1
|
Zhang L, Dong W, Li J, Gao S, Sheng H, Kong Q, Guan F, Zhang L. C1ql3 knockout affects microglia activation, neuronal integrity, and spontaneous behavior in Wistar rats. Animal Model Exp Med 2025; 8:332-343. [PMID: 38379452 PMCID: PMC11871103 DOI: 10.1002/ame2.12383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 12/27/2023] [Indexed: 02/22/2024] Open
Abstract
BACKGROUND C1QL3 is widely expressed in the brain and is specifically produced by a subset of excitatory neurons. However, its function is still not clear. We established C1ql3-deficient rats to investigate the role of C1QL3 in the brain. METHODS C1ql3 knockout (KO) rats were generated using CRISPR/Cas9. C1ql3 KO was determined by polymerase chain reaction (PCR), DNA sequencing, and western blotting. Microglia morphology and cytokine expression with or without lipopolysaccharide (LPS) stimulus were analyzed using immunohistochemistry and real-time PCR. The brain structure changes in KO rats were examined using magnetic resonance imaging. Neuronal architecture alteration was analyzed by performing Golgi staining. Behavior was evaluated using the open field test, Morris water maze test, and Y maze test. RESULTS C1ql3 KO significantly increased the number of ramified microglia and decreased the number of hypertrophic microglia, whereas C1ql3 KO did not influence the expression of pro-inflammatory factors and anti-inflammatory factors except IL-10. C1ql3 KO brains had more amoeboid microglia types and higher Arg-1 expression compared with the WT rats after LPS stimulation. The brain weights and HPC sizes of C1ql3 KO rats did not differ from WT rats. C1ql3 KO damaged neuronal integrity including neuron dendritic arbors and spine density. C1ql3 KO rats demonstrated an increase in spontaneous activity and an impairment in short working memory. CONCLUSIONS C1ql3 KO not only interrupts the neuronal integrity but also affects the microglial activation, resulting in hyperactive behavior and impaired short memory in rats, which highlights the role of C1QL3 in the regulation of structure and function of both neuronal and microglial cells.
Collapse
Affiliation(s)
- Li Zhang
- Beijing Engineering Research Center for Experimental Animal Models of Human Diseases, Institute of Laboratory Animal SciencePeking Union Medicine College, Chinese Academy of Medical SciencesBeijingChina
| | - Wei Dong
- Key Laboratory of Human Disease Comparative Medicine, National Health Commission of China (NHC)Institute of Laboratory Animal Science, Peking Union Medicine College, Chinese Academy of Medical SciencesBeijingChina
| | - Jingwen Li
- Key Laboratory of Human Disease Comparative Medicine, National Health Commission of China (NHC)Institute of Laboratory Animal Science, Peking Union Medicine College, Chinese Academy of Medical SciencesBeijingChina
| | - Shan Gao
- Key Laboratory of Human Disease Comparative Medicine, National Health Commission of China (NHC)Institute of Laboratory Animal Science, Peking Union Medicine College, Chinese Academy of Medical SciencesBeijingChina
| | - Hanxuan Sheng
- Key Laboratory of Human Disease Comparative Medicine, National Health Commission of China (NHC)Institute of Laboratory Animal Science, Peking Union Medicine College, Chinese Academy of Medical SciencesBeijingChina
| | - Qi Kong
- Key Laboratory of Human Disease Comparative Medicine, National Health Commission of China (NHC)Institute of Laboratory Animal Science, Peking Union Medicine College, Chinese Academy of Medical SciencesBeijingChina
| | - Feifei Guan
- Beijing Engineering Research Center for Experimental Animal Models of Human Diseases, Institute of Laboratory Animal SciencePeking Union Medicine College, Chinese Academy of Medical SciencesBeijingChina
| | - Lianfeng Zhang
- Key Laboratory of Human Disease Comparative Medicine, National Health Commission of China (NHC)Institute of Laboratory Animal Science, Peking Union Medicine College, Chinese Academy of Medical SciencesBeijingChina
- Neuroscience Center, Chinese Academy of Medical SciencesBeijingChina
| |
Collapse
|
2
|
Chang CY, Dai W, Hu SSJ. Cannabidiol enhances socially transmitted food preference: a role of acetylcholine in the mouse basal forebrain. Psychopharmacology (Berl) 2025; 242:247-269. [PMID: 39158618 DOI: 10.1007/s00213-024-06670-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2024] [Accepted: 08/05/2024] [Indexed: 08/20/2024]
Abstract
RATIONALE AND OBJECTIVE Rodents acquire food information from their conspecifics and display a preference for the conspecifics' consumed food. This social learning of food information from others promotes the survival of a species, and it is introduced as the socially transmitted food preference (STFP) task. The cholinergic system in the basal forebrain plays a role in the acquisition of STFP. Cannabidiol (CBD), one of the most abundant phytocannabinoids, exerts its therapeutic potential for cognitive deficits through versatile mechanisms of action, including its interaction with the cholinergic system. We hypothesize a positive relationship between CBD and STFP because acetylcholine (ACh) is involved in STFP, and CBD increases the ACh levels in the basal forebrain. MATERIALS AND METHODS Male C57BL/6J mice were trained to acquire the STFP task. We examined whether CBD affects STFP memory by administering CBD (20 mg/kg, i.p.) before the STFP social training. The involvement of cholinergic system in CBD's effect on STFP was examined by knockdown of brain acetylcholinesterase (AChE), applying a nonselective muscarinic antagonist SCO (3 mg/kg, i.p.) before CBD treatment, and measuring the basal forebrain ACh levels in the CBD-treated mice. RESULTS We first showed that CBD enhanced STFP memory. Knockdown of brain AChE also enhanced STFP memory, which mimicked CBD's effect on STFP. SCO blocked CBD's memory-enhancing effect on STFP. Our most significant finding is that the basal forebrain ACh levels in the CBD-treated mice, but not their control counterparts, were positively correlated with mice's STFP memory performance. CONCLUSION This study indicates that CBD enhances STFP memory in mice. Specifically, those which respond to CBD by increasing the muscarinic-mediated ACh signaling perform better in their STFP memory.
Collapse
Affiliation(s)
- Chih-Yu Chang
- Cannabinoid Signaling Laboratory, Department of Psychology, National Cheng Kung University, 1 University Rd, Tainan, 70101, Taiwan
| | - Wen Dai
- Cannabinoid Signaling Laboratory, Department of Psychology, National Cheng Kung University, 1 University Rd, Tainan, 70101, Taiwan
| | - Sherry Shu-Jung Hu
- Cannabinoid Signaling Laboratory, Department of Psychology, National Cheng Kung University, 1 University Rd, Tainan, 70101, Taiwan.
| |
Collapse
|
3
|
Altunay ZM, Biswas J, Cheung HW, Pijewski RS, Papile LE, Akinlaja YO, Tang A, Kresic LC, Schouw AD, Ugrak MV, Caro K, Peña Palomino PA, Ressl S, Nishiyama A, Crocker SJ, Martinelli DC. C1ql1 expression in oligodendrocyte progenitor cells promotes oligodendrocyte differentiation. FEBS J 2025; 292:52-74. [PMID: 39257292 PMCID: PMC11706710 DOI: 10.1111/febs.17256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 07/16/2024] [Accepted: 08/14/2024] [Indexed: 09/12/2024]
Abstract
Myelinating oligodendrocytes arise from the stepwise differentiation of oligodendrocyte progenitor cells (OPCs). Approximately 5% of all adult brain cells are OPCs. Why would a mature brain need such a large number of OPCs? New myelination is possibly required for higher-order functions such as cognition and learning. Additionally, this pool of OPCs represents a source of new oligodendrocytes to replace those lost during injury, inflammation, or in diseases such as multiple sclerosis (MS). How OPCs are instructed to differentiate into oligodendrocytes is poorly understood, and for reasons presently unclear, resident pools of OPCs are progressively less utilized in MS. The complement component 1, q subcomponent-like (C1QL) protein family has been studied for their functions at neuron-neuron synapses, but we show that OPCs express C1ql1. We created OPC-specific conditional knockout mice and show that C1QL1 deficiency reduces the differentiation of OPCs into oligodendrocytes and reduces myelin production during both development and recovery from cuprizone-induced demyelination. In vivo over-expression of C1QL1 causes the opposite phenotype: increased oligodendrocyte density and myelination during recovery from demyelination. We further used primary cultured OPCs to show that C1QL1 levels can bidirectionally regulate the extent of OPC differentiation in vitro. Our results suggest that C1QL1 may initiate a previously unrecognized signaling pathway to promote differentiation of OPCs into oligodendrocytes. This study has relevance for possible novel therapies for demyelinating diseases and may illuminate a previously undescribed mechanism to regulate the function of myelination in cognition and learning.
Collapse
Affiliation(s)
- Zeynep M. Altunay
- Department of Neuroscience, University of Connecticut Health, Farmington, CT, USA
| | - Joyshree Biswas
- Department of Neuroscience, University of Connecticut Health, Farmington, CT, USA
| | - Hiu W. Cheung
- Department of Neuroscience, University of Connecticut Health, Farmington, CT, USA
| | - Robert S. Pijewski
- Department of Neuroscience, University of Connecticut Health, Farmington, CT, USA
- Department of Biology, Anna Maria College, Paxton, MA, USA
| | - Lucille E. Papile
- Department of Neuroscience, University of Connecticut Health, Farmington, CT, USA
| | - Yetunde O. Akinlaja
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, CT, USA
| | - Andrew Tang
- Department of Neuroscience, University of Connecticut Health, Farmington, CT, USA
| | - Lyndsay C. Kresic
- Department of Neuroscience, University of Connecticut Health, Farmington, CT, USA
| | - Alexander D. Schouw
- Department of Neuroscience, University of Connecticut Health, Farmington, CT, USA
| | - Maksym V. Ugrak
- Department of Neuroscience, University of Connecticut Health, Farmington, CT, USA
| | - Keaven Caro
- Department of Neuroscience, University of Connecticut Health, Farmington, CT, USA
| | | | - Susanne Ressl
- Department of Neuroscience, The University of Texas at Austin, Austin, TX, USA
| | - Akiko Nishiyama
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, CT, USA
- The Connecticut Institute for the Brain and Cognitive Sciences (IBACS), Storrs, CT, USA
| | - Stephen J. Crocker
- Department of Neuroscience, University of Connecticut Health, Farmington, CT, USA
- The Connecticut Institute for the Brain and Cognitive Sciences (IBACS), Storrs, CT, USA
| | - David C. Martinelli
- Department of Neuroscience, University of Connecticut Health, Farmington, CT, USA
- The Connecticut Institute for the Brain and Cognitive Sciences (IBACS), Storrs, CT, USA
| |
Collapse
|
4
|
周 彤, 吴 祎, 胡 蒙, 唐 欣, 朱 平, 杜 立, 吴 春. [Application of optogenetic technology in the research on olfactory bulb neural projection from advanced brain regions to regulate olfactory signal processing]. SHENG WU YI XUE GONG CHENG XUE ZA ZHI = JOURNAL OF BIOMEDICAL ENGINEERING = SHENGWU YIXUE GONGCHENGXUE ZAZHI 2024; 41:1265-1270. [PMID: 40000218 PMCID: PMC11955376 DOI: 10.7507/1001-5515.202404009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 10/15/2024] [Indexed: 02/27/2025]
Abstract
Olfactory bulb is a critical component in encoding and processing olfactory signals, characterized by its intricate neural projections and networks dedicated to this function. It has been found that descending neural projections from the olfactory cortex and other advanced brain regions can modulate the excitability of olfactory bulb output neurons in the olfactory bulb, either directly or indirectly, which can further influence olfactory discrimination, learning, and other abilities. In recent years, advancements in optogenetic technology have facilitated extensive application of neuron manipulation for studying neural circuits, thereby greatly accelerating research into olfactory mechanisms. This review summarizes the latest research progress on the regulatory effects of neural projections from the olfactory cortex, basal forebrain, raphe nucleus, and locus coeruleus on olfactory bulb function. Furthermore, the important role that photogenetic technology plays in olfactory mechanism research is evaluated. Finally, the existing problems and future development trends in current research are preliminarily proposed and explained. This review aims to provide new insights into the mechanisms underlying olfactory neural regulation as well as applications of optogenetic technology, which are crucial for advancing the research on olfactory mechanism and the application of optogenetic technology.
