chromatin programs that underlie heterogeneous but specific genomic responses that shape both postnatal circuit development and neuroplasticity in the adult. In this review, we focus on our knowledge of the developmental processes regulated by spontaneous activity and the underlying transcriptional mechanisms. We also review novel findings on how chromatin regulates the specificity and developmental induction of the experience-dependent program, and speculate their relevance for our understanding of how spontaneous activity may act at the genomic level to instruct circuit assembly and prepare developing neurons for sensory-dependent connectivity refinement and processing.
Collapse
Affiliation(s)
- Gabriele M. Pumo
- Laboratory of Neurodevelopmental Epigenetics, Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland
- Department Biozentrum, University of Basel, Basel, Switzerland
| | - Taro Kitazawa
- Laboratory of Neurodevelopmental Epigenetics, Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland
| | - Filippo M. Rijli
- Laboratory of Neurodevelopmental Epigenetics, Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland
- Department Biozentrum, University of Basel, Basel, Switzerland
| |
Collapse
|
19
|
Adaptive erasure of spurious sequences in sensory cortical circuits. Neuron 2022; 110:1857-1868.e5. [PMID: 35358415 PMCID: PMC9616807 DOI: 10.1016/j.neuron.2022.03.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 11/12/2021] [Accepted: 03/07/2022] [Indexed: 12/02/2022]
Abstract
Sequential activity reflecting previously experienced temporal sequences is considered a hallmark of learning across cortical areas. However, it is unknown how cortical circuits avoid the converse problem: producing spurious sequences that are not reflecting sequences in their inputs. We develop methods to quantify and study sequentiality in neural responses. We show that recurrent circuit responses generally include spurious sequences, which are specifically prevented in circuits that obey two widely known features of cortical microcircuit organization: Dale’s law and Hebbian connectivity. In particular, spike-timing-dependent plasticity in excitation-inhibition networks leads to an adaptive erasure of spurious sequences. We tested our theory in multielectrode recordings from the visual cortex of awake ferrets. Although responses to natural stimuli were largely non-sequential, responses to artificial stimuli initially included spurious sequences, which diminished over extended exposure. These results reveal an unexpected role for Hebbian experience-dependent plasticity and Dale’s law in sensory cortical circuits. Recurrent circuits generate spurious sequences without sequential inputs A principled measure of total sequentiality in population responses is developed Theory predicts that Hebbian plasticity should abolish spurious sequences Spurious sequences in the visual cortex diminish with experience
Collapse
|
20
|
Shan L, Swaab DF. Changes in histaminergic system in neuropsychiatric disorders and the potential treatment consequences. Curr Neuropharmacol 2021; 20:403-411. [PMID: 34521328 PMCID: PMC9413789 DOI: 10.2174/1570159x19666210909144930] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 08/05/2021] [Accepted: 08/26/2021] [Indexed: 11/22/2022] Open
Abstract
In contrast to that of other monoamine neurotransmitters, the association of the histaminergic system with neuropsychiatric disorders is not well documented. In the last two decades, several clinical studies involved in the development of drugs targeting the histaminergic system have been reported. These include the H3R-antagonist/inverse agonist, pitolisant, used for the treatment of excessive sleepiness in narcolepsy, and the H1R antagonist, doxepin, used to alleviate symptoms of insomnia. The current review summarizes reports from animal models, including genetic and neuroimaging studies, as well as human brain samples and cerebrospinal fluid measurements from clinical trials, on the possible role of the histaminergic system in neuropsychiatric disorders. These studies will potentially pave the way for novel histamine-related therapeutic strategies.
Collapse
Affiliation(s)
- Ling Shan
- Department of Neuropsychiatric Disorders, Netherlands Institute for Neuroscience, an Institute of the Royal Netherlands Academy of Arts and Sciences, Amsterdam. Netherlands
| | - Dick F Swaab
- Department of Neuropsychiatric Disorders, Netherlands Institute for Neuroscience, an Institute of the Royal Netherlands Academy of Arts and Sciences, Amsterdam. Netherlands
| |
Collapse
|
21
|
Chen Q, Shen W, Sun H, Shen D, Cai X, Ke J, Zhang L, Fang Q. Effects of mirror therapy on motor aphasia after acute cerebral infarction: A randomized controlled trial. NeuroRehabilitation 2021; 49:103-117. [PMID: 34180428 DOI: 10.3233/nre-210125] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Mirror therapy (MT) has proven to be beneficial for treating patients suffering from motor aphasia after stroke. However, the impacts of MT on neuroplasticity remain unexplored. OBJECTIVE In this paper we conducted a randomized controlled trial to evaluate the treatment using the MT on motor aphasia following acute cerebral infarction. METHODS We randomly assigned 30 patients into test and control groups, with test group patients treated with MT, whereas control group patients were treated with sham MT. At 24 hours prior to and after the intervention, we obtained functional magnetic resonance imaging (fMRI) data from study subjects. At baseline, after treatment and 12-week follow-up, we additionally evaluated patients with the Modified Rankin Scale (mRS), the National Institutes of Health Stroke Scale (NIHSS), and the aphasia quotient (AQ) in the western aphasia test. RESULTS After 2 weeks of treatment, the test group demonstrated significant improvements in AQ values, naming, repetition, spontaneous speech, and mRS scores compared to the control group (P < 0.05). Furthermore, in the follow-up time point (12 weeks), we found that the test group exhibited significantly better NIHSS scores and AQ evaluation indicators than the control group (P < 0.05). Specifically, the fMRI study shows that functional connectivity significantly improved in test group patients mainly among frontal, temporal, and parietal lobes of the left hemisphere with each other than controls group. Meanwhile, we found significantly enhanced functional connectivity with the hippocampus (P < 0.01). CONCLUSIONS Our results indicate that the MT can expedite the recovery of language function during the early phases of stroke recovery. These findings may elucidate the underlying mechanism of MT and the application of this therapy as an adjunct rehabilitation technique in language recovery.
Collapse
Affiliation(s)
- Qingmei Chen
- Department of Neurology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China.,Department of Physical Medicine & Rehabilitation, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Wenjun Shen
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Haiwei Sun
- Department of Emergency Medicine, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Dan Shen
- Department of Neurology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Xiuying Cai
- Department of Neurology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Jun Ke
- Department of Radiology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Lichi Zhang
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Qi Fang
- Department of Neurology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| |
Collapse
|
22
|
Reyes-Pinto R, Ferrán JL, Vega-Zuniga T, González-Cabrera C, Luksch H, Mpodozis J, Puelles L, Marín GJ. Change in the neurochemical signature and morphological development of the parvocellular isthmic projection to the avian tectum. J Comp Neurol 2021; 530:553-573. [PMID: 34363623 DOI: 10.1002/cne.25229] [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: 05/04/2021] [Revised: 07/27/2021] [Accepted: 07/30/2021] [Indexed: 11/05/2022]
Abstract
Neurons can change their classical neurotransmitters during ontogeny, sometimes going through stages of dual release. Here, we explored the development of the neurotransmitter identity of neurons of the avian nucleus isthmi parvocellularis (Ipc), whose axon terminals are retinotopically arranged in the optic tectum (TeO) and exert a focal gating effect upon the ascending transmission of retinal inputs. Although cholinergic and glutamatergic markers are both found in Ipc neurons and terminals of adult pigeons and chicks, the mRNA expression of the vesicular acetylcholine transporter, VAChT, is weak or absent. To explore how the Ipc neurotransmitter identity is established during ontogeny, we analyzed the expression of mRNAs coding for cholinergic (ChAT, VAChT, and CHT) and glutamatergic (VGluT2 and VGluT3) markers in chick embryos at different developmental stages. We found that between E12 and E18, Ipc neurons expressed all cholinergic mRNAs and also VGluT2 mRNA; however, from E16 through posthatch stages, VAChT mRNA expression was specifically diminished. Our ex vivo deposits of tracer crystals and intracellular filling experiments revealed that Ipc axons exhibit a mature paintbrush morphology late in development, experiencing marked morphological transformations during the period of presumptive dual vesicular transmitter release. Additionally, although ChAT protein immunoassays increasingly label the growing Ipc axon, this labeling was consistently restricted to sparse portions of the terminal branches. Combined, these results suggest that the synthesis of glutamate and acetylcholine, and their vesicular release, is complexly linked to the developmental processes of branching, growing and remodeling of these unique axons.
