1
|
Sheriff A, Zhou G, Sagar V, Morgenthaler JB, Cyr C, Hauner KK, Omidbeigi M, Rosenow JM, Schuele SU, Lane G, Zelano C. Breathing orchestrates synchronization of sleep oscillations in the human hippocampus. Proc Natl Acad Sci U S A 2024; 121:e2405395121. [PMID: 39680758 DOI: 10.1073/pnas.2405395121] [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: 03/18/2024] [Accepted: 11/07/2024] [Indexed: 12/18/2024] Open
Abstract
Nested sleep oscillations, emerging from asynchronous states in coordinated bursts, are critical for memory consolidation. Whether these bursts emerge intrinsically or from an underlying rhythm is unknown. Here, we show a previously undescribed respiratory-driven oscillation in the human hippocampus that couples with cardinal sleep oscillations. Further, breathing promotes nesting of ripples in slow oscillations, together suggesting that respiration acts as an intrinsic rhythm to coordinate synchronization of sleep oscillations, providing a unique framework to characterize sleep-related respiratory and memory processes.
Collapse
Affiliation(s)
- Andrew Sheriff
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
| | - Guangyu Zhou
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
| | - Vivek Sagar
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
| | - Justin B Morgenthaler
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
| | - Christopher Cyr
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
| | - Katherina K Hauner
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
- Department Medical Social Sciences, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
| | - Mahmoud Omidbeigi
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
| | - Joshua M Rosenow
- Department of Neurosurgery, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
| | - Stephan U Schuele
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
| | - Gregory Lane
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
| | - Christina Zelano
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
| |
Collapse
|
2
|
Fox AS, Shackman AJ. An Honest Reckoning With the Amygdala and Mental Illness. Am J Psychiatry 2024; 181:1059-1075. [PMID: 39616453 PMCID: PMC11611071 DOI: 10.1176/appi.ajp.20240941] [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] [Indexed: 12/18/2024]
Abstract
Anxiety disorders are a leading source of human misery, morbidity, and premature mortality. Existing treatments are far from curative for many, underscoring the need to clarify the underlying neural mechanisms. Although many brain regions contribute, the amygdala has received the most intense scientific attention. Over the past several decades, this scrutiny has yielded a detailed understanding of amygdala function, but it has failed to produce new clinical assays, biomarkers, or cures. Rising to this urgent public health challenge demands an honest reckoning with the functional-neuroanatomical complexity of the amygdala and a shift from theories anchored on "the amygdala" to models centered on specific amygdala nuclei and cell types. This review begins by examining evidence from studies of rodents, monkeys, and humans for the "canonical model," the idea that the amygdala plays a central role in fear- and anxiety-related states, traits, and disorders. Next, the authors selectively highlight work indicating that the canonical model, while true, is overly simplistic and fails to adequately capture the actual state of the evidentiary record, the breadth of amygdala-associated functions and illnesses, or the complexity of the amygdala's functional architecture. The authors describe the implications of these facts for basic and clinical neuroimaging research. The review concludes with some general recommendations for grappling with the complexity of the amygdala and accelerating efforts to understand and more effectively treat amygdala-related psychopathology.
Collapse
Affiliation(s)
- Andrew S. Fox
- Department of Psychology, University of California, Davis, CA 95616 USA
- California National Primate Research Center, University of California, Davis, CA 95616 USA
| | - Alexander J. Shackman
- Department of Psychology, University of Maryland, College Park, MD 20742 USA
- Department of Neuroscience and Cognitive Science Program, University of Maryland, College Park, MD 20742 USA
- Department of Maryland Neuroimaging Center, University of Maryland, College Park, MD 20742 USA
| |
Collapse
|
3
|
Deng XH, Liu XY, Wei YH, Wang K, Zhu JR, Zhong JJ, Zheng JY, Guo R, Zhu YF, Ye QH, Wang MD, Chen YJ, He JQ, Chen ZX, Huang SQ, Lv CS, Zheng GQ, Liu SF, Wen L. ErbB4 deficiency exacerbates olfactory dysfunction in an early-stage Alzheimer's disease mouse model. Acta Pharmacol Sin 2024; 45:2497-2512. [PMID: 38982150 PMCID: PMC11579518 DOI: 10.1038/s41401-024-01332-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Accepted: 06/02/2024] [Indexed: 07/11/2024] Open
Abstract
Olfactory dysfunction is increasingly recognized as an early indicator of Alzheimer's disease (AD). Aberrations in GABAergic function and the excitatory/inhibitory (E/I) balance within the olfactory bulb (OB) have been implicated in olfactory impairment during the initial stages of AD. While the neuregulin 1 (NRG1)/ErbB4 signaling pathway is known to regulate GABAergic transmission in the brain and is associated with various neuropsychiatric disorders, its specific role in early AD-related olfactory impairment remains incompletely understood. This study demonstrated that olfactory dysfunction preceded cognitive decline in young adult APP/PS1 mice and was characterized by reduced levels of NRG1 and ErbB4 in the OB. Further investigation revealed that deletion of ErbB4 in parvalbumin interneurons reduced GABAergic transmission and increased hyperexcitability in mitral and tufted cells (M/Ts) in the OB, thereby accelerating olfactory dysfunction in young adult APP/PS1 mice. Additionally, ErbB4 deficiency was associated with increased accumulation of Aβ and BACE1-mediated cleavage of APP, along with enhanced CDK5 signaling in the OB. NRG1 infusion into the OB was found to enhance GABAergic transmission in M/Ts and alleviate olfactory dysfunction in young adult APP/PS1 mice. These findings underscore the critical role of NRG1/ErbB4 signaling in regulating GABAergic transmission and E/I balance within the OB, contributing to olfactory impairment in young adult APP/PS1 mice, and provide novel insights for early intervention strategies in AD. This work has shown that ErbB4 deficiency increased the burden of Aβ, impaired GABAergic transmission, and disrupted the E/I balance of mitral and tufted cells (M/Ts) in the OB, ultimately resulting in olfactory dysfunction in young adult APP/PS1 mice. NRG1 could enhance GABAergic transmission, rescue E/I imbalance in M/Ts, and alleviate olfactory dysfunction in young adult APP/PS1 mice. OB: olfactory bulb, E/I: excitation/inhibition, Pr: probability of release, PV: parvalbumin interneurons, Aβ: β-amyloid, GABA: gamma-aminobutyric acid.
Collapse
Affiliation(s)
- Xian-Hua Deng
- Center for Brain Sciences, The First Affiliated Hospital of Xiamen University, State Key Laboratory of Cellular Stress Biology, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, 361000, China
- Department of Neurology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, 310006, China
| | - Xing-Yang Liu
- Center for Brain Sciences, The First Affiliated Hospital of Xiamen University, State Key Laboratory of Cellular Stress Biology, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, 361000, China
- Department of Neurology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, 310006, China
| | - Yi-Hua Wei
- Center for Brain Sciences, The First Affiliated Hospital of Xiamen University, State Key Laboratory of Cellular Stress Biology, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, 361000, China
- Department of Neurology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, 310006, China
| | - Ke Wang
- Center for Brain Sciences, The First Affiliated Hospital of Xiamen University, State Key Laboratory of Cellular Stress Biology, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, 361000, China
- Department of Neurology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, 310006, China
| | - Jun-Rong Zhu
- Center for Brain Sciences, The First Affiliated Hospital of Xiamen University, State Key Laboratory of Cellular Stress Biology, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, 361000, China
| | - Jia-Jun Zhong
- Center for Brain Sciences, The First Affiliated Hospital of Xiamen University, State Key Laboratory of Cellular Stress Biology, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, 361000, China
| | - Jing-Yuan Zheng
- Center for Brain Sciences, The First Affiliated Hospital of Xiamen University, State Key Laboratory of Cellular Stress Biology, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, 361000, China
| | - Rui Guo
- Center for Brain Sciences, The First Affiliated Hospital of Xiamen University, State Key Laboratory of Cellular Stress Biology, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, 361000, China
| | - Yi-Fan Zhu
- Center for Brain Sciences, The First Affiliated Hospital of Xiamen University, State Key Laboratory of Cellular Stress Biology, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, 361000, China
| | - Qiu-Hong Ye
- Center for Brain Sciences, The First Affiliated Hospital of Xiamen University, State Key Laboratory of Cellular Stress Biology, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, 361000, China
| | - Meng-Dan Wang
- Center for Brain Sciences, The First Affiliated Hospital of Xiamen University, State Key Laboratory of Cellular Stress Biology, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, 361000, China
| | - Ying-Jie Chen
- Center for Brain Sciences, The First Affiliated Hospital of Xiamen University, State Key Laboratory of Cellular Stress Biology, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, 361000, China
| | - Jian-Quan He
- Zhongshan Hospital, School of Medicine, Xiamen University, Xiamen, 361000, China
| | - Ze-Xu Chen
- Center for Brain Sciences, The First Affiliated Hospital of Xiamen University, State Key Laboratory of Cellular Stress Biology, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, 361000, China
| | - Shu-Qiong Huang
- Center for Brain Sciences, The First Affiliated Hospital of Xiamen University, State Key Laboratory of Cellular Stress Biology, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, 361000, China
| | - Chong-Shan Lv
- Center for Brain Sciences, The First Affiliated Hospital of Xiamen University, State Key Laboratory of Cellular Stress Biology, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, 361000, China
| | - Guo-Qing Zheng
- Department of Neurology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, 310006, China.
| | - Sui-Feng Liu
- Zhongshan Hospital, School of Medicine, Xiamen University, Xiamen, 361000, China.
| | - Lei Wen
- Center for Brain Sciences, The First Affiliated Hospital of Xiamen University, State Key Laboratory of Cellular Stress Biology, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, 361000, China.
| |
Collapse
|
4
|
Mercado E, Zhuo J. Do rodents smell with sound? Neurosci Biobehav Rev 2024; 167:105908. [PMID: 39343078 DOI: 10.1016/j.neubiorev.2024.105908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 09/09/2024] [Accepted: 09/24/2024] [Indexed: 10/01/2024]
Abstract
Chemosensation via olfaction is a critical process underlying social interactions in many different species. Past studies of olfaction in mammals often have focused on its mechanisms in isolation from other systems, limiting the generalizability of findings from olfactory research to perceptual processes in other modalities. Studies of chemical communication, in particular, have progressed independently of research on vocal behavior and acoustic communication. Those bioacousticians who have considered how sound production and reception might interact with olfaction often portray odors as cues to the kinds of vocalizations that might be functionally useful. In the olfaction literature, vocalizations are rarely mentioned. Here, we propose that ultrasonic vocalizations may affect what rodents smell by altering the deposition of inhaled particles and that rodents coordinate active sniffing with sound production specifically to enhance reception of pheromones. In this scenario, rodent vocalizations may contribute to a unique mode of active olfactory sensing, in addition to whatever roles they serve as social signals. Consideration of this hypothesis highlights the perceptual advantages that parallel coordination of multiple sensorimotor processes may provide to individuals exploring novel situations and environments, especially those involving dynamic social interactions.
Collapse
Affiliation(s)
- Eduardo Mercado
- University at Buffalo, The State University of New York, USA.
| | | |
Collapse
|
5
|
Saad MZH, Ryan V WG, Edwards CA, Szymanski BN, Marri AR, Jerow LG, McCullumsmith R, Bamber BA. Olfactory combinatorial coding supports risk-reward decision making in C. elegans. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.19.599745. [PMID: 39484578 PMCID: PMC11526860 DOI: 10.1101/2024.06.19.599745] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
Olfactory-driven behaviors are essential for animal survival, but mechanisms for decoding olfactory inputs remain poorly understood. We have used whole-network Ca ++ imaging to study olfactory coding in Caenorhabditis elegans. We show that the odorant 1-octanol is encoded combinatorially in the periphery as both an attractant and a repellant. These inputs are integrated centrally, and their relative strengths determine the sensitivity and valence of the behavioral response through modulation of locomotory reversals and speed. The balance of these pathways also dictates the activity of the locomotory command interneurons, which control locomotory reversals. This balance serves as a regulatory node for response modulation, allowing C. elegans to weigh opportunities and hazards in its environment when formulating behavioral responses. Thus, an odorant can be encoded simultaneously as inputs of opposite valence, focusing attention on the integration of these inputs in determining perception, response, and plasticity.
Collapse
|
6
|
Wang P, Li S, Li A. Odor representation and coding by the mitral/tufted cells in the olfactory bulb. J Zhejiang Univ Sci B 2024; 25:824-840. [PMID: 39420520 PMCID: PMC11494158 DOI: 10.1631/jzus.b2400051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Accepted: 07/14/2024] [Indexed: 10/19/2024]
Abstract
The olfactory bulb (OB) is the first relay station in the olfactory system and functions as a crucial hub. It can represent odor information precisely and accurately in an ever-changing environment. As the only output neurons in the OB, mitral/tufted cells encode information such as odor identity and concentration. Recently, the neural strategies and mechanisms underlying odor representation and encoding in the OB have been investigated extensively. Here we review the main progress on this topic. We first review the neurons and circuits involved in odor representation, including the different cell types in the OB and the neural circuits within and beyond the OB. We will then discuss how two different coding strategies-spatial coding and temporal coding-work in the rodent OB. Finally, we discuss potential future directions for this research topic. Overall, this review provides a comprehensive description of our current understanding of how odor information is represented and encoded by mitral/tufted cells in the OB.
Collapse
Affiliation(s)
- Panke Wang
- School of Biomedical Engineering, Guangdong Medical University, Dongguan 523808, China
| | - Shan Li
- Jiangsu Key Laboratory of Brain Disease and Bioinformation, Research Center for Biochemistry and Molecular Biology, Xuzhou Medical University, Xuzhou 221002, China
| | - An'an Li
- Jiangsu Key Laboratory of Brain Disease and Bioinformation, Research Center for Biochemistry and Molecular Biology, Xuzhou Medical University, Xuzhou 221002, China.
| |
Collapse
|
7
|
Rokni D, Ben-Shaul Y. Object-oriented olfaction: challenges for chemosensation and for chemosensory research. Trends Neurosci 2024; 47:834-848. [PMID: 39245626 DOI: 10.1016/j.tins.2024.08.008] [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: 04/10/2024] [Revised: 08/02/2024] [Accepted: 08/16/2024] [Indexed: 09/10/2024]
Abstract
Many animal species use olfaction to extract information about objects in their environment. Yet, the specific molecular signature that any given object emits varies due to various factors. Here, we detail why such variability makes chemosensory-mediated object recognition such a hard problem, and we propose that a major function of the elaborate chemosensory network is to overcome it. We describe previous work addressing different elements of the problem and outline future research directions that we consider essential for a full understanding of object-oriented olfaction. In particular, we call for extensive representation of olfactory object variability in chemical, behavioral, and electrophysiological analyses. While written with an emphasis on macrosmatic mammalian species, our arguments apply to all organisms that employ chemosensation to navigate complex environments.