Collapse
Affiliation(s)
- 彤 周
- 西安交通大学 基础医学院 生物物理学系 医学工程研究所(西安 710061)Institute of Medical Engineering, Department of Biophysics, School of Basic Medicine, Xi'an Jiaotong University, Xi'an 710061, P. R. China
| | - 祎凡 吴
- 西安交通大学 基础医学院 生物物理学系 医学工程研究所(西安 710061)Institute of Medical Engineering, Department of Biophysics, School of Basic Medicine, Xi'an Jiaotong University, Xi'an 710061, P. R. China
| | - 蒙 胡
- 西安交通大学 基础医学院 生物物理学系 医学工程研究所(西安 710061)Institute of Medical Engineering, Department of Biophysics, School of Basic Medicine, Xi'an Jiaotong University, Xi'an 710061, P. R. China
| | - 欣 唐
- 西安交通大学 基础医学院 生物物理学系 医学工程研究所(西安 710061)Institute of Medical Engineering, Department of Biophysics, School of Basic Medicine, Xi'an Jiaotong University, Xi'an 710061, P. R. China
| | - 平 朱
- 西安交通大学 基础医学院 生物物理学系 医学工程研究所(西安 710061)Institute of Medical Engineering, Department of Biophysics, School of Basic Medicine, Xi'an Jiaotong University, Xi'an 710061, P. R. China
| | - 立萍 杜
- 西安交通大学 基础医学院 生物物理学系 医学工程研究所(西安 710061)Institute of Medical Engineering, Department of Biophysics, School of Basic Medicine, Xi'an Jiaotong University, Xi'an 710061, P. R. China
| | - 春生 吴
- 西安交通大学 基础医学院 生物物理学系 医学工程研究所(西安 710061)Institute of Medical Engineering, Department of Biophysics, School of Basic Medicine, Xi'an Jiaotong University, Xi'an 710061, P. R. China
| |
Collapse
|
5
|
Tang Y, Wang C, Li Q, Liu G, Song D, Quan Z, Yan Y, Qing H. Neural Network Excitation/Inhibition: A Key to Empathy and Empathy Impairment. Neuroscientist 2024; 30:644-665. [PMID: 38347700 DOI: 10.1177/10738584231223119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2024]
Abstract
Empathy is an ability to fully understand and feel the mental states of others. We emphasize that empathy is elicited by the transmission of pain, fear, and sensory information. In clinical studies, impaired empathy has been observed in most psychiatric conditions. However, the precise impairment mechanism of the network systems on the pathogenesis of empathy impairment in psychiatric disorders is still unclear. Multiple lines of evidence suggest that disturbances in the excitatory/inhibitory balance in neurologic disorders are key to empathetic impairment in psychiatric disorders. Therefore, we here describe the roles played by the anterior cingulate cortex- and medial prefrontal cortex-dependent neural circuits and their impairments in psychiatric disorders, including anxiety, depression, and autism. In addition, we review recent studies on the role of microglia in neural network excitation/inhibition imbalance, which contributes to a better understanding of the neural network excitation/inhibition imbalance and may open up innovative psychiatric therapies.
Collapse
Affiliation(s)
- Yuanhong Tang
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Science, Beijing Institute of Technology, Beijing, China
| | - Chunjian Wang
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Science, Beijing Institute of Technology, Beijing, China
| | - Qingquan Li
- Department of Cardiology, The First Hospital of Hebei Medical University, Shijiazhuang, China
| | - Gang Liu
- Department of Cardiology, The First Hospital of Hebei Medical University, Shijiazhuang, China
| | - Da Song
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Science, Beijing Institute of Technology, Beijing, China
| | - Zhenzhen Quan
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Science, Beijing Institute of Technology, Beijing, China
| | - Yan Yan
- Department of Cardiology, The First Hospital of Hebei Medical University, Shijiazhuang, China
| | - Hong Qing
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Science, Beijing Institute of Technology, Beijing, China
- Department of Biology, Shenzhen MSU-BIT University, Shenzhen, China
| |
Collapse
|
6
|
Ma C, Shen B, Chen L, Yang G. Impacts of circadian disruptions on behavioral rhythms in mice. FASEB J 2024; 38:e70183. [PMID: 39570004 DOI: 10.1096/fj.202401536r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 10/11/2024] [Accepted: 11/04/2024] [Indexed: 11/22/2024]
Abstract
Circadian rhythms are fundamental biological processes that recur approximately every 24 h, with the sleep-wake cycle or circadian behavior being a well-known example. In the field of chronobiology, mice serve as valuable model animals for studying mammalian circadian rhythms due to their genetic similarity to humans and the availability of various genetic tools for manipulation. Monitoring locomotor activity in mice provides valuable insights into the impact of various conditions or disturbances on circadian behavior. In this review, we summarized the effects of disturbance of biological rhythms on circadian behavior in mice. External factors, especially light exert a significant impact on circadian behavior. Additionally, feeding timing, food composition, ambient temperature, and physical exercise contribute to variations in the behavior of the mouse. Internal factors, including gender, age, genetic background, and clock gene mutation or deletion, are effective as well. Understanding the effects of circadian disturbances on murine behavior is essential for gaining insights into the underlying mechanisms of circadian regulation and developing potential therapeutic interventions for circadian-related disorders in humans.
Collapse
Affiliation(s)
- Changxiao Ma
- Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
- School of Bioengineering, Dalian University of Technology, Dalian, China
| | - Bingyi Shen
- School of Bioengineering, Dalian University of Technology, Dalian, China
| | - Lihong Chen
- Health Science Center, East China Normal University, Shanghai, China
| | - Guangrui Yang
- Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
- School of Clinical Medicine, Shanghai University of Medicine & Health Sciences, Shanghai, China
| |
Collapse
|
7
|
A brain circuit that cements the memory of socially learnt food preferences. Nature 2024:10.1038/d41586-024-03012-1. [PMID: 39294284 DOI: 10.1038/d41586-024-03012-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/20/2024]
|
8
|
Liu Z, Sun W, Ng YH, Dong H, Quake SR, Südhof TC. The cortical amygdala consolidates a socially transmitted long-term memory. Nature 2024; 632:366-374. [PMID: 38961294 PMCID: PMC11306109 DOI: 10.1038/s41586-024-07632-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Accepted: 05/30/2024] [Indexed: 07/05/2024]
Abstract
Social communication guides decision-making, which is essential for survival. Social transmission of food preference (STFP) is an ecologically relevant memory paradigm in which an animal learns a desirable food odour from another animal in a social context, creating a long-term memory1,2. How food-preference memory is acquired, consolidated and stored is unclear. Here we show that the posteromedial nucleus of the cortical amygdala (COApm) serves as a computational centre in long-term STFP memory consolidation by integrating social and sensory olfactory inputs. Blocking synaptic signalling by the COApm-based circuit selectively abolished STFP memory consolidation without impairing memory acquisition, storage or recall. COApm-mediated STFP memory consolidation depends on synaptic inputs from the accessory olfactory bulb and on synaptic outputs to the anterior olfactory nucleus. STFP memory consolidation requires protein synthesis, suggesting a gene-expression mechanism. Deep single-cell and spatially resolved transcriptomics revealed robust but distinct gene-expression signatures induced by STFP memory formation in the COApm that are consistent with synapse restructuring. Our data thus define a neural circuit for the consolidation of a socially communicated long-term memory, thereby mechanistically distinguishing protein-synthesis-dependent memory consolidation from memory acquisition, storage or retrieval.
Collapse
Affiliation(s)
- Zhihui Liu
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA, USA.
- Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA, USA.
| | - Wenfei Sun
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA, USA
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| | - Yi Han Ng
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA, USA
| | - Hua Dong
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Stephen R Quake
- Department of Bioengineering, Stanford University, Stanford, CA, USA.
- Chan Zuckerberg Initiative, Redwood City, CA, USA.
| | - Thomas C Südhof
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA, USA.
- Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
9
|
Matúš D, Lopez JM, Sando RC, Südhof TC. Essential Role of Latrophilin-1 Adhesion GPCR Nanoclusters in Inhibitory Synapses. J Neurosci 2024; 44:e1978232024. [PMID: 38684366 PMCID: PMC11154861 DOI: 10.1523/jneurosci.1978-23.2024] [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: 10/19/2023] [Revised: 02/03/2024] [Accepted: 02/20/2024] [Indexed: 05/02/2024] Open
Abstract
Latrophilin-1 (Lphn1, aka CIRL1 and CL1; gene symbol Adgrl1) is an adhesion GPCR that has been implicated in excitatory synaptic transmission as a candidate receptor for α-latrotoxin. Here we analyzed conditional knock-in/knock-out mice for Lphn1 that contain an extracellular myc epitope tag. Mice of both sexes were used in all experiments. Surprisingly, we found that Lphn1 is localized in cultured neurons to synaptic nanoclusters that are present in both excitatory and inhibitory synapses. Conditional deletion of Lphn1 in cultured neurons failed to elicit a detectable impairment in excitatory synapses but produced a decrease in inhibitory synapse numbers and synaptic transmission that was most pronounced for synapses close to the neuronal soma. No changes in axonal or dendritic outgrowth or branching were observed. Our data indicate that Lphn1 is among the few postsynaptic adhesion molecules that are present in both excitatory and inhibitory synapses and that Lphn1 by itself is not essential for excitatory synaptic transmission but is required for some inhibitory synaptic connections.
