1
|
Francis TC, Klawonn AM. Editorial: Mechanisms of cholinergic transmission in motivation and cognition. Front Mol Neurosci 2025; 18:1586940. [PMID: 40206189 PMCID: PMC11979246 DOI: 10.3389/fnmol.2025.1586940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2025] [Accepted: 03/12/2025] [Indexed: 04/11/2025] Open
Affiliation(s)
- T. Chase Francis
- Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, Columbia, SC, United States
| | - Anna M. Klawonn
- Danish Institute of Translational Neuroscience (DANDRITE), Nordic EMBL Partnership for Molecular Medicine, Aarhus University, Aarhus, Denmark
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| |
Collapse
|
2
|
Li M, Ma C, Li Y, Wang H, Xiu X, Zhao X, Liu P, Yang H, Cheng M. Design, synthesis and biological evaluation of galantamine analogues for cognitive improvement in Alzheimer's disease. Eur J Med Chem 2025; 284:117198. [PMID: 39733484 DOI: 10.1016/j.ejmech.2024.117198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 12/18/2024] [Accepted: 12/19/2024] [Indexed: 12/31/2024]
Abstract
Galantamine plays a crucial role in the management of brain disorders. In this study, a series of galantamine analogues were designed, synthesized and evaluated as potential therapeutic agents for Alzheimer's disease (AD). Compound C2, a dual inhibitor of cholinesterase, was obtained by introducing a benzylpyridine ring to the hydroxyl group of galantamine. Compared to galantamine (hAChE, IC50 = 1529 ± 6 nM), C2 exhibited excellent inhibitory activities against hAChE (IC50 = 513.90 ± 9.60 nM) and hBuChE (IC50 = 357.77 ± 10.24 nM). Further studies revealed that C2 possessed significant abilities to protect PC12 cells from H2O2-induced apoptosis and reactive oxygen species (ROS) production. The acute toxicity test in vivo indicated that C2 exhibited a remarkable safety profile. Whether in the acute memory impairment induced by the Aβ1-42 model or in the amnesia induced by the scopolamine model, oral administration of C2 demonstrated superior improvement on cognition and spatial memory. In summary, both in vitro and in vivo results suggest that C2 deserves to be further explored as an anti-AD agent.
Collapse
Affiliation(s)
- Mengzhen Li
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Liaoning, Shenyang, 110016, China
| | - Chao Ma
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Liaoning, Shenyang, 110016, China
| | - Yao Li
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Liaoning, Shenyang, 110016, China
| | - Hanxun Wang
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Liaoning, Shenyang, 110016, China
| | - Xiaomeng Xiu
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Liaoning, Shenyang, 110016, China
| | - Xueqi Zhao
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Liaoning, Shenyang, 110016, China
| | - Peng Liu
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, Liaoning, 110016, China.
| | - Huali Yang
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Liaoning, Shenyang, 110016, China.
| | - Maosheng Cheng
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Liaoning, Shenyang, 110016, China.
| |
Collapse
|
3
|
Ratna DD, Francis TC. Extrinsic and intrinsic control of striatal cholinergic interneuron activity. Front Mol Neurosci 2025; 18:1528419. [PMID: 40018010 PMCID: PMC11865219 DOI: 10.3389/fnmol.2025.1528419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Accepted: 01/31/2025] [Indexed: 03/01/2025] Open
Abstract
The striatum is an integrated component of the basal ganglia responsible for associative learning and response. Besides the presence of the most abundant γ-aminobutyric acid (GABA-ergic) medium spiny neurons (MSNs), the striatum also contains distributed populations of cholinergic interneurons (ChIs), which bidirectionally communicate with many of these neuronal subtypes. Despite their sparse distribution, ChIs provide the largest source of acetylcholine (ACh) to striatal cells, have a prominent level of arborization and activity, and are potent modulators of striatal output and play prominent roles in plasticity underlying associative learning and reinforcement. Deviations from this tonic activity, including phasic bursts or pauses caused by region-selective excitatory input, neuromodulator, or neuropeptide release can exert strong influences on intrinsic activity and synaptic plasticity via diverse receptor signaling. Recent studies and new tools have allowed improved identification of factors driving or suppressing cholinergic activity, including peptides. This review aims to outline our current understanding of factors that control tonic and phasic ChI activity, specifically focusing on how neuromodulators and neuropeptides interact to facilitate or suppress phasic ChI responses underlying learning and plasticity.
Collapse
Affiliation(s)
| | - Tanner Chase Francis
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, Columbia, SC, United States
| |
Collapse
|
4
|
Yin D, Zhao L, Deng S, Xie Y, Ro KS, Yang Z, Du L, Xie J, Wei D. Lactiplantibacillus plantarum X7022 Plays Roles on Aging Mice with Memory Impairment Induced by D-Galactose Through Restoring Neuronal Damage, Relieving Inflammation and Oxidative Stress. Probiotics Antimicrob Proteins 2025; 17:1-14. [PMID: 38183568 DOI: 10.1007/s12602-023-10208-w] [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] [Accepted: 12/16/2023] [Indexed: 01/08/2024]
Abstract
In this study, Lactiplantibacillus plantarum X7022 was applied to ameliorate memory impairment of aging mice induced by D-galactose. The strain showed specific choloylglycine hydrolysis ability based on in vitro investigation. Morris water maze test showed L. plantarum X7022 administration improved learning ability and spatial memory of aging mice. The gavage of L. plantarum X7022 displayed a promising ability of relieving cerebral oxidative stress and hippocampal inflammatory condition according to the increased GSH level and SOD activity and decreased MDA level, as well as decreased TNF-α, IL-1β, and IL-6 levels. The intervention with the strain could protect neuron by regulating cell apoptosis and AChE overexpression and inhibiting amyloid-β deposition, as well as affect neuron functions by regulating CREB-BDNF signaling pathways and iNOS expression. Besides, the strain could improve fecal SCFA contents and increase the abundance of anti-inflammatory and antioxidant-related genera such as Lactobacillus, Akkermansia, and Adlercreutzia. These results suggest that L. plantarum X7022 could be a prospective therapeutic alternative for the improvement of memory impairment among the elderly.
Collapse
Affiliation(s)
- Deyi Yin
- State Key Laboratory of Bioreactor Engineering, Department of Food Science and Technology, School of Biotechnology, East China University of Science and Technology, 130 # Meilong Rd, Shanghai, 200237, People's Republic of China
| | - Li Zhao
- State Key Laboratory of Bioreactor Engineering, Department of Food Science and Technology, School of Biotechnology, East China University of Science and Technology, 130 # Meilong Rd, Shanghai, 200237, People's Republic of China
| | - Sijing Deng
- State Key Laboratory of Bioreactor Engineering, Department of Food Science and Technology, School of Biotechnology, East China University of Science and Technology, 130 # Meilong Rd, Shanghai, 200237, People's Republic of China
| | - Yaqi Xie
- State Key Laboratory of Bioreactor Engineering, Department of Food Science and Technology, School of Biotechnology, East China University of Science and Technology, 130 # Meilong Rd, Shanghai, 200237, People's Republic of China
| | - Kum-Song Ro
- State Key Laboratory of Bioreactor Engineering, Department of Food Science and Technology, School of Biotechnology, East China University of Science and Technology, 130 # Meilong Rd, Shanghai, 200237, People's Republic of China
- Department of Biotechnology, Faculty of Life Science, Kim Hyong Jik University of Education, Pyongyang, 999093, Democratic People's Republic of Korea
| | - Zeyong Yang
- Department of Anesthesiology, International Peace Maternity and Child Health Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Embryo Original Disease, Shanghai Municipal Key Clinical Specialty, Shanghai, 200030, People's Republic of China
| | - Lei Du
- State Key Laboratory of Bioreactor Engineering, Department of Food Science and Technology, School of Biotechnology, East China University of Science and Technology, 130 # Meilong Rd, Shanghai, 200237, People's Republic of China.
| | - Jingli Xie
- State Key Laboratory of Bioreactor Engineering, Department of Food Science and Technology, School of Biotechnology, East China University of Science and Technology, 130 # Meilong Rd, Shanghai, 200237, People's Republic of China.
- Shanghai Collaborative Innovation Center for Biomanufacturing (SCICB), Shanghai, 200237, People's Republic of China.
| | - Dongzhi Wei
- State Key Laboratory of Bioreactor Engineering, Department of Food Science and Technology, School of Biotechnology, East China University of Science and Technology, 130 # Meilong Rd, Shanghai, 200237, People's Republic of China
- Shanghai Collaborative Innovation Center for Biomanufacturing (SCICB), Shanghai, 200237, People's Republic of China
| |
Collapse
|
5
|
Dittmann NL, Chen L, Voronova A. Regulation of neural stem cells by innervating neurons. J Neurochem 2025; 169:e16287. [PMID: 39775528 PMCID: PMC11707326 DOI: 10.1111/jnc.16287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 10/25/2024] [Accepted: 10/28/2024] [Indexed: 01/11/2025]
Abstract
The adult central nervous system (CNS) hosts several niches, in which the neural stem and precursor cells (NPCs) reside. The subventricular zone (SVZ) lines the lateral brain ventricles and the subgranular zone (SGZ) is located in the dentate gyrus of the hippocampus. SVZ and SGZ NPCs replace neurons and glia in the homeostatic as well as diseased or injured states. Recently, NPCs have been found to express neurotransmitter receptors, respond to electrical stimulation and interact with neurons, suggesting that neuron-NPC communication is an emerging critical regulator of NPC biology. In this review, we discuss reports that demonstrate neuronal innervation and control of the neurogenic niches. We discuss the role of innervating neurons in regulating NPC fates, such as activation, proliferation, and differentiation. Our review focuses primarily on the innervation of the SVZ niche by the following neuronal types: glutamatergic, GABAergic projection and interneurons, cholinergic, dopaminergic, serotonergic, neuropeptidergic, nitrergic, and noradrenergic. We also discuss the origins of SVZ niche innervating neurons, such as striatum, cortex, basal ganglia, raphe nuclei, substantia nigra and ventral tegmental area, hypothalamus, and locus coeruleus. Our review highlights the various roles of innervating neurons in SVZ NPC fates in a spatiotemporal manner and emphasizes a need for future investigation into the impact of neuronal innervation on NPC gliogenesis.
Collapse
Affiliation(s)
| | - Lauren Chen
- Department of Medical Genetics, Faculty of Medicine & DentistryUniversity of AlbertaEdmontonAlbertaCanada
- Department of Cell Biology, Faculty of Medicine & DentistryUniversity of AlbertaEdmontonAlbertaCanada
| | - Anastassia Voronova
- Neurosciences and Mental Health InstituteUniversity of AlbertaEdmontonAlbertaCanada
- Department of Medical Genetics, Faculty of Medicine & DentistryUniversity of AlbertaEdmontonAlbertaCanada
- Department of Cell Biology, Faculty of Medicine & DentistryUniversity of AlbertaEdmontonAlbertaCanada
- Faculty of Medicine & Dentistry, MS CentreUniversity of AlbertaEdmontonAlbertaCanada
- Women and Children's Health Research InstituteUniversity of AlbertaEdmontonAlbertaCanada
| |
Collapse
|
6
|
da Costa P, Schetinger MRC, Baldissarelli J, Stefanello N, Lopes TF, Reichert KP, Assmann CE, Bottari NB, Miron VV, Vargas FFA, Gutierres JM, da Cruz IBM, Morsch VM. Blackcurrant ( Ribes nigrum L.) improves cholinergic signaling and protects against chronic Scopolamine-induced memory impairment in mice. J Psychopharmacol 2024; 38:1170-1183. [PMID: 39262284 DOI: 10.1177/02698811241273776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/13/2024]
Abstract
BACKGROUND Blackcurrant (Ribes nigrum L.) is a berry rich in anthocyanins, bioactive compounds known for their antioxidant and neuroprotective properties that benefit human health. AIMS This study aimed to investigate the effects of blackcurrant and its association with Donepezil on memory impairment, cholinergic neurotransmission, and antioxidant systems in a mouse model of amnesia induced by chronic administration of Scopolamine. METHODS Adult male Swiss mice were given saline, blackcurrant (50 mg/kg, orally), and/or Donepezil (5 mg/kg, orally) and/or Scopolamine (1 mg/kg, intraperitoneally). RESULTS Behavioral tests revealed that blackcurrant and/or Donepezil prevented the learning and memory deficits induced by Scopolamine. In the cerebral cortex and hippocampus, blackcurrant and/or Donepezil treatments prevented the increase in acetylcholinesterase and butyrylcholinesterase activities induced by Scopolamine. Scopolamine also disrupted the glutathione redox system and increased levels of reactive species; nevertheless, blackcurrant and/or Donepezil treatments were able to prevent oxidative stress. Furthermore, these treatments prevented the increase in gene expression and protein density of acetylcholinesterase and the decrease in gene expression of the choline acetyltransferase enzyme induced by Scopolamine. CONCLUSIONS Findings suggest that blackcurrant and Donepezil, either alone or in combination, have anti-amnesic effects by modulating cholinergic system enzymes and improving the redox profile. Therefore, blackcurrant could be used as a natural supplement for the prevention and treatment of memory impairment in neurodegenerative diseases.
Collapse
Affiliation(s)
- Pauline da Costa
- Postgraduate Program in Toxicological Biochemistry, Department of Biochemistry and Molecular Biology, Federal University of Santa Maria, Santa Maria, RS, Brazil
| | - Maria Rosa C Schetinger
- Postgraduate Program in Toxicological Biochemistry, Department of Biochemistry and Molecular Biology, Federal University of Santa Maria, Santa Maria, RS, Brazil
| | - Jucimara Baldissarelli
- Multicenter Postgraduate Program in Physiological Sciences, Department of Physiology and Pharmacology, Federal University of Pelotas, Pelotas, RS, Brazil
- Postgraduate Program in Biochemistry and Bioprospecting, Federal University of Pelotas, Pelotas, RS, Brazil
| | - Naiara Stefanello
- Postgraduate Program in Toxicological Biochemistry, Department of Biochemistry and Molecular Biology, Federal University of Santa Maria, Santa Maria, RS, Brazil
| | - Thauan F Lopes
- Postgraduate Program in Toxicological Biochemistry, Department of Biochemistry and Molecular Biology, Federal University of Santa Maria, Santa Maria, RS, Brazil
| | - Karine P Reichert
- Postgraduate Program in Toxicological Biochemistry, Department of Biochemistry and Molecular Biology, Federal University of Santa Maria, Santa Maria, RS, Brazil
| | - Charles E Assmann
- Postgraduate Program in Toxicological Biochemistry, Department of Biochemistry and Molecular Biology, Federal University of Santa Maria, Santa Maria, RS, Brazil
| | - Nathieli B Bottari
- Postgraduate Program in Toxicological Biochemistry, Department of Biochemistry and Molecular Biology, Federal University of Santa Maria, Santa Maria, RS, Brazil
- Department of Microbiology and Parasitology, Federal University of Pelotas, Pelotas, RS, Brazil
| | - Vanessa V Miron
- Postgraduate Program in Toxicological Biochemistry, Department of Biochemistry and Molecular Biology, Federal University of Santa Maria, Santa Maria, RS, Brazil
| | - Fermina Francesca A Vargas
- Multicenter Postgraduate Program in Physiological Sciences, Department of Physiology and Pharmacology, Federal University of Pelotas, Pelotas, RS, Brazil
| | - Jessié M Gutierres
- Institute of Biomedical Sciences, Federal University of Alfenas, Alfenas, MG, Brazil
| | - Ivana Beatrice M da Cruz
- Post graduate Program in Gerontology, Federal University of Santa Maria, Santa Maria, RS, Brazil
| | - Vera Maria Morsch
- Postgraduate Program in Toxicological Biochemistry, Department of Biochemistry and Molecular Biology, Federal University of Santa Maria, Santa Maria, RS, Brazil
| |
Collapse
|
7
|
Stedehouder J, Roberts BM, Raina S, Bossi S, Liu AKL, Doig NM, McGerty K, Magill PJ, Parkkinen L, Cragg SJ. Rapid modulation of striatal cholinergic interneurons and dopamine release by satellite astrocytes. Nat Commun 2024; 15:10017. [PMID: 39562551 PMCID: PMC11577008 DOI: 10.1038/s41467-024-54253-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Accepted: 11/01/2024] [Indexed: 11/21/2024] Open
Abstract
Astrocytes are increasingly appreciated to possess underestimated and important roles in modulating neuronal circuits. Astrocytes in striatum can regulate dopamine transmission by governing the extracellular tone of axonal neuromodulators, including GABA and adenosine. However, here we reveal that striatal astrocytes occupy a cell type-specific anatomical and functional relationship with cholinergic interneurons (ChIs), through which they rapidly excite ChIs and govern dopamine release via nicotinic acetylcholine receptors on subsecond timescales. We identify that ChI somata are in unexpectedly close proximity to astrocyte somata, in mouse and human, forming a "soma-to-soma" satellite-like configuration not typically observed for other striatal neurons. We find that transient depolarization of astrocytes in mouse striatum reversibly regulates ChI excitability by decreasing extracellular calcium. These findings reveal a privileged satellite astrocyte-interneuron interaction for striatal ChIs operating on subsecond timescales via regulation of extracellular calcium dynamics to shape downstream striatal circuit activity and dopamine signaling.