Collapse
Affiliation(s)
- Rosana Reyes-Pinto
- Departamento de Biología, Facultad de Ciencias, Universidad de Chile, Santiago, Chile
| | - José L Ferrán
- Department of Human Anatomy and Psychobiology and IMIB-Arrixaca Institute, University of Murcia, Murcia, Spain
| | - Tomas Vega-Zuniga
- Lehrstuhl für Zoologie, Technical University of Munich, Freising, Germany.,Institute of Science and Technology Austria (IST Austria), Klosterneuburg, Austria
| | | | - Harald Luksch
- Lehrstuhl für Zoologie, Technical University of Munich, Freising, Germany
| | - Jorge Mpodozis
- Departamento de Biología, Facultad de Ciencias, Universidad de Chile, Santiago, Chile
| | - Luis Puelles
- Department of Human Anatomy and Psychobiology and IMIB-Arrixaca Institute, University of Murcia, Murcia, Spain
| | - Gonzalo J Marín
- Departamento de Biología, Facultad de Ciencias, Universidad de Chile, Santiago, Chile.,Facultad de Medicina, Universidad Finis Terrae, Santiago, Chile
| |
Collapse
|
23
|
Ahmad SS, Khan MB, Ahmad K, Lim JH, Shaikh S, Lee EJ, Choi I. Biocomputational Screening of Natural Compounds against Acetylcholinesterase. Molecules 2021; 26:2641. [PMID: 33946559 PMCID: PMC8125523 DOI: 10.3390/molecules26092641] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 04/27/2021] [Accepted: 04/29/2021] [Indexed: 12/19/2022] Open
Abstract
Alzheimer's disease (AD) is the most common form of dementia and is characterized by irreversible and progressive neurodegeneration. Cholinergic dysfunction has been reported in AD, and several cholinesterase inhibitors, including natural compounds and synthetic analogs, have been developed to treat the disease. However, there is currently no treatment for AD, as most drug-like compounds have failed in clinical trials. Acetylcholinesterase (AChE) is the target of most drugs used commercially to treat AD. This work focused on screening natural compounds obtained from the ZINC database (224, 205 compounds) against AChE to identify those possibly capable of enabling the management of AD. Indirubin and dehydroevodiamine were the best potential AChE inhibitors with free binding energies of -10.03 and -9.00 kcal/mol, respectively. The key residue (His447) of the active site of AChE was found to participate in complex interactions with these two molecules. Six H-bonds were involved in the 'indirubin-AChE' interaction and three H-bonds in the 'dehydroevodiamine-AChE' interaction. These compounds were predicted to cross the blood-brain barrier (BBB) and to exhibit high levels of intestinal absorption. Furthermore, 'indirubin-AChE' and 'dehydroevodiamine-AChE' complexes were found to be stable, as determined by root mean square deviation (RMSD) during a 50 ns molecular dynamics simulation study. Based on the free binding energies and stabilities obtained by simulation studies, we recommend that experimental studies be undertaken on indirubin and dehydroevodiamine with a view towards their potential use as treatments for AD.
Collapse
Affiliation(s)
- Syed Sayeed Ahmad
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan 38541, Korea; (S.S.A.); (J.-H.L.); (S.S.); (E.-J.L.)
- Research Institute of Cell Culture, Yeungnam University, Gyeongsan 38541, Korea;
| | - Mohd Babu Khan
- Centre for Bioinformatics, School of Life Sciences, Pondicherry University, Puducherry 605014, India;
| | - Khurshid Ahmad
- Research Institute of Cell Culture, Yeungnam University, Gyeongsan 38541, Korea;
| | - Jeong-Ho Lim
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan 38541, Korea; (S.S.A.); (J.-H.L.); (S.S.); (E.-J.L.)
| | - Sibhghatulla Shaikh
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan 38541, Korea; (S.S.A.); (J.-H.L.); (S.S.); (E.-J.L.)
- Research Institute of Cell Culture, Yeungnam University, Gyeongsan 38541, Korea;
| | - Eun-Ju Lee
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan 38541, Korea; (S.S.A.); (J.-H.L.); (S.S.); (E.-J.L.)
- Research Institute of Cell Culture, Yeungnam University, Gyeongsan 38541, Korea;
| | - Inho Choi
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan 38541, Korea; (S.S.A.); (J.-H.L.); (S.S.); (E.-J.L.)
- Research Institute of Cell Culture, Yeungnam University, Gyeongsan 38541, Korea;
| |
Collapse
|
24
|
Chantranupong L, Saulnier JL, Wang W, Jones DR, Pacold ME, Sabatini BL. Rapid purification and metabolomic profiling of synaptic vesicles from mammalian brain. eLife 2020; 9:59699. [PMID: 33043885 PMCID: PMC7575323 DOI: 10.7554/elife.59699] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Accepted: 10/11/2020] [Indexed: 12/14/2022] Open
Abstract
Neurons communicate by the activity-dependent release of small-molecule neurotransmitters packaged into synaptic vesicles (SVs). Although many molecules have been identified as neurotransmitters, technical limitations have precluded a full metabolomic analysis of SV content. Here, we present a workflow to rapidly isolate SVs and to interrogate their metabolic contents at high-resolution using mass spectrometry. We validated the enrichment of glutamate in SVs of primary cortical neurons using targeted polar metabolomics. Unbiased and extensive global profiling of SVs isolated from these neurons revealed that the only detectable polar metabolites they contain are the established neurotransmitters glutamate and GABA. In addition, we adapted the approach to enable quick capture of SVs directly from brain tissue and determined the neurotransmitter profiles of diverse brain regions in a cell-type-specific manner. The speed, robustness, and precision of this method to interrogate SV contents will facilitate novel insights into the chemical basis of neurotransmission.
Collapse
Affiliation(s)
- Lynne Chantranupong
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, Boston, United States
| | - Jessica L Saulnier
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, Boston, United States
| | - Wengang Wang
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, Boston, United States
| | - Drew R Jones
- New York University School of Medicine, Metabolomics Core Resource Laboratory at NYU Langone Health, New York, United States
| | - Michael E Pacold
- Department of Radiation Oncology, New York University Langone Medical Center, New York, United States
| | - Bernardo L Sabatini
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, Boston, United States
| |
Collapse
|
25
|
Sar-El R, Sharon H, Lubianiker N, Hendler T, Raz G. Inducing a Functional-Pharmacological Coupling in the Human Brain to Achieve Improved Drug Effect. Front Neurosci 2020; 14:557874. [PMID: 33154714 PMCID: PMC7586318 DOI: 10.3389/fnins.2020.557874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 09/07/2020] [Indexed: 11/13/2022] Open
Affiliation(s)
- Roy Sar-El
- Sagol Brain Institute, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- *Correspondence: Roy Sar-El,
| | - Haggai Sharon
- Sagol Brain Institute, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Nitzan Lubianiker
- Sagol Brain Institute, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
- The School of Psychological Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Talma Hendler
- Sagol Brain Institute, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- The School of Psychological Sciences, Tel Aviv University, Tel Aviv, Israel
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Gal Raz
- Sagol Brain Institute, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
- Steve Tisch School of Film and Television, Tel Aviv University, Tel Aviv, Israel
- Gal Raz,
| |
Collapse
|
26
|
Abstract
Neurotransmitter switching is a form of brain plasticity in which an environmental stimulus causes neurons to replace one neurotransmitter with another, often resulting in changes in behavior. This raises the possibility of applying a specific environmental stimulus to induce a switch that can enhance a desirable behavior or ameliorate symptoms of a specific pathology. For example, a stimulus inducing an increase in the number of neurons expressing dopamine could treat Parkinson's disease, or one affecting the number expressing serotonin could alleviate depression. This may already be producing successful treatment outcomes without our knowing that transmitter switching is involved, with improvement of motor function through physical activity and cure of seasonal depression with phototherapy. This review presents prospects for future investigation of neurotransmitter switching, considering opportunities and challenges for future research and describing how the investigation of transmitter switching is likely to evolve with new tools, thus reshaping our understanding of both normal brain function and mental illness.
Collapse
|
27
|
Klocke C, Lein PJ. Evidence Implicating Non-Dioxin-Like Congeners as the Key Mediators of Polychlorinated Biphenyl (PCB) Developmental Neurotoxicity. Int J Mol Sci 2020; 21:E1013. [PMID: 32033061 PMCID: PMC7037228 DOI: 10.3390/ijms21031013] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Revised: 01/31/2020] [Accepted: 02/02/2020] [Indexed: 12/15/2022] Open
Abstract
Despite being banned from production for decades, polychlorinated biphenyls (PCBs) continue to pose a significant risk to human health. This is due to not only the continued release of legacy PCBs from PCB-containing equipment and materials manufactured prior to the ban on PCB production, but also the inadvertent production of PCBs as byproducts of contemporary pigment and dye production. Evidence from human and animal studies clearly identifies developmental neurotoxicity as a primary endpoint of concern associated with PCB exposures. However, the relative role(s) of specific PCB congeners in mediating the adverse effects of PCBs on the developing nervous system, and the mechanism(s) by which PCBs disrupt typical neurodevelopment remain outstanding questions. New questions are also emerging regarding the potential developmental neurotoxicity of lower chlorinated PCBs that were not present in the legacy commercial PCB mixtures, but constitute a significant proportion of contemporary human PCB exposures. Here, we review behavioral and mechanistic data obtained from experimental models as well as recent epidemiological studies that suggest the non-dioxin-like (NDL) PCBs are primarily responsible for the developmental neurotoxicity associated with PCBs. We also discuss emerging data demonstrating the potential for non-legacy, lower chlorinated PCBs to cause adverse neurodevelopmental outcomes. Molecular targets, the relevance of PCB interactions with these targets to neurodevelopmental disorders, and critical data gaps are addressed as well.