Collapse
Affiliation(s)
- Daniel Matúš
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, California 94305
| | - Jaybree M Lopez
- Department of Pharmacology, Vanderbilt Brain Institute, Vanderbilt University, Nashville, Tennessee 37240
| | - Richard C Sando
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, California 94305
- Department of Pharmacology, Vanderbilt Brain Institute, Vanderbilt University, Nashville, Tennessee 37240
| | - Thomas C Südhof
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, California 94305
- Hughes Medical Institute, Stanford University School of Medicine, Stanford, California 94305
| |
Collapse
|
10
|
Brosnan A, Knapska E. Cheerful tails: Delving into positive emotional contagion. Neurosci Biobehav Rev 2024; 161:105674. [PMID: 38614451 DOI: 10.1016/j.neubiorev.2024.105674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Revised: 03/29/2024] [Accepted: 04/10/2024] [Indexed: 04/15/2024]
Abstract
This review delves into the phenomenon of positive emotional contagion (PEC) in rodents, an area that remains relatively understudied compared to the well-explored realm of negative emotions such as fear or pain. Rodents exhibit clear preferences for individuals expressing positive emotions over neutral counterparts, underscoring the importance of detecting and responding to positive emotional signals from others. We thoroughly examine the adaptive function of PEC, highlighting its pivotal role in social learning and environmental adaptation. The developmental aspect of the ability to interpret positive emotions is explored, intricately linked to maternal care and social interactions, with oxytocin playing a central role in these processes. We discuss the potential involvement of the reward system and draw attention to persisting gaps in our understanding of the neural mechanisms governing PEC. Presenting a comprehensive overview of the existing literature, we focus on food-related protocols such as the Social Transmission of Food Preferences paradigm and tickling behaviour. Our review emphasizes the pressing need for further research to address lingering questions and advance our comprehension of positive emotional contagion.
Collapse
Affiliation(s)
- Adam Brosnan
- Laboratory of Emotions Neurobiology, BRAINCITY - Centre of Excellence for Neural Plasticity and Brain Disorders, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Ewelina Knapska
- Laboratory of Emotions Neurobiology, BRAINCITY - Centre of Excellence for Neural Plasticity and Brain Disorders, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland.
| |
Collapse
|
11
|
Saegusa C, Kakegawa W, Miura E, Aimi T, Mogi S, Harada T, Yamashita T, Yuzaki M, Fujioka M. Brain-Specific Angiogenesis Inhibitor 3 Is Expressed in the Cochlea and Is Necessary for Hearing Function in Mice. Int J Mol Sci 2023; 24:17092. [PMID: 38069416 PMCID: PMC10707444 DOI: 10.3390/ijms242317092] [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: 11/13/2023] [Revised: 11/27/2023] [Accepted: 11/29/2023] [Indexed: 12/18/2023] Open
Abstract
Mammalian auditory hair cells transduce sound-evoked traveling waves in the cochlea into nerve stimuli, which are essential for hearing function. Pillar cells located between the inner and outer hair cells are involved in the formation of the tunnel of Corti, which incorporates outer-hair-cell-driven fluid oscillation and basilar membrane movement, leading to the fine-tuned frequency-specific perception of sounds by the inner hair cells. However, the detailed molecular mechanism underlying the development and maintenance of pillar cells remains to be elucidated. In this study, we examined the expression and function of brain-specific angiogenesis inhibitor 3 (Bai3), an adhesion G-protein-coupled receptor, in the cochlea. We found that Bai3 was expressed in hair cells in neonatal mice and pillar cells in adult mice, and, interestingly, Bai3 knockout mice revealed the abnormal formation of pillar cells, with the elevation of the hearing threshold in a frequency-dependent manner. Furthermore, old Bai3 knockout mice showed the degeneration of hair cells and spiral ganglion neurons in the basal turn. The results suggest that Bai3 plays a crucial role in the development and/or maintenance of pillar cells, which, in turn, are necessary for normal hearing function. Our results may contribute to understanding the mechanisms of hearing loss in human patients.
Collapse
Affiliation(s)
- Chika Saegusa
- Department of Molecular Genetics, Kitasato University School of Medicine, Kanagawa 252-0374, Japan;
- Department of Otorhinolaryngology, Head and Neck Surgery, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Wataru Kakegawa
- Department of Physiology, Keio University School of Medicine, Tokyo 160-8582, Japan; (W.K.); (E.M.); (T.A.); (M.Y.)
| | - Eriko Miura
- Department of Physiology, Keio University School of Medicine, Tokyo 160-8582, Japan; (W.K.); (E.M.); (T.A.); (M.Y.)
| | - Takahiro Aimi
- Department of Physiology, Keio University School of Medicine, Tokyo 160-8582, Japan; (W.K.); (E.M.); (T.A.); (M.Y.)
| | - Sachiyo Mogi
- Department of Otorhinolaryngology, Head and Neck Surgery, Kitasato University, Kanagawa 252-0374, Japan; (S.M.); (T.Y.)
| | - Tatsuhiko Harada
- Department of Otolaryngology, International University of Health and Welfare, Shizuoka 413-0012, Japan;
| | - Taku Yamashita
- Department of Otorhinolaryngology, Head and Neck Surgery, Kitasato University, Kanagawa 252-0374, Japan; (S.M.); (T.Y.)
| | - Michisuke Yuzaki
- Department of Physiology, Keio University School of Medicine, Tokyo 160-8582, Japan; (W.K.); (E.M.); (T.A.); (M.Y.)
| | - Masato Fujioka
- Department of Molecular Genetics, Kitasato University School of Medicine, Kanagawa 252-0374, Japan;
- Department of Otorhinolaryngology, Head and Neck Surgery, Keio University School of Medicine, Tokyo 160-8582, Japan
- Clinical and Translational Research Center, Keio University Hospital, Tokyo 162-8582, Japan
| |
Collapse
|
12
|
Chen F, Sarver DC, Saqib M, Zhou M, Aja S, Seldin MM, Wong GW. CTRP13 ablation improves systemic glucose and lipid metabolism. Mol Metab 2023; 78:101824. [PMID: 37844630 PMCID: PMC10598410 DOI: 10.1016/j.molmet.2023.101824] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 09/30/2023] [Accepted: 10/11/2023] [Indexed: 10/18/2023] Open
Abstract
OBJECTIVE Tissue crosstalk mediated by secreted hormones underlies the integrative control of metabolism. We previously showed that CTRP13/C1QL3, a secreted protein of the C1q family, can improve glucose metabolism and insulin action in vitro and reduce food intake and body weight in mice when centrally delivered. A role for CTRP13 in regulating insulin secretion in isolated islets has also been demonstrated. It remains unclear, however, whether the effects of CTRP13 on cultured cells and in mice reflect the physiological function of the protein. Here, we use a loss-of-function mouse model to address whether CTRP13 is required for metabolic homeostasis. METHODS WT and Ctrp13 knockout (KO) mice fed a standard chow or a high-fat diet were subjected to comprehensive metabolic phenotyping. Transcriptomic analyses were carried out on visceral and subcutaneous fat, liver, and skeletal muscle to identify pathways altered by CTRP13 deficiency. RNA-seq data was further integrated with the Metabolic Syndrome in Man (METSIM) cohort data. Adjusted regression analysis was used to demonstrate that genetic variation of CTRP13 expression accounts for a significant proportion of variance between differentially expressed genes (DEGs) in adipose tissue and metabolic traits in humans. RESULTS Contrary to expectation, chow-fed Ctrp13-KO male mice had elevated physical activity, lower body weight, and improved lipid handling. On a high-fat diet (HFD), Ctrp13-KO mice of either sex were consistently more active and leaner. Loss of CTRP13 reduced hepatic glucose output and improved glucose tolerance, insulin sensitivity, and triglyceride clearance, though with notable sex differences. Consistent with the lean phenotype, transcriptomic analyses revealed a lower inflammatory profile in visceral fat and liver. Reduced hepatic steatosis was correlated with the suppression of lipid synthesis and enhanced lipid catabolism gene expression. Visceral fat had the largest number of DEGs and mediation analyses on the human orthologs of the DEGs suggested the potential causal contribution of CTRP13 to human metabolic syndrome. CONCLUSIONS Our results suggest that CTRP13 is a negative metabolic regulator, and its deficiency improves systemic metabolic profiles. Our data also suggest the reduction in circulating human CTRP13 levels seen in obesity and diabetes may reflect a compensatory physiologic response to counteract insulin resistance.
Collapse
Affiliation(s)
- Fangluo Chen
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Center for Metabolism and Obesity Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Dylan C Sarver
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Center for Metabolism and Obesity Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Muzna Saqib
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Center for Metabolism and Obesity Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Mingqi Zhou
- Department of Biological Chemistry, University of California, Irvine, Irvine, USA; Center for Epigenetics and Metabolism, University of California, Irvine, Irvine, USA
| | - Susan Aja
- Center for Metabolism and Obesity Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Marcus M Seldin
- Department of Biological Chemistry, University of California, Irvine, Irvine, USA; Center for Epigenetics and Metabolism, University of California, Irvine, Irvine, USA
| | - G William Wong
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Center for Metabolism and Obesity Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
13
|
Shiu FH, Wong JC, Bhattacharya D, Kuranaga Y, Parag RR, Alsharif HA, Bhatnagar S, Van Meir EG, Escayg A. Generation and initial characterization of mice lacking full-length BAI3 (ADGRB3) expression. Basic Clin Pharmacol Toxicol 2023; 133:353-363. [PMID: 37337931 PMCID: PMC10730119 DOI: 10.1111/bcpt.13917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 05/24/2023] [Accepted: 06/15/2023] [Indexed: 06/21/2023]
Abstract
Brain-specific angiogenesis inhibitor 3 (ADGRB3/BAI3) belongs to the family of adhesion G protein-coupled receptors. It is most highly expressed in the brain where it plays a role in synaptogenesis and synapse maintenance. Genome-wide association studies have implicated ADGRB3 in disorders such as schizophrenia and epilepsy. Somatic mutations in ADGRB3 have also been identified in cancer. To better understand the in vivo physiological role of ADGRB3, we used CRISPR/Cas9 editing to generate a mouse line with a 7-base pair deletion in Adgrb3 exon 10. Western blot analysis confirmed that homozygous mutants (Adgrb3∆7/∆7 ) lack full-length ADGRB3 expression. The mutant mice were viable and reproduced in Mendelian ratios but demonstrated reduced brain and body weights and deficits in social interaction. Measurements of locomotor function, olfaction, anxiety levels and prepulse inhibition were comparable between heterozygous and homozygous mutants and wild-type littermates. Since ADGRB3 is also expressed in organs such as lung and pancreas, this new mouse model will facilitate elucidation of ADGRB3's role in non-central nervous system-related functions. Finally, since somatic mutations in ADGRB3 were identified in patients with several cancer types, these mice can be used to determine whether loss of ADGRB3 function contributes to tumour development.
