1
|
Lugenbühl JF, Viho EMG, Binder EB, Daskalakis NP. Stress Molecular Signaling in Interaction With Cognition. Biol Psychiatry 2025; 97:349-358. [PMID: 39368530 DOI: 10.1016/j.biopsych.2024.09.023] [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: 06/02/2024] [Revised: 09/02/2024] [Accepted: 09/27/2024] [Indexed: 10/07/2024]
Abstract
Exposure to stressful life events is associated with a high risk of developing psychiatric disorders with a wide variety of symptoms. Cognitive symptoms in stress-related psychiatric disorders can be particularly challenging to understand, both for those experiencing them and for health care providers. To gain insights, it is important to capture stress-induced structural, epigenomic, transcriptomic, and proteomic changes in relevant brain regions such as the amygdala, hippocampus, locus coeruleus, and prefrontal cortex that result in long-lasting alterations in brain function. In this review, we will emphasize a subset of stress molecular mechanisms that alter neuroplasticity, neurogenesis, and balance between excitatory and inhibitory neurons. Then, we discuss how to identify genetic risk factors that may accelerate stress-driven or stress-induced cognitive impairment. Despite the development of new technologies such as single-cell resolution sequencing, our understanding of the molecular effects of stress in the brain remains to be deepened. A better understanding of the diversity of stress effects in different brain regions and cell types is a prerequisite to open new avenues for mechanism-informed prevention and treatment of stress-related cognitive symptoms.
Collapse
Affiliation(s)
- Justina F Lugenbühl
- Department of Psychiatry, McLean Hospital, Harvard Medical School, Belmont, Massachusetts; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, Massachusetts; Department of Psychiatry and Neuropsychology, School for Mental Health, and Neuroscience, Maastricht University, Maastricht, the Netherlands
| | - Eva M G Viho
- Department Genes and Environment, Max Planck Institute of Psychiatry, Munich, Germany
| | - Elisabeth B Binder
- Department Genes and Environment, Max Planck Institute of Psychiatry, Munich, Germany.
| | - Nikolaos P Daskalakis
- Department of Psychiatry, McLean Hospital, Harvard Medical School, Belmont, Massachusetts; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, Massachusetts
| |
Collapse
|
2
|
Chen R, Lu X, Xiao A, Ma J. Role of insulin-like growth factor-2 in Alzheimer's disease induced memory impairment and underlying mechanisms. Front Cell Neurosci 2025; 18:1520253. [PMID: 39830039 PMCID: PMC11739150 DOI: 10.3389/fncel.2024.1520253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Accepted: 12/13/2024] [Indexed: 01/22/2025] Open
Abstract
Alzheimer's disease (AD) is the most prevalent type of dementia. Treatments for AD do not reverse the loss of brain function; rather, they decrease the rate of cognitive deterioration. Current treatments are ineffective in part because they do not address neurotrophic mechanisms, which are believed to be critical for functional recovery. Given that structural losses are assumed to be the root cause of cognitive impairment in AD, strengthening neurotrophic pathways may be a useful preventative therapeutic approach. Insulin-like growth factor-2 (IGF2), which is widely expressed in the central nervous system (CNS), has emerged as a crucial mechanism of synaptic plasticity and learning and memory, and many studies have indicated that this neurotrophic peptide is a viable candidate for treating and preventing AD-induced cognitive decline. An increase in IGF2 levels improves memory in healthy animals and alleviates several symptoms associated with neurodegenerative disorders. These effects are primarily caused by the IGF2 receptor, which is widely expressed in neurons and controls protein trafficking, synthesis, and degradation. However, the use of IGF2 as a potential target for the development of novel pharmaceuticals to treat AD-induced memory impairment needs further investigation. We compiled recent studies on the role of IGF2 in AD-associated memory issues and summarized the current knowledge regarding IGF2 expression and function in the brain, specifically in AD-induced memory impairment.
Collapse
Affiliation(s)
- Ruiqi Chen
- Department of Neurosurgery, West China Hospital of Sichuan University, Chengdu, China
| | - Xing Lu
- Department of Gynecological Nursing, West China Second Hospital, Sichuan University, Chengdu, China
| | - Anqi Xiao
- Department of Neurosurgery, West China Hospital of Sichuan University, Chengdu, China
| | - Junpeng Ma
- Department of Neurosurgery, West China Hospital of Sichuan University, Chengdu, China
- Department of Neurosurgery, West China Tianfu Hospital of Sichuan University, Chengdu, China
| |
Collapse
|
3
|
Kochunov P, Hong LE, Summerfelt A, Gao S, Brown PL, Terzi M, Acheson A, Woldorff MG, Fieremans E, Abdollahzadeh A, Sathyasaikumar KV, Clark SM, Schwarcz R, Shepard PD, Elmer GI. White matter and latency of visual evoked potentials during maturation: A miniature pig model of adolescent development. J Neurosci Methods 2024; 411:110252. [PMID: 39159872 DOI: 10.1016/j.jneumeth.2024.110252] [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: 01/10/2024] [Revised: 07/17/2024] [Accepted: 08/16/2024] [Indexed: 08/21/2024]
Abstract
BACKGROUND Continuous myelination of cerebral white matter (WM) during adolescence overlaps with the formation of higher cognitive skills and the onset of many neuropsychiatric disorders. We developed a miniature-pig model of adolescent brain development for neuroimaging and neurophysiological assessment during this critical period. Minipigs have gyroencephalic brains with a large cerebral WM compartment and a well-defined adolescence period. METHODS Eight Sinclair™ minipigs (Sus scrofa domestica) were evaluated four times during weeks 14-28 (40, 28 and 28 days apart) of adolescence using monocular visual stimulation (1 Hz)-evoked potentials and diffusion MRI (dMRI) of WM. The latency for the pre-positive 30 ms (PP30), positive 30 ms (P30) and negative 50 ms (N50) components of the flash visual evoked potentials (fVEPs) and their interhemispheric latency (IL) were recorded in the frontal, central and occipital areas during ten 60-second stimulations for each eye. The dMRI imaging protocol consisted of fifteen b-shells (b = 0-3500 s/mm2) with 32 directions/shell, providing measurements that included fractional anisotropy (FA), radial kurtosis, kurtosis anisotropy (KA), axonal water fraction (AWF), and the permeability-diffusivity index (PDI). RESULTS Significant reductions (p < 0.05) in the latency and IL of fVEP measurements paralleled significant rises in FA, KA, AWF and PDI over the same period. The longitudinal latency changes in fVEPs were primarily associated with whole-brain changes in diffusion parameters, while fVEP IL changes were related to maturation of the corpus callosum. CONCLUSIONS Good agreement between reduction in the latency of fVEPs and maturation of cerebral WM was interpreted as evidence for ongoing myelination and confirmation of the minipig as a viable research platform. Adolescent development in minipigs can be studied using human neuroimaging and neurophysiological protocols and followed up with more invasive assays to investigate key neurodevelopmental hypotheses in psychiatry.
Collapse
Affiliation(s)
- Peter Kochunov
- Faillace Department of Psychiatry and Behavioral Sciences at McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA; Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA.
| | - L Elliot Hong
- Faillace Department of Psychiatry and Behavioral Sciences at McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Ann Summerfelt
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Si Gao
- Faillace Department of Psychiatry and Behavioral Sciences at McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA; Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA
| | - P Leon Brown
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Matthew Terzi
- Department of Pathology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Ashley Acheson
- Department of Psychiatry, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Marty G Woldorff
- Center for Cognitive Neuroscience, Duke University, Durham, NC. USA
| | - Els Fieremans
- Center for Biomedical Imaging, Department of Radiology, New York University School of Medicine, New York, NY, USA
| | - Ali Abdollahzadeh
- Center for Biomedical Imaging, Department of Radiology, New York University School of Medicine, New York, NY, USA
| | - Korrapati V Sathyasaikumar
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Sarah M Clark
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA; Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Robert Schwarcz
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Paul D Shepard
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Greg I Elmer
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
4
|
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
|
5
|
Simard S, Rahimian R, Davoli MA, Théberge S, Matosin N, Turecki G, Nagy C, Mechawar N. Spatial transcriptomic analysis of adult hippocampal neurogenesis in the human brain. J Psychiatry Neurosci 2024; 49:E319-E333. [PMID: 39414359 PMCID: PMC11495544 DOI: 10.1503/jpn.240026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 06/21/2024] [Accepted: 08/18/2024] [Indexed: 10/18/2024] Open
Abstract
BACKGROUND Adult hippocampal neurogenesis has been extensively characterized in rodent models, but its existence in humans remains controversial. We sought to assess the phenomenon in postmortem human hippocampal samples by combining spatial transcriptomics and multiplexed fluorescent in situ hybridization. METHODS We computationally examined the spatial expression of various canonical neurogenesis markers in postmortem dentate gyrus (DG) sections from young and middle-aged sudden-death males. We conducted in situ assessment of markers expressed in neural stem cells, proliferative cells, and immature granule neurons in postmortem DG sections from infant, adolescent, and middle-aged males. RESULTS We examined frozen DG tissue from infant (n = 1, age 2 yr), adolescent (n = 1, age 16 yr), young adult (n = 2, mean age 23.5 yr), and middle-aged (n = 2, mean age 42.5 yr) males, and frozen-fixed DG tissue from middle-aged males (n = 6, mean age 43.5 yr). We detected very few cells expressing neural stem cell and proliferative markers in the human DG from childhood to middle age. However, at all ages, we observed a substantial number of DG cells expressing the immature neuronal marker DCX. Most DCX + cells displayed an inhibitory phenotype, while the remainder were non-committed or excitatory in nature. LIMITATIONS The study was limited by small sample sizes and included samples only from males. CONCLUSION Our findings indicate very low levels of hippocampal neurogenesis throughout life and the existence of a local reserve of plasticity in the adult human hippocampus. Overall, our study provides important insight into the distribution and phenotype of cells expressing neurogenesis markers in the adult human hippocampus.
