1
|
Just N, Chevillard PM, Batailler M, Dubois JP, Vaudin P, Pillon D, Migaud M. Multiparametric MR Evaluation of the Photoperiodic Regulation of Hypothalamic Structures in Sheep. Neuroscience 2023; 535:142-157. [PMID: 37913859 DOI: 10.1016/j.neuroscience.2023.10.022] [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: 08/25/2023] [Revised: 10/16/2023] [Accepted: 10/24/2023] [Indexed: 11/03/2023]
Abstract
Most organisms on earth, humans included, have developed strategies to cope with environmental day-night and seasonal cycles to survive. For most of them, their physiological and behavioral functions, including the reproductive function, are synchronized with the annual changes of day length, to ensure winter survival and subsequent reproductive success in the following spring. Sheep are sensitive to photoperiod, which also regulates natural adult neurogenesis in their hypothalamus. We postulate that the ovine model represents a good alternative to study the functional and metabolic changes occurring in response to photoperiodic changes in hypothalamic structures of the brain. Here, the impact of the photoperiod on the neurovascular coupling and the metabolism of the hypothalamic structures was investigated at 3T using BOLD fMRI, perfusion-MRI and proton magnetic resonance spectroscopy (1H-MRS). A longitudinal study involving 8 ewes was conducted during long days (LD) and short days (SD) revealing significant BOLD, rCBV and metabolic changes in hypothalamic structures of the ewe brain between LD and SD. More specifically, the transition between LD and SD revealed negative BOLD responses to hypercapnia at the beginning of SD period followed by significant increases in BOLD, rCBV, Glx and tNAA concentrations towards the end of the SD period. These observations suggest longitudinal mechanisms promoting the proliferation and differentiation of neural stem cells within the hypothalamic niche of breeding ewes. We conclude that multiparametric MRI studies including 1H-MRS could be promising non-invasive translational techniques to investigate the existence of natural adult neurogenesis in-vivo in gyrencephalic brains.
Collapse
Affiliation(s)
- Nathalie Just
- INRAE Centre Val de Loire, UMR Physiologie de la Reproduction et des Comportements CNRS, IFCE, INRAE, Université de Tours, 37380 Nouzilly France; Danish Research Centre for Magnetic Resonance (DRCMR), Hvidovre, Denmark.
| | - Pierre Marie Chevillard
- INRAE Centre Val de Loire, UMR Physiologie de la Reproduction et des Comportements CNRS, IFCE, INRAE, Université de Tours, 37380 Nouzilly France
| | - Martine Batailler
- INRAE Centre Val de Loire, UMR Physiologie de la Reproduction et des Comportements CNRS, IFCE, INRAE, Université de Tours, 37380 Nouzilly France
| | - Jean-Philippe Dubois
- INRAE Centre Val de Loire, UMR Physiologie de la Reproduction et des Comportements CNRS, IFCE, INRAE, Université de Tours, 37380 Nouzilly France
| | - Pascal Vaudin
- INRAE Centre Val de Loire, UMR Physiologie de la Reproduction et des Comportements CNRS, IFCE, INRAE, Université de Tours, 37380 Nouzilly France
| | - Delphine Pillon
- INRAE Centre Val de Loire, UMR Physiologie de la Reproduction et des Comportements CNRS, IFCE, INRAE, Université de Tours, 37380 Nouzilly France
| | - Martine Migaud
- INRAE Centre Val de Loire, UMR Physiologie de la Reproduction et des Comportements CNRS, IFCE, INRAE, Université de Tours, 37380 Nouzilly France
| |
Collapse
|
2
|
Bonfanti L, Couillard-Després S. Neuron and Brain Maturation 2.0. Int J Mol Sci 2023; 24:17113. [PMID: 38069434 PMCID: PMC10707153 DOI: 10.3390/ijms242317113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Accepted: 11/23/2023] [Indexed: 12/18/2023] Open
Abstract
The mammalian central nervous system (CNS) is built up during embryogenesis by neural stem cells located in the periventricular germinal layers which undergo multiple division cycles [...].
Collapse
Affiliation(s)
- Luca Bonfanti
- Neuroscience Institute Cavalieri Ottolenghi (NICO), 10043 Orbassano, Italy
- Department of Veterinary Sciences, University of Turin, 10095 Torino, Italy
| | - Sébastien Couillard-Després
- Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Paracelsus Medical University, 5020 Salzburg, Austria
- Institute of Experimental Neuroregeneration, Paracelsus Medical University, 5020 Salzburg, Austria
- Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria
| |
Collapse
|
3
|
Faykoo-Martinez M, Collins T, Peragine D, Malik M, Javed F, Kolisnyk M, Ziolkowski J, Jeewa I, Cheng AH, Lowden C, Mascarenhas B, Cheng HYM, Holmes MM. Protracted neuronal maturation in a long-lived, highly social rodent. PLoS One 2022; 17:e0273098. [PMID: 36107951 PMCID: PMC9477366 DOI: 10.1371/journal.pone.0273098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Accepted: 08/02/2022] [Indexed: 11/19/2022] Open
Abstract
Naked mole-rats are a long-lived rodent species (current lifespan >37 years) and an increasingly popular biomedical model. Naked mole-rats exhibit neuroplasticity across their long lifespan. Previous studies have begun to investigate their neurogenic patterns. Here, we test the hypothesis that neuronal maturation is extended in this long-lived rodent. We characterize cell proliferation and neuronal maturation in established rodent neurogenic regions over 12 months following seven days of consecutive BrdU injection. Given that naked mole-rats are eusocial (high reproductive skew where only a few socially-dominant individuals reproduce), we also looked at proliferation in brain regions relevant to the social-decision making network. Finally, we measured co-expression of EdU (newly-born cells), DCX (immature neuron marker), and NeuN (mature neuron marker) to assess the timeline of neuronal maturation in adult naked mole-rats. This work reaffirms the subventricular zone as the main source of adult cell proliferation and suggests conservation of the rostral migratory stream in this species. Our profiling of socially-relevant brain regions suggests that future work which manipulates environmental context can unveil how newly-born cells integrate into circuitry and facilitate adult neuroplasticity. We also find naked mole-rat neuronal maturation sits at the intersection of rodents and long-lived, non-rodent species: while neurons can mature by 3 weeks (rodent-like), most neurons mature at 5 months and hippocampal neurogenic levels are low (like long-lived species). These data establish a timeline for future investigations of longevity- and socially-related manipulations of naked mole-rat adult neurogenesis.
Collapse
Affiliation(s)
| | - Troy Collins
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON, Canada
| | - Diana Peragine
- Department of Psychology, University of Toronto Mississauga, Mississauga, ON, Canada
| | - Manahil Malik
- Department of Psychology, University of Toronto Mississauga, Mississauga, ON, Canada
| | - Fiza Javed
- Department of Psychology, University of Toronto Mississauga, Mississauga, ON, Canada
| | - Matthew Kolisnyk
- Department of Psychology, University of Toronto Mississauga, Mississauga, ON, Canada
| | - Justine Ziolkowski
- Department of Psychology, University of Toronto Mississauga, Mississauga, ON, Canada
| | - Imaan Jeewa
- Department of Psychology, University of Toronto Mississauga, Mississauga, ON, Canada
| | - Arthur H. Cheng
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON, Canada
| | - Christopher Lowden
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON, Canada
| | - Brittany Mascarenhas
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON, Canada
| | - Hai-Ying Mary Cheng
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON, Canada
- Department of Biology, University of Toronto Mississauga, Mississauga, ON, Canada
| | - Melissa M. Holmes
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON, Canada
- Department of Psychology, University of Toronto Mississauga, Mississauga, ON, Canada
- Department of Ecology and Evolutionary Biology, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
4
|
Singular Adult Neural Stem Cells Do Not Exist. Cells 2022; 11:cells11040722. [PMID: 35203370 PMCID: PMC8870225 DOI: 10.3390/cells11040722] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 02/04/2022] [Accepted: 02/10/2022] [Indexed: 12/13/2022] Open
Abstract
Adult neural stem cells (aNSCs) are the source for the continuous production of new neurons throughout life. This so-called adult neurogenesis has been extensively studied; the intermediate cellular stages are well documented. Recent discoveries have raised new controversies in the field, such as the notion that progenitor cells hold similar self-renewal potential as stem cells, or whether different types of aNSCs exist. Here, we discuss evidence for heterogeneity of aNSCs, including short-term and long-term self-renewing aNSCs, regional and temporal differences in aNSC function, and single cell transcriptomics. Reviewing various genetic mouse models used for targeting aNSCs and lineage tracing, we consider potential lineage relationships between Ascl1-, Gli1-, and Nestin-targeted aNSCs. We present a multidimensional model of adult neurogenesis that incorporates recent findings and conclude that stemness is a phenotype, a state of properties that can change with time, rather than a cell property, which is static and immutable. We argue that singular aNSCs do not exist.
