1
|
Vassal M, Martins F, Monteiro B, Tambaro S, Martinez-Murillo R, Rebelo S. Emerging Pro-neurogenic Therapeutic Strategies for Neurodegenerative Diseases: A Review of Pre-clinical and Clinical Research. Mol Neurobiol 2025; 62:46-76. [PMID: 38816676 PMCID: PMC11711580 DOI: 10.1007/s12035-024-04246-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Accepted: 05/14/2024] [Indexed: 06/01/2024]
Abstract
The neuroscience community has largely accepted the notion that functional neurons can be generated from neural stem cells in the adult brain, especially in two brain regions: the subventricular zone of the lateral ventricles and the subgranular zone in the dentate gyrus of the hippocampus. However, impaired neurogenesis has been observed in some neurodegenerative diseases, particularly in Alzheimer's, Parkinson's, and Huntington's diseases, and also in Lewy Body dementia. Therefore, restoration of neurogenic function in neurodegenerative diseases emerges as a potential therapeutic strategy to counteract, or at least delay, disease progression. Considering this, the present study summarizes the different neuronal niches, provides a collection of the therapeutic potential of different pro-neurogenic strategies in pre-clinical and clinical research, providing details about their possible modes of action, to guide future research and clinical practice.
Collapse
Affiliation(s)
- Mariana Vassal
- Department of Medical Sciences, Institute of Biomedicine (iBiMED), University of Aveiro, Aveiro, Portugal
| | - Filipa Martins
- Department of Medical Sciences, Institute of Biomedicine (iBiMED), University of Aveiro, Aveiro, Portugal
| | - Bruno Monteiro
- Department of Medical Sciences, Institute of Biomedicine (iBiMED), University of Aveiro, Aveiro, Portugal
| | - Simone Tambaro
- Department of Neurobiology, Care Sciences and Society, Division of Neurogeriatrics, Karolinska Institutet, Huddinge, Sweden
| | - Ricardo Martinez-Murillo
- Neurovascular Research Group, Department of Translational Neurobiology, Cajal Institute (CSIC), Madrid, Spain
| | - Sandra Rebelo
- Department of Medical Sciences, Institute of Biomedicine (iBiMED), University of Aveiro, Aveiro, Portugal.
| |
Collapse
|
2
|
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
|
3
|
Zhang L, Zetter MA, Hernández VS, Hernández-Pérez OR, Jáuregui-Huerta F, Krabichler Q, Grinevich V. Morphological Signatures of Neurogenesis and Neuronal Migration in Hypothalamic Vasopressinergic Magnocellular Nuclei of the Adult Rat. Int J Mol Sci 2024; 25:6988. [PMID: 39000096 PMCID: PMC11241681 DOI: 10.3390/ijms25136988] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 06/17/2024] [Accepted: 06/17/2024] [Indexed: 07/16/2024] Open
Abstract
The arginine vasopressin (AVP)-magnocellular neurosecretory system (AVPMNS) in the hypothalamus plays a critical role in homeostatic regulation as well as in allostatic motivational behaviors. However, it remains unclear whether adult neurogenesis exists in the AVPMNS. By using immunoreaction against AVP, neurophysin II, glial fibrillar acidic protein (GFAP), cell division marker (Ki67), migrating neuroblast markers (doublecortin, DCX), microglial marker (Ionized calcium binding adaptor molecule 1, Iba1), and 5'-bromo-2'-deoxyuridine (BrdU), we report morphological evidence that low-rate neurogenesis and migration occur in adult AVPMNS in the rat hypothalamus. Tangential AVP/GFAP migration routes and AVP/DCX neuronal chains as well as ascending AVP axonal scaffolds were observed. Chronic water deprivation significantly increased the BrdU+ nuclei within both the supraaoptic (SON) and paraventricular (PVN) nuclei. These findings raise new questions about AVPMNS's potential hormonal role for brain physiological adaptation across the lifespan, with possible involvement in coping with homeostatic adversities.
Collapse
Affiliation(s)
- Limei Zhang
- Department of Physiology, School of Medicine, National Autonomous University of Mexico, Mexico City 04510, Mexico; (M.A.Z.); (V.S.H.); (O.R.H.-P.)
- Section on Molecular Neuroscience, National Institute of Mental Health, NIH, Bethesda, MD 20892, USA
| | - Mario A. Zetter
- Department of Physiology, School of Medicine, National Autonomous University of Mexico, Mexico City 04510, Mexico; (M.A.Z.); (V.S.H.); (O.R.H.-P.)
- Department of Medicine and Health, University of La Salle, Mexico City 14000, Mexico
| | - Vito S. Hernández
- Department of Physiology, School of Medicine, National Autonomous University of Mexico, Mexico City 04510, Mexico; (M.A.Z.); (V.S.H.); (O.R.H.-P.)
- Section on Molecular Neuroscience, National Institute of Mental Health, NIH, Bethesda, MD 20892, USA
| | - Oscar R. Hernández-Pérez
- Department of Physiology, School of Medicine, National Autonomous University of Mexico, Mexico City 04510, Mexico; (M.A.Z.); (V.S.H.); (O.R.H.-P.)
| | - Fernando Jáuregui-Huerta
- Department of Physiology, School of Medicine, National Autonomous University of Mexico, Mexico City 04510, Mexico; (M.A.Z.); (V.S.H.); (O.R.H.-P.)
| | - Quirin Krabichler
- Department of Neuropeptide Research in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, 69120 Mannheim, Germany; (Q.K.); (V.G.)
| | - Valery Grinevich
- Department of Neuropeptide Research in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, 69120 Mannheim, Germany; (Q.K.); (V.G.)
| |
Collapse
|
4
|
Lathe R, St Clair D. Programmed ageing: decline of stem cell renewal, immunosenescence, and Alzheimer's disease. Biol Rev Camb Philos Soc 2023; 98:1424-1458. [PMID: 37068798 DOI: 10.1111/brv.12959] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 03/27/2023] [Accepted: 03/30/2023] [Indexed: 04/19/2023]
Abstract
The characteristic maximum lifespan varies enormously across animal species from a few hours to hundreds of years. This argues that maximum lifespan, and the ageing process that itself dictates lifespan, are to a large extent genetically determined. Although controversial, this is supported by firm evidence that semelparous species display evolutionarily programmed ageing in response to reproductive and environmental cues. Parabiosis experiments reveal that ageing is orchestrated systemically through the circulation, accompanied by programmed changes in hormone levels across a lifetime. This implies that, like the circadian and circannual clocks, there is a master 'clock of age' (circavital clock) located in the limbic brain of mammals that modulates systemic changes in growth factor and hormone secretion over the lifespan, as well as systemic alterations in gene expression as revealed by genomic methylation analysis. Studies on accelerated ageing in mice, as well as human longevity genes, converge on evolutionarily conserved fibroblast growth factors (FGFs) and their receptors, including KLOTHO, as well as insulin-like growth factors (IGFs) and steroid hormones, as key players mediating the systemic effects of ageing. Age-related changes in these and multiple other factors are inferred to cause a progressive decline in tissue maintenance through failure of stem cell replenishment. This most severely affects the immune system, which requires constant renewal from bone marrow stem cells. Age-related immune decline increases risk of infection whereas lifespan can be extended in germfree animals. This and other evidence suggests that infection is the major cause of death in higher organisms. Immune decline is also associated with age-related diseases. Taking the example of Alzheimer's disease (AD), we assess the evidence that AD is caused by immunosenescence and infection. The signature protein of AD brain, Aβ, is now known to be an antimicrobial peptide, and Aβ deposits in AD brain may be a response to infection rather than a cause of disease. Because some cognitively normal elderly individuals show extensive neuropathology, we argue that the location of the pathology is crucial - specifically, lesions to limbic brain are likely to accentuate immunosenescence, and could thus underlie a vicious cycle of accelerated immune decline and microbial proliferation that culminates in AD. This general model may extend to other age-related diseases, and we propose a general paradigm of organismal senescence in which declining stem cell proliferation leads to programmed immunosenescence and mortality.
Collapse
Affiliation(s)
- Richard Lathe
- Division of Infection Medicine, Chancellor's Building, University of Edinburgh Medical School, Little France, Edinburgh, EH16 4SB, UK
| | - David St Clair
- Institute of Medical Sciences, School of Medicine, University of Aberdeen, Aberdeen, AB25 2ZD, UK
| |
Collapse
|
5
|
Sufieva DA, Korzhevskii DE. Proliferative Markers and Neural Stem Cells Markers in Tanycytes of the Third Cerebral Ventricle in Rats. Bull Exp Biol Med 2023; 174:564-570. [PMID: 36894817 DOI: 10.1007/s10517-023-05748-8] [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: 09/06/2022] [Indexed: 03/11/2023]
Abstract
The proliferative properties of tanycyte subpopulations during postnatal development and aging were studied. Using immunohistochemical markers, we described the distribution of proliferative markers and markers of neural stem cells (NSC) in 4 tanycyte subpopulations (α1-, α2-, β1-, and β2-tanycytes). During the first postnatal week, all tanycyte subpopulations exhibit proliferative activity. During aging, β-tanycytes lose their proliferative activity and retain a limited set of NSC markers, whereas α-tanycytes maintain both the ability to proliferate and the properties of NSC throughout the entire postnatal development including aging. The data obtained significantly improve modern understanding of the proliferative potential of tanycytes and their subpopulation differences in early postnatal period and during aging.
Collapse
Affiliation(s)
- D A Sufieva
- Institute of Experimental Medicine, St. Petersburg, Russia.
| | | |
Collapse
|
6
|
Inada H, Corales LG, Osumi N. A novel feature of the ancient organ: A possible involvement of the subcommissural organ in neurogenic/gliogenic potential in the adult brain. Front Neurosci 2023; 17:1141913. [PMID: 36960167 PMCID: PMC10027738 DOI: 10.3389/fnins.2023.1141913] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 02/20/2023] [Indexed: 03/09/2023] Open
Abstract
The subcommissural organ (SCO) is a circumventricular organ highly conserved in vertebrates from Cyclostomata such as lamprey to mammals including human. The SCO locates in the boundary between the third ventricle and the entrance of the aqueduct of Sylvius. The SCO functions as a secretory organ producing a variety of proteins such as SCO-spondin, transthyretin, and basic fibroblast growth factor (FGF) into the cerebrospinal fluid (CSF). A significant contribution of the SCO has been thought to maintain the homeostasis of CSF dynamics. However, evidence has shown a possible role of SCO on neurogenesis in the adult brain. This review highlights specific features of the SCO related to adult neurogenesis, suggested by the progress of understanding SCO functions. We begin with a brief history of the SCO discovery and continue to structural features, gene expression, and a possible role in adult neurogenesis suggested by the SCO transplant experiment.
Collapse
Affiliation(s)
- Hitoshi Inada
- Laboratory of Health and Sports Sciences, Division of Biomedical Engineering for Health and Welfare, Graduate School of Biomedical Engineering, Tohoku University, Sendai, Japan
- Department of Developmental Neuroscience, Graduate School of Medicine, Tohoku University, Sendai, Japan
- *Correspondence: Hitoshi Inada,
| | - Laarni Grace Corales
- Department of Developmental Neuroscience, Graduate School of Medicine, Tohoku University, Sendai, Japan
| | - Noriko Osumi
- Department of Developmental Neuroscience, Graduate School of Medicine, Tohoku University, Sendai, Japan
| |
Collapse
|
7
|
Ball GF, Balthazart J. In memoriam - Mei-Fang Cheng (1938 to 2022). Horm Behav 2022; 146:105256. [PMID: 36103748 DOI: 10.1016/j.yhbeh.2022.105256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Affiliation(s)
- Gregory F Ball
- Department of Psychology, University of Maryland, College Park, United States of America
| | | |
Collapse
|
8
|
Ou Y, Zhou M, Che M, Gong H, Wu G, Peng J, Li K, Yang R, Wang X, Zhang X, Liu Y, Feng Z, Qi S. Adult neurogenesis of the median eminence contributes to structural reconstruction and recovery of body fluid metabolism in hypothalamic self-repair after pituitary stalk lesion. Cell Mol Life Sci 2022; 79:458. [PMID: 35907165 PMCID: PMC11073094 DOI: 10.1007/s00018-022-04457-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 06/26/2022] [Accepted: 06/27/2022] [Indexed: 01/25/2023]
Abstract
Body fluid homeostasis is critical to survival. The integrity of the hypothalamo-neurohypophysial system (HNS) is an important basis of the precise regulation of body fluid metabolism and arginine vasopressin (AVP) hormone release. Clinically, some patients with central diabetes insipidus (CDI) due to HNS lesions can experience recovery compensation of body fluid metabolism. However, whether the hypothalamus has the potential for structural plasticity and self-repair under pathological conditions remains unclear. Here, we report the repair and reconstruction of a new neurohypophysis-like structure in the hypothalamic median eminence (ME) after pituitary stalk electrical lesion (PEL). We show that activated and proliferating adult neural progenitor cells differentiate into new mature neurons, which then integrate with remodeled AVP fibers to reconstruct the local AVP hormone release neural circuit in the ME after PEL. We found that the transcription factor of NK2 homeobox 1 (NKX2.1) and the sonic hedgehog signaling pathway, mediated by NKX2.1, are the key regulators of adult hypothalamic neurogenesis. Taken together, our study provides evidence that adult ME neurogenesis is involved in the structural reconstruction of the AVP release circuit and eventually restores body fluid metabolic homeostasis during hypothalamic self-repair.
