1
|
Eid F, Boushehri M, Boucher C, Rajkanth N, Sa AF, Alhoutan T, Vavetsi K, Dibart S, Ma Y. Chronic Lipopolysaccharide Exposure Causes AD-Like Pathology in Male Mice With Intact Blood-Brain Barrier. FASEB J 2025; 39:e70601. [PMID: 40317532 DOI: 10.1096/fj.202403117rr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 04/08/2025] [Accepted: 04/24/2025] [Indexed: 05/07/2025]
Abstract
Chronic inflammatory conditions like periodontitis and inflammatory bowel disease (IBD) are reported to contribute to the pathogenesis of late-onset Alzheimer's disease (AD). Gram-negative bacteria are the main bacterial species causing oral and gut mucosal infections. Lipopolysaccharide (LPS) is a major inflammation-inducing molecule in Gram-negative bacteria. LPS derived from the oral bacterium Porphyromonas gingivalis exhibits heterogeneous tetra-acylated and penta-acylated lipid A, while LPS from Escherichia coli exhibits the classical hexa-acylated lipid A. Whether P. gingivalis-LPS and E. coli-LPS play a similar role in the progression of late-onset AD is unknown. Using adult, wild-type C57BL/6J mice to mimic the adult population without genetically determined predisposition to AD, we showed that chronic inflammation induced by a 28-day, subcutaneous infusion of P. gingivalis-LPS or E. coli-LPS can lead to neuroinflammation and AD-like cognitive decline and pathology in male mice. At this relatively early stage (4 weeks) of chronic inflammation when the blood-brain barrier is intact, both P. gingivalis-LPS and E. coli-LPS cause neuroinflammation through Toll-like receptor 4 (TLR4) and Toll-like receptor 2 (TLR2) expressed at microglia in the brain. Notably, only E. coli-LPS induces significant inflammatory responses systemically. In short, our results suggest that chronic P. gingivalis-LPS release (occurring in chronic periodontitis) or E. coli-LPS release (occurring in IBD) could harm the brain before the blood-brain barrier is disrupted; continuous local P. gingivalis-LPS release might do harm to the brain before exhibiting adverse effects systemically.
Collapse
Affiliation(s)
- Fady Eid
- Department of Periodontology, Boston University Henry M. Goldman School of Dental Medicine, Boston, Massachusetts, USA
| | - Mohammad Boushehri
- Department of Periodontology, Boston University Henry M. Goldman School of Dental Medicine, Boston, Massachusetts, USA
| | - Chloé Boucher
- Department of Periodontology, Boston University Henry M. Goldman School of Dental Medicine, Boston, Massachusetts, USA
| | - Nischwethaa Rajkanth
- Department of Periodontology, Boston University Henry M. Goldman School of Dental Medicine, Boston, Massachusetts, USA
| | - Ana Flor Sa
- Department of Periodontology, Boston University Henry M. Goldman School of Dental Medicine, Boston, Massachusetts, USA
| | - Thamir Alhoutan
- Department of Periodontology, Boston University Henry M. Goldman School of Dental Medicine, Boston, Massachusetts, USA
| | - Konstantina Vavetsi
- Department of Periodontology, Boston University Henry M. Goldman School of Dental Medicine, Boston, Massachusetts, USA
| | - Serge Dibart
- Department of Periodontology, Boston University Henry M. Goldman School of Dental Medicine, Boston, Massachusetts, USA
| | - Yun Ma
- Department of Periodontology, Boston University Henry M. Goldman School of Dental Medicine, Boston, Massachusetts, USA
| |
Collapse
|
2
|
Han H, Yao J, Wu J, Mao S, Pan H, Qv L, Zhu G, Ren J, Yu Y, Xuan F, Zeng L, Ma Y, Yang Z, Zhu Z, Zhu F, Li MD. Implications of neurogenesis in depression through BDNF: rodent models, regulatory pathways, gut microbiota, and potential therapy. Mol Psychiatry 2025:10.1038/s41380-025-03044-7. [PMID: 40341897 DOI: 10.1038/s41380-025-03044-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 03/05/2025] [Accepted: 04/30/2025] [Indexed: 05/11/2025]
Abstract
Major Depressive Disorder (MDD) is a prevalent psychiatric disorder with a profound impact on global health, necessitating a deeper understanding of its pathophysiology. This review synthesizes current evidence linking neurogenesis, particularly in the hippocampal region, with MDD. Accumulating data showed a significant reduction of neurogenesis in the hippocampal region of both MDD patients and various MDD rodent models. We highlight the role of brain-derived neurotrophic factor (BDNF) and its associated signaling pathways in regulating neurogenesis and depressive symptoms. Additionally, the influence of gut microbiota on the neurogenesis in depression is presented, offering a novel perspective on environmental modulation of neurogenesis. This review also underscores the potential antidepressant interventions targeting neurogenesis and BDNF's regulation, such as therapeutic benefits of environmental enrichment, physical activity, and pharmacological agents in enhancing neurogenesis and alleviating depressive symptoms. Together, this systemic review provides a foundation for future research aiming at developing personalized treatments by targeting neurogenesis in MDD, potentially leading to novel biomarkers and therapeutic strategies.
Collapse
Affiliation(s)
- Haijun Han
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, Institute of Brain and Cognitive Science, School of Medicine, Hangzhou City University, Hangzhou, China
| | - Jianhua Yao
- Joint Institute of Tobacco and Health, Kunming, Yunnan, China
| | - Jinhan Wu
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, Institute of Brain and Cognitive Science, School of Medicine, Hangzhou City University, Hangzhou, China
| | - Shiqi Mao
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, Institute of Brain and Cognitive Science, School of Medicine, Hangzhou City University, Hangzhou, China
| | - Hongyi Pan
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, Institute of Brain and Cognitive Science, School of Medicine, Hangzhou City University, Hangzhou, China
| | - Lingling Qv
- Central Laboratory, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Guanqi Zhu
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, Institute of Brain and Cognitive Science, School of Medicine, Hangzhou City University, Hangzhou, China
| | - Juntian Ren
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, Institute of Brain and Cognitive Science, School of Medicine, Hangzhou City University, Hangzhou, China
| | - Yaning Yu
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, Institute of Brain and Cognitive Science, School of Medicine, Hangzhou City University, Hangzhou, China
| | - Feiyang Xuan
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, Institute of Brain and Cognitive Science, School of Medicine, Hangzhou City University, Hangzhou, China
| | - Linghui Zeng
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, Institute of Brain and Cognitive Science, School of Medicine, Hangzhou City University, Hangzhou, China
| | - Yunlong Ma
- Department of Psychiatry, Perelman School of Medicine, Lifespan Brain Institute at Penn Med and the Children's Hospital of Philadelphia, University of Pennsylvania, Philadelphia, PA, USA
| | - Zhongli Yang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Zhijing Zhu
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, Institute of Brain and Cognitive Science, School of Medicine, Hangzhou City University, Hangzhou, China
| | - Feng Zhu
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, Institute of Brain and Cognitive Science, School of Medicine, Hangzhou City University, Hangzhou, China
| | - Ming D Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.
| |
Collapse
|
3
|
Feighan KM, Nesan D, Kurrasch DM. Gestational bisphenol A exposure alters energy homeostasis and adult hypothalamic neurogenesis in female mice. Sci Rep 2024; 14:16082. [PMID: 38992091 PMCID: PMC11239822 DOI: 10.1038/s41598-024-66726-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Accepted: 07/03/2024] [Indexed: 07/13/2024] Open
Abstract
Regulation of physiological homeostasis, including energy balance, is thought to be modified by low levels of adult neurogenesis in the hypothalamus. Hormones such as oestradiol can influence both embryonic and adult hypothalamic neurogenic programs, demonstrating a sensitivity of hypothalamic neural progenitor cells to endogenous hormones. Previously we showed that gestational exposure to environmental levels of the xenoestrogen bisphenol A (BPA) changed neural progenitor cell behaviors in the embryo; however, we did not examine if these changes were permanent to affect adult neurogenesis. Here we investigated whether adult neuro- and/or gliogenesis were altered in mice prenatally exposed to BPA and placed on a high-fat diet challenge. Gestationally exposed adult female mice on a standard diet gained less weight than non-BPA controls, whereas gestationally exposed BPA females on a high-fat diet gained more weight than controls. Males exposed to gestational BPA showed no differences in weight gain relative to control males. Concomitantly, adult neurogenesis was increased in the VMH, DMH, and PVN of adult female mice exposed to BPA on standard diet, suggesting that disrupted adult neurogenesis might perturb normal energy balance regulation in females. These results add to growing evidence that low-dose BPA exposure in utero causes changes to adult hypothalamic function.
Collapse
Affiliation(s)
- Kira M Feighan
- Department of Medical Genetics, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
- Montreal Neurological Institute, McGill University, Montreal, QC, Canada
| | - Dinushan Nesan
- Department of Medical Genetics, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
| | - Deborah M Kurrasch
- Department of Medical Genetics, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, Canada.
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada.
| |
Collapse
|
4
|
Noche JA, Radhakrishnan H, Ubele MF, Boaz K, Mefford JL, Jones ED, van Rooyen HY, Perpich JA, McCarty K, Meacham B, Smiley J, Bembenek Bailey SA, Puskás LG, Powell DK, Sordo L, Phelan MJ, Norris CM, Head E, Stark CEL. Age-Related Brain Atrophy and the Positive Effects of Behavioral Enrichment in Middle-Aged Beagles. J Neurosci 2024; 44:e2366232024. [PMID: 38561226 PMCID: PMC11097262 DOI: 10.1523/jneurosci.2366-23.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 02/08/2024] [Accepted: 02/28/2024] [Indexed: 04/04/2024] Open
Abstract
Aging dogs serve as a valuable preclinical model for Alzheimer's disease (AD) due to their natural age-related development of β-amyloid (Aβ) plaques, human-like metabolism, and large brains that are ideal for studying structural brain aging trajectories from serial neuroimaging. Here we examined the effects of chronic treatment with the calcineurin inhibitor (CNI) tacrolimus or the nuclear factor of activated T cells (NFAT)-inhibiting compound Q134R on age-related canine brain atrophy from a longitudinal study in middle-aged beagles (36 females, 7 males) undergoing behavioral enrichment. Annual MRI was analyzed using modern, automated techniques for region-of-interest-based and voxel-based volumetric assessments. We found that the frontal lobe showed accelerated atrophy with age, while the caudate nucleus remained relatively stable. Remarkably, the hippocampus increased in volume in all dogs. None of these changes were influenced by tacrolimus or Q134R treatment. Our results suggest that behavioral enrichment can prevent atrophy and increase the volume of the hippocampus but does not prevent aging-associated prefrontal cortex atrophy.
Collapse
Affiliation(s)
| | - Hamsanandini Radhakrishnan
- University of California, Irvine, California 92697
- University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | | | - Kathy Boaz
- University of Kentucky, Lexington, Kentucky 40506
| | | | - Erin D Jones
- University of Kentucky, Lexington, Kentucky 40506
| | | | | | | | | | | | | | | | | | - Lorena Sordo
- University of California, Irvine, California 92697
| | | | | | | | | |
Collapse
|
5
|
Giatti S, Diviccaro S, Cioffi L, Cosimo Melcangi R. Post-Finasteride Syndrome And Post-Ssri Sexual Dysfunction: Two Clinical Conditions Apparently Distant, But Very Close. Front Neuroendocrinol 2024; 72:101114. [PMID: 37993021 DOI: 10.1016/j.yfrne.2023.101114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 10/31/2023] [Accepted: 11/19/2023] [Indexed: 11/24/2023]
Abstract
Post-finasteride syndrome and post-SSRI sexual dysfunction, are two poorly explored clinical conditions in which men treated for androgenetic alopecia with finasteride or for depression with SSRI antidepressants show persistent side effects despite drug suspension (e.g., sexual dysfunction, psychological complaints, sleep disorders). Because of some similarities in the symptoms, common pathological mechanisms are proposed here. Indeed, as discussed, clinical studies and preclinical data obtained so far suggest an important role for brain modulators (i.e., neuroactive steroids), neurotransmitters (i.e., serotonin, and cathecolamines), and gut microbiota in the context of the gut-brain axis. In particular, the observed interconnections of these signals in these two clinical conditions may suggest similar etiopathogenetic mechanisms, such as the involvement of the enzyme converting norepinephrine into epinephrine (i.e., phenylethanolamine N-methyltransferase). However, despite the current efforts, more work is still needed to advance the understanding of these clinical conditions in terms of diagnostic markers and therapeutic strategies.