Collapse
Affiliation(s)
| | - Pamela J. Lein
- Department of Molecular Biosciences, University of California, Davis, School of Veterinary Medicine, Davis, CA 95616, USA;
| |
Collapse
|
28
|
Wirz-Justice A, Benedetti F. Perspectives in affective disorders: Clocks and sleep. Eur J Neurosci 2019; 51:346-365. [PMID: 30702783 DOI: 10.1111/ejn.14362] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2018] [Revised: 12/30/2018] [Accepted: 01/22/2019] [Indexed: 12/17/2022]
Abstract
Mood disorders are often characterised by alterations in circadian rhythms, sleep disturbances and seasonal exacerbation. Conversely, chronobiological treatments utilise zeitgebers for circadian rhythms such as light to improve mood and stabilise sleep, and manipulations of sleep timing and duration as rapid antidepressant modalities. Although sleep deprivation ("wake therapy") can act within hours, and its mood-elevating effects be maintained by regular morning light administration/medication/earlier sleep, it has not entered the regular guidelines for treating affective disorders as a first-line treatment. The hindrances to using chronotherapeutics may lie in their lack of patentability, few sponsors to carry out large multi-centre trials, non-reimbursement by medical insurance and their perceived difficulty or exotic "alternative" nature. Future use can be promoted by new technology (single-sample phase measurements, phone apps, movement and sleep trackers) that provides ambulatory documentation over long periods and feedback to therapist and patient. Light combinations with cognitive behavioural therapy and sleep hygiene practice may speed up and also maintain response. The urgent need for new antidepressants should hopefully lead to reconsideration and implementation of these non-pharmacological methods, as well as further clinical trials. We review the putative neurochemical mechanisms underlying the antidepressant effect of sleep deprivation and light therapy, and current knowledge linking clocks and sleep with affective disorders: neurotransmitter switching, stress and cortico-limbic reactivity, clock genes, cortical neuroplasticity, connectomics and neuroinflammation. Despite the complexity of multi-system mechanisms, more insight will lead to fine tuning and better application of circadian and sleep-related treatments of depression.
Collapse
Affiliation(s)
- Anna Wirz-Justice
- Centre for Chronobiology, Transfaculty Research Platform Molecular and Cognitive Neurosciences, Psychiatric Hospital of the University of Basel, Basel, Switzerland
| | - Francesco Benedetti
- University Vita-Salute San Raffaele, Milano, Italy.,Psychiatry & Clinical Psychobiology, Division of Neuroscience, San Raffaele Scientific Institute, Milano, Italy
| |
Collapse
|
29
|
Chen WL, Ye Q, Zhang SC, Xia Y, Yang X, Yuan TF, Shan CL, Li JA. Aphasia rehabilitation based on mirror neuron theory: a randomized-block-design study of neuropsychology and functional magnetic resonance imaging. Neural Regen Res 2019; 14:1004-1012. [PMID: 30762012 PMCID: PMC6404486 DOI: 10.4103/1673-5374.250580] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
When watching someone performs an action, mirror neurons are activated in a way that is very similar to the activation that occurs when actually performing that action. Previous single-sample case studies indicate that hand-action observation training may lead to activation and remodeling of mirror neuron systems, which include important language centers, and may improve language function in aphasia patients. In this randomized-block-design experiment, we recruited 24 aphasia patients from, Zhongda Hospital, Southeast University, China. The patients were divided into three groups where they underwent hand-action observation and repetition, dynamic-object observation and repetition, or conventional speech therapy. Training took place 5 days per week, 35 minutes per day, for 2 weeks. We assessed language function via picture naming tests for objects and actions and the Western Aphasia Battery. Among the participants, one patient, his wife and four healthy student volunteers underwent functional magnetic resonance imaging to analyze changes in brain activation during hand-action observation and dynamic-object observation. Results demonstrated that, compared with dynamic-object observation, hand-action observation led to greater performance with respect to the aphasia quotient and affiliated naming sub-tests and a greater Western Aphasia Battery test score. The overall effect was similar to that of conventional aphasia training, yet hand-action observation had advantages compared with conventional training in terms of vocabulary extraction and spontaneous speech. Thus, hand-action observation appears to more strongly activate the mirror neuron system compared with dynamic-object observation. The activated areas included Broca’s area, Wernicke’s area, and the supramarginal gyrus. These results suggest that hand-action observation combined with repetition might better improve language function in aphasia patients compared with dynamic-object observation combined with repetition. The therapeutic mechanism of this intervention may be associated with activation of additional mirror neuron systems, and may have implications for the possible repair and remodeling of damaged nerve networks. The study protocol was approved by the Ethical Committee of Nanjing Medical University, China (approval number: 2011-SRFA-086) on March 11, 2011. This trial has been registered in the ISRCTN Registry (ISRCTN84827527).
Collapse
Affiliation(s)
- Wen-Li Chen
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing; Department of Rehabilitation Medicine, Zhangjiagang Hospital Affiliated to Nanjing University of Chinese Medicine, Zhangjiagang, Jiangsu Province, China
| | - Qian Ye
- School of Rehabilitation Sciences, Nanjing Normal University of Special Education, Nanjing, Jiangsu Province, China
| | - Si-Cong Zhang
- Yueyang Hospital of Integrated Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yang Xia
- Department of Rehabilitation Medicine, Zhongda Hospital, Southeast University, Nanjing, Jiangsu Province, China
| | - Xi Yang
- Department of Rehabilitation Medicine, Zhongda Hospital, Southeast University, Nanjing, Jiangsu Province, China
| | - Ti-Fei Yuan
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chun-Lei Shan
- Yueyang Hospital of Integrated Chinese and Western Medicine; School of Rehabilitation Science, Shanghai University of Traditional Chinese Medicine, Shanghai, China; Institute of Rehabilitation Medicine, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China
| | - Jian-An Li
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| |
Collapse
|
30
|
Abstract
Given the failure of psychiatry to develop clinically useful biomarkers for psychiatric disorders, and the concomitant failure to develop significant advances in diagnosis and treatment, the National Institute of Mental Health (NIMH) in 2010 launched the Research Domain Criteria (RDoC), a framework for research based on the assumption that mental disorders are disorders of identifiable brain neural circuits, with neural circuitry at the center of units of analysis ranging from genes, molecules, and cells to behavior, self-reports, and paradigms. These were to be integrated with five validated dimensional psychological constructs such as negative and positive valence systems. Four years later, the NIMH stated that the ultimate goal of RDoC is precision medicine for psychiatry, with the assumption that precision medications will normalize dysfunctional neural circuits. How this could be accomplished is not obvious, given that neural circuits are widely distributed, have unclear boundaries, and exhibit a significant degree of neuroplasticity, with multiple circuits present in any given disorder. Moreover, the early focus on neural circuitry has been criticized for its reductionism and neglect of the more recent RDoC emphasis on the integration and equivalence of biological and psychological phenomena. Yet this seems inconsistent with the priorities of the NIMH director, an advocate of the central role of neural circuitry and projects such as the Brain Initiative and the Human Connectome Project. Will such projects, at a cost of at least $10 billion, lead to precision medications for mental disorders, or further diminish funding for clinical care and research?
Collapse
Affiliation(s)
- Charles E Dean
- Mental Health Service Line,Minneapolis Veteran Administration Medical Center,One Veterans Drive, Minneapolis Minnesota, 55147,USA
| |
Collapse
|
31
|
Adult spinal motoneurons change their neurotransmitter phenotype to control locomotion. Proc Natl Acad Sci U S A 2018; 115:E9926-E9933. [PMID: 30275331 PMCID: PMC6196516 DOI: 10.1073/pnas.1809050115] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
An intriguing feature of the nervous system is its plasticity—the remarkable lifelong capacity to change and adapt in light of intrinsic and extrinsic stimuli. Among the many different adaptive mechanisms that occur within the nervous system, changes in neurotransmission form an important plasticity-bestowing mechanism in the reconfiguration of neuronal circuits. Here, we reveal that physical activity and spinal cord injury can switch the neurotransmitter phenotype of the fast axial motoneurons to coexpress glutamate. Furthermore, our study shows that the adult vertebrate spinal motoneurons corelease glutamate alongside ACh in neuromuscular junctions to regulate motor behaviors. Thus, our findings suggest that fast motoneuron glutamatergic respecification enables a motor function-enhancing mechanism in vertebrates. A particularly essential determinant of a neuron’s functionality is its neurotransmitter phenotype. While the prevailing view is that neurotransmitter phenotypes are fixed and determined early during development, a growing body of evidence suggests that neurons retain the ability to switch between different neurotransmitters. However, such changes are considered unlikely in motoneurons due to their crucial functional role in animals’ behavior. Here we describe the expression and dynamics of glutamatergic neurotransmission in the adult zebrafish spinal motoneuron circuit assembly. We demonstrate that part of the fast motoneurons retain the ability to switch their neurotransmitter phenotype under physiological (exercise/training) and pathophysiological (spinal cord injury) conditions to corelease glutamate in the neuromuscular junctions to enhance animals’ motor output. Our findings suggest that motoneuron neurotransmitter switching is an important plasticity-bestowing mechanism in the reconfiguration of spinal circuits that control movements.