Collapse
Affiliation(s)
- Sophie Simard
- From the McGill Group for Suicide Studies, Douglas Mental Health University Institute, McGill University, Montréal, Que. (Simard, Rahimian, Antonietta, Théberge, Turecki, Nagy, Mechawar); the School of Medical Sciences, Faculty of Medicine and Health, Charles Perkins Centre, The University of Sydney, Camperdown, Australia (Matosin); the Department of Psychiatry, McGill University, Montréal, Que. (Turecki, Nagy, Mechawar)
| | - Reza Rahimian
- From the McGill Group for Suicide Studies, Douglas Mental Health University Institute, McGill University, Montréal, Que. (Simard, Rahimian, Antonietta, Théberge, Turecki, Nagy, Mechawar); the School of Medical Sciences, Faculty of Medicine and Health, Charles Perkins Centre, The University of Sydney, Camperdown, Australia (Matosin); the Department of Psychiatry, McGill University, Montréal, Que. (Turecki, Nagy, Mechawar)
| | - Maria Antonietta Davoli
- From the McGill Group for Suicide Studies, Douglas Mental Health University Institute, McGill University, Montréal, Que. (Simard, Rahimian, Antonietta, Théberge, Turecki, Nagy, Mechawar); the School of Medical Sciences, Faculty of Medicine and Health, Charles Perkins Centre, The University of Sydney, Camperdown, Australia (Matosin); the Department of Psychiatry, McGill University, Montréal, Que. (Turecki, Nagy, Mechawar)
| | - Stéphanie Théberge
- From the McGill Group for Suicide Studies, Douglas Mental Health University Institute, McGill University, Montréal, Que. (Simard, Rahimian, Antonietta, Théberge, Turecki, Nagy, Mechawar); the School of Medical Sciences, Faculty of Medicine and Health, Charles Perkins Centre, The University of Sydney, Camperdown, Australia (Matosin); the Department of Psychiatry, McGill University, Montréal, Que. (Turecki, Nagy, Mechawar)
| | - Natalie Matosin
- From the McGill Group for Suicide Studies, Douglas Mental Health University Institute, McGill University, Montréal, Que. (Simard, Rahimian, Antonietta, Théberge, Turecki, Nagy, Mechawar); the School of Medical Sciences, Faculty of Medicine and Health, Charles Perkins Centre, The University of Sydney, Camperdown, Australia (Matosin); the Department of Psychiatry, McGill University, Montréal, Que. (Turecki, Nagy, Mechawar)
| | - Gustavo Turecki
- From the McGill Group for Suicide Studies, Douglas Mental Health University Institute, McGill University, Montréal, Que. (Simard, Rahimian, Antonietta, Théberge, Turecki, Nagy, Mechawar); the School of Medical Sciences, Faculty of Medicine and Health, Charles Perkins Centre, The University of Sydney, Camperdown, Australia (Matosin); the Department of Psychiatry, McGill University, Montréal, Que. (Turecki, Nagy, Mechawar)
| | - Corina Nagy
- From the McGill Group for Suicide Studies, Douglas Mental Health University Institute, McGill University, Montréal, Que. (Simard, Rahimian, Antonietta, Théberge, Turecki, Nagy, Mechawar); the School of Medical Sciences, Faculty of Medicine and Health, Charles Perkins Centre, The University of Sydney, Camperdown, Australia (Matosin); the Department of Psychiatry, McGill University, Montréal, Que. (Turecki, Nagy, Mechawar)
| | - Naguib Mechawar
- From the McGill Group for Suicide Studies, Douglas Mental Health University Institute, McGill University, Montréal, Que. (Simard, Rahimian, Antonietta, Théberge, Turecki, Nagy, Mechawar); the School of Medical Sciences, Faculty of Medicine and Health, Charles Perkins Centre, The University of Sydney, Camperdown, Australia (Matosin); the Department of Psychiatry, McGill University, Montréal, Que. (Turecki, Nagy, Mechawar).
| |
Collapse
|
6
|
Melgar-Locatelli S, Mañas-Padilla MC, Castro-Zavala A, Rivera P, Del Carmen Razola-Díaz M, Monje FJ, Rodríguez-Pérez C, Castilla-Ortega E. Diet enriched with high-phenolic cocoa potentiates hippocampal brain-derived neurotrophic factor expression and neurogenesis in healthy adult micewith subtle effects on memory. Food Funct 2024; 15:8310-8329. [PMID: 39069830 DOI: 10.1039/d4fo01201a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/30/2024]
Abstract
Cocoa is widely known for its health benefits, but its neurocognitive impact remains underexplored. This preclinical study aimed to investigate the effects of cocoa and cocoa polyphenols on hippocampal neuroplasticity, cognitive function and emotional behavior. Seventy young-adult C57BL/6JRj male and female mice were fed either a standard diet (CTR) or a diet enriched with 10% high-phenolic content cocoa (HPC) or low-phenolic content cocoa (LPC) for at least four weeks. In a first experiment, behavioral tests assessing exploratory behavior, emotional responses and hippocampal-dependent memory were conducted four weeks into the diet, followed by animal sacrifice a week later. Adult hippocampal neurogenesis and brain-derived neurotrophic factor (BDNF) expression in the hippocampus and prefrontal cortex were evaluated using immunohistochemistry and western blot. In a different experiment, hippocampal synaptic response, long-term potentiation and presynaptic-dependent short-term plasticity were studied by electrophysiology. Cocoa-enriched diets had minimal effects on exploratory activity and anxiety-like behavior, except for reduced locomotion in the LPC group. Only the HPC diet enhanced object recognition memory, while place recognition memory and spatial navigation remained unaffected. The HPC diet also increased adult hippocampal neurogenesis, boosting the proliferation, survival and number of young adult-born neurons. However, both cocoa-enriched diets increased immobility in the forced swimming test and hippocampal BDNF expression. Hippocampal electrophysiology revealed no alterations in neuroplasticity among diets. The results were mostly unaffected by sex. Overall, the HPC diet demonstrated greater potential regarding cognitive and neuroplastic benefits, suggesting a key role of cocoa flavanols in dietary interventions aimed at enhancing brain health.
Collapse
Affiliation(s)
- Sonia Melgar-Locatelli
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, Spain
- Departamento de Psicobiología y Metodología de las Ciencias del Comportamiento, Universidad de Málaga, Spain
- Departamento de Nutrición y Bromatología, Universidad de Granada, Campus Universitario de Cartuja, Spain
| | - M Carmen Mañas-Padilla
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, Spain
- Departamento de Psicobiología y Metodología de las Ciencias del Comportamiento, Universidad de Málaga, Spain
- Universidad Internacional de la Rioja (UNIR), Spain
| | - Adriana Castro-Zavala
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, Spain
- Departamento de Psicobiología y Metodología de las Ciencias del Comportamiento, Universidad de Málaga, Spain
| | - Patricia Rivera
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, Spain
- Unidad de Gestión Clínica de Salud Mental, Hospital Regional Universitario de Málaga, Spain
| | - María Del Carmen Razola-Díaz
- Departamento de Nutrición y Bromatología, Universidad de Granada, Campus Universitario de Cartuja, Spain
- Instituto de Nutrición y Tecnología de los Alimentos 'José Mataix' (INYTA), Universidad de Granada, Granada, Spain
| | - Francisco J Monje
- Center for Physiology and Pharmacology, Department of Neurophysiology and Neuropharma-cology, Medical University of Vienna, 1090 Vienna, Austria
| | - Celia Rodríguez-Pérez
- Departamento de Nutrición y Bromatología, Universidad de Granada, Campus Universitario de Cartuja, Spain
- Instituto de Nutrición y Tecnología de los Alimentos 'José Mataix' (INYTA), Universidad de Granada, Granada, Spain
- Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), 18012, Granada, Spain
| | - Estela Castilla-Ortega
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, Spain
- Departamento de Psicobiología y Metodología de las Ciencias del Comportamiento, Universidad de Málaga, Spain
| |
Collapse
|
7
|
Dejou J, Mandairon N, Didier A. Olfactory neurogenesis plays different parts at successive stages of life, implications for mental health. Front Neural Circuits 2024; 18:1467203. [PMID: 39175668 PMCID: PMC11338910 DOI: 10.3389/fncir.2024.1467203] [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: 07/19/2024] [Accepted: 07/31/2024] [Indexed: 08/24/2024] Open
Abstract
The olfactory bulb is a unique site of continuous neurogenesis, primarily generating inhibitory interneurons, a process that begins at birth and extends through infancy and adulthood. This review examines the characteristics of olfactory bulb neurogenesis, focusing on granule cells, the most numerous interneurons, and how their age and maturation affect their function. Adult-born granule cells, while immature, contribute to the experience-dependent plasticity of the olfactory circuit by enabling structural and functional synaptic changes. In contrast, granule cells born early in life form the foundational elements of the olfactory bulb circuit, potentially facilitating innate olfactory information processing. The implications of these neonatal cells on early life olfactory memory and their impact on adult perception, particularly in response to aversive events and susceptibility to emotional disorders, warrant further investigation.