Collapse
|
5
|
Núñez Y, Radović Č, Savić R, García-Casco JM, Čandek-Potokar M, Benítez R, Radojković D, Lukić M, Gogić M, Muñoz M, Fontanesi L, Óvilo C. Muscle Transcriptome Analysis Reveals Molecular Pathways Related to Oxidative Phosphorylation, Antioxidant Defense, Fatness and Growth in Mangalitsa and Moravka Pigs. Animals (Basel) 2021; 11:ani11030844. [PMID: 33809803 PMCID: PMC8002519 DOI: 10.3390/ani11030844] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 03/04/2021] [Accepted: 03/13/2021] [Indexed: 12/28/2022] Open
Abstract
This work was aimed at evaluating loin transcriptome and metabolic pathway differences between the two main Serbian local pig breeds with divergent characteristics regarding muscle growth and fatness, as well as exploring nutrigenomic effects of tannin supplementation in Mangalitsa (MA) pigs. The study comprised 24 Mangalitsa and 10 Moravka (MO) males, which were kept under identical management conditions. Mangalitsa animals were divided in two nutritional groups (n = 12) receiving a standard (control) or tannin-supplemented diet (1.5%; MAT). Moravka pigs were fed the standard mixture. All animals were slaughtered at a similar age; 120 kg of average live weight (LW) and loin tissue was used for RNA-seq analysis. Results showed 306 differentially expressed genes (DEGs) according to breed, enriched in genes involved in growth, lipid metabolism, protein metabolism and muscle development, such as PDK4, FABP4, MYOD1 and STAT3, as well as a relevant number of genes involved in mitochondrial respiratory activity (MT-NDs, NDUFAs among others). Oxidative phosphorylation was the most significantly affected pathway, activated in Mangalitsa muscle, revealing the basis of a different muscle metabolism. Also, many other relevant pathways were affected by breed and involved in oxidative stress response, fat accumulation and development of skeletal muscle. Results also allowed the identification of potential regulators and causal networks such as those controlled by FLCN, PPARGC1A or PRKAB1 with relevant regulatory roles on DEGs involved in mitochondrial and lipid metabolism, or IL3 and TRAF2 potentially controlling DEGs involved in muscle development. The Tannin effect on transcriptome was small, with only 23 DEGs, but included interesting ones involved in lipid deposition such as PPARGC1B. The results indicate a significant effect of the breed on muscle tissue gene expression, affecting relevant biological pathways and allowing the identification of strong regulatory candidate genes to underlie the gene expression and phenotypic differences between the compared groups.
Collapse
Affiliation(s)
- Yolanda Núñez
- Departamento de Mejora Genética Animal, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA), 28040 Madrid, Spain; (Y.N.); (J.M.G.-C.); (R.B.); (M.M.)
| | - Čedomir Radović
- Institute for Animal Husbandry, 11080 Belgrade, Serbia; (Č.R.); (M.L.); (M.G.)
| | - Radomir Savić
- Faculty of Agriculture, University of Belgrade, 11080 Belgrade, Serbia; (R.S.); (D.R.)
| | - Juan M. García-Casco
- Departamento de Mejora Genética Animal, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA), 28040 Madrid, Spain; (Y.N.); (J.M.G.-C.); (R.B.); (M.M.)
| | | | - Rita Benítez
- Departamento de Mejora Genética Animal, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA), 28040 Madrid, Spain; (Y.N.); (J.M.G.-C.); (R.B.); (M.M.)
| | - Dragan Radojković
- Faculty of Agriculture, University of Belgrade, 11080 Belgrade, Serbia; (R.S.); (D.R.)
| | - Miloš Lukić
- Institute for Animal Husbandry, 11080 Belgrade, Serbia; (Č.R.); (M.L.); (M.G.)
| | - Marija Gogić
- Institute for Animal Husbandry, 11080 Belgrade, Serbia; (Č.R.); (M.L.); (M.G.)
| | - María Muñoz
- Departamento de Mejora Genética Animal, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA), 28040 Madrid, Spain; (Y.N.); (J.M.G.-C.); (R.B.); (M.M.)
| | - Luca Fontanesi
- Department of Agricultural and Food Sciences, University of Bologna, 40126 Bologna, Italy;
| | - Cristina Óvilo
- Departamento de Mejora Genética Animal, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA), 28040 Madrid, Spain; (Y.N.); (J.M.G.-C.); (R.B.); (M.M.)
- Correspondence: ; Tel.: +34-913471492
| |
Collapse
|
6
|
Cellular Mechanisms Participating in Brain Repair of Adult Zebrafish and Mammals after Injury. Cells 2021; 10:cells10020391. [PMID: 33672842 PMCID: PMC7917790 DOI: 10.3390/cells10020391] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 01/28/2021] [Accepted: 02/05/2021] [Indexed: 12/12/2022] Open
Abstract
Adult neurogenesis is an evolutionary conserved process occurring in all vertebrates. However, striking differences are observed between the taxa, considering the number of neurogenic niches, the neural stem cell (NSC) identity, and brain plasticity under constitutive and injury-induced conditions. Zebrafish has become a popular model for the investigation of the molecular and cellular mechanisms involved in adult neurogenesis. Compared to mammals, the adult zebrafish displays a high number of neurogenic niches distributed throughout the brain. Furthermore, it exhibits a strong regenerative capacity without scar formation or any obvious disabilities. In this review, we will first discuss the similarities and differences regarding (i) the distribution of neurogenic niches in the brain of adult zebrafish and mammals (mainly mouse) and (ii) the nature of the neural stem cells within the main telencephalic niches. In the second part, we will describe the cascade of cellular events occurring after telencephalic injury in zebrafish and mouse. Our study clearly shows that most early events happening right after the brain injury are shared between zebrafish and mouse including cell death, microglia, and oligodendrocyte recruitment, as well as injury-induced neurogenesis. In mammals, one of the consequences following an injury is the formation of a glial scar that is persistent. This is not the case in zebrafish, which may be one of the main reasons that zebrafish display a higher regenerative capacity.
Collapse
|
7
|
Tsintou M, Dalamagkas K, Makris N. Taking central nervous system regenerative therapies to the clinic: curing rodents versus nonhuman primates versus humans. Neural Regen Res 2020; 15:425-437. [PMID: 31571651 PMCID: PMC6921352 DOI: 10.4103/1673-5374.266048] [Citation(s) in RCA: 25] [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: 04/01/2019] [Accepted: 06/04/2019] [Indexed: 12/17/2022] Open
Abstract
The central nervous system is known to have limited regenerative capacity. Not only does this halt the human body's reparative processes after central nervous system lesions, but it also impedes the establishment of effective and safe therapeutic options for such patients. Despite the high prevalence of stroke and spinal cord injury in the general population, these conditions remain incurable and place a heavy burden on patients' families and on society more broadly. Neuroregeneration and neural engineering are diverse biomedical fields that attempt reparative treatments, utilizing stem cells-based strategies, biologically active molecules, nanotechnology, exosomes and highly tunable biodegradable systems (e.g., certain hydrogels). Although there are studies demonstrating promising preclinical results, safe clinical translation has not yet been accomplished. A key gap in clinical translation is the absence of an ideal animal or ex vivo model that can perfectly simulate the human microenvironment, and also correspond to all the complex pathophysiological and neuroanatomical factors that affect functional outcomes in humans after central nervous system injury. Such an ideal model does not currently exist, but it seems that the nonhuman primate model is uniquely qualified for this role, given its close resemblance to humans. This review considers some regenerative therapies for central nervous system repair that hold promise for future clinical translation. In addition, it attempts to uncover some of the main reasons why clinical translation might fail without the implementation of nonhuman primate models in the research pipeline.
Collapse
Affiliation(s)
- Magdalini Tsintou
- Departments of Psychiatry and Neurology Services, Center for Neural Systems Investigations, Center for Morphometric Analysis, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Department of Psychiatry, Psychiatry Neuroimaging Laboratory, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- University College of London Division of Surgery & Interventional Science, Center for Nanotechnology & Regenerative Medicine, University College London, London, UK
| | - Kyriakos Dalamagkas
- University College of London Division of Surgery & Interventional Science, Center for Nanotechnology & Regenerative Medicine, University College London, London, UK
- Department of Physical Medicine and Rehabilitation, The University of Texas Health Science Center at Houston, Houston, TX, USA
- The Institute for Rehabilitation and Research Memorial Hermann Research Center, The Institute for Rehabilitation and Research Memorial Hermann Hospital, Houston, TX, USA
| | - Nikos Makris
- Departments of Psychiatry and Neurology Services, Center for Neural Systems Investigations, Center for Morphometric Analysis, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Department of Psychiatry, Psychiatry Neuroimaging Laboratory, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Anatomy & Neurobiology, Boston University School of Medicine, Boston, MA, USA
| |
Collapse
|
8
|
Bekiari C, Grivas I, Tsingotjidou A, Papadopoulos GC. Adult neurogenesis and gliogenesis in the dorsal and ventral canine hippocampus. J Comp Neurol 2019; 528:1216-1230. [PMID: 31743444 DOI: 10.1002/cne.24818] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Revised: 11/08/2019] [Accepted: 11/08/2019] [Indexed: 12/13/2022]
Abstract
Dentate gyrus (DG) of the mammalian hippocampus gives rise to new neurons and astrocytes all through adulthood. Canine hippocampus presents many similarities in fetal development, anatomy, and physiology with human hippocampus, establishing canines as excellent animal models for the study of adult neurogenesis. In the present study, BrdU-dated cells of the structurally and functionally dissociated dorsal (dDG) and ventral (vDG) adult canine DG were comparatively examined over a period of 30 days. Each part's neurogenic potential, radial glia-like neural stem cells (NSCs) proliferation and differentiation, migration, and maturation of their progenies were evaluated at 2, 5, 14, and 30 days post BrdU administration, with the use of selected markers (glial fibrillary acidic protein, doublecortin, calretinin and calbindin). Co-staining of BrdU+ cells with NeuN or S100B permitted the parallel study of the ongoing neurogenesis and gliogenesis. Our findings reveal the comparatively higher populations of residing granule cells, proliferating NSCs and BrdU+ neurons in the dDG, whereas newborn neurons of the vDG showed a prolonged differentiation, migration, and maturation. Newborn astrocytes were found all along the dorso-ventral axis, counting however for only 11% of newborn cell population. Comparative evaluation of adult canine and rat neurogenesis revealed significant differences in the distribution of resident and newborn granule cells along the dorso-ventral axis, division pattern of adult NSCs, maturation time plan of newborn neurons, and ongoing gliogenesis. Concluding, spatial and temporal features of adult canine neurogenesis are similar to that of other gyrencephalic species, including humans, and justify the comparative examination of adult neurogenesis across mammalian species.