Collapse
Affiliation(s)
- Jeffrey Stedehouder
- Centre for Cellular and Molecular Neurobiology, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, OX1 3PT, UK
- Oxford Parkinson's Disease Centre, University of Oxford, Oxford, OX1 3PT, UK
| | - Bradley M Roberts
- Centre for Cellular and Molecular Neurobiology, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, OX1 3PT, UK
- Oxford Parkinson's Disease Centre, University of Oxford, Oxford, OX1 3PT, UK
| | - Shinil Raina
- Centre for Cellular and Molecular Neurobiology, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, OX1 3PT, UK
- Oxford Parkinson's Disease Centre, University of Oxford, Oxford, OX1 3PT, UK
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA
| | - Simon Bossi
- Centre for Cellular and Molecular Neurobiology, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, OX1 3PT, UK
- Oxford Parkinson's Disease Centre, University of Oxford, Oxford, OX1 3PT, UK
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA
| | - Alan King Lun Liu
- Oxford Parkinson's Disease Centre, University of Oxford, Oxford, OX1 3PT, UK
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, OX3 9DU, UK
| | - Natalie M Doig
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA
- Medical Research Council Brain Network Dynamics Unit, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, OX1 3QT, UK
| | - Kevin McGerty
- Mathematical Institute, University of Oxford, Oxford, OX2 6GG, UK
| | - Peter J Magill
- Oxford Parkinson's Disease Centre, University of Oxford, Oxford, OX1 3PT, UK
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA
- Medical Research Council Brain Network Dynamics Unit, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, OX1 3QT, UK
| | - Laura Parkkinen
- Oxford Parkinson's Disease Centre, University of Oxford, Oxford, OX1 3PT, UK
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, OX3 9DU, UK
| | - Stephanie J Cragg
- Centre for Cellular and Molecular Neurobiology, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, OX1 3PT, UK.
- Oxford Parkinson's Disease Centre, University of Oxford, Oxford, OX1 3PT, UK.
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA.
| |
Collapse
|
8
|
Lu J, Chen J, Li SY, Pan GJ, Ou Y, Yuan LF, Jiang JP, Zeng LH, Zhao J. Naringin and Naringenin: Potential Multi-Target Agents for Alzheimer's Disease. Curr Med Sci 2024; 44:867-882. [PMID: 39347923 DOI: 10.1007/s11596-024-2921-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Accepted: 07/15/2024] [Indexed: 10/01/2024]
Abstract
Alzheimer's disease (AD) is one of the most common forms of neurodegenerative dementia. The etiology of AD is multifactorial, and its complex pathophysiology involves tau and amyloid-β deposition, increased oxidative stress, neuroinflammation, metabolic disorders, and massive neuronal loss. Due to its complex pathology, no effective cure for AD has been found to date. Therefore, there is an unmet clinical need for the development of new drugs against AD. Natural products are known to be good sources of compounds with pharmacological activity and have potential for the development of new therapeutic agents. Naringin, a naturally occurring flavanone glycoside, is predominantly found in citrus fruits and Chinese medicinal herbs. Mounting evidence shows that naringin and its aglycone, naringenin, have direct neuroprotective effects on AD, such as anti-amyloidogenic, antioxidant, anti-acetylcholinesterase, and anti-neuroinflammatory effects, as well as metal chelation. Furthermore, they are known to improve disordered glucose/lipid metabolism, which is a high risk factor for AD. In this review, we summarize the latest data on the impact of naringin and naringenin on the molecular mechanisms involved in AD pathophysiology. Additionally, we provide an overview of the current clinical applications of naringin and naringenin. The novel delivery systems for naringin and naringenin, which can address their widespread pharmacokinetic limitations, are also discussed. The literature indicates that naringin and naringenin could be multilevel, multitargeted, and multifaceted for preventing and treating AD.
Collapse
Affiliation(s)
- Jing Lu
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, 310015, China
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, 310015, China
| | - Jie Chen
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, 310015, China
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, 310015, China
| | - Shu-Yue Li
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, 310015, China
| | - Guang-Jie Pan
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, 310015, China
| | - Yi Ou
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, 310015, China
| | - Li-Fu Yuan
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, 310015, China
| | - Jian-Ping Jiang
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, 310015, China.
- Affiliated Hospital, Hangzhou City University School of Medicine, Hangzhou, 310015, China.
| | - Ling-Hui Zeng
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, 310015, China.
| | - Jie Zhao
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, 310015, China.
| |
Collapse
|
9
|
Wang H, Huang M, Yang S, Xu J, Li J, Qin H, Liang S, Teng T, Yang C, Gong M, He Y, Li X, Wang H, Liao X, Chen X, Yang Z, Zhang K. Mapping multi-regional functional connectivity of astrocyte-neuronal networks during behaviors. NEUROPHOTONICS 2024; 11:045010. [PMID: 39554692 PMCID: PMC11566604 DOI: 10.1117/1.nph.11.4.045010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 10/21/2024] [Accepted: 10/22/2024] [Indexed: 11/19/2024]
Abstract
Significance Diverse behaviors rely on coordinated activity and multi-regional functional connectivity within astrocyte-neuronal networks. However, current techniques for simultaneously measuring astrocytic and neuronal activities across multiple brain regions during behaviors remain limited. Aim We propose a multi-fiber solution that can simultaneously record activities of astrocyte-neuronal networks across multiple regions during behaviors. Approach We employed cell-specific dual-color genetically encoded calcium indicators (GECIs) and multi-fiber photometry to simultaneously measure astrocytic and neuronal Ca2+ transients across multiple brain regions in freely behaving animals. Results Our findings demonstrate that both movements and sensory stimuli induce synchronized and highly correlated Ca2+ transients in astrocytes and neurons of freely behaving mice. In addition, we recorded astrocytic and neuronal Ca2+ transients from multiple brain regions during mouse behaviors. Our observations reveal heightened synchronization of astrocytic and neuronal Ca2+ transients across different brain regions during movements or sensory stimuli, indicating enhanced functional connectivity within brain-wide astrocyte-neuronal networks. Conclusions Multi-fiber photometry, combined with cell-specific dual-color GECIs, represents a powerful approach for investigating astrocytic and neuronal activities across different brain regions during behaviors. This technique serves as a versatile tool for analyzing the multi-regional functional connectivity map of astrocyte-neuronal networks associated with specific behaviors.
Collapse
Affiliation(s)
- Haoyu Wang
- Third Military Medical University, Brain Research Center and State Key Laboratory of Trauma and Chemical Poisoning, Chongqing, China
| | - Mingzhu Huang
- Chongqing University, College of Bioengineering, Chongqing, China
| | - Shaofan Yang
- Third Military Medical University, Brain Research Center and State Key Laboratory of Trauma and Chemical Poisoning, Chongqing, China
| | - Jiameng Xu
- Tiangong University, The School of Control Science and Engineering, Tianjin, China
| | - Jin Li
- Third Military Medical University, Brain Research Center and State Key Laboratory of Trauma and Chemical Poisoning, Chongqing, China
| | - Han Qin
- Chongqing Institute for Brain and Intelligence, Guangyang Bay Laboratory, Chongqing, China
| | - Shanshan Liang
- Third Military Medical University, Brain Research Center and State Key Laboratory of Trauma and Chemical Poisoning, Chongqing, China
| | - Teng Teng
- Third Military Medical University, Brain Research Center and State Key Laboratory of Trauma and Chemical Poisoning, Chongqing, China
| | - Chuanyan Yang
- Third Military Medical University, Brain Research Center and State Key Laboratory of Trauma and Chemical Poisoning, Chongqing, China
| | - Mingyue Gong
- Third Military Medical University, Brain Research Center and State Key Laboratory of Trauma and Chemical Poisoning, Chongqing, China
| | - Yong He
- Peking university, School of Electronics, Beijing, China
| | - Xingyi Li
- Chongqing University, School of Medicine, Center for Neurointelligence, Chongqing, China
| | - Huiquan Wang
- Tiangong University, The School of Control Science and Engineering, Tianjin, China
| | - Xiang Liao
- Chongqing University, School of Medicine, Center for Neurointelligence, Chongqing, China
| | - Xiaowei Chen
- Third Military Medical University, Brain Research Center and State Key Laboratory of Trauma and Chemical Poisoning, Chongqing, China
| | - Zhiqi Yang
- Gansu Provincial Central Hospital, Department of Neurology, Lanzhou, China
| | - Kuan Zhang
- Third Military Medical University, Brain Research Center and State Key Laboratory of Trauma and Chemical Poisoning, Chongqing, China
| |
Collapse
|
10
|
Uribe-Cano S, Kottmann AH. The primary cilium of cholinergic neurons may be a linchpin in the progression of Parkinson's Disease. Proc Natl Acad Sci U S A 2024; 121:e2414226121. [PMID: 39288184 PMCID: PMC11441531 DOI: 10.1073/pnas.2414226121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/19/2024] Open
Affiliation(s)
- Santiago Uribe-Cano
- City University of New York (CUNY) Graduate Center, Neuroscience Collaborative, New York, NY 10016
- Department of Molecular, Cellular and Biomedical Sciences, The City University of New York (CUNY) School of Medicine, New York, NY 10031
| | - Andreas H Kottmann
- City University of New York (CUNY) Graduate Center, Neuroscience Collaborative, New York, NY 10016
- Department of Molecular, Cellular and Biomedical Sciences, The City University of New York (CUNY) School of Medicine, New York, NY 10031
| |
Collapse
|
11
|
Fagerli E, Jackson CW, Escobar I, Ferrier FJ, Perez Lao EJ, Saul I, Gomez J, Dave KR, Bracko O, Perez-Pinzon MA. Resveratrol Mitigates Cognitive Impairments and Cholinergic Cell Loss in the Medial Septum in a Mouse Model of Gradual Cerebral Hypoperfusion. Antioxidants (Basel) 2024; 13:984. [PMID: 39199230 PMCID: PMC11351397 DOI: 10.3390/antiox13080984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 08/03/2024] [Accepted: 08/06/2024] [Indexed: 09/01/2024] Open
Abstract
Vascular cognitive impairment and dementia (VCID) is the second leading cause of dementia. There is currently no effective treatment for VCID. Resveratrol (RSV) is considered an antioxidant; however, our group has observed pleiotropic effects in stroke paradigms, suggesting more effects may contribute to mechanistic changes beyond antioxidative properties. The main goal of this study was to investigate if administering RSV twice a week could alleviate cognitive declines following the induction of a VCID model. Additionally, our aim was to further describe whether this treatment regimen could decrease cell death in brain areas vulnerable to changes in cerebral blood flow, such as the hippocampus and medial septum. We hypothesized RSV treatments in a mouse model of gradual cerebral hypoperfusion protect against cognitive impairment. We utilized gradual bilateral common carotid artery stenosis (GBCCAS) via the surgical implantation of ameroid constrictor devices. RSV treatment was administered on the day of implantation and twice a week thereafter. Cerebral perfusion was measured by laser speckle contrast imaging, and cognitive functions, including the recognition memory, the spatial working memory, and associative learning, were assessed by novel object recognition (NOR), Y-maze testing, and contextual fear conditioning (CFC), respectively. RSV treatment did not alleviate cerebral perfusion deficits but mitigated cognitive deficits in CFC and NOR after GBCCAS. Despite these deficits, no hippocampal pathology was observed; however, cholinergic cell loss in the medial septum was significantly increased after GBCCAS. This cholinergic cell loss was mitigated by RSV. This study describes a novel mechanism by which chronic RSV treatments protect against a VCID-induced cognitive decline through the preservation of cholinergic cell viability to improve memory performance.
Collapse
Affiliation(s)
- Eric Fagerli
- Peritz Scheinberg Cerebral Vascular Disease Research Laboratories, University of Miami Leonard M. Miller School of Medicine, Miami, FL 33136, USA; (E.F.); (C.W.J.); (I.E.); (F.J.F.); (E.J.P.L.); (I.S.); (J.G.); (K.R.D.)
- Department of Neurology, University of Miami Leonard M. Miller School of Medicine, Miami, FL 33136, USA;
- Neuroscience Program, University of Miami Leonard M. Miller School of Medicine, Miami, FL 33136, USA
| | - Charles W. Jackson
- Peritz Scheinberg Cerebral Vascular Disease Research Laboratories, University of Miami Leonard M. Miller School of Medicine, Miami, FL 33136, USA; (E.F.); (C.W.J.); (I.E.); (F.J.F.); (E.J.P.L.); (I.S.); (J.G.); (K.R.D.)
- Department of Neurology, University of Miami Leonard M. Miller School of Medicine, Miami, FL 33136, USA;
- Neuroscience Program, University of Miami Leonard M. Miller School of Medicine, Miami, FL 33136, USA
| | - Iris Escobar
- Peritz Scheinberg Cerebral Vascular Disease Research Laboratories, University of Miami Leonard M. Miller School of Medicine, Miami, FL 33136, USA; (E.F.); (C.W.J.); (I.E.); (F.J.F.); (E.J.P.L.); (I.S.); (J.G.); (K.R.D.)
- Department of Neurology, University of Miami Leonard M. Miller School of Medicine, Miami, FL 33136, USA;
- Neuroscience Program, University of Miami Leonard M. Miller School of Medicine, Miami, FL 33136, USA
| | - Fernando J. Ferrier
- Peritz Scheinberg Cerebral Vascular Disease Research Laboratories, University of Miami Leonard M. Miller School of Medicine, Miami, FL 33136, USA; (E.F.); (C.W.J.); (I.E.); (F.J.F.); (E.J.P.L.); (I.S.); (J.G.); (K.R.D.)
- Department of Neurology, University of Miami Leonard M. Miller School of Medicine, Miami, FL 33136, USA;
- Neuroscience Program, University of Miami Leonard M. Miller School of Medicine, Miami, FL 33136, USA
| | - Efrain J. Perez Lao
- Peritz Scheinberg Cerebral Vascular Disease Research Laboratories, University of Miami Leonard M. Miller School of Medicine, Miami, FL 33136, USA; (E.F.); (C.W.J.); (I.E.); (F.J.F.); (E.J.P.L.); (I.S.); (J.G.); (K.R.D.)
- Department of Neurology, University of Miami Leonard M. Miller School of Medicine, Miami, FL 33136, USA;
- Neuroscience Program, University of Miami Leonard M. Miller School of Medicine, Miami, FL 33136, USA
| | - Isabel Saul
- Peritz Scheinberg Cerebral Vascular Disease Research Laboratories, University of Miami Leonard M. Miller School of Medicine, Miami, FL 33136, USA; (E.F.); (C.W.J.); (I.E.); (F.J.F.); (E.J.P.L.); (I.S.); (J.G.); (K.R.D.)
- Department of Neurology, University of Miami Leonard M. Miller School of Medicine, Miami, FL 33136, USA;
| | - Jorge Gomez
- Peritz Scheinberg Cerebral Vascular Disease Research Laboratories, University of Miami Leonard M. Miller School of Medicine, Miami, FL 33136, USA; (E.F.); (C.W.J.); (I.E.); (F.J.F.); (E.J.P.L.); (I.S.); (J.G.); (K.R.D.)
- Department of Neurology, University of Miami Leonard M. Miller School of Medicine, Miami, FL 33136, USA;
| | - Kunjan R. Dave
- Peritz Scheinberg Cerebral Vascular Disease Research Laboratories, University of Miami Leonard M. Miller School of Medicine, Miami, FL 33136, USA; (E.F.); (C.W.J.); (I.E.); (F.J.F.); (E.J.P.L.); (I.S.); (J.G.); (K.R.D.)
- Department of Neurology, University of Miami Leonard M. Miller School of Medicine, Miami, FL 33136, USA;
- Neuroscience Program, University of Miami Leonard M. Miller School of Medicine, Miami, FL 33136, USA
| | - Oliver Bracko
- Department of Neurology, University of Miami Leonard M. Miller School of Medicine, Miami, FL 33136, USA;
- Neuroscience Program, University of Miami Leonard M. Miller School of Medicine, Miami, FL 33136, USA
- Department of Biology, University of Miami, Coral Gables, FL 33146, USA
| | - Miguel A. Perez-Pinzon
- Peritz Scheinberg Cerebral Vascular Disease Research Laboratories, University of Miami Leonard M. Miller School of Medicine, Miami, FL 33136, USA; (E.F.); (C.W.J.); (I.E.); (F.J.F.); (E.J.P.L.); (I.S.); (J.G.); (K.R.D.)
- Department of Neurology, University of Miami Leonard M. Miller School of Medicine, Miami, FL 33136, USA;
- Neuroscience Program, University of Miami Leonard M. Miller School of Medicine, Miami, FL 33136, USA
| |
Collapse
|
12
|
Khan SS, Jaimon E, Lin YE, Nikoloff J, Tonelli F, Alessi DR, Pfeffer SR. Loss of primary cilia and dopaminergic neuroprotection in pathogenic LRRK2-driven and idiopathic Parkinson's disease. Proc Natl Acad Sci U S A 2024; 121:e2402206121. [PMID: 39088390 PMCID: PMC11317616 DOI: 10.1073/pnas.2402206121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 06/13/2024] [Indexed: 08/03/2024] Open
Abstract
Activating leucine-rich repeat kinase 2 (LRRK2) mutations cause Parkinson's and phosphorylation of Rab10 by pathogenic LRRK2 blocks primary ciliogenesis in cultured cells. In the mouse brain, LRRK2 blockade of primary cilia is highly cell type specific: For example, cholinergic interneurons and astrocytes but not medium spiny neurons of the dorsal striatum lose primary cilia in LRRK2-pathway mutant mice. We show here that the cell type specificity of LRRK2-mediated cilia loss is also seen in human postmortem striatum from patients with LRRK2 pathway mutations and idiopathic Parkinson's. Single nucleus RNA sequencing shows that cilia loss in mouse cholinergic interneurons is accompanied by decreased glial-derived neurotrophic factor transcription, decreasing neuroprotection for dopamine neurons. Nevertheless, LRRK2 expression differences cannot explain the unique vulnerability of cholinergic neurons to LRRK2 kinase as much higher LRRK2 expression is seen in medium spiny neurons that have normal cilia. In parallel with decreased striatal dopaminergic neurite density, LRRK2 G2019S neurons show increased autism-linked CNTN5 adhesion protein expression; glial cells show significant loss of ferritin heavy chain. These data strongly suggest that loss of cilia in specific striatal cell types decreases neuroprotection for dopamine neurons in mice and human Parkinson's.