Collapse
Affiliation(s)
- Silvia Giatti
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milano, Italy
| | - Silvia Diviccaro
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milano, Italy
| | - Lucia Cioffi
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milano, Italy
| | - Roberto Cosimo Melcangi
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milano, Italy.
| |
Collapse
|
6
|
Ávila-González D, Romero-Morales I, Caro L, Martínez-Juárez A, Young LJ, Camacho-Barrios F, Martínez-Alarcón O, Castro AE, Paredes RG, Díaz NF, Portillo W. Increased proliferation and neuronal fate in prairie vole brain progenitor cells cultured in vitro: effects by social exposure and sexual dimorphism. Biol Sex Differ 2023; 14:77. [PMID: 37919790 PMCID: PMC10623709 DOI: 10.1186/s13293-023-00563-2] [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: 06/09/2023] [Accepted: 10/23/2023] [Indexed: 11/04/2023] Open
Abstract
BACKGROUND The prairie vole (Microtus ochrogaster) is a socially monogamous rodent that establishes an enduring pair bond after cohabitation, with (6 h) or without (24 h) mating. Previously, we reported that social interaction and mating increased cell proliferation and differentiation to neuronal fate in neurogenic niches in male voles. We hypothesized that neurogenesis may be a neural plasticity mechanism involved in mating-induced pair bond formation. Here, we evaluated the differentiation potential of neural progenitor cells (NPCs) isolated from the subventricular zone (SVZ) of both female and male adult voles as a function of sociosexual experience. Animals were assigned to one of the following groups: (1) control (Co), sexually naive female and male voles that had no contact with another vole of the opposite sex; (2) social exposure (SE), males and females exposed to olfactory, auditory, and visual stimuli from a vole of the opposite sex, but without physical contact; and (3) social cohabitation with mating (SCM), male and female voles copulating to induce pair bonding formation. Subsequently, the NPCs were isolated from the SVZ, maintained, and supplemented with growth factors to form neurospheres in vitro. RESULTS Notably, we detected in SE and SCM voles, a higher proliferation of neurosphere-derived Nestin + cells, as well as an increase in mature neurons (MAP2 +) and a decrease in glial (GFAP +) differentiated cells with some sex differences. These data suggest that when voles are exposed to sociosexual experiences that induce pair bonding, undifferentiated cells of the SVZ acquire a commitment to a neuronal lineage, and the determined potential of the neurosphere is conserved despite adaptations under in vitro conditions. Finally, we repeated the culture to obtain neurospheres under treatments with different hormones and factors (brain-derived neurotrophic factor, estradiol, prolactin, oxytocin, and progesterone); the ability of SVZ-isolated cells to generate neurospheres and differentiate in vitro into neurons or glial lineages in response to hormones or factors is also dependent on sex and sociosexual context. CONCLUSION Social interactions that promote pair bonding in voles change the properties of cells isolated from the SVZ. Thus, SE or SCM induces a bias in the differentiation potential in both sexes, while SE is sufficient to promote proliferation in SVZ-isolated cells from male brains. In females, proliferation increases when mating is performed. The next question is whether the rise in proliferation and neurogenesis of cells from the SVZ are plastic processes essential for establishing, enhancing, maintaining, or accelerating pair bond formation. Highlights 1. Sociosexual experiences that promote pair bonding (social exposure and social cohabitation with mating) induce changes in the properties of neural stem/progenitor cells isolated from the SVZ in adult prairie voles. 2. Social interactions lead to increased proliferation and induce a bias in the differentiation potential of SVZ-isolated cells in both male and female voles. 3. The differentiation potential of SVZ-isolated cells is conserved under in vitro conditions, suggesting a commitment to a neuronal lineage under a sociosexual context. 4. Hormonal and growth factors treatments (brain-derived neurotrophic factor, estradiol, prolactin, oxytocin, and progesterone) affect the generation and differentiation of neurospheres, with dependencies on sex and sociosexual context. 5. Proliferation and neurogenesis in the SVZ may play a crucial role in establishing, enhancing, maintaining, or accelerating pair bond formation.
Collapse
Affiliation(s)
- Daniela Ávila-González
- Departamento de Neurobiología Conductual y Cognitiva, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Juriquilla, Querétaro, Mexico
- Departamento de Fisiología y Desarrollo Celular, Instituto Nacional de Perinatología Isidro Espinosa de los Reyes, Mexico City, Mexico
| | - Italo Romero-Morales
- Departamento de Fisiología y Desarrollo Celular, Instituto Nacional de Perinatología Isidro Espinosa de los Reyes, Mexico City, Mexico
| | - Lizette Caro
- Departamento de Fisiología y Desarrollo Celular, Instituto Nacional de Perinatología Isidro Espinosa de los Reyes, Mexico City, Mexico
| | - Alejandro Martínez-Juárez
- Departamento de Fisiología y Desarrollo Celular, Instituto Nacional de Perinatología Isidro Espinosa de los Reyes, Mexico City, Mexico
| | - Larry J Young
- Silvio O. Conte Center for Oxytocin and Social Cognition, Center for Translational Social Neuroscience, Emory National Primate Research Center, Department of Psychiatry and Behavioral Sciences, Emory University, Atlanta, USA
| | - Francisco Camacho-Barrios
- Departamento de Neurobiología Conductual y Cognitiva, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Juriquilla, Querétaro, Mexico
| | - Omar Martínez-Alarcón
- Departamento de Fisiología y Desarrollo Celular, Instituto Nacional de Perinatología Isidro Espinosa de los Reyes, Mexico City, Mexico
| | - Analía E Castro
- Departamento de Neurobiología Conductual y Cognitiva, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Juriquilla, Querétaro, Mexico
| | - Raúl G Paredes
- Departamento de Neurobiología Conductual y Cognitiva, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Juriquilla, Querétaro, Mexico
- Escuela Nacional de Estudios Superiores Juriquilla, Universidad Nacional Autónoma de México, Querétaro, Mexico
| | - Néstor F Díaz
- Departamento de Fisiología y Desarrollo Celular, Instituto Nacional de Perinatología Isidro Espinosa de los Reyes, Mexico City, Mexico.
| | - Wendy Portillo
- Departamento de Neurobiología Conductual y Cognitiva, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Juriquilla, Querétaro, Mexico.
| |
Collapse
|
7
|
Roeder SS, Burkardt P, Rost F, Rode J, Brusch L, Coras R, Englund E, Håkansson K, Possnert G, Salehpour M, Primetzhofer D, Csiba L, Molnár S, Méhes G, Tonchev AB, Schwab S, Bergmann O, Huttner HB. Evidence for postnatal neurogenesis in the human amygdala. Commun Biol 2022; 5:366. [PMID: 35440676 PMCID: PMC9018740 DOI: 10.1038/s42003-022-03299-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 03/22/2022] [Indexed: 12/12/2022] Open
Abstract
The human amygdala is involved in processing of memory, decision-making, and emotional responses. Previous studies suggested that the amygdala may represent a neurogenic niche in mammals. By combining two distinct methodological approaches, lipofuscin quantification and 14C-based retrospective birth dating of neurons, along with mathematical modelling, we here explored whether postnatal neurogenesis exists in the human amygdala. We investigated post-mortem samples of twelve neurologically healthy subjects. The average rate of lipofuscin-negative neurons was 3.4%, representing a substantial proportion of cells substantially younger than the individual. Mass spectrometry analysis of genomic 14C-concentrations in amygdala neurons compared with atmospheric 14C-levels provided evidence for postnatal neuronal exchange. Mathematical modelling identified a best-fitting scenario comprising of a quiescent and a renewing neuronal population with an overall renewal rate of >2.7% per year. In conclusion, we provide evidence for postnatal neurogenesis in the human amygdala with cell turnover rates comparable to the hippocampus. Lipofuscin labeling and 14 C retrospective birth-dating of neurons, along with mathematical modelling, here suggest continued postnatal neurogenesis in the human amygdala, rather than protracted maturation of developmentally generated neurons.
Collapse
Affiliation(s)
- Sebastian S Roeder
- Department of Neurology, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Petra Burkardt
- Department of Neurology, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Fabian Rost
- Center for Regenerative Therapies (CRTD), TU Dresden, Dresden, Germany.,Max Planck Institute for the Physics of Complex Systems, Dresden, Germany.,Center for Information Services and High Performance Computing (ZIH), TU Dresden, Dresden, Germany.,Center for Molecular and Cellular Bioengineering, DRESDEN-concept Genome Center, TU Dresden, Dresden, Germany
| | - Julian Rode
- Center for Information Services and High Performance Computing (ZIH), TU Dresden, Dresden, Germany
| | - Lutz Brusch
- Center for Information Services and High Performance Computing (ZIH), TU Dresden, Dresden, Germany
| | - Roland Coras
- Department of Neuropathology, University of Erlangen-Nuremberg, Erlangen, Germany
| | | | - Karl Håkansson
- Tandem Laboratory, Uppsala University, Uppsala, Sweden.,Department of Physics and Astronomy, Uppsala University, Uppsala, Sweden
| | | | - Mehran Salehpour
- Department of Physics and Astronomy, Uppsala University, Uppsala, Sweden
| | - Daniel Primetzhofer
- Tandem Laboratory, Uppsala University, Uppsala, Sweden.,Department of Physics and Astronomy, Uppsala University, Uppsala, Sweden
| | - László Csiba
- Department of Neurology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary.,MTA-DE Cerebrovascular and Neurodegenerative Research Group, Debrecen, Hungary
| | - Sarolta Molnár
- Department of Pathology, University of Debrecen, Debrecen, Hungary
| | - Gábor Méhes
- Department of Pathology, University of Debrecen, Debrecen, Hungary
| | - Anton B Tonchev
- Departments of Anatomy, Cell Biology and Stem Cell Biology, Medical University Varna, Varna, Bulgaria
| | - Stefan Schwab
- Department of Neurology, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Olaf Bergmann
- Center for Regenerative Therapies (CRTD), TU Dresden, Dresden, Germany.,Department of Cell and Molecular Biology, Karolinska Institute, Stockholm, Sweden
| | - Hagen B Huttner
- Department of Neurology, University of Erlangen-Nuremberg, Erlangen, Germany. .,Department of Neurology, Justus Liebig University Giessen, Giessen, Germany.
| |
Collapse
|
8
|
Beiriger J, Habib A, Jovanovich N, Kodavali CV, Edwards L, Amankulor N, Zinn PO. The Subventricular Zone in Glioblastoma: Genesis, Maintenance, and Modeling. Front Oncol 2022; 12:790976. [PMID: 35359410 PMCID: PMC8960165 DOI: 10.3389/fonc.2022.790976] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Accepted: 02/07/2022] [Indexed: 12/13/2022] Open
Abstract
Glioblastoma (GBM) is a malignant tumor with a median survival rate of 15-16 months with standard care; however, cases of successful treatment offer hope that an enhanced understanding of the pathology will improve the prognosis. The cell of origin in GBM remains controversial. Recent evidence has implicated stem cells as cells of origin in many cancers. Neural stem/precursor cells (NSCs) are being evaluated as potential initiators of GBM tumorigenesis. The NSCs in the subventricular zone (SVZ) have demonstrated similar molecular profiles and share several distinctive characteristics to proliferative glioblastoma stem cells (GSCs) in GBM. Genomic and proteomic studies comparing the SVZ and GBM support the hypothesis that the tumor cells and SVZ cells are related. Animal models corroborate this connection, demonstrating migratory patterns from the SVZ to the tumor. Along with laboratory and animal research, clinical studies have demonstrated improved progression-free survival in patients with GBM after radiation to the ipsilateral SVZ. Additionally, key genetic mutations in GBM for the most part carry regulatory roles in the SVZ as well. An exciting avenue towards SVZ modeling and determining its role in gliomagenesis in the human context is human brain organoids. Here we comprehensively discuss and review the role of the SVZ in GBM genesis, maintenance, and modeling.
Collapse
Affiliation(s)
- Jamison Beiriger
- Department of Neurosurgery, University of Pittsburgh Medical Center, Pittsburgh, PA, United States
- Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh PA, United States
| | - Ahmed Habib
- Department of Neurosurgery, University of Pittsburgh Medical Center, Pittsburgh, PA, United States
- Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh PA, United States
| | - Nicolina Jovanovich
- Department of Neurosurgery, University of Pittsburgh Medical Center, Pittsburgh, PA, United States
- Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh PA, United States
| | - Chowdari V. Kodavali
- Department of Neurosurgery, University of Pittsburgh Medical Center, Pittsburgh, PA, United States
- Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh PA, United States
| | - Lincoln Edwards
- Department of Neurosurgery, University of Pittsburgh Medical Center, Pittsburgh, PA, United States
- Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh PA, United States
| | - Nduka Amankulor
- Department of Neurosurgery, University of Pittsburgh Medical Center, Pittsburgh, PA, United States
- Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh PA, United States
| | - Pascal O. Zinn
- Department of Neurosurgery, University of Pittsburgh Medical Center, Pittsburgh, PA, United States
- Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh PA, United States
| |
Collapse
|
9
|
Rodrigues RS, Paulo SL, Moreira JB, Tanqueiro SR, Sebastião AM, Diógenes MJ, Xapelli S. Adult Neural Stem Cells as Promising Targets in Psychiatric Disorders. Stem Cells Dev 2021; 29:1099-1117. [PMID: 32723008 DOI: 10.1089/scd.2020.0100] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The development of new therapies for psychiatric disorders is of utmost importance, given the enormous toll these disorders pose to society nowadays. This should be based on the identification of neural substrates and mechanisms that underlie disease etiopathophysiology. Adult neural stem cells (NSCs) have been emerging as a promising platform to counteract brain damage. In this perspective article, we put forth a detailed view of how NSCs operate in the adult brain and influence brain homeostasis, having profound implications at both behavioral and functional levels. We appraise evidence suggesting that adult NSCs play important roles in regulating several forms of brain plasticity, particularly emotional and cognitive flexibility, and that NSC dynamics are altered upon brain pathology. Furthermore, we discuss the potential therapeutic value of utilizing adult endogenous NSCs as vessels for regeneration, highlighting their importance as targets for the treatment of multiple mental illnesses, such as affective disorders, schizophrenia, and addiction. Finally, we speculate on strategies to surpass current challenges in neuropsychiatric disease modeling and brain repair.
Collapse
Affiliation(s)
- Rui S Rodrigues
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal.,Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Sara L Paulo
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal.,Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - João B Moreira
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal.,Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Sara R Tanqueiro
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal.,Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Ana M Sebastião
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal.,Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Maria J Diógenes
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal.,Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Sara Xapelli
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal.,Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| |
Collapse
|
10
|
Drummond KD, Waring ML, Faulkner GJ, Blewitt ME, Perry CJ, Kim JH. Hippocampal neurogenesis mediates sex-specific effects of social isolation and exercise on fear extinction in adolescence. Neurobiol Stress 2021; 15:100367. [PMID: 34337114 PMCID: PMC8313755 DOI: 10.1016/j.ynstr.2021.100367] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 06/30/2021] [Accepted: 07/12/2021] [Indexed: 12/15/2022] Open
Abstract
Impaired extinction of conditioned fear is associated with anxiety disorders. Common lifestyle factors, like isolation stress and exercise, may alter the ability to extinguish fear. However, the effect of and interplay between these factors on adolescent fear extinction, and the relevant underlying neural mechanisms are unknown. Here we examined the effects of periadolescent social isolation and physical activity on adolescent fear extinction in rats and explored neurogenesis as a potential mechanism. Isolation stress impaired extinction recall in male adolescents, an effect prevented by exercise. Extinction recall in female adolescents was unaffected by isolation stress. However, exercise disrupted extinction recall in isolated females. Extinction recall in isolated females was positively correlated to the number of immature neurons in the ventral hippocampus, suggesting that exercise affected extinction recall via neurogenesis in females. Pharmacologically suppressing cellular proliferation in isolated adolescents using temozolomide blocked the effect of exercise on extinction recall in both sexes. Together, these findings highlight sex-specific outcomes of isolation stress and exercise on adolescent brain and behavior, and highlights neurogenesis as a potential mechanism underlying lifestyle effects on adolescent fear extinction. Periadolescent isolation stress disrupted extinction recall in male adolescents. Running prevented isolation-induced extinction recall deficit in male adolescents. Exercise impaired extinction recall in isolated female adolescents. Exercise increased hippocampal neurogenesis, except in isolated males. Suppression of neurogenesis blocked exercise effects in isolated adolescents.