Collapse
Affiliation(s)
- Jules Dejou
- INSERM, U1028; CNRS, UMR5292; Lyon Neuroscience Research Center, Neuroplasticity and Neuropathology of Olfactory Perception Team, Lyon, France
| | - Nathalie Mandairon
- INSERM, U1028; CNRS, UMR5292; Lyon Neuroscience Research Center, Neuroplasticity and Neuropathology of Olfactory Perception Team, Lyon, France
| | - Anne Didier
- INSERM, U1028; CNRS, UMR5292; Lyon Neuroscience Research Center, Neuroplasticity and Neuropathology of Olfactory Perception Team, Lyon, France
- Institut Universitaire de France, Paris, France
| |
Collapse
|
8
|
Alonso M, Petit AC, Lledo PM. The impact of adult neurogenesis on affective functions: of mice and men. Mol Psychiatry 2024; 29:2527-2542. [PMID: 38499657 DOI: 10.1038/s41380-024-02504-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 02/22/2024] [Accepted: 02/27/2024] [Indexed: 03/20/2024]
Abstract
In most mammals, new neurons are not only produced during embryogenesis but also after birth. Soon after adult neurogenesis was discovered, the influence of recruiting new neurons on cognitive functions, especially on memory, was documented. Likewise, the late process of neuronal production also contributes to affective functions, but this outcome was recognized with more difficulty. This review covers hypes and hopes of discovering the influence of newly-generated neurons on brain circuits devoted to affective functions. If the possibility of integrating new neurons into the adult brain is a commonly accepted faculty in the realm of mammals, the reluctance is strong when it comes to translating this concept to humans. Compiling data suggest now that new neurons are derived not only from stem cells, but also from a population of neuroblasts displaying a protracted maturation and ready to be engaged in adult brain circuits, under specific signals. Here, we discuss the significance of recruiting new neurons in the adult brain circuits, specifically in the context of affective outcomes. We also discuss the fact that adult neurogenesis could be the ultimate cellular process that integrates elements from both the internal and external environment to adjust brain functions. While we must be critical and beware of the unreal promises that Science could generate sometimes, it is important to continue exploring the potential of neural recruitment in adult primates. Reporting adult neurogenesis in humankind contributes to a new vision of humans as mammals whose brain continues to develop throughout life. This peculiar faculty could one day become the target of treatment for mental health, cognitive disorders, and elderly-associated diseases. The vision of an adult brain which never stops integrating new neurons is a real game changer for designing new therapeutic interventions to treat mental disorders associated with substantial morbidity, mortality, and social costs.
Collapse
Affiliation(s)
- Mariana Alonso
- Institut Pasteur, Université Paris Cité, CNRS UMR 3571, Perception and Action Unit, F-75015, Paris, France
| | - Anne-Cécile Petit
- Institut Pasteur, Université Paris Cité, CNRS UMR 3571, Perception and Action Unit, F-75015, Paris, France
- Pôle Hospitalo-Universitaire Psychiatrie Paris 15, GHU Paris Psychiatry and Neurosciences, Hôpital Sainte-Anne, Paris, France
| | - Pierre-Marie Lledo
- Institut Pasteur, Université Paris Cité, CNRS UMR 3571, Perception and Action Unit, F-75015, Paris, France.
| |
Collapse
|
9
|
Kopsidas CA, Lowe CC, McDaniel DP, Zhou X, Feng Y. Sustained generation of neurons destined for neocortex with oxidative metabolic upregulation upon filamin abrogation. iScience 2024; 27:110199. [PMID: 38989458 PMCID: PMC11233971 DOI: 10.1016/j.isci.2024.110199] [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: 09/12/2023] [Revised: 04/01/2024] [Accepted: 06/03/2024] [Indexed: 07/12/2024] Open
Abstract
Neurons in the neocortex are generated during embryonic development. While the adult ventricular-subventricular zone (V-SVZ) contains cells with neural stem/progenitors' characteristics, it remains unclear whether it has the capacity of producing neocortical neurons. Here, we show that generating neurons with transcriptomic resemblance to upper layer neocortical neurons continues in the V-SVZ of mouse models of a human condition known as periventricular heterotopia by abrogating Flna and Flnb. We found such surplus neurogenesis was associated with V-SVZ's upregulation of oxidative phosphorylation, mitochondrial biogenesis, and vascular abundance. Additionally, spatial transcriptomics analyses showed V-SVZ's neurogenic activation was coupled with transcriptional enrichment of genes in diverse pathways for energy metabolism, angiogenesis, cell signaling, synaptic transmission, and turnovers of nucleic acids and proteins in upper cortical layers. These findings support the potential of generating neocortical neurons in adulthood through boosting brain-wide vascular circulation, aerobic adenosine triphosphate synthesis, metabolic turnover, and neuronal activity.
Collapse
Affiliation(s)
- Caroline A. Kopsidas
- Department of Biochemistry and Molecular Biology, Uniformed Services University, 4301 Jones Bridge Road, Bethesda, MD 20814, USA
| | - Clara C. Lowe
- Department of Biochemistry and Molecular Biology, Uniformed Services University, 4301 Jones Bridge Road, Bethesda, MD 20814, USA
| | - Dennis P. McDaniel
- Biomedical Instrumentation Center, Uniformed Services University, 4301 Jones Bridge Road, Bethesda, MD 20814, USA
| | - Xiaoming Zhou
- Department of Medicine, Uniformed Services University, 4301 Jones Bridge Road, Bethesda, MD 20814, USA
| | - Yuanyi Feng
- Department of Biochemistry and Molecular Biology, Uniformed Services University, 4301 Jones Bridge Road, Bethesda, MD 20814, USA
| |
Collapse
|
10
|
Luo Y, Wang Y, Qiu F, Hou G, Liu J, Yang H, Wu M, Dong X, Guo D, Zhong Z, Zhang X, Ge J, Meng P. Ablated Sonic Hedgehog Signaling in the Dentate Gyrus of the Dorsal and Ventral Hippocampus Impairs Hippocampal-Dependent Memory Tasks and Emotion in a Rat Model of Depression. Mol Neurobiol 2024; 61:4352-4368. [PMID: 38087166 DOI: 10.1007/s12035-023-03796-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Accepted: 11/08/2023] [Indexed: 07/11/2024]
Abstract
Specific memory processes and emotional aberrations in depression can be attributed to the different dorsal-ventral regions of the hippocampus. However, the molecular mechanisms underlying the differential functions of the dorsal hippocampus (dHip) and ventral hippocampus (vHip) remain unclear. As Sonic Hedgehog (Shh) is involved in the dorsal-ventral patterning of the neural tube and its signaling is dysregulated by chronic unpredictable mild stress (CUMS), we investigated its role in influencing the differential functions of the dHip and vHip. Here, CUMS downregulated the expression of Shh signaling markers, including Shh and its downstream effectors GLI family zinc finger 12 (Gli1/2), Patched (Ptch), and smoothened (Smo), in both the dHip and vHip of rats, though more so in the vHip. Additionally, Shh knockdown in the dorsal or ventral dentate gyrus (DG) resulted in restrained neurogenic activity in newborn neurons, especially in immature neurons through decreased expression of Shh signaling markers. Furthermore, Shh knockdown in the DG of the dHip led to memory impairment by inhibiting experience-dependent activation of immature neurons, whereas its knockdown in the DG of the vHip led to an emotional handicap by delaying the maturation of immature neurons. Finally, Shh knockdown in either the dDG or vDG of hippocampus abolished the corresponding cognitive enhancement and emotional recovery of fluoxetine. In conclusion, Shh is essential to maintain the functional heterogeneity of dHip and vHip in depressed rat, which was mainly mediating by local changes of dependent activation and maturity of immature neurons, respectively.
Collapse
Affiliation(s)
- Yan Luo
- Hunan University of Chinese Medicine, Changsha, 410208, Hunan, China
| | - Yan Wang
- Yiyang Central Hospital, Yiyang, 413000, Hunan, China
| | - Feng Qiu
- Hunan University of Chinese Medicine, Changsha, 410208, Hunan, China
| | - Guanghan Hou
- Fourth Hospital of Changsha, Hunan, 410000, China
| | - Jian Liu
- Hunan University of Chinese Medicine, Changsha, 410208, Hunan, China
- First Affiliated Hospital, Hunan University of Chinese Medicine, Changsha, 410007, Hunan, China
| | - Hui Yang
- Hunan University of Chinese Medicine, Changsha, 410208, Hunan, China
- First Affiliated Hospital, Hunan University of Chinese Medicine, Changsha, 410007, Hunan, China
| | - Mei Wu
- Hunan University of Chinese Medicine, Changsha, 410208, Hunan, China
| | - Xuanqi Dong
- The Second People's Hospital of Hunan Province, Changsha, 410000, Hunan, China
| | - Dongwei Guo
- The Second People's Hospital of Hunan Province, Changsha, 410000, Hunan, China
| | - Ziyan Zhong
- Hunan University of Chinese Medicine, Changsha, 410208, Hunan, China
| | - Xi Zhang
- The Second People's Hospital of Hunan Province, Changsha, 410000, Hunan, China.
| | - Jinwen Ge
- Hunan Academy of Chinese Medicine, Changsha, 410300, Hunan, China.
| | - Pan Meng
- Hunan University of Chinese Medicine, Changsha, 410208, Hunan, China.
| |
Collapse
|
11
|
Simard S, Matosin N, Mechawar N. Adult Hippocampal Neurogenesis in the Human Brain: Updates, Challenges, and Perspectives. Neuroscientist 2024:10738584241252581. [PMID: 38757781 DOI: 10.1177/10738584241252581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/18/2024]
Abstract
The existence of neurogenesis in the adult human hippocampus has been under considerable debate within the past three decades due to the diverging conclusions originating mostly from immunohistochemistry studies. While some of these reports conclude that hippocampal neurogenesis in humans occurs throughout physiologic aging, others indicate that this phenomenon ends by early childhood. More recently, some groups have adopted next-generation sequencing technologies to characterize with more acuity the extent of this phenomenon in humans. Here, we review the current state of research on adult hippocampal neurogenesis in the human brain with an emphasis on the challenges and limitations of using immunohistochemistry and next-generation sequencing technologies for its study.