Collapse
Affiliation(s)
- Fu Hung Shiu
- Department of Human Genetics, Emory University School of Medicine, Atlanta, Georgia, USA
- Neuroscience Graduate Program, Graduate Division of Biological and Biomedical Sciences, Laney Graduate School, Emory University, Atlanta, Georgia, USA
| | - Jennifer C. Wong
- Department of Human Genetics, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Debanjan Bhattacharya
- Department of Neurology and Rehabilitation Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Yuki Kuranaga
- Department of Neurosurgery, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Rashed R. Parag
- Department of Neurosurgery, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
- O’Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Haifa A. Alsharif
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
- Comprehensive Diabetes Center, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Sushant Bhatnagar
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
- Comprehensive Diabetes Center, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Erwin G. Van Meir
- Department of Neurosurgery, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
- O’Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Andrew Escayg
- Department of Human Genetics, Emory University School of Medicine, Atlanta, Georgia, USA
| |
Collapse
|
14
|
Aimi T, Matsuda K, Yuzaki M. C1ql1-Bai3 signaling is necessary for climbing fiber synapse formation in mature Purkinje cells in coordination with neuronal activity. Mol Brain 2023; 16:61. [PMID: 37488606 PMCID: PMC10367388 DOI: 10.1186/s13041-023-01048-4] [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: 02/27/2023] [Accepted: 07/05/2023] [Indexed: 07/26/2023] Open
Abstract
Changes in neural activity induced by learning and novel environments have been reported to lead to the formation of new synapses in the adult brain. However, the underlying molecular mechanism is not well understood. Here, we show that Purkinje cells (PCs), which have established adult-type monosynaptic innervation by climbing fibers (CFs) after elimination of weak CFs during development, can be reinnervated by multiple CFs by increased expression of the synaptic organizer C1ql1 in CFs or Bai3, a receptor for C1ql1, in PCs. In the adult cerebellum, CFs are known to have transverse branches that run in a mediolateral direction without forming synapses with PCs. Electrophysiological, Ca2+-imaging and immunohistochemical studies showed that overexpression of C1ql1 or Bai3 caused these CF transverse branches to elongate and synapse on the distal dendrites of mature PCs. Mature PCs were also reinnervated by multiple CFs when the glutamate receptor GluD2, which is essential for the maintenance of synapses between granule cells and PCs, was deleted. Interestingly, the effect of GluD2 knockout was not observed in Bai3 knockout PCs. In addition, C1ql1 levels were significantly upregulated in CFs of GluD2 knockout mice, suggesting that endogenous, not overexpressed, C1ql1-Bai3 signaling could regulate the reinnervation of mature PCs by CFs. Furthermore, the effects of C1ql1 and Bai3 overexpression required neuronal activity in the PC and CF, respectively. C1ql1 immunoreactivity at CF-PC synapses was reduced when the neuronal activity of CFs was suppressed. These results suggest that C1ql1-Bai3 signaling may mediate CF synaptogenesis in mature PCs, potentially in concert with neuronal activity.
Collapse
Affiliation(s)
- Takahiro Aimi
- Department of Physiology, Keio University School of Medicine, Tokyo, 160-8582, Japan
| | - Keiko Matsuda
- Department of Physiology, Keio University School of Medicine, Tokyo, 160-8582, Japan
| | - Michisuke Yuzaki
- Department of Physiology, Keio University School of Medicine, Tokyo, 160-8582, Japan.
| |
Collapse
|
15
|
Ramanadham S, Turk J, Bhatnagar S. Noncanonical Regulation of cAMP-Dependent Insulin Secretion and Its Implications in Type 2 Diabetes. Compr Physiol 2023; 13:5023-5049. [PMID: 37358504 PMCID: PMC10809800 DOI: 10.1002/cphy.c220031] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/27/2023]
Abstract
Impaired glucose tolerance (IGT) and β-cell dysfunction in insulin resistance associated with obesity lead to type 2 diabetes (T2D). Glucose-stimulated insulin secretion (GSIS) from β-cells occurs via a canonical pathway that involves glucose metabolism, ATP generation, inactivation of K ATP channels, plasma membrane depolarization, and increases in cytosolic concentrations of [Ca 2+ ] c . However, optimal insulin secretion requires amplification of GSIS by increases in cyclic adenosine monophosphate (cAMP) signaling. The cAMP effectors protein kinase A (PKA) and exchange factor activated by cyclic-AMP (Epac) regulate membrane depolarization, gene expression, and trafficking and fusion of insulin granules to the plasma membrane for amplifying GSIS. The widely recognized lipid signaling generated within β-cells by the β-isoform of Ca 2+ -independent phospholipase A 2 enzyme (iPLA 2 β) participates in cAMP-stimulated insulin secretion (cSIS). Recent work has identified the role of a G-protein coupled receptor (GPCR) activated signaling by the complement 1q like-3 (C1ql3) secreted protein in inhibiting cSIS. In the IGT state, cSIS is attenuated, and the β-cell function is reduced. Interestingly, while β-cell-specific deletion of iPLA 2 β reduces cAMP-mediated amplification of GSIS, the loss of iPLA 2 β in macrophages (MØ) confers protection against the development of glucose intolerance associated with diet-induced obesity (DIO). In this article, we discuss canonical (glucose and cAMP) and novel noncanonical (iPLA 2 β and C1ql3) pathways and how they may affect β-cell (dys)function in the context of impaired glucose intolerance associated with obesity and T2D. In conclusion, we provide a perspective that in IGT states, targeting noncanonical pathways along with canonical pathways could be a more comprehensive approach for restoring β-cell function in T2D. © 2023 American Physiological Society. Compr Physiol 13:5023-5049, 2023.
Collapse
Affiliation(s)
- Sasanka Ramanadham
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Alabama, USA
- Comprehensive Diabetes Center, University of Alabama at Birmingham, Alabama, USA
| | - John Turk
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Sushant Bhatnagar
- Comprehensive Diabetes Center, University of Alabama at Birmingham, Alabama, USA
- Department of Medicine, University of Alabama at Birmingham, Alabama, USA
| |
Collapse
|
16
|
Brunert D, Quintela RM, Rothermel M. The anterior olfactory nucleus revisited - an emerging role for neuropathological conditions? Prog Neurobiol 2023:102486. [PMID: 37343762 DOI: 10.1016/j.pneurobio.2023.102486] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 06/14/2023] [Accepted: 06/16/2023] [Indexed: 06/23/2023]
Abstract
Olfaction is an important sensory modality for many species and greatly influences animal and human behavior. Still, much about olfactory perception remains unknown. The anterior olfactory nucleus is one of the brain's central early olfactory processing areas. Located directly posterior to the olfactory bulb in the olfactory peduncle with extensive in- and output connections and unique cellular composition, it connects olfactory processing centers of the left and right hemispheres. Almost 20 years have passed since the last comprehensive review on the anterior olfactory nucleus has been published and significant advances regarding its anatomy, function, and pathophysiology have been made in the meantime. Here we briefly summarize previous knowledge on the anterior olfactory nucleus, give detailed insights into the progress that has been made in recent years, and map out its emerging importance in translational research of neurological diseases.
Collapse
Affiliation(s)
- Daniela Brunert
- Institute of Physiology, Medical Faculty, Otto-von-Guericke-University, 39120 Magdeburg, Germany
| | | | - Markus Rothermel
- Institute of Physiology, Medical Faculty, Otto-von-Guericke-University, 39120 Magdeburg, Germany.
| |
Collapse
|
17
|
Alsharif H, Latimer MN, Perez KC, Alexander J, Rahman MM, Challa AK, Kim JA, Ramanadham S, Young M, Bhatnagar S. Loss of Brain Angiogenesis Inhibitor-3 (BAI3) G-Protein Coupled Receptor in Mice Regulates Adaptive Thermogenesis by Enhancing Energy Expenditure. Metabolites 2023; 13:711. [PMID: 37367869 PMCID: PMC10301052 DOI: 10.3390/metabo13060711] [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: 03/16/2023] [Revised: 04/24/2023] [Accepted: 05/18/2023] [Indexed: 06/28/2023] Open
Abstract
Effective energy expenditure is critical for maintaining body weight (BW). However, underlying mechanisms contributing to increased BW remain unknown. We characterized the role of brain angiogenesis inhibitor-3 (BAI3/ADGRB3), an adhesion G-protein coupled receptor (aGPCR), in regulating BW. A CRISPR/Cas9 gene editing approach was utilized to generate a whole-body deletion of the BAI3 gene (BAI3-/-). In both BAI3-/- male and female mice, a significant reduction in BW was observed compared to BAI3+/+ control mice. Quantitative magnetic imaging analysis showed that lean and fat masses were reduced in male and female mice with BAI3 deficiency. Total activity, food intake, energy expenditure (EE), and respiratory exchange ratio (RER) were assessed in mice housed at room temperature using a Comprehensive Lab Animal Monitoring System (CLAMS). While no differences were observed in the activity between the two genotypes in male or female mice, energy expenditure was increased in both sexes with BAI3 deficiency. However, at thermoneutrality (30 °C), no differences in energy expenditure were observed between the two genotypes for either sex, suggesting a role for BAI3 in adaptive thermogenesis. Notably, in male BAI3-/- mice, food intake was reduced, and RER was increased, but these attributes remained unchanged in the female mice upon BAI3 loss. Gene expression analysis showed increased mRNA abundance of thermogenic genes Ucp1, Pgc1α, Prdm16, and Elov3 in brown adipose tissue (BAT). These outcomes suggest that adaptive thermogenesis due to enhanced BAT activity contributes to increased energy expenditure and reduced BW with BAI3 deficiency. Additionally, sex-dependent differences were observed in food intake and RER. These studies identify BAI3 as a novel regulator of BW that can be potentially targeted to improve whole-body energy expenditure.
Collapse
Affiliation(s)
- Haifa Alsharif
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (H.A.); (K.C.P.); (J.A.); (M.M.R.); (J.-A.K.)
- Comprehensive Diabetes Center, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Mary N. Latimer
- Division of Cardiovascular Disease, Department of Medicine, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (M.Y.)
| | - Katherine C. Perez
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (H.A.); (K.C.P.); (J.A.); (M.M.R.); (J.-A.K.)
- Comprehensive Diabetes Center, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Justin Alexander
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (H.A.); (K.C.P.); (J.A.); (M.M.R.); (J.-A.K.)
- Comprehensive Diabetes Center, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Md Mostafizur Rahman
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (H.A.); (K.C.P.); (J.A.); (M.M.R.); (J.-A.K.)
- Comprehensive Diabetes Center, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Anil K. Challa
- Department of Biology, University of Alabama at Birmingham, Birmingham, AL 35294, USA;
| | - Jeong-A. Kim
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (H.A.); (K.C.P.); (J.A.); (M.M.R.); (J.-A.K.)
- Comprehensive Diabetes Center, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Sasanka Ramanadham
- Comprehensive Diabetes Center, University of Alabama at Birmingham, Birmingham, AL 35294, USA
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Martin Young
- Division of Cardiovascular Disease, Department of Medicine, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (M.Y.)
| | - Sushant Bhatnagar
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (H.A.); (K.C.P.); (J.A.); (M.M.R.); (J.-A.K.)
