1
|
Yagi S, Mohammad A, Wen Y, Batallán Burrowes AA, Blankers SA, Galea LAM. Estrogens dynamically regulate neurogenesis in the dentate gyrus of adult female rats. Hippocampus 2024; 34:583-597. [PMID: 39166359 DOI: 10.1002/hipo.23633] [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: 01/26/2024] [Revised: 08/01/2024] [Accepted: 08/09/2024] [Indexed: 08/22/2024]
Abstract
Estrone and estradiol differentially modulate neuroplasticity and cognition. How they influence the maturation of new neurons in the adult hippocampus, however, is not known. The present study assessed the effects of estrone and estradiol on the maturation timeline of neurogenesis in the dentate gyrus (DG) of ovariectomized (a model of surgical menopause) young adult Sprague-Dawley rats using daily subcutaneous injections of 17β-estradiol, estrone or vehicle. Rats were injected with a DNA synthesis marker, 5-bromo-2-deoxyuridine (BrdU), and were perfused 1, 2, or 3 weeks after BrdU injection and daily hormone treatment. Brains were sectioned and processed for various markers including: sex-determining region Y-box 2 (Sox2), glial fibrillary acidic protein (GFAP), antigen kiel 67 (Ki67), doublecortin (DCX), and neuronal nuclei (NeuN). Immunofluorescent labeling or co-labelling of BrdU with Sox2 (progenitor cells), Sox2/GFAP (neural progenitor cells), Ki67 (cell proliferation), DCX (immature neurons), NeuN (mature neurons) was used to examine the trajectory and maturation of adult-born neurons over time. Estrogens had early (1 week of exposure) effects on different stages of neurogenesis (neural progenitor cells, cell proliferation and early maturation of new cells into neurons) but these effects were less pronounced after prolonged treatment. Estradiol enhanced, whereas estrone reduced cell proliferation after 1 week but not after longer exposure to either estrogen. Both estrogens increased the density of immature neurons (BrdU/DCX-ir) after 1 week of exposure compared to vehicle treatment but this increased density was not sustained over longer durations of treatments to estrogens, suggesting that the enhancing effects of estrogens on neurogenesis were short-lived. Longer duration post-ovariectomy, without treatments with either of the estrogens, was associated with reduced neural progenitor cells in the DG. These results demonstrate that estrogens modulate several aspects of adult hippocampal neurogenesis differently in the short term, but may lose their ability to influence neurogenesis after long-term exposure. These findings have potential implications for treatments involving estrogens after surgical menopause.
Collapse
Affiliation(s)
- Shunya Yagi
- Graduate Program in Neuroscience, University of British Columbia, Vancouver, British Columbia, Canada
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, British Columbia, Canada
| | - Ahmad Mohammad
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
| | - Yanhua Wen
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, British Columbia, Canada
| | - Ariel A Batallán Burrowes
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
| | - Samantha A Blankers
- Graduate Program in Neuroscience, University of British Columbia, Vancouver, British Columbia, Canada
- Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
| | - Liisa A M Galea
- Graduate Program in Neuroscience, University of British Columbia, Vancouver, British Columbia, Canada
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, British Columbia, Canada
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
- Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
2
|
Kramer M, Hoang TH, Yang H, Shchyglo O, Böge J, Neubacher U, Colitti-Klausnitzer J, Manahan-Vaughan D. Intracerebral inoculation of healthy non-transgenic rats with a single aliquot of oligomeric amyloid-β (1-42) profoundly and progressively alters brain function throughout life. Front Aging Neurosci 2024; 16:1397901. [PMID: 39156737 PMCID: PMC11327071 DOI: 10.3389/fnagi.2024.1397901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Accepted: 07/09/2024] [Indexed: 08/20/2024] Open
Abstract
One of the puzzling aspects of sporadic Alzheimer's disease (AD) is how it commences. Changes in one key brain peptide, amyloid-beta (Aβ), accompany disease progression, but whether this comprises a trigger or a consequence of AD is still a topic of debate. It is clear however that the cerebral presence of oligomeric Aβ (1-42) is a key factor in early AD-pathogenesis. Furthermore, treatment of rodent brains with oligomeric Aβ (1-42) either in vitro or in vivo, acutely impairs hippocampal synaptic plasticity, creating a link between Aβ-pathology and learning impairments. Here, we show that a once-off inoculation of the brains of healthy adult rats with oligomeric Aβ (1-42) exerts debilitating effects on the long-term viability of the hippocampus, one of the primary targets of AD. Changes are progressive: months after treatment, synaptic plasticity, neuronal firing and spatial learning are impaired and expression of plasticity-related proteins are changed, in the absence of amyloid plaques. Early changes relate to activation of microglia, whereas later changes are associated with a reconstruction of astroglial morphology. These data suggest that a disruption of Aβ homeostasis may suffice to trigger an irreversible cascade, underlying progressive loss of hippocampal function, that parallels the early stages of AD.
