1
|
Naffaa MM. Neurogenesis dynamics in the olfactory bulb: deciphering circuitry organization, function, and adaptive plasticity. Neural Regen Res 2025; 20:1565-1581. [PMID: 38934393 PMCID: PMC11688548 DOI: 10.4103/nrr.nrr-d-24-00312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 05/20/2024] [Accepted: 05/31/2024] [Indexed: 06/28/2024] Open
Abstract
Adult neurogenesis persists after birth in the subventricular zone, with new neurons migrating to the granule cell layer and glomerular layers of the olfactory bulb, where they integrate into existing circuitry as inhibitory interneurons. The generation of these new neurons in the olfactory bulb supports both structural and functional plasticity, aiding in circuit remodeling triggered by memory and learning processes. However, the presence of these neurons, coupled with the cellular diversity within the olfactory bulb, presents an ongoing challenge in understanding its network organization and function. Moreover, the continuous integration of new neurons in the olfactory bulb plays a pivotal role in regulating olfactory information processing. This adaptive process responds to changes in epithelial composition and contributes to the formation of olfactory memories by modulating cellular connectivity within the olfactory bulb and interacting intricately with higher-order brain regions. The role of adult neurogenesis in olfactory bulb functions remains a topic of debate. Nevertheless, the functionality of the olfactory bulb is intricately linked to the organization of granule cells around mitral and tufted cells. This organizational pattern significantly impacts output, network behavior, and synaptic plasticity, which are crucial for olfactory perception and memory. Additionally, this organization is further shaped by axon terminals originating from cortical and subcortical regions. Despite the crucial role of olfactory bulb in brain functions and behaviors related to olfaction, these complex and highly interconnected processes have not been comprehensively studied as a whole. Therefore, this manuscript aims to discuss our current understanding and explore how neural plasticity and olfactory neurogenesis contribute to enhancing the adaptability of the olfactory system. These mechanisms are thought to support olfactory learning and memory, potentially through increased complexity and restructuring of neural network structures, as well as the addition of new granule granule cells that aid in olfactory adaptation. Additionally, the manuscript underscores the importance of employing precise methodologies to elucidate the specific roles of adult neurogenesis amidst conflicting data and varying experimental paradigms. Understanding these processes is essential for gaining insights into the complexities of olfactory function and behavior.
Collapse
Affiliation(s)
- Moawiah M. Naffaa
- Department of Psychology and Neuroscience, Duke University, Durham, NC, USA
- Department of Cell Biology, Duke University School of Medicine, Durham, NC, USA
| |
Collapse
|
2
|
Castelló ME, Olivera-Pasilio V, Rosillo JC, Fernández AS. Adult neurogenesis in the Uruguayan teleost species Austrolebias charrua and Gymnotus omarorum. Neuroscience 2025; 573:143-153. [PMID: 40101892 DOI: 10.1016/j.neuroscience.2025.03.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 02/19/2025] [Accepted: 03/14/2025] [Indexed: 03/20/2025]
Abstract
Neurogenesis -the process by which new neurons are generated in the brain- is critical for nervous system development and plasticity. Adult neurogenesis underlies growth, repair, and adaptation to environmental changes. Vertebrates differ in their neurogenic and regenerative capacity, being particularly prominent in teleost as adult neurogenesis occurs throughout the rostral-caudal brain axis. This review examines adult proliferation and neurogenesis in the autochthonous Uruguayan teleost Austrolebias charrua and Gymnotus omarorum. A. charrua are annual fishes that live in temporary freshwater pools that dry up in the summer. The luminosity of the puddles varies greatly, and both vision and olfaction are crucial for the survival of this species. G. omarorum inhabits freshwater lagoons and rivers beneath dense masses of floating plants and have nocturnal habits. They rely on the electrosensory modality to navigate and communicate with conspecifics. These differences in habitats and predominant sensory modalities are reflected in the distinct brain morphotypes of G. omarorum and A. charrua. While G. omarorum is characterized by the hypertrophy of rhombencephalic cerebellum and electrosensory lateral line lobe, A. charrua has a well-developed olfactory bulb, mesencephalic tectum opticum, and torus longitudinalis. Accordingly, these regions have notorious neurogenic activity. Differences in neuroanatomy and distribution of neurogenesis in the brains of both species are discussed considering their life cycle and lifestyle. The comparison of these results with those reported in other teleost and vertebrates contributes to the understanding of the key role of neurogenesis in brain plasticity and evolution.
Collapse
Affiliation(s)
- María E Castelló
- Laboratorio de Desarrollo Y Evolución Neural, Departamento de Neurociencias Integrativas Y Computacionales, Instituto de Investigaciones Biológicas Clemente Estable, (IIBCE-MEC), Avenida. Italia 3318 11600 Montevideo, Uruguay.
| | - Valentina Olivera-Pasilio
- Laboratorio de Desarrollo Y Evolución Neural, Departamento de Neurociencias Integrativas Y Computacionales, Instituto de Investigaciones Biológicas Clemente Estable, (IIBCE-MEC), Avenida. Italia 3318 11600 Montevideo, Uruguay; Department of Psychiatry and Behavioral Sciences, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA.
| | - Juan Carlos Rosillo
- Laboratorio de Neurobiología Comparada, Departamento Neurociencias Integrativas Y Computacionales, Instituto de Investigaciones Biológicas Clemente Estable (IIBCE-MEC), Uruguay; Departamento de Histología Y Embriología, Facultad de Medicina, UdelaR. Avda. General Flores 2125 11800 Montevideo, Uruguay; Departamento de Neurobiología Y Neuropatología, Instituto de Investigaciones Biológicas Clemente Estable (IIBCE-MEC), Avenida. Italia 3318 11600 Montevideo, Uruguay.
| | - Anabel S Fernández
- Laboratorio de Neurobiología Comparada, Departamento Neurociencias Integrativas Y Computacionales, Instituto de Investigaciones Biológicas Clemente Estable (IIBCE-MEC), Uruguay; Laboratorio de Neurociencias, Instituto de Biología, Facultad de Ciencias, UdelaR, Iguá 4225 11400 Montevideo, Uruguay.
| |
Collapse
|
3
|
Sepúlveda-Cuéllar RD, Soria-Medina DA, Cañedo-Solares I, Gómez-Chávez F, Molina-López LM, Cruz-Martínez MY, Correa D. Controversies and insights into cytokine regulation of neurogenesis and behavior in adult rodents. Front Immunol 2025; 16:1550660. [PMID: 40352932 PMCID: PMC12061686 DOI: 10.3389/fimmu.2025.1550660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Accepted: 03/24/2025] [Indexed: 05/14/2025] Open
Abstract
Adult learning, memory, and social interaction partially depend on neurogenesis in two regions: the hippocampus and the subventricular zone. There is evidence that the immune system is important for these processes in pathological situations, but there is no review of its role in non-pathological or near-physiological conditions. Although further research is warranted in this area, some conclusions can be drawn. Intrusive LyC6hi monocytes and autoreactive CD4+ T cells have a positive impact on neurogenesis and behavior, but the latter are deleterious if specific to external antigens. Mildly activated microglia play a crucial role in promoting these processes, by eliminating apoptotic neuronal progenitors and producing low levels of interleukins, which increase if the cells are activated, leading to inhibition of neurogenesis. Chemokines are poorly studied, but progenitor cells and neurons express their receptors, which appear important for migration and maturation. The few works that jointly analyzed neurogenesis and behavior showed congruent effects of immune cells and cytokines. In conclusion, the immune system components -mostly local- seem of utmost importance for the control of behavior under non-pathological conditions.
Collapse
Affiliation(s)
- Rodrigo Daniel Sepúlveda-Cuéllar
- Programa de Doctorado en Ciencias Biomédicas, Universidad Nacional Autónoma de México (UNAM), Ciudad de México, Mexico
- Centro de Investigación en Ciencias de la Salud, Facultad de Ciencias de la Salud, Universidad Anáhuac México, Huixquilucan, EdoMex, Mexico
| | - Diego Alberto Soria-Medina
- Centro de Investigación en Ciencias de la Salud, Facultad de Ciencias de la Salud, Universidad Anáhuac México, Huixquilucan, EdoMex, Mexico
- Facultad de Psicología, Universidad Nacional Autónoma de México (UNAM), Ciudad de México, Mexico
| | - Irma Cañedo-Solares
- Laboratorio de Inmunología Experimental, Instituto Nacional de Pediatría (INP), Secretaría de Salud, Ciudad de México, Mexico
| | - Fernando Gómez-Chávez
- Laboratorio de Enfermedades Osteoarticulares e Inmunológicas, Sección de Estudios de Posgrado e Investigación, Escuela Nacional de Medicina y Homeopatía (ENMyH), Instituto Politécnico Nacional (IPN), Ciudad de México, Mexico
| | - Liliana Monserrat Molina-López
- Centro de Investigación en Ciencias de la Salud, Facultad de Ciencias de la Salud, Universidad Anáhuac México, Huixquilucan, EdoMex, Mexico
| | - María Yolanda Cruz-Martínez
- Centro de Investigación en Ciencias de la Salud, Facultad de Ciencias de la Salud, Universidad Anáhuac México, Huixquilucan, EdoMex, Mexico
| | - Dolores Correa
- Centro de Investigación en Ciencias de la Salud, Facultad de Ciencias de la Salud, Universidad Anáhuac México, Huixquilucan, EdoMex, Mexico
| |
Collapse
|
4
|
Liao SF, Chan TC, Su MH, Lin MC, Wu CS, Fan CC, Wang SH. The independent role of fine particulate matter and genetic liability on cognition in older adults. Ann Gen Psychiatry 2025; 24:20. [PMID: 40181397 PMCID: PMC11969746 DOI: 10.1186/s12991-025-00559-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Accepted: 03/23/2025] [Indexed: 04/05/2025] Open
Abstract
BACKGROUND Genetic susceptibility to mental health and cognitive traits, as well as air pollution, significantly impact cognition. The interplay between polygenic liability and fine particulate matter (PM2.5) remains unclear due to the limited number of large-scale studies in Asia. This study utilized the Taiwan Biobank, a nationwide community-based database, to investigate the main and modified effect of PM2.5 on individuals' polygenic susceptibility in cognition. METHODS Polygenic risk score (PRS) for cognitive performance (CP PRS), Alzheimer's disease (AD PRS), schizophrenia (SCZ PRS), and major depression (MDD PRS) were computed representing genetic susceptibility for an individual. APOE genotype was classified into E3/E3, E3/E4, and E4/E4. The five-year average concentration of PM2.5 from satellite images was used for defining environmental exposure. Cognitive performance was evaluated via the Mini-Mental State Examination (MMSE) score. The association between personal genetic susceptibility, PM2.5, and cognitive performance was examined using multilevel linear regression with the adjustment of age, sex, batch effect, and population stratification effect. The gene-environment synergism was examined with the inclusion of product term of PM2.5 and PRS in the multivariate model. RESULTS Our analyses included 25,593 participants from 164 townships. Participants exposed to higher PM2.5 concentrations had a lower MMSE score (Beta=-0.0830 corresponding to a 1 µg/m3 increase in PM2.5 concentration, 95% CI, -0.0973 to -0.0688, p-value < 0.0001). After controlling for PM2.5 concentration, CP PRS (Beta = 0.1729, 95% CI, 0.1470 to 0.1988, p-value < 0.0001), SCZ PRS (Beta=-0.0632, 95% CI, -0.0891 to -0.0374, p-value < 0.0001), and AD PRS (Beta=-0.0321, 95% CI, -0.0580 to -0.0062, p-value = 0.0153) were associated with MMSE score. After further examination of gene-environment synergism, no interaction effect was identified, indicating different mechanism of PM2.5 and genetic liability to influence cognitive performance. CONCLUSIONS Human polygenic loading and PM2.5 may impact cognition via an independent pathway. A prevention strategy targeting air pollution reduction may effectively improve the cognitive performance. Multiple exposures and their influences on the long-term change of cognition were required in future research.
Collapse
Affiliation(s)
- Shu-Fen Liao
- Department of Medical Research, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
- School of Public Health, College of Public Health, Taipei Medical University, Taipei, Taiwan
| | - Ta-Chien Chan
- Research Center for Humanities and Social Sciences, Academia Sinica, Taipei, Taiwan
- Institute of Public Health, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Mei-Hsin Su
- Department of Public Health, College of Public Health, China Medical University, Taichung, Taiwan
- Department of Psychiatry, Virginia Institute for Psychiatric Behavioral Genetics, Virginia Commonwealth University, Richmond, VA, USA
| | - Mei-Chen Lin
- National Center for Geriatrics and Welfare Research, National Health Research Institutes, 35, Keyan Road, Zhunan Town, Miaoli County 350, Miaoli, Taiwan
| | - Chi-Shin Wu
- National Center for Geriatrics and Welfare Research, National Health Research Institutes, 35, Keyan Road, Zhunan Town, Miaoli County 350, Miaoli, Taiwan
- Department of Psychiatry, National Taiwan University Hospital, Yunlin branch, Douliu, Taiwan
| | - Chun-Chieh Fan
- Center for Population Neuroscience and Genetics, Laureate Institute for Brain Research, Tulsa, OK, USA
- Department of Radiology, School of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Shi-Heng Wang
- National Center for Geriatrics and Welfare Research, National Health Research Institutes, 35, Keyan Road, Zhunan Town, Miaoli County 350, Miaoli, Taiwan.
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan.
| |
Collapse
|
5
|
Hossain K, Smith M, Rufenacht KE, O’Rourke R, Santoro SW. In mice, discrete odors can selectively promote the neurogenesis of sensory neuron subtypes that they stimulate. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.02.10.579748. [PMID: 38405728 PMCID: PMC10888860 DOI: 10.1101/2024.02.10.579748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/27/2024]
Abstract
In mammals, olfactory sensory neurons (OSNs) are born throughout life, ostensibly solely to replace neurons lost via turnover or injury. This assumption follows from the hypothesis that olfactory neurogenesis is stochastic with respect to neuron subtype, as defined by the single odorant receptor that each neural precursor stochastically chooses out of hundreds of possibilities. This assumption is challenged, however, by recent findings that the birthrates of a fraction of OSN subtypes are selectively reduced by olfactory deprivation. These findings raise questions about how, and why, olfactory stimuli are required to accelerate the neurogenesis rates of some subtypes, including whether the stimuli are specific (e.g., discrete odorants) or generic (e.g., broadly activating odors or mechanical stimuli). Based on previous findings that the exposure of mice to sex-specific odors can increase the representations of subtypes responsive to those odors, we hypothesized that the neurogenic stimuli comprise discrete odorants that selectively stimulate OSNs of the same subtypes whose birthrates are accelerated. In support of this, we have found, using scRNA-seq and subtype-specific OSN birthdating, that exposure to male and exogenous musk odors can accelerate the birthrates of subtypes responsive to those odors. These findings reveal that certain odor experiences can selectively 'amplify' specific OSN subtypes and suggest that persistent OSN neurogenesis serves, in part, an adaptive function.
Collapse
Affiliation(s)
- Kawsar Hossain
- Department of Pediatrics, Section of Developmental Biology, University of Colorado School of Medicine, Aurora, CO, USA
- Current affiliation: Department of Molecular and Cellular Biosciences, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Madeline Smith
- Department of Pediatrics, Section of Developmental Biology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Karlin E. Rufenacht
- Department of Pediatrics, Section of Developmental Biology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Rebecca O’Rourke
- Department of Pediatrics, Section of Developmental Biology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Stephen W. Santoro
- Department of Pediatrics, Section of Developmental Biology, University of Colorado School of Medicine, Aurora, CO, USA
| |
Collapse
|
6
|
Meller SJ, Greer CA. Olfactory Development and Dysfunction: Involvement of Microglia. Physiology (Bethesda) 2025; 40:0. [PMID: 39499248 DOI: 10.1152/physiol.00037.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 10/31/2024] [Accepted: 11/01/2024] [Indexed: 11/07/2024] Open
Abstract
Olfactory deficits are increasingly recognized in a variety of neurological, neurodevelopmental, psychiatric, and viral diseases. While the pathology underlying olfactory loss is likely to differ across diseases, one shared feature may be an immune response mediated by microglia. Microglia orchestrate the brain's response to environmental insults and maintain neurodevelopmental homeostasis. Here, we explore the potential involvement of microglia in olfactory development and loss in disease. The effects of microglia-mediated immune response during development may be of special relevance to the olfactory system, which is unique in both its vulnerability to environmental insults as well as its extended period of neurogenesis and neuronal migration.