Collapse
Affiliation(s)
- Chryssa Bekiari
- Laboratory of Anatomy, Histology & Embryology, Faculty of Veterinary Medicine, School of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Ioannis Grivas
- Laboratory of Anatomy, Histology & Embryology, Faculty of Veterinary Medicine, School of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Anastasia Tsingotjidou
- Laboratory of Anatomy, Histology & Embryology, Faculty of Veterinary Medicine, School of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Georgios C Papadopoulos
- Laboratory of Anatomy, Histology & Embryology, Faculty of Veterinary Medicine, School of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki, Greece
| |
Collapse
|
9
|
Keller M, Vandenberg LN, Charlier TD. The parental brain and behavior: A target for endocrine disruption. Front Neuroendocrinol 2019; 54:100765. [PMID: 31112731 PMCID: PMC6708493 DOI: 10.1016/j.yfrne.2019.100765] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Revised: 05/15/2019] [Accepted: 05/17/2019] [Indexed: 12/25/2022]
Abstract
During pregnancy, the sequential release of progesterone, 17β-estradiol, prolactin, oxytocin and placental lactogens reorganize the female brain. Brain structures such as the medial preoptic area, the bed nucleus of the stria terminalis and the motivation network including the ventral tegmental area and the nucleus accumbens are reorganized by this specific hormonal schedule such that the future mother will be ready to provide appropriate care for her offspring right at parturition. Any disruption to this hormone pattern, notably by exposures to endocrine disrupting chemicals (EDC), is therefore likely to affect the maternal brain and result in maladaptive maternal behavior. Development effects of EDCs have been the focus of intense study, but relatively little is known about how the maternal brain and behavior are affected by EDCs. We encourage further research to better understand how the physiological hormone sequence prepares the mother's brain and how EDC exposure could disturb this reorganization.
Collapse
Affiliation(s)
- Matthieu Keller
- Laboratoire de Physiologie de la Reproduction & des Comportements, UMR 7247 INRA/CNRS/Université de Tours/IFCE, Nouzilly, France
| | - Laura N Vandenberg
- School of Public Health and Health Sciences, University of Massachusetts, Amherst, USA
| | - Thierry D Charlier
- Univ Rennes, Inserm, EHESP, Irset (Institut de Recherche en Santé, Environnement et Travail), UMR_S 1085, F-35000 Rennes, France.
| |
Collapse
|
10
|
Lei W, Li W, Ge L, Chen G. Non-engineered and Engineered Adult Neurogenesis in Mammalian Brains. Front Neurosci 2019; 13:131. [PMID: 30872991 PMCID: PMC6401632 DOI: 10.3389/fnins.2019.00131] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2018] [Accepted: 02/05/2019] [Indexed: 12/31/2022] Open
Abstract
Adult neurogenesis has been extensively studied in rodent animals, with distinct niches found in the hippocampus and subventricular zone (SVZ). In non-human primates and human postmortem samples, there has been heated debate regarding adult neurogenesis, but it is largely agreed that the rate of adult neurogenesis is much reduced comparing to rodents. The limited adult neurogenesis may partly explain why human brains do not have self-repair capability after injury or disease. A new technology called “in vivo cell conversion” has been invented to convert brain internal glial cells in the injury areas directly into functional new neurons to replenish the lost neurons. Because glial cells are abundant throughout the brain and spinal cord, such engineered glia-to-neuron conversion technology can be applied throughout the central nervous system (CNS) to regenerate new neurons. Thus, compared to cell transplantation or the non-engineered adult neurogenesis, in vivo engineered neuroregeneration technology can provide a large number of functional new neurons in situ to repair damaged brain and spinal cord.
Collapse
Affiliation(s)
- Wenliang Lei
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, China
| | - Wen Li
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, China
| | - Longjiao Ge
- Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Gong Chen
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, China.,Department of Biology, Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, PA, United States
| |
Collapse
|
11
|
Boisgontier J, Tacchella JM, Lemaître H, Lehman N, Saitovitch A, Gatinois V, Boursier G, Sanchez E, Rechtman E, Fillon L, Lyonnet S, Le Quang Sang KH, Baujat G, Rio M, Boute O, Faivre L, Schaefer E, Sanlaville D, Zilbovicius M, Grévent D, Geneviève D, Boddaert N. Anatomical and functional abnormalities on MRI in kabuki syndrome. NEUROIMAGE-CLINICAL 2018; 21:101610. [PMID: 30497982 PMCID: PMC6413468 DOI: 10.1016/j.nicl.2018.11.020] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Revised: 11/16/2018] [Accepted: 11/18/2018] [Indexed: 01/12/2023]
Abstract
Kabuki syndrome (KS) is a rare congenital disorder (1/32000 births) characterized by distinctive facial features, intellectual disability, short stature, and dermatoglyphic and skeletal abnormalities. In the last decade, mutations in KMT2D and KDM6A were identified as a major cause of kabuki syndrome. Although genetic abnormalities have been highlighted in KS, brain abnormalities have been little explored. Here, we have investigated brain abnormalities in 6 patients with KS (4 males; Mage = 10.96 years, SD = 2.97 years) with KMT2D mutation in comparison with 26 healthy controls (17 males; Mage = 10.31 years, SD = 2.96 years). We have used MRI to explore anatomical and functional brain abnormalities in patients with KS. Anatomical abnormalities in grey matter volume were assessed by cortical and subcortical analyses. Functional abnormalities were assessed by comparing rest cerebral blood flow measured with arterial spin labeling-MRI. When compared to healthy controls, KS patients had anatomical alterations characterized by grey matter decrease localized in the bilateral precentral gyrus and middle frontal gyrus. In addition, KS patients also presented functional alterations characterized by cerebral blood flow decrease in the left precentral gyrus and middle frontal gyrus. Moreover, subcortical analyses revealed significantly decreased grey matter volume in the bilateral hippocampus and dentate gyrus in patients with KS. Our results strongly indicate anatomical and functional brain abnormalities in KS. They suggest a possible neural basis of the cognitive symptoms observed in KS, such as fine motor impairment, and indicate the need to further explore the consequences of such brain abnormalities in this disorder. Finally, our results encourage further imaging-genetics studies investigating the link between genetics, anatomical and functional brain alterations in KS.
Collapse
Affiliation(s)
- Jennifer Boisgontier
- Service de radiologie pédiatrique, Hôpital Necker Enfants Malades, INSERM U1000, AP-HP, Université René Descartes, Pres Sorbonne Paris Cité, Institut Imagine, UMR 1163, France.
| | - Jean Marc Tacchella
- Service de radiologie pédiatrique, Hôpital Necker Enfants Malades, INSERM U1000, AP-HP, Université René Descartes, Pres Sorbonne Paris Cité, Institut Imagine, UMR 1163, France
| | - Hervé Lemaître
- Service de radiologie pédiatrique, Hôpital Necker Enfants Malades, INSERM U1000, AP-HP, Université René Descartes, Pres Sorbonne Paris Cité, Institut Imagine, UMR 1163, France; Faculté de Médecine, Université Paris Sud, France
| | - Natacha Lehman
- Département de Génétique Médicale, maladies Rares et Médecine Personnalisée, génétique clinique, CHU Montpellier, Université Montpellier, Centre de référence anomalies du développement SORO, INSERM U1183, Montpellier, France
| | - Ana Saitovitch
- Service de radiologie pédiatrique, Hôpital Necker Enfants Malades, INSERM U1000, AP-HP, Université René Descartes, Pres Sorbonne Paris Cité, Institut Imagine, UMR 1163, France
| | - Vincent Gatinois
- Département de Génétique Médicale, maladies Rares et Médecine Personnalisée, génétique clinique, CHU Montpellier, Université Montpellier, Centre de référence anomalies du développement SORO, INSERM U1183, Montpellier, France
| | - Guilaine Boursier
- Département de Génétique Médicale, maladies Rares et Médecine Personnalisée, génétique clinique, CHU Montpellier, Université Montpellier, Centre de référence anomalies du développement SORO, INSERM U1183, Montpellier, France
| | - Elodie Sanchez
- Département de Génétique Médicale, maladies Rares et Médecine Personnalisée, génétique clinique, CHU Montpellier, Université Montpellier, Centre de référence anomalies du développement SORO, INSERM U1183, Montpellier, France
| | - Elza Rechtman
- Service de radiologie pédiatrique, Hôpital Necker Enfants Malades, INSERM U1000, AP-HP, Université René Descartes, Pres Sorbonne Paris Cité, Institut Imagine, UMR 1163, France
| | - Ludovic Fillon
- Service de radiologie pédiatrique, Hôpital Necker Enfants Malades, INSERM U1000, AP-HP, Université René Descartes, Pres Sorbonne Paris Cité, Institut Imagine, UMR 1163, France
| | - Stanislas Lyonnet
- Service de Génétique Médicale, Institut IMAGINE, AP-HP Necker Enfants Malades, France
| | | | - Genevieve Baujat
- Service de Génétique Médicale, Institut IMAGINE, AP-HP Necker Enfants Malades, France
| | - Marlene Rio
- Service de Génétique Médicale, Institut IMAGINE, AP-HP Necker Enfants Malades, France
| | - Odile Boute
- Service de génétique Clinique, Hôpital Jeanne de Flandre, France
| | - Laurence Faivre
- Service de génétique médicale, Centre de référence anomalies du développement, Fédération Hospitalo-Universitaire Médecine Translationnelle et Anomalies Du Développement (TRANSLAD), Centre Hospitalier Universitaire Dijon, Dijon, France
| | - Elise Schaefer