Collapse
|
3
|
Koca RO, Gormus ZIS, Solak H, Celik FS, Kurar E, Kutlu S. Are the promnestic effects of neurokinin 3 receptor mediated by hippocampal neurogenesis in a Aβ-induced rat model of Alzheimer's disease? Int J Dev Neurosci 2024. [PMID: 39010691 DOI: 10.1002/jdn.10362] [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: 04/26/2024] [Revised: 05/27/2024] [Accepted: 06/26/2024] [Indexed: 07/17/2024] Open
Abstract
Alzheimer's disease (AD) is an age-related neurodegenerative disorder characterised by cognitive dysfunction, memory loss and mood changes. Hippocampal neurogenesis has been suggested to play a role in learning and memory. Neurokinin 3 receptor (NK3R) has been shown to be prevalent in the hippocampus region. The aim of the project was to investigate the role of hippocampal neurogenesis in the promnestic effects of NK3R agonist administration in an amyloid beta-induced AD rat model. Wistar albino rats were divided into control, Alzheimer, NK3R agonist and Alzheimer + NK3R agonist groups. The open field (OF) test and Morris water maze (MWM) test were performed for locomotor activity and memory analysis. Peptide gene expression levels (Nestin, DCX, Neuritin, MASH1, Neun, BDNF) were analysed by quantitative reverse transcription polymerase chain reaction (RT-PCR). In the OF test, the group-time relationship was found to be statistically different in the parameters of distance travelled and percentage of movement (p < 0.05). In MWM, the time to reach the platform and the time spent in the target quadrant were statistically significant between the groups (p < 0.05). Statistically significant differences were observed in gene expression levels (Nestin, DCX, Neuritin, MASH1) in the hippocampal tissue of rats between the groups (p < 0.05). NK3 receptor agonism favourably affected hippocampal neurogenesis in AD model rats. It was concluded that NK3 receptor agonism in the hippocampus, which is the first affected region in the physiopathology of AD, may be effective in both the formation of neural precursor cells and the reduction of neuronal degeneration. The positive effect of NK3R on cognitive functions may be mediated by hippocampal neurogenesis.
Collapse
Affiliation(s)
- Raviye Ozen Koca
- Department of Physiology, Faculty of Medicine, Necmettin Erbakan University, Konya, Turkey
| | - Z Isık Solak Gormus
- Department of Physiology, Faculty of Medicine, Necmettin Erbakan University, Konya, Turkey
| | - Hatice Solak
- Department of Physiology, Faculty of Medicine, Kütahya Health Sciences University, Kutahya, Turkey
| | - Fatma Secer Celik
- Department of Medical Biology, Faculty of Medicine, Ankara Medipol University, Ankara, Turkey
| | - Ercan Kurar
- Department of Medical Biology, Faculty of Medicine, Necmettin Erbakan University, Konya, Turkey
| | - Selim Kutlu
- Department of Physiology, Faculty of Medicine, Necmettin Erbakan University, Konya, Turkey
| |
Collapse
|
4
|
Murata Y, Yoshimitsu S, Senoura C, Araki T, Kanayama S, Mori M, Ohe K, Mine K, Enjoji M. Sleep rebound leads to marked recovery of prolonged sleep deprivation-induced adversities in the stress response and hippocampal neuroplasticity of male rats. J Affect Disord 2024; 355:478-486. [PMID: 38574868 DOI: 10.1016/j.jad.2024.04.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 03/26/2024] [Accepted: 04/01/2024] [Indexed: 04/06/2024]
Abstract
BACKGROUND Sleep disturbances are not only frequent symptoms, but also risk factors for major depressive disorder. We previously reported that depressed patients who experienced "Hypersomnia" showed a higher and more rapid response rate under paroxetine treatment, but the underlying mechanism remains unclear. The present study was conducted to clarify the beneficial effects of sleep rebound through an experimental "Hypersomnia" rat model on glucocorticoid and hippocampal neuroplasticity associated with antidepressive potency. METHODS Thirty-four male Sprague-Dawley rats were subjected to sham treatment, 72-h sleep deprivation, or sleep deprivation and subsequent follow-up for one week. Approximately half of the animals were sacrificed to evaluate adrenal weight, plasma corticosterone level, hippocampal content of mRNA isoforms, and protein of the brain-derived neurotrophic factor (Bdnf) gene. In the other half of the rats, Ki-67- and doublecortin (DCX)-positive cells in the hippocampus were counted via immunostaining to quantify adult neurogenesis. RESULTS Prolonged sleep deprivation led to adrenal hypertrophy and an increase in the plasma corticosterone level, which had returned to normal after one week follow-up. Of note, sleep deprivation-induced decreases in hippocampal Bdnf transcripts containing exons II, IV, VI, and IX and BDNF protein levels, Ki-67-(+)-proliferating cells, and DCX-(+)-newly-born neurons were not merely reversed, but overshot their normal levels with sleep rebound. LIMITATIONS The present study did not record electroencephalogram or assess behavioral changes of the sleep-deprived rats. CONCLUSIONS The present study demonstrated that prolonged sleep deprivation-induced adversities are reversed or recovered by sleep rebound, which supports "Hypersomnia" in depressed patients as having a beneficial pharmacological effect.