Collapse
Affiliation(s)
- Sarah J Meller
- Departments of Neuroscience, Yale University School of Medicine, New Haven, Connecticut, United States
- Neurosurgery, Yale University School of Medicine, New Haven, Connecticut, United States
- The Interdepartmental Neuroscience Graduate Program, Yale University School of Medicine, New Haven, Connecticut, United States
| | - Charles A Greer
- Departments of Neuroscience, Yale University School of Medicine, New Haven, Connecticut, United States
- Neurosurgery, Yale University School of Medicine, New Haven, Connecticut, United States
- The Interdepartmental Neuroscience Graduate Program, Yale University School of Medicine, New Haven, Connecticut, United States
| |
Collapse
|
7
|
Zhou W, Munoz JR, Henry Ho HY, Hanamura K, Dalva MB. Specific neuroblast-derived signals control both cell migration and fate in the rostral migratory stream. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.18.638163. [PMID: 40027825 PMCID: PMC11870606 DOI: 10.1101/2025.02.18.638163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Functional neuronal circuits require neuroblasts migrate to appropriate locations and then differentiate into neuronal subtypes. However, it remains unknown how neuroblasts in the subventricular zone (SVZ) are guided through the rostral migratory stream (RMS) to the olfactory bulb (OB). Here we define EphB2 as a neuroblast-derived cue that controls migration along the RMS and helps to determine cell fate. Within the RMS, EphB2 is expressed selectively in, kinase-active in, and required for the migration of neuroblasts. As neuroblasts enter the OB and differentiate, EphB kinase activity is down-regulated, and in the granule cell layer (GCL), EphB2 expression is down-regulated. Blocking EphB kinase activity or knocking down EphB2 results in defects in migration and premature cellular differentiation in the RMS. Unexpectedly, premature loss of EphB2 expression causes neuroblasts to stop migrating and differentiate into astrocyte-like cells. Thus, EphB2 kinase activity and expression are linked to migration and specification of neuroblast fate.
Collapse
Affiliation(s)
- Wei Zhou
- Department of Neuroscience and the Jefferson Synaptic Biology Center, Thomas Jefferson University, 900 Walnut Street, Philadelphia, PA 19107, USA
| | - James R. Munoz
- Present address: Department of Psychology and Neuroscience, Nova Southeastern University, 3301 College Avenue, Fort Lauderdale, FL 33314, USA
| | - Hsin-Yi Henry Ho
- Department of Cell Biology and Human Anatomy, UC Davis School of Medicine, One Shields Avenue, Davis, CA 95616
| | - Kenji Hanamura
- Department of Neuroscience and the Jefferson Synaptic Biology Center, Thomas Jefferson University, 900 Walnut Street, Philadelphia, PA 19107, USA
- Present address: Faculty of Medical Technology, Department of Radiological Technology, Niigata University of Health and Welfare, 1398 Shimami-cho, Kita-ku, Niigata-City, 950-3198, Japan
| | - Matthew B. Dalva
- Department of Neuroscience and the Jefferson Synaptic Biology Center, Thomas Jefferson University, 900 Walnut Street, Philadelphia, PA 19107, USA
- Present Address: Tulane Brain Institute, Tulane University, 201 Flower Hall, 6823 St. Charles Avenue, New Orleans, LA 70118
| |
Collapse
|
8
|
Li J, Raina M, Wang Y, Xu C, Su L, Guo Q, Ferreira RM, Eadon MT, Ma Q, Wang J, Xu D. scBSP: A fast and accurate tool for identifying spatially variable features from high-resolution spatial omics data. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.02.636138. [PMID: 39974940 PMCID: PMC11838397 DOI: 10.1101/2025.02.02.636138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Emerging spatial omics technologies empower comprehensive exploration of biological systems from multi-omics perspectives in their native tissue location in two and three-dimensional space. However, sparse sequencing capacity and growing spatial resolution in spatial omics present significant computational challenges in identifying biologically meaningful molecules that exhibit variable spatial distributions across different omics. We introduce scBSP, an open-source, versatile, and user-friendly package for identifying spatially variable features in high-resolution spatial omics data. scBSP leverages sparse matrix operation to significantly increase computational efficiency in both computational time and memory usage. In diverse spatial sequencing data and simulations, scBSP consistently and rapidly identifies spatially variable genes and spatially variable peaks across various sequencing techniques and spatial resolutions, handling two- and three-dimensional data with up to millions of cells. It can process high-definition spatial transcriptomics data for 19,950 genes across 181,367 spots within 10 seconds on a typical desktop computer, making it the fastest tool available for handling such high-resolution, sparse spatial omics data while maintaining high accuracy. In a case study of kidney disease using 10x Xenium data, scBSP identified spatially variable genes representative of critical pathological mechanisms associated with histology.
Collapse
Affiliation(s)
- Jinpu Li
- Institute for Data Science and Informatics, University of Missouri, Columbia, MO 65211, USA
- Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, MO 65211, USA
| | - Mauminah Raina
- Department of Biomedical Engineering and Informatics, Indiana University Indianapolis, Indianapolis, IN 46202, USA
| | - Yiqing Wang
- Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, MO 65211, USA
| | - Chunhui Xu
- Institute for Data Science and Informatics, University of Missouri, Columbia, MO 65211, USA
- Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, MO 65211, USA
| | - Li Su
- Institute for Data Science and Informatics, University of Missouri, Columbia, MO 65211, USA
- Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, MO 65211, USA
| | - Qi Guo
- Department of Biomedical Informatics, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Ricardo Melo Ferreira
- Department of Medicine, Indiana University Indianapolis, Indianapolis, IN 46202, USA
| | - Michael T Eadon
- Department of Medicine, Indiana University Indianapolis, Indianapolis, IN 46202, USA
| | - Qin Ma
- Department of Biomedical Informatics, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
- Pelotonia Institute for Immuno-Oncology, The James Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA
| | - Juexin Wang
- Department of Biomedical Engineering and Informatics, Indiana University Indianapolis, Indianapolis, IN 46202, USA
| | - Dong Xu
- Institute for Data Science and Informatics, University of Missouri, Columbia, MO 65211, USA
- Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, MO 65211, USA
- Department of Electrical Engineering and Computer Science, University of Missouri, Columbia, MO 65211, USA
| |
Collapse
|
9
|
Azargoonjahromi A, Abutalebian F, Hoseinpour F. The role of resveratrol in neurogenesis: a systematic review. Nutr Rev 2025; 83:e257-e272. [PMID: 38511504 DOI: 10.1093/nutrit/nuae025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/22/2024] Open
Abstract
CONTEXT Resveratrol (RV) is a natural compound found in grapes, wine, berries, and peanuts and has potential health benefits-namely, neurogenesis improvement. Neurogenesis, which is the process through which new neurons or nerve cells are generated in the brain, occurs in the subventricular zone and hippocampus and is influenced by various factors. RV has been shown to increase neural stem cell proliferation and survival, improving cognitive function in hippocampus-dependent tasks. Thus, to provide a convergent and unbiased conclusion of the available evidence on the correlation between the RV and neurogenesis, a systematic review needs to be undertaken meticulously and with appropriate attention. OBJECTIVE This study aimed to systematically review any potential connection between the RV and neurogenesis in animal models. DATA SOURCES AND EXTRACTION Based on the particular selection criteria, 8 original animal studies that investigated the relationship between RV and neurogenesis were included. Studies written in English and published in peer-reviewed journals with no restrictions on the starting date of publication on August 17, 2023, were searched in the Google Scholar and PubMed databases. Furthermore, data were extracted and analyzed independently by 2 researchers and then reviewed by a third researcher, and discrepancies were resolved by consensus. This project followed PRISMA reporting standards. DATA ANALYSIS In the studies analyzed in this review, there is a definite correlation between RV and neurogenesis, meaning that RV intake, irrespective of the mechanisms thereof, can boost neurogenesis in both the subventricular zone and hippocampus. CONCLUSION This finding, albeit with some limitations, provides a plausible indication of RV's beneficial function in neurogenesis. Indeed, RV intake may result in neurogenesis benefits-namely, cognitive function, mood regulation, stress resilience, and neuroprotection, potentially preventing cognitive decline.
Collapse
Affiliation(s)
| | - Fatemeh Abutalebian
- Department of Biotechnology and Medicine, Islamic Azad University of Tehran Central Branch, Tehran, Iran
| | - Fatemeh Hoseinpour
- Department of Occupational Therapy, Semnan University of Medical Sciences and Health Services, Semnan, Iran
| |
Collapse
|
10
|
Saxena R, McNaughton BL. Bridging Neuroscience and AI: Environmental Enrichment as a model for forward knowledge transfer in continual learning. ARXIV 2025:arXiv:2405.07295v3. [PMID: 38947919 PMCID: PMC11213130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
Continual learning (CL) refers to an agent's capability to learn from a continuous stream of data and transfer knowledge without forgetting old information. One crucial aspect of CL is forward transfer, i.e., improved and faster learning on a new task by leveraging information from prior knowledge. While this ability comes naturally to biological brains, it poses a significant challenge for artificial intelligence (AI). Here, we suggest that environmental enrichment (EE) can be used as a biological model for studying forward transfer, inspiring human-like AI development. EE refers to animal studies that enhance cognitive, social, motor, and sensory stimulation and is a model for what, in humans, is referred to as 'cognitive reserve'. Enriched animals show significant improvement in learning speed and performance on new tasks, typically exhibiting forward transfer. We explore anatomical, molecular, and neuronal changes post-EE and discuss how artificial neural networks (ANNs) can be used to predict neural computation changes after enriched experiences. Finally, we provide a synergistic way of combining neuroscience and AI research that paves the path toward developing AI capable of rapid and efficient new task learning.
Collapse
Affiliation(s)
- Rajat Saxena
- Department of Neurobiology and Behavior, University of California, Irvine, Irvine, CA 92697, USA
| | - Bruce L McNaughton
- Department of Neurobiology and Behavior, University of California, Irvine, Irvine, CA 92697, USA
- Canadian Centre for Behavioural Neuroscience, University of Lethbridge, Lethbridge, AB, T1K 3M4 Canada
| |
Collapse
|
11
|
Bonfanti L, La Rosa C, Ghibaudi M, Sherwood CC. Adult neurogenesis and "immature" neurons in mammals: an evolutionary trade-off in plasticity? Brain Struct Funct 2024; 229:1775-1793. [PMID: 37833544 PMCID: PMC11485216 DOI: 10.1007/s00429-023-02717-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Accepted: 09/26/2023] [Indexed: 10/15/2023]
Abstract
Neuronal plasticity can vary remarkably in its form and degree across animal species. Adult neurogenesis, namely the capacity to produce new neurons from neural stem cells through adulthood, appears widespread in non-mammalian vertebrates, whereas it is reduced in mammals. A growing body of comparative studies also report variation in the occurrence and activity of neural stem cell niches between mammals, with a general trend of reduction from small-brained to large-brained species. Conversely, recent studies have shown that large-brained mammals host large amounts of neurons expressing typical markers of neurogenesis in the absence of cell division. In layer II of the cerebral cortex, populations of prenatally generated, non-dividing neurons continue to express molecules indicative of immaturity throughout life (cortical immature neurons; cINs). After remaining in a dormant state for a very long time, these cINs retain the potential of differentiating into mature neurons that integrate within the preexisting neural circuits. They are restricted to the paleocortex in small-brained rodents, while extending into the widely expanded neocortex of highly gyrencephalic, large-brained species. The current hypothesis is that these populations of non-newly generated "immature" neurons might represent a reservoir of developmentally plastic cells for mammalian species that are characterized by reduced stem cell-driven adult neurogenesis. This indicates that there may be a trade-off between various forms of plasticity that coexist during brain evolution. This balance may be necessary to maintain a "reservoir of plasticity" in brain regions that have distinct roles in species-specific socioecological adaptations, such as the neocortex and olfactory structures.
Collapse
Affiliation(s)
- Luca Bonfanti
- Neuroscience Institute Cavalieri Ottolenghi, Orbassano, Italy.
- Department of Veterinary Sciences, University of Turin, Largo Braccini 2, 10095, Turin, Grugliasco, Italy.
| | - Chiara La Rosa
- Neuroscience Institute Cavalieri Ottolenghi, Orbassano, Italy
| | - Marco Ghibaudi
- Neuroscience Institute Cavalieri Ottolenghi, Orbassano, Italy
- Department of Veterinary Sciences, University of Turin, Largo Braccini 2, 10095, Turin, Grugliasco, Italy
| | - Chet C Sherwood
- Department of Anthropology and Center for the Advanced Study of Human Paleobiology, The George Washington University, Washington, DC, USA.
| |
Collapse
|
12
|
Chen YN, Kostka JK. Beyond anosmia: olfactory dysfunction as a common denominator in neurodegenerative and neurodevelopmental disorders. Front Neurosci 2024; 18:1502779. [PMID: 39539496 PMCID: PMC11557544 DOI: 10.3389/fnins.2024.1502779] [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: 09/27/2024] [Accepted: 10/21/2024] [Indexed: 11/16/2024] Open
Abstract
Olfactory dysfunction has emerged as a hallmark feature shared among several neurological conditions, including both neurodevelopmental and neurodegenerative disorders. While diseases of both categories have been extensively studied for decades, their association with olfaction has only recently gained attention. Olfactory deficits often manifest already during prodromal stages of these diseases, yet it remains unclear whether common pathophysiological changes along olfactory pathways cause such impairments. Here we probe into the intricate relationship between olfactory dysfunction and neurodegenerative and neurodevelopmental disorders, shedding light on their commonalities and underlying mechanisms. We begin by providing a brief overview of the olfactory circuit and its connections to higher-associated brain areas. Additionally, we discuss olfactory deficits in these disorders, focusing on potential common mechanisms that may contribute to olfactory dysfunction across both types of disorders. We further debate whether olfactory deficits contribute to the disease propagation or are simply an epiphenomenon. We conclude by emphasizing the significance of olfactory function as a potential pre-clinical diagnostic tool to identify individuals with neurological disorders that offers the opportunity for preventive intervention before other symptoms manifest.
Collapse
Affiliation(s)
- Yu-Nan Chen
- Institute of Developmental Neuroscience, Center of Molecular Neurobiology, Hamburg Center of Neuroscience, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Johanna Katharina Kostka
- Institute of Developmental Neuroscience, Center of Molecular Neurobiology, Hamburg Center of Neuroscience, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
13
|
Salimi M, Nazari M, Shahsavar P, Dehghan S, Javan M, Mirnajafi‐Zadeh J, Raoufy MR. Olfactory bulb stimulation mitigates Alzheimer's-like disease progression. CNS Neurosci Ther 2024; 30:e70056. [PMID: 39404073 PMCID: PMC11474698 DOI: 10.1111/cns.70056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2024] [Revised: 09/03/2024] [Accepted: 09/06/2024] [Indexed: 10/19/2024] Open
Abstract
BACKGROUND Deep brain stimulation (DBS) has demonstrated potential in mitigating Alzheimer's disease (AD). However, the invasive nature of DBS presents challenges for its application. The olfactory bulb (OB), showing early AD-related changes and extensive connections with memory regions, offers an attractive entry point for intervention, potentially restoring normal activity in deteriorating memory circuits. AIMS Our study examined the impact of electrically stimulating the OB on working memory as well as pathological and electrophysiological alterations in the OB, medial prefrontal cortex, hippocampus, and entorhinal cortex in amyloid beta (Aβ) AD model rats. METHODS Male Wistar rats underwent surgery for electrode implantation in brain regions, inducing Alzheimer's-like disease. Bilateral olfactory bulb (OB) electrical stimulation was performed for 1 hour daily to the OB of stimulation group animals for 18 consecutive days, followed by the evaluations of histological, behavioral, and local field potential signal processing. RESULTS OB stimulation counteracted Aβ plaque accumulation and prevented AD-induced working memory impairments. Furthermore, it prompted an increase in power across diverse frequency bands and enhanced functional connectivity, particularly in the gamma band, within the investigated regions during a working memory task. CONCLUSION This preclinical investigation highlights the potential of olfactory pathway-based brain stimulation to modulate the activity of deep-seated memory networks for AD treatment. Importantly, the accessibility of this pathway via the nasal cavity lays the groundwork for the development of minimally invasive approaches targeting the olfactory pathway for brain modulation.