- Service de génétique médicale, Institut de Génétique Médicale d'Alsace, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - Damien Sanlaville
- Hospices civils de Lyon, Service de génétique, Centre de Recherche en Neurosciences de Lyon, Inserm U1028, UMR CNRS 5292, GENDEV Team, Université Claude Bernard Lyon 1, Lyon, France
| | - Monica Zilbovicius
- Service de radiologie pédiatrique, Hôpital Necker Enfants Malades, INSERM U1000, AP-HP, Université René Descartes, Pres Sorbonne Paris Cité, Institut Imagine, UMR 1163, France
| | - David Grévent
- Service de radiologie pédiatrique, Hôpital Necker Enfants Malades, INSERM U1000, AP-HP, Université René Descartes, Pres Sorbonne Paris Cité, Institut Imagine, UMR 1163, France
| | - David Geneviève
- Département de Génétique Médicale, maladies Rares et Médecine Personnalisée, génétique clinique, CHU Montpellier, Université Montpellier, Centre de référence anomalies du développement SORO, INSERM U1183, Montpellier, France
| | - Nathalie Boddaert
- Service de radiologie pédiatrique, Hôpital Necker Enfants Malades, INSERM U1000, AP-HP, Université René Descartes, Pres Sorbonne Paris Cité, Institut Imagine, UMR 1163, France
| |
Collapse
|
12
|
|
13
|
La Rosa C, Parolisi R, Palazzo O, Lévy F, Meurisse M, Bonfanti L. Clusters of DCX+ cells "trapped" in the subcortical white matter of early postnatal Cetartiodactyla (Tursiops truncatus, Stenella coeruloalba and Ovis aries). Brain Struct Funct 2018; 223:3613-3632. [PMID: 29980931 DOI: 10.1007/s00429-018-1708-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Accepted: 07/02/2018] [Indexed: 01/08/2023]
Abstract
The cytoskeletal protein doublecortin (DCX) is a marker for neuronal cells retaining high potential for structural plasticity, originating from both embryonic and adult neurogenic processes. Some of these cells have been described in the subcortical white matter of neonatal and postnatal mammals. In mice and humans it has been shown they are young neurons migrating through the white matter after birth, reaching the cortex in a sort of protracted neurogenesis. Here we show that DCX+ cells in the white matter of neonatal and young Cetartiodactyla (dolphin and sheep) form large clusters which are not newly generated (in sheep, and likely neither in dolphins) and do not reach the cortical layers, rather appearing "trapped" in the white matter tissue. No direct contact or continuity can be observed between the subventricular zone region and the DCX+ clusters, thus indicating their independence from any neurogenic source (in dolphins further confirmed by the recent demonstration that periventricular neurogenesis is inactive since birth). Cetartiodactyla include two orders of large-brained, relatively long-living mammals (cetaceans and artiodactyls) which were recognized as two separate monophyletic clades until recently, yet, despite the evident morphological distinctions, they are monophyletic in origin. The brain of Cetartiodactyla is characterized by an advanced stage of development at birth, a feature that might explain the occurrence of "static" cell clusters confined within their white matter. These results further confirm the existence of high heterogeneity in the occurrence, distribution and types of structural plasticity among mammals, supporting the emerging view that multiple populations of DCX+, non-newly generated cells can be abundant in large-brained, long-living species.
Collapse
Affiliation(s)
- Chiara La Rosa
- Neuroscience Institute Cavalieri Ottolenghi (NICO), Orbassano, Italy.,Department of Veterinary Sciences, University of Turin, Largo Braccini 2, 10095, Grugliasco, TO, Italy
| | - Roberta Parolisi
- Neuroscience Institute Cavalieri Ottolenghi (NICO), Orbassano, Italy
| | - Ottavia Palazzo
- Neuroscience Institute Cavalieri Ottolenghi (NICO), Orbassano, Italy
| | - Frederic Lévy
- UMR INRA, CNRS/Universitè F. Rabelais, IFCE Physiologie de la Reproduction et des Comportements, Nouzilly, France
| | - Maryse Meurisse
- UMR INRA, CNRS/Universitè F. Rabelais, IFCE Physiologie de la Reproduction et des Comportements, Nouzilly, France
| | - Luca Bonfanti
- Neuroscience Institute Cavalieri Ottolenghi (NICO), Orbassano, Italy. .,Department of Veterinary Sciences, University of Turin, Largo Braccini 2, 10095, Grugliasco, TO, Italy.
| |
Collapse
|
14
|
Krzisch M, Fülling C, Jabinet L, Armida J, Gebara E, Cassé F, Habbas S, Volterra A, Hornung JP, Toni N. Synaptic Adhesion Molecules Regulate the Integration of New Granule Neurons in the Postnatal Mouse Hippocampus and their Impact on Spatial Memory. Cereb Cortex 2018; 27:4048-4059. [PMID: 27473321 DOI: 10.1093/cercor/bhw217] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2016] [Accepted: 06/21/2016] [Indexed: 11/14/2022] Open
Abstract
Postnatal hippocampal neurogenesis induces network remodeling and may participate to mechanisms of learning. In turn, the maturation and survival of newborn neurons is regulated by their activity. Here, we tested the effect of a cell-autonomous overexpression of synaptic adhesion molecules on the maturation and survival of neurons born postnatally and on hippocampal-dependent memory performances. Families of adhesion molecules are known to induce pre- and post-synaptic assembly. Using viral targeting, we overexpressed three different synaptic adhesion molecules, SynCAM1, Neuroligin-1B and Neuroligin-2A in newborn neurons in the dentate gyrus of 7- to 9-week-old mice. We found that SynCAM1 increased the morphological maturation of dendritic spines and mossy fiber terminals while Neuroligin-1B increased spine density. In contrast, Neuroligin-2A increased both spine density and size as well as GABAergic innervation and resulted in a drastic increase of neuronal survival. Surprisingly, despite increased neurogenesis, mice overexpressing Neuroligin-2A in new neurons showed decreased memory performances in a Morris water maze task. These results indicate that the cell-autonomous overexpression of synaptic adhesion molecules can enhance different aspects of synapse formation on new neurons and increase their survival. Furthermore, they suggest that the mechanisms by which new neurons integrate in the postnatal hippocampus conditions their functional implication in learning and memory.
Collapse
Affiliation(s)
- Marine Krzisch
- Department of Fundamental Neurosciences, University of Lausanne, 9, rue du Bugnon, Lausanne, Switzerland
| | - Christine Fülling
- Department of Fundamental Neurosciences, University of Lausanne, 9, rue du Bugnon, Lausanne, Switzerland
| | - Laura Jabinet
- Department of Fundamental Neurosciences, University of Lausanne, 9, rue du Bugnon, Lausanne, Switzerland
| | - Jan Armida
- Department of Fundamental Neurosciences, University of Lausanne, 9, rue du Bugnon, Lausanne, Switzerland
| | - Elias Gebara
- Department of Fundamental Neurosciences, University of Lausanne, 9, rue du Bugnon, Lausanne, Switzerland
| | - Frédéric Cassé
- Department of Fundamental Neurosciences, University of Lausanne, 9, rue du Bugnon, Lausanne, Switzerland
| | - Samia Habbas
- Department of Fundamental Neurosciences, University of Lausanne, 9, rue du Bugnon, Lausanne, Switzerland
| | - Andrea Volterra
- Department of Fundamental Neurosciences, University of Lausanne, 9, rue du Bugnon, Lausanne, Switzerland
| | - Jean-Pierre Hornung
- Department of Fundamental Neurosciences, University of Lausanne, 9, rue du Bugnon, Lausanne, Switzerland
| | - Nicolas Toni
- Department of Fundamental Neurosciences, University of Lausanne, 9, rue du Bugnon, Lausanne, Switzerland
| |
Collapse
|
15
|
Liu J, Reeves C, Jacques T, McEvoy A, Miserocchi A, Thompson P, Sisodiya S, Thom M. Nestin-expressing cell types in the temporal lobe and hippocampus: Morphology, differentiation, and proliferative capacity. Glia 2018; 66:62-77. [PMID: 28925561 PMCID: PMC5724502 DOI: 10.1002/glia.23211] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Revised: 07/30/2017] [Accepted: 08/09/2017] [Indexed: 12/13/2022]
Abstract
Nestin is expressed in immature neuroepithelial and progenitor cell types and transiently upregulated in proliferative neuroglial cells responding to acute brain injury, including following seizures. In 36 temporal lobe (TLobe) specimens from patients with TLobe epilepsy (age range 8-60 years) we studied the number, distribution and morphology of nestin-expressing cells (NEC) in the pes, hippocampus body, parahippocampal gyrus, amygdala, temporal cortex and pole compared with post mortem control tissues from 26 cases (age range 12 gestational weeks to 76 years). The proliferative fraction of NEC was evaluated in selected regions, including recognized niches, using MCM2. Their differentiation was explored with neuronal (DCX, mushashi, βIII tubulin, NeuN) and glial (GFAP, GFAPdelta, glutamine synthetase, aquaporin4, EAAT1) markers, both in sections or following culture. Findings were correlated with clinical parameters. A stereotypical pattern in the distribution and morphologies of NEC was observed, reminiscent of patterns in the developing brain, with increased densities in epilepsy than adult controls (p < .001). Findings included MCM2-positive radial glial-like cells in the periventricular white matter and rows of NEC in the hippocampal fimbria and sulcus. Nestin cells represented 29% of the hippocampal proliferative fraction in epilepsy cases; 20% co-expressed βIII tubulin in culture compared with 28% with GFAP. Significant correlations were noted between age at surgery, memory deficits and nestin populations. TLobe NEC with ongoing proliferative capacity likely represent vestiges of developmental migratory streams and resident reactive cell populations of potential relevance to hippocampal epileptogenesis, TLobe pathology, and co-morbidities, including memory decline.