Collapse
|
32
|
Jabeen S, Thirumalai V. The interplay between electrical and chemical synaptogenesis. J Neurophysiol 2018; 120:1914-1922. [PMID: 30067121 PMCID: PMC6230774 DOI: 10.1152/jn.00398.2018] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Neurons communicate with each other via electrical or chemical synaptic connections. The pattern and strength of connections between neurons are critical for generating appropriate output. What mechanisms govern the formation of electrical and/or chemical synapses between two neurons? Recent studies indicate that common molecular players could regulate the formation of both of these classes of synapses. In addition, electrical and chemical synapses can mutually coregulate each other’s formation. Electrical activity, generated spontaneously by the nervous system or initiated from sensory experience, plays an important role in this process, leading to the selection of appropriate connections and the elimination of inappropriate ones. In this review, we discuss recent studies that shed light on the formation and developmental interactions of chemical and electrical synapses.
Collapse
Affiliation(s)
- Shaista Jabeen
- National Centre for Biological Sciences, Tata Institute for Fundamental Research , Bangalore , India.,Manipal Academy of Higher Education, Madhav Nagar, Manipal , India
| | - Vatsala Thirumalai
- National Centre for Biological Sciences, Tata Institute for Fundamental Research , Bangalore , India
| |
Collapse
|
33
|
Vitali I, Fièvre S, Telley L, Oberst P, Bariselli S, Frangeul L, Baumann N, McMahon JJ, Klingler E, Bocchi R, Kiss JZ, Bellone C, Silver DL, Jabaudon D. Progenitor Hyperpolarization Regulates the Sequential Generation of Neuronal Subtypes in the Developing Neocortex. Cell 2018; 174:1264-1276.e15. [PMID: 30057116 DOI: 10.1016/j.cell.2018.06.036] [Citation(s) in RCA: 96] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Revised: 04/20/2018] [Accepted: 06/18/2018] [Indexed: 12/29/2022]
Abstract
During corticogenesis, ventricular zone progenitors sequentially generate distinct subtypes of neurons, accounting for the diversity of neocortical cells and the circuits they form. While activity-dependent processes are critical for the differentiation and circuit assembly of postmitotic neurons, how bioelectrical processes affect nonexcitable cells, such as progenitors, remains largely unknown. Here, we reveal that, in the developing mouse neocortex, ventricular zone progenitors become more hyperpolarized as they generate successive subtypes of neurons. Experimental in vivo hyperpolarization shifted the transcriptional programs and division modes of these progenitors to a later developmental status, with precocious generation of intermediate progenitors and a forward shift in the laminar, molecular, morphological, and circuit features of their neuronal progeny. These effects occurred through inhibition of the Wnt-beta-catenin signaling pathway by hyperpolarization. Thus, during corticogenesis, bioelectric membrane properties are permissive for specific molecular pathways to coordinate the temporal progression of progenitor developmental programs and thus neocortical neuron diversity.
Collapse
Affiliation(s)
- Ilaria Vitali
- Department of Basic Neurosciences, University of Geneva, 1 Rue Michel Servet, 1211 Geneva, Switzerland
| | - Sabine Fièvre
- Department of Basic Neurosciences, University of Geneva, 1 Rue Michel Servet, 1211 Geneva, Switzerland
| | - Ludovic Telley
- Department of Basic Neurosciences, University of Geneva, 1 Rue Michel Servet, 1211 Geneva, Switzerland
| | - Polina Oberst
- Department of Basic Neurosciences, University of Geneva, 1 Rue Michel Servet, 1211 Geneva, Switzerland
| | - Sebastiano Bariselli
- Department of Basic Neurosciences, University of Geneva, 1 Rue Michel Servet, 1211 Geneva, Switzerland
| | - Laura Frangeul
- Department of Basic Neurosciences, University of Geneva, 1 Rue Michel Servet, 1211 Geneva, Switzerland
| | - Natalia Baumann
- Department of Basic Neurosciences, University of Geneva, 1 Rue Michel Servet, 1211 Geneva, Switzerland
| | - John J McMahon
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, 224 Carl Building, Durham, NC 27710, USA
| | - Esther Klingler
- Department of Basic Neurosciences, University of Geneva, 1 Rue Michel Servet, 1211 Geneva, Switzerland
| | - Riccardo Bocchi
- Department of Basic Neurosciences, University of Geneva, 1 Rue Michel Servet, 1211 Geneva, Switzerland
| | - Jozsef Z Kiss
- Department of Basic Neurosciences, University of Geneva, 1 Rue Michel Servet, 1211 Geneva, Switzerland
| | - Camilla Bellone
- Department of Basic Neurosciences, University of Geneva, 1 Rue Michel Servet, 1211 Geneva, Switzerland
| | - Debra L Silver
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, 224 Carl Building, Durham, NC 27710, USA
| | - Denis Jabaudon
- Department of Basic Neurosciences, University of Geneva, 1 Rue Michel Servet, 1211 Geneva, Switzerland; Department of Neurology, Geneva University Hospital, 4 Rue Gabrielle-Perret-Gentil, 1205 Geneva, Switzerland.
| |
Collapse
|
34
|
Morris M, Shaw A, Lambert M, Perry HH, Lowenstein E, Valenzuela D, Velazquez-Ulloa NA. Developmental nicotine exposure affects larval brain size and the adult dopaminergic system of Drosophila melanogaster. BMC DEVELOPMENTAL BIOLOGY 2018; 18:13. [PMID: 29898654 PMCID: PMC6001141 DOI: 10.1186/s12861-018-0172-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Accepted: 05/21/2018] [Indexed: 01/15/2023]
Abstract
BACKGROUND Pregnant women may be exposed to nicotine if they smoke or use tobacco products, nicotine replacement therapy, or via e-cigarettes. Prenatal nicotine exposure has been shown to have deleterious effects on the nervous system in mammals including changes in brain size and in the dopaminergic system. The genetic and molecular mechanisms for these changes are not well understood. A Drosophila melanogaster model for these effects of nicotine exposure could contribute to faster identification of genes and molecular pathways underlying these effects. The purpose of this study was to determine if developmental nicotine exposure affects the nervous system of Drosophila melanogaster, focusing on changes to brain size and the dopaminergic system at two developmental stages. RESULTS We reared flies on control or nicotine food from egg to 3rd instar larvae or from egg to adult and determined effectiveness of the nicotine treatment. We used immunohistochemistry to visualize the whole brain and dopaminergic neurons, using tyrosine hydroxylase as the marker. We measured brain area, tyrosine hydroxylase fluorescence, and counted the number of dopaminergic neurons in brain clusters. We detected an increase in larval brain hemisphere area, a decrease in tyrosine hydroxylase fluorescence in adult central brains, and a decrease in the number of neurons in the PPM3 adult dopaminergic cluster. We tested involvement of Dα7, one of the nicotinic acetylcholine receptor subunits, and found it was involved in eclosion, as previously described, but not involved in brain size. CONCLUSIONS We conclude that developmental nicotine exposure in Drosophila melanogaster affects brain size and the dopaminergic system. Prenatal nicotine exposure in mammals has also been shown to have effects on brain size and in the dopaminergic system. This study further establishes Drosophila melanogaster as model organism to study the effects of developmental nicotine exposure. The genetic and molecular tools available for Drosophila research will allow elucidation of the mechanisms underlying the effects of nicotine exposure during development.