Collapse
Affiliation(s)
- Morteza Salimi
- Department of Physiology, Faculty of Medical SciencesTarbiat Modares UniversityTehranIran
| | - Milad Nazari
- Department of Molecular Biology and GeneticsAarhus UniversityAarhusDenmark
- The Danish Research Institute of Translational Neuroscience, DANDRITEAarhus UniversityAarhusDenmark
| | - Payam Shahsavar
- Department of Physiology, Faculty of Medical SciencesTarbiat Modares UniversityTehranIran
| | - Samaneh Dehghan
- Stem Cell and Regenerative Medicine Research CenterIran University of Medical SciencesTehranIran
- The Five Senses Institute, Eye Research CenterRassoul Akram Hospital, Iran University of Medical SciencesTehranIran
| | - Mohammad Javan
- Department of Physiology, Faculty of Medical SciencesTarbiat Modares UniversityTehranIran
- Institute for Brain Sciences and CognitionTarbiat Modares UniversityTehranIran
| | - Javad Mirnajafi‐Zadeh
- Department of Physiology, Faculty of Medical SciencesTarbiat Modares UniversityTehranIran
- Institute for Brain Sciences and CognitionTarbiat Modares UniversityTehranIran
| | - Mohammad Reza Raoufy
- Department of Physiology, Faculty of Medical SciencesTarbiat Modares UniversityTehranIran
- Institute for Brain Sciences and CognitionTarbiat Modares UniversityTehranIran
| |
Collapse
|
14
|
Wang K, Liu XY, Liu SF, Wang XX, Wei YH, Zhu JR, Liu J, Xu XQ, Wen L. Rbm24/Notch1 signaling regulates adult neurogenesis in the subventricular zone and mediates Parkinson-associated olfactory dysfunction. Theranostics 2024; 14:4499-4518. [PMID: 39113792 PMCID: PMC11303084 DOI: 10.7150/thno.96045] [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: 03/07/2024] [Accepted: 07/11/2024] [Indexed: 08/10/2024] Open
Abstract
Rationale: Adult neurogenesis in the subventricular zone (SVZ) is essential for maintaining neural homeostasis, and its dysregulation contributes to anosmia and delayed tissue healing in neurological disorders, such as Parkinson's disease (PD). Despite intricate regulatory networks identified in SVZ neurogenesis, the molecular mechanisms dynamically maintaining neural stem/progenitor cells (NSPCs) in response to physiological and pathological stimuli remain incompletely elucidated. Methods: We generated an RNA binding motif protein 24 (Rbm24) knockout model to investigate its impact on adult neurogenesis in the SVZ, employing immunofluorescence, immunoblot, electrophysiology, RNA-sequencing, and in vitro experiments. Further investigations utilized a PD mouse model, along with genetic and pharmacological manipulations, to elucidate Rbm24 involvement in PD pathology. Results: Rbm24, a multifaceted post-transcriptional regulator of cellular homeostasis, exhibited broad expression in the SVZ from development to aging. Deletion of Rbm24 significantly impaired NSPC proliferation in the adult SVZ, ultimately resulting in collapsed neurogenesis in the olfactory bulb. Notably, Rbm24 played a specific role in maintaining Notch1 mRNA stability in adult NSPCs. The Rbm24/Notch1 signaling axis was significantly downregulated in the SVZ of PD mice. Remarkably, overexpression of Rbm24 rescued disruption of adult neurogenesis and olfactory dysfunction in PD mice, and these effects were hindered by DAPT, a potent inhibitor of Notch1. Conclusions: Our findings highlight the critical role of the Rbm24/Notch1 signaling axis in regulating adult SVZ neurogenesis under physiological and pathological circumstances. This provides valuable insights into the dynamic regulation of NSPC homeostasis and offers a potential targeted intervention for PD and related neurological disorders.
Collapse
Affiliation(s)
- Ke Wang
- Institute of Stem Cell and Regenerative Medicine, Women and Children's Hospital of Xiamen University, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, State Key Laboratory of Cellular Stress Biology, School of Medicine, Xiamen University, Xiamen, Fujian 361000, China
- Center for Brain Sciences, Department of Traditional Chinese Medicine, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian 361000, China
| | - Xing-Yang Liu
- Institute of Stem Cell and Regenerative Medicine, Women and Children's Hospital of Xiamen University, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, State Key Laboratory of Cellular Stress Biology, School of Medicine, Xiamen University, Xiamen, Fujian 361000, China
- Center for Brain Sciences, Department of Traditional Chinese Medicine, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian 361000, China
| | - Sui-Feng Liu
- Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian 361000, China
| | - Xiao-Xia Wang
- Institute of Stem Cell and Regenerative Medicine, Women and Children's Hospital of Xiamen University, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, State Key Laboratory of Cellular Stress Biology, School of Medicine, Xiamen University, Xiamen, Fujian 361000, China
| | - Yi-Hua Wei
- Institute of Stem Cell and Regenerative Medicine, Women and Children's Hospital of Xiamen University, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, State Key Laboratory of Cellular Stress Biology, School of Medicine, Xiamen University, Xiamen, Fujian 361000, China
- Center for Brain Sciences, Department of Traditional Chinese Medicine, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian 361000, China
| | - Jun-Rong Zhu
- Institute of Stem Cell and Regenerative Medicine, Women and Children's Hospital of Xiamen University, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, State Key Laboratory of Cellular Stress Biology, School of Medicine, Xiamen University, Xiamen, Fujian 361000, China
- Center for Brain Sciences, Department of Traditional Chinese Medicine, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian 361000, China
| | - Jing Liu
- Institute of Stem Cell and Regenerative Medicine, Women and Children's Hospital of Xiamen University, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, State Key Laboratory of Cellular Stress Biology, School of Medicine, Xiamen University, Xiamen, Fujian 361000, China
| | - Xiu Qin Xu
- Institute of Stem Cell and Regenerative Medicine, Women and Children's Hospital of Xiamen University, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, State Key Laboratory of Cellular Stress Biology, School of Medicine, Xiamen University, Xiamen, Fujian 361000, China
| | - Lei Wen
- Institute of Stem Cell and Regenerative Medicine, Women and Children's Hospital of Xiamen University, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, State Key Laboratory of Cellular Stress Biology, School of Medicine, Xiamen University, Xiamen, Fujian 361000, China
- Center for Brain Sciences, Department of Traditional Chinese Medicine, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian 361000, China
| |
Collapse
|
15
|
Chen Z, Padmanabhan K. Adult-neurogenesis allows for representational stability and flexibility in early olfactory system. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.02.601573. [PMID: 39005290 PMCID: PMC11244980 DOI: 10.1101/2024.07.02.601573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/16/2024]
Abstract
In the early olfactory system, adult-neurogenesis, a process of neuronal replacement results in the continuous reorganization of synaptic connections and network architecture throughout the animal's life. This poses a critical challenge: How does the olfactory system maintain stable representations of odors and therefore allow for stable sensory perceptions amidst this ongoing circuit instability? Utilizing a detailed spiking network model of early olfactory circuits, we uncovered dual roles for adult-neurogenesis: one that both supports representational stability to faithfully encode odor information and also one that facilitates plasticity to allow for learning and adaptation. In the main olfactory bulb, adult-neurogenesis affects neural codes in individual mitral and tufted cells but preserves odor representations at the neuronal population level. By contrast, in the olfactory piriform cortex, both individual cell responses and overall population dynamics undergo progressive changes due to adult-neurogenesis. This leads to representational drift, a gradual alteration in sensory perception. Both processes are dynamic and depend on experience such that repeated exposure to specific odors reduces the drift due to adult-neurogenesis; thus, when the odor environment is stable over the course of adult-neurogenesis, it is neurogenesis that actually allows the representations to remain stable in piriform cortex; when those olfactory environments change, adult-neurogenesis allows the cortical representations to track environmental change. Whereas perceptual stability and plasticity due to learning are often thought of as two distinct, often contradictory processing in neuronal coding, we find that adult-neurogenesis serves as a shared mechanism for both. In this regard, the quixotic presence of adult-neurogenesis in the mammalian olfactory bulb that has been the focus of considerable debate in chemosensory neuroscience may be the mechanistic underpinning behind an array of complex computations.
Collapse
Affiliation(s)
- Zhen Chen
- Department of Brain and Cognitive Sciences, University of Rochester, Rochester, NY14627
| | - Krishnan Padmanabhan
- Department of Neuroscience, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642
| |
Collapse
|
16
|
Hawkins SJ, Gärtner Y, Offner T, Weiss L, Maiello G, Hassenklöver T, Manzini I. The olfactory network of larval Xenopus laevis regenerates accurately after olfactory nerve transection. Eur J Neurosci 2024; 60:3719-3741. [PMID: 38758670 DOI: 10.1111/ejn.16375] [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: 12/07/2023] [Revised: 04/10/2024] [Accepted: 04/14/2024] [Indexed: 05/19/2024]
Abstract
Across vertebrate species, the olfactory epithelium (OE) exhibits the uncommon feature of lifelong neuronal turnover. Epithelial stem cells give rise to new neurons that can adequately replace dying olfactory receptor neurons (ORNs) during developmental and adult phases and after lesions. To relay olfactory information from the environment to the brain, the axons of the renewed ORNs must reconnect with the olfactory bulb (OB). In Xenopus laevis larvae, we have previously shown that this process occurs between 3 and 7 weeks after olfactory nerve (ON) transection. In the present study, we show that after 7 weeks of recovery from ON transection, two functionally and spatially distinct glomerular clusters are reformed in the OB, akin to those found in non-transected larvae. We also show that the same odourant response tuning profiles observed in the OB of non-transected larvae are again present after 7 weeks of recovery. Next, we show that characteristic odour-guided behaviour disappears after ON transection but recovers after 7-9 weeks of recovery. Together, our findings demonstrate that the olfactory system of larval X. laevis regenerates with high accuracy after ON transection, leading to the recovery of odour-guided behaviour.
Collapse
Affiliation(s)
- Sara J Hawkins
- Institute of Animal Physiology, Department of Animal Physiology and Molecular Biomedicine, Justus Liebig University Gießen, Gießen, Germany
- School of Biological Sciences, Faculty of Environmental and Life Sciences, University of Southampton, Southampton, UK
| | - Yvonne Gärtner
- Institute of Animal Physiology, Department of Animal Physiology and Molecular Biomedicine, Justus Liebig University Gießen, Gießen, Germany
- School of Biological Sciences, Faculty of Environmental and Life Sciences, University of Southampton, Southampton, UK
- Department of Neurology, Focus Program Translational Neuroscience (FTN) and Immunotherapy (FZI), Rhine-Main Neuroscience Network (rmn2), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Thomas Offner
- Institute of Animal Physiology, Department of Animal Physiology and Molecular Biomedicine, Justus Liebig University Gießen, Gießen, Germany
| | - Lukas Weiss
- Institute of Animal Physiology, Department of Animal Physiology and Molecular Biomedicine, Justus Liebig University Gießen, Gießen, Germany
| | - Guido Maiello
- Department of Experimental Psychology, Justus Liebig University Gießen, Gießen, Germany
- School of Psychology, Faculty of Environmental and Life Sciences, University of Southampton, Southampton, UK
| | - Thomas Hassenklöver
- Institute of Animal Physiology, Department of Animal Physiology and Molecular Biomedicine, Justus Liebig University Gießen, Gießen, Germany
| | - Ivan Manzini
- Institute of Animal Physiology, Department of Animal Physiology and Molecular Biomedicine, Justus Liebig University Gießen, Gießen, Germany
| |
Collapse
|
17
|
Fulton KA, Zimmerman D, Samuel A, Vogt K, Datta SR. Common principles for odour coding across vertebrates and invertebrates. Nat Rev Neurosci 2024; 25:453-472. [PMID: 38806946 DOI: 10.1038/s41583-024-00822-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/30/2024] [Indexed: 05/30/2024]
Abstract
The olfactory system is an ideal and tractable system for exploring how the brain transforms sensory inputs into behaviour. The basic tasks of any olfactory system include odour detection, discrimination and categorization. The challenge for the olfactory system is to transform the high-dimensional space of olfactory stimuli into the much smaller space of perceived objects and valence that endows odours with meaning. Our current understanding of how neural circuits address this challenge has come primarily from observations of the mechanisms of the brain for processing other sensory modalities, such as vision and hearing, in which optimized deep hierarchical circuits are used to extract sensory features that vary along continuous physical dimensions. The olfactory system, by contrast, contends with an ill-defined, high-dimensional stimulus space and discrete stimuli using a circuit architecture that is shallow and parallelized. Here, we present recent observations in vertebrate and invertebrate systems that relate the statistical structure and state-dependent modulation of olfactory codes to mechanisms of perception and odour-guided behaviour.
Collapse
Affiliation(s)
- Kara A Fulton
- Department of Neuroscience, Harvard Medical School, Boston, MA, USA
| | - David Zimmerman
- Department of Physics, Harvard University, Cambridge, MA, USA
| | - Aravi Samuel
- Department of Physics, Harvard University, Cambridge, MA, USA
| | - Katrin Vogt
- Department of Physics, Harvard University, Cambridge, MA, USA.
- Department of Biology, University of Konstanz, Konstanz, Germany.
- Centre for the Advanced Study of Collective Behaviour, University of Konstanz, Konstanz, Germany.
| | | |
Collapse
|
18
|
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
|
19
|
Rufenacht KE, Asson AJ, Hossain K, Santoro SW. The influence of olfactory experience on the birthrates of olfactory sensory neurons with specific odorant receptor identities. Genesis 2024; 62:e23611. [PMID: 38888221 PMCID: PMC11189617 DOI: 10.1002/dvg.23611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 05/31/2024] [Accepted: 06/03/2024] [Indexed: 06/20/2024]
Abstract
Olfactory sensory neurons (OSNs) are one of a few neuron types that are generated continuously throughout life in mammals. The persistence of olfactory sensory neurogenesis beyond early development has long been thought to function simply to replace neurons that are lost or damaged through exposure to environmental insults. The possibility that olfactory sensory neurogenesis may also serve an adaptive function has received relatively little consideration, largely due to the assumption that the generation of new OSNs is stochastic with respect to OSN subtype, as defined by the single odorant receptor gene that each neural precursor stochastically chooses for expression out of hundreds of possibilities. Accordingly, the relative birthrates of different OSN subtypes are predicted to be constant and impervious to olfactory experience. This assumption has been called into question, however, by evidence that the birthrates of specific OSN subtypes can be selectively altered by manipulating olfactory experience through olfactory deprivation, enrichment, and conditioning paradigms. Moreover, studies of recovery of the OSN population following injury provide further evidence that olfactory sensory neurogenesis may not be strictly stochastic with respect to subtype. Here we review this evidence and consider mechanistic and functional implications of the prospect that specific olfactory experiences can regulate olfactory sensory neurogenesis rates in a subtype-selective manner.
Collapse
Affiliation(s)
- Karlin E Rufenacht
- Department of Pediatrics, Section of Developmental Biology, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Alexa J Asson
- Department of Pediatrics, Section of Developmental Biology, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Kawsar Hossain
- Department of Pediatrics, Section of Developmental Biology, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Stephen W Santoro
- Department of Pediatrics, Section of Developmental Biology, University of Colorado School of Medicine, Aurora, Colorado, USA
| |
Collapse
|
20
|
Vergil Andrews JF, Selvaraj DB, Bhavani Radhakrishnan A, Kandasamy M. Low-dose aspirin increases olfactory sensitivity in association with enhanced neurogenesis and reduced activity of AChE in the experimental aging mice. MEDICINE IN DRUG DISCOVERY 2024; 22:100191. [DOI: 10.1016/j.medidd.2024.100191] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/11/2024] Open
|
21
|
Li J, Wang Y, Raina MA, Xu C, Su L, Guo Q, Ma Q, Wang J, Xu D. scBSP: A fast and accurate tool for identifying spatially variable genes from spatial transcriptomic data. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.06.592851. [PMID: 38765956 PMCID: PMC11100755 DOI: 10.1101/2024.05.06.592851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
Spatially resolved transcriptomics have enabled the inference of gene expression patterns within two and three-dimensional space, while introducing computational challenges due to growing spatial resolutions and sparse expressions. Here, we introduce scBSP, an open-source, versatile, and user-friendly package designed for identifying spatially variable genes in large-scale spatial transcriptomics. scBSP implements sparse matrix operation to significantly increase the computational efficiency in both computational time and memory usage, processing the high-definition spatial transcriptomics data for 19,950 genes on 181,367 spots within 10 seconds. Applied to diverse sequencing data and simulations, scBSP efficiently identifies spatially variable genes, demonstrating fast computational speed and consistency across various sequencing techniques and spatial resolutions for both two and three-dimensional data with up to millions of cells. On a sample with hundreds of thousands of sports, scBSP identifies SVGs accurately in seconds to on a typical desktop computer.