Collapse
Affiliation(s)
- Joan Liu
- Department of Clinical and Experimental EpilepsyUCL Institute of Neurology, Queen SquareLondonWC1N 3BGUnited Kingdom
- Divisions of NeuropathologyNational Hospital for Neurology and Neurosurgery, Queen SquareLondonWCN1BGUK
- Department of Biomedical SciencesUniversity of WestminsterLondonW1W 6UWUnited Kingdom
| | - Cheryl Reeves
- Department of Clinical and Experimental EpilepsyUCL Institute of Neurology, Queen SquareLondonWC1N 3BGUnited Kingdom
- Divisions of NeuropathologyNational Hospital for Neurology and Neurosurgery, Queen SquareLondonWCN1BGUK
| | - Thomas Jacques
- Department of NeuropathologyUCL Institute of Child Health and Great Ormond Street Hospital for ChildrenLondonUnited Kingdom
| | - Andrew McEvoy
- Department of Clinical and Experimental EpilepsyUCL Institute of Neurology, Queen SquareLondonWC1N 3BGUnited Kingdom
- Neurosurgery at the National Hospital for Neurology and Neurosurgery, Queen SquareLondonWCN1BGUnited Kingdom
| | - Anna Miserocchi
- Neurosurgery at the National Hospital for Neurology and Neurosurgery, Queen SquareLondonWCN1BGUnited Kingdom
| | - Pamela Thompson
- Department of Clinical and Experimental EpilepsyUCL Institute of Neurology, Queen SquareLondonWC1N 3BGUnited Kingdom
- The Chalfont Centre for Epilepsy, Chesham Lane, Chalfont St PeterBuckinghamshireSL9 0RJUnited Kingdom
- Department of NeuropsychologyNational Hospital for Neurology and Neurosurgery, Queen SquareLondonWCN1BGUK
| | - Sanjay Sisodiya
- Department of Clinical and Experimental EpilepsyUCL Institute of Neurology, Queen SquareLondonWC1N 3BGUnited Kingdom
- The Chalfont Centre for Epilepsy, Chesham Lane, Chalfont St PeterBuckinghamshireSL9 0RJUnited Kingdom
- Department of NeurologyNational Hospital for Neurology and Neurosurgery, Queen SquareLondonWCN1BGUK
| | - Maria Thom
- Department of Clinical and Experimental EpilepsyUCL Institute of Neurology, Queen SquareLondonWC1N 3BGUnited Kingdom
- Divisions of NeuropathologyNational Hospital for Neurology and Neurosurgery, Queen SquareLondonWCN1BGUK
| |
Collapse
|
16
|
Non-Newly Generated, "Immature" Neurons in the Sheep Brain Are Not Restricted to Cerebral Cortex. J Neurosci 2017; 38:826-842. [PMID: 29217680 DOI: 10.1523/jneurosci.1781-17.2017] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Revised: 10/24/2017] [Accepted: 11/16/2017] [Indexed: 12/15/2022] Open
Abstract
A newly proposed form of brain structural plasticity consists of non-newly generated, "immature" neurons of the adult cerebral cortex. Similar to newly generated neurons, these cells express the cytoskeletal protein Doublecortin (DCX), yet they are generated prenatally and then remain in a state of immaturity for long periods. In rodents, the immature neurons are restricted to the paleocortex, whereas in other mammals, they are also found in neocortex. Here, we analyzed the DCX-expressing cells in the whole sheep brain of both sexes to search for an indicator of structural plasticity at a cellular level in a relatively large-brained, long-living mammal. Brains from adult and newborn sheep (injected with BrdU and analyzed at different survival times) were processed for DCX, cell proliferation markers (Ki-67, BrdU), pallial/subpallial developmental origin (Tbr1, Sp8), and neuronal/glial antigens for phenotype characterization. We found immature-like neurons in the whole sheep cortex and in large populations of DCX-expressing cells within the external capsule and the surrounding gray matter (claustrum and amygdala). BrdU and Ki-67 detection at neonatal and adult ages showed that all of these DCX+ cells were generated during embryogenesis, not after birth. These results show that the adult sheep, unlike rodents, is largely endowed with non-newly generated neurons retaining immature features, suggesting that such plasticity might be particularly important in large-brained, long-living mammals.SIGNIFICANCE STATEMENT Brain plasticity is important in adaptation and brain repair. Structural changes span from synaptic plasticity to adult neurogenesis, the latter being highly reduced in large-brained, long-living mammals (e.g., humans). The cerebral cortex contains "immature" neurons, which are generated prenatally and then remain in an undifferentiated state for long periods, being detectable with markers of immaturity. We studied the distribution and developmental origin of these cells in the whole brain of sheep, relatively large-brained, long-living mammals. In addition to the expected cortical location, we also found populations of non-newly generated neurons in several subcortical regions (external capsule, claustrum, and amygdala). These results suggests that non-neurogenic, parenchymal structural plasticity might be more important in large mammals with respect to adult neurogenesis.
Collapse
|
17
|
Powers AS. Preface. BRAIN, BEHAVIOR AND EVOLUTION 2016; 87:143-145. [PMID: 27560929 DOI: 10.1159/000447046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
|
18
|
Brus M, Trouillet AC, Hellier V, Bakker J. Estradiol-induced neurogenesis in the female accessory olfactory bulb is required for the learning of the male odor. J Neurochem 2016; 138:457-68. [PMID: 27216894 DOI: 10.1111/jnc.13677] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Revised: 04/20/2016] [Accepted: 05/12/2016] [Indexed: 01/05/2023]
Abstract
Odors processed by the main and accessory olfactory bulbs (MOB, AOB) are important for sexual behavior. Interestingly, both structures continue to receive new neurons during adulthood. A role for olfactory neurogenesis in sexual behavior in female mice has recently been shown and gonadal hormones such as estradiol can modulate adult neurogenesis. Therefore, we wanted to determine the role of estradiol in learning the odors of sexual partners and in the adult neurogenesis of female aromatase knockout mice (ArKO), unable to produce estradiol. Female wild-type (WT) and ArKO mice were exposed to male odors during 7 days, and olfactory preferences, cell proliferation, cell survival and functional involvement of newborn neurons were analyzed, using BrdU injections, in combination with a marker of cell activation (Zif268) and neuronal fate (doublecortin, NeuN). Behavioral tasks indicated that both WT and ArKO females were able to discriminate between the odors of two different males, but ArKO mice failed to learn the familiar male odor. Proliferation of newborn cells was reduced in ArKO mice only in the dentate gyrus of the hippocampus. Olfactory exposure decreased cell survival in the AOB in WT females, suggesting a role for estradiol in a structure involved in sexual behavior. Finally, newborn neurons do not seem to be functionally involved in the AOB of ArKO mice compared with WT, when females were exposed to the odor of a familiar male, suggesting that estradiol-induced neurogenesis in the AOB is required for the learning of the male odor in female mice. Aromatase knockout mice (ArKO) presented deficits in olfactory preferences without affecting their olfactory discrimination abilities, and showed no functional involvement of newborn neurons in the accessory olfactory bulb (AOB) in response to the odor of a familiar male. These results suggest that estradiol-induced neurogenesis in the female AOB is required for the learning of the male odor.
Collapse
Affiliation(s)
- Maïna Brus
- Laboratory of Neuroendocrinology, Groupe Interdisciplinaire de Génoprotéomique Appliquée (GIGA) Neurosciences, University of Liège, Liège, Belgium
| | - Anne-Charlotte Trouillet
- Laboratory of Neuroendocrinology, Groupe Interdisciplinaire de Génoprotéomique Appliquée (GIGA) Neurosciences, University of Liège, Liège, Belgium
| | - Vincent Hellier
- Laboratory of Neuroendocrinology, Groupe Interdisciplinaire de Génoprotéomique Appliquée (GIGA) Neurosciences, University of Liège, Liège, Belgium
| | - Julie Bakker
- Laboratory of Neuroendocrinology, Groupe Interdisciplinaire de Génoprotéomique Appliquée (GIGA) Neurosciences, University of Liège, Liège, Belgium
| |
Collapse
|
19
|
Induced pluripotent stem cells in Alzheimer's disease: applications for disease modeling and cell-replacement therapy. Mol Neurodegener 2016; 11:39. [PMID: 27184028 PMCID: PMC4869261 DOI: 10.1186/s13024-016-0106-3] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Accepted: 05/12/2016] [Indexed: 02/06/2023] Open
Abstract
Alzheimer's disease (AD) is the most common cause of dementia in those over the age of 65. While a numerous of disease-causing genes and risk factors have been identified, the exact etiological mechanisms of AD are not yet completely understood, due to the inability to test theoretical hypotheses on non-postmortem and patient-specific research systems. The use of recently developed and optimized induced pluripotent stem cells (iPSCs) technology may provide a promising platform to create reliable models, not only for better understanding the etiopathological process of AD, but also for efficient anti-AD drugs screening. More importantly, human-sourced iPSCs may also provide a beneficial tool for cell-replacement therapy against AD. Although considerable progress has been achieved, a number of key challenges still require to be addressed in iPSCs research, including the identification of robust disease phenotypes in AD modeling and the clinical availabilities of iPSCs-based cell-replacement therapy in human. In this review, we highlight recent progresses of iPSCs research and discuss the translational challenges of AD patients-derived iPSCs in disease modeling and cell-replacement therapy.