Collapse
Affiliation(s)
- Dan Rokni
- Department of Medical Neurobiology, The Hebrew University Faculty of Medicine, Institute for Medical Research, Israel-Canada (IMRIC), Jerusalem, Israel.
| | - Yoram Ben-Shaul
- Department of Medical Neurobiology, The Hebrew University Faculty of Medicine, Institute for Medical Research, Israel-Canada (IMRIC), Jerusalem, Israel.
| |
Collapse
|
8
|
Wang P, Xiang J, Niu Y, Wei J, Lan C, Li X, Xu L, Yin Y, Wang H, Zhang T, Yang L, Xing H, Fan S, Niu Q, Kang H, Liang Y. Study of a precise treatment protocol for patients with consciousness disorders based on the brain network analysis of functional magnetic resonance imaging. Front Neurosci 2024; 18:1443478. [PMID: 39351395 PMCID: PMC11439825 DOI: 10.3389/fnins.2024.1443478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 08/19/2024] [Indexed: 10/04/2024] Open
Abstract
Objective How to conduct objective and accurate individualized assessments of patients with disorders of consciousness (DOC) and carry out precision rehabilitation treatment technology is a major rehabilitation problem that needs to be solved urgently. Methods In this study, a multi-layer brain network was constructed based on functional magnetic resonance imaging (fMRI) to analyze the structural and functional brain networks of patients with DOC at different levels and to find regulatory targets (imaging markers) with recovery potential for DOC. Then repeated transcranial magnetic stimulation (rTMS) was performed in DOC patients to clinically validate. Results The brain network connectivity of DOC patients with different consciousness states is different, and the most obvious brain regions appeared in the olfactory cortex and precuneus. rTMS stimulation could effectively improve the consciousness level of DOC patients and stimulate the occipital lobe (specific regions found in this study) and the dorsolateral prefrontal cortex (DLPFC), and both parts had a good consciousness recovery effect. Conclusion In clinical work, personalized stimulation regimen treatment combined with the brain network characteristics of DOC patients can improve the treatment effect.
Collapse
Affiliation(s)
- Pingzhi Wang
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Tongji Shanxi Hospital, Shanxi Academy of Medical Sciences, Taiyuan, China
| | - Jie Xiang
- College of Computer Science and Technology (College of Data Science), Taiyuan University of Technology, Taiyuan, China
| | - Yan Niu
- College of Computer Science and Technology (College of Data Science), Taiyuan University of Technology, Taiyuan, China
| | - Jing Wei
- College of Computer Science and Technology (College of Data Science), Taiyuan University of Technology, Taiyuan, China
| | - Caiqin Lan
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Tongji Shanxi Hospital, Shanxi Academy of Medical Sciences, Taiyuan, China
| | - Xiangping Li
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Tongji Shanxi Hospital, Shanxi Academy of Medical Sciences, Taiyuan, China
| | - Liying Xu
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Tongji Shanxi Hospital, Shanxi Academy of Medical Sciences, Taiyuan, China
| | - Yajie Yin
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Tongji Shanxi Hospital, Shanxi Academy of Medical Sciences, Taiyuan, China
| | - Hongxiong Wang
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Tongji Shanxi Hospital, Shanxi Academy of Medical Sciences, Taiyuan, China
| | - Tao Zhang
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Tongji Shanxi Hospital, Shanxi Academy of Medical Sciences, Taiyuan, China
| | - Lei Yang
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Tongji Shanxi Hospital, Shanxi Academy of Medical Sciences, Taiyuan, China
| | - Hao Xing
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Tongji Shanxi Hospital, Shanxi Academy of Medical Sciences, Taiyuan, China
| | - Shasha Fan
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Tongji Shanxi Hospital, Shanxi Academy of Medical Sciences, Taiyuan, China
| | - Qing Niu
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Tongji Shanxi Hospital, Shanxi Academy of Medical Sciences, Taiyuan, China
| | - Huicong Kang
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Tongji Shanxi Hospital, Shanxi Academy of Medical Sciences, Taiyuan, China
- Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Ying Liang
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Tongji Shanxi Hospital, Shanxi Academy of Medical Sciences, Taiyuan, China
| |
Collapse
|
9
|
Mori K, Sakano H. One respiratory cycle as a minimum time unit for making behavioral decisions in the mammalian olfactory system. Front Neurosci 2024; 18:1423694. [PMID: 39315076 PMCID: PMC11417025 DOI: 10.3389/fnins.2024.1423694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Accepted: 08/26/2024] [Indexed: 09/25/2024] Open
Abstract
Voluntary behaviors such as sniffing, moving, and eating require decision-making accompanied by intentional respiration. Based on the study of respiration-coherent activity of rodent olfactory networks, we infer that during the inhalation phase of respiration, olfactory cortical areas process environmental odor information and transmit it to the higher multisensory cognitive areas via feedforward pathways to comprehensively evaluate the surrounding situation. We also infer that during the exhalation phase, the higher multisensory areas generate cognitive-signals and transmit them not only to the behavioral output system but also back to the olfactory cortical areas. We presume that the cortical mechanism couples the intentional respiration with the voluntary behaviors. Thus, in one respiratory cycle, the mammalian brain may transmit and process sensory information to cognize and evaluate the multisensory image of the external world, leading to one behavioral decision and one emotional expression. In this perspective article, we propose that one respiratory cycle provides a minimum time unit for decision making during wakefulness.
Collapse
Affiliation(s)
- Kensaku Mori
- RIKEN Center for Brain Science, Wako, Saitama, Japan
| | - Hitoshi Sakano
- Department of Brain Function, School of Medical Sciences, University of Fukui, Matsuoka, Japan
| |
Collapse
|
10
|
Tsuboi A. A specific olfactory bulb interneuron subtype Tpbg/5T4 generated at embryonic and neonatal stages. Front Neural Circuits 2024; 18:1427378. [PMID: 38933598 PMCID: PMC11203798 DOI: 10.3389/fncir.2024.1427378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Accepted: 05/24/2024] [Indexed: 06/28/2024] Open
Abstract
Various mammals have shown that sensory stimulation plays a crucial role in regulating the development of diverse structures, such as the olfactory bulb (OB), cerebral cortex, hippocampus, and retina. In the OB, the dendritic development of excitatory projection neurons like mitral/tufted cells is influenced by olfactory experiences. Odor stimulation is also essential for the dendritic development of inhibitory OB interneurons, such as granule and periglomerular cells, which are continuously produced in the ventricular-subventricular zone throughout life. Based on the morphological and molecular features, OB interneurons are classified into several subtypes. The role for each interneuron subtype in the control of olfactory behavior remains poorly understood due to lack of each specific marker. Among the several OB interneuron subtypes, a specific granule cell subtype, which expresses the oncofetal trophoblast glycoprotein (Tpbg or 5T4) gene, has been reported to be required for odor detection and discrimination behavior. This review will primarily focus on elucidating the contribution of different granule cell subtypes, including the Tpbg/5T4 subtype, to olfactory processing and behavior during the embryonic and adult stages.
Collapse
Affiliation(s)
- Akio Tsuboi
- Graduate School of Pharmaceutical Sciences, Osaka University, Toyonaka, Japan
| |
Collapse
|
11
|
Kikuta S, Nagayama S, Hasegawa-Ishii S. Structures and functions of the normal and injured human olfactory epithelium. Front Neural Circuits 2024; 18:1406218. [PMID: 38903957 PMCID: PMC11188711 DOI: 10.3389/fncir.2024.1406218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2024] [Accepted: 05/20/2024] [Indexed: 06/22/2024] Open
Abstract
The olfactory epithelium (OE) is directly exposed to environmental agents entering the nasal cavity, leaving OSNs prone to injury and degeneration. The causes of olfactory dysfunction are diverse and include head trauma, neurodegenerative diseases, and aging, but the main causes are chronic rhinosinusitis (CRS) and viral infections. In CRS and viral infections, reduced airflow due to local inflammation, inflammatory cytokine production, release of degranulated proteins from eosinophils, and cell injury lead to decreased olfactory function. It is well known that injury-induced loss of mature OSNs in the adult OE causes massive regeneration of new OSNs within a few months through the proliferation and differentiation of progenitor basal cells that are subsequently incorporated into olfactory neural circuits. Although normal olfactory function returns after injury in most cases, prolonged olfactory impairment and lack of improvement in olfactory function in some cases poses a major clinical problem. Persistent inflammation or severe injury in the OE results in morphological changes in the OE and respiratory epithelium and decreases the number of mature OSNs, resulting in irreversible loss of olfactory function. In this review, we discuss the histological structure and distribution of the human OE, and the pathogenesis of olfactory dysfunction associated with CRS and viral infection.
Collapse
Affiliation(s)
- Shu Kikuta
- Department of Otolaryngology-Head and Neck Surgery, Faculty of Medicine, Nihon University, Tokyo, Japan
| | - Shin Nagayama
- Department of Neurobiology and Anatomy, McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, TX, United States
| | | |
Collapse
|
12
|
Kim S, Choi T, Bang E. Investigation of odor pollution by utilizing selected ion flow tube mass spectrometry (SIFT-MS) and principal component analysis (PCA). ENVIRONMENTAL MONITORING AND ASSESSMENT 2024; 196:550. [PMID: 38743156 DOI: 10.1007/s10661-024-12708-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Accepted: 05/04/2024] [Indexed: 05/16/2024]
Abstract
Odor pollution, also referred to as odor nuisance, is a growing environmental concern that is significantly associated with mental health. Once emitted into the air, the concentration of odorous substances varies considerably with wind conditions, leading to difficulties in timely sampling. In the present study, we employed selected ion flow tube mass spectrometry (SIFT-MS) to measure 22 odor-producing molecules continuously in an urban-rural complex city. In addition, we applied statistical analyses, principal component analysis (PCA), and a conditional probability function (CPF) to the datasets obtained from SIFT-MS to identify the odor characteristics at two study sites. At site A, odorants related to livestock farming and industry showed high factor loadings on principal components (PCs) from the PCA. In contrast, we estimated that the odorous gaseous chemicals affecting site B were closely related to sewage treatment and municipal solid waste disposal. Similar CPF patterns of grouped substances from the PCA supported the association between potential odor sources and specific odorants at site B, which helped estimate possible source locations. Consequently, our findings indicate that continuous monitoring of odorous substances using SIFT-MS can be an effective way to provide sufficient information on odor-producing molecules, leading to the clear identification of odor characteristics despite the high variability of odorous substances.
Collapse
Affiliation(s)
- Sangcheol Kim
- Sejong Institute of Health and Environment, Twelve Seobukbu 2-Ro, Jochiwon-Eup, Sejong, 30015, Republic of Korea.
| | - Taeryeong Choi
- Sejong Institute of Health and Environment, Twelve Seobukbu 2-Ro, Jochiwon-Eup, Sejong, 30015, Republic of Korea
| | - Eunok Bang
- Sejong Institute of Health and Environment, Twelve Seobukbu 2-Ro, Jochiwon-Eup, Sejong, 30015, Republic of Korea
| |
Collapse
|
13
|
Yang K, Hasegawa Y, Bhattarai JP, Hua J, Dower M, Etyemez S, Prasad N, Duvall L, Paez A, Smith A, Wang Y, Zhang YF, Lane AP, Ishizuka K, Kamath V, Ma M, Kamiya A, Sawa A. Inflammation-related pathology in the olfactory epithelium: its impact on the olfactory system in psychotic disorders. Mol Psychiatry 2024; 29:1453-1464. [PMID: 38321120 PMCID: PMC11189720 DOI: 10.1038/s41380-024-02425-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 01/03/2024] [Accepted: 01/08/2024] [Indexed: 02/08/2024]
Abstract
Smell deficits and neurobiological changes in the olfactory bulb (OB) and olfactory epithelium (OE) have been observed in schizophrenia and related disorders. The OE is the most peripheral olfactory system located outside the cranium, and is connected with the brain via direct neuronal projections to the OB. Nevertheless, it is unknown whether and how a disturbance of the OE affects the OB in schizophrenia and related disorders. Addressing this gap would be the first step in studying the impact of OE pathology in the disease pathophysiology in the brain. In this cross-species study, we observed that chronic, local OE inflammation with a set of upregulated genes in an inducible olfactory inflammation (IOI) mouse model led to a volume reduction, layer structure changes, and alterations of neuron functionality in the OB. Furthermore, IOI model also displayed behavioral deficits relevant to negative symptoms (avolition) in parallel to smell deficits. In first episode psychosis (FEP) patients, we observed a significant alteration in immune/inflammation-related molecular signatures in olfactory neuronal cells (ONCs) enriched from biopsied OE and a significant reduction in the OB volume, compared with those of healthy controls (HC). The increased expression of immune/inflammation-related molecules in ONCs was significantly correlated to the OB volume reduction in FEP patients, but no correlation was found in HCs. Moreover, the increased expression of human orthologues of the IOI genes in ONCs was significantly correlated with the OB volume reduction in FEP, but not in HCs. Together, our study implies a potential mechanism of the OE-OB pathology in patients with psychotic disorders (schizophrenia and related disorders). We hope that this mechanism may have a cross-disease implication, including COVID-19-elicited mental conditions that include smell deficits.