Collapse
|
22
|
Goodkey K, Wischmeijer A, Perrin L, Watson AES, Qureshi L, Cordelli DM, Toni F, Gnazzo M, Benedicenti F, Elmaleh-Bergès M, Low KJ, Voronova A. Olfactory bulb anomalies in KBG syndrome mouse model and patients. BMC Med 2024; 22:158. [PMID: 38616269 PMCID: PMC11017579 DOI: 10.1186/s12916-024-03363-6] [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: 08/02/2023] [Accepted: 03/18/2024] [Indexed: 04/16/2024] Open
Abstract
ANKRD11 (ankyrin repeat domain 11) is a chromatin regulator and the only gene associated with KBG syndrome, a rare neurodevelopmental disorder. We have previously shown that Ankrd11 regulates murine embryonic cortical neurogenesis. Here, we show a novel olfactory bulb phenotype in a KBG syndrome mouse model and two diagnosed patients. Conditional knockout of Ankrd11 in murine embryonic neural stem cells leads to aberrant postnatal olfactory bulb development and reduced size due to reduction of the olfactory bulb granule cell layer. We further show that the rostral migratory stream has incomplete migration of neuroblasts, reduced cell proliferation as well as aberrant differentiation of neurons. This leads to reduced neuroblasts and neurons in the olfactory bulb granule cell layer. In vitro, Ankrd11-deficient neural stem cells from the postnatal subventricular zone display reduced migration, proliferation, and neurogenesis. Finally, we describe two clinically and molecularly confirmed KBG syndrome patients with anosmia and olfactory bulb and groove hypo-dysgenesis/agenesis. Our report provides evidence that Ankrd11 is a novel regulator of olfactory bulb development and neuroblast migration. Moreover, our study highlights a novel clinical sign of KBG syndrome linked to ANKRD11 perturbations in mice and humans.
Collapse
Affiliation(s)
- Kara Goodkey
- Department of Medical Genetics, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, AB, T6G 2H7, Canada
- Women and Children's Health Research Institute, University of Alberta, 5-083 Edmonton Clinic Health Academy, Edmonton, AB, T6G 1C9, Canada
| | - Anita Wischmeijer
- Clinical Genetics Service and Coordination Center for Rare Diseases, Department of Pediatrics, Regional Hospital of Bolzano, Bolzano, Italy
| | | | - Adrianne E S Watson
- Department of Medical Genetics, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, AB, T6G 2H7, Canada
- Women and Children's Health Research Institute, University of Alberta, 5-083 Edmonton Clinic Health Academy, Edmonton, AB, T6G 1C9, Canada
| | - Leenah Qureshi
- Department of Medical Genetics, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, AB, T6G 2H7, Canada
| | - Duccio Maria Cordelli
- IRCCS Istituto Delle Scienze Neurologiche Di Bologna, UOC Neuropsichiatria Dell'età Pediatrica, Bologna, Italy
| | - Francesco Toni
- IRCCS Istituto Delle Scienze Neurologiche Di Bologna, Programma Di Neuroradiologia Con Tecniche Ad Elevata Complessità (PNTEC), Bologna, Italy
| | - Maria Gnazzo
- Laboratory of Medical Genetics, Translational Cytogenomics Research Unit, Bambino Gesù Children's Hospital, IRCCS, 00165, Rome, Italy
| | - Francesco Benedicenti
- Clinical Genetics Service and Coordination Center for Rare Diseases, Department of Pediatrics, Regional Hospital of Bolzano, Bolzano, Italy
| | | | - Karen J Low
- Department of Academic Child Health, Bristol Medical School, Population Health Sciences, University of Bristol, Bristol, UK
- Clinical Genetics Service, St. Michaels Hospital, Bristol, UK
| | - Anastassia Voronova
- Department of Medical Genetics, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, AB, T6G 2H7, Canada.
- Women and Children's Health Research Institute, University of Alberta, 5-083 Edmonton Clinic Health Academy, Edmonton, AB, T6G 1C9, Canada.
- Department of Cell Biology, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, AB, T6G 2H7, Canada.
- Faculty of Medicine & Dentistry, Neuroscience and Mental Health Institute, Edmonton, AB, T6G 2E1, Canada.
| |
Collapse
|
23
|
Senovilla-Ganzo R, García-Moreno F. The Phylotypic Brain of Vertebrates, from Neural Tube Closure to Brain Diversification. BRAIN, BEHAVIOR AND EVOLUTION 2024; 99:45-68. [PMID: 38342091 DOI: 10.1159/000537748] [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: 11/30/2023] [Accepted: 02/04/2024] [Indexed: 02/13/2024]
Abstract
BACKGROUND The phylotypic or intermediate stages are thought to be the most evolutionary conserved stages throughout embryonic development. The contrast with divergent early and later stages derived from the concept of the evo-devo hourglass model. Nonetheless, this developmental constraint has been studied as a whole embryo process, not at organ level. In this review, we explore brain development to assess the existence of an equivalent brain developmental hourglass. In the specific case of vertebrates, we propose to split the brain developmental stages into: (1) Early: Neurulation, when the neural tube arises after gastrulation. (2) Intermediate: Brain patterning and segmentation, when the neuromere identities are established. (3) Late: Neurogenesis and maturation, the stages when the neurons acquire their functionality. Moreover, we extend this analysis to other chordates brain development to unravel the evolutionary origin of this evo-devo constraint. SUMMARY Based on the existing literature, we hypothesise that a major conservation of the phylotypic brain might be due to the pleiotropy of the inductive regulatory networks, which are predominantly expressed at this stage. In turn, earlier stages such as neurulation are rather mechanical processes, whose regulatory networks seem to adapt to environment or maternal geometries. The later stages are also controlled by inductive regulatory networks, but their effector genes are mostly tissue-specific and functional, allowing diverse developmental programs to generate current brain diversity. Nonetheless, all stages of the hourglass are highly interconnected: divergent neurulation must have a vertebrate shared end product to reproduce the vertebrate phylotypic brain, and the boundaries and transcription factor code established during the highly conserved patterning will set the bauplan for the specialised and diversified adult brain. KEY MESSAGES The vertebrate brain is conserved at phylotypic stages, but the highly conserved mechanisms that occur during these brain mid-development stages (Inducing Regulatory Networks) are also present during other stages. Oppositely, other processes as cell interactions and functional neuronal genes are more diverse and majoritarian in early and late stages of development, respectively. These phenomena create an hourglass of transcriptomic diversity during embryonic development and evolution, with a really conserved bottleneck that set the bauplan for the adult brain around the phylotypic stage.
Collapse
Affiliation(s)
- Rodrigo Senovilla-Ganzo
- Achucarro Basque Center for Neuroscience, Scientific Park of the University of the Basque Country (UPV/EHU), Leioa, Spain
- Department of Neuroscience, Faculty of Medicine and Odontology, UPV/EHU, Leioa, Spain
| | - Fernando García-Moreno
- Achucarro Basque Center for Neuroscience, Scientific Park of the University of the Basque Country (UPV/EHU), Leioa, Spain
- Department of Neuroscience, Faculty of Medicine and Odontology, UPV/EHU, Leioa, Spain
- IKERBASQUE Foundation, Bilbao, Spain
| |
Collapse
|
24
|
Kelly CJ, Lindsay SL, Smith RS, Keh S, Cunningham KT, Thümmler K, Maizels RM, Campbell JDM, Barnett SC. Development of Good Manufacturing Practice-Compatible Isolation and Culture Methods for Human Olfactory Mucosa-Derived Mesenchymal Stromal Cells. Int J Mol Sci 2024; 25:743. [PMID: 38255817 PMCID: PMC10815924 DOI: 10.3390/ijms25020743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 12/21/2023] [Accepted: 12/27/2023] [Indexed: 01/24/2024] Open
Abstract
Demyelination in the central nervous system (CNS) resulting from injury or disease can cause loss of nerve function and paralysis. Cell therapies intended to promote remyelination of axons are a promising avenue of treatment, with mesenchymal stromal cells (MSCs) a prominent candidate. We have previously demonstrated that MSCs derived from human olfactory mucosa (hOM-MSCs) promote myelination to a greater extent than bone marrow-derived MSCs (hBM-MSCs). However, hOM-MSCs were developed using methods and materials that were not good manufacturing practice (GMP)-compliant. Before considering these cells for clinical use, it is necessary to develop a method for their isolation and expansion that is readily adaptable to a GMP-compliant environment. We demonstrate here that hOM-MSCs can be derived without enzymatic tissue digestion or cell sorting and without culture antibiotics. They grow readily in GMP-compliant media and express typical MSC surface markers. They robustly produce CXCL12 (a key secretory factor in promoting myelination) and are pro-myelinating in in vitro rodent CNS cultures. GMP-compliant hOM-MSCs are comparable in this respect to those grown in non-GMP conditions. However, when assessed in an in vivo model of demyelinating disease (experimental autoimmune encephalitis, EAE), they do not significantly improve disease scores compared with controls, indicating further pre-clinical evaluation is necessary before their advancement to clinical trials.
Collapse
Affiliation(s)
- Christopher J. Kelly
- School of Infection and Immunity, 120 University Place, Glasgow G12 8TA, UK; (C.J.K.); (S.L.L.); (R.M.M.)
| | - Susan L. Lindsay
- School of Infection and Immunity, 120 University Place, Glasgow G12 8TA, UK; (C.J.K.); (S.L.L.); (R.M.M.)
| | - Rebecca Sherrard Smith
- School of Infection and Immunity, 120 University Place, Glasgow G12 8TA, UK; (C.J.K.); (S.L.L.); (R.M.M.)
| | - Siew Keh
- New Victoria Hospital, 55 Grange Road, Glasgow G42 9LF, UK
| | - Kyle T. Cunningham
- School of Infection and Immunity, 120 University Place, Glasgow G12 8TA, UK; (C.J.K.); (S.L.L.); (R.M.M.)
| | - Katja Thümmler
- School of Infection and Immunity, 120 University Place, Glasgow G12 8TA, UK; (C.J.K.); (S.L.L.); (R.M.M.)
| | - Rick M. Maizels
- School of Infection and Immunity, 120 University Place, Glasgow G12 8TA, UK; (C.J.K.); (S.L.L.); (R.M.M.)
| | - John D. M. Campbell
- School of Infection and Immunity, 120 University Place, Glasgow G12 8TA, UK; (C.J.K.); (S.L.L.); (R.M.M.)
- Tissues Cells and Advanced Therapeutics, SNBTS, Jack Copland Centre, Edinburgh EH14 4BE, UK
| | - Susan C. Barnett
- School of Infection and Immunity, 120 University Place, Glasgow G12 8TA, UK; (C.J.K.); (S.L.L.); (R.M.M.)
| |
Collapse
|
25
|
Mattova S, Simko P, Urbanska N, Kiskova T. Bioactive Compounds and Their Influence on Postnatal Neurogenesis. Int J Mol Sci 2023; 24:16614. [PMID: 38068936 PMCID: PMC10706651 DOI: 10.3390/ijms242316614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 11/10/2023] [Accepted: 11/16/2023] [Indexed: 12/18/2023] Open
Abstract
Since postnatal neurogenesis was revealed to have significant implications for cognition and neurological health, researchers have been increasingly exploring the impact of natural compounds on this process, aiming to uncover strategies for enhancing brain plasticity. This review provides an overview of postnatal neurogenesis, neurogenic zones, and disorders characterized by suppressed neurogenesis and neurogenesis-stimulating bioactive compounds. Examining recent studies, this review underscores the multifaceted effects of natural compounds on postnatal neurogenesis. In essence, understanding the interplay between postnatal neurogenesis and natural compounds could bring novel insights into brain health interventions. Exploiting the therapeutic abilities of these compounds may unlock innovative approaches to enhance cognitive function, mitigate neurodegenerative diseases, and promote overall brain well-being.
Collapse
Affiliation(s)
| | | | | | - Terezia Kiskova
- Department of Animal Physiology, Institute of Biology and Ecology, Faculty of Science, Pavol Jozef Safarik University in Kosice, 041 54 Kosice, Slovakia; (S.M.); (P.S.); (N.U.)
| |
Collapse
|
26
|
Chandwani MN, Kamte YS, Singh VR, Hemerson ME, Michaels AC, Leak RK, O'Donnell LA. The anti-viral immune response of the adult host robustly modulates neural stem cell activity in spatial, temporal, and sex-specific manners. Brain Behav Immun 2023; 114:61-77. [PMID: 37516388 DOI: 10.1016/j.bbi.2023.07.008] [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: 02/23/2023] [Revised: 06/20/2023] [Accepted: 07/14/2023] [Indexed: 07/31/2023] Open
Abstract
Viruses induce a wide range of neurological sequelae through the dysfunction and death of infected cells and persistent inflammation in the brain. Neural stem cells (NSCs) are often disturbed during viral infections. Although some viruses directly infect and kill NSCs, the antiviral immune response may also indirectly affect NSCs. To better understand how NSCs are influenced by a productive immune response, where the virus is successfully resolved and the host survives, we used the CD46+ mouse model of neuron-restricted measles virus (MeV) infection. As NSCs are spared from direct infection in this model, they serve as bystanders to the antiviral immune response initiated by selective infection of mature neurons. MeV-infected mice showed distinct regional and temporal changes in NSCs in the primary neurogenic niches of the brain, the hippocampus and subventricular zone (SVZ). Hippocampal NSCs increased throughout the infection (7 and 60 days post-infection; dpi), while mature neurons transiently declined at 7 dpi and then rebounded to basal levels by 60 dpi. In the SVZ, NSC numbers were unchanged, but mature neurons declined even after the infection was controlled at 60 dpi. Further analyses demonstrated sex, temporal, and region-specific changes in NSC proliferation and neurogenesis throughout the infection. A relatively long-term increase in NSC proliferation and neurogenesis was observed in the hippocampus; however, neurogenesis was reduced in the SVZ. This decline in SVZ neurogenesis was associated with increased immature neurons in the olfactory bulb in female, but not male mice, suggesting potential migration of newly-made neurons out of the female SVZ. These sex differences in SVZ neurogenesis were accompanied by higher infiltration of B cells and greater expression of interferon-gamma and interleukin-6 in female mice. Learning, memory, and olfaction tests revealed no overt behavioral changes after the acute infection subsided. These results indicate that antiviral immunity modulates NSC activity in adult mice without inducing gross behavioral deficits among those tested, suggestive of mechanisms to restore neurons and maintain adaptive behavior, but also revealing the potential for robust NSC disruption in subclinical infections.
Collapse
Affiliation(s)
- Manisha N Chandwani
- Duquesne University School of Pharmacy, Graduate School of Pharmaceutical Sciences, Pittsburgh, PA, USA
| | - Yashika S Kamte
- Duquesne University School of Pharmacy, Graduate School of Pharmaceutical Sciences, Pittsburgh, PA, USA
| | - Vivek R Singh
- Duquesne University School of Pharmacy, Graduate School of Pharmaceutical Sciences, Pittsburgh, PA, USA
| | - Marlo E Hemerson
- Duquesne University School of Pharmacy, Graduate School of Pharmaceutical Sciences, Pittsburgh, PA, USA
| | - Alexa C Michaels
- Duquesne University School of Pharmacy, Graduate School of Pharmaceutical Sciences, Pittsburgh, PA, USA
| | - Rehana K Leak
- Duquesne University School of Pharmacy, Graduate School of Pharmaceutical Sciences, Pittsburgh, PA, USA
| | - Lauren A O'Donnell
- Duquesne University School of Pharmacy, Graduate School of Pharmaceutical Sciences, Pittsburgh, PA, USA.
| |
Collapse
|
27
|
Chang SY, Lee MY. Photobiomodulation of Neurogenesis through the Enhancement of Stem Cell and Neural Progenitor Differentiation in the Central and Peripheral Nervous Systems. Int J Mol Sci 2023; 24:15427. [PMID: 37895108 PMCID: PMC10607539 DOI: 10.3390/ijms242015427] [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: 09/15/2023] [Revised: 10/06/2023] [Accepted: 10/20/2023] [Indexed: 10/29/2023] Open
Abstract
Photobiomodulation (PBM) is the regulation of biological processes using light energy from sources such as lasers or light-emitting diodes. Components of the nervous system, such as the brain and peripheral nerves, are important candidate PBM targets due to the lack of therapeutic modalities for the complete cure of neurological diseases. PBM can be applied either to regenerate damaged organs or to prevent or reduce damage caused by disease. Although recent findings have suggested that neural cells can be regenerated, which contradicts our previous understanding, neural structures are still thought to have weaker regenerative capacity than other systems. Therefore, enhancing the regenerative capacity of the nervous system would aid the future development of therapeutics for neural degeneration. PBM has been shown to enhance cell differentiation from stem or progenitor cells to near-target or target cells. In this review, we have reviewed research on the effects of PBM on neurogenesis in the central nervous system (e.g., animal brains) and the peripheral nervous system (e.g., peripheral sensory neural structures) and sought its potential as a therapeutic tool for intractable neural degenerative disorders.