Collapse
Affiliation(s)
- Shahzad S. Khan
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA94305-5307
- Aligning Science Across Parkinson’s Collaborative Research Network, Chevy Chase, MD20815
| | - Ebsy Jaimon
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA94305-5307
- Aligning Science Across Parkinson’s Collaborative Research Network, Chevy Chase, MD20815
| | - Yu-En Lin
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA94305-5307
- Aligning Science Across Parkinson’s Collaborative Research Network, Chevy Chase, MD20815
| | - Jonas Nikoloff
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA94305-5307
- Aligning Science Across Parkinson’s Collaborative Research Network, Chevy Chase, MD20815
| | - Francesca Tonelli
- Aligning Science Across Parkinson’s Collaborative Research Network, Chevy Chase, MD20815
- Medical Research Council Protein Phosphorylation and Ubiquitylation Unit, University of Dundee, DundeeDD1 5EH, Scotland, United Kingdom
| | - Dario R. Alessi
- Aligning Science Across Parkinson’s Collaborative Research Network, Chevy Chase, MD20815
- Medical Research Council Protein Phosphorylation and Ubiquitylation Unit, University of Dundee, DundeeDD1 5EH, Scotland, United Kingdom
| | - Suzanne R. Pfeffer
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA94305-5307
- Aligning Science Across Parkinson’s Collaborative Research Network, Chevy Chase, MD20815
| |
Collapse
|
13
|
Fitzsimons LA, Atif-Sheikh M, Lovely J, Mueth M, Rice M, Kotredes K, Howell G, Harrison BJ. CD2AP is Co-Expressed with Tropomyosin-Related Kinase A and Ras-Related Protein Rab-5A in Cholinergic Neurons of the Murine Basal Forebrain. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.24.604961. [PMID: 39211110 PMCID: PMC11361140 DOI: 10.1101/2024.07.24.604961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Basal forebrain cholinergic neurons project to the hippocampus and cortex, are critical for learning and memory, and are central to the pathogenesis of Alzheimer's disease (AD). GWAS have consistently shown that genomic variants at the CD2AP gene locus are associated with significant increased risk of AD. GWAS studies have also shown that genetic variants in endocytosis genes, including RAB5A , significantly increase susceptibility to AD. Previous work in our lab has shown that CD2AP functions as a docking-scaffold/adaptor protein as a coordinator of nerve growth factor (NGF) and trophic signaling in neurons. We have also demonstrated that CD2AP positively regulates Rab5-mediated mechanisms of endocytosis in primary sensory neurons. The purpose of this study was to perform an in vivo characterization of CD2AP expression in cholinergic neurons of the brain regions most relevant to AD pathogenesis and to investigate the colocalization of CD2AP and Rab5 in cholinergic neurons of the murine basal forebrain. Brain tissue was perfused, harvested from ChAT BAC -eGFP transgenic mice (N=4 male, N=4 female; aged 10 mo), where cholinergic neurons (co-) express green fluorescence protein (GFP) in central and peripheral neurons that express choline acetyltransferase (ChAT). Frozen tissue sections were used to assess the specificity of the reporter in mouse brain along with localization of both CD2AP and Rab5 (co-) expression using immunofluorescence (IF) analysis of ChAT-GFP+ neurons and primary antibodies against ChAT, CD2AP and Rab5. Image J software was used to develop and optimize a colocalization assay for CD2AP and Rab5 puncta. Experiments were repeated in a follow-up cohort of aged-adult mice (N=2 male, N=2 female; aged 18 mo). IF expression of CD2AP was quantified in the basal forebrain, diagonal band of Broca (vDB), and striatal regions and compared to results from the cortical regions of the adult mouse brain. Colocalization of CD2AP was observed in the cell bodies of ChAT-GFP+ neurons of the striatum, vDB and basal forebrain regions, where CD2AP expression intensity as well as the number of cell bodies with positive signal increased incrementally. Colocalization analyses revealed near-complete overlap of CD2AP and Rab5 expression in ChAT-GFP+ cholinergic neurons of the basal forebrain region. We conclude that cholinergic neurons express CD2AP in healthy adult and aged-adult mouse brains. These data provide the first evidence of quantifiable CD2AP protein expression of cholinergic neurons specific to the diagonal band of Broca (vDB) and basal forebrain. Together with previous research from our lab, these data support a role for CD2AP in the pathogenesis of AD through orchestration of endocytosis and retrograde signaling. Ongoing studies are underway to verify these findings in a novel AD mouse model that incorporates the humanized variant of CD2AP , created by MODEL-AD, where we aim to further investigate how CD2AP variants may affect mechanistic components of Rab5 endocytosis as well as subsequent survival of cholinergic neurons in the context of known amyloid beta and Tau pathologies.
Collapse
|
14
|
Oliver Goral R, Lamb PW, Yakel JL. Acetylcholine Neurons Become Cholinergic during Three Time Windows in the Developing Mouse Brain. eNeuro 2024; 11:ENEURO.0542-23.2024. [PMID: 38942474 PMCID: PMC11253243 DOI: 10.1523/eneuro.0542-23.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 06/12/2024] [Accepted: 06/22/2024] [Indexed: 06/30/2024] Open
Abstract
Acetylcholine (ACh) neurons in the central nervous system are required for the coordination of neural network activity during higher brain functions, such as attention, learning, and memory, as well as locomotion. Disturbed cholinergic signaling has been described in many neurodevelopmental and neurodegenerative disorders. Furthermore, cotransmission of other signaling molecules, such as glutamate and GABA, with ACh has been associated with essential roles in brain function or disease. However, it is unknown when ACh neurons become cholinergic during development. Thus, understanding the timeline of how the cholinergic system develops and becomes active in the healthy brain is a crucial part of understanding brain development. To study this, we used transgenic mice to selectively label ACh neurons with tdTomato. We imaged serial sectioned brains and generated whole-brain reconstructions at different time points during pre- and postnatal development. We found three crucial time windows-two in the prenatal and one in the postnatal brain-during which most ACh neuron populations become cholinergic in the brain. We also found that cholinergic gene expression is initiated in cortical ACh interneurons, while the cerebral cortex is innervated by cholinergic projection neurons from the basal forebrain. Taken together, we show that ACh neuron populations are present and become cholinergic before postnatal day 12, which is the onset of major sensory processes, such as hearing and vision. We conclude that the birth of ACh neurons and initiation of cholinergic gene expression are temporally separated during development but highly coordinated by brain anatomical structure.
Collapse
Affiliation(s)
- Rene Oliver Goral
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, North Carolina 27709
- Center on Compulsive Behaviors, National Institutes of Health, Bethesda, Maryland 20892
| | - Patricia W Lamb
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, North Carolina 27709
| | - Jerrel L Yakel
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, North Carolina 27709
| |
Collapse
|
15
|
Lozovaya N, Moumen A, Hammond C. Basal Forebrain Cholinergic Neurons Have Specific Characteristics during the Perinatal Period. eNeuro 2024; 11:ENEURO.0538-23.2024. [PMID: 38755010 PMCID: PMC11137802 DOI: 10.1523/eneuro.0538-23.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 04/09/2024] [Accepted: 05/05/2024] [Indexed: 05/18/2024] Open
Abstract
Cholinergic neurons of the basal forebrain represent the main source of cholinergic innervation of large parts of the neocortex and are involved in adults in the modulation of attention, memory, and arousal. During the first postnatal days, they play a crucial role in the development of cortical neurons and cortical cytoarchitecture. However, their characteristics, during this period have not been studied. To understand how they can fulfill this role, we investigated the morphological and electrophysiological maturation of cholinergic neurons of the substantia innominata-nucleus basalis of Meynert (SI/NBM) complex in the perinatal period in mice. We show that cholinergic neurons, whether or not they express gamma-aminobutyric acid (GABA) as a cotransmitter, are already functional at Embryonic Day 18. Until the end of the first postnatal week, they constitute a single population of neurons with a well developed dendritic tree, a spontaneous activity including bursting periods, and a short-latency response to depolarizations (early-firing). They are excited by both their GABAergic and glutamatergic afferents. During the second postnatal week, a second, less excitable, neuronal population emerges, with a longer delay response to depolarizations (late-firing), together with the hyperpolarizing action of GABAA receptor-mediated currents. This classification into early-firing (40%) and late-firing (60%) neurons is again independent of the coexpression of GABAergic markers. These results strongly suggest that during the first postnatal week, the specific properties of developing SI/NBM cholinergic neurons allow them to spontaneously release acetylcholine (ACh), or ACh and GABA, into the developing cortex.
Collapse
|
16
|
Chakraborty S, Lee SK, Arnold SM, Haast RAM, Khan AR, Schmitz TW. Focal acetylcholinergic modulation of the human midcingulo-insular network during attention: Meta-analytic neuroimaging and behavioral evidence. J Neurochem 2024; 168:397-413. [PMID: 37864501 DOI: 10.1111/jnc.15990] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2023] [Revised: 09/18/2023] [Accepted: 09/26/2023] [Indexed: 10/23/2023]
Abstract
The basal forebrain cholinergic neurons provide acetylcholine to the cortex via large projections. Recent molecular imaging work in humans indicates that the cortical cholinergic innervation is not uniformly distributed, but rather may disproportionately innervate cortical areas relevant to supervisory attention. In this study, we therefore reexamined the spatial relationship between acetylcholinergic modulation and attention in the human cortex using meta-analytic strategies targeting both pharmacological and non-pharmacological neuroimaging studies. We found that pharmaco-modulation of acetylcholine evoked both increased activity in the anterior cingulate and decreased activity in the opercular and insular cortex. In large independent meta-analyses of non-pharmacological neuroimaging research, we demonstrate that during attentional engagement these cortical areas exhibit (1) task-related co-activation with the basal forebrain, (2) task-related co-activation with one another, and (3) spatial overlap with dense cholinergic innervations originating from the basal forebrain, as estimated by multimodal positron emission tomography and magnetic resonance imaging. Finally, we provide meta-analytic evidence that pharmaco-modulation of acetylcholine also induces a speeding of responses to targets with no apparent tradeoff in accuracy. In sum, we demonstrate in humans that acetylcholinergic modulation of midcingulo-insular hubs of the ventral attention/salience network via basal forebrain afferents may coordinate selection of task relevant information, thereby facilitating cognition and behavior.
Collapse
Affiliation(s)
- Sudesna Chakraborty
- Neuroscience Graduate Program, Western University, London, Ontario, Canada
- Department of Medical Biophysics, Western University, London, Ontario, Canada
- Robarts Research Institute, Western University, London, Ontario, Canada
- Western Institute for Neuroscience, Western University, London, Ontario, Canada
| | - Sun Kyun Lee
- Western Institute for Neuroscience, Western University, London, Ontario, Canada
- Faculty of Dentistry, University of Toronto, Toronto, Ontario, Canada
| | - Sarah M Arnold
- Western Institute for Neuroscience, Western University, London, Ontario, Canada
- Department of Chemical Engineering, McMaster University, Hamilton, Ontario, Canada
| | - Roy A M Haast
- Western Institute for Neuroscience, Western University, London, Ontario, Canada
- CRMBM, CNRS UMR 7339, Aix-Marseille University, Marseille, France
| | - Ali R Khan
- Department of Medical Biophysics, Western University, London, Ontario, Canada
- Robarts Research Institute, Western University, London, Ontario, Canada
- Western Institute for Neuroscience, Western University, London, Ontario, Canada
| | - Taylor W Schmitz
- Robarts Research Institute, Western University, London, Ontario, Canada
- Western Institute for Neuroscience, Western University, London, Ontario, Canada
- Department of Physiology and Pharmacology, Western University, London, Ontario, Canada
- Lawson Health Research Institute, London, Ontario, Canada
| |
Collapse
|
17
|
Khan SS, Jaimon E, Lin YE, Nikoloff J, Tonelli F, Alessi DR, Pfeffer SR. Loss of primary cilia and dopaminergic neuroprotection in pathogenic LRRK2-driven and idiopathic Parkinson's disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.15.575737. [PMID: 38293195 PMCID: PMC10827083 DOI: 10.1101/2024.01.15.575737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2024]
Abstract
Activating LRRK2 mutations cause Parkinson's disease. Previously, we showed that cholinergic interneurons and astrocytes but not medium spiny neurons of the dorsal striatum lose primary cilia in LRRK2 mutant mice. Single nucleus RNA sequencing shows that cilia loss in cholinergic interneurons correlates with higher LRRK2 expression and decreased glial derived neurotrophic factor transcription. Nevertheless, much higher LRRK2 expression is seen in medium spiny neurons that have normal cilia in mice and humans. In parallel with decreased striatal dopaminergic neurite density, LRRK2 G2019S neurons show increased autism-linked CNTN5 adhesion protein expression; glial cells show significant loss of ferritin heavy chain. Human striatal tissue from LRRK2 pathway mutation carriers and idiopathic Parkinson's disease show similar cilia loss in cholinergic interneurons and astrocytes and overall loss of such neurons. These data strongly suggest that loss of cilia in specific striatal cell types decreases neuroprotection for dopamine neurons in mice and human Parkinson's disease.
Collapse
Affiliation(s)
- Shahzad S. Khan
- Department of Biochemistry, Stanford University School of Medicine, United States
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, United States
- Current address: Departments of Cell Biology & Physiology and Neurology, University of North Carolina, Chapel Hill, United States
| | - Ebsy Jaimon
- Department of Biochemistry, Stanford University School of Medicine, United States
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, United States
| | - Yu-En Lin
- Department of Biochemistry, Stanford University School of Medicine, United States
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, United States
| | - Jonas Nikoloff
- Department of Biochemistry, Stanford University School of Medicine, United States
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, United States
| | - Francesca Tonelli
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, United States
- MRC Protein Phosphorylation and Ubiquitylation Unit, University of Dundee, United Kingdom
| | - Dario R. Alessi
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, United States
- MRC Protein Phosphorylation and Ubiquitylation Unit, University of Dundee, United Kingdom
| | - Suzanne R. Pfeffer
- Department of Biochemistry, Stanford University School of Medicine, United States
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, United States
| |
Collapse
|
18
|
D'Aloia A, Pastori V, Blasa S, Campioni G, Peri F, Sacco E, Ceriani M, Lecchi M, Costa B. A new advanced cellular model of functional cholinergic-like neurons developed by reprogramming the human SH-SY5Y neuroblastoma cell line. Cell Death Discov 2024; 10:24. [PMID: 38216593 PMCID: PMC10786877 DOI: 10.1038/s41420-023-01790-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 12/11/2023] [Accepted: 12/20/2023] [Indexed: 01/14/2024] Open
Abstract
Modeling human neuronal properties in physiological and pathological conditions is essential to identify novel potential drugs and to explore pathological mechanisms of neurological diseases. For this purpose, we generated a three-dimensional (3D) neuronal culture, by employing the readily available human neuroblastoma SH-SY5Y cell line, and a new differentiation protocol. The entire differentiation process occurred in a matrix and lasted 47 days, with 7 days of pre-differentiation phase and 40 days of differentiation, and allowed the development of a 3D culture in conditions consistent with the physiological environment. Neurons in the culture were electrically active, were able to establish functional networks, and showed features of cholinergic neurons. Hence here we provide an easily accessible, reproducible, and suitable culture method that might empower studies on synaptic function, vesicle trafficking, and metabolism, which sustain neuronal activity and cerebral circuits. Moreover, this novel differentiation protocol could represent a promising cellular tool to study physiological cellular processes, such as migration, differentiation, maturation, and to develop novel therapeutic approaches.