Collapse
|
20
|
Ryan SM, Kelly ÁM. Exercise as a pro-cognitive, pro-neurogenic and anti-inflammatory intervention in transgenic mouse models of Alzheimer's disease. Ageing Res Rev 2016; 27:77-92. [PMID: 27039886 DOI: 10.1016/j.arr.2016.03.007] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Revised: 03/22/2016] [Accepted: 03/30/2016] [Indexed: 12/20/2022]
Abstract
It is now well established, at least in animal models, that exercise elicits potent pro-cognitive and pro-neurogenic effects. Alzheimer's disease (AD) is one of the leading causes of dementia and represents one of the greatest burdens on healthcare systems worldwide, with no effective treatment for the disease to date. Exercise presents a promising non-pharmacological option to potentially delay the onset of or slow down the progression of AD. Exercise interventions in mouse models of AD have been explored and have been found to reduce amyloid pathology and improve cognitive function. More recent studies have expanded the research question by investigating potential pro-neurogenic and anti-inflammatory effects of exercise. In this review we summarise studies that have examined exercise-mediated effects on AD pathology, cognitive function, hippocampal neurogenesis and neuroinflammation in transgenic mouse models of AD. Furthermore, we attempt to identify the optimum exercise conditions required to elicit the greatest benefits, taking into account age and pathology of the model, as well as type and duration of exercise.
Collapse
|
21
|
Paredes MF, Sorrells SF, Garcia-Verdugo JM, Alvarez-Buylla A. Brain size and limits to adult neurogenesis. J Comp Neurol 2015; 524:646-64. [PMID: 26417888 DOI: 10.1002/cne.23896] [Citation(s) in RCA: 95] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2015] [Revised: 08/28/2015] [Accepted: 09/08/2015] [Indexed: 12/31/2022]
Abstract
The walls of the cerebral ventricles in the developing embryo harbor the primary neural stem cells from which most neurons and glia derive. In many vertebrates, neurogenesis continues postnatally and into adulthood in this region. Adult neurogenesis at the ventricle has been most extensively studied in organisms with small brains, such as reptiles, birds, and rodents. In reptiles and birds, these progenitor cells give rise to young neurons that migrate into many regions of the forebrain. Neurogenesis in adult rodents is also relatively widespread along the lateral ventricles, but migration is largely restricted to the rostral migratory stream into the olfactory bulb. Recent work indicates that the wall of the lateral ventricle is highly regionalized, with progenitor cells giving rise to different types of neurons depending on their location. In species with larger brains, young neurons born in these spatially specified domains become dramatically separated from potential final destinations. Here we hypothesize that the increase in size and topographical complexity (e.g., intervening white matter tracts) in larger brains may severely limit the long-term contribution of new neurons born close to, or in, the ventricular wall. We compare the process of adult neuronal birth, migration, and integration across species with different brain sizes, and discuss how early regional specification of progenitor cells may interact with brain size and affect where and when new neurons are added.
Collapse
Affiliation(s)
- Mercedes F Paredes
- Department of Neurological Surgery, University of California, San Francisco, CA, 94143, USA
| | - Shawn F Sorrells
- Department of Neurological Surgery, University of California, San Francisco, CA, 94143, USA.,University of California, San Francisco, CA, 94143, USA
| | - Jose M Garcia-Verdugo
- Laboratory of Comparative Neurobiology, Instituto Cavanilles, Universidad de Valencia, CIBERNED, 46980 Valencia, Spain
| | - Arturo Alvarez-Buylla
- Department of Neurological Surgery and The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, CA, 94143, USA
| |
Collapse
|
22
|
Construction of an MRI 3D high resolution sheep brain template. Magn Reson Imaging 2015; 33:1329-1337. [PMID: 26363468 DOI: 10.1016/j.mri.2015.09.001] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2014] [Revised: 03/16/2015] [Accepted: 09/02/2015] [Indexed: 01/05/2023]
Abstract
Sheep is a developing animal model used in the field of neurosciences for the study of many behavioral, physiological or pathophysiological mechanisms, including for example, the central control of social behavior, brain injury or neurodegenerative diseases. However, sheep remains an orphan species in the field of magnetic resonance imaging (MRI). Therefore, a mean image (template), resulting of registrations of multiple subject images is needed and currently does not exist. In this study, we: i) computed multimodal high resolution 3D in-vivo sheep brain templates of T1 weighted (T1W) and T2W images, ii) computed gray matter (GM), white matter (WM) and cerebrospinal fluid (CSF) prior probability maps using linear and optimized non-linear registrations iii) used prior probability maps to perform the segmentation of a single brain tissues. Computed multimodal sheep brain templates showed to preserve and underline all brain patterns of a single T1W or T2W image, and prior probability maps allowed to improve the segmentation of brain tissues. Finally, we demonstrated that these templates and prior probability maps were able to be portable in other publicly available imaging software and could be used as standardized spaces for multi-institution neuroimaging studies or other neuroscience methods.
Collapse
|
23
|
Bjornsson HT, Benjamin JS, Zhang L, Weissman J, Gerber EE, Chen YC, Vaurio RG, Potter MC, Hansen KD, Dietz HC. Histone deacetylase inhibition rescues structural and functional brain deficits in a mouse model of Kabuki syndrome. Sci Transl Med 2015; 6:256ra135. [PMID: 25273096 DOI: 10.1126/scitranslmed.3009278] [Citation(s) in RCA: 125] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Kabuki syndrome is caused by haploinsufficiency for either of two genes that promote the opening of chromatin. If an imbalance between open and closed chromatin is central to the pathogenesis of Kabuki syndrome, agents that promote chromatin opening might have therapeutic potential. We have characterized a mouse model of Kabuki syndrome with a heterozygous deletion in the gene encoding the lysine-specific methyltransferase 2D (Kmt2d), leading to impairment of methyltransferase function. In vitro reporter alleles demonstrated a reduction in histone 4 acetylation and histone 3 lysine 4 trimethylation (H3K4me3) activity in mouse embryonic fibroblasts from Kmt2d(+/βGeo) mice. These activities were normalized in response to AR-42, a histone deacetylase inhibitor. In vivo, deficiency of H3K4me3 in the dentate gyrus granule cell layer of Kmt2d(+/βGeo) mice correlated with reduced neurogenesis and hippocampal memory defects. These abnormalities improved upon postnatal treatment with AR-42. Our work suggests that a reversible deficiency in postnatal neurogenesis underlies intellectual disability in Kabuki syndrome.
Collapse
Affiliation(s)
- Hans T Bjornsson
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA. Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| | - Joel S Benjamin
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA. Predoctoral Training Program in Human Genetics, McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Li Zhang
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Jacqueline Weissman
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA. Kennedy Krieger Institute, Baltimore, MD 21205, USA
| | - Elizabeth E Gerber
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Yi-Chun Chen
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | | | - Michelle C Potter
- Brain Science Institute, Neurology Department, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Kasper D Hansen
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA. Department of Biostatistics, Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Harry C Dietz
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA. Division of Pediatric Cardiology, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA. Howard Hughes Medical Institute, Baltimore, MD 21205, USA
| |
Collapse
|
24
|
Amrein I. Adult hippocampal neurogenesis in natural populations of mammals. Cold Spring Harb Perspect Biol 2015; 7:7/5/a021295. [PMID: 25934014 DOI: 10.1101/cshperspect.a021295] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
This review will discuss adult hippocampal neurogenesis in wild mammals of different taxa and outline similarities with and differences from laboratory animals. It begins with a review of evidence for hippocampal neurogenesis in various mammals, and shows the similar patterns of age-dependent decline in cell proliferation in wild and domesticated mammals. In contrast, the pool of immature neurons that originate from proliferative activity varies between species, implying a selective advantage for mammals that can make use of a large number of these functionally special neurons. Furthermore, rapid adaptation of hippocampal neurogenesis to experimental challenges appears to be a characteristic of laboratory rodents. Wild mammals show species-specific, rather stable hippocampal neurogenesis, which appears related to demands that characterize the niche exploited by a species rather than to acute events in the life of its members. Studies that investigate adult neurogenesis in wild mammals are not numerous, but the findings of neurogenesis under natural conditions can provide new insights, and thereby also address the question to which cognitive demands neurogenesis may respond during selection.
Collapse
Affiliation(s)
- Irmgard Amrein
- Institute of Anatomy, University of Zürich-Irchel, CH-8057 Zürich, Switzerland
| |
Collapse
|
25
|
Oliveira-Pinto AV, Santos RM, Coutinho RA, Oliveira LM, Santos GB, Alho ATL, Leite REP, Farfel JM, Suemoto CK, Grinberg LT, Pasqualucci CA, Jacob-Filho W, Lent R. Sexual dimorphism in the human olfactory bulb: females have more neurons and glial cells than males. PLoS One 2014; 9:e111733. [PMID: 25372872 PMCID: PMC4221136 DOI: 10.1371/journal.pone.0111733] [Citation(s) in RCA: 75] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2014] [Accepted: 10/06/2014] [Indexed: 11/30/2022] Open
Abstract
Sex differences in the human olfactory function reportedly exist for olfactory sensitivity, odorant identification and memory, and tasks in which odors are rated based on psychological features such as familiarity, intensity, pleasantness, and others. Which might be the neural bases for these behavioral differences? The number of cells in olfactory regions, and especially the number of neurons, may represent a more accurate indicator of the neural machinery than volume or weight, but besides gross volume measures of the human olfactory bulb, no systematic study of sex differences in the absolute number of cells has yet been undertaken. In this work, we investigate a possible sexual dimorphism in the olfactory bulb, by quantifying postmortem material from 7 men and 11 women (ages 55–94 years) with the isotropic fractionator, an unbiased and accurate method to estimate absolute cell numbers in brain regions. Female bulbs weighed 0.132 g in average, while male bulbs weighed 0.137 g, a non-significant difference; however, the total number of cells was 16.2 million in females, and 9.2 million in males, a significant difference of 43.2%. The number of neurons in females reached 6.9 million, being no more than 3.5 million in males, a difference of 49.3%. The number of non-neuronal cells also proved higher in women than in men: 9.3 million and 5.7 million, respectively, a significant difference of 38.7%. The same differences remained when corrected for mass. Results demonstrate a sex-related difference in the absolute number of total, neuronal and non-neuronal cells, favoring women by 40–50%. It is conceivable that these differences in quantitative cellularity may have functional impact, albeit difficult to infer how exactly this would be, without knowing the specific circuits cells make. However, the reported advantage of women as compared to men may stimulate future work on sex dimorphism of synaptic microcircuitry in the olfactory bulb.