Collapse
Affiliation(s)
- Yusuke Murata
- Department of Pharmacotherapeutics, Faculty of Pharmaceutical Sciences, Fukuoka University, 8-19-1, Nanakuma, Jonan-ku, Fukuoka 814-0180, Japan.
| | - Sakuya Yoshimitsu
- Department of Pharmacotherapeutics, Faculty of Pharmaceutical Sciences, Fukuoka University, 8-19-1, Nanakuma, Jonan-ku, Fukuoka 814-0180, Japan
| | - Chiyo Senoura
- Department of Pharmacotherapeutics, Faculty of Pharmaceutical Sciences, Fukuoka University, 8-19-1, Nanakuma, Jonan-ku, Fukuoka 814-0180, Japan
| | - Toshiki Araki
- Department of Pharmacotherapeutics, Faculty of Pharmaceutical Sciences, Fukuoka University, 8-19-1, Nanakuma, Jonan-ku, Fukuoka 814-0180, Japan
| | - Saki Kanayama
- Department of Pharmacotherapeutics, Faculty of Pharmaceutical Sciences, Fukuoka University, 8-19-1, Nanakuma, Jonan-ku, Fukuoka 814-0180, Japan
| | - Masayoshi Mori
- Department of Pharmacotherapeutics, Faculty of Pharmaceutical Sciences, Fukuoka University, 8-19-1, Nanakuma, Jonan-ku, Fukuoka 814-0180, Japan
| | - Kenji Ohe
- Department of Pharmacotherapeutics, Faculty of Pharmaceutical Sciences, Fukuoka University, 8-19-1, Nanakuma, Jonan-ku, Fukuoka 814-0180, Japan
| | - Kazunori Mine
- Faculty of Neurology and Psychiatry, BOOCS CLINIC FUKUOKA, 6F Random Square Bldg., 6-18, Tenya-Machi, Hakata-ku, Fukuoka 812-0025, Japan
| | - Munechika Enjoji
- Department of Pharmacotherapeutics, Faculty of Pharmaceutical Sciences, Fukuoka University, 8-19-1, Nanakuma, Jonan-ku, Fukuoka 814-0180, Japan
| |
Collapse
|
5
|
Zhang Y, Zhang XQ, Niu WP, Sun M, Zhang Y, Li JT, Si TM, Su YA. TAAR1 in dentate gyrus is involved in chronic stress-induced impairments in hippocampal plasticity and cognitive function. Prog Neuropsychopharmacol Biol Psychiatry 2024; 132:110995. [PMID: 38514038 DOI: 10.1016/j.pnpbp.2024.110995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 03/09/2024] [Accepted: 03/17/2024] [Indexed: 03/23/2024]
Abstract
Multiple lines of evidence suggest that the trace amine-associated receptor 1 (TAAR1) holds promise as a potential target for stress-related disorders, such as treating major depressive disorder (MDD). The role of TAAR1 in the regulation of adult neurogenesis is recently supported by transcriptomic data. However, it remains unknown whether TAAR1 in dentate gyrus (DG) mediate chronic stress-induced negative effects on hippocampal plasticity and related behavior in mice. The present study consisted of a series of experiments using RNAscope, genetic approaches, behavioral tests, immunohistochemical staining, Golgi-Cox technique to unravel the effects of TAAR1 on alterations of dentate neuronal plasticity and cognitive function in the chronic social defeat stress model. The mice subjected to chronic defeat stress exhibited a noteworthy decrease in the mRNA level of TAAR1 in DG. Additionally, they exhibited compromised social memory and spatial object recognition memory, as well as impaired proliferation and maturation of adult-born dentate granule cells. Moreover, the selective knockout TAAR1 in DG mostly mimicked the cognitive function deficits and neurogenesis impairment induced by chronic stress. Importantly, the administration of the selective TAAR1 partial agonist RO5263397 during stress exposure attenuated the adverse effects of chronic stress on cognitive function, adult neurogenesis, dendritic arborization, and the synapse number of dentate neurons in DG. In summary, our findings suggest that TAAR1 plays a crucial role in mediating the detrimental effects of chronic stress on hippocampal plasticity and cognition. TAAR1 agonists exhibit therapeutic potential for individuals suffering from cognitive impairments associated with MDD.
Collapse
Affiliation(s)
- Yue Zhang
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing 100191, China
| | - Xian-Qiang Zhang
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing 100191, China
| | - Wei-Pan Niu
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing 100191, China
| | - Meng Sun
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing 100191, China
| | - Yanan Zhang
- Research Triangle Institute, Research Triangle Park, NC 27709, USA
| | - Ji-Tao Li
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing 100191, China
| | - Tian-Mei Si
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing 100191, China.
| | - Yun-Ai Su
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing 100191, China.