Collapse
Affiliation(s)
- Katherine D Drummond
- Mental Health Theme, The Florey Institute of Neuroscience and Mental Health, Parkville, VIC, 3052, Australia.,Florey Department of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, 3052, Australia
| | - Michelle L Waring
- Florey Department of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, 3052, Australia
| | - Geoffrey J Faulkner
- Mater Research Institute - University of Queensland, Woolloongabba, QLD, 4102, Australia.,Queensland Brain Institute, University of Queensland, St. Lucia, QLD, 4067, Australia
| | - Marnie E Blewitt
- The Epigenetics and Development Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, 3052, Australia.,The Department of Medical Biology, University of Melbourne, Parkville, VIC, 3052, Australia
| | - Christina J Perry
- Mental Health Theme, The Florey Institute of Neuroscience and Mental Health, Parkville, VIC, 3052, Australia.,Florey Department of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, 3052, Australia
| | - Jee Hyun Kim
- Mental Health Theme, The Florey Institute of Neuroscience and Mental Health, Parkville, VIC, 3052, Australia.,Florey Department of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, 3052, Australia.,IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Geelong, Australia
| |
Collapse
|
11
|
Navarrete-Yañez V, Garate-Carrillo A, Ayala M, Rodriguez-Castañeda A, Mendoza-Lorenzo P, Ceballos G, Ordoñez-Razo R, Dugar S, Schreiner G, Villarreal F, Ramirez-Sanchez I. Stimulatory effects of (-)-epicatechin and its enantiomer (+)-epicatechin on mouse frontal cortex neurogenesis markers and short-term memory: proof of concept. Food Funct 2021; 12:3504-3515. [PMID: 33900336 DOI: 10.1039/d0fo03084h] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Consumption of (-)-epicatechin (Epi), a cacao flavanol improves cognition. The aim was to compare the effects of (-)-Epi or its stereoisomer (+)-Epi on mouse frontal cortex-dependent short-term working memory and modulators of neurogenesis. Three-month-old male mice (n = 7 per group) were provided by gavage either water (vehicle; Veh), (-)-Epi, at 1 mg kg-1 or (+)-Epi at 0.1 mg per kg of body weight for 15 days. After treatment, spontaneous alternation was evaluated by Y-maze. Brain frontal cortex was isolated for nitrate/nitrite measurements, Western blotting for nerve growth factor (NGF), microtubule associated protein 2 (MAP2), endothelial and neuronal nitric oxide synthase (eNOS and nNOS) and immunohistochemistry for neuronal specific protein (NeuN), doublecortin (DCX), capillary (CD31) and neurofilaments (NF200). Results demonstrate the stimulatory capacity of (-)-Epi and (+)-Epi on markers of neuronal proliferation as per increases in immunoreactive cells for NeuN (74 and 120% respectively), DCX (70 and 124%) as well as in NGF (34.4, 63.6%) and MAP2 (41.8, 63.8%). Capillary density yielded significant increases with (-)-Epi (∼80%) vs. (+)-Epi (∼160%). CD31 protein levels increased with (-)-Epi (∼70%) and (+)-Epi (∼140%). Effects correlated with nitrate/nitrite stimulation by (-)-Epi and (+)-Epi (110.2, 246.5%) and enhanced eNOS phosphorylation (Ser1177) with (-)-Epi and (+)-Epi (21.4, 41.2%) while nNOS phosphorylation only increased with (+)-Epi (18%). Neurofilament staining was increased in (-)-Epi by 135.6 and 84% with (+)-Epi. NF200 increased with (-)-Epi (116%) vs. (+)-Epi (84.5%). Frontal cortex-dependent short-term spatial working improved with (-)-Epi and (+)-Epi (15, 13%). In conclusion, results suggest that both enantiomers, but more effectively (+)-Epi, upregulate neurogenesis markers likely through stimulation of capillary formation and NO triggering, improvements in memory.
Collapse
Affiliation(s)
- Viridiana Navarrete-Yañez
- Seccion de Estudios de Posgrado e Investigacion, Escuela Superior de Medicina, Instituto Politecnico Nacional, Mexico D.F., Mexico.
| | - Alejandra Garate-Carrillo
- Seccion de Estudios de Posgrado e Investigacion, Escuela Superior de Medicina, Instituto Politecnico Nacional, Mexico D.F., Mexico. and School of Medicine, University of California, San Diego, California, USA
| | - Marcos Ayala
- Seccion de Estudios de Posgrado e Investigacion, Escuela Superior de Medicina, Instituto Politecnico Nacional, Mexico D.F., Mexico.
| | - Antonio Rodriguez-Castañeda
- Seccion de Estudios de Posgrado e Investigacion, Escuela Superior de Medicina, Instituto Politecnico Nacional, Mexico D.F., Mexico.
| | - Patricia Mendoza-Lorenzo
- Division Academica de Ciencias Basicas, Unidad Chontalpa, Universidad Juarez, Autonoma de Tabasco, Tabasco, Mexico
| | - Guillermo Ceballos
- Seccion de Estudios de Posgrado e Investigacion, Escuela Superior de Medicina, Instituto Politecnico Nacional, Mexico D.F., Mexico.
| | - Rosa Ordoñez-Razo
- Unidad de Investigación en Genética Humana, Hospital de Pediatría, Centro Médico SXXI, Instituto Mexicano del Seguro Social, Mexico D.F., Mexico
| | | | | | - Francisco Villarreal
- School of Medicine, University of California, San Diego, California, USA and VA San Diego Health Care System, San Diego, California, USA
| | - Israel Ramirez-Sanchez
- Seccion de Estudios de Posgrado e Investigacion, Escuela Superior de Medicina, Instituto Politecnico Nacional, Mexico D.F., Mexico.
| |
Collapse
|
12
|
Zhou YL, Wu FC, Liu WJ, Zheng W, Wang CY, Zhan YN, Lan XF, Ning YP. Volumetric changes in subcortical structures following repeated ketamine treatment in patients with major depressive disorder: a longitudinal analysis. Transl Psychiatry 2020; 10:264. [PMID: 32747631 PMCID: PMC7400625 DOI: 10.1038/s41398-020-00945-9] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 07/07/2020] [Accepted: 07/14/2020] [Indexed: 12/12/2022] Open
Abstract
Abnormal subcortical structures have been associated with major depressive disorder (MDD) and could be reversed by antidepressant treatment. To date no study has examined the relationship between subcortical volumes and repeated ketamine treatment. The current study investigated volume changes in specific subcortical structures and hippocampal subfields after six ketamine infusions. Forty-four patients with MDD received six subanesthetic dose infusions of ketamine. Depressive symptoms were assessed and magnetic resonance imaging scans were performed before and after six ketamine infusions. FreeSurfer software was used to process the T1 images and analyze the volumes of the subcortical regions and hippocampal subfields. After six ketamine infusions, increases were observed in the volumes of the left amygdala; the right hippocampus; the cornu ammonis 4 body, granule cell and molecular layer of the dentate gyrus body in the left hippocampus; and the cornu ammonis 4 head and molecular layer head in the right hippocampus. Positive correlations were found between symptom improvement and the pretreatment volumes of the right thalamus (r = 0.501; P = 0.001) and left subiculum head of the hippocampus (r = 0.471; P = 0.002), and changes in the volumes of the left amygdala (r = -0.452; P = 0.003) and the left cornu ammonis 4 body (r = -0.537; P < 0.001). Our findings provided evidence for critical roles of the amygdala and specific hippocampal subfields in the antidepressant effect of repeated ketamine treatment. Relatively larger volumes in right thalamus and left subiculum head in the hippocampus can predict a superior clinical outcome of ketamine treatment in MDD patients.
Collapse
Affiliation(s)
- Yan-Ling Zhou
- grid.410737.60000 0000 8653 1072The Affiliated Brain Hospital of Guangzhou Medical University (Guangzhou Huiai Hospital), Guangzhou, China ,Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, China
| | - Feng-Chun Wu
- grid.410737.60000 0000 8653 1072The Affiliated Brain Hospital of Guangzhou Medical University (Guangzhou Huiai Hospital), Guangzhou, China ,Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, China
| | - Wei-Jian Liu
- grid.410737.60000 0000 8653 1072The Affiliated Brain Hospital of Guangzhou Medical University (Guangzhou Huiai Hospital), Guangzhou, China ,Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, China
| | - Wei Zheng
- grid.410737.60000 0000 8653 1072The Affiliated Brain Hospital of Guangzhou Medical University (Guangzhou Huiai Hospital), Guangzhou, China ,Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, China
| | - Cheng-Yu Wang
- grid.410737.60000 0000 8653 1072The Affiliated Brain Hospital of Guangzhou Medical University (Guangzhou Huiai Hospital), Guangzhou, China ,Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, China
| | - Yan-Ni Zhan
- grid.410737.60000 0000 8653 1072The Affiliated Brain Hospital of Guangzhou Medical University (Guangzhou Huiai Hospital), Guangzhou, China ,Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, China
| | - Xiao-Feng Lan
- grid.410737.60000 0000 8653 1072The Affiliated Brain Hospital of Guangzhou Medical University (Guangzhou Huiai Hospital), Guangzhou, China ,Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, China
| | - Yu-Ping Ning
- The Affiliated Brain Hospital of Guangzhou Medical University (Guangzhou Huiai Hospital), Guangzhou, China. .,Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, China. .,The First School of Clinical Medicine, Southern Medical University, Guangzhou, China.
| |
Collapse
|
13
|
Kim GS, Uddin M. Sex-specific and shared expression profiles of vulnerability and resilience to trauma in brain and blood. Biol Sex Differ 2020; 11:13. [PMID: 32228684 PMCID: PMC7106761 DOI: 10.1186/s13293-020-00288-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Accepted: 03/03/2020] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND While post-traumatic stress disorder (PTSD) is defined by behavioral/cognitive symptoms most directly relevant to brain function, it can be considered a systemic disorder characterized by a distinct inability to reinstate homeostasis after trauma. METHODS In this study, we conducted a secondary analysis of gene expression profiles in key PTSD-relevant tissues, namely blood, amygdala, and hippocampus, from a rat model of PTSD, to identify sex-specific and shared processes associated with individual differences in response to recent trauma exposure. RESULTS Our findings suggest both shared and sex-specific mechanisms underlying individual differences associated with vulnerability and resilience to trauma in hippocampus, amygdala, and blood. By disentangling cell composition from transcriptional changes, we found higher proportions of hippocampal oligodendrocytes in the PTSD-like, extreme behavioral response (EBR) group for both sexes and also identified modules for transcriptional activity associated with group differences (i.e., response to trauma) in the hippocampus that appeared to be sex-specific. By contrast, we found prominent sex differences, but no group differences, in amygdalar cell composition, and both shared and sex-specific modules representing PTSD-relevant transcriptional activity in the amygdala. Across amygdala and hippocampus, both sex-specific and shared processes were relevant to an overarching framework for EBR implicating disrupted TNFα/NFκΒ signaling and excitatory/inhibitory imbalance in dysregulated synaptic/structural plasticity with important implications for fear learning and memory. Our main finding in peripheral blood was consistent with the human literature and identified wound healing processes and hemostasis to be upregulated in the resilient, minimal behavioral response (MBR) group across sexes, but disrupted in a sexually dimorphic manner in the EBR group. CONCLUSION In contrast to the varied characterization of the PTSD-like EBR group, characterization of MBR across blood, amygdala, and hippocampus suggests a common theme of upregulated wound healing and extracellular matrix (ECM) remodeling shared between sexes. In all, we identified differential oligodendrocyte proportions in hippocampus between PTSD-like EBR and resilient MBR, and identified processes and pathways that characterize the EBR and MBR-associated transcriptional changes across hippocampus, amygdala, and blood. The sex-specific mechanisms involved in EBR may contribute to the pronounced disparity in risk for PTSD, with women much more likely to develop PTSD.
Collapse
Affiliation(s)
- Grace S Kim
- Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, IL, USA.,Medical Scholars Program, University of Illinois College of Medicine at Urbana-Champaign, Urbana, IL, USA
| | - Monica Uddin
- Genomics Program, Center for Global Health and Infectious Disease Research, College of Public Health, University of South Florida, 3720 Spectrum Blvd., Ste. 304, Tampa, FL, 33612, USA.
| |
Collapse
|
14
|
Bulin SE, Simmons SJ, Richardson DR, Latchney SE, Deutsch HM, Yun S, Eisch AJ. Indices of dentate gyrus neurogenesis are unaffected immediately after or following withdrawal from morphine self-administration compared to saline self-administering control male rats. Behav Brain Res 2020; 381:112448. [PMID: 31870778 PMCID: PMC7036141 DOI: 10.1016/j.bbr.2019.112448] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Revised: 12/01/2019] [Accepted: 12/19/2019] [Indexed: 12/16/2022]
Abstract
Opiates - including morphine - are powerful analgesics with high abuse potential. In rodents, chronic opiate exposure or self-administration negatively impacts hippocampal-dependent function, an effect perhaps due in part to the well-documented opiate-induced inhibition of dentate gyrus (DG) precursor proliferation and neurogenesis. Recently, however, intravenous (i.v.) morphine self-administration (MSA) was reported to enhance the survival of new rat DG neurons. To reconcile these disparate results, we used rat i.v. MSA to assess 1) whether a slightly-higher dose MSA paradigm also increases new DG neuron survival; 2) how MSA influences cells in different stages of DG neurogenesis, particularly maturation and survival; and 3) if MSA-induced changes in DG neurogenesis persist through a period of abstinence. To label basal levels of proliferation, rats received the S-phase marker bromodeoxyuridine (BrdU, i.p.) 24 -h prior to 21 days (D) of i.v. MSA or saline self-administration (SSA). Either immediately after SA (0-D) or after 4 weeks in the home cage (28-D withdrawal), stereology was used to quantify DG proliferating precursors (or cells in cell cycle; Ki67+ cells), neuroblast/immature neurons (DCX+ cells), and surviving DG granule cells (BrdU+ cells). Analysis revealed the number of DG cells immunopositive for these neurogenesis-relevant markers was similar between MSA and SSA rats at the 0-D or 28-D timepoints. These negative data highlight the impact experimental parameters, timepoint selection, and quantification approach have on neurogenesis results, and are discussed in the context of the large literature showing the negative impact of opiates on DG neurogenesis.