Collapse
Affiliation(s)
- Sophie Simard
- McGill Group for Suicide Studies, Douglas Mental Health University Institute, McGill University, Montréal, Canada
| | - Natalie Matosin
- School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Sydney, Australia
| | - Naguib Mechawar
- McGill Group for Suicide Studies, Douglas Mental Health University Institute, McGill University, Montréal, Canada
- Department of Psychiatry, McGill University, Montréal, Canada
| |
Collapse
|
12
|
Xie XH, Xu SX, Yao L, Chen MM, Zhang H, Wang C, Nagy C, Liu Z. Altered in vivo early neurogenesis traits in patients with depression: Evidence from neuron-derived extracellular vesicles and electroconvulsive therapy. Brain Stimul 2024; 17:19-28. [PMID: 38101468 DOI: 10.1016/j.brs.2023.12.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 11/15/2023] [Accepted: 12/08/2023] [Indexed: 12/17/2023] Open
Abstract
BACKGROUND The neurogenesis hypothesis is a promising candidate etiologic hypothesis for depression, and it is associated with electroconvulsive therapy (ECT). However, human in vivo molecular-level evidence is lacking. OBJECTIVE We used neuron-derived extracellular vesicles (NDEVs) as a "window to the neurons" to explore the in vivo neurogenesis status associated with ECT in patients with treatment-resistant depression (TRD). METHODS In this study, we enrolled 40 patients with TRD and 35 healthy controls (HCs). We isolated NDEVs from the plasma of each participant to test the levels of doublecortin (DCX), a marker of neurogenesis, and cluster of differentiation (CD) 81, a marker of EVs. We also assessed the plasma levels of brain-derived neurotrophic factor (BDNF), a protein that is known to be associated with ECT and neuroplastic processes. RESULTS Our findings indicated that both the levels of DCX in NDEVs and BDNF in plasma were significantly lower in TRD patients compared to HCs at baseline, but increased following ECTs. Conversely, levels of CD81 in NDEVs were found higher in TRD patients at baseline, but did not change after the ECT treatments. Exploratory analyses revealed that lower levels of BDNF in plasma and DCX in NDEVs, along with higher CD81 levels in NDEVs, were associated with more severe depressive symptoms and reduced cognitive function at baseline. Furthermore, higher baseline CD81 concentrations in NDEVs were correlated with greater decreases in depression symptoms. CONCLUSIONS We first present human in vivo evidence of early neurogenesis using DCX through NDEVs: decreased in TRD patients, increased after ECTs.
Collapse
Affiliation(s)
- Xin-Hui Xie
- Department of Psychiatry, Renmin Hospital of Wuhan University, Wuhan, Hubei, PR China
| | - Shu-Xian Xu
- Department of Psychiatry, Renmin Hospital of Wuhan University, Wuhan, Hubei, PR China
| | - Lihua Yao
- Department of Psychiatry, Renmin Hospital of Wuhan University, Wuhan, Hubei, PR China
| | - Mian-Mian Chen
- Department of Psychiatry, Renmin Hospital of Wuhan University, Wuhan, Hubei, PR China
| | - Honghan Zhang
- Department of Psychiatry, Renmin Hospital of Wuhan University, Wuhan, Hubei, PR China
| | - Chao Wang
- Department of Psychiatry, Renmin Hospital of Wuhan University, Wuhan, Hubei, PR China
| | - Corina Nagy
- Department of Psychiatry, McGill University, Montreal, QC, Canada; McGill Group for Suicide Studies, Douglas Mental Health University Institute, McGill University, Montreal, QC, Canada
| | - Zhongchun Liu
- Department of Psychiatry, Renmin Hospital of Wuhan University, Wuhan, Hubei, PR China; Taikang center for life and medical sciences, Wuhan University, Wuhan, PR China.
| |
Collapse
|
13
|
Gubert C, Hannan AJ. Microbial manipulation of memories and minds. Brain 2023; 146:4791-4793. [PMID: 37873966 DOI: 10.1093/brain/awad368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Accepted: 10/19/2023] [Indexed: 10/25/2023] Open
Abstract
This scientific commentary refers to ‘Microbiota from Alzheimer’s patients induce deficits in cognition and hippocampal neurogenesis’ by Grabrucker et al. (https://doi.org/10.1093/brain/awad303).
Collapse
Affiliation(s)
- Carolina Gubert
- Florey Institute of Neuroscience and Mental Health, Melbourne Brain Centre, University of Melbourne, Parkville, Victoria, Australia
| | - Anthony J Hannan
- Florey Institute of Neuroscience and Mental Health, Melbourne Brain Centre, University of Melbourne, Parkville, Victoria, Australia
- Department of Anatomy and Physiology, University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
14
|
Stone J, Mitrofanis J, Johnstone DM, Robinson SR. Twelve protections evolved for the brain, and their roles in extending its functional life. Front Neuroanat 2023; 17:1280275. [PMID: 38020212 PMCID: PMC10657866 DOI: 10.3389/fnana.2023.1280275] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2023] [Accepted: 10/20/2023] [Indexed: 12/01/2023] Open
Abstract
As human longevity has increased, we have come to understand the ability of the brain to function into advanced age, but also its vulnerability with age, apparent in the age-related dementias. Against that background of success and vulnerability, this essay reviews how the brain is protected by (by our count) 12 mechanisms, including: the cranium, a bony helmet; the hydraulic support given by the cerebrospinal fluid; the strategically located carotid body and sinus, which provide input to reflexes that protect the brain from blood-gas imbalance and extremes of blood pressure; the blood brain barrier, an essential sealing of cerebral vessels; the secretion of molecules such as haemopexin and (we argue) the peptide Aβ to detoxify haemoglobin, at sites of a bleed; autoregulation of the capillary bed, which stabilises metabolites in extracellular fluid; fuel storage in the brain, as glycogen; oxygen storage, in the haemoprotein neuroglobin; the generation of new neurones, in the adult, to replace cells lost; acquired resilience, the stress-induced strengthening of cell membranes and energy production found in all body tissues; and cognitive reserve, the ability of the brain to maintain function despite damage. Of these 12 protections, we identify 5 as unique to the brain, 3 as protections shared with all body tissues, and another 4 as protections shared with other tissues but specialised for the brain. These protections are a measure of the brain's vulnerability, of its need for protection. They have evolved, we argue, to maintain cognitive function, the ability of the brain to function despite damage that accumulates during life. Several can be tools in the hands of the individual, and of the medical health professional, for the lifelong care of our brains.
Collapse
Affiliation(s)
- Jonathan Stone
- Faculty of Medicine and Health, University of Sydney, Camperdown, NSW, Australia
| | - John Mitrofanis
- Grenoble and Institute of Ophthalmology, Fonds de Dotation Clinatec, Université Grenoble Alpes, University College London, London, United Kingdom
| | - Daniel M. Johnstone
- School of Biomedical Sciences and Pharmacy, University of Newcastle and School of Medical Sciences, The University of Sydney, Camperdown, NSW, Australia
| | - Stephen R. Robinson
- School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC, Australia
- Institute for Breathing and Sleep, Austin Health, Heidelberg, VIC, Australia
| |
Collapse
|
15
|
Zhang R, Quan H, Wang Y, Luo F. Neurogenesis in primates versus rodents and the value of non-human primate models. Natl Sci Rev 2023; 10:nwad248. [PMID: 38025664 PMCID: PMC10659238 DOI: 10.1093/nsr/nwad248] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 08/21/2023] [Accepted: 09/10/2023] [Indexed: 12/01/2023] Open
Abstract
Neurogenesis, the process of generating neurons from neural stem cells, occurs during both embryonic and adult stages, with each stage possessing distinct characteristics. Dysfunction in either stage can disrupt normal neural development, impair cognitive functions, and lead to various neurological disorders. Recent technological advancements in single-cell multiomics and gene-editing have facilitated investigations into primate neurogenesis. Here, we provide a comprehensive overview of neurogenesis across rodents, non-human primates, and humans, covering embryonic development to adulthood and focusing on the conservation and diversity among species. While non-human primates, especially monkeys, serve as valuable models with closer neural resemblance to humans, we highlight the potential impacts and limitations of non-human primate models on both physiological and pathological neurogenesis research.
Collapse
Affiliation(s)
- Runrui Zhang
- State Key Laboratory of Primate Biomedical Research; Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming 650500, China
- Yunnan Key Laboratory of Primate Biomedical Research, Kunming 650500, China
| | - Hongxin Quan
- State Key Laboratory of Primate Biomedical Research; Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming 650500, China
- Yunnan Key Laboratory of Primate Biomedical Research, Kunming 650500, China
| | - Yinfeng Wang
- State Key Laboratory of Primate Biomedical Research; Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming 650500, China
- Yunnan Key Laboratory of Primate Biomedical Research, Kunming 650500, China
| | - Fucheng Luo
- State Key Laboratory of Primate Biomedical Research; Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming 650500, China
- Yunnan Key Laboratory of Primate Biomedical Research, Kunming 650500, China
| |
Collapse
|
16
|
Wang Q, Tang B, Hao S, Wu Z, Yang T, Tang J. Forniceal deep brain stimulation in a mouse model of Rett syndrome increases neurogenesis and hippocampal memory beyond the treatment period. Brain Stimul 2023; 16:1401-1411. [PMID: 37704033 PMCID: PMC11152200 DOI: 10.1016/j.brs.2023.09.002] [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: 04/18/2023] [Revised: 08/30/2023] [Accepted: 09/02/2023] [Indexed: 09/15/2023] Open
Abstract
BACKGROUND Rett syndrome (RTT), caused by mutations in the X-linked gene encoding methyl-CpG binding protein 2 (MeCP2), severely impairs learning and memory. We previously showed that forniceal deep brain stimulation (DBS) stimulates hippocampal neurogenesis with concomitant improvements in hippocampal-dependent learning and memory in a mouse model of RTT. OBJECTIVES To determine the duration of DBS benefits; characterize DBS effects on hippocampal neurogenesis; and determine whether DBS influences MECP2 genotype and survival of newborn dentate granular cells (DGCs) in RTT mice. METHODS Chronic DBS was delivered through an electrode implanted in the fimbria-fornix. We tested separate cohorts of mice in contextual and cued fear memory at different time points after DBS. We then examined neurogenesis, DGC apoptosis, and the expression of brain-derived neurotrophic factor (BDNF) and vascular endothelial growth factor (VEGF) after DBS by immunohistochemistry. RESULTS After two weeks of forniceal DBS, memory improvements lasted between 6 and 9 weeks. Repeating DBS every 6 weeks was sufficient to maintain the improvement. Forniceal DBS stimulated the birth of more MeCP2-positive than MeCP2-negative DGCs and had no effect on DGC survival. It also increased the expression of BDNF but not VEGF in the RTT mouse dentate gyrus. CONCLUSION Improvements in learning and memory from forniceal DBS in RTT mice extends well beyond the treatment period and can be maintained by repeated DBS. Stimulation of BDNF expression correlates with improvements in hippocampal neurogenesis and memory benefits.