| |
Collapse
|
6
|
Song YT, Li SS, Chao CY, Shuang-Guo, Chen GZ, Wang SX, Zhang MX, Yin YL, Li P. Floralozone regulates MiR-7a-5p expression through AMPKα2 activation to improve cognitive dysfunction in vascular dementia. Exp Neurol 2024; 376:114748. [PMID: 38458310 DOI: 10.1016/j.expneurol.2024.114748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 02/07/2024] [Accepted: 03/04/2024] [Indexed: 03/10/2024]
Abstract
BACKGROUND The pathogenesis of vascular dementia (VD) is complex, and currently, no effective treatments have been recommended. Floralozone is a colorless liquid first discovered in Lagotis Gaertn. Recently, its medicinal value has been increasingly recognized. Our previous study has demonstrated that Floralozone can improve cognitive dysfunction in rats with VD by regulating the transient receptor potential melastatin 2 (TRPM2) and N-methyl-D-aspartate receptor (NMDAR) signaling pathways. However, the mechanism by which Floralozone regulates TRPM2 and NMDAR to improve VD remains unclear. AMP-activated protein kinase (AMPK) is an energy regulator in vivo; however, its role of AMPK activation in stroke remains controversial. MiR-7a-5p has been identified to be closely related to neuronal function. PURPOSE To explore whether Floralozone can regulate the miR-7a-5p level in vivo through AMPKα2 activation, affect the TRPM2 and NR2B expression levels, and improve VD symptoms. METHODS The VD model was established by a modified bilateral occlusion of the common carotid arteries (2-VO) of Sprague-Dawley (SD) rats and AMPKα2 KO transgenic (AMPKα2-/-) mice. Primary hippocampal neurons were modeled using oxygen and glucose deprivation (OGD). Morris water maze (MWM) test, hematoxylin-eosin staining (HE staining), and TUNEL staining were used to investigate the effects of Floralozone on behavior and hippocampal morphology in rats. Minichromosome maintenance complex component 2(MCM2) positive cells were used to investigate the effect of Floralozone on neurogenesis. Immunofluorescence staining, qRT-PCR, and western blot analysis were used to investigate the effect of Floralozone on the expression levels of AMPKα2, miR-7a-5p, TRPM2, and NR2B. RESULTS The SD rat experiment revealed that Floralozone improved spatial learning and memory, improved the morphology and structure of hippocampal neurons, reduced apoptosis of hippocampal neurons and promoted neurogenesis in VD rats. Floralozone could increase the miR-7a-5p expression level, activate AMPKα2 and NR2B expressions, and inhibit TRPM2 expression in hippocampal neurons of VD rats. The AMPKα2 KO transgenic (AMPKα2-/-) mice experiment demonstrated that Floralozone could regulate miR-7a-5p, TRPM2, and NR2B expression levels through AMPKα2 activation. The cell experiment revealed that the TRPM2 and NR2B expression levels were regulated by miR-7a-5p, whereas the AMPKα2 expression level was not. CONCLUSION Floralozone could regulate miR-7a-5p expression level by activating the protein expression of AMPKα2, control the protein expression of TRPM2 and NR2B, improve the morphology and structure of hippocampus neurons, reduce the apoptosis of hippocampus neurons, promote neurogenesis and improve the cognitive dysfunction.
Collapse
Affiliation(s)
- Yu-Ting Song
- Henan international joint laboratory of cardiovascular remodeling and drug intervention, Sino-UK Joint Laboratory of Brain Function and Injury and Department of Physiology and Neurobiology, School of Basic Medical Sciences, College of Pharmacy, Xinxiang Medical University, Xinxiang 453003, China; JinShan Hospital of Fudan University, Shanghai 201508, China
| | - Shan-Shan Li
- Henan international joint laboratory of cardiovascular remodeling and drug intervention, Sino-UK Joint Laboratory of Brain Function and Injury and Department of Physiology and Neurobiology, School of Basic Medical Sciences, College of Pharmacy, Xinxiang Medical University, Xinxiang 453003, China
| | - Chun-Yan Chao
- Henan international joint laboratory of cardiovascular remodeling and drug intervention, Sino-UK Joint Laboratory of Brain Function and Injury and Department of Physiology and Neurobiology, School of Basic Medical Sciences, College of Pharmacy, Xinxiang Medical University, Xinxiang 453003, China; Huang Huai University, Zhumadian 463000, China
| | - Shuang-Guo
- Hubei Key Laboratory of Diabetes and Angiopathy, Hubei University of Science and Technology, Xianning 437100, China
| | - Gui-Zi Chen
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Shuang-Xi Wang
- Henan international joint laboratory of cardiovascular remodeling and drug intervention, Sino-UK Joint Laboratory of Brain Function and Injury and Department of Physiology and Neurobiology, School of Basic Medical Sciences, College of Pharmacy, Xinxiang Medical University, Xinxiang 453003, China
| | - Ming-Xiang Zhang
- Henan international joint laboratory of cardiovascular remodeling and drug intervention, Sino-UK Joint Laboratory of Brain Function and Injury and Department of Physiology and Neurobiology, School of Basic Medical Sciences, College of Pharmacy, Xinxiang Medical University, Xinxiang 453003, China
| | - Ya-Ling Yin
- Henan international joint laboratory of cardiovascular remodeling and drug intervention, Sino-UK Joint Laboratory of Brain Function and Injury and Department of Physiology and Neurobiology, School of Basic Medical Sciences, College of Pharmacy, Xinxiang Medical University, Xinxiang 453003, China.