Collapse
Affiliation(s)
- Yichao Ou
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
- Laboratory of Precision Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Mingfeng Zhou
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
- Laboratory of Precision Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Mengjie Che
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
- Laboratory of Precision Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Haodong Gong
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Guangsen Wu
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Junjie Peng
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
- Laboratory of Precision Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Kai Li
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Runwei Yang
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Xingqin Wang
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
- Laboratory of Precision Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Xian Zhang
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
- Laboratory of Precision Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Yawei Liu
- Laboratory of Precision Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Zhanpeng Feng
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China.
- Laboratory of Precision Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China.
| | - Songtao Qi
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China.
- Laboratory of Precision Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China.
| |
Collapse
|
9
|
Yaghmaeian Salmani B, Balderson B, Bauer S, Ekman H, Starkenberg A, Perlmann T, Piper M, Bodén M, Thor S. Selective requirement for polycomb repressor complex 2 in the generation of specific hypothalamic neuronal subtypes. Development 2022; 149:274592. [PMID: 35245348 PMCID: PMC8959139 DOI: 10.1242/dev.200076] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Accepted: 01/18/2022] [Indexed: 11/20/2022]
Abstract
The hypothalamus displays staggering cellular diversity, chiefly established during embryogenesis by the interplay of several signalling pathways and a battery of transcription factors. However, the contribution of epigenetic cues to hypothalamus development remains unclear. We mutated the polycomb repressor complex 2 gene Eed in the developing mouse hypothalamus, which resulted in the loss of H3K27me3, a fundamental epigenetic repressor mark. This triggered ectopic expression of posteriorly expressed regulators (e.g. Hox homeotic genes), upregulation of cell cycle inhibitors and reduced proliferation. Surprisingly, despite these effects, single cell transcriptomic analysis revealed that most neuronal subtypes were still generated in Eed mutants. However, we observed an increase in glutamatergic/GABAergic double-positive cells, as well as loss/reduction of dopamine, hypocretin and Tac2-Pax6 neurons. These findings indicate that many aspects of the hypothalamic gene regulatory flow can proceed without the key H3K27me3 epigenetic repressor mark, but points to a unique sensitivity of particular neuronal subtypes to a disrupted epigenomic landscape. Summary: Polycomb repressor complex 2 inactivation results in selective effects on mouse hypothalamic development, increasing glutamatergic/GABA cells, while reducing dopamine, Hcrt and Tac2-Pax6 cells.
Collapse
Affiliation(s)
- Behzad Yaghmaeian Salmani
- Department of Clinical and Experimental Medicine, Linkoping University, SE-58185 Linkoping, Sweden
- Department of Cell and Molecular Biology, Karolinska Institute, SE-17177 Stockholm, Sweden
| | - Brad Balderson
- School of Chemistry and Molecular Biosciences, University of Queensland, St Lucia, QLD 4072, Australia
| | - Susanne Bauer
- Department of Clinical and Experimental Medicine, Linkoping University, SE-58185 Linkoping, Sweden
| | - Helen Ekman
- Department of Clinical and Experimental Medicine, Linkoping University, SE-58185 Linkoping, Sweden
| | - Annika Starkenberg
- Department of Clinical and Experimental Medicine, Linkoping University, SE-58185 Linkoping, Sweden
| | - Thomas Perlmann
- Department of Cell and Molecular Biology, Karolinska Institute, SE-17177 Stockholm, Sweden
| | - Michael Piper
- School of Biomedical Sciences, University of Queensland, St Lucia, QLD 4072, Australia
| | - Mikael Bodén
- School of Chemistry and Molecular Biosciences, University of Queensland, St Lucia, QLD 4072, Australia
| | - Stefan Thor
- Department of Clinical and Experimental Medicine, Linkoping University, SE-58185 Linkoping, Sweden
- School of Biomedical Sciences, University of Queensland, St Lucia, QLD 4072, Australia
| |
Collapse
|
10
|
Does Sertraline Affect Hypothalamic Food Intake Peptides in the Rat Experimental Model of Chronic Mild Stress-Induced Depression? Neurochem Res 2022; 47:1299-1316. [PMID: 35080689 DOI: 10.1007/s11064-022-03529-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 01/12/2022] [Accepted: 01/15/2022] [Indexed: 10/19/2022]
Abstract
Depression is a chronic, recurrent and life-threatening disease affecting approximately 15% of the world population. Depression is responsible for neuropathologies like decreased neurogenesis and increased dendritic atrophy. Antidepressant treatments increase hippocampal neurogenesis and neurotrophic factor expression. Based on this information, it was aimed to investigate effect of sertraline on depression in rats with chronic mild stress (CMS) model and to determine how it affects cell proliferation and hypothalamic peptide levels in hypothalamus. 56 adult male Wistar albino; control, depression(D), depression + sertraline, sertraline were divided into groups. Various stressors were applied to D for 30 days. Open field test (OFT) and forced swimming test (FST) were conducted to check whether the animals were depressed. On the 16th day osmotic minipump was placed subcutaneously and sertraline (10 mg/kg/day) was administered for 15 days. Behavior tests were done. Hypothalamic peptide gene expression levels were analyzed by quantitative RT-PCR. Statistical evaluations were made using ANOVA. It caused a decrease in the percentage of movement in the D and control groups in the OFT, an increase in the immobility time in the D group in the FST, and an increase in the swimming behavior in the DS group. Animals did not show any anxiological behavior based on the elevated plus maze test results. CMS caused a decrease in GLUT2 and NPY gene expression in the hypothalamus of animals, an increase in POMC and FGFR2, and an increase in IGFIR and GLUT2 gene expression in the DS group. Sertraline has been shown to ameliorate the effects of CMS-induced depression. Sertraline is thought to have a positive regulatory effect on both the formation of neural precursor cells and the survival of newly formed neurons in the hypothalamus. Newly formed neurons in the hypothalamus express food intake-related NPY, POMC, GLUT2 neurons, and thus hypothalamic tanycytes may play a key role in the control of energy metabolism.
Collapse
|
11
|
Soumier A, Habart M, Lio G, Demily C, Sirigu A. Differential fate between oxytocin and vasopressin cells in the developing mouse brain. iScience 2022; 25:103655. [PMID: 35028535 PMCID: PMC8741612 DOI: 10.1016/j.isci.2021.103655] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 12/03/2021] [Accepted: 12/14/2021] [Indexed: 11/25/2022] Open
Abstract
Oxytocin (OXT) and arginine vasopressin (AVP), two neuropeptides involved in socio-emotional behaviors have been anatomically defined in the adult brain. Yet their spatial organization during postnatal development is not clearly defined. We built a developmental atlas using 3D imaging of cleared immunolabeled tissue over four early postnatal (P) stages, from birth (P0, P3, P7, P14) to young adulthood (≥P56). Our atlas-based mapping revealed that the number of OXT neurons doubles according to unique temporal dynamics in selective hypothalamic regions, namely, the periventricular and paraventricular nuclei, and in a novel location we named the antero-lateral preoptic. In the paraventricular nucleus, single-cell densities and fluorescence analysis demonstrated selective expansion of OXT cells in the antero-ventral division, whereas the postero-dorsal division contained cells present at birth. No changes were observed for AVP neurons. Our findings show the coexisting of innate and plastic OXT/AVP brain circuits probably triggered by environmental adaptation of the social brain. The OXT system continues to mature during early development but not the AVP system OXT hypothalamic nuclei emerge at different rates after birth PVN cells gradually acquire an oxytocinergic phenotype OXT cells are organized into innate and adaptive neural networks
Collapse
Affiliation(s)
- Amelie Soumier
- iMIND, Center of Excellence for Autism, le Vinatier Hospital, Bron, France.,Institute of Cognitive Science Marc Jeannerod, CNRS, Bron, France
| | - Marie Habart
- iMIND, Center of Excellence for Autism, le Vinatier Hospital, Bron, France.,Institute of Cognitive Science Marc Jeannerod, CNRS, Bron, France
| | - Guillaume Lio
- iMIND, Center of Excellence for Autism, le Vinatier Hospital, Bron, France.,Institute of Cognitive Science Marc Jeannerod, CNRS, Bron, France
| | - Caroline Demily
- iMIND, Center of Excellence for Autism, le Vinatier Hospital, Bron, France.,Institute of Cognitive Science Marc Jeannerod, CNRS, Bron, France
| | - Angela Sirigu
- iMIND, Center of Excellence for Autism, le Vinatier Hospital, Bron, France.,Institute of Cognitive Science Marc Jeannerod, CNRS, Bron, France
| |
Collapse
|
12
|
Chatterjee O, Gopalakrishnan L, Mol P, Advani J, Nair B, Shankar SK, Mahadevan A, Prasad TSK. The Normal Human Adult Hypothalamus Proteomic Landscape: Rise of Neuroproteomics in Biological Psychiatry and Systems Biology. OMICS-A JOURNAL OF INTEGRATIVE BIOLOGY 2021; 25:693-710. [PMID: 34714154 DOI: 10.1089/omi.2021.0158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
The human hypothalamus is central to the regulation of neuroendocrine and neurovegetative systems, as well as modulation of chronobiology and behavioral aspects in human health and disease. Surprisingly, a deep proteomic analysis of the normal human hypothalamic proteome has been missing for such an important organ so far. In this study, we delineated the human hypothalamus proteome using a high-resolution mass spectrometry approach which resulted in the identification of 5349 proteins, while a multiple post-translational modification (PTM) search identified 191 additional proteins, which were missed in the first search. A proteogenomic analysis resulted in the discovery of multiple novel protein-coding regions as we identified proteins from noncoding regions (pseudogenes) and proteins translated from short open reading frames that can be missed using the traditional pipeline of prediction of protein-coding genes as a part of genome annotation. We also identified several PTMs of hypothalamic proteins that may be required for normal hypothalamic functions. Moreover, we observed an enrichment of proteins pertaining to autophagy and adult neurogenesis in the proteome data. We believe that the hypothalamic proteome reported herein would help to decipher the molecular basis for the diverse range of physiological functions attributed to it, as well as its role in neurological and psychiatric diseases. Extensive proteomic profiling of the hypothalamic nuclei would further elaborate on the role and functional characterization of several hypothalamus-specific proteins and pathways to inform future research and clinical discoveries in biological psychiatry, neurology, and system biology.