Collapse
Affiliation(s)
- So-Young Chang
- Beckman Laser Institute Korea, Dankook University, Cheonan 31116, Republic of Korea;
| | - Min Young Lee
- Beckman Laser Institute Korea, Dankook University, Cheonan 31116, Republic of Korea;
- Department of Otolaryngology-Head &Neck Surgery, College of Medicine, Dankook University, Cheonan 31116, Republic of Korea
| |
Collapse
|
28
|
Chitra U, Arnold BJ, Sarkar H, Ma C, Lopez-Darwin S, Sanno K, Raphael BJ. Mapping the topography of spatial gene expression with interpretable deep learning. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.10.561757. [PMID: 37873258 PMCID: PMC10592770 DOI: 10.1101/2023.10.10.561757] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
Spatially resolved transcriptomics technologies provide high-throughput measurements of gene expression in a tissue slice, but the sparsity of this data complicates the analysis of spatial gene expression patterns such as gene expression gradients. We address these issues by deriving a topographic map of a tissue slice-analogous to a map of elevation in a landscape-using a novel quantity called the isodepth. Contours of constant isodepth enclose spatial domains with distinct cell type composition, while gradients of the isodepth indicate spatial directions of maximum change in gene expression. We develop GASTON, an unsupervised and interpretable deep learning algorithm that simultaneously learns the isodepth, spatial gene expression gradients, and piecewise linear functions of the isodepth that model both continuous gradients and discontinuous spatial variation in the expression of individual genes. We validate GASTON by showing that it accurately identifies spatial domains and marker genes across several biological systems. In SRT data from the brain, GASTON reveals gradients of neuronal differentiation and firing, and in SRT data from a tumor sample, GASTON infers gradients of metabolic activity and epithelial-mesenchymal transition (EMT)-related gene expression in the tumor microenvironment.
Collapse
Affiliation(s)
- Uthsav Chitra
- Department of Computer Science, Princeton University, Princeton, NJ, USA
| | - Brian J. Arnold
- Department of Computer Science, Princeton University, Princeton, NJ, USA
- Center for Statistics and Machine Learning, Princeton University, Princeton, NJ, USA
| | - Hirak Sarkar
- Department of Computer Science, Princeton University, Princeton, NJ, USA
- Ludwig Cancer Institute, Princeton Branch, Princeton University, Princeton, NJ, USA
| | - Cong Ma
- Department of Computer Science, Princeton University, Princeton, NJ, USA
| | | | - Kohei Sanno
- Department of Computer Science, Princeton University, Princeton, NJ, USA
- Center for Statistics and Machine Learning, Princeton University, Princeton, NJ, USA
| | | |
Collapse
|
29
|
Silvas-Baltazar M, López-Oropeza G, Durán P, Martínez-Canabal A. Olfactory neurogenesis and its role in fear memory modulation. Front Behav Neurosci 2023; 17:1278324. [PMID: 37840547 PMCID: PMC10569173 DOI: 10.3389/fnbeh.2023.1278324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Accepted: 09/12/2023] [Indexed: 10/17/2023] Open
Abstract
Olfaction is a critical sense that allows animals to navigate and understand their environment. In mammals, the critical brain structure to receive and process olfactory information is the olfactory bulb, a structure characterized by a laminated pattern with different types of neurons, some of which project to distant telencephalic structures, like the piriform cortex, the amygdala, and the hippocampal formation. Therefore, the olfactory bulb is the first structure of a complex cognitive network that relates olfaction to different types of memory, including episodic memories. The olfactory bulb continuously adds inhibitory newborn neurons throughout life; these cells locate both in the granule and glomerular layers and integrate into the olfactory circuits, inhibiting projection neurons. However, the roles of these cells modulating olfactory memories are unclear, particularly their role in fear memories. We consider that olfactory neurogenesis might modulate olfactory fear memories by a plastic process occurring in the olfactory bulb.
Collapse
Affiliation(s)
- Monserrat Silvas-Baltazar
- Licenciatura en Neurociencias, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
- Departamento de Biología Celular, Facultad de Ciencias, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Grecia López-Oropeza
- Licenciatura en Neurociencias, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
- Departamento de Biología Celular, Facultad de Ciencias, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
- Posgrado en Ciencias Biológicas, Facultad de Ciencias, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Pilar Durán
- Licenciatura en Neurociencias, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
- Departamento de Biología Celular, Facultad de Ciencias, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
- Posgrado en Ciencias Biológicas, Facultad de Ciencias, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Alonso Martínez-Canabal
- Licenciatura en Neurociencias, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
- Departamento de Biología Celular, Facultad de Ciencias, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
- Posgrado en Ciencias Biológicas, Facultad de Ciencias, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| |
Collapse
|
30
|
Laaker C, Baenen C, Kovács KG, Sandor M, Fabry Z. Immune cells as messengers from the CNS to the periphery: the role of the meningeal lymphatic system in immune cell migration from the CNS. Front Immunol 2023; 14:1233908. [PMID: 37662908 PMCID: PMC10471710 DOI: 10.3389/fimmu.2023.1233908] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 07/31/2023] [Indexed: 09/05/2023] Open
Abstract
In recent decades there has been a large focus on understanding the mechanisms of peripheral immune cell infiltration into the central nervous system (CNS) in neuroinflammatory diseases. This intense research led to several immunomodulatory therapies to attempt to regulate immune cell infiltration at the blood brain barrier (BBB), the choroid plexus (ChP) epithelium, and the glial barrier. The fate of these infiltrating immune cells depends on both the neuroinflammatory environment and their type-specific interactions with innate cells of the CNS. Although the fate of the majority of tissue infiltrating immune cells is death, a percentage of these cells could become tissue resident immune cells. Additionally, key populations of immune cells can possess the ability to "drain" out of the CNS and act as messengers reporting signals from the CNS toward peripheral lymphatics. Recent data supports that the meningeal lymphatic system is involved not just in fluid homeostatic functions in the CNS but also in facilitating immune cell migration, most notably dendritic cell migration from the CNS to the meningeal borders and to the draining cervical lymph nodes. Similar to the peripheral sites, draining immune cells from the CNS during neuroinflammation have the potential to coordinate immunity in the lymph nodes and thus influence disease. Here in this review, we will evaluate evidence of immune cell drainage from the brain via the meningeal lymphatics and establish the importance of this in animal models and humans. We will discuss how targeting immune cells at sites like the meningeal lymphatics could provide a new mechanism to better provide treatment for a variety of neurological conditions.
Collapse
Affiliation(s)
- Collin Laaker
- Neuroscience Training Program, University of Wisconsin Madison, Madison, WI, United States
| | - Cameron Baenen
- Department of Pathology and Laboratory Medicine, School of Medicine and Public Health, University of Wisconsin Madison, Madison, WI, United States
| | - Kristóf G. Kovács
- Department of Pathology and Laboratory Medicine, School of Medicine and Public Health, University of Wisconsin Madison, Madison, WI, United States
| | - Matyas Sandor
- Department of Pathology and Laboratory Medicine, School of Medicine and Public Health, University of Wisconsin Madison, Madison, WI, United States
| | - Zsuzsanna Fabry
- Department of Pathology and Laboratory Medicine, School of Medicine and Public Health, University of Wisconsin Madison, Madison, WI, United States
| |
Collapse
|
31
|
Athanassi A, Breton M, Chalençon L, Brunelin J, Didier A, Bath K, Mandairon N. Chronic unpredictable mild stress alters odor hedonics and adult olfactory neurogenesis in mice. Front Neurosci 2023; 17:1224941. [PMID: 37600017 PMCID: PMC10435088 DOI: 10.3389/fnins.2023.1224941] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Accepted: 07/17/2023] [Indexed: 08/22/2023] Open
Abstract
Experiencing chronic stress significantly increases the risk for depression. Depression is a complex disorder with varied symptoms across patients. However, feeling of sadness and decreased motivation, and diminished feeling of pleasure (anhedonia) appear to be core to most depressive pathology. Odorants are potent signals that serve a critical role in social interactions, avoiding danger, and consummatory behaviors. Diminished quality of olfactory function is associated with negative effects on quality of life leading to and aggravating the symptoms of depression. Odor hedonic value (I like or I dislike this smell) is a dominant feature of olfaction and guides approach or avoidance behavior of the odor source. The neural representation of the hedonic value of odorants is carried by the granule cells in the olfactory bulb, which functions to modulate the cortical relay of olfactory information. The granule cells of the olfactory bulb and those of the dentate gyrus are the two major populations of cells in the adult brain with continued neurogenesis into adulthood. In hippocampus, decreased neurogenesis has been linked to development or maintenance of depression symptoms. Here, we hypothesize that chronic mild stress can alter olfactory hedonics through effects on the olfactory bulb neurogenesis, contributing to the broader anhedonia phenotype in stress-associated depression. To test this, mice were subjected to chronic unpredictable mild stress and then tested on measures of depressive-like behaviors, odor hedonics, and measures of olfactory neurogenesis. Chronic unpredictable mild stress led to a selective effect on odor hedonics, diminishing attraction to pleasant but not unpleasant odorants, an effect that was accompanied by a specific decrease in adult neurogenesis and of the percentage of adult-born cells responding to pleasant odorants in the olfactory bulb.
Collapse
Affiliation(s)
- Anna Athanassi
- INSERM, U1028, CNRS UMR5292, Neuropop Team, Lyon Neuroscience Research Center, Université Claude Bernard Lyon 1, Université Jean Monnet, Bron, France
| | - Marine Breton
- INSERM, U1028, CNRS UMR5292, Neuropop Team, Lyon Neuroscience Research Center, Université Claude Bernard Lyon 1, Université Jean Monnet, Bron, France
| | - Laura Chalençon
- INSERM, U1028, CNRS UMR5292, Neuropop Team, Lyon Neuroscience Research Center, Université Claude Bernard Lyon 1, Université Jean Monnet, Bron, France
| | - Jérome Brunelin
- Centre Hospitalier Le Vinatier, Bron, France
- INSERM, U1028, CNRS UMR5292, PSYR2 Team, Lyon Neuroscience Research Center, Université Claude Bernard Lyon 1, Université Jean Monnet, Bron, France
| | - Anne Didier
- INSERM, U1028, CNRS UMR5292, Neuropop Team, Lyon Neuroscience Research Center, Université Claude Bernard Lyon 1, Université Jean Monnet, Bron, France
| | - Kevin Bath
- Division of Developmental Neuroscience, New York State Psychiatric Institute, Research Foundation for Mental Hygiene, New York, NY, United States
- Department of Psychiatry, Columbia University Medical College, New York, NY, United States
| | - Nathalie Mandairon
- INSERM, U1028, CNRS UMR5292, Neuropop Team, Lyon Neuroscience Research Center, Université Claude Bernard Lyon 1, Université Jean Monnet, Bron, France
| |
Collapse
|
32
|
Lathe R, St Clair D. Programmed ageing: decline of stem cell renewal, immunosenescence, and Alzheimer's disease. Biol Rev Camb Philos Soc 2023; 98:1424-1458. [PMID: 37068798 DOI: 10.1111/brv.12959] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 03/27/2023] [Accepted: 03/30/2023] [Indexed: 04/19/2023]
Abstract
The characteristic maximum lifespan varies enormously across animal species from a few hours to hundreds of years. This argues that maximum lifespan, and the ageing process that itself dictates lifespan, are to a large extent genetically determined. Although controversial, this is supported by firm evidence that semelparous species display evolutionarily programmed ageing in response to reproductive and environmental cues. Parabiosis experiments reveal that ageing is orchestrated systemically through the circulation, accompanied by programmed changes in hormone levels across a lifetime. This implies that, like the circadian and circannual clocks, there is a master 'clock of age' (circavital clock) located in the limbic brain of mammals that modulates systemic changes in growth factor and hormone secretion over the lifespan, as well as systemic alterations in gene expression as revealed by genomic methylation analysis. Studies on accelerated ageing in mice, as well as human longevity genes, converge on evolutionarily conserved fibroblast growth factors (FGFs) and their receptors, including KLOTHO, as well as insulin-like growth factors (IGFs) and steroid hormones, as key players mediating the systemic effects of ageing. Age-related changes in these and multiple other factors are inferred to cause a progressive decline in tissue maintenance through failure of stem cell replenishment. This most severely affects the immune system, which requires constant renewal from bone marrow stem cells. Age-related immune decline increases risk of infection whereas lifespan can be extended in germfree animals. This and other evidence suggests that infection is the major cause of death in higher organisms. Immune decline is also associated with age-related diseases. Taking the example of Alzheimer's disease (AD), we assess the evidence that AD is caused by immunosenescence and infection. The signature protein of AD brain, Aβ, is now known to be an antimicrobial peptide, and Aβ deposits in AD brain may be a response to infection rather than a cause of disease. Because some cognitively normal elderly individuals show extensive neuropathology, we argue that the location of the pathology is crucial - specifically, lesions to limbic brain are likely to accentuate immunosenescence, and could thus underlie a vicious cycle of accelerated immune decline and microbial proliferation that culminates in AD. This general model may extend to other age-related diseases, and we propose a general paradigm of organismal senescence in which declining stem cell proliferation leads to programmed immunosenescence and mortality.
Collapse
Affiliation(s)
- Richard Lathe
- Division of Infection Medicine, Chancellor's Building, University of Edinburgh Medical School, Little France, Edinburgh, EH16 4SB, UK
| | - David St Clair
- Institute of Medical Sciences, School of Medicine, University of Aberdeen, Aberdeen, AB25 2ZD, UK
| |
Collapse
|
33
|
Venegas JP, Navarrete M, Orellana-Garcia L, Rojas M, Avello-Duarte F, Nunez-Parra A. Basal Forebrain Modulation of Olfactory Coding In Vivo. Int J Psychol Res (Medellin) 2023; 16:62-86. [PMID: 38106956 PMCID: PMC10723750 DOI: 10.21500/20112084.6486] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 08/23/2022] [Accepted: 12/07/2022] [Indexed: 12/19/2023] Open
Abstract
Sensory perception is one of the most fundamental brain functions, allowing individuals to properly interact and adapt to a constantly changing environment. This process requires the integration of bottom-up and topdown neuronal activity, which is centrally mediated by the basal forebrain, a brain region that has been linked to a series of cognitive processes such as attention and alertness. Here, we review the latest research using optogenetic approaches in rodents and in vivo electrophysiological recordings that are shedding light on the role of this region, in regulating olfactory processing and decisionmaking. Moreover, we summarize evidence highlighting the anatomical and physiological differences in the basal forebrain of individuals with autism spectrum disorder, which could underpin the sensory perception abnormalities they exhibit, and propose this research line as a potential opportunity to understand the neurobiological basis of this disorder.