| | - Peng Li
- Henan international joint laboratory of cardiovascular remodeling and drug intervention, Sino-UK Joint Laboratory of Brain Function and Injury and Department of Physiology and Neurobiology, School of Basic Medical Sciences, College of Pharmacy, Xinxiang Medical University, Xinxiang 453003, China.
| |
Collapse
|
7
|
Maheshwari M, Singla A, Rawat A, Banerjee T, Pati S, Shah S, Maiti S, Vaidya VA. Chronic chemogenetic activation of hippocampal progenitors enhances adult neurogenesis and modulates anxiety-like behavior and fear extinction learning. IBRO Neurosci Rep 2024; 16:168-181. [PMID: 39007086 PMCID: PMC11240292 DOI: 10.1016/j.ibneur.2024.01.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Accepted: 01/18/2024] [Indexed: 07/16/2024] Open
Abstract
Adult hippocampal neurogenesis is a lifelong process that involves the integration of newborn neurons into the hippocampal network, and plays a role in cognitive function and the modulation of mood-related behavior. Here, we sought to address the impact of chemogenetic activation of adult hippocampal progenitors on distinct stages of progenitor development, including quiescent stem cell activation, progenitor turnover, differentiation and morphological maturation. We find that hM3Dq-DREADD-mediated activation of nestin-positive adult hippocampal progenitors recruits quiescent stem cells, enhances progenitor proliferation, increases doublecortin-positive newborn neuron number, accompanied by an acceleration of differentiation and morphological maturation, associated with increased dendritic complexity. Behavioral analysis indicated anxiolytic behavioral responses in transgenic mice subjected to chemogenetic activation of adult hippocampal progenitors at timepoints when newborn neurons are predicted to integrate into the mature hippocampal network. Furthermore, we noted an enhanced fear memory extinction on a contextual fear memory learning task in transgenic mice subjected to chemogenetic activation of adult hippocampal progenitors. Our findings indicate that hM3Dq-DREAD-mediated chemogenetic activation of adult hippocampal progenitors impacts distinct aspects of hippocampal neurogenesis, associated with the regulation of anxiety-like behavior and fear memory extinction.
Collapse
Affiliation(s)
| | | | - Anoop Rawat
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai, Maharashtra 400005, India
| | - Toshali Banerjee
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai, Maharashtra 400005, India
| | - Sthitapranjya Pati
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai, Maharashtra 400005, India
| | - Sneha Shah
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai, Maharashtra 400005, India
| | - Sudipta Maiti
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai, Maharashtra 400005, India
| | - Vidita A. Vaidya
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai, Maharashtra 400005, India
| |
Collapse
|
8
|
Oh HN, Yoo D, Park S, Lee S, Kim WK. Assessment of poly(hexamethylenebicyanoguanide-hexamethylenediamine) hydrochloride-induced developmental neurotoxicity via oxidative stress mechanism: Integrative approaches with neuronal cells and zebrafish. JOURNAL OF HAZARDOUS MATERIALS 2024; 465:133146. [PMID: 38064952 DOI: 10.1016/j.jhazmat.2023.133146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 11/22/2023] [Accepted: 11/28/2023] [Indexed: 02/08/2024]
Abstract
Poly(hexamethylenebicyanoguanide-hexamethylenediamine) hydrochloride (PHMB) is a biocide with a broad spectrum of antibacterial activity. Its use as a disinfectant and preservative in consumer products results in human exposure to PHMB. Toxicity studies on PHMB mainly focus on systemic toxicity or skin irritation; however, its effects on developmental neurotoxicity (DNT) and the underlying mechanisms are poorly understood. In this study, the DNT effects of PHMB were evaluated using IMR-32 and SH-SY5Y cell lines and zebrafish. In both cell lines, PHMB concentrations ≥ 10 µM reduced neurite outgrowth, and cytotoxicity was observed at concentrations up to 40 µM. PHMB regulated expression of neurodevelopmental genes and induced reactive oxygen species (ROS) production and mitochondrial dysfunction. Treatment with N-acetylcysteine reversed the toxic effects of PHMB. Toxicity tests on zebrafish embryos showed that PHMB reduced viability and heart rate and caused irregular hatching. PHMB concentrations of 1-4 µM reduced the width of the brain and spinal cord of transgenic zebrafish and attenuated myelination processes. Furthermore, PHMB modulated expression of neurodevelopmental genes in zebrafish and induced ROS accumulation. These results suggested that PHMB exerted DNT effects in vitro and in vivo through a ROS-dependent mechanism, highlighting the risk of PHMB exposure.