Collapse
Affiliation(s)
- Melanie Morris
- School of Medicine, University of Washington, Seattle, USA
| | - Ariel Shaw
- Biochemistry, Cell and Molecular Biology Program, Lewis & Clark College, Portland, USA
| | | | | | - Eve Lowenstein
- Biology Department, Lewis & Clark College, Portland, USA
| | | | | |
Collapse
|
35
|
Gopurappilly R, Deb BK, Chakraborty P, Hasan G. Stable STIM1 Knockdown in Self-Renewing Human Neural Precursors Promotes Premature Neural Differentiation. Front Mol Neurosci 2018; 11:178. [PMID: 29942250 PMCID: PMC6004407 DOI: 10.3389/fnmol.2018.00178] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2018] [Accepted: 05/09/2018] [Indexed: 12/31/2022] Open
Abstract
Ca2+ signaling plays a significant role in the development of the vertebrate nervous system where it regulates neurite growth as well as synapse and neurotransmitter specification. Elucidating the role of Ca2+ signaling in mammalian neuronal development has been largely restricted to either small animal models or primary cultures. Here we derived human neural precursor cells (NPCs) from human embryonic stem cells to understand the functional significance of a less understood arm of calcium signaling, Store-operated Ca2+ entry or SOCE, in neuronal development. Human NPCs exhibited robust SOCE, which was significantly attenuated by expression of a stable shRNA-miR targeted toward the SOCE molecule, STIM1. Along with the plasma membrane channel Orai, STIM is an essential component of SOCE in many cell types, where it regulates gene expression. Therefore, we measured global gene expression in human NPCs with and without STIM1 knockdown. Interestingly, pathways down-regulated through STIM1 knockdown were related to cell proliferation and DNA replication processes, whereas post-synaptic signaling was identified as an up-regulated process. To understand the functional significance of these gene expression changes we measured the self-renewal capacity of NPCs with STIM1 knockdown. The STIM1 knockdown NPCs demonstrated significantly reduced neurosphere size and number as well as precocious spontaneous differentiation toward the neuronal lineage, as compared to control cells. These findings demonstrate that STIM1 mediated SOCE in human NPCs regulates gene expression changes, that in vivo are likely to physiologically modulate the self-renewal and differentiation of NPCs.
Collapse
Affiliation(s)
- Renjitha Gopurappilly
- National Centre for Biological Sciences, Tata Institute of Fundamental Research, Bengaluru, India
| | - Bipan Kumar Deb
- National Centre for Biological Sciences, Tata Institute of Fundamental Research, Bengaluru, India
| | - Pragnya Chakraborty
- National Centre for Biological Sciences, Tata Institute of Fundamental Research, Bengaluru, India
| | - Gaiti Hasan
- National Centre for Biological Sciences, Tata Institute of Fundamental Research, Bengaluru, India
| |
Collapse
|
36
|
Hessel EVS, Staal YCM, Piersma AH. Design and validation of an ontology-driven animal-free testing strategy for developmental neurotoxicity testing. Toxicol Appl Pharmacol 2018; 354:136-152. [PMID: 29544899 DOI: 10.1016/j.taap.2018.03.013] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Revised: 02/26/2018] [Accepted: 03/11/2018] [Indexed: 12/26/2022]
Abstract
Developmental neurotoxicity entails one of the most complex areas in toxicology. Animal studies provide only limited information as to human relevance. A multitude of alternative models have been developed over the years, providing insights into mechanisms of action. We give an overview of fundamental processes in neural tube formation, brain development and neural specification, aiming at illustrating complexity rather than comprehensiveness. We also give a flavor of the wealth of alternative methods in this area. Given the impressive progress in mechanistic knowledge of human biology and toxicology, the time is right for a conceptual approach for designing testing strategies that cover the integral mechanistic landscape of developmental neurotoxicity. The ontology approach provides a framework for defining this landscape, upon which an integral in silico model for predicting toxicity can be built. It subsequently directs the selection of in vitro assays for rate-limiting events in the biological network, to feed parameter tuning in the model, leading to prediction of the toxicological outcome. Validation of such models requires primary attention to coverage of the biological domain, rather than classical predictive value of individual tests. Proofs of concept for such an approach are already available. The challenge is in mining modern biology, toxicology and chemical information to feed intelligent designs, which will define testing strategies for neurodevelopmental toxicity testing.
Collapse
Affiliation(s)
- Ellen V S Hessel
- Center for Health Protection, National Institute for Public Health and The Environment (RIVM), P.O. Box 1, 3720BA Bilthoven, The Netherlands.
| | - Yvonne C M Staal
- Center for Health Protection, National Institute for Public Health and The Environment (RIVM), P.O. Box 1, 3720BA Bilthoven, The Netherlands
| | - Aldert H Piersma
- Center for Health Protection, National Institute for Public Health and The Environment (RIVM), P.O. Box 1, 3720BA Bilthoven, The Netherlands; Institute for Risk Assessment Sciences (IRAS), Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
37
|
Fabbiani G, Rehermann MI, Aldecosea C, Trujillo-Cenóz O, Russo RE. Emergence of Serotonergic Neurons After Spinal Cord Injury in Turtles. Front Neural Circuits 2018; 12:20. [PMID: 29593503 PMCID: PMC5859367 DOI: 10.3389/fncir.2018.00020] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Accepted: 02/22/2018] [Indexed: 01/04/2023] Open
Abstract
Plasticity of neural circuits takes many forms and plays a fundamental role in regulating behavior to changing demands while maintaining stability. For example, during spinal cord development neurotransmitter identity in neurons is dynamically adjusted in response to changes in the activity of spinal networks. It is reasonable to speculate that this type of plasticity might occur also in mature spinal circuits in response to injury. Because serotonergic signaling has a central role in spinal cord functions, we hypothesized that spinal cord injury (SCI) in the fresh water turtle Trachemys scripta elegans may trigger homeostatic changes in serotonergic innervation. To test this possibility we performed immunohistochemistry for serotonin (5-HT) and key molecules involved in the determination of the serotonergic phenotype before and after SCI. We found that as expected, in the acute phase after injury the dense serotonergic innervation was strongly reduced. However, 30 days after SCI the population of serotonergic cells (5-HT+) increased in segments caudal to the lesion site. These cells expressed the neuronal marker HuC/D and the transcription factor Nkx6.1. The new serotonergic neurons did not incorporate the thymidine analog 5-bromo-2'-deoxyuridine (BrdU) and did not express the proliferating cell nuclear antigen (PCNA) indicating that novel serotonergic neurons were not newborn but post-mitotic cells that have changed their neurochemical identity. Switching towards a serotonergic neurotransmitter phenotype may be a spinal cord homeostatic mechanism to compensate for the loss of descending serotonergic neuromodulation, thereby helping the outstanding functional recovery displayed by turtles. The 5-HT1A receptor agonist (±)-8-Hydroxy-2-dipropylaminotetralin hydrobromide (8-OH-DPAT) blocked the increase in 5-HT+ cells suggesting 5-HT1A receptors may trigger the respecification process.
Collapse
Affiliation(s)
- Gabriela Fabbiani
- Departamento de Neurofisiología Celular y Molecular, Instituto de Investigaciones Biológicas Clemente Estable (IIBCE), Montevideo, Uruguay
| | - María I Rehermann
- Departamento de Neurofisiología Celular y Molecular, Instituto de Investigaciones Biológicas Clemente Estable (IIBCE), Montevideo, Uruguay
| | - Carina Aldecosea
- Departamento de Neurofisiología Celular y Molecular, Instituto de Investigaciones Biológicas Clemente Estable (IIBCE), Montevideo, Uruguay
| | - Omar Trujillo-Cenóz
- Departamento de Neurofisiología Celular y Molecular, Instituto de Investigaciones Biológicas Clemente Estable (IIBCE), Montevideo, Uruguay
| | - Raúl E Russo
- Departamento de Neurofisiología Celular y Molecular, Instituto de Investigaciones Biológicas Clemente Estable (IIBCE), Montevideo, Uruguay
| |
Collapse
|
38
|
Photoperiod-Induced Neuroplasticity in the Circadian System. Neural Plast 2018; 2018:5147585. [PMID: 29681926 PMCID: PMC5851158 DOI: 10.1155/2018/5147585] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Accepted: 01/11/2018] [Indexed: 01/01/2023] Open
Abstract
Seasonal changes in light exposure have profound effects on behavioral and physiological functions in many species, including effects on mood and cognitive function in humans. The mammalian brain's master circadian clock, the suprachiasmatic nucleus (SCN), transmits information about external light conditions to other brain regions, including some implicated in mood and cognition. Although the detailed mechanisms are not yet known, the SCN undergoes highly plastic changes at the cellular and network levels under different light conditions. We therefore propose that the SCN may be an essential mediator of the effects of seasonal changes of day length on mental health. In this review, we explore various forms of neuroplasticity that occur in the SCN and other brain regions to facilitate seasonal adaptation, particularly altered phase distribution of cellular circadian oscillators in the SCN and changes in hypothalamic neurotransmitter expression.