Collapse
Affiliation(s)
- Juan Pablo Venegas
- Physiology Laboratory, Biology Department, Faculty of Science, University of Chile, Chile.Universidad de ChileUniversity of ChileChile
| | - Marcela Navarrete
- Physiology Laboratory, Biology Department, Faculty of Science, University of Chile, Chile.Universidad de ChileUniversity of ChileChile
| | - Laura Orellana-Garcia
- Physiology Laboratory, Biology Department, Faculty of Science, University of Chile, Chile.Universidad de ChileUniversity of ChileChile
| | - Marcelo Rojas
- Physiology Laboratory, Biology Department, Faculty of Science, University of Chile, Chile.Universidad de ChileUniversity of ChileChile
| | - Felipe Avello-Duarte
- Physiology Laboratory, Biology Department, Faculty of Science, University of Chile, Chile.Universidad de ChileUniversity of ChileChile
| | - Alexia Nunez-Parra
- Physiology Laboratory, Biology Department, Faculty of Science, University of Chile, Chile.Universidad de ChileUniversity of ChileChile
| |
Collapse
|
34
|
Fang L, Kuniya T, Harada Y, Yasuda O, Maeda N, Suzuki Y, Kawaguchi D, Gotoh Y. TIMP3 promotes the maintenance of neural stem-progenitor cells in the mouse subventricular zone. Front Neurosci 2023; 17:1149603. [PMID: 37456993 PMCID: PMC10338847 DOI: 10.3389/fnins.2023.1149603] [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: 01/22/2023] [Accepted: 06/05/2023] [Indexed: 07/18/2023] Open
Abstract
Adult neural stem cells (NSCs) in the mouse subventricular zone (SVZ) serve as a lifelong reservoir for newborn olfactory bulb neurons. Recent studies have identified a slowly dividing subpopulation of embryonic neural stem-progenitor cells (NPCs) as the embryonic origin of adult NSCs. Yet, little is known about how these slowly dividing embryonic NPCs are maintained until adulthood while other NPCs are extinguished by the completion of brain development. The extracellular matrix (ECM) is an essential component of stem cell niches and thus a key determinant of stem cell fate. Here we investigated tissue inhibitors of metalloproteinases (TIMPs)-regulators of ECM remodeling-for their potential roles in the establishment of adult NSCs. We found that Timp2, Timp3, and Timp4 were expressed at high levels in slowly dividing NPCs compared to rapidly dividing NPCs. Deletion of TIMP3 reduced the number of adult NSCs and neuroblasts in the lateral SVZ. In addition, overexpression of TIMP3 in the embryonic NPCs suppressed neuronal differentiation and upregulated the expression levels of Notch signaling relating genes. These results thus suggest that TIMP3 keeps the undifferentiated state of embryonic NPCs, leading to the establishment and maintenance of adult NSCs.
Collapse
Affiliation(s)
- Lingyan Fang
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Takaaki Kuniya
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Yujin Harada
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Osamu Yasuda
- Department of Sports and Life Sciences, National Institute of Fitness and Sports in Kanoya, Kanoya, Japan
| | - Nobuyo Maeda
- Department of Sports and Life Sciences, National Institute of Fitness and Sports in Kanoya, Kanoya, Japan
| | - Yutaka Suzuki
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Chiba, Japan
| | - Daichi Kawaguchi
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Yukiko Gotoh
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
- International Research Center for Neurointelligence (WPI-IRCN), The University of Tokyo, Tokyo, Japan
| |
Collapse
|
35
|
Exploring Whether Iron Sequestration within the CNS of Patients with Alzheimer’s Disease Causes a Functional Iron Deficiency That Advances Neurodegeneration. Brain Sci 2023; 13:brainsci13030511. [PMID: 36979320 PMCID: PMC10046656 DOI: 10.3390/brainsci13030511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 03/08/2023] [Accepted: 03/13/2023] [Indexed: 03/22/2023] Open
Abstract
The involvement of iron in the pathogenesis of Alzheimer’s disease (AD) may be multifaceted. Besides potentially inducing oxidative damage, the bioavailability of iron may be limited within the central nervous system, creating a functionally iron-deficient state. By comparing staining results from baseline and modified iron histochemical protocols, iron was found to be more tightly bound within cortical sections from patients with high levels of AD pathology compared to subjects with a diagnosis of something other than AD. To begin examining whether the bound iron could cause a functional iron deficiency, a protein-coding gene expression dataset of initial, middle, and advanced stages of AD from olfactory bulb tissue was analyzed for iron-related processes with an emphasis on anemia-related changes in initial AD to capture early pathogenic events. Indeed, anemia-related processes had statistically significant alterations, and the significance of these changes exceeded those for AD-related processes. Other changes in patients with initial AD included the expressions of transcripts with iron-responsive elements and for genes encoding proteins for iron transport and mitochondrial-related processes. In the latter category, there was a decreased expression for the gene encoding pitrilysin metallopeptidase 1 (PITRM1). Other studies have shown that PITRM1 has an altered activity in patients with AD and is associated with pathological changes in this disease. Analysis of a gene expression dataset from PITRM1-deficient or sufficient organoids also revealed statistically significant changes in anemia-like processes. These findings, together with supporting evidence from the literature, raise the possibility that a pathogenic mechanism of AD could be a functional deficiency of iron contributing to neurodegeneration.
Collapse
|
36
|
Characterization by Gene Expression Analysis of Two Groups of Dopaminergic Cells Isolated from the Mouse Olfactory Bulb. BIOLOGY 2023; 12:biology12030367. [PMID: 36979058 PMCID: PMC10045757 DOI: 10.3390/biology12030367] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 02/02/2023] [Accepted: 02/23/2023] [Indexed: 03/02/2023]
Abstract
The olfactory bulb (OB) is one of two regions of the mammalian brain which undergo continuous neuronal replacement during adulthood. A significant fraction of the cells added in adulthood to the bulbar circuitry is constituted by dopaminergic (DA) neurons. We took advantage of a peculiar property of dopaminergic neurons in transgenic mice expressing eGFP under the tyrosine hydroxylase (TH) promoter: while DA neurons located in the glomerular layer (GL) display full electrophysiological maturation, eGFP+ cells in the mitral layer (ML) show characteristics of immature cells. In addition, they also display a lower fluorescence intensity, possibly reflecting different degrees of maturation. To investigate whether this difference in maturation might be confirmed at the gene expression level, we used a fluorescence-activated cell sorting technique on enzymatically dissociated cells of the OB. The cells were divided into two groups based on their level of fluorescence, possibly corresponding to immature ML cells and fully mature DA neurons from the GL. Semiquantitative real-time PCR was performed to detect the level of expression of genes linked to the degree of maturation of DA neurons. We showed that indeed the cells expressing low eGFP fluorescence are immature neurons. Our method can be further used to explore the differences between these two groups of DA neurons.
Collapse
|
37
|
Ghaffari M, Razi S, Zalpoor H, Nabi-Afjadi M, Mohebichamkhorami F, Zali H. Association of MicroRNA-146a with Type 1 and 2 Diabetes and their Related Complications. J Diabetes Res 2023; 2023:2587104. [PMID: 36911496 PMCID: PMC10005876 DOI: 10.1155/2023/2587104] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 01/17/2023] [Accepted: 02/04/2023] [Indexed: 03/06/2023] Open
Abstract
Most medical investigations have found a reduced blood level of miR-146a in type 2 diabetes (T2D) patients, suggesting an important role for miR-146a (microRNA-146a) in the etiology of diabetes mellitus (DM) and its consequences. Furthermore, injection of miR-146a mimic has been confirmed to alleviate diabetes mellitus in diabetic animal models. In this line, deregulation of miR-146a expression has been linked to the progression of nephropathy, neuropathy, wound healing, olfactory dysfunction, cardiovascular disorders, and retinopathy in diabetic patients. In this review, besides a comprehensive review of the function of miR-146a in DM, we discussed new findings on type 1 (T1MD) and type 2 (T2DM) diabetes mellitus, highlighting the discrepancies between clinical and preclinical investigations and elucidating the biological pathways regulated through miR-146a in DM-affected tissues.
Collapse
Affiliation(s)
- Mahyar Ghaffari
- Department of Biology, Parand Branch, Islamic Azad University, Parand, Iran
| | - Sara Razi
- Vira Pioneers of Modern Science (VIPOMS), Tehran, Iran
| | - Hamidreza Zalpoor
- Shiraz Neuroscience Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Network of Immunity in Infection, Malignancy & Autoimmunity (NIIMA), Universal Scientific Education & Research Network (USERN), Tehran, Iran
| | - Mohsen Nabi-Afjadi
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | | | - Hakimeh Zali
- Proteomics Research Center, Shahid Beheshti University of Medical Science, Tehram, Iran
| |
Collapse
|
38
|
Differential vulnerability of adult neurogenic niches to dosage of the neurodevelopmental-disorder linked gene Foxg1. Mol Psychiatry 2023; 28:497-514. [PMID: 35318461 PMCID: PMC9812795 DOI: 10.1038/s41380-022-01497-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 02/14/2022] [Accepted: 02/22/2022] [Indexed: 01/13/2023]
Abstract
The transcription factor FOXG1 serves pleiotropic functions in brain development ranging from the regulation of precursor proliferation to the control of cortical circuit formation. Loss-of-function mutations and duplications of FOXG1 are associated with neurodevelopmental disorders in humans illustrating the importance of FOXG1 dosage for brain development. Aberrant FOXG1 dosage has been found to disrupt the balanced activity of glutamatergic and GABAergic neurons, but the underlying mechanisms are not fully understood. We report that FOXG1 is expressed in the main adult neurogenic niches in mice, i.e. the hippocampal dentate gyrus and the subependymal zone/olfactory bulb system, where neurogenesis of glutamatergic and GABAergic neurons persists into adulthood. These niches displayed differential vulnerability to increased FOXG1 dosage: high FOXG1 levels severely compromised survival and glutamatergic dentate granule neuron fate acquisition in the hippocampal neurogenic niche, but left neurogenesis of GABAergic neurons in the subependymal zone/olfactory bulb system unaffected. Comparative transcriptomic analyses revealed a significantly higher expression of the apoptosis-linked nuclear receptor Nr4a1 in FOXG1-overexpressing hippocampal neural precursors. Strikingly, pharmacological interference with NR4A1 function rescued FOXG1-dependent death of hippocampal progenitors. Our results reveal differential vulnerability of neuronal subtypes to increased FOXG1 dosage and suggest that activity of a FOXG1/NR4A1 axis contributes to such subtype-specific response.
Collapse
|
39
|
Sancaktar M, Kocamer Şahin Ş, Demir B, Elboğa U, Elboğa G, Altındağ A. Is abnormal metabolism in the olfactory bulb and amygdala associated with bipolar disorder? J Neural Transm (Vienna) 2023; 130:145-152. [PMID: 36680695 PMCID: PMC9862245 DOI: 10.1007/s00702-023-02587-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2022] [Accepted: 01/04/2023] [Indexed: 01/22/2023]
Abstract
Accumulated evidence has demonstrated abnormal amygdala activation in bipolar disorder (BD). The olfactory bulb (OB) has vigorous connections with the amygdala. Although odor-related functions of the OB decreased during the evolutionary process, we hypothesized that an evolved OB with increased activation in emotion regulation may be one of the main factors affecting amygdala functions in BD. Our aim was to investigate metabolism in the OB and amygdala in patients with BD. Twenty-six patients diagnosed with BD according to DSM-5 diagnostic criteria were included in this cross-sectional study. Metabolism in the OB and amygdala was assessed using fluorodeoxyglucose positron emission tomography/CT in patients with BD. The OB and amygdala metabolism was compared with the patients' Z scores. Both OB and amygdala metabolic activities were significantly higher than in the controls. A positive correlation was detected between right/left amygdala metabolism and right OB metabolism (p < 0.05, r:467 and r:662, respectively). This study increased our understanding of the etiopathogenesis of BD. In BD, the main cause of hypermetabolism in the amygdala may be increased metabolism in the OB. During evolution, the OB may have assumed a dominant role in emotional processing rather than olfactory functions.
Collapse
Affiliation(s)
- Muhammet Sancaktar
- Department of Psychiatry, Faculty of Medicine, Gaziantep University, Gaziantep, Turkey
| | - Şengül Kocamer Şahin
- Department of Psychiatry, Faculty of Medicine, Gaziantep University, Gaziantep, Turkey
- Department of Psychiatry Osmangazi Neighbourhood, Gaziantep University, University Avenue- 27310 Şehitkamil, Gaziantep, Turkey
| | - Bahadır Demir
- Department of Psychiatry, Faculty of Medicine, Gaziantep University, Gaziantep, Turkey
| | - Umut Elboğa
- Department of NuclearMedıcıne, Faculty of Medicine, Gaziantep University, Gaziantep, Turkey
| | - Gülçin Elboğa
- Department of Psychiatry, Faculty of Medicine, Gaziantep University, Gaziantep, Turkey
| | - Abdurrahman Altındağ
- Department of Psychiatry, Faculty of Medicine, Gaziantep University, Gaziantep, Turkey
| |
Collapse
|
40
|
García-Gómez L, Castillo-Fernández I, Perez-Villalba A. In the pursuit of new social neurons. Neurogenesis and social behavior in mice: A systematic review. Front Cell Dev Biol 2022; 10:1011657. [PMID: 36407114 PMCID: PMC9672322 DOI: 10.3389/fcell.2022.1011657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Accepted: 10/17/2022] [Indexed: 11/06/2022] Open
Abstract
Social behaviors have become more relevant to our understanding of the human nervous system because relationships with our peers may require and modulate adult neurogenesis. Here, we review the pieces of evidence we have to date for the divergence of social behaviors in mice by modulation of adult neurogenesis or if social behaviors and the social environment can drive a change in neurogenic processes. Social recognition and memory are deeply affected by antimitotic drugs and irradiation, while NSC transgenic mice may run with lower levels of social discrimination. Interestingly, social living conditions can create a big impact on neurogenesis. Social isolation and social defeat reduce the number of new neurons, while social dominance and enrichment of the social environment increase their number. These new “social neurons” trigger functional modifications with amazing transgenerational effects. All of these suggest that we are facing two bidirectional intertwined variables, and the great challenge now is to understand the cellular and genetic mechanisms that allow this relationship to be used therapeutically.
Collapse
|
41
|
Saikarthik J, Saraswathi I, Alarifi A, Al-Atram AA, Mickeymaray S, Paramasivam A, Shaikh S, Jeraud M, Alothaim AS. Role of neuroinflammation mediated potential alterations in adult neurogenesis as a factor for neuropsychiatric symptoms in Post-Acute COVID-19 syndrome-A narrative review. PeerJ 2022; 10:e14227. [PMID: 36353605 PMCID: PMC9639419 DOI: 10.7717/peerj.14227] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 09/22/2022] [Indexed: 11/06/2022] Open
Abstract
Persistence of symptoms beyond the initial 3 to 4 weeks after infection is defined as post-acute COVID-19 syndrome (PACS). A wide range of neuropsychiatric symptoms like anxiety, depression, post-traumatic stress disorder, sleep disorders and cognitive disturbances have been observed in PACS. The review was conducted based on PRISMA-S guidelines for literature search strategy for systematic reviews. A cytokine storm in COVID-19 may cause a breach in the blood brain barrier leading to cytokine and SARS-CoV-2 entry into the brain. This triggers an immune response in the brain by activating microglia, astrocytes, and other immune cells leading to neuroinflammation. Various inflammatory biomarkers like inflammatory cytokines, chemokines, acute phase proteins and adhesion molecules have been implicated in psychiatric disorders and play a major role in the precipitation of neuropsychiatric symptoms. Impaired adult neurogenesis has been linked with a variety of disorders like depression, anxiety, cognitive decline, and dementia. Persistence of neuroinflammation was observed in COVID-19 survivors 3 months after recovery. Chronic neuroinflammation alters adult neurogenesis with pro-inflammatory cytokines supressing anti-inflammatory cytokines and chemokines favouring adult neurogenesis. Based on the prevalence of neuropsychiatric symptoms/disorders in PACS, there is more possibility for a potential impairment in adult neurogenesis in COVID-19 survivors. This narrative review aims to discuss the various neuroinflammatory processes during PACS and its effect on adult neurogenesis.