Collapse
Affiliation(s)
- Qi Wang
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, 77030, USA; Department of Pediatrics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Bin Tang
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, 77030, USA; Department of Pediatrics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Shuang Hao
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, 77030, USA; Department of Pediatrics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Zhenyu Wu
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, 77030, USA; Department of Pediatrics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Tingting Yang
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, 77030, USA; Department of Pediatrics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Jianrong Tang
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, 77030, USA; Department of Pediatrics, Baylor College of Medicine, Houston, TX, 77030, USA.
| |
Collapse
|
17
|
Wolniczak E, Meyer F, Albrecht A. [The abdominal brain: neuroanatomic perspectives for the abdominal surgeon]. ZEITSCHRIFT FUR GASTROENTEROLOGIE 2023; 61:1037-1045. [PMID: 37142237 DOI: 10.1055/a-2013-7633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/06/2023]
Abstract
The "abdominal brain" does not only consist of a separate enteric nervous system but also of bidirectional connections to the autonomous nerve system with parasympathicus und sympathicus as well as brain and spinal cord. Novel studies have shown that these connections can quickly transfer information on the ingested nutrients to the brain to conduct the feeling of hunger and more complex behaviour, such as "reward-related learning". However, even emotional experience, in particular, stress, has a strong impact onto the gastrointestinal system. The immune system, motility and barrier function of the gastrointestinal tract are modulated by the intestinal microbiota. Local bacteria may directly influence neuronal communication by released metabolic products and neuropeptides as well as may control inflammatory factors. Intensive research over the last 10 years was able to provide evidence that intestinal microbiota may affect emotional and cognitive aspects of our behaviour and, thus, it might be in the focus of numerous neuropsychiatric diseases, such as depressions and anxiety disorders.The presented review is to provide a short summary of the I): anatomic basics of the so-called gut-brain axis and II): modi of the bidirectional regulation. Through indirect connections to the limbic system, gut-brain axis can substantially influence stress and anxiety but also the pain processing. In addition, the role of microbiota is outlined and future paths are shown, e.g., how the (microbiota-)gut-brain axis may alter emotional experience, pain processing and intestinal function. Such associations are relevant for further development of visceral medicine, and, thus, also for the abdominal surgeon to derive future treatment concepts with interdisciplinary orientation.
Collapse
Affiliation(s)
- Erik Wolniczak
- Institut für Anatomie, Otto-von-Guericke-Universität zu Magdeburg, Magdeburg, Deutschland
| | - Frank Meyer
- Klinik für Allgemein-, Viszeral-, Gefäß- und Transplantationschirurgie, Universitätsklinikum Magdeburg A.ö.R., Magdeburg, Deutschland
| | - Anne Albrecht
- Institut für Anatomie, Otto-von-Guericke-Universität zu Magdeburg, Magdeburg, Deutschland
- Center for Behavioral Brain Science (CBBS), Magdeburg, Germany
- Center for Intervention and Research on adaptive and maladaptive brain Circuits underlying mental health (C-I-R-C), Jena-Magdeburg-Halle, Germany
| |
Collapse
|
18
|
Ghibaudi M, Marchetti N, Vergnano E, La Rosa C, Benedetti B, Couillard-Despres S, Farioli-Vecchioli S, Bonfanti L. Age-related changes in layer II immature neurons of the murine piriform cortex. Front Cell Neurosci 2023; 17:1205173. [PMID: 37576566 PMCID: PMC10416627 DOI: 10.3389/fncel.2023.1205173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Accepted: 07/14/2023] [Indexed: 08/15/2023] Open
Abstract
The recent identification of a population of non-newly born, prenatally generated "immature" neurons in the layer II of the piriform cortex (cortical immature neurons, cINs), raises questions concerning their maintenance or depletion through the lifespan. Most forms of brain structural plasticity progressively decline with age, a feature that is particularly prominent in adult neurogenesis, due to stem cell depletion. By contrast, the entire population of the cINs is produced during embryogenesis. Then these cells simply retain immaturity in postnatal and adult stages, until they "awake" to complete their maturation and ultimately integrate into neural circuits. Hence, the question remains open whether the cINs, which are not dependent on stem cell division, might follow a similar pattern of age-related reduction, or in alternative, might leave a reservoir of young, undifferentiated cells in the adult and aging brain. Here, the number and features of cINs were analyzed in the mouse piriform cortex from postnatal to advanced ages, by using immunocytochemistry for the cytoskeletal marker doublecortin. The abundance and stage of maturation of cINs, along with the expression of other markers of maturity/immaturity were investigated. Despite a marked decrease in this neuronal population during juvenile stages, reminiscent of that observed in hippocampal neurogenesis, a small amount of highly immature cINs persisted up to advanced ages. Overall, albeit reducing in number with increasing age, we report that the cINs are present through the entire animal lifespan.
Collapse
Affiliation(s)
- Marco Ghibaudi
- Neuroscience Institute Cavalieri Ottolenghi (NICO), Orbassano, Italy
- Department of Veterinary Sciences, University of Turin, Turin, Italy
| | - Nicole Marchetti
- Institute of Biochemistry and Cell Biology, National Research Council, Rome, Italy
| | - Elena Vergnano
- Neuroscience Institute Cavalieri Ottolenghi (NICO), Orbassano, Italy
| | - Chiara La Rosa
- Neuroscience Institute Cavalieri Ottolenghi (NICO), Orbassano, Italy
| | - Bruno Benedetti
- Institute of Experimental Neuroregeneration, Paracelsus Medical University, Salzburg, Austria
- Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Salzburg, Austria
- Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Sebastien Couillard-Despres
- Institute of Experimental Neuroregeneration, Paracelsus Medical University, Salzburg, Austria
- Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Salzburg, Austria
- Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | | | - Luca Bonfanti
- Neuroscience Institute Cavalieri Ottolenghi (NICO), Orbassano, Italy
- Department of Veterinary Sciences, University of Turin, Turin, Italy
| |
Collapse
|
19
|
Blasco-Chamarro L, Fariñas I. Fine-tuned rest: unveiling the regulatory landscape of adult quiescent neural stem cells. Neuroscience 2023:S0306-4522(23)00298-1. [PMID: 37437796 DOI: 10.1016/j.neuroscience.2023.07.003] [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: 06/09/2023] [Revised: 06/29/2023] [Accepted: 07/03/2023] [Indexed: 07/14/2023]
Abstract
Cell quiescence is an essential mechanism that allows cells to temporarily halt proliferation while preserving the potential to resume it at a later time. The molecular mechanisms underlying cell quiescence are complex and involve the regulation of various signaling pathways, transcription factors and epigenetic modifications. The importance of unveiling the mechanisms regulating the quiescent state is undeniable, as its long-term maintenance is key to sustain tissue homeostasis throughout life. Neural stem cells (NSCs) are maintained in the subependymal zone (SEZ) niche of adult mammalian brains mostly as long-lasting quiescent cells, owing to multiple intrinsic and extrinsic cues that actively regulate this state. Differently from other non-proliferative states, quiescence is a reversible and tightly regulated condition that can re-activate to support the formation of new neurons throughout adult lifespan. Decoding its regulatory mechanisms in homeostasis and unveiling how it is modulated in the context of the aged brain or during tumorigenesis, could bring us closer to the development of new potential strategies to intervene in adult neurogenesis with therapeutic purposes. Starting with a general conceptualization of the quiescent state in different stem cell niches, we here review what we have learned about NSC quiescence in the SEZ, encompassing the experimental strategies used for its study, to end up discussing the modulation of quiescence in the context of a physiology or pathological NSC dysregulation.
Collapse
Affiliation(s)
- Laura Blasco-Chamarro
- Biomedical Research Network on Neurodegenerative Diseases (CIBERNED); Department of Cell Biology; Biotechnology and Biomedicine Institute (BioTecMed), University of Valencia, Spain
| | - Isabel Fariñas
- Biomedical Research Network on Neurodegenerative Diseases (CIBERNED); Department of Cell Biology; Biotechnology and Biomedicine Institute (BioTecMed), University of Valencia, Spain.
| |
Collapse
|
20
|
Tosoni G, Ayyildiz D, Bryois J, Macnair W, Fitzsimons CP, Lucassen PJ, Salta E. Mapping human adult hippocampal neurogenesis with single-cell transcriptomics: Reconciling controversy or fueling the debate? Neuron 2023; 111:1714-1731.e3. [PMID: 37015226 DOI: 10.1016/j.neuron.2023.03.010] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 02/06/2023] [Accepted: 03/08/2023] [Indexed: 04/05/2023]
Abstract
The notion of exploiting the regenerative potential of the human brain in physiological aging or neurological diseases represents a particularly attractive alternative to conventional strategies for enhancing or restoring brain function. However, a major first question to address is whether the human brain does possess the ability to regenerate. The existence of human adult hippocampal neurogenesis (AHN) has been at the center of a fierce scientific debate for many years. The advent of single-cell transcriptomic technologies was initially viewed as a panacea to resolving this controversy. However, recent single-cell RNA sequencing studies in the human hippocampus yielded conflicting results. Here, we critically discuss and re-analyze previously published AHN-related single-cell transcriptomic datasets. We argue that, although promising, the single-cell transcriptomic profiling of AHN in the human brain can be confounded by methodological, conceptual, and biological factors that need to be consistently addressed across studies and openly discussed within the scientific community.