Collapse
Affiliation(s)
- Kun Yang
- Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Yuto Hasegawa
- Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Janardhan P Bhattarai
- Department of Neuroscience, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Jun Hua
- Department of Radiology and Radiological Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, MD, USA
| | - Milan Dower
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Semra Etyemez
- Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Neal Prasad
- Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Lauren Duvall
- Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Adrian Paez
- F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, MD, USA
| | - Amy Smith
- Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Yingqi Wang
- Department of Neuroscience, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Yun-Feng Zhang
- Department of Neuroscience, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Andrew P Lane
- Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Koko Ishizuka
- Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Vidyulata Kamath
- Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Minghong Ma
- Department of Neuroscience, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Atsushi Kamiya
- Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | - Akira Sawa
- Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Department of Pharmacology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA.
| |
Collapse
|
14
|
Fang S, Luo Z, Wei Z, Qin Y, Zheng J, Zhang H, Jin J, Li J, Miao C, Yang S, Li Y, Liang Z, Yu XD, Zhang XM, Xiong W, Zhu H, Gan WB, Huang L, Li B. Sexually dimorphic control of affective state processing and empathic behaviors. Neuron 2024; 112:1498-1517.e8. [PMID: 38430912 DOI: 10.1016/j.neuron.2024.02.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Revised: 12/08/2023] [Accepted: 02/01/2024] [Indexed: 03/05/2024]
Abstract
Recognizing the affective states of social counterparts and responding appropriately fosters successful social interactions. However, little is known about how the affective states are expressed and perceived and how they influence social decisions. Here, we show that male and female mice emit distinct olfactory cues after experiencing distress. These cues activate distinct neural circuits in the piriform cortex (PiC) and evoke sexually dimorphic empathic behaviors in observers. Specifically, the PiC → PrL pathway is activated in female observers, inducing a social preference for the distressed counterpart. Conversely, the PiC → MeA pathway is activated in male observers, evoking excessive self-grooming behaviors. These pathways originate from non-overlapping PiC neuron populations with distinct gene expression signatures regulated by transcription factors and sex hormones. Our study unveils how internal states of social counterparts are processed through sexually dimorphic mechanisms at the molecular, cellular, and circuit levels and offers insights into the neural mechanisms underpinning sex differences in higher brain functions.
Collapse
Affiliation(s)
- Shunchang Fang
- Neuroscience Program, Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine and the Fifth Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China
| | - Zhengyi Luo
- Neuroscience Program, Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine and the Fifth Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China
| | - Zicheng Wei
- Neuroscience Program, Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine and the Fifth Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China
| | - Yuxin Qin
- Neuroscience Program, Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine and the Fifth Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China
| | - Jieyan Zheng
- Neuroscience Program, Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine and the Fifth Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China
| | - Hongyang Zhang
- Neuroscience Program, Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine and the Fifth Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China
| | - Jianhua Jin
- Neuroscience Program, Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine and the Fifth Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China
| | - Jiali Li
- Neuroscience Program, Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine and the Fifth Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China
| | - Chenjian Miao
- Institute on Aging, Hefei, China and Brain Disorders, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China
| | - Shana Yang
- Neuroscience Program, Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine and the Fifth Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China
| | - Yonglin Li
- Neuroscience Program, Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine and the Fifth Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China
| | - Zirui Liang
- Neuroscience Program, Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine and the Fifth Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China
| | - Xiao-Dan Yu
- Neuroscience Program, Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine and the Fifth Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China
| | - Xiao Min Zhang
- Neuroscience Program, Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine and the Fifth Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China
| | - Wei Xiong
- Institute on Aging, Hefei, China and Brain Disorders, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China
| | - Hongying Zhu
- Institute on Aging, Hefei, China and Brain Disorders, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China
| | | | - Lianyan Huang
- Neuroscience Program, Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine and the Fifth Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China; Key Laboratory of Human Microbiome and Chronic Diseases (Sun Yat-Sen University), Ministry of Education, Guangzhou 510655, China.
| | - Boxing Li
- Neuroscience Program, Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine and the Fifth Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China; Advanced Medical Technology Center, the First Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou 510080, China; Key Laboratory of Human Microbiome and Chronic Diseases (Sun Yat-Sen University), Ministry of Education, Guangzhou 510655, China.
| |
Collapse
|
15
|
Hirata T. Olfactory information processing viewed through mitral and tufted cell-specific channels. Front Neural Circuits 2024; 18:1382626. [PMID: 38523698 PMCID: PMC10957668 DOI: 10.3389/fncir.2024.1382626] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Accepted: 02/29/2024] [Indexed: 03/26/2024] Open
Abstract
Parallel processing is a fundamental strategy of sensory coding. Through this processing, unique and distinct features of sensations are computed and projected to the central targets. This review proposes that mitral and tufted cells, which are the second-order projection neurons in the olfactory bulb, contribute to parallel processing within the olfactory system. Based on anatomical and functional evidence, I discuss potential features that could be conveyed through the unique channel formed by these neurons.
Collapse
Affiliation(s)
- Tatsumi Hirata
- Brain Function Laboratory, National Institute of Genetics, SOKENDAI, Mishima, Japan
| |
Collapse
|
16
|
Wang H, Wang Q, Cui L, Feng X, Dong P, Tan L, Lin L, Lian H, Cao S, Huang H, Cao P, Li XM. A molecularly defined amygdala-independent tetra-synaptic forebrain-to-hindbrain pathway for odor-driven innate fear and anxiety. Nat Neurosci 2024; 27:514-526. [PMID: 38347199 DOI: 10.1038/s41593-023-01562-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Accepted: 12/14/2023] [Indexed: 03/08/2024]
Abstract
Fear-related disorders (for example, phobias and anxiety) cause a substantial public health problem. To date, studies of the neural basis of fear have mostly focused on the amygdala. Here we identify a molecularly defined amygdala-independent tetra-synaptic pathway for olfaction-evoked innate fear and anxiety in male mice. This pathway starts with inputs from the olfactory bulb mitral and tufted cells to pyramidal neurons in the dorsal peduncular cortex that in turn connect to cholecystokinin-expressing (Cck+) neurons in the superior part of lateral parabrachial nucleus, which project to tachykinin 1-expressing (Tac1+) neurons in the parasubthalamic nucleus. Notably, the identified pathway is specifically involved in odor-driven innate fear. Selective activation of this pathway induces innate fear, while its inhibition suppresses odor-driven innate fear. In addition, the pathway is both necessary and sufficient for stress-induced anxiety-like behaviors. These findings reveal a forebrain-to-hindbrain neural substrate for sensory-triggered fear and anxiety that bypasses the amygdala.
Collapse
Affiliation(s)
- Hao Wang
- Department of Neurobiology and Department of Neurology of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Affiliated Mental Health Center and Hangzhou Seventh People's Hospital, Zhejiang University School of Medicine / Nanhu Brain-computer Interface Institute, Hangzhou, China
| | - Qin Wang
- Department of Neurobiology and Department of Neurology of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Liuzhe Cui
- Department of Neurobiology and Department of Neurology of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaoyang Feng
- Department of Neurobiology and Department of Neurology of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Ping Dong
- Department of Neurobiology and Department of Neurology of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Liheng Tan
- Department of Neurobiology and Department of Neurology of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Lin Lin
- Department of Neurobiology and Department of Neurology of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Hong Lian
- Department of Neurobiology and Department of Neurology of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Shuxia Cao
- Department of Neurobiology and Department of Neurology of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Huiqian Huang
- Department of Neurobiology and Department of Neurology of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Peng Cao
- National Institute of Biological Sciences, Beijing, China
- Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing, China
| | - Xiao-Ming Li
- Department of Neurobiology and Department of Neurology of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
- National Health Commission and Chinese Academy of Medical Sciences Key Laboratory of Medical Neurobiology, Ministry of Education Frontier Center of Brain Science and Brain-machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, China.
- Center for Brain Science and Brain-Inspired Intelligence, Research Units for Emotion and Emotion Disorders, Chinese Academy of Medical Sciences / Nanhu Brain-Computer Interface Institute, Hangzhou, China.
| |
Collapse
|
17
|
Mori K, Sakano H. Circuit formation and sensory perception in the mouse olfactory system. Front Neural Circuits 2024; 18:1342576. [PMID: 38434487 PMCID: PMC10904487 DOI: 10.3389/fncir.2024.1342576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Accepted: 02/01/2024] [Indexed: 03/05/2024] Open
Abstract
In the mouse olfactory system, odor information is converted to a topographic map of activated glomeruli in the olfactory bulb (OB). Although the arrangement of glomeruli is genetically determined, the glomerular structure is plastic and can be modified by environmental stimuli. If the pups are exposed to a particular odorant, responding glomeruli become larger recruiting the dendrites of connecting projection neurons and interneurons. This imprinting not only increases the sensitivity to the exposed odor, but also imposes the positive quality on imprinted memory. External odor information represented as an odor map in the OB is transmitted to the olfactory cortex (OC) and amygdala for decision making to elicit emotional and behavioral outputs using two distinct neural pathways, innate and learned. Innate olfactory circuits start to work right after birth, whereas learned circuits become functional later on. In this paper, the recent progress will be summarized in the study of olfactory circuit formation and odor perception in mice. We will also propose new hypotheses on the timing and gating of olfactory circuit activity in relation to the respiration cycle.
Collapse
Affiliation(s)
| | - Hitoshi Sakano
- Department of Brain Function, School of Medical Sciences, University of Fukui, Matsuoka, Japan
| |
Collapse
|
18
|
Liu D, Lu J, Wei L, Yao M, Yang H, Lv P, Wang H, Zhu Y, Zhu Z, Zhang X, Chen J, Yang QX, Zhang B. Olfactory deficit: a potential functional marker across the Alzheimer's disease continuum. Front Neurosci 2024; 18:1309482. [PMID: 38435057 PMCID: PMC10907997 DOI: 10.3389/fnins.2024.1309482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Accepted: 02/02/2024] [Indexed: 03/05/2024] Open
Abstract
Alzheimer's disease (AD) is a prevalent form of dementia that affects an estimated 32 million individuals globally. Identifying early indicators is vital for screening at-risk populations and implementing timely interventions. At present, there is an urgent need for early and sensitive biomarkers to screen individuals at risk of AD. Among all sensory biomarkers, olfaction is currently one of the most promising indicators for AD. Olfactory dysfunction signifies a decline in the ability to detect, identify, or remember odors. Within the spectrum of AD, impairment in olfactory identification precedes detectable cognitive impairments, including mild cognitive impairment (MCI) and even the stage of subjective cognitive decline (SCD), by several years. Olfactory impairment is closely linked to the clinical symptoms and neuropathological biomarkers of AD, accompanied by significant structural and functional abnormalities in the brain. Olfactory behavior examination can subjectively evaluate the abilities of olfactory identification, threshold, and discrimination. Olfactory functional magnetic resonance imaging (fMRI) can provide a relatively objective assessment of olfactory capabilities, with the potential to become a promising tool for exploring the neural mechanisms of olfactory damage in AD. Here, we provide a timely review of recent literature on the characteristics, neuropathology, and examination of olfactory dysfunction in the AD continuum. We focus on the early changes in olfactory indicators detected by behavioral and fMRI assessments and discuss the potential of these techniques in MCI and preclinical AD. Despite the challenges and limitations of existing research, olfactory dysfunction has demonstrated its value in assessing neurodegenerative diseases and may serve as an early indicator of AD in the future.
Collapse
Affiliation(s)
- Dongming Liu
- Department of Radiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- Institute of Medical Imaging and Artificial Intelligence, Nanjing University, Nanjing, China
- Medical Imaging Center, The Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
| | - Jiaming Lu
- Department of Radiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- Institute of Medical Imaging and Artificial Intelligence, Nanjing University, Nanjing, China
- Medical Imaging Center, The Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
| | - Liangpeng Wei
- Department of Radiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- Institute of Medical Imaging and Artificial Intelligence, Nanjing University, Nanjing, China
- Medical Imaging Center, The Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
| | - Mei Yao
- Department of Radiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- Institute of Medical Imaging and Artificial Intelligence, Nanjing University, Nanjing, China
- Medical Imaging Center, The Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
| | - Huiquan Yang
- Department of Radiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- Institute of Medical Imaging and Artificial Intelligence, Nanjing University, Nanjing, China
- Medical Imaging Center, The Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
| | - Pin Lv
- Department of Radiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- Institute of Medical Imaging and Artificial Intelligence, Nanjing University, Nanjing, China
- Medical Imaging Center, The Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
| | - Haoyao Wang
- Department of Radiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- Institute of Medical Imaging and Artificial Intelligence, Nanjing University, Nanjing, China
- Medical Imaging Center, The Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
| | - Yajing Zhu
- Department of Radiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Zhengyang Zhu
- Department of Radiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Xin Zhang
- Department of Radiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- Institute of Medical Imaging and Artificial Intelligence, Nanjing University, Nanjing, China
- Medical Imaging Center, The Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
| | - Jiu Chen
- Department of Radiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- Institute of Medical Imaging and Artificial Intelligence, Nanjing University, Nanjing, China
- Medical Imaging Center, The Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
| | - Qing X. Yang
- Department of Radiology, Center for NMR Research, Penn State University College of Medicine, Hershey, PA, United States
| | - Bing Zhang
- Department of Radiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- Institute of Medical Imaging and Artificial Intelligence, Nanjing University, Nanjing, China
- Medical Imaging Center, The Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, China
- Jiangsu Key Laboratory of Molecular Medicine, Nanjing, China
- Institute of Brain Science, Nanjing University, Nanjing, China
- Jiangsu Provincial Medical Key Discipline (Laboratory), Nanjing, China
| |
Collapse
|
19
|
Fang A, Yu CR. Activity-dependent formation of the topographic map and the critical period in the development of mammalian olfactory system. Genesis 2024; 62:e23586. [PMID: 38593162 PMCID: PMC11003738 DOI: 10.1002/dvg.23586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 01/03/2024] [Accepted: 01/05/2024] [Indexed: 04/11/2024]
Abstract
Neural activity influences every aspect of nervous system development. In olfactory systems, sensory neurons expressing the same odorant receptor project their axons to stereotypically positioned glomeruli, forming a spatial map of odorant receptors in the olfactory bulb. As individual odors activate unique combinations of glomeruli, this map forms the basis for encoding olfactory information. The establishment of this stereotypical olfactory map requires coordinated regulation of axon guidance molecules instructed by spontaneous activity. Recent studies show that sensory experiences also modify innervation patterns in the olfactory bulb, especially during a critical period of the olfactory system development. This review examines evidence in the field to suggest potential mechanisms by which various aspects of neural activity regulate axon targeting. We also discuss the precise functions served by neural plasticity during the critical period.