Collapse
Affiliation(s)
- Ha-Na Oh
- Department of Predictive Toxicology, Korea Institute of Toxicology, Daejeon 34114, Republic of Korea
| | - Donggon Yoo
- Department of Predictive Toxicology, Korea Institute of Toxicology, Daejeon 34114, Republic of Korea; Human and Environmental Toxicology, University of Science and Technology, Daejeon 34113, Republic of Korea
| | - Seungmin Park
- Department of Predictive Toxicology, Korea Institute of Toxicology, Daejeon 34114, Republic of Korea; Human and Environmental Toxicology, University of Science and Technology, Daejeon 34113, Republic of Korea
| | - Sangwoo Lee
- Department of Predictive Toxicology, Korea Institute of Toxicology, Daejeon 34114, Republic of Korea; Human and Environmental Toxicology, University of Science and Technology, Daejeon 34113, Republic of Korea
| | - Woo-Keun Kim
- Department of Predictive Toxicology, Korea Institute of Toxicology, Daejeon 34114, Republic of Korea; Human and Environmental Toxicology, University of Science and Technology, Daejeon 34113, Republic of Korea.
| |
Collapse
|
9
|
Cogut V, McNeely TL, Bussian TJ, Graves SI, Baker DJ. Caloric Restriction Improves Spatial Learning Deficits in Tau Mice. J Alzheimers Dis 2024; 98:925-940. [PMID: 38517786 PMCID: PMC11068089 DOI: 10.3233/jad-231117] [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] [Indexed: 03/24/2024]
Abstract
Background Caloric restriction (CR) has been recognized for its benefits in delaying age-related diseases and extending lifespan. While its effects on amyloid pathology in Alzheimer's disease (AD) mouse models are well-documented, its effects on tauopathy, another hallmark of AD, are less explored. Objective To assess the impact of a short-term 30% CR regimen on age-dependent spatial learning deficits and pathological features in a tauopathy mouse model. Methods We subjected male PS19 tau P301S (hereafter PS19) and age-matched wildtype mice from two age cohorts (4.5 and 7.5 months old) to a 6-week 30% CR regimen. Spatial learning performance was assessed using the Barnes Maze test. Tau pathology, neuroinflammation, hippocampal cell proliferation, and neurogenesis were evaluated in the older cohort by immunohistochemical staining and RT-qPCR. Results CR mitigated age-dependent spatial learning deficits in PS19 mice but exhibited limited effects on tau pathology and the associated neuroinflammation. Additionally, we found a decrease in hippocampal cell proliferation, predominantly of Iba1+ cells. Conclusions Our findings reinforce the cognitive benefits conferred by CR despite its limited modulation of disease pathology. Given the pivotal role of microglia in tau-driven pathology, the observed reduction in Iba1+ cells under CR suggests potential therapeutic implications, particularly if CR would be introduced early in disease progression.
Collapse
Affiliation(s)
- Valeria Cogut
- Department of Biochemistry and Molecular Biology, Mayo Clinic, 200 First St SW, Rochester, MN 55905, USA
| | - Taylor L. McNeely
- Department of Biochemistry and Molecular Biology, Mayo Clinic, 200 First St SW, Rochester, MN 55905, USA
| | - Tyler J. Bussian
- Department of Biochemistry and Molecular Biology, Mayo Clinic, 200 First St SW, Rochester, MN 55905, USA
| | - Sara I. Graves
- Department of Pediatric and Adolescent Medicine, Mayo Clinic, 200 First St SW, Rochester, MN 55905, USA
| | - Darren J. Baker
- Department of Biochemistry and Molecular Biology, Mayo Clinic, 200 First St SW, Rochester, MN 55905, USA
- Department of Pediatric and Adolescent Medicine, Mayo Clinic, 200 First St SW, Rochester, MN 55905, USA
- Paul F. Glenn Center for Biology of Aging Research at Mayo Clinic, Mayo Clinic, 200 First St SW, Rochester, MN 55905, USA
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, 200 First St SW, Rochester, MN 55905, USA
| |
Collapse
|
10
|
Nayab DE, Din FU, Ali H, Kausar WA, Urooj S, Zafar M, Khan I, Shabbir K, Khan GM. Nano biomaterials based strategies for enhanced brain targeting in the treatment of neurodegenerative diseases: an up-to-date perspective. J Nanobiotechnology 2023; 21:477. [PMID: 38087359 PMCID: PMC10716964 DOI: 10.1186/s12951-023-02250-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Accepted: 12/03/2023] [Indexed: 12/18/2023] Open
Abstract
Neurons and their connecting axons gradually degenerate in neurodegenerative diseases (NDs), leading to dysfunctionality of the neuronal cells and eventually their death. Drug delivery for the treatment of effected nervous system is notoriously complicated because of the presence of natural barriers, i.e., the blood-brain barrier and the blood cerebrospinal fluid barrier. Palliative care is currently the standard care for many diseases. Therefore, treatment programs that target the disease's origin rather than its symptoms are recommended. Nanotechnology-based drug delivery platforms offer an innovative way to circumvent these obstacles and deliver medications directly to the central nervous system, thereby enabling treatment of several common neurological problems, i.e., Alzheimer's, Parkinson's, Huntington's, and amyotrophic lateral sclerosis. Interestingly, the combination of nanomedicine and gene therapy enables targeting of selective mutant genes responsible for the progression of NDs, which may provide a much-needed boost in the struggle against these diseases. Herein, we discussed various central nervous system delivery obstacles, followed by a detailed insight into the recently developed techniques to restore neurological function via the differentiation of neural stem cells. Moreover, a comprehensive background on the role of nanomedicine in controlling neurogenesis via differentiation of neural stem cells is explained. Additionally, numerous phytoconstituents with their neuroprotective properties and molecular targets in the identification and management of NDs are also deliberated. Furthermore, a detailed insight of the ongoing clinical trials and currently marketed products for the treatment of NDs is provided in this manuscript.