Collapse
Affiliation(s)
- Jayakumar Saikarthik
- Department of Basic Medical Sciences, College of Dentistry, Al Zulfi, Majmaah University, Al-Majmaah, Riyadh, Kingdom of Saudi Arabia,Department of Medical Education, College of Dentistry, Al Zulfi, Majmaah University, Al Majmaah, Riyadh, Kingdom of Saudi Arabia
| | - Ilango Saraswathi
- Department of Physiology, Madha Medical College and Research Institute, Chennai, Tamil Nadu, India
| | - Abdulaziz Alarifi
- Department of Basic Sciences, College of Science and Health Professions, King Saud Bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia,King Abdullah International Medical Research Centre, Riyadh, Saudi Arabia
| | - Abdulrahman A. Al-Atram
- Department of Psychiatry, College of Medicine, Majmaah University, Al Majmaah, Riyadh, Kingdom of Saudi Arabia
| | - Suresh Mickeymaray
- Department of Biology, College of Science, Al Zulfi, Majmaah University, Al Majmaah, Riyadh, Kingdom of Saudi Arabia
| | - Anand Paramasivam
- Department of Physiology, RVS Dental College and Hospital, Kumaran Kottam Campus, Kannampalayan, Coimbatore, Tamilnadu, India
| | - Saleem Shaikh
- Department of Medical Education, College of Dentistry, Al Zulfi, Majmaah University, Al Majmaah, Riyadh, Kingdom of Saudi Arabia,Department of Maxillofacial Surgery and Diagnostic Sciences, College of Dentistry, Al Zulfi, Majmaah University, Al Majmaah, Riyadh, Kingdom of Saudi Arabia
| | - Mathew Jeraud
- Department of Physiology, Ibn Sina National College for Medical Studies, Jeddah, Saudi Arabia
| | - Abdulaziz S. Alothaim
- Department of Biology, College of Science, Al Zulfi, Majmaah University, Al Majmaah, Riyadh, Kingdom of Saudi Arabia
| |
Collapse
|
42
|
Castro AE, Domínguez-Ordoñez R, Young LJ, Camacho FJ, Ávila-González D, Paredes RG, Díaz NF, Portillo W. Pair-bonding and social experience modulate new neurons survival in adult male and female prairie voles ( Microtus ochrogaster). Front Neuroanat 2022; 16:987229. [PMID: 36189119 PMCID: PMC9520527 DOI: 10.3389/fnana.2022.987229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Accepted: 08/16/2022] [Indexed: 12/04/2022] Open
Abstract
Prairie voles are a socially monogamous species that, after cohabitation with mating, form enduring pair bonds. The plastic mechanisms involved in this social behavior are not well-understood. Neurogenesis in adult rodents is a plastic neural process induced in specific brain areas like the olfactory bulbs (OB) and dentate gyrus (DG) of the hippocampus. However, it is unknown how cell survival is modulated by social or sexual experience in prairie voles. This study aimed to evaluate if cohabitation with mating and/or social exposure to a vole of the opposite sex increased the survival of the new cells in the main and accessory OB and DG. To identify the new cells and evaluate their survival, voles were injected with the DNA synthesis marker 5-bromo-2'-deoxyuridine (BrdU) and were randomly distributed into one of the following groups: (A) Control (C), voles that did not receive any sexual stimulation and were placed alone during the behavioral test. (B) Social exposure (SE), voles were individually placed in a cage equally divided into two compartments by an acrylic screen with small holes. One male and one female were placed in opposite compartments. (C) Social cohabitation with mating (SCM), animals mated freely. Our findings demonstrated that SCM females had increases in the number of new cells (BrdU-positive cells) in the main olfactory bulb and new mature neurons (BrdU/NeuN-positive cells) in the glomerular layer (GlL). In contrast, these new cells decrease in males in the SE and SCM conditions. In the granular cell layer (GrL), SCM females had more new cells and neurons than the SE group. In the accessory olfactory bulb, in the anterior GlL, SCM decreased the number of new cells and neurons in females. On the other hand, in the DG, SCM and SE increase the number of new cells in the suprapyramidal blade in female voles. Males from SCM express more new cells and neurons in the infrapyramidal blade compared with SE group. Comparison between male and females showed that new cells/neurons survival was sex dependent. These results suggest that social interaction and sexual behavior modulate cell survival and influence the neuronal fate in a sex-dependent manner, in the OB and DG. This study will contribute to understand neural mechanisms of complex social and pair bond behaviors in the prairie voles; supporting adult neurogenesis as a plastic mechanism potentially involved in social monogamous strategy.
Collapse
Affiliation(s)
- Analía E. Castro
- Instituto de Neurobiología, Universidad Nacional Autónoma de México (UNAM), Querétaro, Mexico
| | - Raymundo Domínguez-Ordoñez
- Instituto de Neurobiología, Universidad Nacional Autónoma de México (UNAM), Querétaro, Mexico
- Benemérita Universidad Autónoma de Puebla, Complejo Regional Centro, Puebla, Mexico
| | - Larry J. Young
- Silvio O. Conte Center for Oxytocin and Social Cognition, Center for Translational Social Neuroscience, Department of Psychiatry and Behavioral Sciences, Emory National Primate Research Center, Emory University, Atlanta, GA, United States
| | - Francisco J. Camacho
- Instituto de Neurobiología, Universidad Nacional Autónoma de México (UNAM), Querétaro, Mexico
| | - Daniela Ávila-González
- Instituto de Neurobiología, Universidad Nacional Autónoma de México (UNAM), Querétaro, Mexico
| | - Raúl G. Paredes
- Instituto de Neurobiología, Universidad Nacional Autónoma de México (UNAM), Querétaro, Mexico
- Escuela Nacional de Estudios Superiores, Unidad Juriquilla, Universidad Nacional Autónoma de México (UNAM), Mexico City, Mexico
| | - Nestor F. Díaz
- Instituto Nacional de Perinatología Isidro Espinosa de los Reyes, Mexico City, Mexico
| | - Wendy Portillo
- Instituto de Neurobiología, Universidad Nacional Autónoma de México (UNAM), Querétaro, Mexico
| |
Collapse
|
43
|
Kumaria A, Noah A, Kirkman MA. Does covid-19 impair endogenous neurogenesis? J Clin Neurosci 2022; 105:79-85. [PMID: 36113246 DOI: 10.1016/j.jocn.2022.09.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 08/14/2022] [Accepted: 09/03/2022] [Indexed: 10/31/2022]
Abstract
Endogenous neural stem cells are thought to continue to generate new neurons throughout life in the human brain. Endogenous neurogenesis has been proposed to contribute to physiological roles in maintaining and regenerating olfaction, as well as promoting normal cognition, learning and memory. Specific impairments in these processes in COVID-19 - impaired olfaction and cognition - may implicate the SARS-CoV-2 virus in attenuating neurogenesis. Furthermore, neurogenesis has been linked with neuroregeneration; and impaired neuroregeneration has previously been linked with neurodegenerative diseases. Emerging evidence supports an association between COVID-19 infection and accelerated neurodegeneration. Also, structural changes indicating global reduction in brain size and specific reduction in the size of limbic structures - including orbitofrontal cortex, olfactory cortex and parahippocampal gyrus - as a result of SARS-CoV-2 infection have been demonstrated. This paper proposes the hypothesis that SARS-CoV-2 infection may impair endogenous neural stem cell activity. An attenuation of neurogenesis may contribute to reduction in brain size and/or neurodegenerative processes following SARS-CoV-2 infection. Furthermore, as neural stem cells are thought to be the cell of origin in glioma, better understanding of SARS-CoV-2 interaction with tumorigenic stem cells is indicated, with a view to informing therapeutic modulation. The subacute and chronic implications of attenuated endogenous neurogenesis are explored in the context of long COVID. Modulating endogenous neurogenesis may be a novel therapeutic strategy to address specific neurological manifestations of COVID-19 and potential applicability in tumour virotherapy.
Collapse
Affiliation(s)
- Ashwin Kumaria
- Department of Neurosurgery, Queen's Medical Centre, Nottingham University Hospitals NHS Trust, Nottingham, UK
| | - Abiodun Noah
- Anaesthesia and Critical Care, Academic Unit of Injury, Inflammation and Recovery Sciences, Queen's Medical Centre, University of Nottingham, Nottingham, UK
| | - Matthew A Kirkman
- Department of Neurosurgery, Queen's Medical Centre, Nottingham University Hospitals NHS Trust, Nottingham, UK
| |
Collapse
|
44
|
Bayat AH, Azimi H, Hassani Moghaddam M, Ebrahimi V, Fathi M, Vakili K, Mahmoudiasl GR, Forouzesh M, Boroujeni ME, Nariman Z, Abbaszadeh HA, Aryan A, Aliaghaei A, Abdollahifar MA. COVID-19 causes neuronal degeneration and reduces neurogenesis in human hippocampus. Apoptosis 2022; 27:852-868. [PMID: 35876935 PMCID: PMC9310365 DOI: 10.1007/s10495-022-01754-9] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/15/2022] [Indexed: 11/30/2022]
Abstract
Recent investigations of COVID-19 have largely focused on the effects of this novel virus on the vital organs in order to efficiently assist individuals who have recovered from the disease. In the present study we used hippocampal tissue samples extracted from people who died after COVID-19. Utilizing histological techniques to analyze glial and neuronal cells we illuminated a massive degeneration of neuronal cells and changes in glial cells morphology in hippocampal samples. The results showed that in hippocampus of the studied brains there were morphological changes in pyramidal cells, an increase in apoptosis, a drop in neurogenesis, and change in spatial distribution of neurons in the pyramidal and granular layer. It was also demonstrated that COVID-19 alter the morphological characteristics and distribution of astrocyte and microglia cells. While the exact mechanism(s) by which the virus causes neuronal loss and morphology in the central nervous system (CNS) remains to be determined, it is necessary to monitor the effect of SARS-CoV-2 infection on CNS compartments like the hippocampus in future investigations. As a result of what happened in the hippocampus secondary to COVID-19, memory impairment may be a long-term neurological complication which can be a predisposing factor for neurodegenerative disorders through neuroinflammation and oxidative stress mechanisms.
Collapse
Affiliation(s)
- Amir-Hossein Bayat
- Department of Basic Sciences, Saveh University of Medical Sciences, Saveh, Iran
| | - Helia Azimi
- Skull Base Research Center, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Meysam Hassani Moghaddam
- Department of Anatomical Sciences, Faculty of Medicine, AJA University of Medical Sciences, Tehran, Iran
| | - Vahid Ebrahimi
- Department of Anatomy and Cell Biology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mobina Fathi
- Student Research Committee, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Kimia Vakili
- Student Research Committee, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Mahdi Forouzesh
- Legal Medicine Research Center, Iranian Legal Medicine Organization, Tehran, Iran
| | - Mahdi Eskandarian Boroujeni
- Laboratory of Human Molecular Genetics, Institute of Molecular Biology and Biotechnology, Faculty of Biology, Adam Mickiewicz University, Poznan, Poland
| | - Zahra Nariman
- Skull Base Research Center, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hojjat-Allah Abbaszadeh
- Laser Application in Medical Sciences Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Arefeh Aryan
- Anatomy Department, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Abbas Aliaghaei
- Skull Base Research Center, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran. .,Department of Cell Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran. .,Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Mohammad-Amin Abdollahifar
- Skull Base Research Center, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran. .,Department of Cell Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
45
|
Culig L, Chu X, Bohr VA. Neurogenesis in aging and age-related neurodegenerative diseases. Ageing Res Rev 2022; 78:101636. [PMID: 35490966 PMCID: PMC9168971 DOI: 10.1016/j.arr.2022.101636] [Citation(s) in RCA: 86] [Impact Index Per Article: 28.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 04/14/2022] [Accepted: 04/25/2022] [Indexed: 12/11/2022]
Abstract
Adult neurogenesis, the process by which neurons are generated in certain areas of the adult brain, declines in an age-dependent manner and is one potential target for extending cognitive healthspan. Aging is a major risk factor for neurodegenerative diseases and, as lifespans are increasing, these health challenges are becoming more prevalent. An age-associated loss in neural stem cell number and/or activity could cause this decline in brain function, so interventions that reverse aging in stem cells might increase the human cognitive healthspan. In this review, we describe the involvement of adult neurogenesis in neurodegenerative diseases and address the molecular mechanistic aspects of neurogenesis that involve some of the key aggregation-prone proteins in the brain (i.e., tau, Aβ, α-synuclein, …). We summarize the research pertaining to interventions that increase neurogenesis and regulate known targets in aging research, such as mTOR and sirtuins. Lastly, we share our outlook on restoring the levels of neurogenesis to physiological levels in elderly individuals and those with neurodegeneration. We suggest that modulating neurogenesis represents a potential target for interventions that could help in the fight against neurodegeneration and cognitive decline.
Collapse
Affiliation(s)
- Luka Culig
- Section on DNA Repair, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | - Xixia Chu
- Section on DNA Repair, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | - Vilhelm A Bohr
- Section on DNA Repair, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA.
| |
Collapse
|
46
|
Gao C, Wu M, Du Q, Deng J, Shen J. Naringin Mediates Adult Hippocampal Neurogenesis for Antidepression via Activating CREB Signaling. Front Cell Dev Biol 2022; 10:731831. [PMID: 35478969 PMCID: PMC9037031 DOI: 10.3389/fcell.2022.731831] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Accepted: 03/07/2022] [Indexed: 11/17/2022] Open
Abstract
The brain-derived neurotrophic factor/tropomyosin receptor kinase B/cAMP response element-binding protein (BDNF/TrkB/CREB) signaling pathway is a critical therapeutic target for inducing adult hippocampal neurogenesis and antidepressant therapy. In this study, we tested the hypothesis that naringin, a natural medicinal compound, could promote adult hippocampal neurogenesis and improve depression-like behaviors via regulating the BDNF/TrkB/CREB signaling pathway. We first investigated the effects of naringin on promoting adult hippocampal neurogenesis in both normal and chronic corticosterone (CORT)-induced depressive mice. Under physiological condition, naringin treatment enhanced the proliferation of neural stem/progenitor cells (NSPCs) and accelerated neuronal differentiation. In CORT-induced depression mouse model, naringin treatment promoted neuronal differentiation and maturation of NSPCs for hippocampal neurogenesis. Forced swim test, tail suspension test, and open field test confirmed the antidepressant and anxiolytic effects of naringin. Co-treatment of temozolomide (TMZ), a neurogenic inhibitor, abolished these antidepressant and anxiolytic effects. Meanwhile, naringin treatment increased phosphorylation of cAMP response element binding protein (CREB) but had no effect on the expression of brain-derived neurotrophic factor and phosphorylation of TrkB in the hippocampus of CORT-induced depressive mice. Co-treatment of CREB inhibitor 666-15, rather than TrkB inhibitor Cyc-B, abolished the neurogenesis-promoting and antidepressant effects of naringin. Taken together, naringin has antidepressant and anxiolytic effects, and the underlying mechanisms could be attributed to enhance hippocampal neurogenesis via activating CREB signaling.
Collapse
Affiliation(s)
- Chong Gao
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hon Kong SAR, China
- The Institute of Brain and Cognitive Sciences, School of Medicine, Zhejiang University City College, Hangzhou, China
| | - Meiling Wu
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hon Kong SAR, China
| | - Qiaohui Du
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hon Kong SAR, China
| | - Jiagang Deng
- Guangxi Key Laboratory of Efficacy Study on Chinese Materia Medica, Guangxi University of Chinese Medicine, Nanning, China
| | - Jiangang Shen
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hon Kong SAR, China
- *Correspondence: Jiangang Shen,
| |
Collapse
|
47
|
Patel ZM, Holbrook EH, Turner JH, Adappa ND, Albers MW, Altundag A, Appenzeller S, Costanzo RM, Croy I, Davis GE, Dehgani-Mobaraki P, Doty RL, Duffy VB, Goldstein BJ, Gudis DA, Haehner A, Higgins TS, Hopkins C, Huart C, Hummel T, Jitaroon K, Kern RC, Khanwalkar AR, Kobayashi M, Kondo K, Lane AP, Lechner M, Leopold DA, Levy JM, Marmura MJ, Mclelland L, Miwa T, Moberg PJ, Mueller CA, Nigwekar SU, O'Brien EK, Paunescu TG, Pellegrino R, Philpott C, Pinto JM, Reiter ER, Roalf DR, Rowan NR, Schlosser RJ, Schwob J, Seiden AM, Smith TL, Soler ZM, Sowerby L, Tan BK, Thamboo A, Wrobel B, Yan CH. International consensus statement on allergy and rhinology: Olfaction. Int Forum Allergy Rhinol 2022; 12:327-680. [PMID: 35373533 DOI: 10.1002/alr.22929] [Citation(s) in RCA: 59] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 01/01/2021] [Accepted: 11/19/2021] [Indexed: 12/11/2022]
Abstract
BACKGROUND The literature regarding clinical olfaction, olfactory loss, and olfactory dysfunction has expanded rapidly over the past two decades, with an exponential rise in the past year. There is substantial variability in the quality of this literature and a need to consolidate and critically review the evidence. It is with that aim that we have gathered experts from around the world to produce this International Consensus on Allergy and Rhinology: Olfaction (ICAR:O). METHODS Using previously described methodology, specific topics were developed relating to olfaction. Each topic was assigned a literature review, evidence-based review, or evidence-based review with recommendations format as dictated by available evidence and scope within the ICAR:O document. Following iterative reviews of each topic, the ICAR:O document was integrated and reviewed by all authors for final consensus. RESULTS The ICAR:O document reviews nearly 100 separate topics within the realm of olfaction, including diagnosis, epidemiology, disease burden, diagnosis, testing, etiology, treatment, and associated pathologies. CONCLUSION This critical review of the existing clinical olfaction literature provides much needed insight and clarity into the evaluation, diagnosis, and treatment of patients with olfactory dysfunction, while also clearly delineating gaps in our knowledge and evidence base that we should investigate further.