Collapse
Affiliation(s)
- Ai Fang
- Stowers Institute for Medical Research, Kansas City, MO 64110, USA
| | - C. Ron Yu
- Stowers Institute for Medical Research, Kansas City, MO 64110, USA
- Department of Cell Biology and Physiology, University of Kansas Medical Center, 3901 Rainbow Boulevard, Kansas City, KS 66160, USA
| |
Collapse
|
20
|
Gao K, Hu M, Li J, Li Z, Xu W, Qian Z, Gao F, Ma T. Drug-detecting bioelectronic nose based on odor cue memory combined with a brain computer interface. Biosens Bioelectron 2024; 244:115797. [PMID: 37922809 DOI: 10.1016/j.bios.2023.115797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 10/12/2023] [Accepted: 10/26/2023] [Indexed: 11/07/2023]
Abstract
The international drug situation is increasingly, various new drugs are hidden in public places through changing forms and packaging, which brings new challenges to drug enforcement. This study proposes a drug-detecting bioelectronic nose based on odor cue memory combined with brain-computer interface and optogenetic regulation technologies. First, the rats were trained to generate positive memories of drug odors through food reward training, and multichannel microelectrodes were implanted into the DG region of the hippocampus for responsible memory retrieval, the spike signals of individual neurons and the local field potential signals of population neurons in the brain region were collected for pattern recognition and analysis. Preliminary experimental results have shown that when low-dose drugs are buried in a hidden area, rats can find the location of the drugs in a very short time, and when close to the relevant area, there is a significant change in the energy value and time-frequency spectrum signal coupling of the returned data, which can be extracted to indicate that the rats have found the drugs. Second, we labled the neuronal activity marker c-fos and revealed more robust activation in the DG region following odor detection. We modulated these neurons through neuroregulatory technology, so that the rats could recognize drugs by retrieving memories more quickly. We conceive that the drug-detecting rat robot can detect trace amounts of various drugs in complex terrain and multiple scenes, which is of great significance for anti-drug work in the future.
Collapse
Affiliation(s)
- Keqiang Gao
- School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China
| | - Mengxi Hu
- School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China
| | - Jiyang Li
- School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China
| | - Ziyi Li
- School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China
| | - Wei Xu
- Department of Biomedical Engineering, Nanjing University of Aeronautics and Astronautics, Nanjing, 210016, China
| | - Zhiyu Qian
- Department of Biomedical Engineering, Nanjing University of Aeronautics and Astronautics, Nanjing, 210016, China
| | - Fan Gao
- Department of Biomedical Engineering, Nanjing University of Aeronautics and Astronautics, Nanjing, 210016, China.
| | - Tengfei Ma
- School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China.
| |
Collapse
|
21
|
Yang K, Hasegawa Y, Bhattarai JP, Hua J, Dower M, Etyemez S, Prasad N, Duvall L, Paez A, Smith A, Wang Y, Zhang YF, Lane AP, Ishizuka K, Kamath V, Ma M, Kamiya A, Sawa A. Inflammation-related pathology in the olfactory epithelium: its impact on the olfactory system in psychotic disorders. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2022.09.23.509224. [PMID: 36203543 PMCID: PMC9536041 DOI: 10.1101/2022.09.23.509224] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/25/2023]
Abstract
Smell deficits and neurobiological changes in the olfactory bulb (OB) and olfactory epithelium (OE) have been observed in schizophrenia and related disorders. The OE is the most peripheral olfactory system located outside the cranium, and is connected with the brain via direct neuronal projections to the OB. Nevertheless, it is unknown whether and how a disturbance of the OE affects the OB in schizophrenia and related disorders. Addressing this gap would be the first step in studying the impact of OE pathology in the disease pathophysiology in the brain. In this cross-species study, we observed that chronic, local OE inflammation with a set of upregulated genes in an inducible olfactory inflammation (IOI) mouse model led to a volume reduction, layer structure changes, and alterations of neuron functionality in the OB. Furthermore, IOI model also displayed behavioral deficits relevant to negative symptoms (avolition) in parallel to smell deficits. In first episode psychosis (FEP) patients, we observed a significant alteration in immune/inflammation-related molecular signatures in olfactory neuronal cells (ONCs) enriched from biopsied OE and a significant reduction in the OB volume, compared with those of healthy controls (HC). The increased expression of immune/inflammation-related molecules in ONCs was significantly correlated to the OB volume reduction in FEP patients, but no correlation was found in HCs. Moreover, the increased expression of human orthologues of the IOI genes in ONCs was significantly correlated with the OB volume reduction in FEP, but not in HCs. Together, our study implies a potential mechanism of the OE-OB pathology in patients with psychotic disorders (schizophrenia and related disorders). We hope that this mechanism may have a cross-disease implication, including COVID-19-elicited mental conditions that include smell deficits.
Collapse
|
22
|
Gutiérrez-García AG, Contreras CM. Olfactory Epithelium Infection by SARS-CoV-2: Possible Neuroinflammatory Consequences of COVID-19. Complex Psychiatry 2024; 10:59-70. [PMID: 39545135 PMCID: PMC11560153 DOI: 10.1159/000540982] [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: 11/07/2023] [Accepted: 07/25/2024] [Indexed: 11/17/2024] Open
Abstract
Background The loss of smell is a typical diagnostic symptom of coronavirus disease 2019 (COVID-19). This sensorial deprivation may be expressed as quantitative (anosmia or hyposmia) or qualitative (dysosmia) alterations as a consequence of anatomical disturbances of the nasal epithelium structure. The olfactory system sends direct neuronal connections to brain structures that are involved in emotional processing, including deep temporal nuclei. This anatomical and functional feature may be related to the occurrence of emotional disorders among COVID-19 patients. Summary We identify a possible sequence of events, from typical olfactory dysfunction that is associated with COVID-19 and caused by olfactory epithelium damage to disturbances in the quality of life and emotional state of infected patients that is attributable to possible neuroinflammatory processes. Sensorial deprivation causes deleterious actions on mood, negatively affecting quality of life. Olfactory dysfunction that is associated with COVID-19 occurs concurrently with psychological distress, symptoms of anxiety, and depressive disorders and impinges on self-perceived quality of life. Key Messages Changes in mood are certainly associated with multiple factors, including the environment and isolation, but the observation that the virus may penetrate the central nervous system through the olfactory bulb and the connection between the olfactory system and prefrontal and orbitofrontal cortices and the amygdala-hippocampus do not allow one to discard neural factors that are involved in the pathophysiology of emotional symptoms in post-COVID-19 patients. Behavioral symptoms of COVID-19 involve local olfactory actions and the participation of central neuronal systems.
Collapse
Affiliation(s)
- Ana G. Gutiérrez-García
- Laboratorio de Neurofarmacología, Instituto de Neuroetología, Universidad Veracruzana, Xalapa, Mexico
| | - Carlos M. Contreras
- Unidad Periférica-Xalapa, Instituto de Investigaciones Biomédicas, National Autonomous University of Mexico (UNAM), Xalapa, Mexico
| |
Collapse
|
23
|
Yang K, Ayala-Grosso C, Bhattarai JP, Sheriff A, Takahashi T, Cristino AS, Zelano C, Ma M. Unraveling the Link between Olfactory Deficits and Neuropsychiatric Disorders. J Neurosci 2023; 43:7501-7510. [PMID: 37940584 PMCID: PMC10634556 DOI: 10.1523/jneurosci.1380-23.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 07/27/2023] [Accepted: 07/31/2023] [Indexed: 11/10/2023] Open
Abstract
Smell loss has caught public attention during the recent COVID-19 pandemic. Research on olfactory function in health and disease gains new momentum. Smell deficits have long been recognized as an early clinical sign associated with neuropsychiatric disorders. Here we review research on the associations between olfactory deficits and neuropathological conditions, focusing on recent progress in four areas: (1) human clinical studies of the correlations between smell deficits and neuropsychiatric disorders; (2) development of olfactory mucosa-derived tissue and cell models for studying the molecular pathologic mechanisms; (3) recent findings in brain imaging studies of structural and functional connectivity changes in olfactory pathways in neuropsychiatric disorders; and (4) application of preclinical animal models to validate and extend the findings from human subjects. Together, these studies have provided strong evidence of the link between the olfactory system and neuropsychiatric disorders, highlighting the relevance of deepening our understanding of the role of the olfactory system in pathophysiological processes. Following the lead of studies reviewed here, future research in this field may open the door to the early detection of neuropsychiatric disorders, personalized treatment approaches, and potential therapeutic interventions through nasal administration techniques, such as nasal brush or nasal spray.
Collapse
Affiliation(s)
- Kun Yang
- Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287
| | - Carlos Ayala-Grosso
- Unit of Cellular Therapy, Centre of Experimental Medicine, Instituto Venezolano de Investigaciones Cientificas, Caracas, 1020-A, Venezuela
- Unit of Advanced Therapies, Instituto Distrital de Ciencia Biotecnología e Innovación en Salud, Bogotá, Colombia 111-611
| | - Janardhan P Bhattarai
- Department of Neuroscience, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania 19104
| | - Andrew Sheriff
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611
| | - Tsutomu Takahashi
- Department of Neuropsychiatry, University of Toyama Graduate School of Medicine and Pharmaceutical Sciences, Toyama, 930-0194, Japan
| | - Alexandre S Cristino
- Griffith Institute for Drug Discovery, Griffith University, Brisbane, Queensland 4111, Australia
| | - Christina Zelano
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611
| | - Minghong Ma
- Department of Neuroscience, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania 19104
| |
Collapse
|
24
|
Hernández-Pérez OR, Hernández VS, Zetter MA, Eiden LE, Zhang L. Nucleus of the lateral olfactory tract: A hub linking the water homeostasis-associated supraoptic nucleus-arginine vasopressin circuit and neocortical regions to promote social behavior under osmotic challenge. J Neuroendocrinol 2023; 35:e13202. [PMID: 36283814 PMCID: PMC10027625 DOI: 10.1111/jne.13202] [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: 07/01/2022] [Revised: 09/14/2022] [Accepted: 09/14/2022] [Indexed: 11/30/2022]
Abstract
Homeostatic challenges may alter the drive for social interaction. The neural activity that prompts this motivation remains poorly understood. In the present study, we identify direct projections from the hypothalamic supraoptic nucleus to the cortico-amygdalar nucleus of the lateral olfactory tract (NLOT). Dual in situ hybridization with probes for pituitary adenylate cyclase-activating polypeptide (PACAP), as well as vesicular glutamate transporter (VGLUT)1, VGLUT2, V1a and V1b, revealed a population of vasopressin-receptive PACAPergic neurons in NLOT layer 2 (NLOT2). Water deprivation (48 h, WD48) increased sociability compared to euhydrated subjects, as assessed with the three-chamber social interaction test (3CST). Fos expression immunohistochemistry showed NLOT and its main efferent regions had further increases in rats subjected to WD48 + 3CST. These regions strongly expressed PAC1 mRNA. Microinjections of arginine vasopressin (AVP) into the NLOT produced similar changes in sociability to water deprivation, and these were reduced by co-injection of V1a or V1b antagonists along with AVP. We conclude that, during challenge to water homeostasis, there is a recruitment of a glutamatergic-multi-peptidergic cooperative circuit that promotes social behavior.
Collapse
Affiliation(s)
- Oscar R. Hernández-Pérez
- Department of Physiology, School of Medicine, National Autonomous University of Mexico, Mexico
- Authors contributed equally to this work
| | - Vito S. Hernández
- Department of Physiology, School of Medicine, National Autonomous University of Mexico, Mexico
- Authors contributed equally to this work
| | - Mario A. Zetter
- Department of Physiology, School of Medicine, National Autonomous University of Mexico, Mexico
| | - Lee E. Eiden
- National Institute of Mental Health, National Institute of Health, Bethesda, MD, USA
| | - Limei Zhang
- Department of Physiology, School of Medicine, National Autonomous University of Mexico, Mexico
| |
Collapse
|
25
|
Liu P, Gao C, Wu J, Wu T, Zhang Y, Liu C, Sun C, Li A. Negative valence encoding in the lateral entorhinal cortex during aversive olfactory learning. Cell Rep 2023; 42:113204. [PMID: 37804511 DOI: 10.1016/j.celrep.2023.113204] [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: 09/30/2022] [Revised: 08/24/2023] [Accepted: 09/19/2023] [Indexed: 10/09/2023] Open
Abstract
Olfactory learning is widely regarded as a substrate for animal survival. The exact brain areas involved in olfactory learning and how they function at various stages during learning remain elusive. Here, we investigate the role of the lateral entorhinal cortex (LEC) and the posterior piriform cortex (PPC), two important olfactory areas, in aversive olfactory learning. We find that the LEC is involved in the acquisition of negative odor value during olfactory fear conditioning, whereas the PPC is involved in the memory-retrieval phase. Furthermore, inhibition of LEC CaMKIIα+ neurons affects fear encoding, fear memory recall, and PPC responses to a conditioned odor. These findings provide direct evidence for the involvement of LEC CaMKIIα+ neurons in negative valence encoding.
Collapse
Affiliation(s)
- Penglai Liu
- Jiangsu Key Laboratory of Brain Disease and Bioinformation, Research Center for Biochemistry and Molecular Biology, Xuzhou Medical University, Xuzhou 221004, China
| | - Cheng Gao
- Jiangsu Key Laboratory of Brain Disease and Bioinformation, Research Center for Biochemistry and Molecular Biology, Xuzhou Medical University, Xuzhou 221004, China
| | - Jing Wu
- Jiangsu Key Laboratory of Brain Disease and Bioinformation, Research Center for Biochemistry and Molecular Biology, Xuzhou Medical University, Xuzhou 221004, China
| | - Tingting Wu
- Jiangsu Key Laboratory of Brain Disease and Bioinformation, Research Center for Biochemistry and Molecular Biology, Xuzhou Medical University, Xuzhou 221004, China
| | - Ying Zhang
- Jiangsu Key Laboratory of Brain Disease and Bioinformation, Research Center for Biochemistry and Molecular Biology, Xuzhou Medical University, Xuzhou 221004, China
| | - Changyu Liu
- Jiangsu Key Laboratory of Brain Disease and Bioinformation, Research Center for Biochemistry and Molecular Biology, Xuzhou Medical University, Xuzhou 221004, China
| | - Changcheng Sun
- Jiangsu Key Laboratory of Brain Disease and Bioinformation, Research Center for Biochemistry and Molecular Biology, Xuzhou Medical University, Xuzhou 221004, China.
| | - Anan Li
- Jiangsu Key Laboratory of Brain Disease and Bioinformation, Research Center for Biochemistry and Molecular Biology, Xuzhou Medical University, Xuzhou 221004, China.
| |
Collapse
|
26
|
Sha MFR, Koga Y, Murata Y, Taniguchi M, Yamaguchi M. Learning-dependent structural plasticity of intracortical and sensory connections to functional domains of the olfactory tubercle. Front Neurosci 2023; 17:1247375. [PMID: 37680965 PMCID: PMC10480507 DOI: 10.3389/fnins.2023.1247375] [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: 06/26/2023] [Accepted: 08/09/2023] [Indexed: 09/09/2023] Open
Abstract
The olfactory tubercle (OT), which is a component of the olfactory cortex and ventral striatum, has functional domains that play a role in odor-guided motivated behaviors. Learning odor-guided attractive and aversive behavior activates the anteromedial (am) and lateral (l) domains of the OT, respectively. However, the mechanism driving learning-dependent activation of specific OT domains remains unknown. We hypothesized that the neuronal connectivity of OT domains is plastically altered through olfactory experience. To examine the plastic potential of synaptic connections to OT domains, we optogenetically stimulated intracortical inputs from the piriform cortex or sensory inputs from the olfactory bulb to the OT in mice in association with a food reward for attractive learning and electrical foot shock for aversive learning. For both intracortical and sensory connections, axon boutons that terminated in the OT domains were larger in the amOT than in the lOT for mice exhibiting attractive learning and larger in the lOT than in the amOT for mice exhibiting aversive learning. These results indicate that both intracortical and sensory connections to the OT domains have learning-dependent plastic potential, suggesting that this plasticity underlies learning-dependent activation of specific OT domains and the acquisition of appropriate motivated behaviors.