Collapse
Affiliation(s)
- Oishi Chatterjee
- Institute of Bioinformatics, Bangalore India.,Amrita School of Biotechnology, Amrita University, Kollam, India.,Center for Systems Biology and Molecular Medicine, Yenepoya Research Center, Yenepoya (Deemed to be University), Mangalore, India
| | - Lathika Gopalakrishnan
- Institute of Bioinformatics, Bangalore India.,Center for Systems Biology and Molecular Medicine, Yenepoya Research Center, Yenepoya (Deemed to be University), Mangalore, India.,Manipal Academy of Higher Education, Manipal, India
| | - Praseeda Mol
- Institute of Bioinformatics, Bangalore India.,Amrita School of Biotechnology, Amrita University, Kollam, India
| | | | - Bipin Nair
- Amrita School of Biotechnology, Amrita University, Kollam, India
| | - Susarla Krishna Shankar
- Department of Neuropathology, National Institute of Mental Health and Neurosciences, Bangalore, India.,Human Brain Tissue Repository, National Institute of Mental Health and Neurosciences, Bangalore, India
| | - Anita Mahadevan
- Department of Neuropathology, National Institute of Mental Health and Neurosciences, Bangalore, India.,Human Brain Tissue Repository, National Institute of Mental Health and Neurosciences, Bangalore, India
| | | |
Collapse
|
13
|
Suh JS, Fiori LM, Ali M, Harkness KL, Ramonas M, Minuzzi L, Hassel S, Strother SC, Zamyadi M, Arnott SR, Farzan F, Foster JA, Lam RW, MacQueen GM, Milev R, Müller DJ, Parikh SV, Rotzinger S, Sassi RB, Soares CN, Uher R, Kennedy SH, Turecki G, Frey BN. Hypothalamus volume and DNA methylation of stress axis genes in major depressive disorder: A CAN-BIND study report. Psychoneuroendocrinology 2021; 132:105348. [PMID: 34229186 DOI: 10.1016/j.psyneuen.2021.105348] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 06/14/2021] [Accepted: 06/25/2021] [Indexed: 11/28/2022]
Abstract
Dysfunction of the hypothalamic-pituitary-adrenal (HPA) axis is considered one of the mechanisms underlying the development of major depressive disorder (MDD), but the exact nature of this dysfunction is unknown. We investigated the relationship between hypothalamus volume (HV) and blood-derived DNA methylation in MDD. We obtained brain MRI, clinical and molecular data from 181 unmedicated MDD and 90 healthy control (HC) participants. MDD participants received a 16-week standardized antidepressant treatment protocol, as part of the first Canadian Biomarker Integration Network in Depression (CAN-BIND) study. We collected bilateral HV measures via manual segmentation by two independent raters. DNA methylation and RNA sequencing were performed for three key HPA axis-regulating genes coding for the corticotropin-binding protein (CRHBP), glucocorticoid receptor (NR3C1) and FK506 binding protein 5 (FKBP5). We used elastic net regression to perform variable selection and assess predictive ability of methylation variables on HV. Left HV was negatively associated with duration of current episode (ρ = -0.17, p = 0.035). We did not observe significant differences in HV between MDD and HC or any associations between HV and treatment response at weeks 8 or 16, overall depression severity, illness duration or childhood maltreatment. We also did not observe any differentially methylated CpG sites between MDD and HC groups. After assessing functionality by correlating methylation levels with RNA expression of the respective genes, we observed that the number of functionally relevant CpG sites differed between MDD and HC groups in FKBP5 (χ2 = 77.25, p < 0.0001) and NR3C1 (χ2 = 7.29, p = 0.007). Cross-referencing functionally relevant CpG sites to those that were highly ranked in predicting HV in elastic net modeling identified one site from FKBP5 (cg03591753) and one from NR3C1 (cg20728768) within the MDD group. Stronger associations between DNA methylation, gene expression and HV in MDD suggest a novel putative molecular pathway of stress-related sensitivity in depression. Future studies should consider utilizing the epigenome and ultra-high field MR data which would allow the investigation of HV sub-fields.
Collapse
Affiliation(s)
- Jee Su Suh
- Neuroscience Graduate Program, McMaster University, Hamilton, ON, Canada; Mood Disorders Program and Women's Health Concerns Clinic, St. Joseph's Healthcare Hamilton, ON, Canada
| | - Laura M Fiori
- Douglas Mental Health University Institute, Department of Psychiatry, McGill University, Montreal, QC, Canada
| | - Mohammad Ali
- Neuroscience Graduate Program, McMaster University, Hamilton, ON, Canada; Mood Disorders Program and Women's Health Concerns Clinic, St. Joseph's Healthcare Hamilton, ON, Canada
| | - Kate L Harkness
- Department of Psychology, Queen's University, Kingston, ON, Canada
| | - Milita Ramonas
- Juravinski Hospital and Cancer Centre, Hamilton Health Sciences, Hamilton, ON, Canada
| | - Luciano Minuzzi
- Neuroscience Graduate Program, McMaster University, Hamilton, ON, Canada; Mood Disorders Program and Women's Health Concerns Clinic, St. Joseph's Healthcare Hamilton, ON, Canada; Department of Psychiatry and Behavioural Neurosciences, McMaster University, Hamilton, ON, Canada
| | - Stefanie Hassel
- Department of Psychiatry, University of Calgary, Calgary, AB, Canada
| | | | - Mojdeh Zamyadi
- Rotman Research Institute, Baycrest, Toronto, ON, Canada
| | | | - Faranak Farzan
- eBrain Lab, School of Mechatronic Systems Engineering, Simon Fraser University, Surrey, BC, Canada
| | - Jane A Foster
- Department of Psychiatry and Behavioural Neurosciences, McMaster University, Hamilton, ON, Canada
| | - Raymond W Lam
- Department of Psychiatry, University of British Columbia, Vancouver, BC, Canada
| | - Glenda M MacQueen
- Department of Psychiatry, University of Calgary, Calgary, AB, Canada
| | - Roumen Milev
- Departments of Psychiatry and Psychology, Queen's University, and Providence Care Hospital, Kingston, ON, Canada
| | - Daniel J Müller
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada; Centre for Addiction and Mental Health, Pharmacogenetics Research Clinic, Toronto, ON, Canada
| | - Sagar V Parikh
- University of Michigan Depression Center, Ann Arbor, MI, United States
| | - Susan Rotzinger
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada
| | - Roberto B Sassi
- Department of Psychiatry and Behavioural Neurosciences, McMaster University, Hamilton, ON, Canada
| | - Claudio N Soares
- Departments of Psychiatry and Psychology, Queen's University, and Providence Care Hospital, Kingston, ON, Canada
| | - Rudolf Uher
- Department of Psychiatry, Dalhousie University, Halifax, NS, Canada
| | - Sidney H Kennedy
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada; Centre for Depression and Suicide Studies, St. Michael's Hospital, Toronto, ON, Canada
| | - Gustavo Turecki
- Douglas Mental Health University Institute, Department of Psychiatry, McGill University, Montreal, QC, Canada
| | - Benicio N Frey
- Neuroscience Graduate Program, McMaster University, Hamilton, ON, Canada; Mood Disorders Program and Women's Health Concerns Clinic, St. Joseph's Healthcare Hamilton, ON, Canada; Department of Psychiatry and Behavioural Neurosciences, McMaster University, Hamilton, ON, Canada.
| |
Collapse
|
14
|
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
|
15
|
Hamblin MH, Lee JP. Neural Stem Cells for Early Ischemic Stroke. Int J Mol Sci 2021; 22:ijms22147703. [PMID: 34299322 PMCID: PMC8306669 DOI: 10.3390/ijms22147703] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 07/14/2021] [Accepted: 07/15/2021] [Indexed: 12/11/2022] Open
Abstract
Clinical treatments for ischemic stroke are limited. Neural stem cell (NSC) transplantation can be a promising therapy. Clinically, ischemia and subsequent reperfusion lead to extensive neurovascular injury that involves inflammation, disruption of the blood-brain barrier, and brain cell death. NSCs exhibit multiple potentially therapeutic actions against neurovascular injury. Currently, tissue plasminogen activator (tPA) is the only FDA-approved clot-dissolving agent. While tPA’s thrombolytic role within the vasculature is beneficial, tPA’s non-thrombolytic deleterious effects aggravates neurovascular injury, restricting the treatment time window (time-sensitive) and tPA eligibility. Thus, new strategies are needed to mitigate tPA’s detrimental effects and quickly mediate vascular repair after stroke. Up to date, clinical trials focus on the impact of stem cell therapy on neuro-restoration by delivering cells during the chronic stroke stage. Also, NSCs secrete factors that stimulate endogenous repair mechanisms for early-stage ischemic stroke. This review will present an integrated view of the preclinical perspectives of NSC transplantation as a promising treatment for neurovascular injury, with an emphasis on early-stage ischemic stroke. Further, this will highlight the impact of early sub-acute NSC delivery on improving short-term and long-term stroke outcomes.
Collapse
Affiliation(s)
- Milton H. Hamblin
- Department of Pharmacology, Tulane University School of Medicine, 1430 Tulane Ave, New Orleans, LA 70112, USA
- Correspondence: (M.H.H.); (J.-P.L.)
| | - Jean-Pyo Lee
- Department of Physiology, Tulane University School of Medicine, 1430 Tulane Ave, New Orleans, LA 70112, USA
- Tulane Brain Institute, Tulane University, 1430 Tulane Ave, New Orleans, LA 70112, USA
- Correspondence: (M.H.H.); (J.-P.L.)
| |
Collapse
|
16
|
Kostin A, Alam MA, McGinty D, Alam MN. Adult hypothalamic neurogenesis and sleep-wake dysfunction in aging. Sleep 2021; 44:5986548. [PMID: 33202015 DOI: 10.1093/sleep/zsaa173] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 07/22/2020] [Indexed: 12/21/2022] Open
Abstract
In the mammalian brain, adult neurogenesis has been extensively studied in the hippocampal sub-granular zone and the sub-ventricular zone of the anterolateral ventricles. However, growing evidence suggests that new cells are not only "born" constitutively in the adult hypothalamus, but many of these cells also differentiate into neurons and glia and serve specific functions. The preoptic-hypothalamic area plays a central role in the regulation of many critical functions, including sleep-wakefulness and circadian rhythms. While a role for adult hippocampal neurogenesis in regulating hippocampus-dependent functions, including cognition, has been extensively studied, adult hypothalamic neurogenic process and its contributions to various hypothalamic functions, including sleep-wake regulation are just beginning to unravel. This review is aimed at providing the current understanding of the hypothalamic adult neurogenic processes and the extent to which it affects hypothalamic functions, including sleep-wake regulation. We propose that hypothalamic neurogenic processes are vital for maintaining the proper functioning of the hypothalamic sleep-wake and circadian systems in the face of regulatory challenges. Sleep-wake disturbance is a frequent and challenging problem of aging and age-related neurodegenerative diseases. Aging is also associated with a decline in the neurogenic process. We discuss a hypothesis that a decrease in the hypothalamic neurogenic process underlies the aging of its sleep-wake and circadian systems and associated sleep-wake disturbance. We further discuss whether neuro-regenerative approaches, including pharmacological and non-pharmacological stimulation of endogenous neural stem and progenitor cells in hypothalamic neurogenic niches, can be used for mitigating sleep-wake and other hypothalamic dysfunctions in aging.
Collapse
Affiliation(s)
- Andrey Kostin
- Research Service (151A3), Veterans Affairs Greater Los Angeles Healthcare System, Sepulveda, CA
| | - Md Aftab Alam
- Research Service (151A3), Veterans Affairs Greater Los Angeles Healthcare System, Sepulveda, CA.,Department of Psychiatry, University of California, Los Angeles, CA
| | - Dennis McGinty
- Research Service (151A3), Veterans Affairs Greater Los Angeles Healthcare System, Sepulveda, CA.,Department of Psychology, University of California, Los Angeles, CA
| | - Md Noor Alam
- Research Service (151A3), Veterans Affairs Greater Los Angeles Healthcare System, Sepulveda, CA.,Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA
| |
Collapse
|
17
|
Sinet F, Soty M, Zemdegs J, Guiard B, Estrada J, Malleret G, Silva M, Mithieux G, Gautier-Stein A. Dietary Fibers and Proteins Modulate Behavior via the Activation of Intestinal Gluconeogenesis. Neuroendocrinology 2021; 111:1249-1265. [PMID: 33429400 DOI: 10.1159/000514289] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Accepted: 01/07/2021] [Indexed: 11/19/2022]
Abstract
INTRODUCTION Several studies have suggested that diet, especially the one enriched in microbiota-fermented fibers or fat, regulates behavior. The underlying mechanisms are currently unknown. We previously reported that certain macronutrients (fermentable fiber and protein) regulate energy homeostasis via the activation of intestinal gluconeogenesis (IGN), which generates a neural signal to the brain. We hypothesized that these nutriments might control behavior using the same gut-brain circuit. METHODS Wild-type and IGN-deficient mice were fed chow or diets enriched in protein or fiber. Changes in their behavior were assessed using suited tests. Hippocampal neurogenesis, extracellular levels of serotonin, and protein expression levels were assessed by immunofluorescence, in vivo dialysis, and Western blotting, respectively. IGN was rescued by infusing glucose into the portal vein of IGN-deficient mice. RESULTS We show here that both fiber- and protein-enriched diets exert beneficial actions on anxiety-like and depressive-like behaviors. These benefits do not occur in mice lacking IGN. Consistently, IGN-deficient mice display hallmarks of depressive-like disorders, including decreased hippocampal neurogenesis, basal hyperactivity, and deregulation of the hypothalamic-pituitary-adrenal axis, which are associated with increased expression of the precursor of corticotropin-releasing hormone in the hypothalamus and decreased expression of the glucocorticoid receptor in the hippocampus. These neurobiological alterations are corrected by portal glucose infusion mimicking IGN. CONCLUSION IGN translates nutritional information, allowing the brain to finely coordinate energy metabolism and behavior.