Collapse
Affiliation(s)
- Ana V. Oliveira-Pinto
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Raquel M. Santos
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Renan A. Coutinho
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Lays M. Oliveira
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Gláucia B. Santos
- Aging Brain Study Group, University of São Paulo Medical School, São Paulo, Brazil
- Brain Institute, Hospital Israelita Albert Einstein, São Paulo, Brazil
| | - Ana T. L. Alho
- Aging Brain Study Group, University of São Paulo Medical School, São Paulo, Brazil
- Brain Institute, Hospital Israelita Albert Einstein, São Paulo, Brazil
| | - Renata E. P. Leite
- Aging Brain Study Group, University of São Paulo Medical School, São Paulo, Brazil
| | - José M. Farfel
- Aging Brain Study Group, University of São Paulo Medical School, São Paulo, Brazil
- Discipline of Geriatrics, University of São Paulo Medical School, São Paulo, Brazil
| | - Claudia K. Suemoto
- Aging Brain Study Group, University of São Paulo Medical School, São Paulo, Brazil
- Discipline of Geriatrics, University of São Paulo Medical School, São Paulo, Brazil
| | - Lea T. Grinberg
- Aging Brain Study Group, University of São Paulo Medical School, São Paulo, Brazil
- Department of Neurology and Pathology, University of California San Francisco, San Francisco, United States of America
| | | | - Wilson Jacob-Filho
- Aging Brain Study Group, University of São Paulo Medical School, São Paulo, Brazil
- Discipline of Geriatrics, University of São Paulo Medical School, São Paulo, Brazil
| | - Roberto Lent
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- National Institute of Translational Neuroscience, Ministry of Science and Technology, São Paulo, Brazil
- * E-mail:
| |
Collapse
|
26
|
Larriva-Sahd J. Structural variation and interactions among astrocytes of the rostral migratory stream and olfactory bulb: II. Golgi and electron microscopic study of the adult rat. Neurosci Res 2014; 89:10-30. [PMID: 25205624 DOI: 10.1016/j.neures.2014.08.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2014] [Revised: 08/12/2014] [Accepted: 08/22/2014] [Indexed: 10/24/2022]
Abstract
Cytological characteristics of a cell sharing structure of both an astrocyte and a neuron, previously termed amphomorphic cell (AC), were defined here in adult rat rostral migratory stream (RMS). The AC perikaryon corresponds to that of the B1 cell of the adult mouse subventricular zone (SVZ)-RMS. The AC and its processes are confined to the RMS. Each AC originates four sets of processes that overlap with those from its homologues and adjacent neural and stromal elements. ACs interact between them via reciprocal sets of processes: those directed caudally bear spheroidal vesicles (SVP) and form gap junctions with pleomorphic vesicles (PVs) associated with the anterior set from the adjacent AC. Large asymmetric synapses, a set of them arising from the anterior olfactory nucleus, converge on each SV. The interlacing processes of the AC, together with a set of perikaryal out-growths form the glial cuff surrounding migrating neuroblasts described earlier. Small asymmetrical and symmetrical synapses terminate in subsets of differentiated A-cells, termed here A1, in the bulbar part of the RMS. Both AC- and A1-cells form electrical synapses between them and with their homologues. The strategic, wide-spread distribution between the neuropil and blood vessels of the AC, its processes, and migrating neuroblasts, suggests that the AC might mediate between both endogenous inductors and neurotransmitters, influencing the adult-born neurons it had previously originated.
Collapse
Affiliation(s)
- Jorge Larriva-Sahd
- Instituto de Neurobiología, Universidad Nacional Autónoma de México, Campus Juriquilla, Qro., Mexico.
| |
Collapse
|
27
|
Phillips KA, Bales KL, Capitanio JP, Conley A, Czoty PW, ‘t Hart BA, Hopkins WD, Hu SL, Miller LA, Nader MA, Nathanielsz PW, Rogers J, Shively CA, Voytko ML. Why primate models matter. Am J Primatol 2014; 76:801-27. [PMID: 24723482 PMCID: PMC4145602 DOI: 10.1002/ajp.22281] [Citation(s) in RCA: 399] [Impact Index Per Article: 39.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2014] [Revised: 03/01/2014] [Accepted: 03/02/2014] [Indexed: 12/13/2022]
Abstract
Research involving nonhuman primates (NHPs) has played a vital role in many of the medical and scientific advances of the past century. NHPs are used because of their similarity to humans in physiology, neuroanatomy, reproduction, development, cognition, and social complexity-yet it is these very similarities that make the use of NHPs in biomedical research a considered decision. As primate researchers, we feel an obligation and responsibility to present the facts concerning why primates are used in various areas of biomedical research. Recent decisions in the United States, including the phasing out of chimpanzees in research by the National Institutes of Health and the pending closure of the New England Primate Research Center, illustrate to us the critical importance of conveying why continued research with primates is needed. Here, we review key areas in biomedicine where primate models have been, and continue to be, essential for advancing fundamental knowledge in biomedical and biological research.
Collapse
Affiliation(s)
- Kimberley A. Phillips
- Department of Psychology, Trinity University, San Antonio TX 78212
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio TX
| | - Karen L. Bales
- Department of Psychology, University of California, Davis CA 95616
- California National Primate Research Center, Davis CA 95616
| | - John P. Capitanio
- Department of Psychology, University of California, Davis CA 95616
- California National Primate Research Center, Davis CA 95616
| | - Alan Conley
- Department of Population Health & Reproduction, School of Veterinary Medicine, University of California, Davis CA 95616
| | - Paul W. Czoty
- Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Winston-Salem NC 27157
| | - Bert A. ‘t Hart
- Department of Immunobiology, Biomedical Primate Research Center, Rijswick, The Netherlands
| | - William D. Hopkins
- Neuroscience Institute and Language Research Center, Georgia State University, Atlanta GA 30302
- Division of Cognitive and Developmental Neuroscience, Yerkes National Primate Research Center, Atlanta GA 30030
| | - Shiu-Lok Hu
- Department of Pharmaceutics and Washington National Primate Research Center, University of Washington, Seattle WA
| | - Lisa A. Miller
- California National Primate Research Center, Davis CA 95616
- Department of Anatomy, Physiology and Cell Biology, University of California, Davis CA 95616
| | - Michael A. Nader
- Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Winston-Salem NC 27157
| | - Peter W. Nathanielsz
- Center for Pregnancy and Newborn Research, University of Texas Health Science Center, San Antonio TX 78229
| | - Jeffrey Rogers
- Human Genome Sequencing Center, Baylor College of Medicine, Houston TX
- Wisconsin National Primate Research Center, Madison, WI
| | - Carol A. Shively
- Department of Pathology, Section on Comparative Medicine, Wake Forest University School of Medicine, Winston-Salem NC 27157
| | - Mary Lou Voytko
- Department of Neurobiology and Anatomy, Wake Forest University School of Medicine, Winston-Salem NC 27157
| |
Collapse
|
28
|
Seo Y, Kim HS, Shin Y, Kang I, Choi SW, Yu KR, Seo KW, Kang KS. Excessive microglial activation aggravates olfactory dysfunction by impeding the survival of newborn neurons in the olfactory bulb of Niemann-Pick disease type C1 mice. Biochim Biophys Acta Mol Basis Dis 2014; 1842:2193-203. [PMID: 25132229 DOI: 10.1016/j.bbadis.2014.08.005] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2014] [Revised: 08/07/2014] [Accepted: 08/08/2014] [Indexed: 12/26/2022]
Abstract
Progressive olfactory impairment is one of the earliest markers of neurodegeneration. However, the underlying mechanism for this dysfunction remains unclear. The present study investigated the possible role of microgliosis in olfactory deficits using a mouse model of Niemann-Pick disease type C1 (NPC1), which is an incurable neurodegenerative disorder with disrupted lipid trafficking. At 7weeks of age, NPC1 mutants showed a distinct olfactory impairment in an olfactory test compared with age-matched wild-type controls (WT). The marked loss of olfactory sensory neurons within the NPC1 affected olfactory bulb (NPC1-OB) suggests that NPC1 dysfunction impairs olfactory structure. Furthermore, the pool of neuroblasts in the OB was diminished in NPC1 mice despite the intact proliferative capacity of neural stem/progenitor cells in the subventricular zone. Instead, pro-inflammatory proliferating microglia accumulated extensively in the NPC1-OB as the disease progressed. To evaluate the impact of abnormal microglial activation on olfaction in NPC1 mice, a microglial inhibition study was performed using the anti-inflammatory agent Cyclosporin A (CsA). Importantly, long-term CsA treatment in NPC1 mice reduced reactive microgliosis, restored the survival of newly generated neurons in the OB and improved overall performance on the olfactory test. Therefore, our study highlights the possible role of microglia in the regulation of neuronal turnover in the OB and provides insight into the possible therapeutic applications of microglial inhibition in the attenuation or reversal of olfactory impairment.