Collapse
Affiliation(s)
| | | | | | | | - Masahiro Yamaguchi
- Department of Physiology, Kochi Medical School, Kochi University, Kochi, Japan
| |
Collapse
|
27
|
Wu J, Liu P, Geng C, Liu C, Li J, Zhu Q, Li A. Principal neurons in the olfactory cortex mediate bidirectional modulation of seizures. J Physiol 2023; 601:3557-3584. [PMID: 37384845 DOI: 10.1113/jp284731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 06/15/2023] [Indexed: 07/01/2023] Open
Abstract
Although the piriform cortex (PC) has been previously implicated as a critical node for seizure generation and propagation, the underlying neural mechanism has remained unclear. Here, we found increased excitability in PC neurons during amygdala kindling acquisition. Optogenetic or chemogenetic activation of PC pyramidal neurons promoted kindling progression, whereas inhibition of these neurons retarded seizure activities induced by electrical kindling in the amygdala. Furthermore, chemogenetic inhibition of PC pyramidal neurons alleviated the severity of kainic acid-induced acute seizures. These results demonstrate that PC pyramidal neurons bidirectionally modulate seizures in temporal lobe epilepsy, providing evidence for the efficacy of PC pyramidal neurons as a potential therapeutic target for epileptogenesis. KEY POINTS: While the piriform cortex (PC) is an important olfactory centre critically involved in olfactory processing and plays a crucial role in epilepsy due to its close connection with the limbic system, how the PC regulates epileptogenesis is largely unknown. In this study, we evaluated the neuronal activity and the role of pyramidal neurons in the PC in the mouse amygdala kindling model of epilepsy. PC pyramidal neurons are hyperexcited during epileptogenesis. Optogenetic and chemogenetic activation of PC pyramidal neurons significantly promoted seizures in the amygdala kindling model, whereas selective inhibition of these neurons produced an anti-epileptic effect for both electrical kindling and kainic acid-induced acute seizures. The results of the present study indicate that PC pyramidal neurons bidirectionally modulate seizure activity.
Collapse
Affiliation(s)
- Jing Wu
- Jiangsu Key Laboratory of Brain Disease and Bioinformation, Research Center for Biochemistry and Molecular Biology, Xuzhou Medical University, Xuzhou, China
| | - Penglai Liu
- Jiangsu Key Laboratory of Brain Disease and Bioinformation, Research Center for Biochemistry and Molecular Biology, Xuzhou Medical University, Xuzhou, China
| | - Chi Geng
- Jiangsu Key Laboratory of Brain Disease and Bioinformation, Research Center for Biochemistry and Molecular Biology, Xuzhou Medical University, Xuzhou, China
| | - Changyu Liu
- Jiangsu Key Laboratory of Brain Disease and Bioinformation, Research Center for Biochemistry and Molecular Biology, Xuzhou Medical University, Xuzhou, China
| | - Jiaxin Li
- Jiangsu Key Laboratory of Brain Disease and Bioinformation, Research Center for Biochemistry and Molecular Biology, Xuzhou Medical University, Xuzhou, China
| | - Qiuju Zhu
- Jiangsu Key Laboratory of Brain Disease and Bioinformation, Research Center for Biochemistry and Molecular Biology, Xuzhou Medical University, Xuzhou, China
| | - Anan Li
- Jiangsu Key Laboratory of Brain Disease and Bioinformation, Research Center for Biochemistry and Molecular Biology, Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
28
|
López M, Fernández-Real JM, Tomarev SI. Obesity wars: may the smell be with you. Am J Physiol Endocrinol Metab 2023; 324:E569-E576. [PMID: 37166265 PMCID: PMC10259866 DOI: 10.1152/ajpendo.00040.2023] [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: 02/07/2023] [Revised: 05/08/2023] [Accepted: 05/09/2023] [Indexed: 05/12/2023]
Abstract
Classically, the regulation of energy balance has been based on central and peripheral mechanisms sensing energy, nutrients, metabolites, and hormonal cues. Several cellular mechanisms at central level, such as hypothalamic AMP-activated protein kinase (AMPK), integrate this information to elicit counterregulatory responses that control feeding, energy expenditure, and glucose homeostasis, among other processes. Recent data have added more complexity to the homeostatic regulation of metabolism by introducing, for example, the key role of "traditional" senses and sensorial information in this complicated network. In this regard, current evidence is showing that olfaction plays a key and bidirectional role in energy homeostasis. Although nutritional status dynamically and profoundly impacts olfactory sensitivity, the sense of smell is involved in food appreciation and selection, as well as in brown adipose tissue (BAT) thermogenesis and substrate utilization, with some newly described actors, such as olfactomedin 2 (OLFM2), likely playing a major role. Thus, olfactory inputs are contributing to the regulation of both sides of the energy balance equation, namely, feeding and energy expenditure (EE), as well as whole body metabolism. Here, we will review the current knowledge and advances about the role of olfaction in the regulation of energy homeostasis.
Collapse
Affiliation(s)
- Miguel López
- NeurObesity Group, Department of Physiology, CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Spain
| | - José Manuel Fernández-Real
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Spain
- Service of Diabetes, Endocrinology and Nutrition (UDEN), Institut d'Investigació Biomédica de Girona (IDIBGI), Department of Medical Sciences, University of Girona, Girona, Spain
| | - Stanislav I Tomarev
- Section of Retinal Ganglion Cell Biology, Laboratory of Retinal Cell and Molecular Biology, National Eye Institute, National Institutes of Health, Bethesda, Maryland, United States
| |
Collapse
|
29
|
Amos C, Fox MA, Su J. Collagen XIX is required for pheromone recognition and glutamatergic synapse formation in mouse accessory olfactory bulb. Front Cell Neurosci 2023; 17:1157577. [PMID: 37091919 PMCID: PMC10113670 DOI: 10.3389/fncel.2023.1157577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Accepted: 03/21/2023] [Indexed: 04/25/2023] Open
Abstract
In mammals, the accessory olfactory bulb (AOB) receives input from vomeronasal sensory neurons (VSN) which detect pheromones, chemical cues released by animals to regulate the physiology or behaviors of other animals of the same species. Cytoarchitecturally, cells within the AOB are segregated into a glomerular layer (GL), mitral cell layer (MCL), and granule cell layer (GCL). While the cells and circuitry of these layers has been well studied, the molecular mechanism underlying the assembly of such circuitry in the mouse AOB remains unclear. With the goal of identifying synaptogenic mechanisms in AOB, our attention was drawn to Collagen XIX, a non-fibrillar collagen generated by neurons in the mammalian telencephalon that has previously been shown to regulate the assembly of synapses. Here, we used both a targeted mouse mutant that lacks Collagen XIX globally and a conditional allele allowing for cell-specific deletion of this collagen to test if the loss of Collagen XIX causes impaired synaptogenesis in the mouse AOB. These analyses not only revealed defects in excitatory synapse distribution in these Collagen XIX-deficient mutants, but also showed that these mutant mice exhibit altered behavioral responses to pheromones. Although this collagen has been demonstrated to play synaptogenic roles in the telencephalon, those roles are at perisomatic inhibitory synapses, results here are the first to demonstrate the function of this unconventional collagen in glutamatergic synapse formation.
Collapse
Affiliation(s)
- Chase Amos
- Center for Neurobiology Research, Fralin Biomedical Research Institute at Virginia Tech Carilion (VTC), Roanoke, VA, United States
| | - Michael A. Fox
- Center for Neurobiology Research, Fralin Biomedical Research Institute at Virginia Tech Carilion (VTC), Roanoke, VA, United States
- School of Neuroscience, Virginia Tech, Blacksburg, VA, United States
- Department of Biological Sciences, Virginia Tech, Blacksburg, VA, United States
- Department of Pediatrics, Virginia Tech Carilion School of Medicine, Roanoke, VA, United States
| | - Jianmin Su
- Center for Neurobiology Research, Fralin Biomedical Research Institute at Virginia Tech Carilion (VTC), Roanoke, VA, United States
- School of Neuroscience, Virginia Tech, Blacksburg, VA, United States
| |
Collapse
|
30
|
Wang J, Sun L, Chen L, Sun J, Xie Y, Tian D, Gao L, Zhang D, Xia M, Wu T. Common and distinct roles of amygdala subregional functional connectivity in non-motor symptoms of Parkinson's disease. NPJ Parkinsons Dis 2023; 9:28. [PMID: 36806219 PMCID: PMC9938150 DOI: 10.1038/s41531-023-00469-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 02/02/2023] [Indexed: 02/19/2023] Open
Abstract
Neuroimaging studies suggest a pivotal role of amygdala dysfunction in non-motor symptoms (NMS) of Parkinson's disease (PD). However, the relationship between amygdala subregions (the centromedial (CMA), basolateral (BLA) and superficial amygdala (SFA)) and NMS has not been delineated. We used resting-state functional MRI to examine the PD-related alterations in functional connectivity for amygdala subregions. The left three subregions and right BLA exhibited between-group differences, and were commonly hypo-connected with the frontal, temporal, insular cortex, and putamen in PD. Each subregion displayed distinct hypoconnectivity with the limbic systems. Partial least-squares analysis revealed distinct amygdala subregional involvement in diverse NMS. Hypo-connectivity of all four subregions was associated with emotion, pain, olfaction, and cognition. Hypo-connectivity of the left SFA was associated with sleepiness. Our findings highlight the hypofunction of the amygdala subregions in PD and their preliminary associations with NMS, providing new insights into the pathogenesis of NMS.
Collapse
Affiliation(s)
- Junling Wang
- grid.24696.3f0000 0004 0369 153XCenter for Movement Disorders, Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070 China
| | - Lianglong Sun
- grid.20513.350000 0004 1789 9964State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing, 100091 China ,grid.20513.350000 0004 1789 9964Beijing Key Laboratory of Brain Imaging and Connectomics, Beijing Normal University, Beijing, 100091 China ,grid.20513.350000 0004 1789 9964IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing, 100091 China
| | - Lili Chen
- grid.24696.3f0000 0004 0369 153XCenter for Movement Disorders, Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070 China
| | - Junyan Sun
- grid.24696.3f0000 0004 0369 153XCenter for Movement Disorders, Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070 China
| | - Yapei Xie
- grid.20513.350000 0004 1789 9964State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing, 100091 China ,grid.20513.350000 0004 1789 9964Beijing Key Laboratory of Brain Imaging and Connectomics, Beijing Normal University, Beijing, 100091 China ,grid.20513.350000 0004 1789 9964IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing, 100091 China
| | - Dezheng Tian
- grid.20513.350000 0004 1789 9964State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing, 100091 China ,grid.20513.350000 0004 1789 9964Beijing Key Laboratory of Brain Imaging and Connectomics, Beijing Normal University, Beijing, 100091 China ,grid.20513.350000 0004 1789 9964IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing, 100091 China
| | - Linlin Gao
- grid.417031.00000 0004 1799 2675Department of General Medicine, Tianjin Union Medical Center, Tianjin, 300122 China
| | - Dongling Zhang
- grid.24696.3f0000 0004 0369 153XCenter for Movement Disorders, Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070 China
| | - Mingrui Xia
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing, 100091, China. .,Beijing Key Laboratory of Brain Imaging and Connectomics, Beijing Normal University, Beijing, 100091, China. .,IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing, 100091, China.
| | - Tao Wu
- Center for Movement Disorders, Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China.
| |
Collapse
|
31
|
Osada K, Kujirai R, Hosono A, Tsuda M, Ohata M, Ohta T, Nishimori K. Repeated exposure to kairomone-containing coffee odor improves abnormal olfactory behaviors in heterozygous oxytocin receptor knock-in mice. Front Behav Neurosci 2023; 16:983421. [PMID: 36817409 PMCID: PMC9930907 DOI: 10.3389/fnbeh.2022.983421] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 12/16/2022] [Indexed: 02/04/2023] Open
Abstract
The oxytocin receptor (OXTR) knockout mouse is a model of autism spectrum disorder, characterized by abnormalities in social and olfactory behaviors and learning. Previously, we demonstrated that OXTR plays a crucial role in regulating aversive olfactory behavior to butyric acid odor. In this study, we attempted to determine whether coffee aroma affects the abnormal olfactory behavior of OXTR-Venus knock-in heterozygous mice [heterozygous OXTR (±) mice] using a set of behavioral and molecular experiments. Four-week repeated exposures of heterozygous OXTR (±) mice to coffee odor, containing three kairomone alkylpyrazines, rescued the abnormal olfactory behaviors compared with non-exposed wild-type or heterozygous OXTR (±) mice. Increased Oxtr mRNA expression in the olfactory bulb and amygdala coincided with the rescue of abnormal olfactory behaviors. In addition, despite containing the kairomone compounds, both the wild-type and heterozygous OXTR (±) mice exhibited a preference for the coffee odor and exhibited no stress-like increase in the corticotropin-releasing hormone, instead of a kairomone-associated avoidance response. The repeated exposures to the coffee odor did not change oxytocin and estrogen synthetase/receptors as a regulator of the gonadotropic hormone. These data suggest that the rescue of abnormal olfactory behaviors in heterozygous OXTR (±) mice is due to the coffee odor exposure-induced OXTR expression.