- Comprehensive Diabetes Center, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| |
Collapse
|
18
|
Hale WD, Südhof TC, Huganir RL. Engineered adhesion molecules drive synapse organization. Proc Natl Acad Sci U S A 2023; 120:e2215905120. [PMID: 36638214 PMCID: PMC9934208 DOI: 10.1073/pnas.2215905120] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Accepted: 12/04/2022] [Indexed: 01/15/2023] Open
Abstract
In multicellular organisms, cell-adhesion molecules connect cells into tissues and mediate intercellular signaling between these cells. In vertebrate brains, synaptic cell-adhesion molecules (SAMs) guide the formation, specification, and plasticity of synapses. Some SAMs, when overexpressed in cultured neurons or in heterologous cells co-cultured with neurons, drive formation of synaptic specializations onto the overexpressing cells. However, genetic deletion of the same SAMs from neurons often has no effect on synapse numbers, but frequently severely impairs synaptic transmission, suggesting that most SAMs control the function and plasticity of synapses (i.e., organize synapses) instead of driving their initial establishment (i.e., make synapses). Since few SAMs were identified that mediate initial synapse formation, it is difficult to develop methods that enable experimental control of synaptic connections by targeted expression of these SAMs. To overcome this difficulty, we engineered novel SAMs from bacterial proteins with no eukaryotic homologues that drive synapse formation. We named these engineered adhesion proteins "Barnoligin" and "Starexin" because they were assembled from parts of Barnase and Neuroligin-1 or of Barstar and Neurexin3β, respectively. Barnoligin and Starexin robustly induce the formation of synaptic specializations in a specific and directional manner in cultured neurons. Synapse formation by Barnoligin and Starexin requires both their extracellular Barnase- and Barstar-derived interaction domains and their Neuroligin- and Neurexin-derived intracellular signaling domains. Our findings support a model of synapse formation whereby trans-synaptic interactions by SAMs drive synapse organization via adhesive interactions that activate signaling cascades.
Collapse
Affiliation(s)
- W. Dylan Hale
- Department of Molecular and Cellular Physiology, Stanford University, Stanford, CA94305
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD21205
| | - Thomas C. Südhof
- Department of Molecular and Cellular Physiology, Stanford University, Stanford, CA94305
- HHMI, Stanford University, Stanford, CA94305
| | - Richard L. Huganir
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD21205
| |
Collapse
|
19
|
Chae H, Banerjee A, Dussauze M, Albeanu DF. Long-range functional loops in the mouse olfactory system and their roles in computing odor identity. Neuron 2022; 110:3970-3985.e7. [PMID: 36174573 PMCID: PMC9742324 DOI: 10.1016/j.neuron.2022.09.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 07/12/2022] [Accepted: 09/02/2022] [Indexed: 12/15/2022]
Abstract
Elucidating the neural circuits supporting odor identification remains an open challenge. Here, we analyze the contribution of the two output cell types of the mouse olfactory bulb (mitral and tufted cells) to decode odor identity and concentration and its dependence on top-down feedback from their respective major cortical targets: piriform cortex versus anterior olfactory nucleus. We find that tufted cells substantially outperform mitral cells in decoding both odor identity and intensity. Cortical feedback selectively regulates the activity of its dominant bulb projection cell type and implements different computations. Piriform feedback specifically restructures mitral responses, whereas feedback from the anterior olfactory nucleus preferentially controls the gain of tufted representations without altering their odor tuning. Our results identify distinct functional loops involving the mitral and tufted cells and their cortical targets. We suggest that in addition to the canonical mitral-to-piriform pathway, tufted cells and their target regions are ideally positioned to compute odor identity.
Collapse
Affiliation(s)
- Honggoo Chae
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
| | - Arkarup Banerjee
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA; Cold Spring Harbor Laboratory School for Biological Sciences, Cold Spring Harbor, NY, USA
| | - Marie Dussauze
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA; Cold Spring Harbor Laboratory School for Biological Sciences, Cold Spring Harbor, NY, USA
| | - Dinu F Albeanu
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA; Cold Spring Harbor Laboratory School for Biological Sciences, Cold Spring Harbor, NY, USA.
| |
Collapse
|
20
|
Noguer-Calabús I, Schäble S, Kalenscher T. Lesions of nucleus accumbens shell abolish socially transmitted food preferences. Eur J Neurosci 2022; 56:5795-5809. [PMID: 36151057 DOI: 10.1111/ejn.15827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 06/05/2022] [Accepted: 07/28/2022] [Indexed: 01/10/2023]
Abstract
Rats adapt their food choices to conform to their conspecifics' dietary preferences. The nucleus accumbens shell is a relevant brain region to process reward-related and motivated behaviours and social information. Here, we hypothesize that the integrity of the nucleus accumbens shell is necessary to show socially transmitted food preferences. We made excitotoxic and sham lesions of nucleus accumbens shell in male Long-Evans rats who performed a social transmission of food preference task. In this task, observer rats revealed their original preference for one out of two food options. Afterward, they were exposed to a demonstrator rat who was fed with the observer's originally non-preferred food, and the observer's food choices were sampled again. Sham lesioned observer rats changed their food preferences following interaction with the demonstrator, specifically by increasing the intake of their originally non-preferred food type. This interaction-related change in preference was not found after nucleus accumbens shell lesions. The lesion effects on choice were not the consequence of impaired social or non-social motivation, anxiety or sensory or motor function, suggesting that they reflected a genuine deficit in social reward revaluation. These results highlight the role of nucleus accumbens shell in revaluating food rewards to match a conspecific's preferences.
Collapse
Affiliation(s)
- Irina Noguer-Calabús
- Comparative Psychology, Institute of Experimental Psychology, Heinrich-Heine University Düsseldorf, Düsseldorf, Germany
| | - Sandra Schäble
- Comparative Psychology, Institute of Experimental Psychology, Heinrich-Heine University Düsseldorf, Düsseldorf, Germany
| | - Tobias Kalenscher
- Comparative Psychology, Institute of Experimental Psychology, Heinrich-Heine University Düsseldorf, Düsseldorf, Germany
| |
Collapse
|
21
|
Chen Y, Chen X, Baserdem B, Zhan H, Li Y, Davis MB, Kebschull JM, Zador AM, Koulakov AA, Albeanu DF. High-throughput sequencing of single neuron projections reveals spatial organization in the olfactory cortex. Cell 2022; 185:4117-4134.e28. [PMID: 36306734 PMCID: PMC9681627 DOI: 10.1016/j.cell.2022.09.038] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 07/22/2022] [Accepted: 09/28/2022] [Indexed: 11/07/2022]
Abstract
In most sensory modalities, neuronal connectivity reflects behaviorally relevant stimulus features, such as spatial location, orientation, and sound frequency. By contrast, the prevailing view in the olfactory cortex, based on the reconstruction of dozens of neurons, is that connectivity is random. Here, we used high-throughput sequencing-based neuroanatomical techniques to analyze the projections of 5,309 mouse olfactory bulb and 30,433 piriform cortex output neurons at single-cell resolution. Surprisingly, statistical analysis of this much larger dataset revealed that the olfactory cortex connectivity is spatially structured. Single olfactory bulb neurons targeting a particular location along the anterior-posterior axis of piriform cortex also project to matched, functionally distinct, extra-piriform targets. Moreover, single neurons from the targeted piriform locus also project to the same matched extra-piriform targets, forming triadic circuit motifs. Thus, as in other sensory modalities, olfactory information is routed at early stages of processing to functionally diverse targets in a coordinated manner.
Collapse
Affiliation(s)
- Yushu Chen
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Xiaoyin Chen
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | | | - Huiqing Zhan
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Yan Li
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Martin B Davis
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | | | - Anthony M Zador
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA.
| | | | - Dinu F Albeanu
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA.
| |
Collapse
|
22
|
Lala T, Hall RA. Adhesion G protein-coupled receptors: structure, signaling, physiology, and pathophysiology. Physiol Rev 2022; 102:1587-1624. [PMID: 35468004 PMCID: PMC9255715 DOI: 10.1152/physrev.00027.2021] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 03/11/2022] [Accepted: 04/16/2022] [Indexed: 01/17/2023] Open
Abstract
Adhesion G protein-coupled receptors (AGPCRs) are a family of 33 receptors in humans exhibiting a conserved general structure but diverse expression patterns and physiological functions. The large NH2 termini characteristic of AGPCRs confer unique properties to each receptor and possess a variety of distinct domains that can bind to a diverse array of extracellular proteins and components of the extracellular matrix. The traditional view of AGPCRs, as implied by their name, is that their core function is the mediation of adhesion. In recent years, though, many surprising advances have been made regarding AGPCR signaling mechanisms, activation by mechanosensory forces, and stimulation by small-molecule ligands such as steroid hormones and bioactive lipids. Thus, a new view of AGPCRs has begun to emerge in which these receptors are seen as massive signaling platforms that are crucial for the integration of adhesive, mechanosensory, and chemical stimuli. This review article describes the recent advances that have led to this new understanding of AGPCR function and also discusses new insights into the physiological actions of these receptors as well as their roles in human disease.
Collapse
Affiliation(s)
- Trisha Lala
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, Georgia
| | - Randy A Hall
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, Georgia
| |
Collapse
|
23
|
González-Calvo I, Cizeron M, Bessereau JL, Selimi F. Synapse Formation and Function Across Species: Ancient Roles for CCP, CUB, and TSP-1 Structural Domains. Front Neurosci 2022; 16:866444. [PMID: 35546877 PMCID: PMC9083331 DOI: 10.3389/fnins.2022.866444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 03/28/2022] [Indexed: 11/17/2022] Open
Abstract
The appearance of synapses was a crucial step in the creation of the variety of nervous systems that are found in the animal kingdom. With increased complexity of the organisms came a greater number of synaptic proteins. In this review we describe synaptic proteins that contain the structural domains CUB, CCP, or TSP-1. These domains are found in invertebrates and vertebrates, and CUB and CCP domains were initially described in proteins belonging to the complement system of innate immunity. Interestingly, they are found in synapses of the nematode C. elegans, which does not have a complement system, suggesting an ancient function. Comparison of the roles of CUB-, CCP-, and TSP-1 containing synaptic proteins in various species shows that in more complex nervous systems, these structural domains are combined with other domains and that there is partial conservation of their function. These three domains are thus basic building blocks of the synaptic architecture. Further studies of structural domains characteristic of synaptic proteins in invertebrates such as C. elegans and comparison of their role in mammals will help identify other conserved synaptic molecular building blocks. Furthermore, this type of functional comparison across species will also identify structural domains added during evolution in correlation with increased complexity, shedding light on mechanisms underlying cognition and brain diseases.