Collapse
Affiliation(s)
- Sarah E Bulin
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Steven J Simmons
- Department of Anesthesiology and Critical Care Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Devon R Richardson
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Sarah E Latchney
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, USA; Department of Neurobiology, St. Mary's College of Maryland, St. Mary's City, MD, 20686-3001
| | - Hannah M Deutsch
- Department of Anesthesiology and Critical Care Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Sanghee Yun
- Department of Anesthesiology and Critical Care Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Amelia J Eisch
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, USA; Department of Anesthesiology and Critical Care Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA; Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
15
|
Murine's amygdala microstructure and elevated plus maze activities following R. vomitoria root bark and G. latifolium leaf extracts administration. Anat Sci Int 2020; 95:342-355. [PMID: 32006225 DOI: 10.1007/s12565-020-00527-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2019] [Accepted: 01/22/2020] [Indexed: 12/31/2022]
Abstract
R. vomitoria (RV), a plant used locally in the management of psychotic disorders, adversely affects the brain functionally and structurally. Such adverse reports, as well as the potential of G. latifolium (GL) to mitigate same warranted this investigation on the combined actions of RV and GL on the amygdala. Twenty-four male CD-1 mice weighing 22-27 g were divided into four groups (n = 6): Control (20 ml/kg body weight, b.w., distilled water); RV (200 mg/kg b.w.), GL (200 mg/kg b.w.), and RV (200 mg/kg b.w.) and GL (200 mg/kg b.w.) combination orally, and for 14 days. On day 15, the elevated-plus maze test was carried out and the animals sacrificed, and processed for histological and immunohistochemical studies. Neurobehavioural results showed significant decrease (p[Formula: see text] 0.001) in stretch-attend posture, time spent in closed arms, grooming frequency, protected head-dip, as well as significantly (p [Formula: see text] 0.01) increased time spent in the open arms and unprotected head-dips of the RV group. The combined RV and GL groups showed no such differences in these parameters. Histologically, the amygdala showed hypertrophied cells, with pyknotic and karyorrhectic nuclei, and reduced expression of Nissl substance in the RV group, while the combined RV and GL group showed less degenerative features. Glial fibrillary acidic protein expression (GFAP) was increased in the RV group, while the combined RV and GL group showed reduced expression. In conclusion, RV root bark extract has adverse effects on the microstructure of murines' amygdala and their behaviour, which may be ameliorated by GL.
Collapse
|
16
|
Daun KA, Fuchigami T, Koyama N, Maruta N, Ikenaka K, Hitoshi S. Early Maternal and Social Deprivation Expands Neural Stem Cell Population Size and Reduces Hippocampus/Amygdala-Dependent Fear Memory. Front Neurosci 2020; 14:22. [PMID: 32063832 PMCID: PMC7000530 DOI: 10.3389/fnins.2020.00022] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Accepted: 01/10/2020] [Indexed: 11/13/2022] Open
Abstract
Early life stress can exert detrimental or beneficial effects on neural development and postnatal behavior depending on the timing, duration, strength, and ability to control the stressors. In this study, we utilized a maternal and social deprivation (MSD) model to investigate the effects of early life stress on neural stem cells (NSCs) and neurogenesis in the adult brain. We found that MSD during the stress-hyporesponsive period (SHRP) (early-MSD), when corticosterone secretion is suppressed, increased the size of the NSC population, whereas the same stress beyond the SHRP abrogated these effects. Early-MSD enhanced neurogenesis not only in the dentate gyrus of the hippocampus, one of the classic neurogenic regions, but also in the amygdala. In addition, mice exposed to early-MSD exhibited a reduction in amygdala/hippocampus-dependent fear memory. These results suggest that animals exposed to early life stress during the SHRP have reinforced stress resilience to cope with perceived stressors to maintain a normal homeostatic state.
Collapse
Affiliation(s)
- Kenny Anak Daun
- Department of Integrative Physiology, Shiga University of Medical Science, Otsu, Japan
| | - Takahiro Fuchigami
- Department of Integrative Physiology, Shiga University of Medical Science, Otsu, Japan
| | - Natsu Koyama
- Department of Integrative Physiology, Shiga University of Medical Science, Otsu, Japan
| | - Noriko Maruta
- Department of Psychiatry, Health Center, Hitotsubashi University, Tokyo, Japan
| | - Kazuhiro Ikenaka
- Division of Neurobiology and Bioinformatics, National Institute for Physiological Sciences, Okazaki, Japan.,Department of Physiological Sciences, School of Life Sciences, The Graduate University for Advanced Studies, Okazaki, Japan
| | - Seiji Hitoshi
- Department of Integrative Physiology, Shiga University of Medical Science, Otsu, Japan.,Division of Neurobiology and Bioinformatics, National Institute for Physiological Sciences, Okazaki, Japan.,Department of Physiological Sciences, School of Life Sciences, The Graduate University for Advanced Studies, Okazaki, Japan
| |
Collapse
|
17
|
Lathe R, Singadia S, Jordan C, Riedel G. The interoceptive hippocampus: Mouse brain endocrine receptor expression highlights a dentate gyrus (DG)-cornu ammonis (CA) challenge-sufficiency axis. PLoS One 2020; 15:e0227575. [PMID: 31940330 PMCID: PMC6961916 DOI: 10.1371/journal.pone.0227575] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Accepted: 12/20/2019] [Indexed: 12/19/2022] Open
Abstract
The primeval function of the mammalian hippocampus (HPC) remains uncertain. Implicated in learning and memory, spatial navigation, and neuropsychological disorders, evolutionary theory suggests that the HPC evolved from a primeval chemosensory epithelium. Deficits in sensing of internal body status ('interoception') in patients with HPC lesions argue that internal sensing may be conserved in higher vertebrates. We studied the expression patterns in mouse brain of 250 endocrine receptors that respond to blood-borne ligands. Key findings are (i) the proportions and levels of endocrine receptor expression in the HPC are significantly higher than in all other comparable brain regions. (ii) Surprisingly, the distribution of endocrine receptor expression within mouse HPC was found to be highly structured: receptors signaling 'challenge' are segregated in dentate gyrus (DG), whereas those signaling 'sufficiency' are principally found in cornu ammonis (CA) regions. Selective expression of endocrine receptors in the HPC argues that interoception remains a core feature of hippocampal function. Further, we report that ligands of DG receptors predominantly inhibit both synaptic potentiation and neurogenesis, whereas CA receptor ligands conversely promote both synaptic potentiation and neurogenesis. These findings suggest that the hippocampus acts as an integrator of body status, extending its role in context-dependent memory encoding from 'where' and 'when' to 'how I feel'. Implications for anxiety and depression are discussed.
Collapse
Affiliation(s)
- Richard Lathe
- Division of Infection Medicine, University of Edinburgh Medical School, Little France, Edinburgh, Scotland, United Kingdom
- * E-mail: (RL); (GR)
| | - Sheena Singadia
- Division of Behavioral Neuroscience, Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen, Scotland, United Kingdom
| | - Crispin Jordan
- Division of Biomedical Sciences, University of Edinburgh Medical School, George Square, Edinburgh, Scotland, United Kingdom
| | - Gernot Riedel
- Division of Behavioral Neuroscience, Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen, Scotland, United Kingdom
- * E-mail: (RL); (GR)
| |
Collapse
|
18
|
Mishra A, Singh S, Tiwari V, Bano S, Shukla S. Dopamine D1 receptor agonism induces dynamin related protein-1 inhibition to improve mitochondrial biogenesis and dopaminergic neurogenesis in rat model of Parkinson's disease. Behav Brain Res 2019; 378:112304. [PMID: 31626851 DOI: 10.1016/j.bbr.2019.112304] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Revised: 09/26/2019] [Accepted: 10/14/2019] [Indexed: 12/11/2022]
Abstract
Dopamine (DA) neurotransmitter act on dopamine receptors (D1-D5) to regulate motor functions, reward, addiction and cognitive behavior. The depletion of DA in midbrain due to degeneration of nigral dopaminergic (DAergic) neurons leads to Parkinson's disease (PD). DA agonist and levodopa (L-DOPA) are the only therapies used for symptomatic relief in PD. However, the role of DA receptors in PD pathogenesis and how they are associated with mitochondrial functions and DAergic neurogenesis is still not known. Here, we investigated the mechanistic aspect of DA D1 receptor mediated control of DAergic neurogenesis, motor behavior and mitochondrial functions in rat PD model. The pharmacological activation of D1 receptors markedly improved motor deficits, mitochondrial biogenesis, ATP levels, mitochondrial membrane potential and defended nigral DAergic neurons against 6-hydroxydopamine (6-OHDA) induced neurotoxicity in adult rats. However, the D1 agonist mediated effects were abolished following D1 receptor antagonist treatment in 6-OHDA lesioned rats. Interestingly, pharmacological inhibition of dynamin related protein-1 (Drp-1) by Mdivi-1 in D1 antagonist treated PD rats, significantly restored behavioral deficits, mitochondrial functions, mitochondrial biogenesis and increased the number of newborn DAergic neurons in substantia nigra pars compacta (SNpc). Drp-1 inhibition mediated neuroprotective effects in PD rats were associated with increased level of protein kinase-B/Akt and extracellular-signal-regulated kinase (ERK). Taken together, our data suggests that dopamine D1 receptor mediated reduction in mitochondrial fission and enhanced DAergic neurogenesis may involve Drp-1 inhibition which led to improved behavioral recovery in PD rats.
Collapse
Affiliation(s)
- Akanksha Mishra
- Division of Neuroscience and Ageing biology, CSIR-Central Drug Research Institute, Lucknow (U.P.), India; Academy of Scientific and Innovative Research, New Delhi, India
| | - Sonu Singh
- Division of Neuroscience and Ageing biology, CSIR-Central Drug Research Institute, Lucknow (U.P.), India; L4078, Department of Neuroscience, School of Medicine, University of Connecticut (Uconn) Health Center, 263 Farmington Avenue
| | - Virendra Tiwari
- Division of Neuroscience and Ageing biology, CSIR-Central Drug Research Institute, Lucknow (U.P.), India; Academy of Scientific and Innovative Research, New Delhi, India
| | - Shameema Bano
- Division of Neuroscience and Ageing biology, CSIR-Central Drug Research Institute, Lucknow (U.P.), India
| | - Shubha Shukla
- Division of Neuroscience and Ageing biology, CSIR-Central Drug Research Institute, Lucknow (U.P.), India; Academy of Scientific and Innovative Research, New Delhi, India.
| |
Collapse
|
19
|
Hillerer KM, Slattery DA, Pletzer B. Neurobiological mechanisms underlying sex-related differences in stress-related disorders: Effects of neuroactive steroids on the hippocampus. Front Neuroendocrinol 2019; 55:100796. [PMID: 31580837 PMCID: PMC7115954 DOI: 10.1016/j.yfrne.2019.100796] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Revised: 09/26/2019] [Accepted: 09/27/2019] [Indexed: 12/19/2022]
Abstract
Men and women differ in their vulnerability to a variety of stress-related illnesses, but the underlying neurobiological mechanisms are not well understood. This is likely due to a comparative dearth of neurobiological studies that assess male and female rodents at the same time, while human neuroimaging studies often don't model sex as a variable of interest. These sex differences are often attributed to the actions of sex hormones, i.e. estrogens, progestogens and androgens. In this review, we summarize the results on sex hormone actions in the hippocampus and seek to bridge the gap between animal models and findings in humans. However, while effects of sex hormones on the hippocampus are largely consistent in animals and humans, methodological differences challenge the comparability of animal and human studies on stress effects. We summarise our current understanding of the neurobiological mechanisms that underlie sex-related differences in behavior and discuss implications for stress-related illnesses.
Collapse
Affiliation(s)
- Katharina M Hillerer
- Department of Obstetrics and Gynaecology, Salzburger Landeskrankenhaus (SALK), Paracelsus Medical University (PMU), Clinical Research Center Salzburg (CRCS), Salzburg, Austria.
| | - David A Slattery
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital, Goethe University, Frankfurt, Germany
| | - Belinda Pletzer
- Department of Psychology, University of Salzburg, Salzburg, Austria; Centre for Cognitive Neuroscience, University of Salzburg, Salzburg, Austria
| |
Collapse
|
20
|
Kandasamy M, Radhakrishnan RK, Poornimai Abirami GP, Roshan SA, Yesudhas A, Balamuthu K, Prahalathan C, Shanmugaapriya S, Moorthy A, Essa MM, Anusuyadevi M. Possible Existence of the Hypothalamic-Pituitary-Hippocampal (HPH) Axis: A Reciprocal Relationship Between Hippocampal Specific Neuroestradiol Synthesis and Neuroblastosis in Ageing Brains with Special Reference to Menopause and Neurocognitive Disorders. Neurochem Res 2019; 44:1781-1795. [PMID: 31254250 DOI: 10.1007/s11064-019-02833-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Revised: 05/13/2019] [Accepted: 06/19/2019] [Indexed: 12/18/2022]
Abstract
The hippocampus-derived neuroestradiol plays a major role in neuroplasticity, independent of circulating estradiol that originates from gonads. The response of hypothalamus-pituitary regions towards the synthesis of neuroestradiol in the hippocampus is an emerging scientific concept in cognitive neuroscience. Hippocampal plasticity has been proposed to be regulated via neuroblasts, a major cellular determinant of functional neurogenesis in the adult brain. Defects in differentiation, integration and survival of neuroblasts in the hippocampus appear to be an underlying cause of neurocognitive disorders. Gonadotropin receptors and steroidogenic enzymes have been found to be expressed in neuroblasts in the hippocampus of the brain. However, the reciprocal relationship between hippocampal-specific neuroestradiol synthesis along neuroblastosis and response of pituitary based feedback regulation towards regulation of estradiol level in the hippocampus have not completely been ascertained. Therefore, this conceptual article revisits (1) the cellular basis of neuroestradiol synthesis (2) a potential relationship between neuroestradiol synthesis and neuroblastosis in the hippocampus (3) the possible involvement of aberrant neuroestradiol production with mitochondrial dysfunctions and dyslipidemia in menopause and adult-onset neurodegenerative disorders and (4) provides a hypothesis for the possible existence of the hypothalamic-pituitary-hippocampal (HPH) axis in the adult brain. Eventually, understanding the regulation of hippocampal neurogenesis by abnormal levels of neuroestradiol concentration in association with the feedback regulation of HPH axis might provide additional cues to establish a neuroregenerative therapeutic management for mood swings, depression and cognitive decline in menopause and neurocognitive disorders.