Collapse
Affiliation(s)
- Alessia D'Aloia
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Piazza della Scienza 2, 20126, Milano, Italy.
| | - Valentina Pastori
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Piazza della Scienza 2, 20126, Milano, Italy
| | - Stefania Blasa
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Piazza della Scienza 2, 20126, Milano, Italy
- Milan Center for Neuroscience (NeuroMI), University of Milano-Bicocca, Piazza dell'Ateneo Nuovo 1, 20126, Milano, Italy
| | - Gloria Campioni
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Piazza della Scienza 2, 20126, Milano, Italy
- SYSBIO-ISBE-IT, Europe, 20126, Milano, Italy
| | - Francesco Peri
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Piazza della Scienza 2, 20126, Milano, Italy
| | - Elena Sacco
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Piazza della Scienza 2, 20126, Milano, Italy
- Milan Center for Neuroscience (NeuroMI), University of Milano-Bicocca, Piazza dell'Ateneo Nuovo 1, 20126, Milano, Italy
- SYSBIO-ISBE-IT, Europe, 20126, Milano, Italy
- Inter-University Center for the Promotion of the 3Rs Principles in Teaching & Research, Pisa, Italy
| | - Michela Ceriani
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Piazza della Scienza 2, 20126, Milano, Italy
- Milan Center for Neuroscience (NeuroMI), University of Milano-Bicocca, Piazza dell'Ateneo Nuovo 1, 20126, Milano, Italy
| | - Marzia Lecchi
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Piazza della Scienza 2, 20126, Milano, Italy
- Milan Center for Neuroscience (NeuroMI), University of Milano-Bicocca, Piazza dell'Ateneo Nuovo 1, 20126, Milano, Italy
- Inter-University Center for the Promotion of the 3Rs Principles in Teaching & Research, Pisa, Italy
| | - Barbara Costa
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Piazza della Scienza 2, 20126, Milano, Italy
- Milan Center for Neuroscience (NeuroMI), University of Milano-Bicocca, Piazza dell'Ateneo Nuovo 1, 20126, Milano, Italy
| |
Collapse
|
19
|
Nagori K, Pradhan M, Sharma M, Ajazuddin, Badwaik HR, Nakhate KT. Current Progress on Central Cholinergic Receptors as Therapeutic Targets for Alzheimer's Disease. Curr Alzheimer Res 2024; 21:50-68. [PMID: 38529600 DOI: 10.2174/0115672050306008240321034006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 02/22/2024] [Accepted: 02/26/2024] [Indexed: 03/27/2024]
Abstract
Acetylcholine (ACh) is ubiquitously present in the nervous system and has been involved in the regulation of various brain functions. By modulating synaptic transmission and promoting synaptic plasticity, particularly in the hippocampus and cortex, ACh plays a pivotal role in the regulation of learning and memory. These procognitive actions of ACh are mediated by the neuronal muscarinic and nicotinic cholinergic receptors. The impairment of cholinergic transmission leads to cognitive decline associated with aging and dementia. Therefore, the cholinergic system has been of prime focus when concerned with Alzheimer's disease (AD), the most common cause of dementia. In AD, the extensive destruction of cholinergic neurons occurs by amyloid-β plaques and tau protein-rich neurofibrillary tangles. Amyloid-β also blocks cholinergic receptors and obstructs neuronal signaling. This makes the central cholinergic system an important target for the development of drugs for AD. In fact, centrally acting cholinesterase inhibitors like donepezil and rivastigmine are approved for the treatment of AD, although the outcome is not satisfactory. Therefore, identification of specific subtypes of cholinergic receptors involved in the pathogenesis of AD is essential to develop future drugs. Also, the identification of endogenous rescue mechanisms to the cholinergic system can pave the way for new drug development. In this article, we discussed the neuroanatomy of the central cholinergic system. Further, various subtypes of muscarinic and nicotinic receptors involved in the cognition and pathophysiology of AD are described in detail. The article also reviewed primary neurotransmitters that regulate cognitive processes by modulating basal forebrain cholinergic projection neurons.
Collapse
Affiliation(s)
- Kushagra Nagori
- Department of Pharmaceutical Chemistry, Rungta College of Pharmaceutical Sciences and Research, Kurud Road, Kohka, Bhilai 490024, Chhattisgarh, India
| | - Madhulika Pradhan
- Department of Pharmaceutical Technology, Gracious College of Pharmacy, Abhanpur 493661, Chhattisgarh, India
| | - Mukesh Sharma
- Department of Pharmacognosy, Rungta College of Pharmaceutical Sciences and Research, Kurud Road, Kohka, Bhilai 490024, Chhattisgarh, India
| | - Ajazuddin
- Department of Pharmaceutics, Rungta College of Pharmaceutical Sciences and Research, Kurud Road, Kohka, Bhilai 490024, Chhattisgarh, India
| | - Hemant R Badwaik
- Department of Pharmaceutical Chemistry, Shri Shankaracharya Institute of Pharmaceutical Sciences and Research, Junwani, Bhilai 490020, Chhattisgarh, India
| | - Kartik T Nakhate
- Department of Pharmacology, Shri Vile Parle Kelavani Mandal's Institute of Pharmacy, Dhule 424001, Maharashtra, India
| |
Collapse
|
20
|
Sabbir MG. Cholinergic Receptor Muscarinic 1 Co-Localized with Mitochondria in Cultured Dorsal Root Ganglion Neurons, and Its Deletion Disrupted Mitochondrial Ultrastructure in Peripheral Neurons: Implications in Alzheimer's Disease. J Alzheimers Dis 2024; 98:247-264. [PMID: 38427478 DOI: 10.3233/jad-230883] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/03/2024]
Abstract
Background Loss of Cholinergic Receptor Muscarinic 1 (CHRM1) has been linked to the pathogenesis of Alzheimer's disease (AD). Our recent study found significantly lower CHRM1 protein levels in AD patient cortices, linked to reduced survival. Furthermore, using knockout mice (Chrm1-/-) we demonstrated that deletion of Chrm1 alters cortical mitochondrial structure and function, directly establishing a connection between its loss and mitochondrial dysfunction in the context of AD. While CHRM1's role in the brain has been extensively investigated, its impact on peripheral neurons in AD remains a crucial area of research, especially considering reported declines in peripheral nerve conduction among AD patients. Objective The objective was to characterize Chrm1 localization and mitochondrial deficits in Chrm1-/- dorsal root ganglion (DRG) neurons. Methods Recombinant proteins tagged with Green or Red Fluorescent Protein (GFP/RFP) were transiently expressed to investigate the localization of Chrm1 and mitochondria, as well as mitochondrial movement in the neurites of cultured primary mouse DRG neurons, using confocal time-lapse live cell imaging. Transmission electron microscopy was performed to examine the ultrastructure of mitochondria in both wild-type and Chrm1-/- DRGs. Results Fluorescence imaging revealed colocalization and comigration of N-terminal GFP-tagged Chrm1 and mitochondrial localization signal peptide-tagged RFP-labelled mitochondria in the DRGs neurons. A spectrum of mitochondrial structural abnormalities, including disruption and loss of cristae was observed in 87% neurons in Chrm1-/- DRGs. Conclusions This study suggests that Chrm1 may be localized in the neuronal mitochondria and loss of Chrm1 in peripheral neurons causes sever mitochondrial structural aberrations resembling AD pathology.
Collapse
Affiliation(s)
- Mohammad Golam Sabbir
- Department of Psychology and Neuroscience, Collegeof Psychology, Nova Southeastern University, Fort Lauderdale, FL, USA
- Alzo Biosciences Inc., San Diego, CA, USA
| |
Collapse
|
21
|
Yao Z, van Velthoven CTJ, Kunst M, Zhang M, McMillen D, Lee C, Jung W, Goldy J, Abdelhak A, Aitken M, Baker K, Baker P, Barkan E, Bertagnolli D, Bhandiwad A, Bielstein C, Bishwakarma P, Campos J, Carey D, Casper T, Chakka AB, Chakrabarty R, Chavan S, Chen M, Clark M, Close J, Crichton K, Daniel S, DiValentin P, Dolbeare T, Ellingwood L, Fiabane E, Fliss T, Gee J, Gerstenberger J, Glandon A, Gloe J, Gould J, Gray J, Guilford N, Guzman J, Hirschstein D, Ho W, Hooper M, Huang M, Hupp M, Jin K, Kroll M, Lathia K, Leon A, Li S, Long B, Madigan Z, Malloy J, Malone J, Maltzer Z, Martin N, McCue R, McGinty R, Mei N, Melchor J, Meyerdierks E, Mollenkopf T, Moonsman S, Nguyen TN, Otto S, Pham T, Rimorin C, Ruiz A, Sanchez R, Sawyer L, Shapovalova N, Shepard N, Slaughterbeck C, Sulc J, Tieu M, Torkelson A, Tung H, Valera Cuevas N, Vance S, Wadhwani K, Ward K, Levi B, Farrell C, Young R, Staats B, Wang MQM, Thompson CL, Mufti S, Pagan CM, Kruse L, Dee N, Sunkin SM, Esposito L, Hawrylycz MJ, Waters J, Ng L, Smith K, Tasic B, Zhuang X, et alYao Z, van Velthoven CTJ, Kunst M, Zhang M, McMillen D, Lee C, Jung W, Goldy J, Abdelhak A, Aitken M, Baker K, Baker P, Barkan E, Bertagnolli D, Bhandiwad A, Bielstein C, Bishwakarma P, Campos J, Carey D, Casper T, Chakka AB, Chakrabarty R, Chavan S, Chen M, Clark M, Close J, Crichton K, Daniel S, DiValentin P, Dolbeare T, Ellingwood L, Fiabane E, Fliss T, Gee J, Gerstenberger J, Glandon A, Gloe J, Gould J, Gray J, Guilford N, Guzman J, Hirschstein D, Ho W, Hooper M, Huang M, Hupp M, Jin K, Kroll M, Lathia K, Leon A, Li S, Long B, Madigan Z, Malloy J, Malone J, Maltzer Z, Martin N, McCue R, McGinty R, Mei N, Melchor J, Meyerdierks E, Mollenkopf T, Moonsman S, Nguyen TN, Otto S, Pham T, Rimorin C, Ruiz A, Sanchez R, Sawyer L, Shapovalova N, Shepard N, Slaughterbeck C, Sulc J, Tieu M, Torkelson A, Tung H, Valera Cuevas N, Vance S, Wadhwani K, Ward K, Levi B, Farrell C, Young R, Staats B, Wang MQM, Thompson CL, Mufti S, Pagan CM, Kruse L, Dee N, Sunkin SM, Esposito L, Hawrylycz MJ, Waters J, Ng L, Smith K, Tasic B, Zhuang X, Zeng H. A high-resolution transcriptomic and spatial atlas of cell types in the whole mouse brain. Nature 2023; 624:317-332. [PMID: 38092916 PMCID: PMC10719114 DOI: 10.1038/s41586-023-06812-z] [Show More Authors] [Citation(s) in RCA: 267] [Impact Index Per Article: 133.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Accepted: 10/31/2023] [Indexed: 12/17/2023]
Abstract
The mammalian brain consists of millions to billions of cells that are organized into many cell types with specific spatial distribution patterns and structural and functional properties1-3. Here we report a comprehensive and high-resolution transcriptomic and spatial cell-type atlas for the whole adult mouse brain. The cell-type atlas was created by combining a single-cell RNA-sequencing (scRNA-seq) dataset of around 7 million cells profiled (approximately 4.0 million cells passing quality control), and a spatial transcriptomic dataset of approximately 4.3 million cells using multiplexed error-robust fluorescence in situ hybridization (MERFISH). The atlas is hierarchically organized into 4 nested levels of classification: 34 classes, 338 subclasses, 1,201 supertypes and 5,322 clusters. We present an online platform, Allen Brain Cell Atlas, to visualize the mouse whole-brain cell-type atlas along with the single-cell RNA-sequencing and MERFISH datasets. We systematically analysed the neuronal and non-neuronal cell types across the brain and identified a high degree of correspondence between transcriptomic identity and spatial specificity for each cell type. The results reveal unique features of cell-type organization in different brain regions-in particular, a dichotomy between the dorsal and ventral parts of the brain. The dorsal part contains relatively fewer yet highly divergent neuronal types, whereas the ventral part contains more numerous neuronal types that are more closely related to each other. Our study also uncovered extraordinary diversity and heterogeneity in neurotransmitter and neuropeptide expression and co-expression patterns in different cell types. Finally, we found that transcription factors are major determinants of cell-type classification and identified a combinatorial transcription factor code that defines cell types across all parts of the brain. The whole mouse brain transcriptomic and spatial cell-type atlas establishes a benchmark reference atlas and a foundational resource for integrative investigations of cellular and circuit function, development and evolution of the mammalian brain.
Collapse
Affiliation(s)
- Zizhen Yao
- Allen Institute for Brain Science, Seattle, WA, USA.
| | | | | | - Meng Zhang
- Howard Hughes Medical Institute, Department of Chemistry and Chemical Biology, Department of Physics, Harvard University, Cambridge, MA, USA
| | | | - Changkyu Lee
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Won Jung
- Howard Hughes Medical Institute, Department of Chemistry and Chemical Biology, Department of Physics, Harvard University, Cambridge, MA, USA
| | - Jeff Goldy
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | | | | | - Pamela Baker
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Eliza Barkan
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | | | | | | | | | - Daniel Carey
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | | | | | | | - Min Chen
- University of Pennsylvania, Philadelphia, PA, USA
| | | | - Jennie Close
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | - Scott Daniel
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | - Tim Dolbeare
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | | | | | - James Gee
- University of Pennsylvania, Philadelphia, PA, USA
| | | | | | - Jessica Gloe
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | - James Gray
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | | | | | - Windy Ho
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | - Mike Huang
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Madie Hupp
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Kelly Jin
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | - Kanan Lathia
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Arielle Leon
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Su Li
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Brian Long
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Zach Madigan
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | | | - Zoe Maltzer
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Naomi Martin
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Rachel McCue
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Ryan McGinty
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Nicholas Mei
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Jose Melchor
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | | | | | | | - Sven Otto
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | | | | | | | - Lane Sawyer
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | - Noah Shepard
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | - Josef Sulc
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Michael Tieu
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | - Herman Tung
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | - Shane Vance
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | - Katelyn Ward
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Boaz Levi
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | - Rob Young
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Brian Staats
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | | | - Shoaib Mufti
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | - Lauren Kruse
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Nick Dee
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | | | | | - Jack Waters
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Lydia Ng
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | | | - Xiaowei Zhuang
- Howard Hughes Medical Institute, Department of Chemistry and Chemical Biology, Department of Physics, Harvard University, Cambridge, MA, USA
| | - Hongkui Zeng
- Allen Institute for Brain Science, Seattle, WA, USA.
| |
Collapse
|
22
|
Carreiro EP, Costa AR, Antunes CM, Ernesto S, Pinto F, Rodrigues B, Burke AJ. Quercetin-1,2,3-Triazole Hybrids as Multifunctional Anti-Alzheimer's Agents. Molecules 2023; 28:7495. [PMID: 38005217 PMCID: PMC10673615 DOI: 10.3390/molecules28227495] [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: 10/09/2023] [Revised: 11/03/2023] [Accepted: 11/06/2023] [Indexed: 11/26/2023] Open
Abstract
The number of patients with Alzheimer's disease (AD) continues to rise and, despite the efforts of researchers, there are still no effective treatments for this multifaceted disease. The main objective of this work was the search for multifunctional and more effective anti-Alzheimer agents. Herein, we report the evaluation of a library of quercetin-1,2,3-triazole hybrids (I-IV) in antioxidant, hydrogen peroxide-induced oxidative stress protection, and cholinesterases (AChE and BuChE) inhibitory activities. Hybrids IIf and IVa-d showed potent in vitro inhibitory activity on eqBuChE (IC50 values between 11.2 and 65.7 μM). Hybrid IIf, the best inhibitor, was stronger than galantamine, displaying an IC50 value of 11.2 μM for eqBuChE, and is also a competitive inhibitor. Moreover, toxicity evaluation for the most promising hybrids was performed using the Artemia salina toxicity assay, showing low toxicity. Hybrids IIf, IVb, and IVd did not affect viability at 12.5 μM and also displayed a protective effect against oxidative stress induced by hydrogen peroxide in cell damage in MCF-7 cells. Hybrids IIf, IVb, and IVd act as multifunctional ligands in AD pathologies.
Collapse
Affiliation(s)
- Elisabete P. Carreiro
- Institute for Research and Advanced Training (IIFA), LAQV-REQUIMTE, University of Évora, Rua Romão Ramalho 59, 7000-671 Évora, Portugal
| | - Ana R. Costa
- Department of Medical and Health Sciences, School of Health and Human Development, University of Évora, Rua Romão Ramalho 59, 7000-671 Évora, Portugal
- Institute of Earth Sciences, Institute of Research and Advanced Training, University of Évora, 7000-671 Évora, Portugal
- Academic Clinical Center of Alentejo, C-TRAIL, Rua Romão Ramalho 59, 7000-671 Évora, Portugal
| | - Célia M. Antunes
- Department of Medical and Health Sciences, School of Health and Human Development, University of Évora, Rua Romão Ramalho 59, 7000-671 Évora, Portugal
- Institute of Earth Sciences, Institute of Research and Advanced Training, University of Évora, 7000-671 Évora, Portugal
- Academic Clinical Center of Alentejo, C-TRAIL, Rua Romão Ramalho 59, 7000-671 Évora, Portugal
| | - Sofia Ernesto
- Department of Chemistry and Biochemistry, School of Sciences and Technologies, University of Évora, Rua Romão Ramalho 59, 7000-671 Évora, Portugal
| | - Flávia Pinto
- Department of Chemistry and Biochemistry, School of Sciences and Technologies, University of Évora, Rua Romão Ramalho 59, 7000-671 Évora, Portugal
| | - Beatriz Rodrigues
- Department of Chemistry and Biochemistry, School of Sciences and Technologies, University of Évora, Rua Romão Ramalho 59, 7000-671 Évora, Portugal
| | - Anthony J. Burke
- Department of Chemistry and Biochemistry, School of Sciences and Technologies, University of Évora, Rua Romão Ramalho 59, 7000-671 Évora, Portugal
- Faculty Pharmacy, University of Coimbra, Pólo das Ciências da Saúde, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal
| |
Collapse
|
23
|
Rumpler É, Göcz B, Skrapits K, Sárvári M, Takács S, Farkas I, Póliska S, Papp M, Solymosi N, Hrabovszky E. Development of a versatile LCM-Seq method for spatial transcriptomics of fluorescently tagged cholinergic neuron populations. J Biol Chem 2023; 299:105121. [PMID: 37536628 PMCID: PMC10477691 DOI: 10.1016/j.jbc.2023.105121] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 06/29/2023] [Accepted: 07/20/2023] [Indexed: 08/05/2023] Open
Abstract
Single-cell transcriptomics are powerful tools to define neuronal cell types based on co-expressed gene clusters. Limited RNA input in these technologies necessarily compromises transcriptome coverage and accuracy of differential expression analysis. We propose that bulk RNA-Seq of neuronal pools defined by spatial position offers an alternative strategy to overcome these technical limitations. We report a laser-capture microdissection (LCM)-Seq method which allows deep transcriptome profiling of fluorescently tagged neuron populations isolated with LCM from histological sections of transgenic mice. Mild formaldehyde fixation of ZsGreen marker protein, LCM sampling of ∼300 pooled neurons, followed by RNA isolation, library preparation and RNA-Seq with methods optimized for nanogram amounts of moderately degraded RNA enabled us to detect ∼15,000 different transcripts in fluorescently labeled cholinergic neuron populations. The LCM-Seq approach showed excellent accuracy in quantitative studies, allowing us to detect 2891 transcripts expressed differentially between the spatially defined and clinically relevant cholinergic neuron populations of the dorsal caudate-putamen and medial septum. In summary, the LCM-Seq method we report in this study is a versatile, sensitive, and accurate bulk sequencing approach to study the transcriptome profile and differential gene expression of fluorescently tagged neuronal populations isolated from transgenic mice with high spatial precision.