Collapse
Affiliation(s)
- Dur E Nayab
- Department of Pharmacy, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, 45320, Pakistan
| | - Fakhar Ud Din
- Department of Pharmacy, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, 45320, Pakistan.
- Nanomedicine Research Group, Department of Pharmacy, Faculty of Biological Sciences, Quaid- i-Azam University, Islamabad, 45320, Pakistan.
| | - Hussain Ali
- Department of Pharmacy, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, 45320, Pakistan.
| | - Warda Arooj Kausar
- Department of Pharmacy, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, 45320, Pakistan
| | - Shaiza Urooj
- Department of Pharmacy, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, 45320, Pakistan
- Nanomedicine Research Group, Department of Pharmacy, Faculty of Biological Sciences, Quaid- i-Azam University, Islamabad, 45320, Pakistan
| | - Maryam Zafar
- Department of Pharmacy, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, 45320, Pakistan
| | - Ibrahim Khan
- Department of Pharmacy, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, 45320, Pakistan
| | - Kanwal Shabbir
- Department of Pharmacy, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, 45320, Pakistan
- Nanomedicine Research Group, Department of Pharmacy, Faculty of Biological Sciences, Quaid- i-Azam University, Islamabad, 45320, Pakistan
| | - Gul Majid Khan
- Department of Pharmacy, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, 45320, Pakistan
- Nanomedicine Research Group, Department of Pharmacy, Faculty of Biological Sciences, Quaid- i-Azam University, Islamabad, 45320, Pakistan
- Islamia College University, Peshawar, Khyber Pakhtunkhwa, Pakistan
| |
Collapse
|
11
|
Loef D, Tendolkar I, van Eijndhoven PFP, Hoozemans JJM, Oudega ML, Rozemuller AJM, Lucassen PJ, Dols A, Dijkstra AA. Electroconvulsive therapy is associated with increased immunoreactivity of neuroplasticity markers in the hippocampus of depressed patients. Transl Psychiatry 2023; 13:355. [PMID: 37981649 PMCID: PMC10658169 DOI: 10.1038/s41398-023-02658-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 10/23/2023] [Accepted: 11/07/2023] [Indexed: 11/21/2023] Open
Abstract
Electroconvulsive therapy (ECT) is an effective therapy for depression, but its cellular effects on the human brain remain elusive. In rodents, electroconvulsive shocks increase proliferation and the expression of plasticity markers in the hippocampal dentate gyrus (DG), suggesting increased neurogenesis. Furthermore, MRI studies in depressed patients have demonstrated increases in DG volume after ECT, that were notably paralleled by a decrease in depressive mood scores. Whether ECT also triggers cellular plasticity, inflammation or possibly injury in the human hippocampus, was unknown. We here performed a first explorative, anatomical study on the human post-mortem hippocampus of a unique, well-documented cohort of bipolar or unipolar depressed patients, who had received ECT in the 5 years prior to their death. They were compared to age-matched patients with a depressive disorder who had not received ECT and to matched healthy controls. Upon histopathological examination, no indications were observed for major hippocampal cell loss, overt cytoarchitectural changes or classic neuropathology in these 3 groups, nor were obvious differences present in inflammatory markers for astrocytes or microglia. Whereas the numbers of proliferating cells expressing Ki-67 was not different, we found a significantly higher percentage of cells positive for Doublecortin, a marker commonly used for young neurons and cellular plasticity, in the subgranular zone and CA4 / hilus of the hippocampus of ECT patients. Also, the percentage of positive Stathmin 1 cells was significantly higher in the subgranular zone of ECT patients, indicating neuroplasticity. These first post-mortem observations suggest that ECT has no damaging effects but may rather have induced neuroplasticity in the DG of depressed patients.
Collapse
Affiliation(s)
- Dore Loef
- Amsterdam UMC, location VUmc, Amsterdam, Department of Psychiatry, Amsterdam Neuroscience, Amsterdam, the Netherlands.