Collapse
Affiliation(s)
- Yoojin Seo
- Adult Stem Cell Research Center, College of Veterinary Medicine, Seoul National University, Seoul 151-742, South Korea
| | - Hyung-Sik Kim
- Adult Stem Cell Research Center, College of Veterinary Medicine, Seoul National University, Seoul 151-742, South Korea; Institute for Stem Cell and Regenerative Medicine at Kangstem Biotech, Biotechnology Incubating Center, Seoul National University, Seoul 151-742, South Korea
| | - Yooyoung Shin
- Adult Stem Cell Research Center, College of Veterinary Medicine, Seoul National University, Seoul 151-742, South Korea
| | - Insung Kang
- Adult Stem Cell Research Center, College of Veterinary Medicine, Seoul National University, Seoul 151-742, South Korea
| | - Soon Won Choi
- Adult Stem Cell Research Center, College of Veterinary Medicine, Seoul National University, Seoul 151-742, South Korea
| | - Kyung-Rok Yu
- Adult Stem Cell Research Center, College of Veterinary Medicine, Seoul National University, Seoul 151-742, South Korea; Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul 151-742, South Korea
| | - Kwang-Won Seo
- Adult Stem Cell Research Center, College of Veterinary Medicine, Seoul National University, Seoul 151-742, South Korea; Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul 151-742, South Korea; Institute for Stem Cell and Regenerative Medicine at Kangstem Biotech, Biotechnology Incubating Center, Seoul National University, Seoul 151-742, South Korea.
| | - Kyung-Sun Kang
- Adult Stem Cell Research Center, College of Veterinary Medicine, Seoul National University, Seoul 151-742, South Korea; Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul 151-742, South Korea.
| |
Collapse
|
29
|
Bhumika S, Darras VM. Role of thyroid hormones in different aspects of nervous system regeneration in vertebrates. Gen Comp Endocrinol 2014; 203:86-94. [PMID: 24681191 DOI: 10.1016/j.ygcen.2014.03.017] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2013] [Revised: 03/08/2014] [Accepted: 03/11/2014] [Indexed: 12/19/2022]
Abstract
Spontaneous functional recovery from injury in the adult human nervous system is rare and trying to improve recovery remains a clinical challenge. Nervous system regeneration is a complicated sequence of events involving cell death or survival, cell proliferation, axon extension and remyelination, and finally reinnervation and functional recovery. Successful recovery depends on the cell-specific and time-dependent activation and repression of a wide variety of growth factors and guidance molecules. Thyroid hormones (THs), well known for their regulatory role in neurodevelopment, have recently emerged as important modulators of neuroregeneration. This review focuses on the endogenous changes in the proteins regulating TH availability and action in different cell types of the adult mammalian nervous system during regeneration as well as the impact of TH supplementation on the consecutive steps in this process. It also addresses possible differences in TH involvement between different vertebrate classes, early or late developmental stages and peripheral or central nervous system. The available data show that THs are able to stimulate many signaling pathways necessary for successful neurogeneration. They however also suggest that supplementation with T4 and/or T3 may have beneficial or detrimental influences depending on the dose and more importantly on the specific phase of the regeneration process.
Collapse
Affiliation(s)
- Stitipragyan Bhumika
- Laboratory of Comparative Endocrinology, Division Animal Physiology and Neurobiology, Biology Department, KU Leuven, B-3000 Leuven, Belgium
| | - Veerle M Darras
- Laboratory of Comparative Endocrinology, Division Animal Physiology and Neurobiology, Biology Department, KU Leuven, B-3000 Leuven, Belgium.
| |
Collapse
|
30
|
Abstract
Adult neurogenesis, a developmental process of generating functionally integrated neurons, occurs throughout life in the hippocampus of the mammalian brain and showcases the highly plastic nature of the mature central nervous system. Significant progress has been made in recent years to decipher how adult neurogenesis contributes to brain functions. Here we review recent findings that inform our understanding of adult hippocampal neurogenesis processes and special properties of adult-born neurons. We further discuss potential roles of adult-born neurons at the circuitry and behavioral levels in cognitive and affective functions and how their dysfunction may contribute to various brain disorders. We end by considering a general model proposing that adult neurogenesis is not a cell-replacement mechanism, but instead maintains a plastic hippocampal neuronal circuit via the continuous addition of immature, new neurons with unique properties and structural plasticity of mature neurons induced by new-neuron integration.
Collapse
|
31
|
Di Liegro CM, Schiera G, Di Liegro I. Regulation of mRNA transport, localization and translation in the nervous system of mammals (Review). Int J Mol Med 2014; 33:747-62. [PMID: 24452120 PMCID: PMC3976132 DOI: 10.3892/ijmm.2014.1629] [Citation(s) in RCA: 81] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2013] [Accepted: 12/09/2013] [Indexed: 12/13/2022] Open
Abstract
Post-transcriptional control of mRNA trafficking and metabolism plays a critical role in the actualization and fine tuning of the genetic program of cells, both in development and in differentiated tissues. Cis-acting signals, responsible for post-transcriptional regulation, reside in the RNA message itself, usually in untranslated regions, 5′ or 3′ to the coding sequence, and are recognized by trans-acting factors: RNA-binding proteins (RBPs) and/or non-coding RNAs (ncRNAs). ncRNAs bind short mRNA sequences usually present in the 3′-untranslated (3′-UTR) region of their target messages. RBPs recognize specific nucleotide sequences and/or secondary/tertiary structures. Most RBPs assemble on mRNA at the moment of transcription and shepherd it to its destination, somehow determining its final fate. Regulation of mRNA localization and metabolism has a particularly important role in the nervous system where local translation of pre-localized mRNAs has been implicated in developing axon and dendrite pathfinding, and in synapse formation. Moreover, activity-dependent mRNA trafficking and local translation may underlie long-lasting changes in synaptic efficacy, responsible for learning and memory. This review focuses on the role of RBPs in neuronal development and plasticity, as well as possible connections between ncRNAs and RBPs.
Collapse
Affiliation(s)
- Carlo Maria Di Liegro
- Department of Biological Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), I-90128 Palermo, Italy
| | - Gabriella Schiera
- Department of Biological Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), I-90128 Palermo, Italy
| | - Italia Di Liegro
- Department of Experimental Biomedicine and Clinical Neurosciences (BIONEC), University of Palermo, I-90127 Palermo, Italy
| |
Collapse
|
32
|
Maine AR, Powers SD, Lutterschmidt DI. Seasonal Variation in Cell Proliferation and Cell Migration in the Brain of Adult Red-Sided Garter Snakes(Thamnophis sirtalis parietalis). BRAIN, BEHAVIOR AND EVOLUTION 2014; 84:181-96. [DOI: 10.1159/000364778] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/18/2013] [Accepted: 12/13/2013] [Indexed: 11/19/2022]
|
33
|
Xuan W, Vatansever F, Huang L, Hamblin MR. Transcranial low-level laser therapy enhances learning, memory, and neuroprogenitor cells after traumatic brain injury in mice. JOURNAL OF BIOMEDICAL OPTICS 2014; 19:108003. [PMID: 25292167 PMCID: PMC4189010 DOI: 10.1117/1.jbo.19.10.108003] [Citation(s) in RCA: 86] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/04/2014] [Accepted: 09/15/2014] [Indexed: 05/09/2023]
Abstract
The use of transcranial low-level laser (light) therapy (tLLLT) to treat stroke and traumatic brain injury (TBI) is attracting increasing attention. We previously showed that LLLT using an 810-nm laser 4 h after controlled cortical impact (CCI)-TBI in mice could significantly improve the neurological severity score, decrease lesion volume, and reduce Fluoro-Jade staining for degenerating neurons. We obtained some evidence for neurogenesis in the region of the lesion. We now tested the hypothesis that tLLLT can improve performance on the Morris water maze (MWM, learning, and memory) and increase neurogenesis in the hippocampus and subventricular zone (SVZ) after CCI-TBI in mice. One and (to a greater extent) three daily laser treatments commencing 4-h post-TBI improved neurological performance as measured by wire grip and motion test especially at 3 and 4 weeks post-TBI. Improvements in visible and hidden platform latency and probe tests in MWM were seen at 4 weeks. Caspase-3 expression was lower in the lesion region at 4 days post-TBI. Double-stained BrdU-NeuN (neuroprogenitor cells) was increased in the dentate gyrus and SVZ. Increases in double-cortin (DCX) and TUJ-1 were also seen. Our study results suggest that tLLLT may improve TBI both by reducing cell death in the lesion and by stimulating neurogenesis.
Collapse
Affiliation(s)
- Weijun Xuan
- Ruikang Hospital Affiliated to Guangxi University of Chinese Medicine, Department of Otolaryngology, Nanning 530021, China
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, Massachusetts 02114, United States
- Harvard Medical School, Department of Dermatology, Boston, Massachusetts 02115, United States
| | - Fatma Vatansever
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, Massachusetts 02114, United States
- Harvard Medical School, Department of Dermatology, Boston, Massachusetts 02115, United States
| | - Liyi Huang
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, Massachusetts 02114, United States
- Harvard Medical School, Department of Dermatology, Boston, Massachusetts 02115, United States
- Guangxi Medical University, First Affiliated College and Hospital, Department of Infectious Diseases, Nanning 530021, China
| | - Michael R. Hamblin
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, Massachusetts 02114, United States
- Harvard Medical School, Department of Dermatology, Boston, Massachusetts 02115, United States
- Harvard-MIT Division of Health Sciences and Technology, Cambridge, Massachusetts 02139, United States
- Address all correspondence to: Michael R. Hamblin E-mail:
| |
Collapse
|