Collapse
Affiliation(s)
- Giorgia Tosoni
- Laboratory of Neurogenesis and Neurodegeneration, Netherlands Institute for Neuroscience, 1105 BA, Amsterdam, the Netherlands
| | - Dilara Ayyildiz
- Laboratory of Neurogenesis and Neurodegeneration, Netherlands Institute for Neuroscience, 1105 BA, Amsterdam, the Netherlands
| | - Julien Bryois
- Roche Pharma Research and Early Development, Neuroscience and Rare Diseases, Roche Innovation Center, CH-4070, Basel, Switzerland
| | - Will Macnair
- Roche Pharma Research and Early Development, Neuroscience and Rare Diseases, Roche Innovation Center, CH-4070, Basel, Switzerland
| | - Carlos P Fitzsimons
- Brain Plasticity group, Swammerdam Institute for Life Sciences, Faculty of Science, University of Amsterdam, 1098 XH, Amsterdam, the Netherlands
| | - Paul J Lucassen
- Brain Plasticity group, Swammerdam Institute for Life Sciences, Faculty of Science, University of Amsterdam, 1098 XH, Amsterdam, the Netherlands; Center for Urban Mental Health, University of Amsterdam, 1098 SM, Amsterdam, the Netherlands
| | - Evgenia Salta
- Laboratory of Neurogenesis and Neurodegeneration, Netherlands Institute for Neuroscience, 1105 BA, Amsterdam, the Netherlands.
| |
Collapse
|
21
|
Padmanabhan P, Götz J. Clinical relevance of animal models in aging-related dementia research. NATURE AGING 2023; 3:481-493. [PMID: 37202516 DOI: 10.1038/s43587-023-00402-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 03/21/2023] [Indexed: 05/20/2023]
Abstract
Alzheimer's disease (AD) and other, less prevalent dementias are complex age-related disorders that exhibit multiple etiologies. Over the past decades, animal models have provided pathomechanistic insight and evaluated countless therapeutics; however, their value is increasingly being questioned due to the long history of drug failures. In this Perspective, we dispute this criticism. First, the utility of the models is limited by their design, as neither the etiology of AD nor whether interventions should occur at a cellular or network level is fully understood. Second, we highlight unmet challenges shared between animals and humans, including impeded drug transport across the blood-brain barrier, limiting effective treatment development. Third, alternative human-derived models also suffer from the limitations mentioned above and can only act as complementary resources. Finally, age being the strongest AD risk factor should be better incorporated into the experimental design, with computational modeling expected to enhance the value of animal models.
Collapse
Affiliation(s)
- Pranesh Padmanabhan
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, the University of Queensland, Brisbane, Queensland, Australia
| | - Jürgen Götz
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, the University of Queensland, Brisbane, Queensland, Australia.
| |
Collapse
|
22
|
Charbonneau JA, Bennett JL, Chau K, Bliss-Moreau E. Reorganization in the macaque interoceptive-allostatic network following anterior cingulate cortex damage. Cereb Cortex 2023; 33:4334-4349. [PMID: 36066407 PMCID: PMC10110454 DOI: 10.1093/cercor/bhac346] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 08/08/2022] [Accepted: 08/09/2022] [Indexed: 11/14/2022] Open
Abstract
Accumulating evidence indicates that the adult brain is capable of significant structural change following damage-a capacity once thought to be largely limited to developing brains. To date, most existing research on adult plasticity has focused on how exteroceptive sensorimotor networks compensate for damage to preserve function. Interoceptive networks-those that represent and process sensory information about the body's internal state-are now recognized to be critical for a wide range of physiological and psychological functions from basic energy regulation to maintaining a sense of self, but the extent to which these networks remain plastic in adulthood has not been established. In this report, we used detailed histological analyses to pinpoint precise changes to gray matter volume in the interoceptive-allostatic network in adult rhesus monkeys (Macaca mulatta) who received neurotoxic lesions of the anterior cingulate cortex (ACC) and neurologically intact control monkeys. Relative to controls, monkeys with ACC lesions had significant and selective unilateral expansion of the ventral anterior insula and significant relative bilateral expansion of the lateral nucleus of the amygdala. This work demonstrates the capacity for neuroplasticity in the interoceptive-allostatic network which, given that changes included expansion rather than atrophy, is likely to represent an adaptive response following damage.
Collapse
Affiliation(s)
- Joey A Charbonneau
- Neuroscience Graduate Program, University of California Davis, 1544 Newton Court, Davis, CA 95618, United States
- California National Primate Research Center, University of California Davis, One Shields Avenue, Davis, CA 95616, United States
| | - Jeffrey L Bennett
- California National Primate Research Center, University of California Davis, One Shields Avenue, Davis, CA 95616, United States
- Department of Psychiatry and Behavioral Sciences, University of California Davis School of Medicine, 2230 Stockton Blvd, Sacramento, CA 95817, United States
- The MIND Institute, University of California Davis, 2825 50th Street, Sacramento, CA 95817, United States
| | - Kevin Chau
- California National Primate Research Center, University of California Davis, One Shields Avenue, Davis, CA 95616, United States
| | - Eliza Bliss-Moreau
- California National Primate Research Center, University of California Davis, One Shields Avenue, Davis, CA 95616, United States
- Department of Psychology, University of California Davis, 135 Young Hall One Shields Avenue, Davis, CA 95616, United States
| |
Collapse
|
23
|
Olpe C, Jessberger S. Cell population dynamics in the course of adult hippocampal neurogenesis: Remaining unknowns. Hippocampus 2023; 33:402-411. [PMID: 36256493 DOI: 10.1002/hipo.23475] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 09/26/2022] [Accepted: 09/30/2022] [Indexed: 11/08/2022]
Abstract
Neural stem cells (NSCs) generate new neurons throughout life in the mammalian hippocampus. The distinct developmental steps in the course of adult neurogenesis, including NSC activation, expansion, and neuronal integration, are increasingly well characterized down to the molecular level. However, substantial gaps remain in our knowledge about regulators and mechanisms involved in this biological process. This review highlights three long-standing unknowns. First, we discuss potency and identity of NSCs and the quest for a unifying model of short- and long-term self-renewal dynamics. Next, we examine cell death, specifically focusing on the early demise of newborn cells. Then, we outline the current knowledge on cell integration dynamics, discussing which (if any) neurons are replaced by newly added neurons in the hippocampal circuits. For each of these unknowns, we summarize the trajectory of studies leading to the current state of knowledge. Finally, we offer suggestions on how to fill the remaining gaps by taking advantage of novel technology to reveal currently hidden secrets in the course of adult hippocampal neurogenesis.
Collapse
Affiliation(s)
- Cora Olpe
- Brain Research Institute, Faculties of Medicine and Science, University of Zurich, Zurich, Switzerland
| | - Sebastian Jessberger
- Brain Research Institute, Faculties of Medicine and Science, University of Zurich, Zurich, Switzerland
| |
Collapse
|
24
|
Fitzgerald GS, Chuchta TG, McNay EC. Insulin‐like growth factor‐2 is a promising candidate for the treatment and prevention of Alzheimer's disease. CNS Neurosci Ther 2023; 29:1449-1469. [PMID: 36971212 PMCID: PMC10173726 DOI: 10.1111/cns.14160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 02/06/2023] [Accepted: 02/22/2023] [Indexed: 03/29/2023] Open
Abstract
Alzheimer's disease (AD) is the most common form of dementia. Current AD treatments slow the rate of cognitive decline, but do not restore lost function. One reason for the low efficacy of current treatments is that they fail to target neurotrophic processes, which are thought to be essential for functional recovery. Bolstering neurotrophic processes may also be a viable strategy for preventative treatment, since structural losses are thought to underlie cognitive decline in AD. The challenge of identifying presymptomatic patients who might benefit from preventative treatment means that any such treatment must meet a high standard of safety and tolerability. The neurotrophic peptide insulin-like growth factor-2 (IGF2) is a promising candidate for both treating and preventing AD-induced cognitive decline. Brain IGF2 expression declines in AD patients. In rodent models of AD, exogenous IGF2 modulates multiple aspects of AD pathology, resulting in (1) improved cognitive function; (2) stimulation of neurogenesis and synaptogenesis; and, (3) neuroprotection against cholinergic dysfunction and beta amyloid-induced neurotoxicity. Preclinical evidence suggests that IGF2 is likely to be safe and tolerable at therapeutic doses. In the preventative treatment context, the intranasal route of administration is likely to be the preferred method for achieving the therapeutic effect without risking adverse side effects. For patients already experiencing AD dementia, routes of administration that deliver IGF2 directly access the CNS may be necessary. Finally, we discuss several strategies for improving the translational validity of animal models used to study the therapeutic potential of IGF2.