Collapse
|
39
|
Williams PTJA, Jiang YQ, Martin JH. Motor system plasticity after unilateral injury in the developing brain. Dev Med Child Neurol 2017; 59:1224-1229. [PMID: 28972274 PMCID: PMC5773112 DOI: 10.1111/dmcn.13581] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/10/2017] [Indexed: 11/30/2022]
Abstract
UNLABELLED In maturity, motor skills depend on the corticospinal tract (CST) and brainstem pathways that together synapse on interneurons and motoneurons in the spinal cord. Descending signals to spinal neurons that mediate voluntary control can be distinguished from peripheral sensory signals, primarily for feedback control. These motor system circuits depend initially on developmental genetic mechanisms to establish their connections and neural activity- and use-dependent synaptic refinement during the early postnatal period to enable motor skills to develop. In this review we consider four key activity-dependent developmental mechanisms that provide insights into how the motor systems establish the proper connections for skilled movement control and how the same mechanisms also inform the mechanisms of motor impairments and developmental plasticity after corticospinal system injury: (1) synaptic competition between the CSTs from each hemisphere; (2) interactions between the CST and spinal cord neurons; (3) synaptic competition between the CST and proprioceptive sensory fibres; and (4) interactions between the developing corticospinal motor system and the rubrospinal tract. Our findings suggest that the corticospinal motor system effectively 'oversees' development of its subcortical targets through synaptic competition and trophic-like interactions and this has important implications for motor impairments after perinatal cortical stroke. WHAT THIS PAPER ADDS Neural activity-dependent processes inform the brain and spinal cord response to injury. The corticospinal motor system may 'oversee' development of its downstream subcortical targets through activity, trophic-like interactions, and synaptic competition.
Collapse
Affiliation(s)
- Preston TJA Williams
- Department of Physiology, Pharmacology and Neuroscience, City University of New York School of Medicine at City College, New York, NY, USA
| | - Yu-Qiu Jiang
- Department of Physiology, Pharmacology and Neuroscience, City University of New York School of Medicine at City College, New York, NY, USA
| | - John H Martin
- Department of Physiology, Pharmacology and Neuroscience, City University of New York School of Medicine at City College, New York, NY, USA,City University of New York Graduate Center, New York, NY, USA
| |
Collapse
|
40
|
Lai HC, Seal RP, Johnson JE. Making sense out of spinal cord somatosensory development. Development 2017; 143:3434-3448. [PMID: 27702783 DOI: 10.1242/dev.139592] [Citation(s) in RCA: 126] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The spinal cord integrates and relays somatosensory input, leading to complex motor responses. Research over the past couple of decades has identified transcription factor networks that function during development to define and instruct the generation of diverse neuronal populations within the spinal cord. A number of studies have now started to connect these developmentally defined populations with their roles in somatosensory circuits. Here, we review our current understanding of how neuronal diversity in the dorsal spinal cord is generated and we discuss the logic underlying how these neurons form the basis of somatosensory circuits.
Collapse
Affiliation(s)
- Helen C Lai
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Rebecca P Seal
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15260, USA
| | - Jane E Johnson
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| |
Collapse
|
41
|
Blanquie O, Yang JW, Kilb W, Sharopov S, Sinning A, Luhmann HJ. Electrical activity controls area-specific expression of neuronal apoptosis in the mouse developing cerebral cortex. eLife 2017; 6:27696. [PMID: 28826501 PMCID: PMC5582867 DOI: 10.7554/elife.27696] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Accepted: 08/18/2017] [Indexed: 12/22/2022] Open
Abstract
Programmed cell death widely but heterogeneously affects the developing brain, causing the loss of up to 50% of neurons in rodents. However, whether this heterogeneity originates from neuronal identity and/or network-dependent processes is unknown. Here, we report that the primary motor cortex (M1) and primary somatosensory cortex (S1), two adjacent but functionally distinct areas, display striking differences in density of apoptotic neurons during the early postnatal period. These differences in rate of apoptosis negatively correlate with region-dependent levels of activity. Disrupting this activity either pharmacologically or by electrical stimulation alters the spatial pattern of apoptosis and sensory deprivation leads to exacerbated amounts of apoptotic neurons in the corresponding functional area of the neocortex. Thus, our data demonstrate that spontaneous and periphery-driven activity patterns are important for the structural and functional maturation of the neocortex by refining the final number of cortical neurons in a region-dependent manner.
Collapse
Affiliation(s)
- Oriane Blanquie
- Institute of Physiology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Jenq-Wei Yang
- Institute of Physiology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Werner Kilb
- Institute of Physiology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Salim Sharopov
- Institute of Physiology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Anne Sinning
- Institute of Physiology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Heiko J Luhmann
- Institute of Physiology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| |
Collapse
|
42
|
Richhariya S, Jayakumar S, Abruzzi K, Rosbash M, Hasan G. A pupal transcriptomic screen identifies Ral as a target of store-operated calcium entry in Drosophila neurons. Sci Rep 2017; 7:42586. [PMID: 28195208 PMCID: PMC5307359 DOI: 10.1038/srep42586] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Accepted: 01/12/2017] [Indexed: 12/20/2022] Open
Abstract
Transcriptional regulation by Store-operated Calcium Entry (SOCE) is well studied in non-excitable cells. However, the role of SOCE has been poorly documented in neuronal cells with more complicated calcium dynamics. Previous reports demonstrated a requirement for SOCE in neurons that regulate Drosophila flight bouts. We refine this requirement temporally to the early pupal stage and use RNA-sequencing to identify SOCE mediated gene expression changes in the developing Drosophila pupal nervous system. Down regulation of dStim, the endoplasmic reticular calcium sensor and a principal component of SOCE in the nervous system, altered the expression of 131 genes including Ral, a small GTPase. Disruption of Ral function in neurons impaired flight, whereas ectopic expression of Ral in SOCE-compromised neurons restored flight. Through live imaging of calcium transients from cultured pupal neurons, we confirmed that Ral does not participate in SOCE, but acts downstream of it. These results identify neuronal SOCE as a mechanism that regulates expression of specific genes during development of the pupal nervous system and emphasizes the relevance of SOCE-regulated gene expression to flight circuit maturation.
Collapse
Affiliation(s)
- Shlesha Richhariya
- National Centre for Biological Sciences, Tata Institute of Fundamental Research, Bangalore 560065, India
| | - Siddharth Jayakumar
- National Centre for Biological Sciences, Tata Institute of Fundamental Research, Bangalore 560065, India
- Manipal University, Manipal 576104, India
| | - Katharine Abruzzi
- Howard Hughes Medical Institute, National Center for Behavioral Genomics, Department of Biology, Brandeis University, Waltham, MA 02454, USA
| | - Michael Rosbash
- Howard Hughes Medical Institute, National Center for Behavioral Genomics, Department of Biology, Brandeis University, Waltham, MA 02454, USA
| | - Gaiti Hasan
- National Centre for Biological Sciences, Tata Institute of Fundamental Research, Bangalore 560065, India
| |
Collapse
|
43
|
Defteralı Ç, Verdejo R, Majeed S, Boschetti-de-Fierro A, Méndez-Gómez HR, Díaz-Guerra E, Fierro D, Buhr K, Abetz C, Martínez-Murillo R, Vuluga D, Alexandre M, Thomassin JM, Detrembleur C, Jérôme C, Abetz V, López-Manchado MÁ, Vicario-Abejón C. In Vitro Evaluation of Biocompatibility of Uncoated Thermally Reduced Graphene and Carbon Nanotube-Loaded PVDF Membranes with Adult Neural Stem Cell-Derived Neurons and Glia. Front Bioeng Biotechnol 2016; 4:94. [PMID: 27999773 PMCID: PMC5138223 DOI: 10.3389/fbioe.2016.00094] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Accepted: 11/18/2016] [Indexed: 01/03/2023] Open
Abstract
Graphene, graphene-based nanomaterials (GBNs), and carbon nanotubes (CNTs) are being investigated as potential substrates for the growth of neural cells. However, in most in vitro studies, the cells were seeded on these materials coated with various proteins implying that the observed effects on the cells could not solely be attributed to the GBN and CNT properties. Here, we studied the biocompatibility of uncoated thermally reduced graphene (TRG) and poly(vinylidene fluoride) (PVDF) membranes loaded with multi-walled CNTs (MWCNTs) using neural stem cells isolated from the adult mouse olfactory bulb (termed aOBSCs). When aOBSCs were induced to differentiate on coverslips treated with TRG or control materials (polyethyleneimine-PEI and polyornithine plus fibronectin-PLO/F) in a serum-free medium, neurons, astrocytes, and oligodendrocytes were generated in all conditions, indicating that TRG permits the multi-lineage differentiation of aOBSCs. However, the total number of cells was reduced on both PEI and TRG. In a serum-containing medium, aOBSC-derived neurons and oligodendrocytes grown on TRG were more numerous than in controls; the neurons developed synaptic boutons and oligodendrocytes were more branched. In contrast, neurons growing on PVDF membranes had reduced neurite branching, and on MWCNTs-loaded membranes oligodendrocytes were lower in numbers than in controls. Overall, these findings indicate that uncoated TRG may be biocompatible with the generation, differentiation, and maturation of aOBSC-derived neurons and glial cells, implying a potential use for TRG to study functional neuronal networks.