Collapse
Affiliation(s)
- Flore Sinet
- INSERM UMR-S1213, Université Claude Bernard Lyon 1, Université de Lyon, Lyon, France
| | - Maud Soty
- INSERM UMR-S1213, Université Claude Bernard Lyon 1, Université de Lyon, Lyon, France
| | - Juliane Zemdegs
- CRCA - UMR 5169 - Université Paul Sabatier, Toulouse, France
| | - Bruno Guiard
- CRCA - UMR 5169 - Université Paul Sabatier, Toulouse, France
| | - Judith Estrada
- INSERM UMR-S1213, Université Claude Bernard Lyon 1, Université de Lyon, Lyon, France
| | - Gaël Malleret
- Forgetting and Cortical Dynamics, Lyon Neuroscience Research Center, Université de Lyon, Lyon, France
| | - Marine Silva
- INSERM UMR-S1213, Université Claude Bernard Lyon 1, Université de Lyon, Lyon, France
| | - Gilles Mithieux
- INSERM UMR-S1213, Université Claude Bernard Lyon 1, Université de Lyon, Lyon, France
| | | |
Collapse
|
18
|
Uppal SK, Uhlendorf TL, Nuryyev RL, Saenz J, Shanmugam M, Ochoa J, Van Trigt W, Malone CS, St. Julian AP, Kopyov O, Kopyov A, Cohen RW. Human neural progenitor cells ameliorate NMDA-induced hippocampal degeneration and related functional deficits. AIMS MEDICAL SCIENCE 2021. [DOI: 10.3934/medsci.2021021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
<abstract>
<p>It has been established that the CA3 region of the hippocampus is involved in consolidating short-term memory to long-term memory and aids in spatial navigation retention. Seizures and many neurologic diseases induce damage to that region of the hippocampus, resulting in deficits in memory consolidation and spatial navigation. Drug treatments have been proven to have limited effectiveness, but cell replacement therapy has demonstrated to be more promising. Celavie Biosciences have developed a multipotent, nontumorigenic human neural progenitor cell (hNPC) line shown to have the ability to migrate <italic>in situ</italic>, reducing structural and functional deficits in neurodegenerative animal models. Here, we examined whether transplanted hNPCs would reestablish the memories of Han-Wistar rats subjected to hippocampal excitotoxic lesioning. The rats were lesioned in the CA3c regions at 50 days bilaterally with the neurotoxin NMDA (1 µl containing 7.5 mg/ml; −3.5 mm AP; ±2.0 L and −2.5 V). At 54 days of age, live hNPCs (500000 cells in 5 µl cell suspension media), frozen-killed hNPCs (500000 cells/5 µl), HEK293T cells (500000 cells/5 µl) or vehicle (cell suspension media; 5 µl) were bilaterally implanted directly into the NMDA damaged area. The rats were tested two weeks later with three different memory tests: novel and place-object assays and the water-maze task. Results showed that rats receiving live hNPC implantation performed significantly better in the water maze task than control groups; yet, novel and place-object test results showed no significant differences among treatments. Histology confirmed the survival of implanted hNPCs after 28 days post-implantation as well as showing neuroprotective effects. This study showed that Celavie's hNPCs were able to survive and improve some but not all hippocampal functionality, emphasizing the promise for cell replacement therapeutics for neurodegenerative disorders.</p>
</abstract>
Collapse
|
19
|
Losurdo M, Grilli M. Extracellular Vesicles, Influential Players of Intercellular Communication within Adult Neurogenic Niches. Int J Mol Sci 2020; 21:E8819. [PMID: 33233420 PMCID: PMC7700666 DOI: 10.3390/ijms21228819] [Citation(s) in RCA: 10] [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: 10/19/2020] [Revised: 11/18/2020] [Accepted: 11/19/2020] [Indexed: 12/14/2022] Open
Abstract
Adult neurogenesis, involving the generation of functional neurons from adult neural stem cells (NSCs), occurs constitutively in discrete brain regions such as hippocampus, sub-ventricular zone (SVZ) and hypothalamus. The intrinsic structural plasticity of the neurogenic process allows the adult brain to face the continuously changing external and internal environment and requires coordinated interplay between all cell types within the specialized microenvironment of the neurogenic niche. NSC-, neuronal- and glia-derived factors, originating locally, regulate the balance between quiescence and self-renewal of NSC, their differentiation programs and the survival and integration of newborn cells. Extracellular Vesicles (EVs) are emerging as important mediators of cell-to-cell communication, representing an efficient way to transfer the biologically active cargos (nucleic acids, proteins, lipids) by which they modulate the function of the recipient cells. Current knowledge of the physiological role of EVs within adult neurogenic niches is rather limited. In this review, we will summarize and discuss EV-based cross-talk within adult neurogenic niches and postulate how EVs might play a critical role in the regulation of the neurogenic process.
Collapse
Affiliation(s)
| | - Mariagrazia Grilli
- Laboratory of Neuroplasticity, Department of Pharmaceutical Sciences, University of Piemonte Orientale, 28100 Novara, Italy;
| |
Collapse
|
20
|
de Araújo TM, Velloso LA. Hypothalamic IRX3: A New Player in the Development of Obesity. Trends Endocrinol Metab 2020; 31:368-377. [PMID: 32035736 DOI: 10.1016/j.tem.2020.01.002] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Revised: 01/06/2020] [Accepted: 01/09/2020] [Indexed: 12/15/2022]
Abstract
Genome-wide association studies (GWASs) have identified SNPs of the fat mass and obesity (FTO) gene as the most important risk alleles for obesity. However, how the presence of risk alleles affect phenotype is still a matter of intense investigation. In 2014, a study revealed that long-range enhancers from the intronic regions of the FTO gene regulate iroquois-class homeobox protein (IRX)3 expression. IRX3 is expressed in hypothalamic pro-opiomelanocortin (POMC) neurons and changes in its expression levels affect body adiposity by modifying food intake and energy expenditure. These findings have placed IRX3 as a potential target for the treatment of obesity. Here, we review studies that evaluated the roles of IRX3 in development, neurogenesis, and body energy homeostasis.
Collapse
Affiliation(s)
- Thiago Matos de Araújo
- Laboratory of Cell Signaling, Obesity and Comorbidities Research Center, State University of Campinas (UNICAMP), Campinas, Brazil
| | - Licio A Velloso
- Laboratory of Cell Signaling, Obesity and Comorbidities Research Center, State University of Campinas (UNICAMP), Campinas, Brazil.
| |
Collapse
|
21
|
Martyniuk CJ, Martínez R, Kostyniuk DJ, Mennigen JA, Zubcevic J. Genetic ablation of bone marrow beta-adrenergic receptors in mice modulates miRNA-transcriptome networks of neuroinflammation in the paraventricular nucleus. Physiol Genomics 2020; 52:169-177. [PMID: 32089076 PMCID: PMC7191424 DOI: 10.1152/physiolgenomics.00001.2020] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Revised: 02/07/2020] [Accepted: 02/16/2020] [Indexed: 12/21/2022] Open
Abstract
Elucidating molecular pathways regulating neuroimmune communication is critical for therapeutic interventions in conditions characterized by overactive immune responses and dysfunctional autonomic nervous system. We generated a bone marrow-specific adrenergic beta 1 and beta 2 knockout mouse chimera (AdrB1.B2 KO) to determine how sympathetic drive to the bone affects transcripts and miRNAs in the hypothalamic paraventricular nucleus (PVN). This model has previously exhibited a dampened systemic immune response and decreased blood pressure compared with control animals. Reduced sympathetic responsiveness of the bone marrow hematopoietic cells of AdrB1.B2 KO chimera led to suppression of transcriptional networks that included leukocyte cell adhesion and migration and T cell-activation and recruitment. Transcriptome responses related to IL-17a signaling and the renin-angiotensin system were also suppressed in the PVN. Based on the transcriptome response, we next computationally predicted miRNAs in the PVN that may underscore the reduced sympathetic responsiveness of the bone marrow cells. These included miR-27b-3p, miR-150, miR-223-3p, and miR-326. Using real-time PCR, we measured a downregulation in the expression of miR-150-5p, miR-205-5p, miR-223-3p, miR-375-5p, miR-499a-5p, miR-27b-3p, let-7a-5p, and miR-21a-5p in the PVN of AdrB1.B2 KO chimera, confirming computational predictions that these miRNAs are associated with reduced neuro-immune responses and the loss of sympathetic responsiveness in the bone marrow. Intriguingly, directional responses of the miRNA corresponded to mRNAs, suggesting complex temporal or circuit-dependent posttranscriptional control of gene expression in the PVN. This study identifies molecular pathways involved in neural-immune interactions that may act as targets of therapeutic intervention for a dysfunctional autonomic nervous system.
Collapse
Affiliation(s)
- Christopher J Martyniuk
- Department of Physiological Sciences, College of Veterinary Medicine, University of Florida, Gainesville, Florida
| | - Ruben Martínez
- Department of Environmental Chemistry, Institute of Environmental Assessment and Water Research, IDAEA-CSIC, Jordi Girona, Barcelona, Spain
- Department of Cellular Biology, Physiology and Immunology, Universidad de Barcelona (UB), Barcelona, Spain
| | | | - Jan A Mennigen
- Department of Biology, University of Ottawa, Ottawa, Ontario, Canada
| | - Jasenka Zubcevic
- Department of Physiological Sciences, College of Veterinary Medicine, University of Florida, Gainesville, Florida
| |
Collapse
|
22
|
Herrera-Morales WV, Herrera-Solís A, Núñez-Jaramillo L. Sexual Behavior and Synaptic Plasticity. ARCHIVES OF SEXUAL BEHAVIOR 2019; 48:2617-2631. [PMID: 31270644 DOI: 10.1007/s10508-019-01483-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Revised: 05/30/2019] [Accepted: 06/01/2019] [Indexed: 06/09/2023]
Abstract
Although sex drive is present in many animal species, sexual behavior is not static and, like many other behaviors, can be modified by experience. This modification relies on synaptic plasticity, a sophisticated mechanism through which neurons change how they process a given stimulus, and the neurophysiological basis of learning. This review addresses the main plastic effects of steroid sex hormones in the central nervous system (CNS) and the effects of sexual experience on the CNS, including effects on neurogenesis, intracellular signaling, gene expression, and changes in dendritic spines, as well as behavioral changes.