Collapse
Affiliation(s)
- Mahesh Kandasamy
- Laboratory of Stem Cells and Neuroregeneration, Department of Animal Science, School of Life Sciences, Bharathidasan University, Tiruchirappalli, Tamil Nadu, 620024, India.
- Department of Animal Science, School of Life Sciences, Bharathidasan University, Tiruchirappalli, Tamil Nadu, 620024, India.
- School of Life Sciences, Bharathidasan University, Tiruchirappalli, Tamil Nadu, 620024, India.
- Faculty Recharge Programme, University Grants Commission(UGC-FRP), New Delhi, India.
| | - Risna Kanjirassery Radhakrishnan
- Laboratory of Stem Cells and Neuroregeneration, Department of Animal Science, School of Life Sciences, Bharathidasan University, Tiruchirappalli, Tamil Nadu, 620024, India
- Department of Animal Science, School of Life Sciences, Bharathidasan University, Tiruchirappalli, Tamil Nadu, 620024, India
| | - G P Poornimai Abirami
- School of Life Sciences, Bharathidasan University, Tiruchirappalli, Tamil Nadu, 620024, India
| | - Syed Aasish Roshan
- Molecular Gerontology Laboratory, Department of Biochemistry, School of Life Sciences, Bharathidasan University, Tiruchirappalli, Tamil Nadu, 620024, India
- Department of Biochemistry, Bharathidasan University, Tiruchirappalli, 620024, India
| | - Ajisha Yesudhas
- Laboratory of Stem Cells and Neuroregeneration, Department of Animal Science, School of Life Sciences, Bharathidasan University, Tiruchirappalli, Tamil Nadu, 620024, India
- Department of Animal Science, School of Life Sciences, Bharathidasan University, Tiruchirappalli, Tamil Nadu, 620024, India
| | - Kadalmani Balamuthu
- Department of Animal Science, School of Life Sciences, Bharathidasan University, Tiruchirappalli, Tamil Nadu, 620024, India
- School of Life Sciences, Bharathidasan University, Tiruchirappalli, Tamil Nadu, 620024, India
| | - Chidambaram Prahalathan
- School of Life Sciences, Bharathidasan University, Tiruchirappalli, Tamil Nadu, 620024, India
- Department of Biochemistry, Bharathidasan University, Tiruchirappalli, 620024, India
| | | | - Anbalagan Moorthy
- Department of Integrative Biology, School of Bioscience and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, 632014, India
| | - Musthafa Mohamed Essa
- Department of Food Science and Nutrition, College of Agricultural and Marine Sciences, Sultan Qaboos University, Muscat, Oman
- Ageing and Dementia Research Group, Sultan Qaboos University, Muscat, Oman
| | - Muthuswamy Anusuyadevi
- School of Life Sciences, Bharathidasan University, Tiruchirappalli, Tamil Nadu, 620024, India
- Molecular Gerontology Laboratory, Department of Biochemistry, School of Life Sciences, Bharathidasan University, Tiruchirappalli, Tamil Nadu, 620024, India
- Department of Biochemistry, Bharathidasan University, Tiruchirappalli, 620024, India
| |
Collapse
|
21
|
Sequeira A, Shen K, Gottlieb A, Limon A. Human brain transcriptome analysis finds region- and subject-specific expression signatures of GABA AR subunits. Commun Biol 2019; 2:153. [PMID: 31069263 PMCID: PMC6494906 DOI: 10.1038/s42003-019-0413-7] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Accepted: 04/03/2019] [Indexed: 11/19/2022] Open
Abstract
Altered expression of GABA receptors (GABAARs) has been implicated in neurological and psychiatric disorders, but limited information about region-specific GABAAR subunit expression in healthy human brains, heteromeric assembly of major isoforms, and their collective organization across healthy individuals, are major roadblocks to understanding their role in non-physiological states. Here, by using microarray and RNA-Seq datasets-from single cell nuclei to global brain expression-from the Allen Institute, we find that transcriptional expression of GABAAR subunits is anatomically organized according to their neurodevelopmental origin. The data show a combination of complementary and mutually-exclusive expression patterns that delineate major isoforms, and which is highly stereotypical across brains from control donors. We summarize the region-specific signature of GABAR subunits per subject and its variability in a control population sample that can be used as a reference for remodeling changes during homeostatic rearrangements of GABAAR subunits after physiological, pharmacological or pathological challenges.
Collapse
Affiliation(s)
- Adolfo Sequeira
- Department of Psychiatry and Human Behavior, School of Medicine, University of California Irvine, Irvine, CA USA
| | - Kevin Shen
- Department of Neurology, Mitchel Center for Neurodegenerative Diseases, School of Medicine, University of Texas Medical Branch, Galveston, TX USA
| | - Assaf Gottlieb
- School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX USA
| | - Agenor Limon
- Department of Neurology, Mitchel Center for Neurodegenerative Diseases, School of Medicine, University of Texas Medical Branch, Galveston, TX USA
| |
Collapse
|
22
|
The Role of SVZ Stem Cells in Glioblastoma. Cancers (Basel) 2019; 11:cancers11040448. [PMID: 30934929 PMCID: PMC6521108 DOI: 10.3390/cancers11040448] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Revised: 03/22/2019] [Accepted: 03/26/2019] [Indexed: 12/27/2022] Open
Abstract
As most common primary brain cancer, glioblastoma is also the most aggressive and malignant form of cancer in the adult central nervous system. Glioblastomas are genetic and transcriptional heterogeneous tumors, which in spite of intensive research are poorly understood. Over the years conventional therapies failed to affect a cure, resulting in low survival rates of affected patients. To improve the clinical outcome, an important approach is to identify the cells of origin. One potential source for these are neural stem cells (NSCs) located in the subventricular zone, which is one of two niches in the adult nervous system where NSCs with the capacity of self-renewal and proliferation reside. These cells normally give rise to neuronal as well as glial progenitor cells. This review summarizes current findings about links between NSCs and cancer stem cells in glioblastoma and discusses current therapeutic approaches, which arise as a result of identifying the cell of origin in glioblastoma.
Collapse
|
23
|
Kim GH, Baek HK, Lee JS, Kim SJ, Yi SS. Chronic Oral Administration of Tenebrio molitor Extract Exhibits Inhibitory Effect on Glucocorticoid Receptor Overexpression in the Hippocampus of Ovariectomy-Induced Estrogen Deficient Mice. J Food Sci 2019; 84:687-694. [PMID: 30714630 DOI: 10.1111/1750-3841.14454] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Revised: 01/03/2019] [Accepted: 01/07/2019] [Indexed: 12/22/2022]
Abstract
It has been reported that estrogen deficiency in female disrupts systemic endocrinologic regulatory mechanisms, finally leading to osteoporotic condition. Estrogen deficiency also down-regulates brain functions due to its deficits of its original roles in a number of neurological events. Therefore, it is necessary to find alternative materials that can prevent osteoporotic condition and maintain normal brain functions to correct such hormone deficiency. In the present study, we found that novel compounds originated from larvae of Tenebrio molitor (TM) possessed anti-osteoporotic effect. They could also prevent abnormal progressive brain function by deaccelerating enhanced HPA-axis negative feedback while maintaining neurogenesis in hippocampus. We daily administered TM to ovariectomized (OVX) ddY mice for 4 weeks and then performed histological and hormonal evaluations for its anti-osteoporotic effects. In addition, we investigated glucocorticoid receptor (GR) expression and neuroblast expression (DCX) in the hippocampal dentate gyrus morphologically by immunohistochemistry analysis. According to our results, TM has anti-osteoporotic effects. It also tends to bring interfered brain environment back to normal condition. These results suggest that TM might have anti-osteoporosis effect and enhancing effects on enrichment of environment in brain by being antidestroyed hormonal deficiency simultaneously. This is the first study to report that TM can be used as source of bioactive substance to prevent decreased neurogenesis and impaired HPA axis driven by high GR expression in the hippocampus in hormonal deficient female animals. PRACTICAL APPLICATION: Anti-osteoporosis effect and stress resistance due to improved brain function caused by the ingestion of Tenebrio molitor extract were observed in postmenopausal women. T. molitor is available as a nutritional supplement for bone and brain health, which menopausal women need most.
Collapse
Affiliation(s)
- Gwang-Ho Kim
- Dept. of Biomedical Lab. Science, College of Medical Sciences, Soonchunhyang Univ., Asan, 31538, Republic of Korea
| | - Hye Kyung Baek
- Dept. of Biomedical Lab. Science, College of Medical Sciences, Soonchunhyang Univ., Asan, 31538, Republic of Korea
| | - Jong Suk Lee
- Biocenter, Gyeonggido Business and Science Accelerator (GBSA), Suwon, 16229, Republic of Korea
| | - Sung-Jo Kim
- Dept. of Biotechnology, Hoseo Univ., Asan, 31499, Republic of Korea
| | - Sun Shin Yi
- Dept. of Biomedical Lab. Science, College of Medical Sciences, Soonchunhyang Univ., Asan, 31538, Republic of Korea
| |
Collapse
|
24
|
Narita Y, Tsutiya A, Nakano Y, Ashitomi M, Sato K, Hosono K, Kaneko T, Chen RD, Lee JR, Tseng YC, Hwang PP, Ohtani-Kaneko R. Androgen induced cellular proliferation, neurogenesis, and generation of GnRH3 neurons in the brain of mature female Mozambique tilapia. Sci Rep 2018; 8:16855. [PMID: 30442908 PMCID: PMC6237963 DOI: 10.1038/s41598-018-35303-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Accepted: 11/05/2018] [Indexed: 01/24/2023] Open
Abstract
The neuroplastic mechanisms in the fish brain that underlie sex reversal remain unknown. Gonadotropin-releasing hormone 3 (GnRH3) neurons control male reproductive behaviours in Mozambique tilapia and show sexual dimorphism, with males having a greater number of GnRH3 neurons. Treatment with androgens such as 11-ketotestosterone (KT), but not 17β-estradiol, increases the number of GnRH3 neurons in mature females to a level similar to that observed in mature males. Compared with oestrogen, the effect of androgen on neurogenesis remains less clear. The present study examined the effects of 11-KT, a non-aromatizable androgen, on cellular proliferation, neurogenesis, generation of GnRH3 neurons and expression of cell cycle-related genes in mature females. The number of proliferating cell nuclear antigen-positive cells was increased by 11-KT. Simultaneous injection of bromodeoxyuridine and 11-KT significantly increased the number of newly-generated (newly-proliferated) neurons, but did not affect radial glial cells, and also resulted in newly-generated GnRH3 neurons. Transcriptome analysis showed that 11-KT modulates the expression of genes related to the cell cycle process. These findings suggest that tilapia could serve as a good animal model to elucidate the effects of androgen on adult neurogenesis and the mechanisms for sex reversal in the fish brain.
Collapse
Affiliation(s)
- Yasuto Narita
- Department of Life Sciences, Toyo University, 1-1-1 Itakura, Oura, Gunma, 374-0193, Japan
| | - Atsuhiro Tsutiya
- Department of Life Sciences, Toyo University, 1-1-1 Itakura, Oura, Gunma, 374-0193, Japan
| | - Yui Nakano
- Department of Life Sciences, Toyo University, 1-1-1 Itakura, Oura, Gunma, 374-0193, Japan
| | - Moe Ashitomi
- Department of Life Sciences, Toyo University, 1-1-1 Itakura, Oura, Gunma, 374-0193, Japan
| | - Kenjiro Sato
- Department of Life Sciences, Toyo University, 1-1-1 Itakura, Oura, Gunma, 374-0193, Japan
| | - Kohei Hosono
- Department of Aquatic Bioscience, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo, Tokyo, 113-8657, Japan
| | - Toyoji Kaneko
- Department of Aquatic Bioscience, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo, Tokyo, 113-8657, Japan
| | - Ruo-Dong Chen
- Institute of Cellular and Organismic Biology, Academia Sinica, Nankang, Taipei City, Taiwan, Republic of China
| | - Jay-Ron Lee
- Institute of Cellular and Organismic Biology, Academia Sinica, Nankang, Taipei City, Taiwan, Republic of China
| | - Yung-Che Tseng
- Institute of Cellular and Organismic Biology, Academia Sinica, Nankang, Taipei City, Taiwan, Republic of China
| | - Pung-Pung Hwang
- Institute of Cellular and Organismic Biology, Academia Sinica, Nankang, Taipei City, Taiwan, Republic of China
| | - Ritsuko Ohtani-Kaneko
- Department of Life Sciences, Toyo University, 1-1-1 Itakura, Oura, Gunma, 374-0193, Japan.
| |
Collapse
|
25
|
Liu PZ, Nusslock R. Exercise-Mediated Neurogenesis in the Hippocampus via BDNF. Front Neurosci 2018; 12:52. [PMID: 29467613 PMCID: PMC5808288 DOI: 10.3389/fnins.2018.00052] [Citation(s) in RCA: 302] [Impact Index Per Article: 43.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2017] [Accepted: 01/23/2018] [Indexed: 12/16/2022] Open
Abstract
Exercise is known to have numerous neuroprotective and cognitive benefits, especially pertaining to memory and learning related processes. One potential link connecting them is exercise-mediated hippocampal neurogenesis, in which new neurons are generated and incorporated into hippocampal circuits. The present review synthesizes the extant literature detailing the relationship between exercise and hippocampal neurogenesis, and identifies a key molecule mediating this process, brain-derived neurotrophic factor (BDNF). As a member of the neurotrophin family, BDNF regulates many of the processes within neurogenesis, such as differentiation and survival. Although much more is known about the direct role that exercise and BDNF have on hippocampal neurogenesis in rodents, their corresponding cognitive benefits in humans will also be discussed. Specifically, what is known about exercise-mediated hippocampal neurogenesis will be presented as it relates to BDNF to highlight the critical role that it plays. Due to the inaccessibility of the human brain, much less is known about the role BDNF plays in human hippocampal neurogenesis. Limitations and future areas of research with regards to human neurogenesis will thus be discussed, including indirect measures of neurogenesis and single nucleotide polymorphisms within the BDNF gene.