Collapse
Affiliation(s)
- Zara M Patel
- Otolaryngology, Stanford University School of Medicine, Stanford, California, USA
| | - Eric H Holbrook
- Otolaryngology, Massachusetts Eye and Ear Infirmary, Boston, Massachusetts, USA
| | - Justin H Turner
- Otolaryngology, Vanderbilt School of Medicine, Nashville, Tennessee, USA
| | - Nithin D Adappa
- Otolaryngology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Mark W Albers
- Neurology, Harvard Medical School, Boston, Massachusetts, USA
| | - Aytug Altundag
- Otolaryngology, Biruni University School of Medicine, İstanbul, Turkey
| | - Simone Appenzeller
- Rheumatology, School of Medical Sciences, University of Campinas, São Paulo, Brazil
| | - Richard M Costanzo
- Physiology and Biophysics and Otolaryngology, Virginia Commonwealth University School of Medicine, Richmond, Virginia, USA
| | - Ilona Croy
- Psychology and Psychosomatic Medicine, TU Dresden, Dresden, Germany
| | - Greg E Davis
- Otolaryngology, Proliance Surgeons, Seattle and Puyallup, Washington, USA
| | - Puya Dehgani-Mobaraki
- Associazione Naso Sano, Umbria Regional Registry of Volunteer Activities, Corciano, Italy
| | - Richard L Doty
- Smell and Taste Center, Otolaryngology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Valerie B Duffy
- Allied Health Sciences, University of Connecticut, Storrs, Connecticut, USA
| | | | - David A Gudis
- Otolaryngology, Columbia University Irving Medical Center, New York, USA
| | - Antje Haehner
- Smell and Taste, Otolaryngology, TU Dresden, Dresden, Germany
| | - Thomas S Higgins
- Otolaryngology, University of Louisville School of Medicine, Louisville, Kentucky, USA
| | - Claire Hopkins
- Otolaryngology, Guy's and St. Thomas' Hospitals, London Bridge Hospital, London, UK
| | - Caroline Huart
- Otorhinolaryngology, Cliniques universitaires Saint-Luc, Institute of Neuroscience, Université catholgique de Louvain, Brussels, Belgium
| | - Thomas Hummel
- Smell and Taste, Otolaryngology, TU Dresden, Dresden, Germany
| | | | - Robert C Kern
- Otolaryngology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Ashoke R Khanwalkar
- Otolaryngology, Stanford University School of Medicine, Stanford, California, USA
| | - Masayoshi Kobayashi
- Otorhinolaryngology-Head and Neck Surgery, Mie University Graduate School of Medicine, Mie, Japan
| | - Kenji Kondo
- Otolaryngology, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
| | - Andrew P Lane
- Otolaryngology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Matt Lechner
- Otolaryngology, Barts Health and University College London, London, UK
| | - Donald A Leopold
- Otolaryngology, University of Vermont Medical Center, Burlington, Vermont, USA
| | - Joshua M Levy
- Otolaryngology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Michael J Marmura
- Neurology Thomas Jefferson University School of Medicine, Philadelphia, Pennsylvania, USA
| | - Lisha Mclelland
- Otolaryngology, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - Takaki Miwa
- Otolaryngology, Kanazawa Medical University, Ishikawa, Japan
| | - Paul J Moberg
- Psychiatry, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
| | | | - Sagar U Nigwekar
- Division of Nephrology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Erin K O'Brien
- Otolaryngology, Mayo Clinic Rochester, Rochester, Minnesota, USA
| | - Teodor G Paunescu
- Division of Nephrology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | | | - Carl Philpott
- Otolaryngology, University of East Anglia, Norwich, UK
| | - Jayant M Pinto
- Otolaryngology, University of Chicago, Chicago, Illinois, USA
| | - Evan R Reiter
- Otolaryngology, Virginia Commonwealth University School of Medicine, Richmond, Virginia, USA
| | - David R Roalf
- Psychiatry, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
| | - Nicholas R Rowan
- Otolaryngology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Rodney J Schlosser
- Otolaryngology, Medical University of South Carolina, Mt Pleasant, South Carolina, USA
| | - James Schwob
- Biomedical Sciences, Tufts University School of Medicine, Boston, Massachusetts, USA
| | - Allen M Seiden
- Otolaryngology, University of Cincinnati School of Medicine, Cincinnati, Ohio, USA
| | - Timothy L Smith
- Otolaryngology, Oregon Health and Sciences University, Portland, Oregon, USA
| | - Zachary M Soler
- Otolaryngology, Medical University of South Carolina, Mt Pleasant, South Carolina, USA
| | - Leigh Sowerby
- Otolaryngology, University of Western Ontario, London, Ontario, Canada
| | - Bruce K Tan
- Otolaryngology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Andrew Thamboo
- Otolaryngology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Bozena Wrobel
- Otolaryngology, Keck School of Medicine, USC, Los Angeles, California, USA
| | - Carol H Yan
- Otolaryngology, School of Medicine, UCSD, La Jolla, California, USA
| |
Collapse
|
48
|
del Águila Á, Adam M, Ullom K, Shaw N, Qin S, Ehrman J, Nardini D, Salomone J, Gebelein B, Lu QR, Potter SS, Waclaw R, Campbell K, Nakafuku M. Olig2 defines a subset of neural stem cells that produce specific olfactory bulb interneuron subtypes in the subventricular zone of adult mice. Development 2022; 149:274286. [PMID: 35132995 PMCID: PMC8959153 DOI: 10.1242/dev.200028] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Accepted: 01/24/2022] [Indexed: 12/12/2022]
Abstract
Distinct neural stem cells (NSCs) reside in different regions of the subventricular zone (SVZ) and generate multiple olfactory bulb (OB) interneuron subtypes in the adult brain. However, the molecular mechanisms underlying such NSC heterogeneity remain largely unknown. Here, we show that the basic helix-loop-helix transcription factor Olig2 defines a subset of NSCs in the early postnatal and adult SVZ. Olig2-expressing NSCs exist broadly but are most enriched in the ventral SVZ along the dorsoventral axis complementary to dorsally enriched Gsx2-expressing NSCs. Comparisons of Olig2-expressing NSCs from early embryonic to adult stages using single cell transcriptomics reveal stepwise developmental changes in their cell cycle and metabolic properties. Genetic studies further show that cross-repression contributes to the mutually exclusive expression of Olig2 and Gsx2 in NSCs/progenitors during embryogenesis, but that their expression is regulated independently from each other in adult NSCs. Finally, lineage-tracing and conditional inactivation studies demonstrate that Olig2 plays an important role in the specification of OB interneuron subtypes. Altogether, our study demonstrates that Olig2 defines a unique subset of adult NSCs enriched in the ventral aspect of the adult SVZ.
Collapse
Affiliation(s)
- Ángela del Águila
- Division of Developmental Biology, Cincinnati Children's Hospital Research Foundation, 3333 Burnet Avenue, Cincinnati, OH 45229-3039, USA
| | - Mike Adam
- Division of Developmental Biology, Cincinnati Children's Hospital Research Foundation, 3333 Burnet Avenue, Cincinnati, OH 45229-3039, USA
| | - Kristy Ullom
- Division of Developmental Biology, Cincinnati Children's Hospital Research Foundation, 3333 Burnet Avenue, Cincinnati, OH 45229-3039, USA
| | - Nicholas Shaw
- Division of Developmental Biology, Cincinnati Children's Hospital Research Foundation, 3333 Burnet Avenue, Cincinnati, OH 45229-3039, USA,Department of Medical Science, University of Cincinnati College of Medicine, 3125 Eden Avenue, Cincinnati, OH 45267-0521, USA
| | - Shenyue Qin
- Division of Developmental Biology, Cincinnati Children's Hospital Research Foundation, 3333 Burnet Avenue, Cincinnati, OH 45229-3039, USA
| | - Jacqueline Ehrman
- Division of Developmental Biology, Cincinnati Children's Hospital Research Foundation, 3333 Burnet Avenue, Cincinnati, OH 45229-3039, USA
| | - Diana Nardini
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Research Foundation, 3333 Burnet Avenue, Cincinnati, OH 45229-3039, USA
| | - Joseph Salomone
- Division of Developmental Biology, Cincinnati Children's Hospital Research Foundation, 3333 Burnet Avenue, Cincinnati, OH 45229-3039, USA
| | - Brian Gebelein
- Division of Developmental Biology, Cincinnati Children's Hospital Research Foundation, 3333 Burnet Avenue, Cincinnati, OH 45229-3039, USA
| | - Q. Richard Lu
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Research Foundation, 3333 Burnet Avenue, Cincinnati, OH 45229-3039, USA,Department of Pediatrics, University of Cincinnati College of Medicine, 3125 Eden Avenue, Cincinnati, OH 45267-0521, USA
| | - Steven S. Potter
- Division of Developmental Biology, Cincinnati Children's Hospital Research Foundation, 3333 Burnet Avenue, Cincinnati, OH 45229-3039, USA,Department of Pediatrics, University of Cincinnati College of Medicine, 3125 Eden Avenue, Cincinnati, OH 45267-0521, USA
| | - Ronald Waclaw
- Division of Developmental Biology, Cincinnati Children's Hospital Research Foundation, 3333 Burnet Avenue, Cincinnati, OH 45229-3039, USA,Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Research Foundation, 3333 Burnet Avenue, Cincinnati, OH 45229-3039, USA,Department of Pediatrics, University of Cincinnati College of Medicine, 3125 Eden Avenue, Cincinnati, OH 45267-0521, USA
| | - Kenneth Campbell
- Division of Developmental Biology, Cincinnati Children's Hospital Research Foundation, 3333 Burnet Avenue, Cincinnati, OH 45229-3039, USA,Department of Pediatrics, University of Cincinnati College of Medicine, 3125 Eden Avenue, Cincinnati, OH 45267-0521, USA,Division of Neurosurgery, Cincinnati Children's Hospital Research Foundation, 3333 Burnet Avenue, Cincinnati, OH 45229-3039, USA
| | - Masato Nakafuku
- Division of Developmental Biology, Cincinnati Children's Hospital Research Foundation, 3333 Burnet Avenue, Cincinnati, OH 45229-3039, USA,Department of Pediatrics, University of Cincinnati College of Medicine, 3125 Eden Avenue, Cincinnati, OH 45267-0521, USA,Department of Neurosurgery, University of Cincinnati College of Medicine, 3125 Eden Avenue, Cincinnati, OH 45267-0521, USA,Author for correspondence ()
| |
Collapse
|
49
|
Kersen DEC, Tavoni G, Balasubramanian V. Connectivity and dynamics in the olfactory bulb. PLoS Comput Biol 2022; 18:e1009856. [PMID: 35130267 PMCID: PMC8853646 DOI: 10.1371/journal.pcbi.1009856] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 02/17/2022] [Accepted: 01/22/2022] [Indexed: 12/22/2022] Open
Abstract
Dendrodendritic interactions between excitatory mitral cells and inhibitory granule cells in the olfactory bulb create a dense interaction network, reorganizing sensory representations of odors and, consequently, perception. Large-scale computational models are needed for revealing how the collective behavior of this network emerges from its global architecture. We propose an approach where we summarize anatomical information through dendritic geometry and density distributions which we use to calculate the connection probability between mitral and granule cells, while capturing activity patterns of each cell type in the neural dynamical systems theory of Izhikevich. In this way, we generate an efficient, anatomically and physiologically realistic large-scale model of the olfactory bulb network. Our model reproduces known connectivity between sister vs. non-sister mitral cells; measured patterns of lateral inhibition; and theta, beta, and gamma oscillations. The model in turn predicts testable relationships between network structure and several functional properties, including lateral inhibition, odor pattern decorrelation, and LFP oscillation frequency. We use the model to explore the influence of cortex on the olfactory bulb, demonstrating possible mechanisms by which cortical feedback to mitral cells or granule cells can influence bulbar activity, as well as how neurogenesis can improve bulbar decorrelation without requiring cell death. Our methodology provides a tractable tool for other researchers.
Collapse
Affiliation(s)
- David E. Chen Kersen
- Computational Neuroscience Initiative, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Gaia Tavoni
- Computational Neuroscience Initiative, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
- Department of Physics and Astronomy, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
- Department of Neuroscience, Washington University in St. Louis, St. Louis, Missouri, United States of America
| | - Vijay Balasubramanian
- Computational Neuroscience Initiative, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
- Department of Physics and Astronomy, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
- Department of Neuroscience, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| |
Collapse
|
50
|
Malloul H, Bonzano S, Bennis M, De Marchis S, Ba-M'hamed S. Chronic thinner inhalation alters olfactory behaviors in adult mice. Behav Brain Res 2022; 417:113597. [PMID: 34563601 DOI: 10.1016/j.bbr.2021.113597] [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: 04/14/2021] [Revised: 09/01/2021] [Accepted: 09/20/2021] [Indexed: 12/17/2022]
Abstract
Volatile solvents exposure can result in various behavioral impairments that have been partly associated to altered adult hippocampal neurogenesis. Despite recent evidence supporting this association, few studies have been devoted to examine the impact on olfactory functioning and olfactory bulb (OB) neurogenesis, although olfactory system is directly in contact with volatile molecules. Thus, this study was designed to evaluate in adult mice the potential modifications of the olfactory functioning after acute (1 day), subchronic (6 weeks) and chronic (12 weeks) exposure to thinner vapor at both behavioral and cellular levels. Firstly, behavioral evaluations showed that chronic thinner exposure impacts on odor detection ability of treated mice but does not affect mice ability to efficiently discriminate between two different odors. Moreover, chronic thinner exposure produces impairment in the olfactory-mediated associative memory. Secondly, analysis of the effects of thinner exposure in the subventricular zone (SVZ) of the lateral ventricle and in the OB revealed that thinner treatments do not induce apoptosis nor glial activation. Thirdly, immunohistochemical quantification of different markers of adult olfactory neurogenesis showed that inhalant treatments do not change the number of proliferating progenitors in the SVZ and the rostral migratory stream (RMS), as well as the number of newborn cells reaching and integrating in the OB circuitry. Altogether, our data highlight that the impaired olfactory performances in chronically-exposed mice are not associated to an alteration of adult neurogenesis in the SVZ-OB system.
Collapse
Affiliation(s)
- Hanaa Malloul
- Laboratory of Pharmacology, Neurobiology, Anthropobiology and Environment, Faculty of Sciences Semlalia, Cadi Ayyad University, Marrakech, Morocco; Department of Life Sciences and Systems Biology, University of Turin, Turin, Italy; Neuroscience Institute Cavalieri Ottolenghi (NICO), Orbassano, Italy
| | - Sara Bonzano
- Department of Life Sciences and Systems Biology, University of Turin, Turin, Italy; Neuroscience Institute Cavalieri Ottolenghi (NICO), Orbassano, Italy
| | - Mohammed Bennis
- Laboratory of Pharmacology, Neurobiology, Anthropobiology and Environment, Faculty of Sciences Semlalia, Cadi Ayyad University, Marrakech, Morocco.
| | - Silvia De Marchis
- Department of Life Sciences and Systems Biology, University of Turin, Turin, Italy; Neuroscience Institute Cavalieri Ottolenghi (NICO), Orbassano, Italy
| | - Saadia Ba-M'hamed
- Laboratory of Pharmacology, Neurobiology, Anthropobiology and Environment, Faculty of Sciences Semlalia, Cadi Ayyad University, Marrakech, Morocco
| |
Collapse
|