Collapse
Affiliation(s)
- Éva Rumpler
- Laboratory of Reproductive Neurobiology, Institute of Experimental Medicine, Budapest, Hungary.
| | - Balázs Göcz
- Laboratory of Reproductive Neurobiology, Institute of Experimental Medicine, Budapest, Hungary; János Szentágothai Doctoral School of Neurosciences, Semmelweis University, Budapest, Hungary.
| | - Katalin Skrapits
- Laboratory of Reproductive Neurobiology, Institute of Experimental Medicine, Budapest, Hungary
| | - Miklós Sárvári
- Laboratory of Reproductive Neurobiology, Institute of Experimental Medicine, Budapest, Hungary
| | - Szabolcs Takács
- Laboratory of Reproductive Neurobiology, Institute of Experimental Medicine, Budapest, Hungary
| | - Imre Farkas
- Laboratory of Reproductive Neurobiology, Institute of Experimental Medicine, Budapest, Hungary
| | - Szilárd Póliska
- Faculty of Medicine, Department of Biochemistry and Molecular Biology, University of Debrecen, Debrecen, Hungary
| | - Márton Papp
- Centre for Bioinformatics, University of Veterinary Medicine, Budapest, Hungary
| | - Norbert Solymosi
- Centre for Bioinformatics, University of Veterinary Medicine, Budapest, Hungary; Department of Physics of Complex Systems, Eötvös Loránd University, Budapest, Hungary
| | - Erik Hrabovszky
- Laboratory of Reproductive Neurobiology, Institute of Experimental Medicine, Budapest, Hungary.
| |
Collapse
|
24
|
Alcaide Martin A, Mayerl S. Local Thyroid Hormone Action in Brain Development. Int J Mol Sci 2023; 24:12352. [PMID: 37569727 PMCID: PMC10418487 DOI: 10.3390/ijms241512352] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 07/28/2023] [Accepted: 07/30/2023] [Indexed: 08/13/2023] Open
Abstract
Proper brain development essentially depends on the timed availability of sufficient amounts of thyroid hormone (TH). This, in turn, necessitates a tightly regulated expression of TH signaling components such as TH transporters, deiodinases, and TH receptors in a brain region- and cell-specific manner from early developmental stages onwards. Abnormal TH levels during critical stages, as well as mutations in TH signaling components that alter the global and/or local thyroidal state, result in detrimental consequences for brain development and neurological functions that involve alterations in central neurotransmitter systems. Thus, the question as to how TH signaling is implicated in the development and maturation of different neurotransmitter and neuromodulator systems has gained increasing attention. In this review, we first summarize the current knowledge on the regulation of TH signaling components during brain development. We then present recent advances in our understanding on how altered TH signaling compromises the development of cortical glutamatergic neurons, inhibitory GABAergic interneurons, cholinergic and dopaminergic neurons. Thereby, we highlight novel mechanistic insights and point out open questions in this evolving research field.
Collapse
Affiliation(s)
| | - Steffen Mayerl
- Department of Endocrinology Diabetes & Metabolism, University Hospital Essen, University of Duisburg-Essen, Hufelandstraße 55, 45147 Essen, Germany
| |
Collapse
|
25
|
Cheng H, Wang Y, Wang Y, Ge L, Liu X, Li F. A visualized sensor based on layered double hydroxides with peroxidase-like activity for sensitive acetylcholinesterase assay. ANALYTICAL METHODS : ADVANCING METHODS AND APPLICATIONS 2023. [PMID: 37470116 DOI: 10.1039/d3ay00776f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/21/2023]
Abstract
Acetylcholinesterase (AChE) plays a crucial role in biological neurotransmission. The aberrant expression of AChE is associated with various neurodegenerative diseases. Therefore, it is of great significance to develop a simple and highly sensitive AChE analysis platform. Herein, a simple colorimetric sensor was constructed for sensitive detection of AChE based on the peroxidase-like catalytic activity of Ni/Co layered double hydroxides (Ni/Co LDHs). In this sensor, the fabricated Ni/Co LDHs possess high peroxidase-like activity, enabling rapid catalysis of o-phenylenediamine (OPD) to produce yellow oxOPD in the presence of H2O2. This peroxidase-like activity of Ni/Co LDHs was found to be effectively inhibited by the presence of AChE. It is speculated that the combination of AChE on the outer surface of Ni/Co LDHs through non-covalent interaction may cover the active sites and hinder their adsorption to the substrates, leading to the failure of OPD oxidation. As a result, the yellow color from oxOPD is related to the AChE concentration, enabling the direct AChE assay in an equipment-free manner. In addition, the fabricated Ni/Co LDHs could be modified on a paper surface to obtain a paper-based analytical device for visualized colorimetric detection of AChE. The as-proposed sensor shows high sensitivity to AChE with a detection limit down to 6.6 μU mL-1. Therefore, this naked-eye paper-based sensor is capable of on-site and real-time detection of AChE, and has outstanding application prospects in clinical diagnosis and biomedical fields.
Collapse
Affiliation(s)
- Hao Cheng
- College of Chemistry and Pharmaceutical Sciences, Qingdao Agricultural University, Qingdao 266109, P. R. China.
- College of Plant Health & Medicine, Qingdao Agricultural University, Qingdao 266109, P. R. China
| | - Yuying Wang
- College of Chemistry and Pharmaceutical Sciences, Qingdao Agricultural University, Qingdao 266109, P. R. China.
- College of Plant Health & Medicine, Qingdao Agricultural University, Qingdao 266109, P. R. China
| | - Yue Wang
- College of Chemistry and Pharmaceutical Sciences, Qingdao Agricultural University, Qingdao 266109, P. R. China.
| | - Lei Ge
- College of Chemistry and Pharmaceutical Sciences, Qingdao Agricultural University, Qingdao 266109, P. R. China.
| | - Xiaojuan Liu
- College of Chemistry and Pharmaceutical Sciences, Qingdao Agricultural University, Qingdao 266109, P. R. China.
| | - Feng Li
- College of Chemistry and Pharmaceutical Sciences, Qingdao Agricultural University, Qingdao 266109, P. R. China.
| |
Collapse
|
26
|
Świt P, Pollap A, Orzeł J. Spectroscopic Determination of Acetylcholine (ACh): A Representative Review. Top Curr Chem (Cham) 2023; 381:16. [PMID: 37169979 PMCID: PMC10175388 DOI: 10.1007/s41061-023-00426-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Accepted: 04/22/2023] [Indexed: 05/13/2023]
Abstract
Acetylcholine (ACh) is one of the most crucial neurotransmitters of the cholinergic system found in vertebrates and invertebrates and is responsible for many processes in living organisms. Disturbances in ACh transmission are closely related to dementia in Alzheimer's and Parkinson's disease. ACh in biological samples is most often determined using chromatographic techniques, radioenzymatic assays, enzyme-linked immunosorbent assay (ELISA), or potentiometric methods. An alternative way to detect and determine acetylcholine is applying spectroscopic techniques, due to low limits of detection and quantification, which is not possible with the methods mentioned above. In this review article, we described a detailed overview of different spectroscopic methods used to determine ACh with a collection of validation parameters as a perspective tool for routine analysis, especially in basic research on animal models on central nervous system. In addition, there is a discussion of examples of other biological materials from clinical and preclinical studies to give the whole spectrum of spectroscopic methods application. Descriptions of the developed chemical sensors, as well as the use of flow technology, were also presented. It is worth emphasizing the inclusion in the article of multi-component analysis referring to other neurotransmitters, as well as the description of the tested biological samples and extraction procedures. The motivation to use spectroscopic techniques to conduct this type of analysis and future perspectives in this field are briefly discussed.
Collapse
Affiliation(s)
- Paweł Świt
- Institute of Chemistry, Faculty of Science and Technology, University of Silesia in Katowice, 9 Szkolna Street, 40-006, Katowice, Poland.
| | | | - Joanna Orzeł
- Institute of Chemistry, Faculty of Science and Technology, University of Silesia in Katowice, 9 Szkolna Street, 40-006, Katowice, Poland
| |
Collapse
|
27
|
Sola E, Moyano P, Flores A, García JM, García J, Anadon MJ, Frejo MT, Pelayo A, de la Cabeza Fernandez M, Del Pino J. Cadmium-promoted thyroid hormones disruption mediates ROS, inflammation, Aβ and Tau proteins production, gliosis, spongiosis and neurodegeneration in rat basal forebrain. Chem Biol Interact 2023; 375:110428. [PMID: 36868496 DOI: 10.1016/j.cbi.2023.110428] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 02/26/2023] [Accepted: 02/28/2023] [Indexed: 03/05/2023]
Abstract
Cadmium (Cd) produces cognition decline following single and repeated treatment, although the complete mechanisms are still unrevealed. Basal forebrain (BF) cholinergic neurons innervate the cortex and hippocampus, regulating cognition. Cd single and repeated exposure induced BF cholinergic neuronal loss, partly through thyroid hormones (THs) disruption, which may cause the cognition decline observed following Cd exposure. However, the mechanisms through which THs disruption mediate this effect remain unknown. To research the possible mechanisms through which Cd-induced THs deficiency may mediate BF neurodegeneration, Wistar male rats were treated with Cd for 1- (1 mg/kg) or 28-days (0.1 mg/kg) with or without triiodothyronine (T3, 40 μg/kg/day). Cd exposure promoted neurodegeneration, spongiosis, gliosis and several mechanisms related to these alterations (increased H202, malondialdehyde, TNF-α, IL-1β, IL-6, BACE1, Aβ and phosphorylated-Tau levels, and decreased phosphorylated-AKT and phosphorylated-GSK-3β levels). T3 supplementation partially reversed the effects observed. Our results show that Cd induces several mechanisms that may be responsible for the neurodegeneration, spongiosis and gliosis observed in the rats' BF, which are partially mediated by a reduction in THs levels. These data may help to explain the mechanisms through which Cd induces BF neurodegeneration, possibly leading to the cognitive decline observed, providing new therapeutic tools to prevent and treat these damages.
Collapse
Affiliation(s)
- Emma Sola
- Department of Pharmacology and Toxicology, Veterinary School, Complutense University of Madrid, 28040, Madrid, Spain
| | - Paula Moyano
- Department of Pharmacology and Toxicology, Veterinary School, Complutense University of Madrid, 28040, Madrid, Spain.
| | - Andrea Flores
- Department of Pharmacology and Toxicology, Veterinary School, Complutense University of Madrid, 28040, Madrid, Spain
| | - José Manuel García
- Department of Pharmacology and Toxicology, Veterinary School, Complutense University of Madrid, 28040, Madrid, Spain
| | - Jimena García
- Department of Pharmacology and Toxicology, Veterinary School, Complutense University of Madrid, 28040, Madrid, Spain
| | - María José Anadon
- Department of Legal Medicine, Psychiatry and Pathology, Medicine School, Complutense University of Madrid, 28041, Madrid, Spain
| | - María Teresa Frejo
- Department of Pharmacology and Toxicology, Veterinary School, Complutense University of Madrid, 28040, Madrid, Spain
| | - Adela Pelayo
- Department of Legal Medicine, Psychiatry and Pathology, Medicine School, Complutense University of Madrid, 28041, Madrid, Spain
| | - Maria de la Cabeza Fernandez
- Department of Chemistry in Pharmaceutical Sciences, Pharmacy School, Complutense University of Madrid, 28041, Madrid, Spain
| | - Javier Del Pino
- Department of Pharmacology and Toxicology, Veterinary School, Complutense University of Madrid, 28040, Madrid, Spain.
| |
Collapse
|
28
|
Wu D, Yu N, Gao Y, Xiong R, Liu L, Lei H, Jin S, Liu J, Liu Y, Xie J, Liu E, Zhou Q, Liu Y, Li S, Wei L, Lv J, Yu H, Zeng W, Zhou Q, Xu F, Luo MH, Zhang Y, Yang Y, Wang JZ. Targeting a vulnerable septum-hippocampus cholinergic circuit in a critical time window ameliorates tau-impaired memory consolidation. Mol Neurodegener 2023; 18:23. [PMID: 37060096 PMCID: PMC10103508 DOI: 10.1186/s13024-023-00614-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 03/12/2023] [Indexed: 04/16/2023] Open
Abstract
BACKGROUND Abnormal tau accumulation and cholinergic degeneration are hallmark pathologies in the brains of patients with Alzheimer's disease (AD). However, the sensitivity of cholinergic neurons to AD-like tau accumulation and strategies to ameliorate tau-disrupted spatial memory in terms of neural circuits still remain elusive. METHODS To investigate the effect and mechanism of the cholinergic circuit in Alzheimer's disease-related hippocampal memory, overexpression of human wild-type Tau (hTau) in medial septum (MS)-hippocampus (HP) cholinergic was achieved by specifically injecting pAAV-EF1α-DIO-hTau-eGFP virus into the MS of ChAT-Cre mice. Immunostaining, behavioral analysis and optogenetic activation experiments were used to detect the effect of hTau accumulation on cholinergic neurons and the MS-CA1 cholinergic circuit. Patch-clamp recordings and in vivo local field potential recordings were used to analyze the influence of hTau on the electrical signals of cholinergic neurons and the activity of cholinergic neural circuit networks. Optogenetic activation combined with cholinergic receptor blocker was used to detect the role of cholinergic receptors in spatial memory. RESULTS In the present study, we found that cholinergic neurons with an asymmetric discharge characteristic in the MS-hippocampal CA1 pathway are vulnerable to tau accumulation. In addition to an inhibitory effect on neuronal excitability, theta synchronization between the MS and CA1 subsets was significantly disrupted during memory consolidation after overexpressing hTau in the MS. Photoactivating MS-CA1 cholinergic inputs within a critical 3 h time window during memory consolidation efficiently improved tau-induced spatial memory deficits in a theta rhythm-dependent manner. CONCLUSIONS Our study not only reveals the vulnerability of a novel MS-CA1 cholinergic circuit to AD-like tau accumulation but also provides a rhythm- and time window-dependent strategy to target the MS-CA1 cholinergic circuit, thereby rescuing tau-induced spatial cognitive functions.
Collapse
Affiliation(s)
- Dongqin Wu
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Department of Pathology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Nana Yu
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yang Gao
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Rui Xiong
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Luping Liu
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, NT, Hong Kong SAR, 999077, China
| | - Huiyang Lei
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Sen Jin
- The Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, 518055, China
| | - Jiale Liu
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yingzhou Liu
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Jiazhao Xie
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Enjie Liu
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Qiuzhi Zhou
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yanchao Liu
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Shihong Li
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Linyu Wei
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Jingru Lv
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Huilin Yu
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Wenbo Zeng
- State Key Laboratory of Virology, CAS Center for Excellence in Brain Science and Intelligence Technology (CEBSIT), Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Qiang Zhou
- State Key Laboratory of Chemical Oncogenomics, Guangdong Provincial Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, 518055, China
| | - Fuqiang Xu
- The Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, 518055, China
- Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Min-Hua Luo
- State Key Laboratory of Virology, CAS Center for Excellence in Brain Science and Intelligence Technology (CEBSIT), Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Yao Zhang
- Endocrine Department of Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430077, China.
| | - Ying Yang
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
- Co-innovation Center of Neuroregeneration, Nantong University, Nantong, 226000, China.
| | - Jian-Zhi Wang
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
- Co-innovation Center of Neuroregeneration, Nantong University, Nantong, 226000, China.
| |
Collapse
|
29
|
Tanaka A, Umatani C, Oka Y. Acetylcholine Inhibits Spontaneous Firing Activity of Terminal Nerve GnRH Neurons in Medaka. Zoolog Sci 2023; 40:151-159. [PMID: 37042694 DOI: 10.2108/zs220070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Accepted: 01/04/2023] [Indexed: 03/17/2023]
Abstract
Vertebrates generally possess hypophysiotropic and non-hypophysiotropic gonadotropin releasing hormone (GnRH) neurons. The terminal nerve (TN) GnRH neurons are known to belong to the non-hypophysiotropic neurons and have been suggested to modulate sexual behaviors. These neurons show spontaneous pacemaker firing activity and release neuropeptides GnRH and neuropeptide FF. Since the spontaneous firing activities of peptidergic neurons, including GnRH neurons, are believed to play important roles in the release of neuropeptides, understanding the regulatory mechanisms of these spontaneous firing activities is important. Here, we analyzed firing activities of the TN-GnRH neurons in medaka during application of acetylcholine (ACh), which is one of the essential neuromodulators in the brain. Whole cell patch clamp recording of TN-GnRH neurons demonstrated that ACh induces hyperpolarization and inhibits their pacemaker firing. Electrophysiological analysis using an antagonist for acetylcholine receptors and in situ hybridization analysis showed that firing of TN-GnRH neurons is inhibited via M2-type muscarinic acetylcholine receptor. These findings, taken together with literature from several other fish species (including teleosts and elasmobranchs), indicate that ACh may generally play an inhibitory role in modulating spontaneous activities of TN-GnRH neurons and thereby sexual behaviors in fish.