Collapse
Affiliation(s)
- Wendy Verónica Herrera-Morales
- División de Ciencias de la Salud, Universidad de Quintana Roo, Av. Erick Paolo Martínez S/N esquina Av 4 de marzo. Colonia Magisterial, 77039, Chetumal, Quintana Roo, Mexico
| | - Andrea Herrera-Solís
- Laboratorio Efectos Terapéuticos de los Canabinoides, Subdirección de Investigación Biomédica, Hospital General Dr. Manuel Gea González, Ciudad de México, Mexico
| | - Luis Núñez-Jaramillo
- División de Ciencias de la Salud, Universidad de Quintana Roo, Av. Erick Paolo Martínez S/N esquina Av 4 de marzo. Colonia Magisterial, 77039, Chetumal, Quintana Roo, Mexico.
| |
Collapse
|
23
|
Remaud S, Demeneix B. [Thyroid hormones regulate neural stem cell fate]. Biol Aujourdhui 2019; 213:7-16. [PMID: 31274098 DOI: 10.1051/jbio/2019007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Indexed: 01/02/2023]
Abstract
Thyroid hormones (THs) are vital for vertebrate brain function throughout life, from early development to ageing. Epidemiological studies show an adequate supply of maternal TH during pregnancy to be necessary for normal brain development, and this from the first trimester of onwards. Maternal TH deficiency irreversibly affects fetal brain development, increasing the risk of offspring cognitive disorders and IQ loss. Mammalian and non-mammalian (zebrafish, xenopus, chicken) models are useful to dissect TH-dependent cellular and molecular mechanisms governing embryonic and fetal brain development: a complex process including cell proliferation, survival, determination, migration, differentiation and maturation of neural stem cells (NSCs). Notably, rodent models have strongly contributed to understand the key neurogenic roles of TH still at work in adult life. Neurogenesis continues in two main areas, the sub-ventricular zone lining the lateral ventricles (essential for olfaction) and the sub-granular zone in the dentate gyrus of the hippocampus (involved in memory, learning and mood control). In both niches, THs tightly regulate the balance between neurogenesis and oligodendrogenesis under physiological and pathological contexts. Understanding how THs modulate NSCs determination toward a neuronal or a glial fate throughout life is a crucial question in neural stem cell biology. Providing answers to this question can offer therapeutic strategies for brain repair, notably in neurodegenerative diseases, demyelinating diseases or stroke where new neurons and/or oligodendrocytes are required. The review focuses on TH regulation of NSC fate in mammals and humans both during development and in the adult.
Collapse
Affiliation(s)
- Sylvie Remaud
- Muséum National d'Histoire Naturelle, CNRS UMR 7221, Laboratoire Physiologie moléculaire de l'adaptation, 7 rue Cuvier 75005 Paris, France
| | - Barbara Demeneix
- Muséum National d'Histoire Naturelle, CNRS UMR 7221, Laboratoire Physiologie moléculaire de l'adaptation, 7 rue Cuvier 75005 Paris, France
| |
Collapse
|
24
|
Kostin A, Alam MA, McGinty D, Szymusiak R, Alam MN. Chronic Suppression of Hypothalamic Cell Proliferation and Neurogenesis Induces Aging-Like Changes in Sleep–Wake Organization in Young Mice. Neuroscience 2019; 404:541-556. [DOI: 10.1016/j.neuroscience.2019.01.053] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2018] [Revised: 12/14/2018] [Accepted: 01/28/2019] [Indexed: 10/27/2022]
|
25
|
Fehily B, Bartlett CA, Lydiard S, Archer M, Milbourn H, Majimbi M, Hemmi JM, Dunlop SA, Yates NJ, Fitzgerald M. Differential responses to increasing numbers of mild traumatic brain injury in a rodent closed-head injury model. J Neurochem 2019; 149:660-678. [PMID: 30702755 DOI: 10.1111/jnc.14673] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Revised: 12/20/2018] [Accepted: 01/14/2019] [Indexed: 01/13/2023]
Abstract
Following mild traumatic brain injury (mTBI), further mild impacts can exacerbate negative outcomes. To compare chronic damage and deficits following increasing numbers of repeated mTBIs, a closed-head weight-drop model of repeated mTBI was used to deliver 1, 2 or 3 mTBIs to adult female rats at 24 h intervals. Outcomes were assessed at 3 months following the first mTBI. No gross motor, sensory or reflex deficits were identified (p > 0.05), consistent with current literature. Cognitive function assessed using a Morris water maze revealed chronic memory deficits following 1 and 2, but not 3 mTBI compared to shams (p ≤ 0.05). Oxidative damage to DNA was assessed immunohistochemically in the dentate hilus of the hippocampus and splenium of the corpus callosum; no changes were observed. IBA1-positive microglia were increased in size in the cortex following 1 mTBI and in the corpus callosum following 2 mTBI compared to shams (p ≤ 0.05); no changes were observed in the dentate hilus. Glial fibrillary acidic protein (GFAP)-positive astrocyte immunoreactivity was assessed in all three brain regions and no chronic changes were observed. Integrity of myelin ultrastructure in the corpus callosum was assessed using transmission electron microscopy. G ratio was decreased following 2 mTBIs compared to shams (p ≤ 0.05) at post hoc level only. The changing patterns of damage and deficits following increasing numbers of mTBI may reflect dynamic responses to small numbers of mTBIs or a conditioning effect such that increasing numbers of mTBIs do not necessarily result in worsening pathology. OPEN SCIENCE BADGES: This article has received a badge for *Open Materials* because it provided all relevant information to reproduce the study in the manuscript. The complete Open Science Disclosure form for this article can be found at the end of the article. More information about the Open Practices badges can be found at https://cos.io/our-services/open-science-badges/. Cover Image for this issue: doi: 10.1111/jnc.14508.
Collapse
Affiliation(s)
- Brooke Fehily
- Experimental and Regenerative Neurosciences, School of Biological Sciences, Crawley, WA, Australia
- School of Human Sciences, The University of Western Australia, Nedlands, WA, Australia
| | - Carole A Bartlett
- Experimental and Regenerative Neurosciences, School of Biological Sciences, Crawley, WA, Australia
| | - Stephen Lydiard
- Experimental and Regenerative Neurosciences, School of Biological Sciences, Crawley, WA, Australia
| | - Michael Archer
- Experimental and Regenerative Neurosciences, School of Biological Sciences, Crawley, WA, Australia
| | - Hannah Milbourn
- Experimental and Regenerative Neurosciences, School of Biological Sciences, Crawley, WA, Australia
| | - Maimuna Majimbi
- Experimental and Regenerative Neurosciences, School of Biological Sciences, Crawley, WA, Australia
- Curtin Health Innovation Research Institute, Faculty of Health Sciences, Bentley, WA, Australia
| | - Jan M Hemmi
- Experimental and Regenerative Neurosciences, School of Biological Sciences, Crawley, WA, Australia
| | - Sarah A Dunlop
- Experimental and Regenerative Neurosciences, School of Biological Sciences, Crawley, WA, Australia
| | - Nathanael J Yates
- School of Human Sciences, The University of Western Australia, Nedlands, WA, Australia
| | - Melinda Fitzgerald
- Experimental and Regenerative Neurosciences, School of Biological Sciences, Crawley, WA, Australia
- Curtin Health Innovation Research Institute, Faculty of Health Sciences, Bentley, WA, Australia
- The Perron Institute for Neurological and Translational Science, Sarich Neuroscience Research Institute Building, Nedlands, WA, Australia
| |
Collapse
|
26
|
Bedos M, Portillo W, Paredes RG. Neurogenesis and sexual behavior. Front Neuroendocrinol 2018; 51:68-79. [PMID: 29438737 DOI: 10.1016/j.yfrne.2018.02.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Revised: 02/06/2018] [Accepted: 02/07/2018] [Indexed: 12/16/2022]
Abstract
Different conditions induce proliferation, migration and integration of new neurons in the adult brain. This process of neurogenesis is a clear example of long lasting plastic changes in the brain of different species. Sexual behavior is a motivated behavior that is crucial for the survival of the species, but an individual can spend all his life without displaying sexual behavior. In the present review, we briefly describe some of the effects of pheromones on neurogenesis. We review in detail studies describing the effects of sexual behavior in both males and females on proliferation, migration and integration of new cells and neurons. It will become evident that most of the studies have been done in rodents, assessing the effects of this behavior on neurogenesis within the dentate gyrus of the hippocampus and in the subventricular zone - rostral migratory stream - olfactory bulb system.
Collapse
Affiliation(s)
- M Bedos
- CONACYT - Instituto de Neurobiología - Universidad Nacional Autónoma de México, Blvd Juriquilla 3001, Campus UNAM-Juriquilla, 76230 Querétaro, QRO, México
| | - W Portillo
- Instituto de Neurobiología - Universidad Nacional Autónoma de México, Blvd Juriquilla 3001, Campus UNAM-Juriquilla, 76230 Querétaro, QRO, México
| | - R G Paredes
- Instituto de Neurobiología - Universidad Nacional Autónoma de México, Blvd Juriquilla 3001, Campus UNAM-Juriquilla, 76230 Querétaro, QRO, México.
| |
Collapse
|
27
|
Safahani M, Aligholi H, Noorbakhsh F, Djalali M, Pishva H, Mousavi SMM, Alipour F, Gorji A, Koohdani F. Resveratrol promotes the arcuate nucleus architecture remodeling to produce more anorexigenic neurons in high-fat-diet–fed mice. Nutrition 2018. [PMID: 29524783 DOI: 10.1016/j.nut.2017.10.019] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
28
|
Ebling FJP, Lewis JE. Tanycytes and hypothalamic control of energy metabolism. Glia 2018; 66:1176-1184. [DOI: 10.1002/glia.23303] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Revised: 12/21/2017] [Accepted: 01/23/2018] [Indexed: 01/15/2023]
Affiliation(s)
- Francis J. P. Ebling
- School of Life Sciences; University of Nottingham Medical School, Queen's Medical Centre; Nottingham NG7 2UH United Kingdom
| | - Jo E. Lewis
- School of Life Sciences; University of Nottingham Medical School, Queen's Medical Centre; Nottingham NG7 2UH United Kingdom
| |
Collapse
|
29
|
Ponti G, Farinetti A, Marraudino M, Panzica G, Gotti S. Sex Steroids and Adult Neurogenesis in the Ventricular-Subventricular Zone. Front Endocrinol (Lausanne) 2018; 9:156. [PMID: 29686651 PMCID: PMC5900029 DOI: 10.3389/fendo.2018.00156] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2017] [Accepted: 03/22/2018] [Indexed: 12/28/2022] Open
Abstract
The forebrain ventricular-subventricular zone (V-SVZ) continuously generates new neurons throughout life. Neural stem cells (type B1 cells) along the lateral ventricle become activated, self-renew, and give rise to proliferating precursors which progress along the neurogenic lineage from intermediate progenitors (type C cells) to neuroblasts (type A cells). Neuroblasts proliferate and migrate into the olfactory bulb and differentiate into different interneuronal types. Multiple factors regulate each step of this process. Newly generated olfactory bulb interneurons are an important relay station in the olfactory circuits, controlling social recognition, reproductive behavior, and parental care. Those behaviors are strongly sexually dimorphic and changes throughout life from puberty through aging and in the reproductive age during estrous cycle and gestation. Despite the key role of sex hormones in regulating those behaviors, their contribution in modulating adult neurogenesis in V-SVZ is underestimated. Here, we compare the literature highlighting the sexual dimorphism and the differences across the physiological phases of the animal for the different cell types and steps through the neurogenic lineage.
Collapse
Affiliation(s)
- Giovanna Ponti
- Department of Veterinary Sciences, University of Turin, Grugliasco,Turin, Italy
- Neuroscience Institute Cavalieri Ottolenghi (NICO), Orbassano, Turin, Italy
- *Correspondence: Giovanna Ponti,
| | - Alice Farinetti
- Neuroscience Institute Cavalieri Ottolenghi (NICO), Orbassano, Turin, Italy
- Department of Neuroscience “Rita Levi-Montalcini”, University of Turin, Turin, Italy
| | - Marilena Marraudino
- Neuroscience Institute Cavalieri Ottolenghi (NICO), Orbassano, Turin, Italy
- Department of Neuroscience “Rita Levi-Montalcini”, University of Turin, Turin, Italy
| | - GianCarlo Panzica
- Neuroscience Institute Cavalieri Ottolenghi (NICO), Orbassano, Turin, Italy
- Department of Neuroscience “Rita Levi-Montalcini”, University of Turin, Turin, Italy
| | - Stefano Gotti
- Neuroscience Institute Cavalieri Ottolenghi (NICO), Orbassano, Turin, Italy
- Department of Neuroscience “Rita Levi-Montalcini”, University of Turin, Turin, Italy
| |
Collapse
|
30
|
Lévy F, Batailler M, Meurisse M, Migaud M. Adult Neurogenesis in Sheep: Characterization and Contribution to Reproduction and Behavior. Front Neurosci 2017; 11:570. [PMID: 29109674 PMCID: PMC5660097 DOI: 10.3389/fnins.2017.00570] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Accepted: 09/28/2017] [Indexed: 01/18/2023] Open
Abstract
Sheep have many advantages to study neurogenesis in comparison to the well-known rodent models. Their development and life expectancy are relatively long and they possess a gyrencephalic brain. Sheep are also seasonal breeders, a characteristic that allows studying the involvement of hypothalamic neurogenesis in the control of seasonal reproduction. Sheep are also able to individually recognize their conspecifics and develop selective and lasting bonds. Adult olfactory neurogenesis could be adapted to social behavior by supporting recognition of conspecifics. The present review reveals the distinctive features of the hippocampal, olfactory, and hypothalamic neurogenesis in sheep. In particular, the organization of the subventricular zone and the dynamic of neuronal maturation differs from that of rodents. In addition, we show that various physiological conditions, such as seasonal reproduction, gestation, and lactation differently modulate these three neurogenic niches. Last, we discuss recent evidence indicating that hypothalamic neurogenesis acts as an important regulator of the seasonal control of reproduction and that olfactory neurogenesis could be involved in odor processing in the context of maternal behavior.