Collapse
Affiliation(s)
- Inés González-Calvo
- Center for Interdisciplinary Research in Biology (CIRB), Collège de France, CNRS, INSERM, PSL Research University, Paris, France
| | - Mélissa Cizeron
- Univ Lyon, Université Claude Bernard Lyon 1, CNRS UMR-5284, INSERM U-1314, MeLiS, Institut NeuroMyoGène, Lyon, France
| | - Jean-Louis Bessereau
- Univ Lyon, Université Claude Bernard Lyon 1, CNRS UMR-5284, INSERM U-1314, MeLiS, Institut NeuroMyoGène, Lyon, France
| | - Fekrije Selimi
- Center for Interdisciplinary Research in Biology (CIRB), Collège de France, CNRS, INSERM, PSL Research University, Paris, France
| |
Collapse
|
24
|
Liu Z, Jiang M, Liakath-Ali K, Sclip A, Ko J, Zhang RS, Südhof TC. Deletion of Calsyntenin-3, an atypical cadherin, suppresses inhibitory synapses but increases excitatory parallel-fiber synapses in cerebellum. eLife 2022; 11:e70664. [PMID: 35420982 PMCID: PMC9064300 DOI: 10.7554/elife.70664] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Accepted: 04/14/2022] [Indexed: 01/11/2023] Open
Abstract
Cadherins contribute to the organization of nearly all tissues, but the functions of several evolutionarily conserved cadherins, including those of calsyntenins, remain enigmatic. Puzzlingly, two distinct, non-overlapping functions for calsyntenins were proposed: As postsynaptic neurexin ligands in synapse formation, or as presynaptic kinesin adaptors in vesicular transport. Here, we show that, surprisingly, acute CRISPR-mediated deletion of calsyntenin-3 in mouse cerebellum in vivo causes a large decrease in inhibitory synapse, but a robust increase in excitatory parallel-fiber synapses in Purkinje cells. As a result, inhibitory synaptic transmission was suppressed, whereas parallel-fiber synaptic transmission was enhanced in Purkinje cells by the calsyntenin-3 deletion. No changes in the dendritic architecture of Purkinje cells or in climbing-fiber synapses were detected. Sparse selective deletion of calsyntenin-3 only in Purkinje cells recapitulated the synaptic phenotype, indicating that calsyntenin-3 acts by a cell-autonomous postsynaptic mechanism in cerebellum. Thus, by inhibiting formation of excitatory parallel-fiber synapses and promoting formation of inhibitory synapses in the same neuron, calsyntenin-3 functions as a postsynaptic adhesion molecule that regulates the excitatory/inhibitory balance in Purkinje cells.
Collapse
Affiliation(s)
- Zhihui Liu
- Department of Molecular and Cellular Physiology, Stanford UniversityStanfordUnited States
- Howard Hughes Medical Institute, Stanford UniversityStanfordUnited States
| | - Man Jiang
- Department of Molecular and Cellular Physiology, Stanford UniversityStanfordUnited States
| | - Kif Liakath-Ali
- Department of Molecular and Cellular Physiology, Stanford UniversityStanfordUnited States
| | - Alessandra Sclip
- Department of Molecular and Cellular Physiology, Stanford UniversityStanfordUnited States
- Howard Hughes Medical Institute, Stanford UniversityStanfordUnited States
| | - Jaewon Ko
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and TechnologyDaeguRepublic of Korea
| | - Roger Shen Zhang
- Department of Molecular and Cellular Physiology, Stanford UniversityStanfordUnited States
| | - Thomas C Südhof
- Department of Molecular and Cellular Physiology, Stanford UniversityStanfordUnited States
- Howard Hughes Medical Institute, Stanford UniversityStanfordUnited States
| |
Collapse
|
25
|
Cnops V, Iyer VR, Parathy N, Wong P, Dawe GS. Test, Rinse, Repeat: A Review of Carryover Effects in Rodent Behavioral Assays. Neurosci Biobehav Rev 2022; 135:104560. [DOI: 10.1016/j.neubiorev.2022.104560] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 01/26/2022] [Accepted: 02/01/2022] [Indexed: 01/21/2023]
|
26
|
Wang J, Miao Y, Wicklein R, Sun Z, Wang J, Jude KM, Fernandes RA, Merrill SA, Wernig M, Garcia KC, Südhof TC. RTN4/NoGo-receptor binding to BAI adhesion-GPCRs regulates neuronal development. Cell 2021; 184:5869-5885.e25. [PMID: 34758294 PMCID: PMC8620742 DOI: 10.1016/j.cell.2021.10.016] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 06/01/2021] [Accepted: 10/13/2021] [Indexed: 10/19/2022]
Abstract
RTN4-binding proteins were widely studied as "NoGo" receptors, but their physiological interactors and roles remain elusive. Similarly, BAI adhesion-GPCRs were associated with numerous activities, but their ligands and functions remain unclear. Using unbiased approaches, we observed an unexpected convergence: RTN4 receptors are high-affinity ligands for BAI adhesion-GPCRs. A single thrombospondin type 1-repeat (TSR) domain of BAIs binds to the leucine-rich repeat domain of all three RTN4-receptor isoforms with nanomolar affinity. In the 1.65 Å crystal structure of the BAI1/RTN4-receptor complex, C-mannosylation of tryptophan and O-fucosylation of threonine in the BAI TSR-domains creates a RTN4-receptor/BAI interface shaped by unusual glycoconjugates that enables high-affinity interactions. In human neurons, RTN4 receptors regulate dendritic arborization, axonal elongation, and synapse formation by differential binding to glial versus neuronal BAIs, thereby controlling neural network activity. Thus, BAI binding to RTN4/NoGo receptors represents a receptor-ligand axis that, enabled by rare post-translational modifications, controls development of synaptic circuits.
Collapse
Affiliation(s)
- Jie Wang
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Yi Miao
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA 94305, USA; Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Rebecca Wicklein
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Zijun Sun
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Jinzhao Wang
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA 94305, USA; Institute for Stem Cell Biology and Regenerative Medicine, Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Kevin M Jude
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA 94305, USA; Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Ricardo A Fernandes
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA 94305, USA; Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Sean A Merrill
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Marius Wernig
- Institute for Stem Cell Biology and Regenerative Medicine, Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - K Christopher Garcia
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA 94305, USA; Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Structural Biology, Stanford University School of Medicine, Stanford, CA 94305, USA.
| | - Thomas C Südhof
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA 94305, USA; Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|
27
|
Yan Y, Aierken A, Wang C, Song D, Ni J, Wang Z, Quan Z, Qing H. A potential biomarker of preclinical Alzheimer's disease: The olfactory dysfunction and its pathogenesis-based neural circuitry impairments. Neurosci Biobehav Rev 2021; 132:857-869. [PMID: 34810025 DOI: 10.1016/j.neubiorev.2021.11.009] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 10/26/2021] [Accepted: 11/07/2021] [Indexed: 01/24/2023]
Abstract
The olfactory dysfunction can signal and act as a potential biomarker of preclinical AD. However, the precise regulatory mechanism of olfactory function on the neural pathogenesis of AD is still unclear. The impairment of neural networks in olfaction system has been shown to be tightly associated with AD. As key brain regions of the olfactory system, the olfactory bulb (OB) and the piriform cortex (PCx) have a profound influence on the olfactory function. Therefore, this review will explore the mechanism of olfactory dysfunction in preclinical AD in the perspective of abnormal neural networks in the OB and PCx and their associated brain regions, especially from two aspects of aberrant oscillations and synaptic plasticity damages, which help better understand the underlying mechanism of olfactory neural network damages related to AD.
Collapse
Affiliation(s)
- Yan Yan
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Science, Beijing Institute of Technology, Beijing, 100081, China
| | - Ailikemu Aierken
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Science, Beijing Institute of Technology, Beijing, 100081, China
| | - Chunjian Wang
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Science, Beijing Institute of Technology, Beijing, 100081, China
| | - Da Song
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Science, Beijing Institute of Technology, Beijing, 100081, China
| | - Junjun Ni
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Science, Beijing Institute of Technology, Beijing, 100081, China
| | - Zhe Wang
- Advanced Innovation Center for Human Brain Protection, Capital Medical University, The National Clinical Research Center for Geriatric Disease, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Zhenzhen Quan
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Science, Beijing Institute of Technology, Beijing, 100081, China.
| | - Hong Qing
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Science, Beijing Institute of Technology, Beijing, 100081, China.
| |
Collapse
|
28
|
Olfactory Optogenetics: Light Illuminates the Chemical Sensing Mechanisms of Biological Olfactory Systems. BIOSENSORS-BASEL 2021; 11:bios11090309. [PMID: 34562900 PMCID: PMC8470751 DOI: 10.3390/bios11090309] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 08/17/2021] [Accepted: 08/27/2021] [Indexed: 01/26/2023]
Abstract
The mammalian olfactory system has an amazing ability to distinguish thousands of odorant molecules at the trace level. Scientists have made great achievements on revealing the olfactory sensing mechanisms in decades; even though many issues need addressing. Optogenetics provides a novel technical approach to solve this dilemma by utilizing light to illuminate specific part of the olfactory system; which can be used in all corners of the olfactory system for revealing the olfactory mechanism. This article reviews the most recent advances in olfactory optogenetics devoted to elucidate the mechanisms of chemical sensing. It thus attempts to introduce olfactory optogenetics according to the structure of the olfactory system. It mainly includes the following aspects: the sensory input from the olfactory epithelium to the olfactory bulb; the influences of the olfactory bulb (OB) neuron activity patterns on olfactory perception; the regulation between the olfactory cortex and the olfactory bulb; and the neuromodulation participating in odor coding by dominating the olfactory bulb. Finally; current challenges and future development trends of olfactory optogenetics are proposed and discussed.
Collapse
|
29
|
Perinatal SSRI Exposure Disrupts G Protein-coupled Receptor BAI3 in Developing Dentate Gyrus and Adult Emotional Behavior: Relevance to Psychiatric Disorders. Neuroscience 2021; 471:32-50. [PMID: 34293414 DOI: 10.1016/j.neuroscience.2021.07.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 07/07/2021] [Accepted: 07/08/2021] [Indexed: 12/16/2022]
Abstract
Selective serotonin reuptake inhibitor (SSRI) antidepressants are widely prescribed to pregnant women suffering with depression, although the long-term impact of these medications on exposed offspring are poorly understood. Perinatal SSRI exposure alters human offspring's neurodevelopment and increases risk for psychiatric illness in later life. Rodent studies suggest that perinatal SSRI-induced behavioral abnormalities are driven by changes in the serotonin system as well as epigenetic and transcriptomic changes in the developing hippocampus. A major gene altered by perinatal SSRI exposure is the G-protein coupled receptor Brain Angiogenesis Inhibitor 3 (BAI3). Our present study shows that perinatal exposure to the SSRI citalopram increases mRNA expression of Bai3 and related molecules (including its C1ql ligands) in the early postnatal dentate gyrus of male and female offspring. Transient Bai3 mRNA knockdown in perinatal SSRI-exposed dentate gyrus lessened behavioral consequences of perinatal SSRI exposure, leading to increased active stress coping. To determine translational implications of this work, we examined expression of BAI3 and related molecules in hippocampus and prefrontal cortex from patients that suffered with depression or schizophrenia relative to healthy control subjects. We found sex- and region-specific changes in mRNA expression of BAI3 and its ligands C1QL2 and C1QL3 in men and women with a history of psychiatric disorders compared to healthy controls. Together these results suggest that abnormal BAI3 signaling may contribute to molecular mechanisms that drive adverse effects of perinatal SSRI exposure, and show evidence for alterations of BAI3 signaling in the hippocampus of patients that suffer depression and schizophrenia.