Collapse
Affiliation(s)
- Aiki Tanaka
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Chie Umatani
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Yoshitaka Oka
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| |
Collapse
|
30
|
Yao Z, van Velthoven CTJ, Kunst M, Zhang M, McMillen D, Lee C, Jung W, Goldy J, Abdelhak A, Baker P, Barkan E, Bertagnolli D, Campos J, Carey D, Casper T, Chakka AB, Chakrabarty R, Chavan S, Chen M, Clark M, Close J, Crichton K, Daniel S, Dolbeare T, Ellingwood L, Gee J, Glandon A, Gloe J, Gould J, Gray J, Guilford N, Guzman J, Hirschstein D, Ho W, Jin K, Kroll M, Lathia K, Leon A, Long B, Maltzer Z, Martin N, McCue R, Meyerdierks E, Nguyen TN, Pham T, Rimorin C, Ruiz A, Shapovalova N, Slaughterbeck C, Sulc J, Tieu M, Torkelson A, Tung H, Cuevas NV, Wadhwani K, Ward K, Levi B, Farrell C, Thompson CL, Mufti S, Pagan CM, Kruse L, Dee N, Sunkin SM, Esposito L, Hawrylycz MJ, Waters J, Ng L, Smith KA, Tasic B, Zhuang X, Zeng H. A high-resolution transcriptomic and spatial atlas of cell types in the whole mouse brain. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.06.531121. [PMID: 37034735 PMCID: PMC10081189 DOI: 10.1101/2023.03.06.531121] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/12/2023]
Abstract
The mammalian brain is composed of millions to billions of cells that are organized into numerous cell types with specific spatial distribution patterns and structural and functional properties. An essential step towards understanding brain function is to obtain a parts list, i.e., a catalog of cell types, of the brain. Here, we report a comprehensive and high-resolution transcriptomic and spatial cell type atlas for the whole adult mouse brain. The cell type atlas was created based on the combination of two single-cell-level, whole-brain-scale datasets: a single-cell RNA-sequencing (scRNA-seq) dataset of ~7 million cells profiled, and a spatially resolved transcriptomic dataset of ~4.3 million cells using MERFISH. The atlas is hierarchically organized into five nested levels of classification: 7 divisions, 32 classes, 306 subclasses, 1,045 supertypes and 5,200 clusters. We systematically analyzed the neuronal, non-neuronal, and immature neuronal cell types across the brain and identified a high degree of correspondence between transcriptomic identity and spatial specificity for each cell type. The results reveal unique features of cell type organization in different brain regions, in particular, a dichotomy between the dorsal and ventral parts of the brain: the dorsal part contains relatively fewer yet highly divergent neuronal types, whereas the ventral part contains more numerous neuronal types that are more closely related to each other. We also systematically characterized cell-type specific expression of neurotransmitters, neuropeptides, and transcription factors. The study uncovered extraordinary diversity and heterogeneity in neurotransmitter and neuropeptide expression and co-expression patterns in different cell types across the brain, suggesting they mediate a myriad of modes of intercellular communications. Finally, we found that transcription factors are major determinants of cell type classification in the adult mouse brain and identified a combinatorial transcription factor code that defines cell types across all parts of the brain. The whole-mouse-brain transcriptomic and spatial cell type atlas establishes a benchmark reference atlas and a foundational resource for deep and integrative investigations of cell type and circuit function, development, and evolution of the mammalian brain.
Collapse
Affiliation(s)
- Zizhen Yao
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | | | - Meng Zhang
- Howard Hughes Medical Institute, Department of Chemistry and Chemical Biology, Department of Physics, Harvard University, Cambridge, MA, USA
| | | | - Changkyu Lee
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Won Jung
- Howard Hughes Medical Institute, Department of Chemistry and Chemical Biology, Department of Physics, Harvard University, Cambridge, MA, USA
| | - Jeff Goldy
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | - Pamela Baker
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Eliza Barkan
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | | | - Daniel Carey
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | | | | | | | - Min Chen
- University of Pennsylvania, Philadelphia, PA, USA
| | | | - Jennie Close
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | - Scott Daniel
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Tim Dolbeare
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | - James Gee
- University of Pennsylvania, Philadelphia, PA, USA
| | | | - Jessica Gloe
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | - James Gray
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | | | | | - Windy Ho
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Kelly Jin
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | - Kanan Lathia
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Arielle Leon
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Brian Long
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Zoe Maltzer
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Naomi Martin
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Rachel McCue
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | | | | | | | | | | | | | - Josef Sulc
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Michael Tieu
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | - Herman Tung
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | | | - Katelyn Ward
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Boaz Levi
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | | | - Shoaib Mufti
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | - Lauren Kruse
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Nick Dee
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | | | | | - Jack Waters
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Lydia Ng
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | | | - Xiaowei Zhuang
- Howard Hughes Medical Institute, Department of Chemistry and Chemical Biology, Department of Physics, Harvard University, Cambridge, MA, USA
| | - Hongkui Zeng
- Allen Institute for Brain Science, Seattle, WA, USA
| |
Collapse
|
31
|
Ahmed NY, Knowles R, Liu L, Yan Y, Li X, Schumann U, Wang Y, Sontani Y, Reynolds N, Natoli R, Wen J, Del Pino I, Mi D, Dehorter N. Developmental deficits of MGE-derived interneurons in the Cntnap2 knockout mouse model of autism spectrum disorder. Front Cell Dev Biol 2023; 11:1112062. [PMID: 36819097 PMCID: PMC9930104 DOI: 10.3389/fcell.2023.1112062] [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: 11/30/2022] [Accepted: 01/17/2023] [Indexed: 02/04/2023] Open
Abstract
Interneurons are fundamental cells for maintaining the excitation-inhibition balance in the brain in health and disease. While interneurons have been shown to play a key role in the pathophysiology of autism spectrum disorder (ASD) in adult mice, little is known about how their maturation is altered in the developing striatum in ASD. Here, we aimed to track striatal developing interneurons and elucidate the molecular and physiological alterations in the Cntnap2 knockout mouse model. Using Stereo-seq and single-cell RNA sequencing data, we first characterized the pattern of expression of Cntnap2 in the adult brain and at embryonic stages in the medial ganglionic eminence (MGE), a transitory structure producing most cortical and striatal interneurons. We found that Cntnap2 is enriched in the striatum, compared to the cortex, particularly in the developing striatal cholinergic interneurons. We then revealed enhanced MGE-derived cell proliferation, followed by increased cell loss during the canonical window of developmental cell death in the Cntnap2 knockout mice. We uncovered specific cellular and molecular alterations in the developing Lhx6-expressing cholinergic interneurons of the striatum, which impacts interneuron firing properties during the first postnatal week. Overall, our work unveils some of the mechanisms underlying the shift in the developmental trajectory of striatal interneurons which greatly contribute to the ASD pathogenesis.
Collapse
Affiliation(s)
- Noorya Yasmin Ahmed
- The Australian National University, The John Curtin School of Medical Research, Canberra, ACT, Australia
| | - Rhys Knowles
- The Australian National University, The John Curtin School of Medical Research, Canberra, ACT, Australia
| | - Lixinyu Liu
- The Australian National University, The John Curtin School of Medical Research, Canberra, ACT, Australia
| | - Yiming Yan
- Tsinghua-Peking Center for Life Sciences, IDG/McGovern Institute for Brain Research, School of Life Sciences, Tsinghua University, Beijing, China
| | - Xiaohan Li
- Tsinghua-Peking Center for Life Sciences, IDG/McGovern Institute for Brain Research, School of Life Sciences, Tsinghua University, Beijing, China
| | - Ulrike Schumann
- The Australian National University, The John Curtin School of Medical Research, Canberra, ACT, Australia
| | - Yumeng Wang
- The Australian National University, The John Curtin School of Medical Research, Canberra, ACT, Australia
| | - Yovina Sontani
- The Australian National University, The John Curtin School of Medical Research, Canberra, ACT, Australia
| | - Nathan Reynolds
- The Australian National University, The John Curtin School of Medical Research, Canberra, ACT, Australia
| | - Riccardo Natoli
- The Australian National University, The John Curtin School of Medical Research, Canberra, ACT, Australia
| | - Jiayu Wen
- The Australian National University, The John Curtin School of Medical Research, Canberra, ACT, Australia
| | - Isabel Del Pino
- Institute of Neurosciences, Spanish National Research Council (CSIC), Sant Joan d’Alacant, Spain
| | - Da Mi
- Tsinghua-Peking Center for Life Sciences, IDG/McGovern Institute for Brain Research, School of Life Sciences, Tsinghua University, Beijing, China
| | - Nathalie Dehorter
- The Australian National University, The John Curtin School of Medical Research, Canberra, ACT, Australia,*Correspondence: Nathalie Dehorter,
| |
Collapse
|
32
|
Khan SA, Akhtar MJ, Gogoi U, Meenakshi DU, Das A. An Overview of 1,2,3-triazole-Containing Hybrids and Their Potential Anticholinesterase Activities. Pharmaceuticals (Basel) 2023; 16:179. [PMID: 37259329 PMCID: PMC9961747 DOI: 10.3390/ph16020179] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 01/18/2023] [Accepted: 01/22/2023] [Indexed: 07/30/2023] Open
Abstract
Acetylcholine (ACh) neurotransmitter of the cholinergic system in the brain is involved in learning, memory, stress responses, and cognitive functioning. It is hydrolyzed into choline and acetic acid by two key cholinesterase enzymes, viz., acetylcholinesterase (AChE) and butyrylcholinesterase (BuChE). A loss or degeneration of cholinergic neurons that leads to a reduction in ACh levels is considered a significant contributing factor in the development of neurodegenerative diseases (NDs) such as Alzheimer's disease (AD). Numerous studies have shown that cholinesterase inhibitors can raise the level of ACh and, therefore, enhance people's quality of life, and, at the very least, it can temporarily lessen the symptoms of NDs. 1,2,3-triazole, a five-membered heterocyclic ring, is a privileged moiety, that is, a central scaffold, and is capable of interacting with a variety of receptors and enzymes to exhibit a broad range of important biological activities. Recently, it has been clubbed with other pharmacophoric fragments/molecules in hope of obtaining potent and selective AChE and/or BuChE inhibitors. The present updated review succinctly summarizes the different synthetic strategies used to synthesize the 1,2,3-triazole moiety. It also highlights the anticholinesterase potential of various 1,2,3-triazole di/trihybrids reported in the past seven years (2015-2022), including a rationale for hybridization and with an emphasis on their structural features for the development and optimization of cholinesterase inhibitors to treat NDs.
Collapse
Affiliation(s)
- Shah Alam Khan
- College of Pharmacy, National University of Science and Technology, Muscat 130, Oman
| | | | - Urvashee Gogoi
- Department of Pharmaceutical Sciences, Dibrugarh University, Dibrugarh 786004, India
| | | | - Aparoop Das
- Department of Pharmaceutical Sciences, Dibrugarh University, Dibrugarh 786004, India
| |
Collapse
|
33
|
Gamage R, Zaborszky L, Münch G, Gyengesi E. Evaluation of eGFP expression in the ChAT-eGFP transgenic mouse brain. BMC Neurosci 2023; 24:4. [PMID: 36650430 PMCID: PMC9847127 DOI: 10.1186/s12868-023-00773-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Accepted: 01/02/2023] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND A historically definitive marker for cholinergic neurons is choline acetyltransferase (ChAT), a synthesizing enzyme for acetylcholine, (ACh), which can be found in high concentrations in cholinergic neurons, both in the central and peripheral nervous systems. ChAT, is produced in the body of the neuron, transported to the nerve terminal (where its concentration is highest), and catalyzes the transfer of an acetyl group from the coenzyme acetyl-CoA to choline, yielding ACh. The creation of bacterial artificial chromosome (BAC) transgenic mice that express promoter-specific fluorescent reporter proteins (green fluorescent protein-[GFP]) provided an enormous advantage for neuroscience. Both in vivo and in vitro experimental methods benefited from the transgenic visualization of cholinergic neurons. Mice were created by adding a BAC clone into the ChAT locus, in which enhanced GFP (eGFP) is inserted into exon 3 at the ChAT initiation codon, robustly and supposedly selectively expressing eGFP in all cholinergic neurons and fibers in the central and peripheral nervous systems as well as in non-neuronal cells. METHODS This project systematically compared the exact distribution of the ChAT-eGFP expressing neurons in the brain with the expression of ChAT by immunohistochemistry using mapping and also made comparisons with in situ hybridization (ISH). RESULTS We qualitatively described the distribution of ChAT-eGFP neurons in the mouse brain by comparing it with the distribution of immunoreactive neurons and ISH data, paying special attention to areas where the expression did not overlap, such as the cortex, striatum, thalamus and hypothalamus. We found a complete overlap between the transgenic expression of eGFP and the immunohistochemical staining in the areas of the cholinergic basal forebrain. However, in the cortex and hippocampus, we found small neurons that were only labeled with the antibody and not expressed eGFP or vice versa. Most importantly, we found no transgenic expression of eGFP in the lateral dorsal, ventral and dorsomedial tegmental nuclei cholinergic cells. CONCLUSION While the majority of the forebrain ChAT expression was aligned in the transgenic animals with immunohistochemistry, other areas of interest, such as the brainstem should be considered before choosing this particular transgenic mouse line.
Collapse
Affiliation(s)
- Rashmi Gamage
- grid.1029.a0000 0000 9939 5719Pharmacology Unit, Group of Pharmacology, School of Medicine, Western Sydney University, Penrith, NSW 2751 Australia
| | - Laszlo Zaborszky
- grid.430387.b0000 0004 1936 8796Center for Molecular and Behavioral Neuroscience, Rutgers The State University of New Jersey, Newark, NJ 07102 USA
| | - Gerald Münch
- grid.1029.a0000 0000 9939 5719Pharmacology Unit, Group of Pharmacology, School of Medicine, Western Sydney University, Penrith, NSW 2751 Australia
| | - Erika Gyengesi
- grid.1029.a0000 0000 9939 5719Pharmacology Unit, Group of Pharmacology, School of Medicine, Western Sydney University, Penrith, NSW 2751 Australia
| |
Collapse
|
34
|
Lysenkov SP, Muzhenya DV, Tuguz AR, Urakova TU, Shumilov DS, Thakushinov IA, Thakushinov RA, Tatarkova EA, Urakova DM. Cholinergic deficiency in the cholinergic system as a pathogenetic link in the formation of various syndromes in COVID-19. CHINESE J PHYSIOL 2023; 66:1-13. [PMID: 36814151 DOI: 10.4103/cjop.cjop-d-22-00072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
According to recent data, several mechanisms of viral invasion of the central nervous system (CNS) have been proposed, one of which is both direct penetration of the virus through afferent nerve fibers and damage to the endothelium of cerebral vessels. It has been proven that the SARS-CoV-2 virus affects pathologically not only the human cardiorespiratory system but is also associated with a wide range of neurological diseases, cerebrovascular accidents, and neuromuscular pathologies. However, the observed post-COVID symptom complex in patients, manifested in the form of headache, "fog in the head," high temperature, muscle weakness, lowering blood pressure, does it make us think about the pathophysiological mechanisms that contribute to the development of this clinical picture? One possible explanation is a disruption in the signaling of the acetylcholine system (AChS) in the body. Viral invasions, and in particular COVID-19, can negatively affect the work of the AChS, disrupting its coordination activities. Therefore, the main goal of this literature review is to analyze the information and substantiate the possible mechanisms for the occurrence of post-COVID syndrome in people who have had COVID-19 from the standpoint of AChS dysfunctions.