Collapse
Affiliation(s)
- Çağla Defteralı
- Instituto Cajal, Consejo Superior de Investigaciones Científicas (IC-CSIC), Madrid, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED-ISCIII), Madrid, Spain
| | - Raquel Verdejo
- Instituto de Ciencia y Tecnología de Polímeros (ICTP-CSIC), Madrid, Spain
| | - Shahid Majeed
- Helmholtz-Zentrum Geesthacht (HZG), Zentrum für Material- und Küstenforschung GmbH, Institut für Polymerforschung, Geesthacht, Germany
| | - Adriana Boschetti-de-Fierro
- Helmholtz-Zentrum Geesthacht (HZG), Zentrum für Material- und Küstenforschung GmbH, Institut für Polymerforschung, Geesthacht, Germany
| | - Héctor R. Méndez-Gómez
- Instituto Cajal, Consejo Superior de Investigaciones Científicas (IC-CSIC), Madrid, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED-ISCIII), Madrid, Spain
| | - Eva Díaz-Guerra
- Instituto Cajal, Consejo Superior de Investigaciones Científicas (IC-CSIC), Madrid, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED-ISCIII), Madrid, Spain
| | - Daniel Fierro
- Helmholtz-Zentrum Geesthacht (HZG), Zentrum für Material- und Küstenforschung GmbH, Institut für Polymerforschung, Geesthacht, Germany
| | - Kristian Buhr
- Helmholtz-Zentrum Geesthacht (HZG), Zentrum für Material- und Küstenforschung GmbH, Institut für Polymerforschung, Geesthacht, Germany
| | - Clarissa Abetz
- Helmholtz-Zentrum Geesthacht (HZG), Zentrum für Material- und Küstenforschung GmbH, Institut für Polymerforschung, Geesthacht, Germany
| | | | - Daniela Vuluga
- Department of Chemistry, Center for Education and Research on Macromolecules (CERM), University of Liège, Liège, Belgium
| | - Michaël Alexandre
- Department of Chemistry, Center for Education and Research on Macromolecules (CERM), University of Liège, Liège, Belgium
| | - Jean-Michel Thomassin
- Department of Chemistry, Center for Education and Research on Macromolecules (CERM), University of Liège, Liège, Belgium
| | - Christophe Detrembleur
- Department of Chemistry, Center for Education and Research on Macromolecules (CERM), University of Liège, Liège, Belgium
| | - Christine Jérôme
- Department of Chemistry, Center for Education and Research on Macromolecules (CERM), University of Liège, Liège, Belgium
| | - Volker Abetz
- Helmholtz-Zentrum Geesthacht (HZG), Zentrum für Material- und Küstenforschung GmbH, Institut für Polymerforschung, Geesthacht, Germany
| | | | - Carlos Vicario-Abejón
- Instituto Cajal, Consejo Superior de Investigaciones Científicas (IC-CSIC), Madrid, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED-ISCIII), Madrid, Spain
| |
Collapse
|
44
|
Regulation of motoneuron excitability and the setting of homeostatic limits. Curr Opin Neurobiol 2016; 43:1-6. [PMID: 27721083 DOI: 10.1016/j.conb.2016.09.014] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Revised: 08/25/2016] [Accepted: 09/24/2016] [Indexed: 01/29/2023]
Abstract
Stability of neural circuits is reliant on homeostatic mechanisms that return neuron activity towards pre-determined and physiologically appropriate levels. Without these mechanisms, changes due to synaptic plasticity, ageing and disease may push neural circuits towards instability. Whilst widely documented, understanding of how and when neurons determine an appropriate activity level, the so-called set-point, remains unknown. Genetically tractable model systems have greatly contributed to our understanding of neuronal homeostasis and continue to offer attractive models to explore these additional questions. This review focuses on the development of Drosophila motoneurons including defining an embryonic critical period during which these neurons encode their set-points to enable homeostatic regulation of activity.
Collapse
|
45
|
Larsen ZH, Chander P, Joyner JA, Floruta CM, Demeter TL, Weick JP. Effects of Ethanol on Cellular Composition and Network Excitability of Human Pluripotent Stem Cell-Derived Neurons. Alcohol Clin Exp Res 2016; 40:2339-2350. [PMID: 27717039 DOI: 10.1111/acer.13218] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Accepted: 08/12/2016] [Indexed: 11/30/2022]
Abstract
BACKGROUND Prenatal alcohol exposure (PAE) in animal models results in excitatory-inhibitory (E/I) imbalance in neocortex due to alterations in the GABAergic interneuron (IN) differentiation and migration. Thus, E/I imbalance is a potential cause for intellectual disability in individuals with fetal alcohol spectrum disorder (FASD), but whether ethanol (EtOH) changes glutamatergic and GABAergic IN specification during human development remains unknown. Here, we created a human cellular model of PAE/FASD and tested the hypothesis that EtOH exposure during differentiation of human pluripotent stem cell-derived neurons (hPSNs) would cause the aberrant production of glutamatergic and GABAergic neurons, resulting in E/I imbalance. METHODS We applied 50 mM EtOH daily to differentiating hPSNs for 50 days to model chronic first-trimester exposure. We used quantitative polymerase chain reaction, immunocytochemical, and electrophysiological analysis to examine the effects of EtOH on hPSN specification and functional E/I balance. RESULTS We found that EtOH did not alter neural induction nor general forebrain patterning and had no effect on the expression of markers of excitatory cortical pyramidal neurons. In contrast, our data revealed highly significant changes to levels of transcripts involved with IN precursor development (e.g., GSX2, DLX1/2/5/6, NR2F2) as well as mature IN specification (e.g., SST, NPY). Interestingly, EtOH did not affect the number of GABAergic neurons generated nor the frequency or amplitude of miniature excitatory and inhibitory postsynaptic currents. CONCLUSIONS Similar to in vivo rodent studies, EtOH significantly and specifically altered the expression of genes involved with IN specification from hPSNs, but did not cause imbalances of synaptic excitation-inhibition. Thus, our findings corroborate previous studies pointing to aberrant neuronal differentiation as an underlying mechanism of intellectual disability in FASD. However, in contrast to rodent binge models, our chronic exposure model suggests possible compensatory mechanisms that may cause more subtle defects of network processing rather than gross alterations in total E/I balance.
Collapse
Affiliation(s)
- Zoe H Larsen
- Department of Neurosciences, University of New Mexico-Health Science Center, Albuquerque, New Mexico
| | - Praveen Chander
- Department of Neurosciences, University of New Mexico-Health Science Center, Albuquerque, New Mexico
| | - Jason A Joyner
- Department of Neurosciences, University of New Mexico-Health Science Center, Albuquerque, New Mexico
| | - Crina M Floruta
- Department of Neurosciences, University of New Mexico-Health Science Center, Albuquerque, New Mexico
| | - Tess L Demeter
- Department of Neurosciences, University of New Mexico-Health Science Center, Albuquerque, New Mexico
| | - Jason P Weick
- Department of Neurosciences, University of New Mexico-Health Science Center, Albuquerque, New Mexico.
| |
Collapse
|
46
|
Hilinski WC, Bostrom JR, England SJ, Juárez-Morales JL, de Jager S, Armant O, Legradi J, Strähle U, Link BA, Lewis KE. Lmx1b is required for the glutamatergic fates of a subset of spinal cord neurons. Neural Dev 2016; 11:16. [PMID: 27553035 PMCID: PMC4995821 DOI: 10.1186/s13064-016-0070-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Accepted: 08/08/2016] [Indexed: 01/27/2023] Open
Abstract
Background Alterations in neurotransmitter phenotypes of specific neurons can cause imbalances in excitation and inhibition in the central nervous system (CNS), leading to diseases. Therefore, the correct specification and maintenance of neurotransmitter phenotypes is vital. As with other neuronal properties, neurotransmitter phenotypes are often specified and maintained by particular transcription factors. However, the specific molecular mechanisms and transcription factors that regulate neurotransmitter phenotypes remain largely unknown. Methods In this paper we use single mutant, double mutant and transgenic zebrafish embryos to elucidate the functions of Lmx1ba and Lmx1bb in the regulation of spinal cord interneuron neurotransmitter phenotypes. Results We demonstrate that lmx1ba and lmx1bb are both expressed in zebrafish spinal cord and that lmx1bb is expressed by both V0v cells and dI5 cells. Our functional analyses demonstrate that these transcription factors are not required for neurotransmitter fate specification at early stages of development, but that in embryos with at least two lmx1ba and/or lmx1bb mutant alleles there is a reduced number of excitatory (glutamatergic) spinal interneurons at later stages of development. In contrast, there is no change in the numbers of V0v or dI5 cells. These data suggest that lmx1b-expressing spinal neurons still form normally, but at least a subset of them lose, or do not form, their normal excitatory fates. As the reduction in glutamatergic cells is only seen at later stages of development, Lmx1b is probably required either for the maintenance of glutamatergic fates or to specify glutamatergic phenotypes of a subset of later forming neurons. Using double labeling experiments, we also show that at least some of the cells that lose their normal glutamatergic phenotype are V0v cells. Finally, we also establish that Evx1 and Evx2, two transcription factors that are required for V0v cells to acquire their excitatory neurotransmitter phenotype, are also required for lmx1ba and lmx1bb expression in these cells, suggesting that Lmx1ba and Lmx1bb act downstream of Evx1 and Evx2 in V0v cells. Conclusions Lmx1ba and Lmx1bb function at least partially redundantly in the spinal cord and three functional lmx1b alleles are required in zebrafish for correct numbers of excitatory spinal interneurons at later developmental stages. Taken together, our data significantly enhance our understanding of how spinal cord neurotransmitter fates are regulated.