Collapse
Affiliation(s)
- Frederic Lévy
- Institut National de la Recherche Agronomique, UMR85, Centre National de la Recherche Scientifique, UMR7247, Université F. Rabelais, IFCE, Physiologie de la Reproduction et des Comportements, Nouzilly, France
| | - Martine Batailler
- Institut National de la Recherche Agronomique, UMR85, Centre National de la Recherche Scientifique, UMR7247, Université F. Rabelais, IFCE, Physiologie de la Reproduction et des Comportements, Nouzilly, France
| | - Maryse Meurisse
- Institut National de la Recherche Agronomique, UMR85, Centre National de la Recherche Scientifique, UMR7247, Université F. Rabelais, IFCE, Physiologie de la Reproduction et des Comportements, Nouzilly, France
| | - Martine Migaud
- Institut National de la Recherche Agronomique, UMR85, Centre National de la Recherche Scientifique, UMR7247, Université F. Rabelais, IFCE, Physiologie de la Reproduction et des Comportements, Nouzilly, France
| |
Collapse
|
31
|
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
|
32
|
Recabal A, Caprile T, García-Robles MDLA. Hypothalamic Neurogenesis as an Adaptive Metabolic Mechanism. Front Neurosci 2017; 11:190. [PMID: 28424582 PMCID: PMC5380718 DOI: 10.3389/fnins.2017.00190] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2017] [Accepted: 03/21/2017] [Indexed: 12/12/2022] Open
Abstract
In the adult brain, well-characterized neurogenic niches are located in the subventricular zone (SVZ) of the lateral ventricles and in the subgranular zone (SGZ) of the hippocampus. In both regions, neural precursor cells (NPCs) share markers of embryonic radial glia and astroglial cells, and in vitro clonal expansion of these cells leads to neurosphere formation. It has also been more recently demonstrated that neurogenesis occurs in the adult hypothalamus, a brain structure that integrates peripheral signals to control energy balance and dietary intake. The NPCs of this region, termed tanycytes, are ependymal-glial cells, which comprise the walls of the infundibular recess of the third ventricle and contact the median eminence. Thus, tanycytes are in a privileged position to detect hormonal, nutritional and mitogenic signals. Recent studies reveal that in response to nutritional signals, tanycytes are capable of differentiating into orexigenic or anorexigenic neurons, suggesting that these cells are crucial for control of feeding behavior. In this review, we discuss evidence, which suggests that hypothalamic neurogenesis may act as an additional adaptive mechanism in order to respond to changes in diet.
Collapse
Affiliation(s)
- Antonia Recabal
- Laboratorio de Biología Celular, Departamento de Biología Celular, Facultad de Ciencias Biológicas, Universidad de ConcepciónConcepción, Chile.,Laboratorio de Guía Axonal, Departamento de Biología Celular, Facultad de Ciencias Biológicas, Universidad de ConcepciónConcepción, Chile
| | - Teresa Caprile
- Laboratorio de Guía Axonal, Departamento de Biología Celular, Facultad de Ciencias Biológicas, Universidad de ConcepciónConcepción, Chile
| | - María de Los Angeles García-Robles
- Laboratorio de Biología Celular, Departamento de Biología Celular, Facultad de Ciencias Biológicas, Universidad de ConcepciónConcepción, Chile
| |
Collapse
|
33
|
Abstract
One of the most hotly pursued topics in neuroscience and therapeutic medicine is the use of stem cells in the adult brain. Growing in parallel to this emerging field is the recognition that the adult brain is indeed capable of generating new cells. While neurogenesis was understood to be restricted to a few areas, recent studies suggest that damage to the adult brain can trigger neurogenesis even in regions outside of these specific areas. This finding raises the possibility that neurons born in response to perturbation in the brain may be involved in the recovery of function in the damaged adult brain. The key is understanding how to cultivate these newborn cells, because they do not remain viable if they are not accepted into the damaged network of interconnected neurons which support specific functions. From a birth site, undifferentiated precursor cells or neurons undergo migration and differentiation. Many factors influence the safe journey of migrating cells and their survival after maturation at their destination. This review will present evidence from ring dove studies that an activity-dependent mechanism underlies the survival of adult newborn neurons and establishment of their functionality. This evidence includes: [1] unique electrophysiological properties or specific connectivity associated with various type of neurons involved in ring dove coo behavior and reproductive function, [2] emergence of electrophysiological properties and specific projection neurons emanating from newborn neurons after hypothalamic lesion, and finally [3] collective behavioral analyses of social stimulations suggesting that sensorimotor events contribute to the integration of new neurons and reinstatement of function.
Collapse
Affiliation(s)
- Mei-Fang Cheng
- Department of Psychology, Rutgers, The State University of New Jersey, Newark, NJ, USA
| |
Collapse
|
34
|
The biology of addiction. Can J Anaesth 2016; 64:141-148. [PMID: 27837404 DOI: 10.1007/s12630-016-0771-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2016] [Revised: 09/13/2016] [Accepted: 11/02/2016] [Indexed: 02/07/2023] Open
Abstract
In this narrative review, the neurobiological mechanisms underlying substance abuse and addiction are discussed with a particular emphasis on the mechanisms that promote ongoing use and relapse. Addiction is estimated to affect 10-15% or more of the adult population, including physicians. Genetic predisposition, psychological and environmental risk factors, the timing of exposure to the substance, the type of substance used, and the frequency of use influence the individual's susceptibility to addiction. Abused substances act on the brain's reward system, a neural circuit that produces pleasurable feelings in response to stimuli that promote survival, thereby modifying future behavior to seek out similar stimuli. Endogenous activators include food, sex, and social interaction. Drugs of abuse hijack the reward circuit, producing intense activation. Repetitive exposure to substances leads to persistent, altered genetic expression and accumulation of ΔFos-B and corticotropin-releasing factor. High levels of these substances suppress the reward circuit and activate the endogenous stress response, resulting in a generalized state of discord. These changes are enduring and can trigger substance use relapse even after long periods of abstinence.
Collapse
|
35
|
Pytte CL. Adult Neurogenesis in the Songbird: Region-Specific Contributions of New Neurons to Behavioral Plasticity and Stability. BRAIN, BEHAVIOR AND EVOLUTION 2016; 87:191-204. [PMID: 27560148 DOI: 10.1159/000447048] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Our understanding of the role of new neurons in learning and encoding new information has been largely based on studies of new neurons in the mammalian dentate gyrus and olfactory bulb - brain regions that may be specialized for learning. Thus the role of new neurons in regions that serve other functions has yet to be fully explored. The song system provides a model for studying new neuron function in brain regions that contribute differently to song learning, song auditory discrimination, and song motor production. These regions subserve learning as well as long-term storage of previously learned information. This review examines the differences between learning-based and activity-based retention of new neurons and explores the potential contributions of new neurons to behavioral stability in the song motor production pathway.
Collapse
Affiliation(s)
- Carolyn L Pytte
- Psychology Department, Queens College and The Graduate Center, City University of New York, Flushing, N.Y., USA
| |
Collapse
|
36
|
Neuropeptide Y (NPY) as a therapeutic target for neurodegenerative diseases. Neurobiol Dis 2016; 95:210-24. [PMID: 27461050 DOI: 10.1016/j.nbd.2016.07.022] [Citation(s) in RCA: 92] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Revised: 06/29/2016] [Accepted: 07/20/2016] [Indexed: 12/16/2022] Open
Abstract
Neuropeptide Y (NPY) and NPY receptors are widely expressed in the mammalian central nervous system. Studies in both humans and rodent models revealed that brain NPY levels are altered in some neurodegenerative disorders, such as Alzheimer's disease, Parkinson's disease, Huntington's disease and Machado-Joseph disease. In this review, we will focus on the roles of NPY in the pathological mechanisms of these disorders, highlighting NPY as a neuroprotective agent, as a neural stem cell proliferative agent, as an agent that increases trophic support, as a stimulator of autophagy and as an inhibitor of excitotoxicity and neuroinflammation. Moreover, the effect of NPY in some clinical manifestations commonly observed in Alzheimer's disease, Parkinson's disease, Huntington's disease and Machado-Joseph disease, such as depressive symptoms and body weight loss, are also discussed. In conclusion, this review highlights NPY system as a potential therapeutic target in neurodegenerative diseases.
Collapse
|
37
|
Mohr MA, Garcia FL, DonCarlos LL, Sisk CL. Neurons and Glial Cells Are Added to the Female Rat Anteroventral Periventricular Nucleus During Puberty. Endocrinology 2016; 157:2393-402. [PMID: 27145006 PMCID: PMC4891785 DOI: 10.1210/en.2015-2012] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The anteroventral periventricular nucleus (AVPV) orchestrates the neuroendocrine-positive feedback response that triggers ovulation in female rodents. The AVPV is larger and more cell-dense in females than in males, and during puberty, only females develop the capacity to show a positive feedback response. We previously reported a potential new mechanism to explain this female-specific gain of function during puberty, namely a female-biased sex difference in the pubertal addition of new cells to the rat AVPV. Here we first asked whether this sex difference is due to greater cell proliferation and/or survival in females. Female and male rats received the cell birthdate marker 5-bromo-2'-deoxyuridine (BrdU; 200 mg/kg, ip) on postnatal day (P) 30; brains were collected at short and long intervals after BrdU administration to assess cell proliferation and survival, respectively. Overall, females had more BrdU-immunoreactive cells in the AVPV than did males, with no sex differences in the rate of cell attrition over time. Thus, the sex difference in pubertal addition of AVPV cells appears to be due to greater cell proliferation in females. Next, to determine the phenotype of pubertally born AVPV cells, daily BrdU injections were given to female rats on P28-56, and tissue was collected on P77 to assess colocalization of BrdU and markers for mature neurons or glia. Of the pubertally born AVPV cells, approximately 15% differentiated into neurons, approximately 19% into astrocytes, and approximately 23% into microglia. Thus, both neuro- and gliogenesis occur in the pubertal female rat AVPV and potentially contribute to maturation of female reproductive function.
Collapse
Affiliation(s)
- Margaret A Mohr
- Neuroscience Program (M.A.M., C.L.S.), Michigan State University, East Lansing, Michigan 48824; and Department of Cell and Molecular Physiology (F.L.G., L.L.D.), Loyola University Chicago, Maywood, Illinois 60153
| | - Francisca L Garcia
- Neuroscience Program (M.A.M., C.L.S.), Michigan State University, East Lansing, Michigan 48824; and Department of Cell and Molecular Physiology (F.L.G., L.L.D.), Loyola University Chicago, Maywood, Illinois 60153
| | - Lydia L DonCarlos
- Neuroscience Program (M.A.M., C.L.S.), Michigan State University, East Lansing, Michigan 48824; and Department of Cell and Molecular Physiology (F.L.G., L.L.D.), Loyola University Chicago, Maywood, Illinois 60153
| | - Cheryl L Sisk
- Neuroscience Program (M.A.M., C.L.S.), Michigan State University, East Lansing, Michigan 48824; and Department of Cell and Molecular Physiology (F.L.G., L.L.D.), Loyola University Chicago, Maywood, Illinois 60153
| |
Collapse
|
38
|
Holmes MM. Social regulation of adult neurogenesis: A comparative approach. Front Neuroendocrinol 2016; 41:59-70. [PMID: 26877107 DOI: 10.1016/j.yfrne.2016.02.001] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2015] [Revised: 02/07/2016] [Accepted: 02/09/2016] [Indexed: 01/25/2023]
Abstract
The social environment sculpts the mammalian brain throughout life. Adult neurogenesis, the birth of new neurons in the mature brain, can be up- or down-regulated by various social manipulations. These include social isolation, social conflict, social status, socio-sexual interactions, and parent/offspring interactions. However, socially-mediated changes in neuron production are often species-, sex-, and/or region-specific. In order to reconcile the variability of social effects on neurogenesis, we need to consider species-specific social adaptations and other contextual variables (e.g. age, social status, reproductive status, etc.) that shift the valence of social stimuli. Using a comparative approach to understand how adult-generated neurons in turn influence social behaviors will shed light on how adult neurogenesis contributes to survival and reproduction in diverse species.