Collapse
Affiliation(s)
- Patrick Z. Liu
- Department of Psychology, Northwestern University, Evanston, IL, United States
| | | |
Collapse
|
26
|
Oosthuizen MK. From Mice to Mole-Rats: Species-Specific Modulation of Adult Hippocampal Neurogenesis. Front Neurosci 2017; 11:602. [PMID: 29163007 PMCID: PMC5670158 DOI: 10.3389/fnins.2017.00602] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Accepted: 10/16/2017] [Indexed: 11/23/2022] Open
Abstract
Rodent populations living in their natural environments have very diverse ecological and life history profiles that may differ substantially from that of conventional laboratory rodents. Free-living rodents show species-specific neurogenesis that are dependent on their unique biology and ecology. This perspective aims to illustrate the benefit of studying wild rodent species in conjunction with laboratory rodents. African mole-rats are discussed in terms of habitat complexity, social structures, and longevity. African mole-rats are a group of subterranean rodents, endemic to Africa, that show major differences in both intrinsic and extrinsic traits compared to the classical rodent models. Mole-rats exhibit a spectrum of sociality within a single family, ranging from solitary to eusocial. This continuum of sociality provides a platform for comparative testing of hypotheses. Indeed, species differences are apparent both in learning ability and hippocampal neurogenesis. In addition, social mole-rat species display a reproductive division of labor that also results in differential hippocampal neurogenesis, independent of age, offering further scope for comparison. In conclusion, it is evident that neurogenesis studies on conventional laboratory rodents are not necessarily representative, specifically because of a lack of diversity in life histories, uniform habitats, and low genetic variability. The observed level of adult neurogenesis in the dentate gyrus is the result of an intricate balance between many contributing factors, which appear to be specific to distinct groups of animals. The ultimate understanding of the functional and adaptive role of adult neurogenesis will involve research on both laboratory animals and natural rodent populations.
Collapse
Affiliation(s)
- Maria K Oosthuizen
- Department of Zoology and Entomology, University of Pretoria, Pretoria, South Africa
| |
Collapse
|
27
|
Mohr MA, DonCarlos LL, Sisk CL. Inhibiting Production of New Brain Cells during Puberty or Adulthood Blunts the Hormonally Induced Surge of Luteinizing Hormone in Female Rats. eNeuro 2017; 4:ENEURO.0133-17.2017. [PMID: 29098175 PMCID: PMC5666323 DOI: 10.1523/eneuro.0133-17.2017] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2017] [Revised: 10/09/2017] [Accepted: 10/16/2017] [Indexed: 01/15/2023] Open
Abstract
New cells are added during both puberty and adulthood to hypothalamic regions that govern reproduction, homeostasis, and social behaviors, yet the functions of these late-born cells remain elusive. Here, we pharmacologically inhibited cell proliferation in ventricular zones during puberty or in adulthood and determined subsequent effects on the hormone-induced surge of luteinizing hormone (LH) in female rats. Initial neuroanatomical analyses focused on verifying incorporation, activation, and pharmacological inhibition of pubertally or adult born cells in the anteroventral periventricular nucleus (AVPV) of the hypothalamus because of the essential role of the AVPV in triggering the preovulatory LH surge in females. We first showed that approximately half of the pubertally born AVPV cells are activated by estradiol plus progesterone (P) treatment, as demonstrated by Fos expression, and that approximately 10% of pubertally born AVPV cells express estrogen receptor alpha (ERα). Next, we found that mitotic inhibition through intracerebroventricular (ICV) administration of cytosine β-D-arabinofuranoside (AraC), whether during puberty or in adulthood, decreased the number of new cells added to the AVPV and the suprachiasmatic nucleus (SCN), and also blunted and delayed the hormone-induced LH surge. These studies do not prove, but are highly suggestive, that ongoing postnatal addition of new cells in periventricular brain regions, including the AVPV and SCN, may be important to the integrity of female reproduction.
Collapse
Affiliation(s)
- Margaret A. Mohr
- Neuroscience Program, Michigan State University, East Lansing, MI 48824
| | - Lydia L. DonCarlos
- Department of Cell and Molecular Physiology, Stritch School of Medicine, Loyola University Chicago, Maywood, IL 60153
| | - Cheryl L. Sisk
- Neuroscience Program, Michigan State University, East Lansing, MI 48824
| |
Collapse
|
28
|
Home sweet home: the neural stem cell niche throughout development and after injury. Cell Tissue Res 2017; 371:125-141. [PMID: 28776186 DOI: 10.1007/s00441-017-2658-0] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Accepted: 05/29/2017] [Indexed: 12/26/2022]
Abstract
Neural stem cells and their progeny reside in two distinct neurogenic niches within the mammalian brain: the subventricular zone and the dentate gyrus. The interplay between the neural stem cells and the niche in which they reside can have significant effects on cell kinetics and neurogenesis. A comprehensive understanding of the changes to the niche that occur through postnatal development and aging, as well as following injury, is relevant for developing therapeutics and interventions to promote neural repair. We discuss changes that occur within the neural stem and progenitor cell populations, the vasculature, extracellular matrix, microglia, and secreted proteins through aging which impact cell behavior within the neurogenic niches. We examine neural precursor cell and niche responses to injury in neonatal hypoxia-ischemia, juvenile cranial irradiation, and adult stroke. This review examines the interplay between the niche and stem cell behavior through aging and following injury as a means to understand intrinsic and extrinsic factors that regulate neurogenesis in vivo.
Collapse
|
29
|
Depression and Anxiety Scores Are Associated with Amygdala Volume in Cushing's Syndrome: Preliminary Study. BIOMED RESEARCH INTERNATIONAL 2017; 2017:2061935. [PMID: 28607927 PMCID: PMC5451775 DOI: 10.1155/2017/2061935] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Revised: 03/31/2017] [Accepted: 04/26/2017] [Indexed: 01/13/2023]
Abstract
Introduction Cushing's syndrome (CS) has repeatedly been associated with hippocampal volume reductions, while little information is available on the amygdala, another structure rich in glucocorticoid receptors. The aim of the study was to analyze amygdala volume in patients with CS and its relationship with anxiety, depression, and hormone levels. Material and Methods 39 CS patients (16 active and 23 patients in remission) and 39 healthy controls matched for age, sex, and education level completed anxiety (STAI) and depression tests (BDI-II) and underwent a 3 Tesla brain MRI and endocrine testing. Amygdala volumes were analysed with FreeSurfer software. Results Active CS patients had smaller right (but not left) amygdala volumes when compared to controls (P = 0.045). Left amygdala volumes negatively correlated with depression scores (r = −0.692, P = 0.003) and current anxiety state scores (r = −0.617, P = 0.011) in active CS patients and with anxiety trait scores (r = −0.440, P = 0.036) in patients in remission. No correlations were found between current ACTH, urinary free cortisol or blood cortisol levels, and amygdala volumes in either patient group. Conclusion Patients with active CS have a smaller right amygdala volume in comparison to controls, while left amygdala volumes are associated with mood state in both patient groups.
Collapse
|
30
|
Lieberwirth C, Pan Y, Liu Y, Zhang Z, Wang Z. Hippocampal adult neurogenesis: Its regulation and potential role in spatial learning and memory. Brain Res 2016; 1644:127-40. [PMID: 27174001 PMCID: PMC5064285 DOI: 10.1016/j.brainres.2016.05.015] [Citation(s) in RCA: 107] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Revised: 05/05/2016] [Accepted: 05/08/2016] [Indexed: 12/24/2022]
Abstract
Adult neurogenesis, defined here as progenitor cell division generating functionally integrated neurons in the adult brain, occurs within the hippocampus of numerous mammalian species including humans. The present review details various endogenous (e.g., neurotransmitters) and environmental (e.g., physical exercise) factors that have been shown to influence hippocampal adult neurogenesis. In addition, the potential involvement of adult-generated neurons in naturally-occurring spatial learning behavior is discussed by summarizing the literature focusing on traditional animal models (e.g., rats and mice), non-traditional animal models (e.g., tree shrews), as well as natural populations (e.g., chickadees and Siberian chipmunk).
Collapse
Affiliation(s)
| | - Yongliang Pan
- Program in Molecular and Translational Medicine, School of Medicine, Huzhou University, Huzhou 313000, PR China; State Key Laboratory of Integrated Management of Pest Insects and Rodents in Agriculture, Institute of Zoology, Chinese Academy of Sciences, Datun Road, Chaoyang District, Beijing 100101, PR China.
| | - Yan Liu
- Department of Psychology and Program in Neuroscience, Florida State University, Tallahassee, FL 32306-1270, USA
| | - Zhibin Zhang
- State Key Laboratory of Integrated Management of Pest Insects and Rodents in Agriculture, Institute of Zoology, Chinese Academy of Sciences, Datun Road, Chaoyang District, Beijing 100101, PR China
| | - Zuoxin Wang
- Department of Psychology and Program in Neuroscience, Florida State University, Tallahassee, FL 32306-1270, USA
| |
Collapse
|
31
|
Castilla-Ortega E, Serrano A, Blanco E, Araos P, Suárez J, Pavón FJ, Rodríguez de Fonseca F, Santín LJ. A place for the hippocampus in the cocaine addiction circuit: Potential roles for adult hippocampal neurogenesis. Neurosci Biobehav Rev 2016; 66:15-32. [PMID: 27118134 DOI: 10.1016/j.neubiorev.2016.03.030] [Citation(s) in RCA: 69] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2015] [Revised: 03/08/2016] [Accepted: 03/08/2016] [Indexed: 02/07/2023]
Abstract
Cocaine addiction is a chronic brain disease in which the drug seeking habits and profound cognitive, emotional and motivational alterations emerge from drug-induced neuroadaptations on a vulnerable brain. Therefore, a 'cocaine addiction brain circuit' has been described to explain this disorder. Studies in both cocaine patients and rodents reveal the hippocampus as a main node in the cocaine addiction circuit. The contribution of the hippocampus to cocaine craving and the associated memories is essential to understand the chronic relapsing nature of addiction, which is the main obstacle for the recovery. Interestingly, the hippocampus holds a particular form of plasticity that is rare in the adult brain: the ability to generate new functional neurons. There is an active scientific debate on the contributions of these new neurons to the addicted brain. This review focuses on the potential role(s) of adult hippocampal neurogenesis (AHN) in cocaine addiction. Although the current evidence primarily originates from animal research, these preclinical studies support AHN as a relevant component for the hippocampal effects of cocaine.
Collapse
Affiliation(s)
- Estela Castilla-Ortega
- Unidad de Gestión Clínica de Salud Mental, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de Málaga, Spain.
| | - Antonia Serrano
- Unidad de Gestión Clínica de Salud Mental, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de Málaga, Spain
| | - Eduardo Blanco
- Departament de Pedagogia i Psicologia, Facultat d'Educació, Psicologia i Treball Social, Universitat de Lleida, Spain
| | - Pedro Araos
- Unidad de Gestión Clínica de Salud Mental, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de Málaga, Spain
| | - Juan Suárez
- Unidad de Gestión Clínica de Salud Mental, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de Málaga, Spain
| | - Francisco J Pavón
- Unidad de Gestión Clínica de Salud Mental, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de Málaga, Spain
| | - Fernando Rodríguez de Fonseca
- Unidad de Gestión Clínica de Salud Mental, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de Málaga, Spain
| | - Luis J Santín
- Departamento de Psicobiología y Metodología de las Ciencias del Comportamiento, Instituto de Investigación Biomédica de Málaga (IBIMA), Facultad de Psicología, Universidad de Málaga, Spain.
| |
Collapse
|
32
|
Nascimento LFR, Souza GFP, Morari J, Barbosa GO, Solon C, Moura RF, Victório SC, Ignácio-Souza LM, Razolli DS, Carvalho HF, Velloso LA. n-3 Fatty Acids Induce Neurogenesis of Predominantly POMC-Expressing Cells in the Hypothalamus. Diabetes 2016; 65:673-86. [PMID: 26512023 DOI: 10.2337/db15-0008] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2015] [Accepted: 10/17/2015] [Indexed: 11/13/2022]
Abstract
Apoptosis of hypothalamic neurons is believed to play an important role in the development and perpetuation of obesity. Similar to the hippocampus, the hypothalamus presents constitutive and stimulated neurogenesis, suggesting that obesity-associated hypothalamic dysfunction can be repaired. Here, we explored the hypothesis that n-3 polyunsaturated fatty acids (PUFAs) induce hypothalamic neurogenesis. Both in the diet and injected directly into the hypothalamus, PUFAs were capable of increasing hypothalamic neurogenesis to levels similar or superior to the effect of brain-derived neurotrophic factor (BDNF). Most of the neurogenic activity induced by PUFAs resulted in increased numbers of proopiomelanocortin but not NPY neurons and was accompanied by increased expression of BDNF and G-protein-coupled receptor 40 (GPR40). The inhibition of GPR40 was capable of reducing the neurogenic effect of a PUFA, while the inhibition of BDNF resulted in the reduction of global hypothalamic cell. Thus, PUFAs emerge as a potential dietary approach to correct obesity-associated hypothalamic neuronal loss.