Collapse
Affiliation(s)
- Kazumi Osada
- Department of Food Bioscience and Biotechnology, College of Bioresource Sciences, Nihon University, Fujisawa, Japan,*Correspondence: Kazumi Osada,
| | - Riyuki Kujirai
- Department of Food Bioscience and Biotechnology, College of Bioresource Sciences, Nihon University, Fujisawa, Japan
| | - Akira Hosono
- Department of Food Bioscience and Biotechnology, College of Bioresource Sciences, Nihon University, Fujisawa, Japan
| | - Masato Tsuda
- Department of Food Bioscience and Biotechnology, College of Bioresource Sciences, Nihon University, Fujisawa, Japan
| | - Motoko Ohata
- Department of Food Bioscience and Biotechnology, College of Bioresource Sciences, Nihon University, Fujisawa, Japan
| | - Tohru Ohta
- The Research Institute of Health Science, Health Sciences University of Hokkaido, Tobetsu, Japan
| | - Katsuhiko Nishimori
- Department of Molecular and Cell Biology, Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
| |
Collapse
|
32
|
Zhao Y, Bhutani S, Kahnt T. Appetite-regulating hormones modulate odor perception and odor-evoked activity in hypothalamus and olfactory cortices. Chem Senses 2023; 48:bjad039. [PMID: 37796827 PMCID: PMC10590159 DOI: 10.1093/chemse/bjad039] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Indexed: 10/07/2023] Open
Abstract
Odors guide food seeking, and food intake modulates olfactory function. This interaction is mediated by appetite-regulating hormones like ghrelin, insulin, and leptin, which alter activity in the rodent olfactory bulb, but their effects on downstream olfactory cortices have not yet been established in humans. The olfactory tract connects the olfactory bulb to the cortex through 3 main striae, terminating in the piriform cortex (PirC), amygdala (AMY), olfactory tubercule (OT), and anterior olfactory nucleus (AON). Here, we test the hypothesis that appetite-regulating hormones modulate olfactory processing in the endpoints of the olfactory tract and the hypothalamus. We collected odor-evoked functional magnetic resonance imaging (fMRI) responses and plasma levels of ghrelin, insulin, and leptin from human subjects (n = 25) after a standardized meal. We found that a hormonal composite measure, capturing variance relating positively to insulin and negatively to ghrelin, correlated inversely with odor intensity ratings and fMRI responses to odorized vs. clean air in the hypothalamus, OT, and AON. No significant correlations were found with activity in PirC or AMY, the endpoints of the lateral stria. Exploratory whole-brain analyses revealed significant correlations near the diagonal band of Broca and parahippocampal gyrus. These results demonstrate that high (low) blood plasma concentrations of insulin (ghrelin) decrease perceived odor intensity and odor-evoked activity in the cortical targets of the medial and intermediate striae of the olfactory tract, as well as the hypothalamus. These findings expand our understanding of the cortical mechanisms by which metabolic hormones in humans modulate olfactory processing after a meal.
Collapse
Affiliation(s)
- Yao Zhao
- National Institute on Drug Abuse Intramural Research Program, Cellular and Neurocomputational Systems Branch, Baltimore, MD,United States
| | - Surabhi Bhutani
- San Diego State University, School of Exercise and Nutritional Sciences, San Diego, CA, United States
| | - Thorsten Kahnt
- National Institute on Drug Abuse Intramural Research Program, Cellular and Neurocomputational Systems Branch, Baltimore, MD,United States
| |
Collapse
|
33
|
Chen HC, Ma YZ, Cao JX, Zhang YS, Zhang L, Gao LP, Jing YH. Synergistic effects of hIAPP and Aβ 1-42 impaired the olfactory function associated with the decline of adult neurogenesis in SVZ. Neuropeptides 2022; 96:102268. [PMID: 35841876 DOI: 10.1016/j.npep.2022.102268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 06/22/2022] [Accepted: 06/26/2022] [Indexed: 10/17/2022]
Abstract
According to many in the field,the prevalence of Alzheimer's disease (AD) in type II diabetes (T2DM) populations is considerably higher than that in the normal population. Human islet amyloid polypeptide (hIAPP) is considered to be a common risk factor for T2DM and AD. Preliminary observations around T2DM animal model show that the decrease of adult neural stem cells (NSCs) in the subventricular zone (SVZ) is accompanied by olfactory dysfunction. Furthermore, impaired olfactory function could serve as to an early predictor of neurodegeneration,which is associated with cognitive impairment. However, the synergistic effects between hIAPP and amyloid-beta (Aβ) 1-42 in the brain and the neurodegeneration remains to be further clarified. In this study, olfactory capacity, synaptic density, status of NSC in SVZ, and status of newborn neurons in olfactory bulb (OB) were assessed 6 months after stereotactic injection of oligomer Aβ1-42 into the dens gyrus (DG) of hIAPP-/+ mice or wild-type homogenous mice. Our results set out that Aβ42 and amylin co-localized into OB and raised Aβ42 deposition in hIAPP-/+ mice compared with wild-type brood mice. In addition, 6 months after injection of Aβ1-42 in hIAPP-/+ mice, these mice showed increased olfactory dysfunction, significant loss of synapses, depletion of NSC in SVZ, and impaired cell renewal in OB. Our present study suggested that the synergistic effects between hIAPP and Aβ1-42 impairs olfactory function and was associated with decreased neurogenesis in adults with SVZ.
Collapse
Affiliation(s)
- Hai-Chao Chen
- Institute of Anatomy and Histology & Embryology, Neuroscience, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu, People's Republic of China
| | - Yue-Zhang Ma
- Institute of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu, People's Republic of China
| | - Jia-Xin Cao
- Institute of Anatomy and Histology & Embryology, Neuroscience, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu, People's Republic of China
| | - Yi-Shu Zhang
- Institute of Anatomy and Histology & Embryology, Neuroscience, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu, People's Republic of China
| | - Lu Zhang
- Institute of Anatomy and Histology & Embryology, Neuroscience, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu, People's Republic of China
| | - Li-Ping Gao
- Institute of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu, People's Republic of China; Key Laboratory of Preclinical Study for New Drugs of Gansu province, Lanzhou University, Lanzhou, Gansu, People's Republic of China
| | - Yu-Hong Jing
- Institute of Anatomy and Histology & Embryology, Neuroscience, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu, People's Republic of China; Key Laboratory of Preclinical Study for New Drugs of Gansu province, Lanzhou University, Lanzhou, Gansu, People's Republic of China.
| |
Collapse
|
34
|
Bentz EJ, Ophir AG. Chromosome-scale genome assembly of the African giant pouched rat (Cricetomys ansorgei) and evolutionary analysis reveals evidence of olfactory specialization. Genomics 2022; 114:110521. [PMID: 36351561 DOI: 10.1016/j.ygeno.2022.110521] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 10/28/2022] [Accepted: 11/04/2022] [Indexed: 11/07/2022]
Abstract
The Southern giant pouched rat, Cricetomys ansorgei, is a large rodent best known for its ability to detect landmines using its impressive sense of smell. Their powerful chemosensory abilities enable subtle discrimination of chemical social signals, and female pouched rats demonstrate a unique reproductive physiology hypothesized to be mediated by pheromonal mechanisms. Thus, C. ansorgei represents a novel mammalian model for chemosensory physiology, social behavior, and pheromonal control of reproductive physiology. We present the first chromosome-scale genomic sequence of the pouched rat encoding 22,671 protein coding genes, including 1571 olfactory receptors, and provide a glance into the evolutionary history of this species. Functional enrichment analysis reveals genetic expansions specific to the pouched rat are enriched for functions related to olfactory specialization. Overall, this assembly is of reference-quality, and will serve as a useful and informative genomic sequence on which we can confidently base future molecular research involving the pouched rat.
Collapse
Affiliation(s)
- Ehren J Bentz
- Department of Psychology, Cornell University, Ithaca, NY, USA.
| | | |
Collapse
|
35
|
Bhattarai JP, Etyemez S, Jaaro-Peled H, Janke E, Leon Tolosa UD, Kamiya A, Gottfried JA, Sawa A, Ma M. Olfactory modulation of the medial prefrontal cortex circuitry: Implications for social cognition. Semin Cell Dev Biol 2022; 129:31-39. [PMID: 33975755 PMCID: PMC8573060 DOI: 10.1016/j.semcdb.2021.03.022] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 03/24/2021] [Accepted: 03/29/2021] [Indexed: 10/21/2022]
Abstract
Olfactory dysfunction is manifested in a wide range of neurological and psychiatric diseases, and often emerges prior to the onset of more classical symptoms and signs. From a behavioral perspective, olfactory deficits typically arise in conjunction with impairments of cognition, motivation, memory, and emotion. However, a conceptual framework for explaining the impact of olfactory processing on higher brain functions in health and disease remains lacking. Here we aim to provide circuit-level insights into this question by synthesizing recent advances in olfactory network connectivity with other cortical brain regions such as the prefrontal cortex. We will focus on social cognition as a representative model for exploring and critically evaluating the relationship between olfactory cortices and higher-order cortical regions in rodent models. Although rodents do not recapitulate all dimensions of human social cognition, they have experimentally accessible neural circuits and well-established behavioral tests for social motivation, memory/recognition, and hierarchy, which can be extrapolated to other species including humans. In particular, the medial prefrontal cortex (mPFC) has been recognized as a key brain region in mediating social cognition in both rodents and humans. This review will highlight the underappreciated connectivity, both anatomical and functional, between the olfactory system and mPFC circuitry, which together provide a neural substrate for olfactory modulation of social cognition and social behaviors. We will provide future perspectives on the functional investigation of the olfactory-mPFC circuit in rodent models and discuss how to translate such animal research to human studies.
Collapse
Affiliation(s)
- Janardhan P Bhattarai
- Department of Neuroscience, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Semra Etyemez
- Department of Psychiatry, John Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Hanna Jaaro-Peled
- Department of Psychiatry, John Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Emma Janke
- Department of Neuroscience, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Usuy D Leon Tolosa
- Department of Neuroscience, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Atsushi Kamiya
- Department of Psychiatry, John Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Jay A Gottfried
- Department of Psychology, University of Pennsylvania, School of Arts and Sciences, Philadelphia, PA 19104, USA; Department of Neurology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Akira Sawa
- Department of Psychiatry, John Hopkins University School of Medicine, Baltimore, MD 21287, USA; Departments of Neuroscience, Biomedical Engineering, and Genetic Medicine, John Hopkins University School of Medicine, Baltimore, MD 21287, USA; Department of Mental Health, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD 21287, USA.
| | - Minghong Ma
- Department of Neuroscience, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA.
| |
Collapse
|
36
|
Kulason S, Ratnanather JT, Miller MI, Kamath V, Hua J, Yang K, Ma M, Ishizuka K, Sawa A. A comparative neuroimaging perspective of olfaction and higher-order olfactory processing: on health and disease. Semin Cell Dev Biol 2022; 129:22-30. [PMID: 34462249 PMCID: PMC9900497 DOI: 10.1016/j.semcdb.2021.08.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2021] [Accepted: 08/18/2021] [Indexed: 02/08/2023]
Abstract
Olfactory dysfunction is often the earliest indicator of disease in a range of neurological and psychiatric disorders. One tempting working hypothesis is that pathological changes in the peripheral olfactory system where the body is exposed to many adverse environmental stressors may have a causal role for the brain alteration. Whether and how the peripheral pathology spreads to more central brain regions may be effectively studied in rodent models, and there is successful precedence in experimental models for Parkinson's disease. It is of interest to study whether a similar mechanism may underlie the pathology of psychiatric illnesses, such as schizophrenia. However, direct comparison between rodent models and humans includes challenges under light of comparative neuroanatomy and experimental methodologies used in these two distinct species. We believe that neuroimaging modality that has been the main methodology of human brain studies may be a useful viewpoint to address and fill the knowledge gap between rodents and humans in this scientific question. Accordingly, in the present review article, we focus on brain imaging studies associated with olfaction in healthy humans and patients with neurological and psychiatric disorders, and if available those in rodents. We organize this review article at three levels: 1) olfactory bulb (OB) and peripheral structures of the olfactory system, 2) primary olfactory cortical and subcortical regions, and 3) associated higher-order cortical regions. This research area is still underdeveloped, and we acknowledge that further validation with independent cohorts may be needed for many studies presented here, in particular those with human subjects. Nevertheless, whether and how peripheral olfactory disturbance impacts brain function is becoming even a hotter topic in the ongoing COVID-19 pandemic, given the risk of long-term changes of mental status associated with olfactory infection of SARS-CoV-2. Together, in this review article, we introduce this underdeveloped but important research area focusing on its implications in neurological and psychiatric disorders, with several pioneered publications.