Collapse
Affiliation(s)
- Melissa M Holmes
- Department of Psychology, University of Toronto, Canada; Department of Cell & Systems Biology, University of Toronto, Canada; Department of Ecology & Evolutionary Biology, University of Toronto, Canada.
| |
Collapse
|
39
|
Understanding Neurogenesis in the Subventricular Zone and the Capacity for Transcriptional Modulation in Ischemic Brain Injury. World Neurosurg 2016; 87:477-80. [PMID: 26828460 DOI: 10.1016/j.wneu.2016.01.045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
40
|
Tsuchiya H, Hohjoh H, Fujiwara Y, Sugimoto Y, Koshimizu TA. Prostaglandin D2 elicits the reversible neurite retraction in hypothalamic cell line. Biochem Biophys Res Commun 2016; 470:804-10. [PMID: 26820529 DOI: 10.1016/j.bbrc.2016.01.091] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Accepted: 01/15/2016] [Indexed: 12/19/2022]
Abstract
Prostaglandins (PGs) play important roles in diverse physiological processes in the central nervous system. PGD2 is the most abundant PG in the brain and acts through specific receptors, DP1 and CRTH2. We investigated the effects of PGD2 on the morphology of the hypothalamic cell line mHypoE-N37 (N37). In N37 cells, serum starvation induced neurite outgrowth and PGD2 elicited neurite retraction, although we failed to detect transcripts for DP1 and CRTH2. Such an effect of PGD2 was efficiently mimicked by its metabolite, 15-deoxy-Δ(12,14)-prostaglandin J2. N-acetyl cysteine completely abolished the effect of PGD2, and reactive oxygen species (ROS) were considered to be important. Notably, neurite outgrowth was restored by PGD2 removal. These results suggest that PGD2 induces reversible neurite retraction in a ROS-mediated mechanism that does not involve any known receptor.
Collapse
Affiliation(s)
- Hiroyoshi Tsuchiya
- Division of Molecular Pharmacology, Department of Pharmacology, Jichi Medical University, Tochigi 329-0498, Japan.
| | - Hirofumi Hohjoh
- Department of Pharmaceutical Biochemistry, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto 862-0973, Japan
| | - Yoko Fujiwara
- Division of Molecular Pharmacology, Department of Pharmacology, Jichi Medical University, Tochigi 329-0498, Japan
| | - Yukihiko Sugimoto
- Department of Pharmaceutical Biochemistry, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto 862-0973, Japan
| | - Taka-Aki Koshimizu
- Division of Molecular Pharmacology, Department of Pharmacology, Jichi Medical University, Tochigi 329-0498, Japan
| |
Collapse
|
41
|
McPherson AD, Barrios JP, Luks-Morgan SJ, Manfredi JP, Bonkowsky JL, Douglass AD, Dorsky RI. Motor Behavior Mediated by Continuously Generated Dopaminergic Neurons in the Zebrafish Hypothalamus Recovers after Cell Ablation. Curr Biol 2016; 26:263-269. [PMID: 26774784 DOI: 10.1016/j.cub.2015.11.064] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2015] [Revised: 10/14/2015] [Accepted: 11/24/2015] [Indexed: 12/21/2022]
Abstract
Postembryonic neurogenesis has been observed in several regions of the vertebrate brain, including the dentate gyrus and rostral migratory stream in mammals, and is required for normal behavior [1-3]. Recently, the hypothalamus has also been shown to undergo continuous neurogenesis as a way to mediate energy balance [4-10]. As the hypothalamus regulates multiple functional outputs, it is likely that additional behaviors may be affected by postembryonic neurogenesis in this brain structure. Here, we have identified a progenitor population in the zebrafish hypothalamus that continuously generates neurons that express tyrosine hydroxylase 2 (th2). We develop and use novel transgenic tools to characterize the lineage of th2(+) cells and demonstrate that they are dopaminergic. Through genetic ablation and optogenetic activation, we then show that th2(+) neurons modulate the initiation of swimming behavior in zebrafish larvae. Finally, we find that the generation of new th2(+) neurons following ablation correlates with restoration of normal behavior. This work thus identifies for the first time a population of dopaminergic neurons that regulates motor behavior capable of functional recovery.
Collapse
Affiliation(s)
- Adam D McPherson
- Department of Neurobiology and Anatomy, University of Utah, Salt Lake City, UT 84112, USA
| | - Joshua P Barrios
- Department of Neurobiology and Anatomy, University of Utah, Salt Lake City, UT 84112, USA
| | - Sasha J Luks-Morgan
- Department of Neurobiology and Anatomy, University of Utah, Salt Lake City, UT 84112, USA
| | - John P Manfredi
- Department of Neurobiology and Anatomy, University of Utah, Salt Lake City, UT 84112, USA
| | - Joshua L Bonkowsky
- Department of Pediatrics, University of Utah, Salt Lake City, UT 84112, USA
| | - Adam D Douglass
- Department of Neurobiology and Anatomy, University of Utah, Salt Lake City, UT 84112, USA.
| | - Richard I Dorsky
- Department of Neurobiology and Anatomy, University of Utah, Salt Lake City, UT 84112, USA.
| |
Collapse
|
42
|
Duncan RN, Xie Y, McPherson AD, Taibi AV, Bonkowsky JL, Douglass AD, Dorsky RI. Hypothalamic radial glia function as self-renewing neural progenitors in the absence of Wnt/β-catenin signaling. Development 2015; 143:45-53. [PMID: 26603385 DOI: 10.1242/dev.126813] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2015] [Accepted: 11/18/2015] [Indexed: 12/12/2022]
Abstract
The vertebrate hypothalamus contains persistent radial glia that have been proposed to function as neural progenitors. In zebrafish, a high level of postembryonic hypothalamic neurogenesis has been observed, but the role of radial glia in generating these new neurons is unclear. We have used inducible Cre-mediated lineage labeling to show that a population of hypothalamic radial glia undergoes self-renewal and generates multiple neuronal subtypes at larval stages. Whereas Wnt/β-catenin signaling has been demonstrated to promote the expansion of other stem and progenitor cell populations, we find that Wnt/β-catenin pathway activity inhibits this process in hypothalamic radial glia and is not required for their self-renewal. By contrast, Wnt/β-catenin signaling is required for the differentiation of a specific subset of radial glial neuronal progeny residing along the ventricular surface. We also show that partial genetic ablation of hypothalamic radial glia or their progeny causes a net increase in their proliferation, which is also independent of Wnt/β-catenin signaling. Hypothalamic radial glia in the zebrafish larva thus exhibit several key characteristics of a neural stem cell population, and our data support the idea that Wnt pathway function may not be homogeneous in all stem or progenitor cells.
Collapse
Affiliation(s)
- Robert N Duncan
- Department of Neurobiology and Anatomy, University of Utah, Salt Lake City, UT 84112, USA
| | - Yuanyuan Xie
- Department of Neurobiology and Anatomy, University of Utah, Salt Lake City, UT 84112, USA
| | - Adam D McPherson
- Department of Neurobiology and Anatomy, University of Utah, Salt Lake City, UT 84112, USA
| | - Andrew V Taibi
- Department of Neurobiology and Anatomy, University of Utah, Salt Lake City, UT 84112, USA
| | - Joshua L Bonkowsky
- Department of Neurobiology and Anatomy, University of Utah, Salt Lake City, UT 84112, USA
| | - Adam D Douglass
- Department of Neurobiology and Anatomy, University of Utah, Salt Lake City, UT 84112, USA
| | - Richard I Dorsky
- Department of Neurobiology and Anatomy, University of Utah, Salt Lake City, UT 84112, USA
| |
Collapse
|
43
|
Jiang H, Modise T, Helm R, Jensen RV, Good DJ. Characterization of the hypothalamic transcriptome in response to food deprivation reveals global changes in long noncoding RNA, and cell cycle response genes. GENES & NUTRITION 2015; 10:48. [PMID: 26475716 PMCID: PMC4608919 DOI: 10.1007/s12263-015-0496-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Accepted: 10/05/2015] [Indexed: 12/15/2022]
Abstract
The hypothalamus integrates energy balance information from the periphery using different neuronal subtypes within each of the hypothalamic areas. However, the effects of prandial state on global mRNA, microRNA and long noncoding (lnc) RNA expression within the whole hypothalamus are largely unknown. In this study, mice were given either a 24-h fast, or ad libitum access to food. RNA samples were analyzed by microarray, and then a subset was confirmed using quantitative real-time PCR (QPCR). A total of 540 mRNAs were either up- or down-regulated with food deprivation. Since gene ontology enrichment analyses identified several categories of mRNAs related to cell cycle processes, ten cell-cycle-related genes were further analyzed using QPCR with six confirmed to be significantly up-regulated and one down-regulated in response to 24-h fasting. While 22 independent microRNAs were differentially expressed by microarray, secondary analysis by QPCR failed to confirm significant changes with fasting. There were 622 lncRNAs identified as differentially expressed, and of three tested by QPCR, two were confirmed. Overall, this is the first time that expression of hypothalamic lncRNAs has been shown to be responsive to food deprivation. In addition, this study is the first to identify a list of lncRNAs with high expression in RNA extracted from hypothalamus. Individual contributions from specific miRNA, lncRNA and mRNAs to the food deprivation response can now be further studied at the physiological and biochemical levels.
Collapse
Affiliation(s)
- Hao Jiang
- Department of Biochemistry, Virginia Tech, Blacksburg, VA, 24061, USA
- Department of Neurology, Washington University in St Louis, St Louis, MO, 63110, USA
| | - Thero Modise
- Program in Genomics, Bioinformatics and Computational Biology, Virginia Tech, Blacksburg, VA, 24061, USA
| | - Richard Helm
- Department of Biochemistry, Virginia Tech, Blacksburg, VA, 24061, USA
- Program in Genomics, Bioinformatics and Computational Biology, Virginia Tech, Blacksburg, VA, 24061, USA
| | - Roderick V Jensen
- Department of Biological Sciences, Virginia Tech, Blacksburg, VA, 24061, USA
| | - Deborah J Good
- Department of Human Nutrition Foods and Exercise, Virginia Tech, 1981 Kraft Drive (0913), Blacksburg, VA, 24061, USA.
- Department of Biochemistry, Virginia Tech, Blacksburg, VA, 24061, USA.
- Program in Genomics, Bioinformatics and Computational Biology, Virginia Tech, Blacksburg, VA, 24061, USA.
| |
Collapse
|
44
|
Goodman T, Hajihosseini MK. Hypothalamic tanycytes-masters and servants of metabolic, neuroendocrine, and neurogenic functions. Front Neurosci 2015; 9:387. [PMID: 26578855 PMCID: PMC4624852 DOI: 10.3389/fnins.2015.00387] [Citation(s) in RCA: 96] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2015] [Accepted: 10/05/2015] [Indexed: 11/13/2022] Open
Abstract
There is a resurgent interest in tanycytes, a radial glial-like cell population occupying the floor and ventro-lateral walls of the third ventricle (3V). Tanycytes reside in close proximity to hypothalamic neuronal nuclei that regulate appetite and energy expenditure, with a subset sending projections into these nuclei. Moreover, tanycytes are exposed to 3V cerebrospinal fluid and have privileged access to plasma metabolites and hormones, through fenestrated capillaries. Indeed, some tanycytes act as conduits for trafficking of these molecules into the brain parenchyma. Tanycytes can also act as neural stem/progenitor cells, supplying the postnatal and adult hypothalamus with new neurons. Collectively, these findings suggest that tanycytes regulate and integrate important trophic and metabolic processes and possibly endow functional malleability to neuronal circuits of the hypothalamus. Hence, manipulation of tanycyte biology could provide a valuable tool for modulating hypothalamic functions such as energy uptake and expenditure in order to tackle prevalent eating disorders such as obesity and anorexia.