Collapse
Affiliation(s)
| | | | - Joseane Morari
- Laboratory of Cell Signaling, University of Campinas, Campinas, Brazil
| | | | - Carina Solon
- Laboratory of Cell Signaling, University of Campinas, Campinas, Brazil
| | - Rodrigo F Moura
- Laboratory of Cell Signaling, University of Campinas, Campinas, Brazil
| | - Sheila C Victório
- Laboratory of Cell Signaling, University of Campinas, Campinas, Brazil
| | | | - Daniela S Razolli
- Laboratory of Cell Signaling, University of Campinas, Campinas, Brazil
| | | | - Lício A Velloso
- Laboratory of Cell Signaling, University of Campinas, Campinas, Brazil
| |
Collapse
|
33
|
Gender Differences in the Neurobiology of Anxiety: Focus on Adult Hippocampal Neurogenesis. Neural Plast 2016; 2016:5026713. [PMID: 26885403 PMCID: PMC4738969 DOI: 10.1155/2016/5026713] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2015] [Revised: 11/30/2015] [Accepted: 12/06/2015] [Indexed: 12/14/2022] Open
Abstract
Although the literature reports a higher incidence of anxiety disorders in women, the majority of basic research has focused on male rodents, thus resulting in a lack of knowledge on the neurobiology of anxiety in females. Bridging this gap is crucial for the design of effective translational interventions in women. One of the key brain mechanisms likely to regulate anxious behavior is adult hippocampal neurogenesis (AHN). This review paper aims to discuss the evidence on the differences between male and female rodents with regard to anxiety-related behavior and physiology, with a special focus on AHN. The differences between male and female physiologies are greatly influenced by hormonal differences. Gonadal hormones and their fluctuations during the estrous cycle have often been identified as agents responsible for sexual dimorphism in behavior and AHN. During sexual maturity, hormone levels fluctuate cyclically in females more than in males, increasing the stress response and the susceptibility to anxiety. It is therefore of great importance that future research investigates anxiety and other neurophysiological aspects in the female model, so that results can be more accurately applicable to the female population.
Collapse
|
34
|
Pan Y, Liu Y, Lieberwirth C, Zhang Z, Wang Z. Species differences in behavior and cell proliferation/survival in the adult brains of female meadow and prairie voles. Neuroscience 2015; 315:259-70. [PMID: 26708743 DOI: 10.1016/j.neuroscience.2015.12.026] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Revised: 12/10/2015] [Accepted: 12/14/2015] [Indexed: 02/03/2023]
Abstract
Microtine rodents display diverse patterns of social organization and behaviors, and thus provide a useful model for studying the effects of the social environment on physiology and behavior. The current study compared the species differences and the effects of oxytocin (OT) on anxiety-like, social affiliation, and social recognition behaviors in female meadow voles (Microtus pennsylvanicus) and prairie voles (Microtus ochrogaster). Furthermore, cell proliferation and survival in the brains of adult female meadow and prairie voles were compared. We found that female meadow voles displayed a higher level of anxiety-like behavior but lower levels of social affiliation and social recognition compared to female prairie voles. In addition, meadow voles showed lower levels of cell proliferation (measured by Ki67 staining) and cell survival (measured by BrdU staining) in the ventromedial hypothalamus (VMH) and amygdala (AMY), but not the dentate gyrus of the hippocampus (DG), than prairie voles. Interestingly, the numbers of new cells in the VMH and AMY, but not DG, also correlated with anxiety-like, social affiliation, and social recognition behaviors in a brain region-specific manner. Finally, central OT treatment (200 ng/kg, icv) did not lead to changes in behavior or cell proliferation/survival in the brain. Together, these data indicate a potential role of cell proliferation/survival in selected brain areas on different behaviors between vole species with distinct life strategies.
Collapse
Affiliation(s)
- Y Pan
- Program in Molecular and Translational Medicine, School of Medicine, Huzhou University, Huzhou 313000, PR China; State Key Laboratory of Integrated Management of Pest Insects and Rodents in Agriculture, Institute of Zoology, Chinese Academy of Sciences, Datun Road, Chaoyang District, Beijing 100101, PR China; Department of Psychology and Program in Neuroscience, Florida State University, Tallahassee, FL 32306-1270, USA
| | - Y Liu
- Department of Psychology and Program in Neuroscience, Florida State University, Tallahassee, FL 32306-1270, USA
| | - C Lieberwirth
- Department of Psychology and Program in Neuroscience, Florida State University, Tallahassee, FL 32306-1270, USA; Behavioral Science Department, Utah Valley University, Orem, UT, 84058, USA
| | - Z Zhang
- State Key Laboratory of Integrated Management of Pest Insects and Rodents in Agriculture, Institute of Zoology, Chinese Academy of Sciences, Datun Road, Chaoyang District, Beijing 100101, PR China
| | - Z Wang
- Department of Psychology and Program in Neuroscience, Florida State University, Tallahassee, FL 32306-1270, USA.
| |
Collapse
|
35
|
Social isolation increases cell proliferation in male and cell survival in female California mice (Peromyscus californicus). Physiol Behav 2015; 151:570-6. [DOI: 10.1016/j.physbeh.2015.08.039] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2014] [Revised: 05/28/2015] [Accepted: 08/29/2015] [Indexed: 11/23/2022]
|
36
|
Wang L, Wang Y, Du H, Jiang Y, Tang Z, Liu H, Xiang H, Xiao H. Impact of ER520, a candidate of selective estrogen receptor modulators, on in vitro cell growth, migration, invasion, angiogenesis and in vivo tumor xenograft of human breast cancer cells. Cancer Chemother Pharmacol 2015; 76:1247-57. [PMID: 26464351 DOI: 10.1007/s00280-015-2838-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2015] [Accepted: 07/27/2015] [Indexed: 12/11/2022]
|
37
|
Kinch CD, Ibhazehiebo K, Jeong JH, Habibi HR, Kurrasch DM. Low-dose exposure to bisphenol A and replacement bisphenol S induces precocious hypothalamic neurogenesis in embryonic zebrafish. Proc Natl Acad Sci U S A 2015; 112:1475-80. [PMID: 25583509 PMCID: PMC4321238 DOI: 10.1073/pnas.1417731112] [Citation(s) in RCA: 356] [Impact Index Per Article: 35.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Bisphenol A (BPA), a ubiquitous endocrine disruptor that is present in many household products, has been linked to obesity, cancer, and, most relevant here, childhood neurological disorders such as anxiety and hyperactivity. However, how BPA exposure translates into these neurodevelopmental disorders remains poorly understood. Here, we used zebrafish to link BPA mechanistically to disease etiology. Strikingly, treatment of embryonic zebrafish with very low-dose BPA (0.0068 μM, 1,000-fold lower than the accepted human daily exposure) and bisphenol S (BPS), a common analog used in BPA-free products, resulted in 180% and 240% increases, respectively, in neuronal birth (neurogenesis) within the hypothalamus, a highly conserved brain region involved in hyperactivity. Furthermore, restricted BPA/BPS exposure specifically during the neurogenic window caused later hyperactive behaviors in zebrafish larvae. Unexpectedly, we show that BPA-mediated precocious neurogenesis and the concomitant behavioral phenotype were not dependent on predicted estrogen receptors but relied on androgen receptor-mediated up-regulation of aromatase. Although human epidemiological results are still emerging, an association between high maternal urinary BPA during gestation and hyperactivity and other behavioral disturbances in the child has been suggested. Our studies here provide mechanistic support that the neurogenic period indeed may be a window of vulnerability and uncovers previously unexplored avenues of research into how endocrine disruptors might perturb early brain development. Furthermore, our results show that BPA-free products are not necessarily safer and support the removal of all bisphenols from consumer merchandise.
Collapse
Affiliation(s)
- Cassandra D Kinch
- Departments of Biological Sciences and Medical Genetics and Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, Canada T2N 4N1
| | - Kingsley Ibhazehiebo
- Medical Genetics and Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, Canada T2N 4N1
| | - Joo-Hyun Jeong
- Medical Genetics and Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, Canada T2N 4N1
| | | | - Deborah M Kurrasch
- Medical Genetics and Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, Canada T2N 4N1
| |
Collapse
|
38
|
Wang P, Liu C, Liu L, Zhang X, Ren B, Li B. The Antidepressant-like Effects of Estrogen-mediated Ghrelin. Curr Neuropharmacol 2015; 13:524-35. [PMID: 26412072 PMCID: PMC4790402 DOI: 10.2174/1570159x1304150831120650] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2014] [Revised: 01/13/2015] [Accepted: 01/24/2015] [Indexed: 12/19/2022] Open
Abstract
Ghrelin, one of the brain-gut peptides, stimulates food-intake. Recently, ghrelin has also shown to play an important role in depression treatment. However, the mechanism of ghrelin's antidepressant-like actions is unknown. On the other hand, sex differences in depression, and the fluctuation of estrogens secretion have been proved to play a key role in depression. It has been reported that women have higher level of ghrelin expression, and ghrelin can stimulate estrogen secretion while estrogen acts as a positive feedback mechanism to up-regulate ghrelin level. Ghrelin may be a potential regulator of reproductive function, and estrogen may have additional effect in ghrelin's antidepressantlike actions. In this review, we summarize antidepressant-like effects of ghrelin and estrogen in basic and clinical studies, and provide new insight on ghrelin's effect in depression.
Collapse
Affiliation(s)
- Pu Wang
- Life Sciences institute, Northeast Normal University, Changchun, China 130024
| | - Changhong Liu
- Life Sciences institute, Northeast Normal University, Changchun, China 130024
| | - Lei Liu
- Life Sciences institute, Northeast Normal University, Changchun, China 130024
| | - Xingyi Zhang
- Jilin provincial key
laboratory on molecular and chemical genetic, Second hospital of Jilin University, Changchun
130024, China
| | - Bingzhong Ren
- Life Sciences institute, Northeast Normal University, Changchun, China 130024
| | - Bingjin Li
- Life Sciences institute, Northeast Normal University, Changchun, China 130024
- Jilin provincial key
laboratory on molecular and chemical genetic, Second hospital of Jilin University, Changchun
130024, China
| |
Collapse
|
39
|
Impact of puberty on the evolution of cerebral perfusion during adolescence. Proc Natl Acad Sci U S A 2014; 111:8643-8. [PMID: 24912164 DOI: 10.1073/pnas.1400178111] [Citation(s) in RCA: 153] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Puberty is the defining biological process of adolescent development, yet its effects on fundamental properties of brain physiology such as cerebral blood flow (CBF) have never been investigated. Capitalizing on a sample of 922 youths ages 8-22 y imaged using arterial spin labeled MRI as part of the Philadelphia Neurodevelopmental Cohort, we studied normative developmental differences in cerebral perfusion in males and females, as well as specific associations between puberty and CBF. Males and females had conspicuously divergent nonlinear trajectories in CBF evolution with development as modeled by penalized splines. Seventeen brain regions, including hubs of the executive and default mode networks, showed a robust nonlinear age-by-sex interaction that surpassed Bonferroni correction. Notably, within these regions the decline in CBF was similar between males and females in early puberty and only diverged in midpuberty, with CBF actually increasing in females. Taken together, these results delineate sex-specific growth curves for CBF during youth and for the first time to our knowledge link such differential patterns of development to the effects of puberty.
Collapse
|
40
|
Pan Y, Li M, Lieberwirth C, Wang Z, Zhang Z. Social defeat and subsequent isolation housing affect behavior as well as cell proliferation and cell survival in the brains of male greater long-tailed hamsters. Neuroscience 2014; 265:226-37. [DOI: 10.1016/j.neuroscience.2014.01.056] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2013] [Revised: 01/28/2014] [Accepted: 01/29/2014] [Indexed: 02/08/2023]
|
41
|
Liu Y, Lieberwirth C, Jia X, Curtis JT, Meredith M, Wang ZX. Chemosensory cues affect amygdaloid neurogenesis and alter behaviors in the socially monogamous prairie vole. Eur J Neurosci 2014; 39:1632-41. [PMID: 24641515 DOI: 10.1111/ejn.12531] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2013] [Revised: 01/24/2014] [Accepted: 01/28/2014] [Indexed: 12/11/2022]
Abstract
The current study examined the effects of pheromonal exposure on adult neurogenesis and revealed the role of the olfactory pathways on adult neurogenesis and behavior in the socially monogamous prairie vole (Microtus ochrogaster). Subjects were injected with a cell proliferation marker [5-bromo-2'-deoxyuridine (BrdU)] and then exposed to their own soiled bedding or bedding soiled by a same- or opposite-sex conspecific. Exposure to opposite-sex bedding increased BrdU labeling in the amygdala (AMY), but not the dentate gyrus (DG), of female, but not male, voles, indicating a sex-, stimulus-, and brain region-specific effect. The removal of the main olfactory bulbs or lesioning of the vomeronasal organ (VNOX) in females reduced BrdU labeling in the AMY and DG, and inhibited the male bedding-induced BrdU labeling in the AMY, revealing the importance of an intact olfactory pathway for amygdaloid neurogenesis. VNOX increased anxiety-like behavior and altered social preference, but it did not affect social recognition memory in female voles. VNOX also reduced the percentage of BrdU-labeled cells that co-expressed the neuronal marker TuJ1 in the AMY, but not the DG. Together, our data indicate the importance of the olfactory pathway in mediating brain plasticity in the limbic system as well as its role in behavior.
Collapse
Affiliation(s)
- Y Liu
- Department of Psychology, Florida State University, 1107 W. Call Street, Tallahassee, FL, 32306, USA; Program in Neuroscience, Florida State University, 1107 W. Call Street, Tallahassee, FL, 32306, USA
| | | | | | | | | | | |
Collapse
|
42
|
Satterthwaite TD, Vandekar S, Wolf DH, Ruparel K, Roalf DR, Jackson C, Elliott MA, Bilker WB, Calkins ME, Prabhakaran K, Davatzikos C, Hakonarson H, Gur RE, Gur RC. Sex differences in the effect of puberty on hippocampal morphology. J Am Acad Child Adolesc Psychiatry 2014; 53:341-50.e1. [PMID: 24565361 PMCID: PMC3935178 DOI: 10.1016/j.jaac.2013.12.002] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2013] [Revised: 10/20/2013] [Accepted: 12/17/2013] [Indexed: 01/07/2023]
Abstract
OBJECTIVE Puberty is the defining process of adolescence, and is accompanied by divergent trajectories of behavior and cognition for males and females. Here we examine whether sex differences exist in the effect of puberty on the morphology of the hippocampus and amygdala. METHOD T1-weighted structural neuroimaging was performed in a sample of 524 pre- or postpubertal individuals ages 10 to 22 years. Hippocampal and amygdala volume and shape were quantified using the Functional Magnetic Resonance Imaging of the Brain (FMRIB) Software Library (FSL) FIRST procedure and scaled by intracranial volume. The effects on regional volume of age, sex, puberty, and their interactions were examined using linear regression. Postpubertal sex differences were examined using a vertex analysis. RESULTS Prepubertal males and females had similar hippocampal volumes, whereas postpubertal females had significantly larger bilateral hippocampi, resulting in a significant puberty-by-sex interaction even when controlling for age and age-by-sex. This effect was regionally specific and was not apparent in the amygdala. Vertex analysis revealed that postpubertal differences were most prominent in the lateral aspect of the hippocampus bilaterally, corresponding to the CA1 subfield. CONCLUSIONS These results establish that there are regionally specific sex differences in the effect of puberty on the hippocampus. These findings are relevant for the understanding of psychiatric disorders that have both hippocampal dysfunction and prominent gender disparities during adolescence.