Collapse
Affiliation(s)
- Sergey Petrovich Lysenkov
- FSBEI HE "Maikop State Technological University", Medical Institute, Maikop, Republic of Adygeya, Russia
| | | | - Aminat Ramazanovna Tuguz
- FSBEI HE "Adyghe State University", Immunogenetic Laboratory of the Research Institute of Complex Problems, Maikop, Republic of Adygeya, Russia
| | - Tamara Ur'evna Urakova
- FSBEI HE "Maikop State Technological University", Medical Institute, Maikop, Republic of Adygeya, Russia
| | - Dmitriy Sergeevich Shumilov
- FSBEI HE "Adyghe State University", Immunogenetic Laboratory of the Research Institute of Complex Problems, Maikop, Republic of Adygeya, Russia
| | | | | | - Elena Anatolevna Tatarkova
- FSBEI HE "Adyghe State University", Immunogenetic Laboratory of the Research Institute of Complex Problems, Maikop, Republic of Adygeya, Russia
| | - Diana Muratovna Urakova
- FSBEI HE "Maikop State Technological University", Medical Institute, Maikop, Republic of Adygeya, Russia
| |
Collapse
|
35
|
Shostak DM, Constantin S, Flannery J, Wray S. Acetylcholine regulation of GnRH neuronal activity: A circuit in the medial septum. Front Endocrinol (Lausanne) 2023; 14:1147554. [PMID: 36950690 PMCID: PMC10025473 DOI: 10.3389/fendo.2023.1147554] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Accepted: 02/22/2023] [Indexed: 03/08/2023] Open
Abstract
In vertebrates, gonadotropin-releasing hormone (GnRH)-secreting neurons control fertility by regulating gonadotrophs in the anterior pituitary. While it is known that acetylcholine (ACh) influences GnRH secretion, whether the effect is direct or indirect, and the specific ACh receptor (AChR) subtype(s) involved remain unclear. Here, we determined 1) whether ACh can modulate GnRH cellular activity and 2) a source of ACh afferents contacting GnRH neurons. Calcium imaging was used to assay GnRH neuronal activity. With GABAergic and glutamatergic transmission blocked, subtype-specific AChR agonists and antagonists were applied to identify direct regulation of GnRH neurons. ACh and nicotine caused a rise in calcium that declined gradually back to baseline after 5-6 min. This response was mimicked by an alpha3-specific agonist. In contrast, muscarine inhibited GnRH calcium oscillations, and blocking M2 and M4 together prevented this inhibition. Labeling for choline acetyltransferase (ChAT) and GnRH revealed ChAT fibers contacting GnRH neurons, primarily in the medial septum (MS), and in greater number in females than males. ChAT positive cells in the MS are known to express p75NGFRs. Labeling for p75NGFR, ChAT and GnRH indicated that ChAT fibers contacting GnRH cells originate from cholinergic cells within these same rostral areas. Together, these results indicate that cholinergic cells in septal areas can directly regulate GnRH neurons.
Collapse
|
36
|
Matsushima T, Miura M, Patzke N, Toji N, Wada K, Ogura Y, Homma KJ, Sgadò P, Vallortigara G. Fetal blockade of nicotinic acetylcholine transmission causes autism-like impairment of biological motion preference in the neonatal chick. Cereb Cortex Commun 2022; 3:tgac041. [PMID: 37674673 PMCID: PMC10478028 DOI: 10.1093/texcom/tgac041] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2022] [Revised: 10/16/2022] [Accepted: 10/25/2022] [Indexed: 09/08/2023] Open
Abstract
Several environmental chemicals are suspected risk factors for autism spectrum disorder (ASD), including valproic acid (VPA) and pesticides acting on nicotinic acetylcholine receptors (nAChRs), if administered during pregnancy. However, their target processes in fetal neuro-development are unknown. We report that the injection of VPA into the fetus impaired imprinting to an artificial object in neonatal chicks, while a predisposed preference for biological motion (BM) remained intact. Blockade of nAChRs acted oppositely, sparing imprinting and impairing BM preference. Beside ketamine and tubocurarine, significant effects of imidacloprid (a neonicotinoid insecticide) appeared at a dose ≤1 ppm. In accord with the behavioral dissociations, VPA enhanced histone acetylation in the primary cell culture of fetal telencephalon, whereas ketamine did not. VPA reduced the brain weight and the ratio of NeuN-positive cells (matured neurons) in the telencephalon of hatchlings, whereas ketamine/tubocurarine did not. Despite the distinct underlying mechanisms, both VPA and nAChR blockade similarly impaired imprinting to biological image composed of point-light animations. Furthermore, both impairments were abolished by postnatal bumetanide treatment, suggesting a common pathology underlying the social attachment malformation. Neurotransmission via nAChR is thus critical for the early social bond formation, which is hindered by ambient neonicotinoids through impaired visual predispositions for animate objects.
Collapse
Affiliation(s)
- Toshiya Matsushima
- Department of Biology, Faculty of Science, Hokkaido University, Sapporo 060-0810, Japan
- Faculty of Pharmaceutical Science, Health Science University of Hokkaido, Tobetsu 061-0293, Japan
- Center for Mind/Brain Sciences, University of Trento, Rovereto 38068, Italy
| | - Momoko Miura
- Department of Biology, Faculty of Science, Hokkaido University, Sapporo 060-0810, Japan
- Faculty of Pharmaceutical Science, Health Science University of Hokkaido, Tobetsu 061-0293, Japan
| | - Nina Patzke
- Department of Biology, Faculty of Science, Hokkaido University, Sapporo 060-0810, Japan
- Health and Medical University, Potsdam 14471, Germany
| | - Noriyuki Toji
- Department of Biology, Faculty of Science, Hokkaido University, Sapporo 060-0810, Japan
| | - Kazuhiro Wada
- Department of Biology, Faculty of Science, Hokkaido University, Sapporo 060-0810, Japan
| | - Yukiko Ogura
- Graduate School of Information Science and Technology, The University of Tokyo, Tokyo 113-8654, Japan
- Japan Science and Technology Agency, PRESTO, Kawaguchi 332-0012, Japan
| | - Koichi J Homma
- Department of Molecular Biology, Faculty of Pharmaceutical Sciences, Teikyo University, Tokyo 173-8605, Japan
| | - Paola Sgadò
- Center for Mind/Brain Sciences, University of Trento, Rovereto 38068, Italy
| | | |
Collapse
|
37
|
Recent advances in enzyme inhibition based-electrochemical biosensors for pharmaceutical and environmental analysis. Talanta 2022. [DOI: 10.1016/j.talanta.2022.124092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
38
|
Delbono O, Wang Z, Messi ML. Brainstem noradrenergic neurons: Identifying a hub at the intersection of cognition, motility, and skeletal muscle regulation. Acta Physiol (Oxf) 2022; 236:e13887. [PMID: 36073023 PMCID: PMC9588743 DOI: 10.1111/apha.13887] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 08/31/2022] [Accepted: 09/05/2022] [Indexed: 01/29/2023]
Abstract
Brainstem noradrenergic neuron clusters form a node integrating efferents projecting to distinct areas such as those regulating cognition and skeletal muscle structure and function, and receive dissimilar afferents through established circuits to coordinate organismal responses to internal and environmental challenges. Genetic lineage tracing shows the remarkable heterogeneity of brainstem noradrenergic neurons, which may explain their varied functions. They project to the locus coeruleus, the primary source of noradrenaline in the brain, which supports learning and cognition. They also project to pre-ganglionic neurons, which lie within the spinal cord and form synapses onto post-ganglionic neurons. The synapse between descending brainstem noradrenergic neurons and pre-ganglionic spinal neurons, and these in turn with post-ganglionic noradrenergic neurons located at the paravertebral sympathetic ganglia, support an anatomical hierarchy that regulates skeletal muscle innervation, neuromuscular transmission, and muscle trophism. Whether any noradrenergic neuron subpopulation is more susceptible to damaged protein deposit and death with ageing and neurodegeneration is a relevant question that answer will help us to detect neurodegeneration at an early stage, establish prognosis, and anticipate disease progression. Loss of muscle mass and strength with ageing, termed sarcopenia, may predict impaired cognition with ageing and neurodegeneration and establish an early time to start interventions aimed at reducing central noradrenergic neurons hyperactivity. Complex multidisciplinary approaches, including genetic tracing, specific circuit labelling, optogenetics and chemogenetics, electrophysiology, and single-cell transcriptomics and proteomics, are required to test this hypothesis pre-clinical.
Collapse
Affiliation(s)
- Osvaldo Delbono
- Department of Internal MedicineSection on Gerontology and Geriatric Medicine. Wake Forest University School of MedicineWinston‐SalemNorth CarolinaUSA
| | - Zhong‐Min Wang
- Department of Internal MedicineSection on Gerontology and Geriatric Medicine. Wake Forest University School of MedicineWinston‐SalemNorth CarolinaUSA
| | - María Laura Messi
- Department of Internal MedicineSection on Gerontology and Geriatric Medicine. Wake Forest University School of MedicineWinston‐SalemNorth CarolinaUSA
| |
Collapse
|
39
|
Metabolic and Cellular Compartments of Acetyl-CoA in the Healthy and Diseased Brain. Int J Mol Sci 2022; 23:ijms231710073. [PMID: 36077475 PMCID: PMC9456256 DOI: 10.3390/ijms231710073] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 08/29/2022] [Accepted: 08/31/2022] [Indexed: 11/25/2022] Open
Abstract
The human brain is characterised by the most diverse morphological, metabolic and functional structure among all body tissues. This is due to the existence of diverse neurons secreting various neurotransmitters and mutually modulating their own activity through thousands of pre- and postsynaptic interconnections in each neuron. Astroglial, microglial and oligodendroglial cells and neurons reciprocally regulate the metabolism of key energy substrates, thereby exerting several neuroprotective, neurotoxic and regulatory effects on neuronal viability and neurotransmitter functions. Maintenance of the pool of mitochondrial acetyl-CoA derived from glycolytic glucose metabolism is a key factor for neuronal survival. Thus, acetyl-CoA is regarded as a direct energy precursor through the TCA cycle and respiratory chain, thereby affecting brain cell viability. It is also used for hundreds of acetylation reactions, including N-acetyl aspartate synthesis in neuronal mitochondria, acetylcholine synthesis in cholinergic neurons, as well as divergent acetylations of several proteins, peptides, histones and low-molecular-weight species in all cellular compartments. Therefore, acetyl-CoA should be considered as the central point of metabolism maintaining equilibrium between anabolic and catabolic pathways in the brain. This review presents data supporting this thesis.
Collapse
|
40
|
Ugbaja RN, Akinhanmi TF, Onunkwor BO, Ugwor EI, James AS, Babalola AA, Babayemi DO, Ezenandu EO, Ugbaja VC. Flavonoid-rich fractions of C. volubile and V. amygdalina alleviates arsenic-induced neurotoxicity by improving neurosignaling and antioxidant capacity in rats' brain. BRAIN DISORDERS 2022. [DOI: 10.1016/j.dscb.2022.100050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
|
41
|
Benevento M, Hökfelt T, Harkany T. Ontogenetic rules for the molecular diversification of hypothalamic neurons. Nat Rev Neurosci 2022; 23:611-627. [PMID: 35906427 DOI: 10.1038/s41583-022-00615-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/14/2022] [Indexed: 11/09/2022]
Abstract
The hypothalamus is an evolutionarily conserved endocrine interface that, among other roles, links central homeostatic control to adaptive bodily responses by releasing hormones and neuropeptides from its many neuronal subtypes. In its preoptic, anterior, tuberal and mammillary subdivisions, a kaleidoscope of magnocellular and parvocellular neuroendocrine command neurons, local-circuit neurons, and neurons that project to extrahypothalamic areas are intermingled in partially overlapping patches of nuclei. Molecular fingerprinting has produced data of unprecedented mass and depth to distinguish and even to predict the synaptic and endocrine competences, connectivity and stimulus selectivity of many neuronal modalities. These new insights support eminent studies from the past century but challenge others on the molecular rules that shape the developmental segregation of hypothalamic neuronal subtypes and their use of morphogenic cues for terminal differentiation. Here, we integrate single-cell RNA sequencing studies with those of mouse genetics and endocrinology to describe key stages of hypothalamus development, including local neurogenesis, the direct terminal differentiation of glutamatergic neurons, transition cascades for GABAergic and GABAergic cell-derived dopamine cells, waves of local neuronal migration, and sequential enrichment in neuropeptides and hormones. We particularly emphasize how transcription factors determine neuronal identity and, consequently, circuit architecture, and whether their deviations triggered by environmental factors and hormones provoke neuroendocrine illnesses.
Collapse
Affiliation(s)
- Marco Benevento
- Department of Molecular Neurosciences, Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - Tomas Hökfelt
- Department of Neuroscience, Biomedicum 7D, Karolinska Institutet, Solna, Sweden
| | - Tibor Harkany
- Department of Molecular Neurosciences, Center for Brain Research, Medical University of Vienna, Vienna, Austria. .,Department of Neuroscience, Biomedicum 7D, Karolinska Institutet, Solna, Sweden.
| |
Collapse
|
42
|
Gao X, Zhang M, Lyu M, Lin S, Luo X, You W, Ke C. Role of Bmal1 in mediating the cholinergic system to regulate the behavioral rhythm of nocturnal marine molluscs. Comput Struct Biotechnol J 2022; 20:2815-2830. [PMID: 35765646 PMCID: PMC9189711 DOI: 10.1016/j.csbj.2022.05.038] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Revised: 05/17/2022] [Accepted: 05/19/2022] [Indexed: 11/18/2022] Open
Abstract
The circadian differential expression of AchE was identified using TMT quantitative proteomics; It was found that the Ach concentration and the expression levels of AchE and Bmal1 exhibit circadian cosine rhythm; The full-length sequences of AchE and nAchR were obtained by cloning technique and made available for phylogenetic analysis; The movement distance and duration of abalone increased after the injection of neostigmine methylsulfate as the AchE inhibitor; Bmal1 as the core circadian clock gene was proven to bind to AchE and nAchR, thereby regulating the movement behavior of abalone.
The circadian rhythm is one of the most general and important rhythms in biological organisms. In this study, continuous 24-h video recordings showed that the cumulative movement distance and duration of the abalone, Haliotis discus hannai, reached their maximum values between 20:00–00:00, but both were significantly lower between 08:00–12:00 than at any other time of day or night (P < 0.05). To investigate the causes of these diel differences in abalone movement behavior, their cerebral ganglia were harvested at 00:00 (group D) and 12:00 (group L) to screen for differentially expressed proteins using tandem mass tagging (TMT) quantitative proteomics. Seventy-five significantly different proteins were identified in group D vs. group L. The differences in acetylcholinesterase (AchE) expression levels between day- and nighttime and the key role in the cholinergic nervous system received particular attention during the investigation. A cosine rhythm analysis found that the concentration of acetylcholine (Ach) and the expression levels of AchE tended to be low during the day and high at night, and high during the day and low at night, respectively. However, the rhythmicity of the diel expression levels of acetylcholine receptor (nAchR) appeared to be insignificant (P > 0.05). Following the injection of three different concentrations of neostigmine methylsulfate, as an AchE inhibitor, the concentration of Ach in the hemolymph, and the expression levels of nAchR in the cerebral ganglia increased significantly (P < 0.05). Four hours after drug injection, the cumulative movement distance and duration of abalones were significantly higher than those in the uninjected control group, and the group injected with saline (P < 0.05). The expression levels of the core diurnal clock Bmal1 over a 24-h period also tended to be high during the day and low at night. First, a co-immunoprecipitation assay demonstrated the binding between Bmal1 and AchE or nAchR. A dual-luciferase gene test and electrophoretic mobility shift assay showed that Bmal1 bound to the promoter regions of AchE and nAchR. Twenty-four hours after silencing the Bmal1 gene, the expression levels of AchE and nAchR decreased significantly compared to those of the dsEGFP and PBS control groups, further showing that Bmal1 mediates the cholinergic system to regulate the behavioral rhythm of abalone. These findings shed light on the endocrine mechanism regulating the rhythmic behavior of abalone, and provide a reference for understanding the life history adaptation strategies of nocturnal organisms and the proliferation and protection of bottom dwelling economically important organisms.
Collapse
Affiliation(s)
- Xiaolong Gao
- State Key Laboratory of Marine Environmental Science, College of Ocean and Earth Sciences, Xiamen University, Xiamen 361102, China
- Fujian Key Laboratory of Genetics and Breeding of Marine Organisms, Xiamen University, Xiamen, China
| | - Mo Zhang
- State Key Laboratory of Marine Environmental Science, College of Ocean and Earth Sciences, Xiamen University, Xiamen 361102, China
- Fujian Key Laboratory of Genetics and Breeding of Marine Organisms, Xiamen University, Xiamen, China
| | - Mingxin Lyu
- State Key Laboratory of Marine Environmental Science, College of Ocean and Earth Sciences, Xiamen University, Xiamen 361102, China
- Fujian Key Laboratory of Genetics and Breeding of Marine Organisms, Xiamen University, Xiamen, China
| | - Shihui Lin
- State Key Laboratory of Marine Environmental Science, College of Ocean and Earth Sciences, Xiamen University, Xiamen 361102, China
- Fujian Key Laboratory of Genetics and Breeding of Marine Organisms, Xiamen University, Xiamen, China
| | - Xuan Luo
- State Key Laboratory of Marine Environmental Science, College of Ocean and Earth Sciences, Xiamen University, Xiamen 361102, China
- Fujian Key Laboratory of Genetics and Breeding of Marine Organisms, Xiamen University, Xiamen, China
| | - Weiwei You
- State Key Laboratory of Marine Environmental Science, College of Ocean and Earth Sciences, Xiamen University, Xiamen 361102, China
- Fujian Key Laboratory of Genetics and Breeding of Marine Organisms, Xiamen University, Xiamen, China
| | - Caihuan Ke
- State Key Laboratory of Marine Environmental Science, College of Ocean and Earth Sciences, Xiamen University, Xiamen 361102, China
- Fujian Key Laboratory of Genetics and Breeding of Marine Organisms, Xiamen University, Xiamen, China
- Corresponding author.
| |
Collapse
|
43
|
Onikanni AS, Lawal B, Oyinloye BE, Mostafa-Hedeab G, Alorabi M, Cavalu S, Olusola AO, Wang CH, Batiha GES. Therapeutic efficacy of Clompanus pubescens leaves fractions via downregulation of neuronal cholinesterases/Na+-K+ATPase/IL-1 β, and improving the neurocognitive and antioxidants status of streptozotocin-induced diabetic rats. Biomed Pharmacother 2022; 148:112730. [DOI: 10.1016/j.biopha.2022.112730] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 02/02/2022] [Accepted: 02/15/2022] [Indexed: 12/29/2022] Open
|
44
|
Caubit X, Gubellini P, Roubertoux PL, Carlier M, Molitor J, Chabbert D, Metwaly M, Salin P, Fatmi A, Belaidouni Y, Brosse L, Kerkerian-Le Goff L, Fasano L. Targeted Tshz3 deletion in corticostriatal circuit components segregates core autistic behaviors. Transl Psychiatry 2022; 12:106. [PMID: 35292625 PMCID: PMC8924251 DOI: 10.1038/s41398-022-01865-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 02/18/2022] [Accepted: 02/24/2022] [Indexed: 01/15/2023] Open
Abstract
We previously linked TSHZ3 haploinsufficiency to autism spectrum disorder (ASD) and showed that embryonic or postnatal Tshz3 deletion in mice results in behavioral traits relevant to the two core domains of ASD, namely social interaction deficits and repetitive behaviors. Here, we provide evidence that cortical projection neurons (CPNs) and striatal cholinergic interneurons (SCINs) are two main and complementary players in the TSHZ3-linked ASD syndrome. In the cerebral cortex, TSHZ3 is expressed in CPNs and in a proportion of GABAergic interneurons, but not in cholinergic interneurons or glial cells. In the striatum, TSHZ3 is expressed in all SCINs, while its expression is absent or partial in the other main brain cholinergic systems. We then characterized two new conditional knockout (cKO) models generated by crossing Tshz3flox/flox with Emx1-Cre (Emx1-cKO) or Chat-Cre (Chat-cKO) mice to decipher the respective role of CPNs and SCINs. Emx1-cKO mice show altered excitatory synaptic transmission onto CPNs and impaired plasticity at corticostriatal synapses, with neither cortical neuron loss nor abnormal layer distribution. These animals present social interaction deficits but no repetitive patterns of behavior. Chat-cKO mice exhibit no loss of SCINs but changes in the electrophysiological properties of these interneurons, associated with repetitive patterns of behavior without social interaction deficits. Therefore, dysfunction in either CPNs or SCINs segregates with a distinct ASD behavioral trait. These findings provide novel insights onto the implication of the corticostriatal circuitry in ASD by revealing an unexpected neuronal dichotomy in the biological background of the two core behavioral domains of this disorder.