Collapse
Affiliation(s)
- Sue Kulason
- Center for Imaging Science, Johns Hopkins University, Baltimore, MD, USA; Institute for Computational Medicine, Johns Hopkins University, Baltimore, MD, USA; Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - J Tilak Ratnanather
- Center for Imaging Science, Johns Hopkins University, Baltimore, MD, USA; Institute for Computational Medicine, Johns Hopkins University, Baltimore, MD, USA; Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Michael I Miller
- Center for Imaging Science, Johns Hopkins University, Baltimore, MD, USA; Institute for Computational Medicine, Johns Hopkins University, Baltimore, MD, USA; Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Vidyulata Kamath
- Department of Psychiatry, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Jun Hua
- Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA; F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, MD, USA
| | - Kun Yang
- Department of Psychiatry, Johns Hopkins School of Medicine, Baltimore, MD, USA; Johns Hopkins Schizophrenia Center, Baltimore, MD, USA
| | - Minghong Ma
- Department of Neuroscience, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Koko Ishizuka
- Department of Psychiatry, Johns Hopkins School of Medicine, Baltimore, MD, USA; Johns Hopkins Schizophrenia Center, Baltimore, MD, USA
| | - Akira Sawa
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA; Department of Psychiatry, Johns Hopkins School of Medicine, Baltimore, MD, USA; Johns Hopkins Schizophrenia Center, Baltimore, MD, USA; Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD, USA; Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA.
| |
Collapse
|
37
|
Chen A, Chen X, Deng J, Wei J, Qian H, Huang Y, Wu S, Gao F, Gong C, Liao Y, Zheng X. Dexmedetomidine alleviates olfactory cognitive dysfunction by promoting neurogenesis in the subventricular zone of hypoxic-ischemic neonatal rats. Front Pharmacol 2022; 13:983920. [PMID: 36059991 PMCID: PMC9437207 DOI: 10.3389/fphar.2022.983920] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 07/25/2022] [Indexed: 11/15/2022] Open
Abstract
Background: Hypoxic-ischemic brain damage (HIBD) is the main cause of neurological dysfunction in neonates. Olfactory cognitive function is important for feeding, the ability to detect hazardous situations and social relationships. However, only a few studies have investigated olfactory cognitive dysfunction in neonates with HIBD; furthermore, the specific mechanisms involved are yet to be elucidated. It has been reported that neurogenesis in the subventricular zone (SVZ) is linked to olfactory cognitive function. Recently, dexmedetomidine (DEX) has been shown to provide neuroprotection in neonates following HIBD. In the present study, we investigated whether DEX could improve olfactory cognitive dysfunction in neonatal rats following HIBD and attempted to determine the underlying mechanisms. Methods: We induced HIBD in rats using the Rice–Vannucci model, and DEX (25 μg/kg, i.p.) was administered immediately after the induction of HIBD. Next, we used triphenyl tetrazolium chloride (TTC) staining and the Zea-longa score to assess the success of modelling. The levels of BDNF, TNF-α, IL-1β and IL-6 were determined by western blotting. Immunofluorescence staining was used to detect microglial activation and microglial M1/M2 polarization as well as to evaluate the extent of neurogenesis in the SVZ. To evaluate the olfactory cognitive function, the rats in each group were raised until post-natal days 28–35; then, we performed the buried food test and the olfactory memory test. Results: Analysis showed that HIBD induced significant brain infarction, neurological deficits, and olfactory cognitive dysfunction. Furthermore, we found that DEX treatment significantly improved olfactory cognitive dysfunction in rat pups with HIBD. DEX treatment also increased the number of newly formed neuroblasts (BrdU/DCX) and neurons (BrdU/NeuN) in the SVZ by increasing the expression of BDNF in rat pups with HIBD. Furthermore, analysis showed that the neurogenic effects of DEX were possibly related to the inhibition of inflammation and the promotion of M1 to M2 conversion in the microglia. Conclusion: Based on the present findings, DEX treatment could improve olfactory cognitive dysfunction in neonatal rats with HIBD by promoting neurogenesis in the SVZ and enhancing the expression of BDNF in the microglia. It was possible associated that DEX inhibited neuroinflammation and promoted M1 to M2 conversion in the microglia.
Collapse
Affiliation(s)
- Andi Chen
- Department of Anesthesiology, Fujian Provincial Hospital, Shengli Clinical Medical College of Fujian Medical University, Fuzhou, China
| | - Xiaohui Chen
- Department of Anesthesiology, Fujian Provincial Hospital, Shengli Clinical Medical College of Fujian Medical University, Fuzhou, China
| | - Jianhui Deng
- Department of Anesthesiology, Fujian Provincial Hospital, Shengli Clinical Medical College of Fujian Medical University, Fuzhou, China
| | - Jianjie Wei
- Department of Anesthesiology, Fujian Provincial Hospital, Shengli Clinical Medical College of Fujian Medical University, Fuzhou, China
| | - Haitao Qian
- Department of Anesthesiology, Fujian Provincial Hospital, Shengli Clinical Medical College of Fujian Medical University, Fuzhou, China
| | - Yongxin Huang
- Department of Anesthesiology, Fujian Provincial Hospital, Shengli Clinical Medical College of Fujian Medical University, Fuzhou, China
| | - Shuyan Wu
- Department of Anesthesiology, Fujian Provincial Hospital, Shengli Clinical Medical College of Fujian Medical University, Fuzhou, China
| | - Fei Gao
- Department of Anesthesiology, Fujian Provincial Hospital, Shengli Clinical Medical College of Fujian Medical University, Fuzhou, China
| | - Cansheng Gong
- Department of Anesthesiology, Fujian Provincial Hospital, Shengli Clinical Medical College of Fujian Medical University, Fuzhou, China
| | - Yanling Liao
- Department of Anesthesiology, Fujian Provincial Hospital, Shengli Clinical Medical College of Fujian Medical University, Fuzhou, China
| | - Xiaochun Zheng
- Department of Anesthesiology, Fujian Provincial Hospital, Shengli Clinical Medical College of Fujian Medical University, Fuzhou, China
- Fujian Provincial Key Laboratory of Emergency Medicine, Fujian Provincial Key Laboratory of Critical care Medicine, Fujian Provincial Co-Constructed Laboratory of “Belt and Road”, Fujian Emergency Medical Center, Fuzhou, China
- *Correspondence: Xiaochun Zheng,
| |
Collapse
|
38
|
Olfactory impairment in psychiatric disorders: Does nasal inflammation impact disease psychophysiology? Transl Psychiatry 2022; 12:314. [PMID: 35927242 PMCID: PMC9352903 DOI: 10.1038/s41398-022-02081-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2021] [Revised: 07/14/2022] [Accepted: 07/20/2022] [Indexed: 12/04/2022] Open
Abstract
Olfactory impairments contribute to the psychopathology of mental illnesses such as schizophrenia and depression. Recent neuroscience research has shed light on the previously underappreciated olfactory neural circuits involved in regulation of higher brain functions. Although environmental factors such as air pollutants and respiratory viral infections are known to contribute to the risk for psychiatric disorders, the role of nasal inflammation in neurobehavioral outcomes and disease pathophysiology remains poorly understood. Here, we will first provide an overview of published findings on the impact of nasal inflammation in the olfactory system. We will then summarize clinical studies on olfactory impairments in schizophrenia and depression, followed by preclinical evidence on the neurobehavioral outcomes produced by olfactory dysfunction. Lastly, we will discuss the potential impact of nasal inflammation on brain development and function, as well as how we can address the role of nasal inflammation in the pathophysiological mechanisms underlying psychiatric disorders. Considering the current outbreak of Coronavirus Disease 2019 (COVID-19), which often causes nasal inflammation and serious adverse effects for olfactory function that might result in long-lasting neuropsychiatric sequelae, this line of research is particularly critical to understanding of the potential significance of nasal inflammation in the pathophysiology of psychiatric disorders.
Collapse
|
39
|
The role of ciliopathy-associated type 3 adenylyl cyclase in infanticidal behavior in virgin adult male mice. iScience 2022; 25:104534. [PMID: 35754726 PMCID: PMC9218507 DOI: 10.1016/j.isci.2022.104534] [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: 10/29/2021] [Revised: 02/11/2022] [Accepted: 06/01/2022] [Indexed: 12/04/2022] Open
Abstract
Virgin adult male mice often display killing of alien newborns, defined as infanticide, and this behavior is dependent on olfactory signaling. Olfactory perception is achieved by the main olfactory system (MOS) or vomeronasal system (VNS). Although it has been established that the VNS is crucial for infanticide in male mice, the role of the MOS in infanticide remains unknown. Herein, by producing lesions via ZnSO4 perfusion and N-methyl-D-aspartic acid stereotactic injection, we demonstrated that the main olfactory epithelium (MOE), anterior olfactory nucleus (AON), or ventromedial hypothalamus (VMH) is crucial for infanticide in adult males. By using CRISPR-Cas9 coupled with adeno-associated viruses to induce specific knockdown of type 3 adenylyl cyclase (AC3) in these tissues, we further demonstrated that AC3, a ciliopathy-associated protein, in the MOE and the expression of related proteins in the AON or VMH are necessary for infanticidal behavior in virgin adult male mice. MOE lesions and knockdown of AC3 in the MOE result in abnormal infanticidal behavior The infanticidal behavior of male mice is impaired by lesioning of the AON or VMH AC3 knockdown in the AON or VMH affects the infanticidal behavior of male mice
Collapse
|
40
|
Mori K, Sakano H. Neural Circuitry for Stress Information of Environmental and Internal Odor Worlds. Front Behav Neurosci 2022; 16:943647. [PMID: 35783233 PMCID: PMC9245520 DOI: 10.3389/fnbeh.2022.943647] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Accepted: 05/27/2022] [Indexed: 11/16/2022] Open
Abstract
In mammals, odor information detected in the olfactory epithelium is converted to a topographic map of activated glomeruli in the olfactory bulb. Odor signals are then conveyed by projection neurons to the olfactory cortex for decision making. Odor information is processed by two distinct pathways, one is innate and the other is learned, which are separately activated during exhalation and inhalation, respectively. There are two types of odor signals, exteroceptive and interoceptive, which are also processed in different phases of respiration. Exteroceptive sensory information whether attractive/pleasant or aversive/stressful, is evaluated by the valence regions in the amygdala. Stress is an alert signal telling the body to take an action so that the normal condition can be recovered. When the odor quality is negative, the brain sets up a behavioral strategy to avoid the danger or to improve the situation. In this review article, we will describe the recent progress in the study of olfactory perception focusing on stress responses to external and internal odors.
Collapse
Affiliation(s)
- Kensaku Mori
- RIKEN Center for Brain Science, Wako, Japan
- *Correspondence: Kensaku Mori,
| | - Hitoshi Sakano
- Department of Brain Function, School of Medical Sciences, University of Fukui, Fukui, Japan
- Hitoshi Sakano,
| |
Collapse
|
41
|
Wang Z, Zheng R, Wang X, Huang X, Huang J, Gu C, He Y, Wu S, Chen J, Yang Q, Qiu P. Aerobic Exercise Improves Methamphetamine-Induced Olfactory Dysfunction Through α-Synuclein Intervention in Male Mice. Front Mol Neurosci 2022; 15:884790. [PMID: 35586307 PMCID: PMC9108672 DOI: 10.3389/fnmol.2022.884790] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Accepted: 04/04/2022] [Indexed: 12/24/2022] Open
Abstract
Methamphetamine (Meth) is a predominantly abused neurostimulant, and its abuse is often associated with multiple neurological symptoms. Olfaction, the sense of smell, is a highly neurotransmission-dependent physiological process; however, the effect of Meth on olfactory function and its underlying mechanisms remain largely unknown. This study aimed to explore the impact of Meth abuse on the olfactory system and the potential mechanisms. Chronic Meth abuse was induced by daily administration of Meth in male mice for 4 weeks, and we then systematically examined olfactory performance. Behavioral tests found that Meth-treated animals showed increased olfactory threshold, decreased olfactory sensitivity, reduced olfactory-dependent discrimination, and difficulty in seeking buried food. Notably, the increased deposition of α-synuclein (α-syn) in the olfactory bulb was detected. Adeno-associated virus (AAV)-mediated α-syn intervention therapy in the olfactory bulb significantly alleviated Meth-induced olfactory function impairment, and 8 weeks of aerobic exercise showed similar effects through the same principle of α-syn intervention. Notably, exercise-mediated reduction of α-syn inhibited abnormal firing activity and restored the inhibitory synaptic regulation of mitral cells in the olfactory bulb. These findings suggest the involvement of α-syn in the pathogenic mechanisms of Meth-induced olfactory dysfunction and shed light on the possible therapeutic applications of aerobic exercise in Meth-induced olfactory dysfunction.
Collapse
|
42
|
Cheng YT, Woo J, Deneen B. Sculpting Astrocyte Diversity through Circuits and Transcription. Neuroscientist 2022:10738584221082620. [PMID: 35373633 PMCID: PMC9526762 DOI: 10.1177/10738584221082620] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Astrocytes are the most abundant glial cell in the central nervous system and occupy a wide range of roles that are essential for brain function. Over the past few years, evidence has emerged that astrocytes exhibit cellular and molecular heterogeneity, raising the possibility that subsets of astrocytes are functionally distinct and that transcriptional mechanisms are involved in encoding this prospective diversity. In this review, we focus on three emerging areas of astrocyte biology: region-specific circuit regulation, molecular diversity, and transcriptional regulation. This review highlights our nascent understanding of how molecular diversity is converted to functional diversity of astrocytes through the lens of brain region-specific circuits. We articulate our understanding of how transcriptional mechanisms regulate this diversity and key areas that need further exploration to achieve the overarching goal of a functional taxonomy of astrocytes in the brain.
Collapse
Affiliation(s)
- Yi-Ting Cheng
- Program in Developmental Biology, Baylor College of Medicine, Houston, Houston, TX, USA.,Center for Cell and Gene Therapy, Texas Children's Hospital, Houston, TX, USA
| | - Junsung Woo
- Center for Cell and Gene Therapy, Texas Children's Hospital, Houston, TX, USA
| | - Benjamin Deneen
- Program in Developmental Biology, Baylor College of Medicine, Houston, Houston, TX, USA.,Center for Cell and Gene Therapy, Texas Children's Hospital, Houston, TX, USA.,Department of Neurosurgery, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
43
|
Mori K, Sakano H. Processing of Odor Information During the Respiratory Cycle in Mice. Front Neural Circuits 2022; 16:861800. [PMID: 35431818 PMCID: PMC9008203 DOI: 10.3389/fncir.2022.861800] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 03/07/2022] [Indexed: 12/23/2022] Open
Abstract
In the mouse olfactory system, odor signals detected in the olfactory epithelium are converted to a topographic map of activated glomeruli in the olfactory bulb. The map information is then conveyed by projection neurons, mitral cells and tufted cells, to various areas in the olfactory cortex. An odor map is transmitted to the anterior olfactory nucleus by tufted cells for odor identification and recollection of associated memory for learned decisions. For instinct decisions, odor information is directly transmitted to the valence regions in the amygdala by specific subsets of mitral cells. Transmission of orthonasal odor signals through these two distinct pathways, innate and learned, are closely related with exhalation and inhalation, respectively. Furthermore, the retronasal/interoceptive and orthonasal/exteroceptive signals are differentially processed during the respiratory cycle, suggesting that these signals are processed in separate areas of the olfactory bulb and olfactory cortex. In this review article, the recent progress is summarized for our understanding of the olfactory circuitry and processing of odor signals during respiration.