Collapse
|
30
|
Südhof TC. The cell biology of synapse formation. J Cell Biol 2021; 220:e202103052. [PMID: 34086051 PMCID: PMC8186004 DOI: 10.1083/jcb.202103052] [Citation(s) in RCA: 170] [Impact Index Per Article: 42.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 05/13/2021] [Accepted: 05/17/2021] [Indexed: 04/25/2023] Open
Abstract
In a neural circuit, synapses transfer information rapidly between neurons and transform this information during transfer. The diverse computational properties of synapses are shaped by the interactions between pre- and postsynaptic neurons. How synapses are assembled to form a neural circuit, and how the specificity of synaptic connections is achieved, is largely unknown. Here, I posit that synaptic adhesion molecules (SAMs) organize synapse formation. Diverse SAMs collaborate to achieve the astounding specificity and plasticity of synapses, with each SAM contributing different facets. In orchestrating synapse assembly, SAMs likely act as signal transduction devices. Although many candidate SAMs are known, only a few SAMs appear to have a major impact on synapse formation. Thus, a limited set of collaborating SAMs likely suffices to account for synapse formation. Strikingly, several SAMs are genetically linked to neuropsychiatric disorders, suggesting that impairments in synapse assembly are instrumental in the pathogenesis of neuropsychiatric disorders.
Collapse
|
31
|
Biswas J, Pijewski RS, Makol R, Miramontes TG, Thompson BL, Kresic LC, Burghard AL, Oliver DL, Martinelli DC. C1ql1 is expressed in adult outer hair cells of the cochlea in a tonotopic gradient. PLoS One 2021; 16:e0251412. [PMID: 33979385 PMCID: PMC8115824 DOI: 10.1371/journal.pone.0251412] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Accepted: 04/26/2021] [Indexed: 01/08/2023] Open
Abstract
Hearing depends on the transduction of sounds into neural signals by the inner hair cells of the cochlea. Cochleae also have outer hair cells with unique electromotile properties that increase auditory sensitivity, but they are particularly susceptible to damage by intense noise exposure, ototoxic drugs, and aging. Although the outer hair cells have synapses on afferent neurons that project to the brain, the function of this neuronal circuit is unclear. Here, we created a novel mouse allele that inserts a fluorescent reporter at the C1ql1 locus which revealed gene expression in the outer hair cells and allowed creation of outer hair cell-specific C1ql1 knockout mice. We found that C1ql1 expression in outer hair cells corresponds to areas with the most sensitive frequencies of the mouse audiogram, and that it has an unexpected adolescence-onset developmental timing. No expression was observed in the inner hair cells. Since C1QL1 in the brain is made by neurons, transported anterogradely in axons, and functions in the synaptic cleft, C1QL1 may serve a similar function at the outer hair cell afferent synapse. Histological analyses revealed that C1ql1 conditional knockout cochleae may have reduced outer hair cell afferent synapse maintenance. However, auditory behavioral and physiological assays did not reveal a compelling phenotype. Nonetheless, this study identifies a potentially useful gene expressed in the cochlea and opens the door for future studies aimed at elucidating the function of C1QL1 and the function of the outer hair cell and its afferent neurons.
Collapse
Affiliation(s)
- Joyshree Biswas
- Department of Neuroscience, University of Connecticut Health, Farmington, CT, United States of America
| | - Robert S. Pijewski
- Department of Neuroscience, University of Connecticut Health, Farmington, CT, United States of America
| | - Rohit Makol
- Department of Neuroscience, University of Connecticut Health, Farmington, CT, United States of America
- The Connecticut Institute for the Brain and Cognitive Sciences (IBACS), Storrs, CT, United States of America
| | - Tania G. Miramontes
- Department of Neuroscience, University of Connecticut Health, Farmington, CT, United States of America
| | - Brianna L. Thompson
- Department of Neuroscience, University of Connecticut Health, Farmington, CT, United States of America
| | - Lyndsay C. Kresic
- Department of Neuroscience, University of Connecticut Health, Farmington, CT, United States of America
| | - Alice L. Burghard
- Department of Neuroscience, University of Connecticut Health, Farmington, CT, United States of America
| | - Douglas L. Oliver
- Department of Neuroscience, University of Connecticut Health, Farmington, CT, United States of America
| | - David C. Martinelli
- Department of Neuroscience, University of Connecticut Health, Farmington, CT, United States of America
- The Connecticut Institute for the Brain and Cognitive Sciences (IBACS), Storrs, CT, United States of America
- * E-mail:
| |
Collapse
|
32
|
Urrieta E, Escobar ML. Metaplastic regulation of neocortical long-term depression in vivo is sensitive to distinct phases of conditioned taste aversion. Neurobiol Learn Mem 2021; 182:107449. [PMID: 33915300 DOI: 10.1016/j.nlm.2021.107449] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2021] [Revised: 04/12/2021] [Accepted: 04/22/2021] [Indexed: 10/21/2022]
Abstract
Metaplasticity refers to the persistent modification, by previous activity, in the ability to induce synaptic plasticity. Accumulated evidence has proposed that metaplasticity contributes to network function and cognitive processes such as learning and memory. In this regard, it has been observed that training in several behavioral tasks modifies the possibility to induce subsequent synaptic plasticity, such as long-term potentiation (LTP) and long-term depression (LTD). For instance, our previous studies have shown that conditioned taste aversion (CTA) training prevents the induction of in vivo LTP in the projection from the basolateral nucleus of the amygdala to the insular cortex (BLA-IC). Likewise, we reported that extinction of CTA allows induction but not maintenance of LTP in the same pathway. Besides, we showed that it is possible to express in vivo low-frequency stimulation LTD in the BLA-IC projection and that its induction prior to CTA training facilitates the extinction of this task. However, until now, little is known about the participation of LTD on metaplastic processes. The present study aimed to analyze whether CTA training modifies the expression of in vivo LTD in the BLA-IC projection. To do so, animals received low-frequency stimulation to induce IC-LTD 48 h after CTA training. Our results show that CTA training occludes the subsequent induction of LTD in the BLA-IC pathway in a retrieval-dependent manner. These findings reveal that CTA elicits a metaplastic regulation of long-lasting changes in the IC synaptic strength, as well as that specific phases of learning differentially take part in adjusting the expression of synaptic plasticity in neocortical regions.
Collapse
Affiliation(s)
- Esteban Urrieta
- División de Investigación y Estudios de Posgrado, Facultad de Psicología, Universidad Nacional Autónoma de México, 04510, Mexico
| | - Martha L Escobar
- División de Investigación y Estudios de Posgrado, Facultad de Psicología, Universidad Nacional Autónoma de México, 04510, Mexico.
| |
Collapse
|
33
|
Sando R, Südhof TC. Latrophilin GPCR signaling mediates synapse formation. eLife 2021; 10:e65717. [PMID: 33646123 PMCID: PMC7954527 DOI: 10.7554/elife.65717] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2020] [Accepted: 02/26/2021] [Indexed: 12/17/2022] Open
Abstract
Neural circuit assembly in the brain requires precise establishment of synaptic connections, but the mechanisms of synapse assembly remain incompletely understood. Latrophilins are postsynaptic adhesion-GPCRs that engage in trans-synaptic complexes with presynaptic teneurins and FLRTs. In mouse CA1-region neurons, Latrophilin-2 and Latrophilin-3 are essential for formation of entorhinal-cortex-derived and Schaffer-collateral-derived synapses, respectively. However, it is unknown whether latrophilins function as GPCRs in synapse formation. Here, we show that Latrophilin-2 and Latrophilin-3 exhibit constitutive GPCR activity that increases cAMP levels, which was blocked by a mutation interfering with G-protein and arrestin interactions of GPCRs. The same mutation impaired the ability of Latrophilin-2 and Latrophilin-3 to rescue the synapse-loss phenotype in Latrophilin-2 and Latrophilin-3 knockout neurons in vivo. Our results suggest that Latrophilin-2 and Latrophilin-3 require GPCR signaling in synapse formation, indicating that latrophilins promote synapse formation in the hippocampus by activating a classical GPCR-signaling pathway.
Collapse
Affiliation(s)
- Richard Sando
- Department of Molecular & Cellular Physiology and Howard Hughes Medical Institute, Stanford University School of MedicineStanfordUnited States
| | - Thomas C Südhof
- Department of Molecular & Cellular Physiology and Howard Hughes Medical Institute, Stanford University School of MedicineStanfordUnited States
| |
Collapse
|
34
|
Hartig R, Wolf D, Schmeisser MJ, Kelsch W. Genetic influences of autism candidate genes on circuit wiring and olfactory decoding. Cell Tissue Res 2021; 383:581-595. [PMID: 33515293 PMCID: PMC7872953 DOI: 10.1007/s00441-020-03390-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Accepted: 12/10/2020] [Indexed: 12/24/2022]
Abstract
Olfaction supports a multitude of behaviors vital for social communication and interactions between conspecifics. Intact sensory processing is contingent upon proper circuit wiring. Disturbances in genetic factors controlling circuit assembly and synaptic wiring can lead to neurodevelopmental disorders, such as autism spectrum disorder (ASD), where impaired social interactions and communication are core symptoms. The variability in behavioral phenotype expression is also contingent upon the role environmental factors play in defining genetic expression. Considering the prevailing clinical diagnosis of ASD, research on therapeutic targets for autism is essential. Behavioral impairments may be identified along a range of increasingly complex social tasks. Hence, the assessment of social behavior and communication is progressing towards more ethologically relevant tasks. Garnering a more accurate understanding of social processing deficits in the sensory domain may greatly contribute to the development of therapeutic targets. With that framework, studies have found a viable link between social behaviors, circuit wiring, and altered neuronal coding related to the processing of salient social stimuli. Here, the relationship between social odor processing in rodents and humans is examined in the context of health and ASD, with special consideration for how genetic expression and neuronal connectivity may regulate behavioral phenotypes.
Collapse
Affiliation(s)
- Renée Hartig
- Department of Psychiatry & Psychotherapy, University Medical Center, Johannes Gutenberg-University, 55131, Mainz, Germany.,Focus Program Translational Neurosciences (FTN), University Medical Center, Johannes Gutenberg-University, 55131, Mainz, Germany.,Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, 68159, Mannheim, Germany.,Institute for Microscopic Anatomy and Neurobiology, University Medical Center, Johannes Gutenberg-University, 55131, Mainz, Germany
| | - David Wolf
- Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, 68159, Mannheim, Germany
| | - Michael J Schmeisser
- Focus Program Translational Neurosciences (FTN), University Medical Center, Johannes Gutenberg-University, 55131, Mainz, Germany.,Institute for Microscopic Anatomy and Neurobiology, University Medical Center, Johannes Gutenberg-University, 55131, Mainz, Germany
| | - Wolfgang Kelsch
- Department of Psychiatry & Psychotherapy, University Medical Center, Johannes Gutenberg-University, 55131, Mainz, Germany. .,Focus Program Translational Neurosciences (FTN), University Medical Center, Johannes Gutenberg-University, 55131, Mainz, Germany. .,Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, 68159, Mannheim, Germany.
| |
Collapse
|
35
|
Kim HY, Um JW, Ko J. Proper synaptic adhesion signaling in the control of neural circuit architecture and brain function. Prog Neurobiol 2021; 200:101983. [PMID: 33422662 DOI: 10.1016/j.pneurobio.2020.101983] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 11/23/2020] [Accepted: 12/22/2020] [Indexed: 12/17/2022]
Abstract
Trans-synaptic cell-adhesion molecules are critical for governing various stages of synapse development and specifying neural circuit properties via the formation of multifarious signaling pathways. Recent studies have pinpointed the putative roles of trans-synaptic cell-adhesion molecules in mediating various cognitive functions. Here, we review the literature on the roles of a diverse group of central synaptic organizers, including neurexins (Nrxns), leukocyte common antigen-related receptor protein tyrosine phosphatases (LAR-RPTPs), and their associated binding proteins, in regulating properties of specific type of synapses and neural circuits. In addition, we highlight the findings that aberrant synaptic adhesion signaling leads to alterations in the structures, transmission, and plasticity of specific synapses across diverse brain areas. These results seem to suggest that proper trans-synaptic signaling pathways by Nrxns, LAR-RPTPs, and their interacting network is likely to constitute central molecular complexes that form the basis for cognitive functions, and that these complexes are heterogeneously and complexly disrupted in many neuropsychiatric and neurodevelopmental disorders.