Collapse
Affiliation(s)
- Timothy Goodman
- School of Biological Sciences, University of East Anglia Norwich, UK
| | | |
Collapse
|
45
|
Segura S, Efthimiadi L, Porcher C, Courtes S, Coronas V, Krantic S, Moyse E. Leptin-dependent neurotoxicity via induction of apoptosis in adult rat neurogenic cells. Front Cell Neurosci 2015; 9:350. [PMID: 26441523 PMCID: PMC4561523 DOI: 10.3389/fncel.2015.00350] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2014] [Accepted: 08/21/2015] [Indexed: 12/30/2022] Open
Abstract
Adipocyte-derived hormone leptin has been recently implicated in the control of neuronal plasticity. To explore whether modulation of adult neurogenesis may contribute to leptin control of neuronal plasticity, we used the neurosphere assay of neural stem cells derived from the adult rat subventricular zone (SVZ). Endogenous expression of specific leptin receptor (ObRb) transcripts, as revealed by RT-PCR, is associated with activation of both ERK and STAT-3 pathways via phosphorylation of the critical ERK/STAT-3 amino acid residues upon addition of leptin to neurospheres. Furthermore, leptin triggered withdrawal of neural stem cells from the cell cycle as monitored by Ki67 labeling. This effect was blocked by pharmacological inhibition of ERK activation thus demonstrating that ERK mediates leptin effects on neural stem cell expansion. Leptin-dependent withdrawal of neural stem cells from the cell cycle was associated with increased apoptosis, as detected by TUNEL, which was preceded by cyclin D1 induction. Cyclin D1 was indeed extensively colocalized with TUNEL-positive, apoptotic nuclei. Cyclin-D1 silencing by specific shRNA prevented leptin-induced decrease of the cell number per neurosphere thus pointing to the causal relationship between leptin actions on apoptosis and cyclin D1 induction. Leptin target cells in SVZ neurospheres were identified by double TUNEL/phenotypic marker immunocytofluorescence as differentiating neurons mostly. The inhibition of neural stem cell expansion via ERK/cyclin D1-triggered apoptosis defines novel biological action of leptin which may be involved in adiposity-dependent neurotoxicity.
Collapse
Affiliation(s)
- Stéphanie Segura
- Physiologie de la Reproduction et des Comportements, UMR 85 Institut National de la Recherche Agronomique, Centre INRA de Tours, Université François Rabelais de Tours Nouzilly, France
| | - Laurie Efthimiadi
- Institut National de la Santé et de la Recherche Médicale Unité 901, Institut de Neurobiologie de la Méditerranée, Parc Scientifique de Luminy, Aix-Marseille Université Marseille, France
| | - Christophe Porcher
- Institut National de la Santé et de la Recherche Médicale Unité 901, Institut de Neurobiologie de la Méditerranée, Parc Scientifique de Luminy, Aix-Marseille Université Marseille, France
| | - Sandrine Courtes
- Institut National de la Santé et de la Recherche Médicale Unité 901, Institut de Neurobiologie de la Méditerranée, Parc Scientifique de Luminy, Aix-Marseille Université Marseille, France
| | - Valérie Coronas
- Signalisation et Transports Ioniques Membranaires, ERL 7368 Centre National de la Recherche Scientifique, Université de Poitiers Poitiers, France
| | - Slavica Krantic
- Institut National de la Santé et de la Recherche Médicale Unité 901, Institut de Neurobiologie de la Méditerranée, Parc Scientifique de Luminy, Aix-Marseille Université Marseille, France ; Centre de Recherche des Cordeliers, UMR_S 1138 INSERM, Paris Descartes University, Sorbonne Paris Cité, Pierre and Marie Curie University Paris, France
| | - Emmanuel Moyse
- Physiologie de la Reproduction et des Comportements, UMR 85 Institut National de la Recherche Agronomique, Centre INRA de Tours, Université François Rabelais de Tours Nouzilly, France
| |
Collapse
|
46
|
Cortés-Campos C, Letelier J, Ceriani R, Whitlock KE. Zebrafish adult-derived hypothalamic neurospheres generate gonadotropin-releasing hormone (GnRH) neurons. Biol Open 2015. [PMID: 26209533 PMCID: PMC4582115 DOI: 10.1242/bio.010447] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Gonadotropin-releasing hormone (GnRH) is a hypothalamic decapeptide essential for fertility in vertebrates. Human male patients lacking GnRH and treated with hormone therapy can remain fertile after cessation of treatment suggesting that new GnRH neurons can be generated during adult life. We used zebrafish to investigate the neurogenic potential of the adult hypothalamus. Previously we have characterized the development of GnRH cells in the zebrafish linking genetic pathways to the differentiation of neuromodulatory and endocrine GnRH cells in specific regions of the brain. Here, we developed a new method to obtain neural progenitors from the adult hypothalamus in vitro. Using this system, we show that neurospheres derived from the adult hypothalamus can be maintained in culture and subsequently differentiate glia and neurons. Importantly, the adult derived progenitors differentiate into neurons containing GnRH and the number of cells is increased through exposure to either testosterone or GnRH, hormones used in therapeutic treatment in humans. Finally, we show in vivo that a neurogenic niche in the hypothalamus contains GnRH positive neurons. Thus, we demonstrated for the first time that neurospheres can be derived from the hypothalamus of the adult zebrafish and that these neural progenitors are capable of producing GnRH containing neurons.
Collapse
Affiliation(s)
- Christian Cortés-Campos
- Centro Interdisciplinario de Neurociencia de Valparaíso (CINV), Facultad de Ciencias, Universidad de Valparaíso, Pasaje Harrington 269, Valparaíso 2340000, Chile Whitehead Institute for Biomedical Research (WIBR), 9 Cambridge Center, Cambridge, MA 02142, USA
| | - Joaquín Letelier
- Centro Interdisciplinario de Neurociencia de Valparaíso (CINV), Facultad de Ciencias, Universidad de Valparaíso, Pasaje Harrington 269, Valparaíso 2340000, Chile Centro Andaluz de Biología del Desarrollo, Universidad Pablo de Olavide, Carretera de Utera km 1, Sevilla 41013, España
| | - Ricardo Ceriani
- Centro Interdisciplinario de Neurociencia de Valparaíso (CINV), Facultad de Ciencias, Universidad de Valparaíso, Pasaje Harrington 269, Valparaíso 2340000, Chile
| | - Kathleen E Whitlock
- Centro Interdisciplinario de Neurociencia de Valparaíso (CINV), Facultad de Ciencias, Universidad de Valparaíso, Pasaje Harrington 269, Valparaíso 2340000, Chile
| |
Collapse
|
47
|
Masana M, Jukic M, Kretzschmar A, Wagner K, Westerholz S, Schmidt M, Rein T, Brodski C, Müller M. Deciphering the spatio-temporal expression and stress regulation of Fam107B, the paralog of the resilience-promoting protein DRR1 in the mouse brain. Neuroscience 2015; 290:147-58. [DOI: 10.1016/j.neuroscience.2015.01.026] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2014] [Revised: 01/12/2015] [Accepted: 01/16/2015] [Indexed: 11/26/2022]
|
48
|
Expression of a novel serine/threonine kinase gene, Ulk4, in neural progenitors during Xenopus laevis forebrain development. Neuroscience 2015; 290:61-79. [DOI: 10.1016/j.neuroscience.2014.12.060] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2014] [Revised: 12/30/2014] [Accepted: 12/31/2014] [Indexed: 01/11/2023]
|
49
|
Niwa A, Nishibori M, Hamasaki S, Kobori T, Liu K, Wake H, Mori S, Yoshino T, Takahashi H. Voluntary exercise induces neurogenesis in the hypothalamus and ependymal lining of the third ventricle. Brain Struct Funct 2015; 221:1653-66. [PMID: 25633473 DOI: 10.1007/s00429-015-0995-x] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2014] [Accepted: 01/21/2015] [Indexed: 11/26/2022]
Abstract
In the adult hypothalamus and ependymal lining of the third ventricle, tanycytes function as multipotential progenitor cells that enable continuous neurogenesis, suggesting that tanycytes may be able to mediate the restoration of homeostatic function after stroke. Voluntary wheel running has been shown to alter neurochemistry and neuronal function and to increase neurogenesis in rodents. In the present study, we found that voluntary exercise improved the survival rate and energy balance of stroke-prone spontaneously hypertensive rats (SHRSP/Kpo). We also investigated the effect of exercise on the proliferation and differentiation of hypothalamic cells using immunoreactivity for tanycytes and neural markers. The proliferation of elongated cells, which may be the tanycytes, was enhanced in exercising SHRSP compared to sedentary rats before and after stroke. In addition, the proliferation of cells was correlated with the induction of fibroblast growth factor-2 in the subependymal cells of the third ventricle and in the cerebrospinal fluid. Some of the newborn cells of exercising SHRSP showed differentiation into mature neurons after stroke. Our results suggest that voluntary exercise correlates with hypothalamic neurogenesis, leading to recovery of homeostatic functions in the adult brain after stroke.
Collapse
Affiliation(s)
- Atsuko Niwa
- Department of Pharmacology, Faculty of Medicine, Kinki University, 377-2 Ohno-Higashi, Osaka-Sayama, Osaka, 589-8511, Japan
| | - Masahiro Nishibori
- Department of Pharmacology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Okayama, Japan
| | - Shinichi Hamasaki
- Department of Anesthesiology, Faculty of Medicine, Kinki University, 377-2 Ohno-Higashi, Osaka-Sayama, Osaka, 589-8511, Japan
| | - Takuro Kobori
- Department of Pharmacology, Faculty of Medicine, Kinki University, 377-2 Ohno-Higashi, Osaka-Sayama, Osaka, 589-8511, Japan
| | - Keyue Liu
- Department of Pharmacology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Okayama, Japan
| | - Hidenori Wake
- Department of Pharmacology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Okayama, Japan
| | - Shuji Mori
- Department of Pharmacy, Shujitsu University, 1-6-1 Nishikawahara, Okayama, Japan
| | - Tadashi Yoshino
- Department of Pathology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Okayama, Japan
| | - Hideo Takahashi
- Department of Pharmacology, Faculty of Medicine, Kinki University, 377-2 Ohno-Higashi, Osaka-Sayama, Osaka, 589-8511, Japan.
| |
Collapse
|
50
|
Genin EC, Caron N, Vandenbosch R, Nguyen L, Malgrange B. Concise review: forkhead pathway in the control of adult neurogenesis. Stem Cells 2015; 32:1398-407. [PMID: 24510844 DOI: 10.1002/stem.1673] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2013] [Revised: 01/09/2014] [Accepted: 01/09/2014] [Indexed: 12/23/2022]
Abstract
New cells are continuously generated from immature proliferating cells in the adult brain in two neurogenic niches known as the subgranular zone (SGZ) of the dentate gyrus (DG) of the hippocampus and the sub-ventricular zone (SVZ) of the lateral ventricles. However, the molecular mechanisms regulating their proliferation, differentiation, migration and functional integration of newborn neurons in pre-existing neural network remain largely unknown. Forkhead box (Fox) proteins belong to a large family of transcription factors implicated in a wide variety of biological processes. Recently, there has been accumulating evidence that several members of this family of proteins play important roles in adult neurogenesis. Here, we describe recent advances in our understanding of regulation provided by Fox factors in adult neurogenesis, and evaluate the potential role of Fox proteins as targets for therapeutic intervention in neurodegenerative diseases.
Collapse
Affiliation(s)
- Emmanuelle C Genin
- GIGA-Neurosciences, Developmental Neurobiology Unit, University of Liège, Liège, Belgium
| | | | | | | | | |
Collapse
|