Collapse
Affiliation(s)
- Kensaku Mori
- RIKEN Center for Brain Science, WAKO, Saitama, Japan
- *Correspondence: Kensaku Mori,
| | - Hitoshi Sakano
- Department of Brain Function, School of Medical Sciences, University of Fukui, Fukui, Japan
- Hitoshi Sakano,
| |
Collapse
|
44
|
Soares GLDS, Leão ERLPD, Freitas SF, Alves RMC, Tavares NDP, Costa MVN, Menezes GCD, Oliveira JHPD, Guerreiro LCF, Assis ACLD, Araújo SC, Franco FTDC, Anaissi AKM, Carmo ELD, Morais RDAPB, Demachki S, Diniz JAP, Nunes HM, Anthony DC, Diniz DG, Diniz CWP. Behavioral and Neuropathological Changes After Toxoplasma gondii Ocular Conjunctival Infection in BALB/c Mice. Front Cell Infect Microbiol 2022; 12:812152. [PMID: 35372100 PMCID: PMC8965508 DOI: 10.3389/fcimb.2022.812152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 02/07/2022] [Indexed: 11/15/2022] Open
Abstract
Ocular infection with Toxoplasma gondii causes toxoplasmosis in mice. However, following ocular infection with tachyzoites, the cause of the accompanying progressive changes in hippocampal-dependent tasks, and their relationship with the morphology and number of microglia, is less well understood. Here, in 6-month-old, female BALB/c mice, 5 μl of a suspension containing 48.5 × 106 tachyzoites/ml was introduced into the conjunctival sac; control received an equal volume of saline. Before and after instillation, all mice were subject to an olfactory discrimination (OD) test, using predator (cat) feces, and to an open-field (OF) task. After the behavioral tests, the animals were culled at either 22 or 44 days post-instillation (dpi), and the brains and retinas were dissected and processed for immunohistochemistry. The total number of Iba-1-immunolabeled microglia in the molecular layer of the dentate gyrus was estimated, and three-dimensional reconstructions of the cells were evaluated. Immobility was increased in the infected group at 12, 22, and 43 dpi, but the greatest immobility was observed at 22 dpi and was associated with reduced line crossing in the OF and distance traveled. In the OD test, infected animals spent more time in the compartment with feline fecal material at 14 and at 43 dpi. No OD changes were observed in the control group. The number of microglia was increased at 22 dpi but returned to control levels by 44 dpi. These changes were associated with the differentiation of T. gondii tachyzoites into bradyzoite-enclosed cysts within the brain and retina. Thus, infection of mice with T. gondii alters exploratory behavior, gives rise to a loss in predator’s odor avoidance from 2 weeks after infection, increased microglia number, and altered their morphology in the molecular layer of the dentate gyrus.
Collapse
|
45
|
Whitlock KE, Palominos MF. The Olfactory Tract: Basis for Future Evolution in Response to Rapidly Changing Ecological Niches. Front Neuroanat 2022; 16:831602. [PMID: 35309251 PMCID: PMC8927807 DOI: 10.3389/fnana.2022.831602] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 02/01/2022] [Indexed: 01/10/2023] Open
Abstract
Within the forebrain the olfactory sensory system is unique from other sensory systems both in the projections of the olfactory tract and the ongoing neurogenic potential, characteristics conserved across vertebrates. Olfaction plays a crucial role in behaviors such as mate choice, food selection, homing, escape from predators, among others. The olfactory forebrain is intimately associated with the limbic system, the region of the brain involved in learning, memory, and emotions through interactions with the endocrine system and the autonomic nervous system. Previously thought to lack a limbic system, we now know that teleost fishes process emotions, have exceptional memories, and readily learn, behaviors that are often associated with olfactory cues. The association of neuromodulatory hormones, and more recently, the immune system, with odor cues underlies behaviors essential for maintenance and adaptation within natural ecological niches. Increasingly anthropogenic perturbations affecting ecosystems are impacting teleost fishes worldwide. Here we examine the role of the olfactory tract as the neural basis for the integration of environmental cues and resulting behaviors necessary for the regulation of biotic interactions that allow for future adaptation as the climate spins out of control.
Collapse
Affiliation(s)
- Kathleen E. Whitlock
- Centro Interdisciplinario de Neurociencia de Valparaíso (CINV), Universidad de ValparaísoValparaíso, Chile
- Instituto de Neurociencia, Universidad de ValparaísoValparaíso, Chile
- *Correspondence: Kathleen E. Whitlock
| | - M. Fernanda Palominos
- Centro Interdisciplinario de Neurociencia de Valparaíso (CINV), Universidad de ValparaísoValparaíso, Chile
- Instituto de Neurociencia, Universidad de ValparaísoValparaíso, Chile
| |
Collapse
|
46
|
Prior NH, Bentz EJ, Ophir AG. Reciprocal processes of sensory perception and social bonding: an integrated social-sensory framework of social behavior. GENES, BRAIN, AND BEHAVIOR 2022; 21:e12781. [PMID: 34905293 PMCID: PMC9744507 DOI: 10.1111/gbb.12781] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/04/2021] [Revised: 11/09/2021] [Accepted: 11/10/2021] [Indexed: 02/06/2023]
Abstract
Organisms filter the complexity of natural stimuli through their individual sensory and perceptual systems. Such perceptual filtering is particularly important for social stimuli. A shared "social umwelt" allows individuals to respond appropriately to the expected diversity of cues and signals during social interactions. In this way, the behavioral and neurobiological mechanisms of sociality and social bonding cannot be disentangled from perceptual mechanisms and sensory processing. While a degree of embeddedness between social and sensory processes is clear, our dominant theoretical frameworks favor treating the social and sensory processes as distinct. An integrated social-sensory framework has the potential to greatly expand our understanding of the mechanisms underlying individual variation in social bonding and sociality more broadly. Here we leverage what is known about sensory processing and pair bonding in two common study systems with significant species differences in their umwelt (rodent chemosensation and avian acoustic communication). We primarily highlight that (1) communication is essential for pair bond formation and maintenance, (2) the neural circuits underlying perception, communication and social bonding are integrated, and (3) candidate neuromodulatory mechanisms that regulate pair bonding also impact communication and perception. Finally, we propose approaches and frameworks that more fully integrate sensory processing, communication, and social bonding across levels of analysis: behavioral, neurobiological, and genomic. This perspective raises two key questions: (1) how is social bonding shaped by differences in sensory processing?, and (2) to what extent is sensory processing and the saliency of signals shaped by social interactions and emerging relationships?
Collapse
Affiliation(s)
- Nora H. Prior
- Department of PsychologyCornell UniversityIthacaNew YorkUSA
| | - Ehren J. Bentz
- Department of PsychologyCornell UniversityIthacaNew YorkUSA
| | | |
Collapse
|
47
|
Schreck MR, Zhuang L, Janke E, Moberly AH, Bhattarai JP, Gottfried JA, Wesson DW, Ma M. State-dependent olfactory processing in freely behaving mice. Cell Rep 2022; 38:110450. [PMID: 35235805 PMCID: PMC8958632 DOI: 10.1016/j.celrep.2022.110450] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 11/07/2021] [Accepted: 02/07/2022] [Indexed: 11/06/2022] Open
Abstract
Decreased responsiveness to sensory stimuli during sleep is presumably mediated via thalamic gating. Without an obligatory thalamic relay in the olfactory system, the anterior piriform cortex (APC) is suggested to be a gate in anesthetized states. However, olfactory processing in natural sleep states remains undetermined. Here, we simultaneously record local field potentials (LFPs) in hierarchical olfactory regions (olfactory bulb [OB], APC, and orbitofrontal cortex) while optogenetically activating olfactory sensory neurons, ensuring consistent peripheral inputs across states in behaving mice. Surprisingly, evoked LFPs in sleep states (both non-rapid eye movement [NREM] and rapid eye movement [REM]) are larger and contain greater gamma-band power and cross-region coherence (compared to wakefulness) throughout the olfactory pathway, suggesting the lack of a central gate. Single-unit recordings from the OB and APC reveal a higher percentage of responsive neurons during sleep with a higher incidence of suppressed firing. Additionally, nasal breathing is slower and shallower during sleep, suggesting a partial peripheral gating mechanism. Schreck et al. examine how the olfactory system responds to the same peripheral stimulus during natural sleep and wake in mice. Larger responses along the pathway during sleep suggest the lack of a central gate, but slower and shallower breathing may act as a partial peripheral gate to reduce olfactory input.
Collapse
Affiliation(s)
- Mary R Schreck
- Department of Neuroscience, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA.
| | - Liujing Zhuang
- Department of Neuroscience, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Emma Janke
- Department of Neuroscience, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Andrew H Moberly
- Department of Neuroscience, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Janardhan P Bhattarai
- Department of Neuroscience, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Jay A Gottfried
- Department of Psychology, University of Pennsylvania, School of Arts and Sciences; Department of Neurology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Daniel W Wesson
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, FL 32610, USA
| | - Minghong Ma
- Department of Neuroscience, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA.
| |
Collapse
|
48
|
Hasegawa Y, Namkung H, Smith A, Sakamoto S, Zhu X, Ishizuka K, Lane AP, Sawa A, Kamiya A. Causal impact of local inflammation in the nasal cavity on higher brain function and cognition. Neurosci Res 2021; 172:110-115. [PMID: 33932551 PMCID: PMC10693917 DOI: 10.1016/j.neures.2021.04.009] [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: 03/16/2021] [Revised: 04/21/2021] [Accepted: 04/25/2021] [Indexed: 12/20/2022]
Abstract
Epidemiological evidence suggests that adverse environmental factors in the nasal cavity may increase the risk for neuropsychiatric diseases. For instance, air pollution and nasal viral infection have been underscored as risk factors for Parkinson's disease, schizophrenia, and mood disorders. These adverse factors can elicit local inflammation in the nasal cavity, which may in turn influence higher brain function. Nevertheless, evidence that directly supports their causal link is missing. To fill this knowledge gap, we used an inducible mouse model for olfactory inflammation and showed the evidence that this local pathological factor can elicit behavioral abnormalities.
Collapse
Affiliation(s)
- Yuto Hasegawa
- Department of Psychiatry, Johns Hopkins University School of Medicine, 600 N. Wolfe Street, Baltimore, MD, 21287, USA
| | - Ho Namkung
- Department of Psychiatry, Johns Hopkins University School of Medicine, 600 N. Wolfe Street, Baltimore, MD, 21287, USA; Department of Biomedical Engineering, Johns Hopkins University School of Medicine, 600 N. Wolfe Street, Baltimore, MD, 21287, USA
| | - Amy Smith
- Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins University School of Medicine, 600 N. Wolfe Street, Baltimore, MD, 21287, USA
| | - Shinji Sakamoto
- Department of Psychiatry, Johns Hopkins University School of Medicine, 600 N. Wolfe Street, Baltimore, MD, 21287, USA
| | - Xiaolei Zhu
- Department of Psychiatry, Johns Hopkins University School of Medicine, 600 N. Wolfe Street, Baltimore, MD, 21287, USA
| | - Koko Ishizuka
- Department of Psychiatry, Johns Hopkins University School of Medicine, 600 N. Wolfe Street, Baltimore, MD, 21287, USA
| | - Andrew P Lane
- Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins University School of Medicine, 600 N. Wolfe Street, Baltimore, MD, 21287, USA
| | - Akira Sawa
- Department of Psychiatry, Johns Hopkins University School of Medicine, 600 N. Wolfe Street, Baltimore, MD, 21287, USA; Department of Biomedical Engineering, Johns Hopkins University School of Medicine, 600 N. Wolfe Street, Baltimore, MD, 21287, USA; Department of Neuroscience, Johns Hopkins University School of Medicine, 600 N. Wolfe Street, Baltimore, MD, 21287, USA; Department of Genetic Medicine, Johns Hopkins University School of Medicine, 600 N. Wolfe Street, Baltimore, MD, 21287, USA; Department of Mental Health, Johns Hopkins University Bloomberg School of Public Health, 600 N. Wolfe Street, Baltimore, MD, 21287, USA.
| | - Atsushi Kamiya
- Department of Psychiatry, Johns Hopkins University School of Medicine, 600 N. Wolfe Street, Baltimore, MD, 21287, USA.
| |
Collapse
|
49
|
Schreck M, Ma M. Sensory neuroscience: Early value-based odor categorization. Curr Biol 2021; 31:R396-R398. [PMID: 33905700 DOI: 10.1016/j.cub.2021.03.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
How the brain categorizes external stimuli in an experience-dependent, behaviorally relevant manner is a fundamental question. A new study reveals that mitral cells in the olfactory bulb of mice dynamically represent value- and category-related odor information in learned behavioral tasks.
Collapse
Affiliation(s)
- Mary Schreck
- Department of Neuroscience, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Minghong Ma
- Department of Neuroscience, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA.
| |
Collapse
|
50
|
Inoue N, Nishizumi H, Ooyama R, Mogi K, Nishimori K, Kikusui T, Sakano H. The olfactory critical period is determined by activity-dependent Sema7A/PlxnC1 signaling within glomeruli. eLife 2021; 10:65078. [PMID: 33780330 PMCID: PMC8007213 DOI: 10.7554/elife.65078] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2020] [Accepted: 03/18/2021] [Indexed: 12/26/2022] Open
Abstract
In mice, early exposure to environmental odors affects social behaviors later in life. A signaling molecule, Semaphorin 7A (Sema7A), is induced in the odor-responding olfactory sensory neurons. Plexin C1 (PlxnC1), a receptor for Sema7A, is expressed in mitral/tufted cells, whose dendrite-localization is restricted to the first week after birth. Sema7A/PlxnC1 signaling promotes post-synaptic events and dendrite selection in mitral/tufted cells, resulting in glomerular enlargement that causes an increase in sensitivity to the experienced odor. Neonatal odor experience also induces positive responses to the imprinted odor. Knockout and rescue experiments indicate that oxytocin in neonates is responsible for imposing positive quality on imprinted memory. In the oxytocin knockout mice, the sensitivity to the imprinted odor increases, but positive responses cannot be promoted, indicating that Sema7A/PlxnC1 signaling and oxytocin separately function. These results give new insights into our understanding of olfactory imprinting during the neonatal critical period.
Collapse
Affiliation(s)
- Nobuko Inoue
- Department of Brain Function, School of Medical Sciences, University of Fukui, Matsuoka, Japan
| | - Hirofumi Nishizumi
- Department of Brain Function, School of Medical Sciences, University of Fukui, Matsuoka, Japan
| | - Rumi Ooyama
- Department of Animal Science and Biotechnology, School of Veterinary Medicine, Azabu University, Sagamihara, Japan
| | - Kazutaka Mogi
- Department of Animal Science and Biotechnology, School of Veterinary Medicine, Azabu University, Sagamihara, Japan
| | - Katsuhiko Nishimori
- Department of Molecular and Cell Biology, Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
| | - Takefumi Kikusui
- Department of Animal Science and Biotechnology, School of Veterinary Medicine, Azabu University, Sagamihara, Japan
| | - Hitoshi Sakano
- Department of Brain Function, School of Medical Sciences, University of Fukui, Matsuoka, Japan
| |
Collapse
|