Collapse
Affiliation(s)
- Xavier Caubit
- grid.462081.90000 0004 0598 4854Aix-Marseille Univ, CNRS, IBDM, UMR7288 Marseille, France
| | - Paolo Gubellini
- grid.462081.90000 0004 0598 4854Aix-Marseille Univ, CNRS, IBDM, UMR7288 Marseille, France
| | - Pierre L. Roubertoux
- grid.5399.60000 0001 2176 4817Aix-Marseille Univ, INSERM, MMG, UMR1251 Marseille, France
| | - Michèle Carlier
- grid.463724.00000 0004 0385 2989Aix-Marseille Univ, CNRS, LPC, UMR7290 Marseille, France
| | - Jordan Molitor
- grid.462081.90000 0004 0598 4854Aix-Marseille Univ, CNRS, IBDM, UMR7288 Marseille, France
| | - Dorian Chabbert
- grid.462081.90000 0004 0598 4854Aix-Marseille Univ, CNRS, IBDM, UMR7288 Marseille, France
| | - Mehdi Metwaly
- grid.462081.90000 0004 0598 4854Aix-Marseille Univ, CNRS, IBDM, UMR7288 Marseille, France
| | - Pascal Salin
- grid.462081.90000 0004 0598 4854Aix-Marseille Univ, CNRS, IBDM, UMR7288 Marseille, France
| | - Ahmed Fatmi
- grid.462081.90000 0004 0598 4854Aix-Marseille Univ, CNRS, IBDM, UMR7288 Marseille, France
| | - Yasmine Belaidouni
- grid.462081.90000 0004 0598 4854Aix-Marseille Univ, CNRS, IBDM, UMR7288 Marseille, France
| | - Lucie Brosse
- grid.462081.90000 0004 0598 4854Aix-Marseille Univ, CNRS, IBDM, UMR7288 Marseille, France
| | | | - Laurent Fasano
- Aix-Marseille Univ, CNRS, IBDM, UMR7288, Marseille, France.
| |
Collapse
|
45
|
Tang Y, Zong H, Kwon H, Qiu Y, Pessin JB, Wu L, Buddo KA, Boykov I, Schmidt CA, Lin CT, Neufer PD, Schwartz GJ, Kurland IJ, Pessin J. TIGAR deficiency enhances skeletal muscle thermogenesis by increasing neuromuscular junction cholinergic signaling. eLife 2022; 11:73360. [PMID: 35254259 PMCID: PMC8947760 DOI: 10.7554/elife.73360] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Accepted: 03/02/2022] [Indexed: 12/03/2022] Open
Abstract
Cholinergic and sympathetic counter-regulatory networks control numerous physiological functions, including learning/memory/cognition, stress responsiveness, blood pressure, heart rate, and energy balance. As neurons primarily utilize glucose as their primary metabolic energy source, we generated mice with increased glycolysis in cholinergic neurons by specific deletion of the fructose-2,6-phosphatase protein TIGAR. Steady-state and stable isotope flux analyses demonstrated increased rates of glycolysis, acetyl-CoA production, acetylcholine levels, and density of neuromuscular synaptic junction clusters with enhanced acetylcholine release. The increase in cholinergic signaling reduced blood pressure and heart rate with a remarkable resistance to cold-induced hypothermia. These data directly demonstrate that increased cholinergic signaling through the modulation of glycolysis has several metabolic benefits particularly to increase energy expenditure and heat production upon cold exposure.
Collapse
Affiliation(s)
- Yan Tang
- Department of Medicine, Albert Einstein College of Medicine, Bronx, United States
| | - Haihong Zong
- Department of Medicine, Albert Einstein College of Medicine, Bronx, United States
| | - Hyokjoon Kwon
- Department of Medicine, Albert Einstein College of Medicine, Bronx, United States
| | - Yunping Qiu
- Department of Medicine, Albert Einstein College of Medicine, Bronx, United States
| | - Jacob B Pessin
- Department of Medicine, Albert Einstein College of Medicine, Bronx, United States
| | - Licheng Wu
- Department of Medicine, Albert Einstein College of Medicine, Bronx, United States
| | - Katherine A Buddo
- Department of Physiology, East Carolina University, Greenville, United States
| | - Ilya Boykov
- Department of Physiology, East Carolina University, Greenville, United States
| | - Cameron A Schmidt
- Department of Physiology, East Carolina University, Greenville, United States
| | - Chien-Te Lin
- Department of Physiology, East Carolina University, Greenville, United States
| | - P Darrell Neufer
- Department of Physiology, East Carolina University, Greenville, United States
| | - Gary J Schwartz
- Department of Medicine, Albert Einstein College of Medicine, Bronx, United States
| | - Irwin J Kurland
- Department of Medicine, Albert Einstein College of Medicine, Bronx, United States
| | - Jeffrey Pessin
- Department of Medicine, Albert Einstein College of Medicine, Bronx, United States
| |
Collapse
|
46
|
Robson DN, Li JM. A dynamical systems view of neuroethology: Uncovering stateful computation in natural behaviors. Curr Opin Neurobiol 2022; 73:102517. [PMID: 35217311 DOI: 10.1016/j.conb.2022.01.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Revised: 01/06/2022] [Accepted: 01/11/2022] [Indexed: 11/03/2022]
Abstract
State-dependent computation is key to cognition in both biological and artificial systems. Alan Turing recognized the power of stateful computation when he created the Turing machine with theoretically infinite computational capacity in 1936. Independently, by 1950, ethologists such as Tinbergen and Lorenz also began to implicitly embed rudimentary forms of state-dependent computation to create qualitative models of internal drives and naturally occurring animal behaviors. Here, we reformulate core ethological concepts in explicitly dynamical systems terms for stateful computation. We examine, based on a wealth of recent neural data collected during complex innate behaviors across species, the neural dynamics that determine the temporal structure of internal states. We will also discuss the degree to which the brain can be hierarchically partitioned into nested dynamical systems and the need for a multi-dimensional state-space model of the neuromodulatory system that underlies motivational and affective states.
Collapse
Affiliation(s)
- Drew N Robson
- Max Planck Institute for Biological Cybernetics, Tuebingen, Germany.
| | - Jennifer M Li
- Max Planck Institute for Biological Cybernetics, Tuebingen, Germany.
| |
Collapse
|
47
|
Cholinergic blockade of neuroinflammation – from tissue to RNA regulators. Neuronal Signal 2022; 6:NS20210035. [PMID: 35211331 PMCID: PMC8837817 DOI: 10.1042/ns20210035] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 01/06/2022] [Accepted: 01/06/2022] [Indexed: 11/17/2022] Open
Abstract
Inflammatory stimuli and consequent pro-inflammatory immune responses may facilitate neurodegeneration and threaten survival following pathogen infection or trauma, but potential controllers preventing these risks are incompletely understood. Here, we argue that small RNA regulators of acetylcholine (ACh) signaling, including microRNAs (miRs) and transfer RNA fragments (tRFs) may tilt the balance between innate and adaptive immunity, avoid chronic inflammation and prevent the neuroinflammation-mediated exacerbation of many neurological diseases. While the restrictive permeability of the blood–brain barrier (BBB) protects the brain from peripheral immune events, this barrier can be disrupted by inflammation and is weakened with age. The consequently dysregulated balance between pro- and anti-inflammatory processes may modify the immune activities of brain microglia, astrocytes, perivascular macrophages, oligodendrocytes and dendritic cells, leading to neuronal damage. Notably, the vagus nerve mediates the peripheral cholinergic anti-inflammatory reflex and underlines the consistent control of body–brain inflammation by pro-inflammatory cytokines, which affect cholinergic functions; therefore, the disruption of this reflex can exacerbate cognitive impairments such as attention deficits and delirium. RNA regulators can contribute to re-balancing the cholinergic network and avoiding its chronic deterioration, and their activities may differ between men and women and/or wear off with age. This can lead to hypersensitivity of aged patients to inflammation and higher risks of neuroinflammation-driven cholinergic impairments such as delirium and dementia following COVID-19 infection. The age- and sex-driven differences in post-transcriptional RNA regulators of cholinergic elements may hence indicate new personalized therapeutic options for neuroinflammatory diseases.
Collapse
|
48
|
Knowles R, Dehorter N, Ellender T. From Progenitors to Progeny: Shaping Striatal Circuit Development and Function. J Neurosci 2021; 41:9483-9502. [PMID: 34789560 PMCID: PMC8612473 DOI: 10.1523/jneurosci.0620-21.2021] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 09/17/2021] [Accepted: 09/27/2021] [Indexed: 12/29/2022] Open
Abstract
Understanding how neurons of the striatum are formed and integrate into complex synaptic circuits is essential to provide insight into striatal function in health and disease. In this review, we summarize our current understanding of the development of striatal neurons and associated circuits with a focus on their embryonic origin. Specifically, we address the role of distinct types of embryonic progenitors, found in the proliferative zones of the ganglionic eminences in the ventral telencephalon, in the generation of diverse striatal interneurons and projection neurons. Indeed, recent evidence would suggest that embryonic progenitor origin dictates key characteristics of postnatal cells, including their neurochemical content, their location within striatum, and their long-range synaptic inputs. We also integrate recent observations regarding embryonic progenitors in cortical and other regions and discuss how this might inform future research on the ganglionic eminences. Last, we examine how embryonic progenitor dysfunction can alter striatal formation, as exemplified in Huntington's disease and autism spectrum disorder, and how increased understanding of embryonic progenitors can have significant implications for future research directions and the development of improved therapeutic options.SIGNIFICANCE STATEMENT This review highlights recently defined novel roles for embryonic progenitor cells in shaping the functional properties of both projection neurons and interneurons of the striatum. It outlines the developmental mechanisms that guide neuronal development from progenitors in the embryonic ganglionic eminences to progeny in the striatum. Where questions remain open, we integrate observations from cortex and other regions to present possible avenues for future research. Last, we provide a progenitor-centric perspective onto both Huntington's disease and autism spectrum disorder. We suggest that future investigations and manipulations of embryonic progenitor cells in both research and clinical settings will likely require careful consideration of their great intrinsic diversity and neurogenic potential.
Collapse
Affiliation(s)
- Rhys Knowles
- The John Curtin School of Medical Research, The Australian National University, Canberra 2601, Australian Capital Territory, Australia
| | - Nathalie Dehorter
- The John Curtin School of Medical Research, The Australian National University, Canberra 2601, Australian Capital Territory, Australia
| | - Tommas Ellender
- Department of Pharmacology, University of Oxford, Oxford OX1 3QT, United Kingdom
- Department of Biomedical Sciences, University of Antwerp, 2610 Wilrijk, Belgium
| |
Collapse
|
49
|
Maruyama T, Mano A, Ishii T, Kakinuma Y, Kaneda M. P2X 2 receptors supply extracellular choline as a substrate for acetylcholine synthesis. FEBS Open Bio 2021; 12:250-257. [PMID: 34787962 PMCID: PMC8727932 DOI: 10.1002/2211-5463.13332] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 11/03/2021] [Accepted: 11/16/2021] [Indexed: 01/14/2023] Open
Abstract
Acetylcholine (ACh), an excitatory neurotransmitter, is biosynthesized from choline in cholinergic neurons. Import from the extracellular space to the intracellular environment through the high-affinity choline transporter is currently regarded to be the only source of choline for ACh synthesis. We recently demonstrated that the P2X2 receptor, through which large cations permeate, functions as an alternative pathway for choline transport in the mouse retina. In the present study, we investigated whether choline entering cells through P2X2 receptors is used for ACh synthesis using a recombinant system. When P2X2 receptors expressed on HEK293 cell lines were stimulated with ATP, intracellular ACh concentrations increased. These results suggest that P2X2 receptors function in a novel pathway that supplies choline for ACh synthesis.
Collapse
Affiliation(s)
- Takuma Maruyama
- Department of Physiology, Nippon Medical School, Tokyo, Japan
| | - Asuka Mano
- Department of Physiology, Nippon Medical School, Tokyo, Japan
| | - Toshiyuki Ishii
- Department of Physiology, Nippon Medical School, Tokyo, Japan
| | | | - Makoto Kaneda
- Department of Physiology, Nippon Medical School, Tokyo, Japan
| |
Collapse
|
50
|
Sumien N, Cunningham JT, Davis DL, Engelland R, Fadeyibi O, Farmer GE, Mabry S, Mensah-Kane P, Trinh OTP, Vann PH, Wilson EN, Cunningham RL. Neurodegenerative Disease: Roles for Sex, Hormones, and Oxidative Stress. Endocrinology 2021; 162:6360925. [PMID: 34467976 PMCID: PMC8462383 DOI: 10.1210/endocr/bqab185] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Indexed: 02/08/2023]
Abstract
Neurodegenerative diseases cause severe impairments in cognitive and motor function. With an increasing aging population and the onset of these diseases between 50 and 70 years, the consequences are bound to be devastating. While age and longevity are the main risk factors for neurodegenerative diseases, sex is also an important risk factor. The characteristic of sex is multifaceted, encompassing sex chromosome complement, sex hormones (estrogens and androgens), and sex hormone receptors. Sex hormone receptors can induce various signaling cascades, ranging from genomic transcription to intracellular signaling pathways that are dependent on the health of the cell. Oxidative stress, associated with aging, can impact the health of the cell. Sex hormones can be neuroprotective under low oxidative stress conditions but not in high oxidative stress conditions. An understudied sex hormone receptor that can induce activation of oxidative stress signaling is the membrane androgen receptor (mAR). mAR can mediate nicotinamide adenine dinucleotide-phosphate (NADPH) oxidase (NOX)-generated oxidative stress that is associated with several neurodegenerative diseases, such as Alzheimer disease. Further complicating this is that aging can alter sex hormone signaling. Prior to menopause, women experience more estrogens than androgens. During menopause, this sex hormone profile switches in women due to the dramatic ovarian loss of 17β-estradiol with maintained ovarian androgen (testosterone, androstenedione) production. Indeed, aging men have higher estrogens than aging women due to aromatization of androgens to estrogens. Therefore, higher activation of mAR-NOX signaling could occur in menopausal women compared with aged men, mediating the observed sex differences. Understanding of these signaling cascades could provide therapeutic targets for neurodegenerative diseases.
Collapse
Affiliation(s)
- Nathalie Sumien
- Department of Pharmacology & Neuroscience, Center for Healthy Aging, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | - J Thomas Cunningham
- Department of Physiology & Anatomy, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | - Delaney L Davis
- Department of Pharmacology & Neuroscience, Center for Healthy Aging, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | - Rachel Engelland
- Department of Pharmaceutical Sciences, School of Pharmacy, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | - Oluwadarasimi Fadeyibi
- Department of Pharmaceutical Sciences, School of Pharmacy, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | - George E Farmer
- Department of Physiology & Anatomy, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | - Steve Mabry
- Department of Pharmaceutical Sciences, School of Pharmacy, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | - Paapa Mensah-Kane
- Department of Pharmacology & Neuroscience, Center for Healthy Aging, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | - Oanh T P Trinh
- Department of Pharmacology & Neuroscience, Center for Healthy Aging, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | - Philip H Vann
- Department of Pharmacology & Neuroscience, Center for Healthy Aging, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | - E Nicole Wilson
- Department of Pharmaceutical Sciences, School of Pharmacy, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | - Rebecca L Cunningham
- Department of Pharmaceutical Sciences, School of Pharmacy, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
- Correspondence: Rebecca L. Cunningham, PhD, Department of Pharmaceutical Sciences, University of North Texas Health Science Center, 3400 Camp Bowie Boulevard, Fort Worth, TX, USA, 76107-2699.
| |
Collapse
|