Collapse
Affiliation(s)
- Hee Young Kim
- Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, 42988, South Korea
| | - Ji Won Um
- Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, 42988, South Korea; Core Protein Resources Center, DGIST, Daegu, 42988, South Korea.
| | - Jaewon Ko
- Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, 42988, South Korea.
| |
Collapse
|
36
|
Zimmerman AD, Munger SD. Olfactory subsystems associated with the necklace glomeruli in rodents. Cell Tissue Res 2021; 383:549-557. [PMID: 33404845 DOI: 10.1007/s00441-020-03388-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Accepted: 12/07/2020] [Indexed: 01/27/2023]
Abstract
The necklace glomeruli are a loosely defined group of glomeruli encircling the caudal main olfactory bulb in rodents. Initially defined by the expression of various immunohistochemical markers, they are now better understood in the context of the specialized chemosensory neurons of the main olfactory epithelium and Grueneberg ganglion that innervate them. It has become clear that the necklace region of the rodent main olfactory bulb is composed of multiple distinct groups of glomeruli, defined at least in part by their afferent inputs. In this review, we will explore the necklace glomeruli and the chemosensory neurons that innervate them.
Collapse
Affiliation(s)
- Arthur D Zimmerman
- Department of Pharmacology and Therapeutics, University of Florida College of Medicine, PO Box 100267, Gainesville, FL, 32610, USA
- Center for Smell and Taste, University of Florida, PO Box 100127, Gainesville, FL, 32610, USA
- Training Program in Chemosensory Science, University of Florida, PO Box 100127, Gainesville, FL, 32610, USA
| | - Steven D Munger
- Department of Pharmacology and Therapeutics, University of Florida College of Medicine, PO Box 100267, Gainesville, FL, 32610, USA.
- Center for Smell and Taste, University of Florida, PO Box 100127, Gainesville, FL, 32610, USA.
- Training Program in Chemosensory Science, University of Florida, PO Box 100127, Gainesville, FL, 32610, USA.
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, University of Florida College of Medicine, PO Box 100266, Gainesville, FL, 32610, USA.
| |
Collapse
|
37
|
Brunert D, Rothermel M. Extrinsic neuromodulation in the rodent olfactory bulb. Cell Tissue Res 2021; 383:507-524. [PMID: 33355709 PMCID: PMC7873007 DOI: 10.1007/s00441-020-03365-9] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Accepted: 11/24/2020] [Indexed: 12/16/2022]
Abstract
Evolutionarily, olfaction is one of the oldest senses and pivotal for an individual's health and survival. The olfactory bulb (OB), as the first olfactory relay station in the brain, is known to heavily process sensory information. To adapt to an animal's needs, OB activity can be influenced by many factors either from within (intrinsic neuromodulation) or outside (extrinsic neuromodulation) the OB which include neurotransmitters, neuromodulators, hormones, and neuropeptides. Extrinsic sources seem to be of special importance as the OB receives massive efferent input from numerous brain centers even outweighing the sensory input from the nose. Here, we review neuromodulatory processes in the rodent OB from such extrinsic sources. We will discuss extrinsic neuromodulation according to points of origin, receptors involved, affected circuits, and changes in behavior. In the end, we give a brief outlook on potential future directions in research on neuromodulation in the OB.
Collapse
Affiliation(s)
- Daniela Brunert
- Department of Chemosensation, AG Neuromodulation, Institute for Biology II, RWTH Aachen University, 52074, Aachen, Germany
| | - Markus Rothermel
- Department of Chemosensation, AG Neuromodulation, Institute for Biology II, RWTH Aachen University, 52074, Aachen, Germany.
| |
Collapse
|
38
|
Sticco MJ, Peña Palomino PA, Lukacsovich D, Thompson BL, Földy C, Ressl S, Martinelli DC. C1QL3 promotes cell-cell adhesion by mediating complex formation between ADGRB3/BAI3 and neuronal pentraxins. FASEB J 2021; 35:e21194. [PMID: 33337553 PMCID: PMC11565477 DOI: 10.1096/fj.202000351rr] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 10/28/2020] [Accepted: 10/30/2020] [Indexed: 11/11/2022]
Abstract
Synapses are the fundamental structural unit by which neurons communicate. An orchestra of proteins regulates diverse synaptic functions, including synapse formation, maintenance, and elimination-synapse homeostasis. Some proteins of the larger C1q super-family are synaptic organizers involved in crucial neuronal processes in various brain regions. C1Q-like (C1QL) proteins bind to the adhesion G protein-coupled receptor B3 (ADGRB3) and act at synapses in a subset of circuits. To investigate the hypothesis that the secreted C1QL proteins mediate tripartite trans-synaptic adhesion complexes, we conducted an in vivo interactome study and identified new binding candidates. We demonstrate that C1QL3 mediates a novel cell-cell adhesion complex involving ADGRB3 and two neuronal pentraxins, NPTX1 and NPTXR. Analysis of single-cell RNA-Seq data from the cerebral cortex shows that C1ql3, Nptx1, and Nptxr are highly co-expressed in the same excitatory neurons. Thus, our results suggest the possibility that in vivo the three co-expressed proteins are presynaptically secreted and form a complex capable of binding to postsynaptically localized ADGRB3, thereby creating a novel trans-synaptic adhesion complex. Identifying new binding partners for C1QL proteins and deciphering their underlying molecular principles will accelerate our understanding of their role in synapse organization.
Collapse
Affiliation(s)
- Matthew J. Sticco
- Department of Neuroscience, University of Connecticut Health, Farmington CT 06030 USA
| | - Perla A. Peña Palomino
- Department of Molecular and Cellular Biochemistry, Indiana University, Bloomington, IN 47405 USA
| | - David Lukacsovich
- Laboratory of Neural Connectivity, Brain Research Institute, Faculties of Medicine and Science, University of Zürich, Switzerland
| | - Brianna L. Thompson
- Department of Neuroscience, University of Connecticut Health, Farmington CT 06030 USA
| | - Csaba Földy
- Laboratory of Neural Connectivity, Brain Research Institute, Faculties of Medicine and Science, University of Zürich, Switzerland
| | - Susanne Ressl
- Department of Molecular and Cellular Biochemistry, Indiana University, Bloomington, IN 47405 USA
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford CA 94305, USA
- Department of Neuroscience, University of Austin Texas, Austin TX 78712, USA
| | - David C. Martinelli
- Department of Neuroscience, University of Connecticut Health, Farmington CT 06030 USA
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford CA 94305, USA
- The Connecticut Institute for the Brain and Cognitive Sciences (IBACS)
| |
Collapse
|
39
|
Nakahara TS, Carvalho VMA, Papes F. From Synapse to Supper: A Food Preference Recipe with Olfactory Synaptic Ingredients. Neuron 2020; 107:8-11. [PMID: 32645309 DOI: 10.1016/j.neuron.2020.06.016] [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: 10/23/2022]
Abstract
C1ql3 protein and its receptor Bai3 are involved in synaptic organization and function. In this issue of Neuron, Wang et al. (2020) report that both are essential for synaptic function between the anterior olfactory nucleus and the olfactory bulb and for the generation, but not recall, of associative olfactory memories determining food preference in mice.
Collapse
Affiliation(s)
- Thiago S Nakahara
- Department of Genetics, Evolution, Microbiology and Immunology, Institute of Biology, University of Campinas, Rua Monteiro Lobato, s/n, Campinas, Sao Paulo 13083-862, Brazil; Department of Biochemistry, Institute of Chemistry, University of Sao Paulo, Av. Prof. Lineu Prestes, 748, Sao Paulo 05508-000, Brazil
| | - Vinicius M A Carvalho
- Department of Genetics, Evolution, Microbiology and Immunology, Institute of Biology, University of Campinas, Rua Monteiro Lobato, s/n, Campinas, Sao Paulo 13083-862, Brazil
| | - Fabio Papes
- Department of Genetics, Evolution, Microbiology and Immunology, Institute of Biology, University of Campinas, Rua Monteiro Lobato, s/n, Campinas, Sao Paulo 13083-862, Brazil.
| |
Collapse
|
40
|
Dynamic Impairment of Olfactory Behavior and Signaling Mediated by an Olfactory Corticofugal System. J Neurosci 2020; 40:7269-7285. [PMID: 32817250 DOI: 10.1523/jneurosci.2667-19.2020] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2019] [Revised: 06/30/2020] [Accepted: 07/05/2020] [Indexed: 01/16/2023] Open
Abstract
Processing of olfactory information is modulated by centrifugal projections from cortical areas, yet their behavioral relevance and underlying neural mechanisms remain unclear in most cases. The anterior olfactory nucleus (AON) is part of the olfactory cortex, and its extensive connections to multiple upstream and downstream brain centers place it in a prime position to modulate early sensory information in the olfactory system. Here, we show that optogenetic activation of AON neurons in awake male and female mice was not perceived as an odorant equivalent cue. However, AON activation during odorant presentation reliably suppressed behavioral odor responses. This AON-mediated effect was fast and constant across odors and concentrations. Likewise, activation of glutamatergic AON projections to the olfactory bulb (OB) transiently inhibited the excitability of mitral/tufted cells (MTCs) that relay olfactory input to the cortex. Single-unit MTC recordings revealed that optogenetic activation of glutamatergic AON terminals in the OB transiently decreased sensory-evoked MTC spiking, regardless of the strength or polarity of the sensory response. The reduction in MTC firing during optogenetic stimulation was confirmed in recordings in awake mice. These findings suggest that glutamatergic AON projections to the OB impede early olfactory signaling by inhibiting OB output neurons, thereby dynamically gating sensory throughput to the cortex.SIGNIFICANCE STATEMENT The anterior olfactory nucleus (AON) as an olfactory information processing area sends extensive projections to multiple brain centers, but the behavioral consequences of its activation have been scarcely investigated. Using behavioral tests in combination with optogenetic manipulation, we show that, in contrast to what has been suggested previously, the AON does not seem to form odor percepts but instead suppresses behavioral odor responses across odorants and concentrations. Furthermore, this study shows that AON activation inhibits olfactory bulb output neurons in both anesthetized as well as awake mice, pointing to a potential mechanism by which the olfactory cortex can actively and dynamically gate sensory throughput to higher brain centers.
Collapse
|