Collapse
Affiliation(s)
- William C Hilinski
- Department of Biology, Syracuse University, 107 College Place, Syracuse, NY, 13244, USA.,Department of Neuroscience and Physiology, SUNY Upstate Medical University, 505 Irving Avenue, Syracuse, NY, 13210, USA
| | - Jonathan R Bostrom
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, 8701 Watertown Plank Rd., Milwaukee, WI, 53226, USA
| | - Samantha J England
- Department of Biology, Syracuse University, 107 College Place, Syracuse, NY, 13244, USA
| | - José L Juárez-Morales
- Department of Biology, Syracuse University, 107 College Place, Syracuse, NY, 13244, USA
| | - Sarah de Jager
- Department of Physiology, Development and Neuroscience, University of Cambridge, Downing Street, Cambridge, CB2 3DY, UK
| | - Olivier Armant
- Institute of Toxicology and Genetics, Karlsruhe Institute of Technology (KIT), Postfach 3640, 76021, Karlsruhe, Germany
| | - Jessica Legradi
- Institute of Toxicology and Genetics, Karlsruhe Institute of Technology (KIT), Postfach 3640, 76021, Karlsruhe, Germany
| | - Uwe Strähle
- Institute of Toxicology and Genetics, Karlsruhe Institute of Technology (KIT), Postfach 3640, 76021, Karlsruhe, Germany
| | - Brian A Link
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, 8701 Watertown Plank Rd., Milwaukee, WI, 53226, USA
| | - Katharine E Lewis
- Department of Biology, Syracuse University, 107 College Place, Syracuse, NY, 13244, USA.
| |
Collapse
|
47
|
Abstract
AbstractMore than 35 years ago, Meltzoff and Moore (1977) published their famous article, “Imitation of facial and manual gestures by human neonates.” Their central conclusion, that neonates can imitate, was and continues to be controversial. Here, we focus on an often-neglected aspect of this debate, namely, neonatal spontaneous behaviors themselves. We present a case study of a paradigmatic orofacial “gesture,” namely tongue protrusion and retraction (TP/R). Against the background of new research on mammalian aerodigestive development, we ask: How does the human aerodigestive system develop, and what role does TP/R play in the neonate's emerging system of aerodigestion? We show that mammalian aerodigestion develops in two phases: (1) from the onset of isolated orofacial movementsin uteroto the postnatal mastery of suckling at 4 months after birth; and (2) thereafter, from preparation to the mastery of mastication and deglutition of solid foods. Like other orofacial stereotypies, TP/R emerges in the first phase and vanishes prior to the second. Based upon recent advances in activity-driven early neural development, we suggest a sequence of three developmental events in which TP/R might participate: the acquisition of tongue control, the integration of the central pattern generator (CPG) for TP/R with other aerodigestive CPGs, and the formation of connections within the cortical maps of S1 and M1. If correct, orofacial stereotypies are crucial to the maturation of aerodigestion in the neonatal period but also unlikely to co-occur with imitative behavior.
Collapse
|
48
|
Vincen-Brown MA, Revill AL, Pilarski JQ. Activity-dependent plasticity in the isolated embryonic avian brainstem following manipulations of rhythmic spontaneous neural activity. Respir Physiol Neurobiol 2016; 229:24-33. [PMID: 27025229 DOI: 10.1016/j.resp.2016.03.013] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2015] [Revised: 02/20/2016] [Accepted: 03/22/2016] [Indexed: 02/06/2023]
Abstract
When rhythmic spontaneous neural activity (rSNA) first appears in the embryonic chick brainstem and cranial nerve motor axons it is principally driven by nicotinic neurotransmission (NT). At this early age, the nicotinic acetylcholine receptor (nAChR) agonist nicotine is known to critically disrupt rSNA at low concentrations (0.1-0.5μM), which are levels that mimic the blood plasma levels of a fetus following maternal cigarette smoking. Thus, we quantified the effect of persistent exposure to exogenous nicotine on rSNA using an in vitro developmental model. We found that rSNA was eliminated by continuous bath application of exogenous nicotine, but rSNA recovered activity within 6-12h despite the persistent activation and desensitization of nAChRs. During the recovery period rSNA was critically driven by chloride-mediated membrane depolarization instead of nicotinic NT. To test whether this observed compensation was unique to the antagonism of nicotinic NT or whether the loss of spiking behavior also played a role, we eliminated rSNA by lowering overall excitatory drive with a low [K(+)]o superfusate. In this context, rSNA again recovered, although the recovery time was much quicker, and exhibited a lower frequency, higher duration, and an increase in the number of bursts per episode when compared to control embryos. Importantly, we show that the main compensatory response to lower overall excitatory drive, similar to nicotinergic block, is a result of potentiated chloride mediated membrane depolarization. These results support increasing evidence that early neural circuits sense spiking behavior to maintain primordial bioelectric rhythms. Understanding the nature of developmental plasticity in the nervous system, especially versions that preserve rhythmic behaviors following clinically meaningful environmental stimuli, both normal and pathological, will require similar studies to determine the consequences of feedback compensation at more mature chronological ages.
Collapse
Affiliation(s)
| | - Ann L Revill
- Department of Physiology, University of Alberta, Edmonton, AB, Canada
| | - Jason Q Pilarski
- Department of Biological Sciences, Idaho State University, Pocatello, ID, United states; Department of Dental Sciences, Idaho State University, Pocatello, ID, United states.
| |
Collapse
|
49
|
Borodinsky LN, Belgacem YH. Crosstalk among electrical activity, trophic factors and morphogenetic proteins in the regulation of neurotransmitter phenotype specification. J Chem Neuroanat 2015; 73:3-8. [PMID: 26686293 DOI: 10.1016/j.jchemneu.2015.12.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2015] [Revised: 11/29/2015] [Accepted: 12/02/2015] [Indexed: 01/11/2023]
Abstract
Morphogenetic proteins are responsible for patterning the embryonic nervous system by enabling cell proliferation that will populate all the neural structures and by specifying neural progenitors that imprint different identities in differentiating neurons. The adoption of specific neurotransmitter phenotypes is crucial for the progression of neuronal differentiation, enabling neurons to connect with each other and with target tissues. Preliminary neurotransmitter specification originates from morphogen-driven neural progenitor specification through the combinatorial expression of transcription factors according to morphogen concentration gradients, which progressively restrict the identity that born neurons adopt. However, neurotransmitter phenotype is not immutable, instead trophic factors released from target tissues and environmental stimuli change expression of neurotransmitter-synthesizing enzymes and specific vesicular transporters modifying neuronal neurotransmitter identity. Here we review studies identifying the mechanisms of catecholaminergic, GABAergic, glutamatergic, cholinergic and serotonergic early specification and of the plasticity of these neurotransmitter phenotypes during development and in the adult nervous system. The emergence of spontaneous electrical activity in developing neurons recruits morphogenetic proteins in the process of neurotransmitter phenotype plasticity, which ultimately equips the nervous system and the whole organism with adaptability for optimal performance in a changing environment.
Collapse
Affiliation(s)
- Laura N Borodinsky
- Department of Physiology & Membrane Biology and Institute for Pediatric Regenerative Medicine, Shriners Hospital for Children, University of California Davis School of Medicine, 2425 Stockton Blvd, Sacramento, CA 95817, United States.
| | - Yesser H Belgacem
- Department of Physiology & Membrane Biology and Institute for Pediatric Regenerative Medicine, Shriners Hospital for Children, University of California Davis School of Medicine, 2425 Stockton Blvd, Sacramento, CA 95817, United States
| |
Collapse
|
50
|
Neurotrophin-dependent plasticity of neurotransmitter segregation in the rat superior cervical ganglionin vivo. Dev Neurobiol 2015; 76:832-46. [DOI: 10.1002/dneu.22362] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2015] [Revised: 10/16/2015] [Accepted: 11/06/2015] [Indexed: 01/26/2023]
|