Collapse
Affiliation(s)
| | | | - E C McNay
- University at Albany, Albany, New York, USA
| |
Collapse
|
25
|
Ghibaudi M, Amenta A, Agosti M, Riva M, Graïc JM, Bifari F, Bonfanti L. Consistency and Variation in Doublecortin and Ki67 Antigen Detection in the Brain Tissue of Different Mammals, including Humans. Int J Mol Sci 2023; 24:2514. [PMID: 36768845 PMCID: PMC9916846 DOI: 10.3390/ijms24032514] [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: 12/16/2022] [Revised: 01/10/2023] [Accepted: 01/12/2023] [Indexed: 01/31/2023] Open
Abstract
Recently, a population of "immature" neurons generated prenatally, retaining immaturity for long periods and finally integrating in adult circuits has been described in the cerebral cortex. Moreover, comparative studies revealed differences in occurrence/rate of different forms of neurogenic plasticity across mammals, the "immature" neurons prevailing in gyrencephalic species. To extend experimentation from laboratory mice to large-brained mammals, including humans, it is important to detect cell markers of neurogenic plasticity in brain tissues obtained from different procedures (e.g., post-mortem/intraoperative specimens vs. intracardiac perfusion). This variability overlaps with species-specific differences in antigen distribution or antibody species specificity, making it difficult for proper comparison. In this work, we detect the presence of doublecortin and Ki67 antigen, markers for neuronal immaturity and cell division, in six mammals characterized by widely different brain size. We tested seven commercial antibodies in four selected brain regions known to host immature neurons (paleocortex, neocortex) and newly born neurons (hippocampus, subventricular zone). In selected human brains, we confirmed the specificity of DCX antibody by performing co-staining with fluorescent probe for DCX mRNA. Our results indicate that, in spite of various types of fixations, most differences were due to the use of different antibodies and the existence of real interspecies variation.
Collapse
Affiliation(s)
- Marco Ghibaudi
- Neuroscience Institute Cavalieri Ottolenghi (NICO), 10043 Orbassano, Italy
- Department of Veterinary Sciences, University of Turin, 10095 Torino, Italy
| | - Alessia Amenta
- Laboratory of Cell Metabolism and Regenerative Medicine, Department of Medical Biotechnology and Translational Medicine, University of Milan, 20133 Milan, Italy
| | - Miriam Agosti
- Laboratory of Cell Metabolism and Regenerative Medicine, Department of Medical Biotechnology and Translational Medicine, University of Milan, 20133 Milan, Italy
| | - Marco Riva
- Department of Biomedical Sciences, Humanitas University, 20090 Pieve Emanuele, Italy
- IRCCS Humanitas Research Hospital, 20089 Rozzano, Italy
| | - Jean-Marie Graïc
- Department of Comparative Biomedicine and Food Science, University of Padova, 35020 Legnaro, Italy
| | - Francesco Bifari
- Laboratory of Cell Metabolism and Regenerative Medicine, Department of Medical Biotechnology and Translational Medicine, University of Milan, 20133 Milan, Italy
| | - Luca Bonfanti
- Neuroscience Institute Cavalieri Ottolenghi (NICO), 10043 Orbassano, Italy
- Department of Veterinary Sciences, University of Turin, 10095 Torino, Italy
| |
Collapse
|
26
|
Afferni P, Cascino-Milani F, Mattera A, Baldassarre G. A neuro-inspired computational model of life-long learning and catastrophic interference, mimicking hippocampus novelty-based dopamine modulation and lateral inhibitory plasticity. Front Comput Neurosci 2022; 16:954847. [PMID: 36157843 PMCID: PMC9500484 DOI: 10.3389/fncom.2022.954847] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 08/15/2022] [Indexed: 11/13/2022] Open
Abstract
The human brain has a remarkable lifelong learning capability to acquire new experiences while retaining previously acquired information. Several hypotheses have been proposed to explain this capability, but the underlying mechanisms are still unclear. Here, we propose a neuro-inspired firing-rate computational model involving the hippocampus and surrounding areas, that encompasses two key mechanisms possibly underlying this capability. The first is based on signals encoded by the neuromodulator dopamine, which is released by novel stimuli and enhances plasticity only when needed. The second is based on a homeostatic plasticity mechanism that involves the lateral inhibitory connections of the pyramidal neurons of the hippocampus. These mechanisms tend to protect neurons that have already been heavily employed in encoding previous experiences. The model was tested with images from the MNIST machine learning dataset, and with more naturalistic images, for its ability to mitigate catastrophic interference in lifelong learning. The results show that the proposed biologically grounded mechanisms can effectively enhance the learning of new stimuli while protecting previously acquired knowledge. The proposed mechanisms could be investigated in future empirical animal experiments and inspire machine learning models.
Collapse
Affiliation(s)
- Pierangelo Afferni
- Department of Engineering, Campus Bio-Medico University, Rome, Italy
- *Correspondence: Pierangelo Afferni
| | - Federico Cascino-Milani
- Department of Genetics and Neurobiology, Julius-Maximilians Universität Würzburg, Würzburg, Germany
| | - Andrea Mattera
- Laboratory of Embodied Natural and Artificial Intelligence, Institute of Cognitive Sciences and Technologies, National Research Council, Rome, Italy
| | - Gianluca Baldassarre
- Laboratory of Embodied Natural and Artificial Intelligence, Institute of Cognitive Sciences and Technologies, National Research Council, Rome, Italy
| |
Collapse
|
27
|
Bartkowska K, Tepper B, Turlejski K, Djavadian R. Postnatal and Adult Neurogenesis in Mammals, Including Marsupials. Cells 2022; 11:cells11172735. [PMID: 36078144 PMCID: PMC9455070 DOI: 10.3390/cells11172735] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2022] [Revised: 08/27/2022] [Accepted: 08/29/2022] [Indexed: 12/11/2022] Open
Abstract
In mammals, neurogenesis occurs during both embryonic and postnatal development. In eutherians, most brain structures develop embryonically; conversely, in marsupials, a number of brain structures develop after birth. The exception is the generation of granule cells in the dentate gyrus, olfactory bulb, and cerebellum of eutherian species. The formation of these structures starts during embryogenesis and continues postnatally. In both eutherians and marsupials, neurogenesis continues in the subventricular zone of the lateral ventricle (SVZ) and the dentate gyrus of the hippocampal formation throughout life. The majority of proliferated cells from the SVZ migrate to the olfactory bulb, whereas, in the dentate gyrus, cells reside within this structure after division and differentiation into neurons. A key aim of this review is to evaluate advances in understanding developmental neurogenesis that occurs postnatally in both marsupials and eutherians, with a particular emphasis on the generation of granule cells during the formation of the olfactory bulb, dentate gyrus, and cerebellum. We debate the significance of immature neurons in the piriform cortex of young mammals. We also synthesize the knowledge of adult neurogenesis in the olfactory bulb and the dentate gyrus of marsupials by considering whether adult-born neurons are essential for the functioning of a given area.
Collapse
Affiliation(s)
- Katarzyna Bartkowska
- Nencki Institute of Experimental Biology, Polish Academy of Sciences, 02-093 Warsaw, Poland
| | - Beata Tepper
- Nencki Institute of Experimental Biology, Polish Academy of Sciences, 02-093 Warsaw, Poland
| | - Krzysztof Turlejski
- Faculty of Biology and Environmental Sciences, Cardinal Stefan Wyszynski University in Warsaw, 01-938 Warsaw, Poland
| | - Ruzanna Djavadian
- Nencki Institute of Experimental Biology, Polish Academy of Sciences, 02-093 Warsaw, Poland
- Correspondence:
| |
Collapse
|
28
|
Mehlhorn J, Niski N, Liu K, Caspers S, Amunts K, Herold C. Regional Patterning of Adult Neurogenesis in the Homing Pigeon’s Brain. Front Psychol 2022; 13:889001. [PMID: 35898980 PMCID: PMC9311432 DOI: 10.3389/fpsyg.2022.889001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Accepted: 05/05/2022] [Indexed: 11/13/2022] Open
Abstract
In the avian brain, adult neurogenesis has been reported in the telencephalon of several species, but the functional significance of this trait is still ambiguous. Homing pigeons (Columba livia f.d.) are well-known for their navigational skills. Their brains are functionally adapted to homing with, e.g., larger hippocampi. So far, no comprehensive mapping of adult neuro- and gliogenesis or studies of different developmental neuronal stages in the telencephalon of homing pigeons exists, although comprehensive analyses in various species surely will result in a higher understanding of the functional significance of adult neurogenesis. Here, adult, free flying homing pigeons were treated with 5-bromo-deoxyuridine (BrdU) to label adult newborn cells. Brains were dissected and immunohistochemically processed with several markers (GFAP, Sox2, S100ß, Tbr2, DCX, Prox1, Ki67, NeuN, Calbindin, Calretinin) to study different stages of adult neurogenesis in a quantitative and qualitative way. Therefore, immature and adult newborn neurons and glial cells were analyzed along the anterior–posterior axis. The analysis proved the existence of different neuronal maturation stages and showed that immature cells, migrating neurons and adult newborn neurons and glia were widely and regionally unequally distributed. Double- and triple-labelling with developmental markers allowed a stage classification of adult neurogenesis in the pigeon brain (1: continuity of stem cells/proliferation, 2: fate specification, 3: differentiation/maturation, 4: integration). The most adult newborn neurons and glia were found in the intercalated hyperpallium (HI) and the hippocampal formation (HF). The highest numbers of immature (DCX+) cells were detected in the nidopallium (N). Generally, the number of newborn glial cells exceeded the number of newborn neurons. Individual structures (e.g., HI, N, and HF) showed further variations along the anterior–posterior axis. Our qualitative classification and the distribution of maturing cells in the forebrain support the idea that there is a functional specialization, respectively, that there is a link between brain-structure and function, species-specific requirements and adult neurogenesis. The high number of immature neurons also suggests a high level of plasticity, which points to the ability for rapid adaption to environmental changes through additive mechanisms. Furthermore, we discuss a possible influence of adult neurogenesis on spatial cognition.