Collapse
Affiliation(s)
| | | | - Daniel H Wolf
- Perelman School of Medicine, University of Pennsylvania
| | - Kosha Ruparel
- Perelman School of Medicine, University of Pennsylvania
| | - David R Roalf
- Perelman School of Medicine, University of Pennsylvania
| | - Chad Jackson
- Perelman School of Medicine, University of Pennsylvania
| | | | | | | | | | | | - Hakon Hakonarson
- Center for Applied Genomics, Children's Hospital of Philadelphia
| | - Raquel E Gur
- Perelman School of Medicine, University of Pennsylvania
| | - Ruben C Gur
- Perelman School of Medicine, University of Pennsylvania; Philadelphia Veterans Administration Medical Center
| |
Collapse
|
43
|
Oyarce K, Bongarzone ER, Nualart F. Unconventional Neurogenic Niches and Neurogenesis Modulation by Vitamins. ACTA ACUST UNITED AC 2014. [PMID: 26203401 DOI: 10.4172/2157-7633.1000184] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Although the generation of new neurons occurs in adult mammals, it has been classically described in two defined regions of the brain denominated neurogenic niches: the subventricular zone of the lateral ventricles and the subgranular zone of the dentate gyrus. In these regions, neural stem cells give rise to new neurons and glia, which functionally integrate into the existing circuits under physiological conditions. However, accumulating evidence indicates the presence of neurogenic potential in other brain regions, from which multipotent precursors can be isolated and differentiated in vitro. In some of these regions, neuron generation occurs at low levels; however, the addition of growth factors, hormones or other signaling molecules increases the proliferation and differentiation of precursor cells. In addition, vitamins, which are micronutrients necessary for normal brain development, and whose deficiency produces neurological impairments, have a regulatory effect on neural stem cells in vitro and in vivo. In the present review, we will describe the progress that has been achieved in determining the neurogenic potential in other regions, known as unconventional niches, as well as the characteristics of the neural stem cells described for each region. Finally, we will revisit the roles of commonly known vitamins as modulators of precursor cell proliferation and differentiation, and their role in the complex and tight molecular signaling that impacts these neurogenic niches.
Collapse
Affiliation(s)
- Karina Oyarce
- Laboratory of Neurobiology and Stem Cells, Center for Advanced Microscopy CMA BIO BIO, Concepcion University, Concepción, Chile
| | - Ernesto R Bongarzone
- Department of Anatomy and Cell Biology, College of Medicine, University of Illinois Chicago, USA
| | - Francisco Nualart
- Laboratory of Neurobiology and Stem Cells, Center for Advanced Microscopy CMA BIO BIO, Concepcion University, Concepción, Chile
| |
Collapse
|
44
|
de Vries GJ, Fields CT, Peters NV, Whylings J, Paul MJ. Sensitive periods for hormonal programming of the brain. Curr Top Behav Neurosci 2014; 16:79-108. [PMID: 24549723 DOI: 10.1007/7854_2014_286] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
During sensitive periods, information from the external and internal environment that occurs during particular phases of development is relayed to the brain to program neural development. Hormones play a central role in this process. In this review, we first discuss sexual differentiation of the brain as an example of hormonal programming. Using sexual differentiation, we define sensitive periods, review cellular and molecular processes that can explain their restricted temporal window, and discuss challenges in determining the precise timing of the temporal window. We then briefly review programming effects of other hormonal systems and discuss how programming of these systems interact with sexual differentiation.
Collapse
Affiliation(s)
- Geert J de Vries
- Neuroscience Institute, Georgia State University, PO Box 5030, Atlanta, GA, 30302-5030, USA,
| | | | | | | | | |
Collapse
|
45
|
Ruan L, Lau BWM, Wang J, Huang L, Zhuge Q, Wang B, Jin K, So KF. Neurogenesis in neurological and psychiatric diseases and brain injury: from bench to bedside. Prog Neurobiol 2013; 115:116-37. [PMID: 24384539 DOI: 10.1016/j.pneurobio.2013.12.006] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2013] [Revised: 12/08/2013] [Accepted: 12/12/2013] [Indexed: 02/08/2023]
Abstract
Researchers who have uncovered the presence of stem cells in an adult's central nervous system have not only challenged the dogma that new neurons cannot be generated during adulthood, but also shed light on the etiology and disease mechanisms underlying many neurological and psychiatric disorders. Brain trauma, neurodegenerative diseases, and psychiatric disorders pose enormous burdens at both personal and societal levels. Although medications for these disorders are widely used, the treatment mechanisms underlying the illnesses remain largely elusive. In the past decade, an increasing amount of evidence indicate that adult neurogenesis (i.e. generating new CNS neurons during adulthood) may be involved in the pathology of different CNS disorders, and thus neurogenesis may be a potential target area for treatments. Although new neurons were shown to be a major player in mediating treatment efficacy of neurological and psychotropic drugs on cognitive functions, it is still debatable if the altered production of new neurons can cause the disorders. This review hence seeks to discuss pre and current clinical studies that demonstrate the functional impact adult neurogenesis have on neurological and psychiatric illnesses while examining the related underlying disease mechanisms.
Collapse
Affiliation(s)
- Linhui Ruan
- Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, First Affiliated Hospital, Wenzhou Medical University, Wenzhou, China; Department of Pharmacology and Neuroscience, University of North Texas Health Science Center at Fort Worth, TX 76107, USA.
| | - Benson Wui-Man Lau
- Department of Rehabilitation Science, The Hong Kong Polytechnic University, Hong Kong, PR China
| | - Jixian Wang
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center at Fort Worth, TX 76107, USA
| | - Lijie Huang
- Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, First Affiliated Hospital, Wenzhou Medical University, Wenzhou, China; Department of Pharmacology and Neuroscience, University of North Texas Health Science Center at Fort Worth, TX 76107, USA
| | - Qichuan Zhuge
- Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, First Affiliated Hospital, Wenzhou Medical University, Wenzhou, China
| | - Brian Wang
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center at Fort Worth, TX 76107, USA
| | - Kunlin Jin
- Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, First Affiliated Hospital, Wenzhou Medical University, Wenzhou, China; Department of Pharmacology and Neuroscience, University of North Texas Health Science Center at Fort Worth, TX 76107, USA.
| | - Kwok-Fai So
- Department of Ophthalmology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, PR China; The State Key Laboratory of Brain and Cognitive Sciences, The University of Hong Kong, Pokfulam, Hong Kong SAR, PR China; Research Centre of Heart, Brain, Hormone and Healthy Aging, Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, PR China; GMH Institute of CNS Regeneration, Jinan University, Guangzhou, PR China.
| |
Collapse
|
46
|
Sorteni C, Clavenzani P, De Giorgio R, Portnoy O, Sirri R, Mordenti O, Di Biase A, Parmeggiani A, Menconi V, Chiocchetti R. Enteric neuroplasticity in seawater-adapted European eel (Anguilla anguilla). J Anat 2013; 224:180-91. [PMID: 24433383 DOI: 10.1111/joa.12131] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/26/2013] [Indexed: 12/01/2022] Open
Abstract
European eels live most of their lives in freshwater until spawning migration to the Sargasso Sea. During seawater adaptation, eels modify their physiology, and their digestive system adapts to the new environment, drinking salt water to compensate for the continuous water loss. In that period, eels stop feeding until spawning. Thus, the eel represents a unique model to understand the adaptive changes of the enteric nervous system (ENS) to modified salinity and starvation. To this purpose, we assessed and compared the enteric neuronal density in the cranial portion of the intestine of freshwater eels (control), lagoon eels captured in brackish water before their migration to the Sargasso Sea (T0), and starved seawater eels hormonally induced to sexual maturity (T18; 18 weeks of starvation and treatment with standardized carp pituitary extract). Furthermore, we analyzed the modification of intestinal neuronal density of hormonally untreated eels during prolonged starvation (10 weeks) in seawater and freshwater. The density of myenteric (MP) and submucosal plexus (SMP) HuC/D-immunoreactive (Hu-IR) neurons was assessed in wholemount preparations and cryosections. The number of MP and SMP HuC/D-IR neurons progressively increased from the freshwater to the salty water habitat (control > T0 > T18; P < 0.05). Compared with freshwater eels, the number of MP and SMP HuC/D-IR neurons significantly increased (P < 0.05) in the intestine of starved untreated salt water eels. In conclusion, high salinity evokes enteric neuroplasticity as indicated by the increasing number of HuC/D-IR MP and SMP neurons, a mechanism likely contributing to maintaining the body homeostasis of this fish in extreme conditions.
Collapse
Affiliation(s)
- C Sorteni
- Department of Veterinary Medical Science (UNI EN ISO 9001:2008), University of Bologna, Bologna, Italy; Centro interdipartimentale di ricerca sull'alimentazione umana, University of Bologna, Bologna, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Cheng MF. Hypothalamic neurogenesis in the adult brain. Front Neuroendocrinol 2013; 34:167-78. [PMID: 23684668 DOI: 10.1016/j.yfrne.2013.05.001] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2012] [Revised: 04/30/2013] [Accepted: 05/03/2013] [Indexed: 12/19/2022]
Abstract
Adult-born new neurons are continuously added to the hippocampus and the olfactory bulb to serve aspects of learning and perceptual functions. Recent evidence establishes a third neurogenic niche in the ventral hypothalamic parenchyma surrounding the third ventricle that ensures the plasticity of specific brain circuits to stabilize physiological functions such as the energy-balance regulatory system. Hypothalamic lesion studies have demonstrated that regions associated with reproduction-related functions are also capable of recruiting newborn neurons to restore physiological functions and courtship behavior. Induced by lesion or other stimulation, elevated neurotrophic factors trigger neurogenic cascades that contribute to remodeling of certain neural circuits to meet specific transient functions. This insight raises the possibility that event-specific changes, such as increased GnRH, may be mediated by courtship-sensitive neurotrophic factors. We will discuss the potentially integral and ubiquitous roles of neurogenesis in physiological and biological phenomena, roles that await future experimental exploration.
Collapse
Affiliation(s)
- Mei-Fang Cheng
- Department of Psychology, Rutgers University, 101 Warren Street, Newark, NJ, USA.
| |
Collapse
|
48
|
|
49
|
Maruska KP, Carpenter RE, Fernald RD. Characterization of cell proliferation throughout the brain of the African cichlid fish Astatotilapia burtoni and its regulation by social status. J Comp Neurol 2013; 520:3471-91. [PMID: 22431175 DOI: 10.1002/cne.23100] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
New cells are added in the brains of all adult vertebrates, but fishes have some of the greatest potential for neurogenesis and gliogenesis among all taxa, partly due to their indeterminate growth. Little is known, however, about how social interactions influence cell proliferation in the brain of these fishes that comprise the largest group of vertebrates. We used 5-bromo-2'-deoxyuridine (BrdU) to identify and localize proliferation zones in the telencephalon, diencephalon, mesencephalon, and rhombencephalon that were primarily associated with ventricular surfaces in the brain of the African cichlid fish Astatotilapia burtoni. Cell migration was evident in some regions by 1 day post injection, and many newborn cells coexpressed the neuronal marker HuC/D at 30 days, suggesting they had differentiated into neurons. To test the hypothesis that social status and perception of an opportunity to rise in rank influenced cell proliferation, we compared numbers of BrdU-labeled cells in multiple brain nuclei among fish of different social status. Socially suppressed subordinate males had the lowest numbers of proliferating cells in all brain regions examined, but males that were given an opportunity to rise in status had higher cell proliferation rates within 1 day, suggesting rapid upregulation of brain mitotic activity associated with this social transition. Furthermore, socially isolated dominant males had similar numbers of BrdU-labeled cells compared with dominant males that were housed in a socially rich environment, suggesting that isolation has little effect on proliferation and that reduced proliferation in subordinates is a result of the social subordination. These results suggest that A. burtoni will be a useful model to analyze the mechanisms of socially induced neurogenesis in vertebrates.
Collapse
Affiliation(s)
- Karen P Maruska
- Department of Biology, Stanford University, Stanford, California 94305, USA.
| | | | | |
Collapse
|
50
|
Pubertally born neurons and glia are functionally integrated into limbic and hypothalamic circuits of the male Syrian hamster. Proc Natl Acad Sci U S A 2013; 110:4792-7. [PMID: 23460698 DOI: 10.1073/pnas.1219443110] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
During puberty, the brain goes through extensive remodeling, involving the addition of new neurons and glia to brain regions beyond the canonical neurogenic regions (i.e., dentate gyrus and olfactory bulb), including limbic and hypothalamic cell groups associated with sex-typical behavior. Whether these pubertally born cells become functionally integrated into neural circuits remains unknown. To address this question, we gave male Syrian hamsters daily injections of the cell birthdate marker bromodeoxyuridine throughout puberty (postnatal day 28-49). Half of the animals were housed in enriched environments with access to a running wheel to determine whether enrichment increased the survival of pubertally born cells compared with the control environment. At 4 wk after the last BrdU injection, animals were allowed to interact with a receptive female and were then killed 1 h later. Triple-label immunofluorescence for BrdU, the mature neuron marker neuronal nuclear antigen, and the astrocytic marker glial fibrillary acidic protein revealed that a proportion of pubertally born cells in the medial preoptic area, arcuate nucleus, and medial amygdala differentiate into either mature neurons or astrocytes. Double-label immunofluorescence for BrdU and the protein Fos revealed that a subset of pubertally born cells in these regions is activated during sociosexual behavior, indicative of their functional incorporation into neural circuits. Enrichment affected the survival and activation of pubertally born cells in a brain region-specific manner. These results demonstrate that pubertally born cells located outside of the traditional neurogenic regions differentiate into neurons and glia and become functionally incorporated into neural circuits that subserve sex-typical behaviors.
Collapse
|