Collapse
Affiliation(s)
- Julia Mehlhorn
- Institute for Anatomy I, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- *Correspondence: Julia Mehlhorn,
| | - Nelson Niski
- C. and O. Vogt-Institute for Brain Research, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Ke Liu
- Institute for Anatomy I, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Svenja Caspers
- Institute for Anatomy I, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- Institute of Neuroscience and Medicine (INM-1), Research Centre Jülich, Jülich, Germany
| | - Katrin Amunts
- C. and O. Vogt-Institute for Brain Research, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- Institute of Neuroscience and Medicine (INM-1), Research Centre Jülich, Jülich, Germany
| | - Christina Herold
- C. and O. Vogt-Institute for Brain Research, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| |
Collapse
|
29
|
Zhou Y, Su Y, Li S, Kennedy BC, Zhang DY, Bond AM, Sun Y, Jacob F, Lu L, Hu P, Viaene AN, Helbig I, Kessler SK, Lucas T, Salinas RD, Gu X, Chen HI, Wu H, Kleinman JE, Hyde TM, Nauen DW, Weinberger DR, Ming GL, Song H. Molecular landscapes of human hippocampal immature neurons across lifespan. Nature 2022; 607:527-533. [PMID: 35794479 PMCID: PMC9316413 DOI: 10.1038/s41586-022-04912-w] [Citation(s) in RCA: 139] [Impact Index Per Article: 46.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 05/26/2022] [Indexed: 01/18/2023]
Abstract
Immature dentate granule cells (imGCs) arising from adult hippocampal neurogenesis contribute to plasticity and unique brain functions in rodents1,2 and are dysregulated in multiple human neurological disorders3-5. Little is known about the molecular characteristics of adult human hippocampal imGCs, and even their existence is under debate1,6-8. Here we performed single-nucleus RNA sequencing aided by a validated machine learning-based analytic approach to identify imGCs and quantify their abundance in the human hippocampus at different stages across the lifespan. We identified common molecular hallmarks of human imGCs across the lifespan and observed age-dependent transcriptional dynamics in human imGCs that suggest changes in cellular functionality, niche interactions and disease relevance, that differ from those in mice9. We also found a decreased number of imGCs with altered gene expression in Alzheimer's disease. Finally, we demonstrated the capacity for neurogenesis in the adult human hippocampus with the presence of rare dentate granule cell fate-specific proliferating neural progenitors and with cultured surgical specimens. Together, our findings suggest the presence of a substantial number of imGCs in the adult human hippocampus via low-frequency de novo generation and protracted maturation, and our study reveals their molecular properties across the lifespan and in Alzheimer's disease.
Collapse
Affiliation(s)
- Yi Zhou
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Yijing Su
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Shiying Li
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Benjamin C Kennedy
- Division of Neurosurgery, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Daniel Y Zhang
- Biochemistry and Molecular Biophysics Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Allison M Bond
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Yusha Sun
- Neuroscience Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Fadi Jacob
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Lu Lu
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Peng Hu
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Angela N Viaene
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Ingo Helbig
- Division of Neurology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- The Epilepsy NeuroGenetics Initiative (ENGIN), Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Biomedical and Health Informatics (DBHi), Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Sudha K Kessler
- Division of Neurology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Timothy Lucas
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ryan D Salinas
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Xiaosong Gu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - H Isaac Chen
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Hao Wu
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA, USA
- The Epigenetics Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Joel E Kleinman
- Lieber Institute for Brain Development, The Solomon H. Snyder Department of Neuroscience, Department of Neurology, and Department of Psychiatry, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Thomas M Hyde
- Lieber Institute for Brain Development, The Solomon H. Snyder Department of Neuroscience, Department of Neurology, and Department of Psychiatry, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - David W Nauen
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Daniel R Weinberger
- Lieber Institute for Brain Development, The Solomon H. Snyder Department of Neuroscience, Department of Neurology, and Department of Psychiatry, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Guo-Li Ming
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| | - Hongjun Song
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- The Epigenetics Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
30
|
Ghibaudi M, Bonfanti L. How Widespread Are the “Young” Neurons of the Mammalian Brain? Front Neurosci 2022; 16:918616. [PMID: 35733930 PMCID: PMC9207312 DOI: 10.3389/fnins.2022.918616] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 05/17/2022] [Indexed: 12/14/2022] Open
Abstract
After the discovery of adult neurogenesis (stem cell-driven production of new neuronal elements), it is conceivable to find young, undifferentiated neurons mixed with mature neurons in the neural networks of the adult mammalian brain. This “canonical” neurogenesis is restricted to small stem cell niches persisting from embryonic germinal layers, yet, the genesis of new neurons has also been reported in various parenchymal brain regions. Whichever the process involved, several populations of “young” neurons can be found at different locations of the brain. Across the years, further complexity emerged: (i) molecules of immaturity can also be expressed by non-dividing cells born during embryogenesis, then maintaining immature features later on; (ii) remarkable interspecies differences exist concerning the types, location, amount of undifferentiated neurons; (iii) re-expression of immaturity can occur in aging (dematuration). These twists are introducing a somewhat different definition of neurogenesis than normally assumed, in which our knowledge of the “young” neurons is less sharp. In this emerging complexity, there is a need for complete mapping of the different “types” of young neurons, considering their role in postnatal development, plasticity, functioning, and interspecies differences. Several important aspects are at stake: the possible role(s) that the young neurons may play in maintaining brain efficiency and in prevention/repair of neurological disorders; nonetheless, the correct translation of results obtained from laboratory rodents. Hence, the open question is: how many types of undifferentiated neurons do exist in the brain, and how widespread are they?
Collapse
Affiliation(s)
- Marco Ghibaudi
- Neuroscience Institute Cavalieri Ottolenghi (NICO), Orbassano, Italy
- Department of Veterinary Sciences, University of Turin, Grugliasco, Italy
| | - Luca Bonfanti
- Neuroscience Institute Cavalieri Ottolenghi (NICO), Orbassano, Italy
- Department of Veterinary Sciences, University of Turin, Grugliasco, Italy
- *Correspondence: Luca Bonfanti,
| |
Collapse
|
31
|
Tartt AN, Mariani MB, Hen R, Mann JJ, Boldrini M. Dysregulation of adult hippocampal neuroplasticity in major depression: pathogenesis and therapeutic implications. Mol Psychiatry 2022; 27:2689-2699. [PMID: 35354926 PMCID: PMC9167750 DOI: 10.1038/s41380-022-01520-y] [Citation(s) in RCA: 154] [Impact Index Per Article: 51.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 02/22/2022] [Accepted: 03/09/2022] [Indexed: 02/07/2023]
Abstract
Major depressive disorder (MDD) was previously hypothesized to be a disease of monoamine deficiency in which low levels of monoamines in the synaptic cleft were believed to underlie depressive symptoms. More recently, however, there has been a paradigm shift toward a neuroplasticity hypothesis of depression in which downstream effects of antidepressants, such as increased neurogenesis, contribute to improvements in cognition and mood. This review takes a top-down approach to assess how changes in behavior and hippocampal-dependent circuits may be attributed to abnormalities at the molecular, structural, and synaptic level. We conclude with a discussion of how antidepressant treatments share a common effect in modulating neuroplasticity and consider outstanding questions and future perspectives.
Collapse
Affiliation(s)
| | | | - Rene Hen
- Departments of Psychiatry, Columbia University, New York, NY, USA
- Neuroscience, Columbia University, New York, NY, USA
- Pharmacology, Columbia University, New York, NY, USA
- Integrative Neuroscience, NYS Psychiatric Institute, New York, NY, USA
| | - J John Mann
- Departments of Psychiatry, Columbia University, New York, NY, USA
- Molecular Imaging and Neuropathology, NYS Psychiatric Institute, New York, NY, USA
| | - Maura Boldrini
- Departments of Psychiatry, Columbia University, New York, NY, USA.
- Molecular Imaging and Neuropathology, NYS Psychiatric Institute, New York, NY, USA.
| |
Collapse
|
32
|
Velloso FJ, Shankar S, Parpura V, Rakic P, Levison SW. Neural Stem Cells in Adult Mammals are not Astrocytes. ASN Neuro 2022; 14:17590914221134739. [PMID: 36330653 PMCID: PMC9638700 DOI: 10.1177/17590914221134739] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 09/27/2022] [Accepted: 10/01/2022] [Indexed: 11/06/2022] Open
Abstract
At the turn of the 21st century studies of the cells that resided in the adult mammalian subventricular zone (SVZ) characterized the neural stem cells (NSCs) as a subtype of astrocyte. Over the ensuing years, numerous studies have further characterized the properties of these NSCs and compared them to parenchymal astrocytes. Here we have evaluated the evidence collected to date to establish whether classifying the NSCs as astrocytes is appropriate and useful. We also performed a meta-analysis with 4 previously published datasets that used cell sorting and unbiased single-cell RNAseq to highlight the distinct gene expression profiles of adult murine NSCs and niche astrocytes. On the basis of our understanding of the properties and functions of astrocytes versus the properties and functions of NSCs, and from our comparative transcriptomic analyses we conclude that classifying the adult mammalian NSC as an astrocyte is potentially misleading. From our vantage point, it is more appropriate to refer to the cells in the adult mammalian SVZ that retain the capacity to produce new neurons and macroglia as NSCs without attaching the term "astrocyte-like."
Collapse
Affiliation(s)
- Fernando Janczur Velloso
- Department of Pharmacology, Physiology & Neuroscience, New
Jersey Medical School, Rutgers University, Newark, NJ, USA
| | - Sandhya Shankar
- Department of Pharmacology, Physiology & Neuroscience, New
Jersey Medical School, Rutgers University, Newark, NJ, USA
| | - Vladimir Parpura
- Department of Neurobiology, The University of Alabama at Birmingham,
Birmingham, AL, USA
| | - Pasko Rakic
- Department of Neuroscience, Yale School of Medicine, New Haven, CT,
USA
- Kavli Institute for Neuroscience, Yale School of Medicine, New
Haven, CT, USA
| | - Steven W. Levison
- Department of Pharmacology, Physiology & Neuroscience, New
Jersey Medical School, Rutgers University, Newark, NJ, USA
| |
Collapse
|