- GGZ inGeest Specialized Mental Health Care, Amsterdam, the Netherlands.
| | - Indira Tendolkar
- Department of Psychiatry, Radboud University Medical Center, Nijmegen, the Netherlands
- Donders Institute for Brain, Cognition and Behavior, Centre for Cognitive Neuroimaging, Nijmegen, the Netherlands
- Department of Psychiatry and Psychotherapy, University Hospital Essen, Essen, Germany
| | - Philip F P van Eijndhoven
- Department of Psychiatry, Radboud University Medical Center, Nijmegen, the Netherlands
- Donders Institute for Brain, Cognition and Behavior, Centre for Cognitive Neuroimaging, Nijmegen, the Netherlands
| | - Jeroen J M Hoozemans
- Department of Pathology, Amsterdam Neuroscience, Amsterdam University Medical Centre, Amsterdam, the Netherlands
| | - Mardien L Oudega
- Amsterdam UMC, location VUmc, Amsterdam, Department of Psychiatry, Amsterdam Neuroscience, Amsterdam, the Netherlands
- GGZ inGeest Specialized Mental Health Care, Amsterdam, the Netherlands
| | - Annemieke J M Rozemuller
- Department of Pathology, Amsterdam Neuroscience, Amsterdam University Medical Centre, Amsterdam, the Netherlands
| | - Paul J Lucassen
- Brain Plasticity Group, Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, the Netherlands
| | - Annemiek Dols
- Amsterdam UMC, location VUmc, Amsterdam, Department of Psychiatry, Amsterdam Neuroscience, Amsterdam, the Netherlands
- Department of Psychiatry, UMC Utrecht Brain Center, University Utrecht, Utrecht, the Netherlands
| | - Anke A Dijkstra
- Molecular Neuroscience Group, Swammerdam Institute for Life Sciences, Center for Neuroscience, University of Amsterdam, Amsterdam, the Netherlands
| |
Collapse
|
12
|
Zhuang H, Li Q, Sun C, Xu D, Gan G, Zhang C, Chen C, Yuan Y, Liu L, Xiao Y, Yao X, Wang C, Kang X, Yang C, Zhao J, Chen W, Wang J, Li J, Luo C, Wang J, Jia X, Yu Z, Liu L. Voluntary wheel exercise ameliorates cognitive impairment, hippocampal neurodegeneration and microglial abnormalities preceded by demyelination in a male mouse model of noise-induced hearing loss. Brain Behav Immun 2023; 114:325-348. [PMID: 37683962 DOI: 10.1016/j.bbi.2023.09.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 08/23/2023] [Accepted: 09/05/2023] [Indexed: 09/10/2023] Open
Abstract
Acquired peripheral hearing loss (APHL) in midlife has been identified as the greatest modifiable risk factor for dementia; however, the pathophysiological neural mechanisms linking APHL with an increased risk of dementia remain to be elucidated. Here, in an adult male mouse model of noise-induced hearing loss (NIHL), one of the most common forms of APHL, we demonstrated accelerated age-related cognitive decline and hippocampal neurodegeneration during a 6-month follow-up period, accompanied by progressive hippocampal microglial aberrations preceded by immediate-onset transient elevation in serum glucocorticoids and delayed-onset sustained myelin disruption in the hippocampus. Pretreatment with the glucocorticoid receptor antagonist RU486 before stressful noise exposure partially mitigated the early activation of hippocampal microglia, which were present at 7 days post noise exposure (7DPN), but had no impact on later microglial aberrations, hippocampal neurodegeneration, or cognitive decline exhibited at 1 month post noise exposure (1MPN). One month of voluntary wheel exercise following noise exposure barely affected either the hearing threshold shift or hippocampal myelin changes but effectively countered cognitive impairment and the decline in hippocampal neurogenesis in NIHL mice at 1MPN, paralleled by the normalization of microglial morphology, which coincided with a reduction in microglial myelin inclusions and a restoration of microglial hypoxia-inducible factor-1α (HIF1α) expression. Our results indicated that accelerated cognitive deterioration and hippocampal neuroplastic decline following NIHL are most likely driven by the maladaptive response of hippocampal microglia to myelin damage secondary to hearing loss, and we also demonstrated the potential of voluntary physical exercise as a promising and cost-effective strategy to alleviate the detrimental impact of APHL on cognitive function and thus curtail the high and continuously increasing global burden of dementia. Furthermore, the findings of the present study highlight the contribution of myelin debris overload to microglial malfunction and identify the microglial HIF1α-related pathway as an attractive candidate for future comprehensive investigation to obtain a more definitive picture of the underlying mechanisms linking APHL and dementia.
Collapse
Affiliation(s)
- Hong Zhuang
- Department of Physiology, School of Medicine, Southeast University, Nanjing 210009, China
| | - Qian Li
- Department of Physiology, School of Medicine, Southeast University, Nanjing 210009, China
| | - Congli Sun
- Department of Physiology, School of Medicine, Southeast University, Nanjing 210009, China
| | - Dan Xu
- School of Public Health, Southeast University, Nanjing 210009, China
| | - Guangming Gan
- School of Medicine, Southeast University, Nanjing 210009, China
| | - Chenchen Zhang
- School of Medicine, Southeast University, Nanjing 210009, China
| | - Chen Chen
- School of Medicine, Southeast University, Nanjing 210009, China
| | - Yang Yuan
- Department of Endocrinology, Zhongda Hospital, Medical School, Southeast University, Nanjing 210009, China
| | - Linchen Liu
- Department of Rheumatology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, China
| | - Yu Xiao
- Department of Physiology, School of Medicine, Southeast University, Nanjing 210009, China
| | - Xiuting Yao
- Department of Physiology, School of Medicine, Southeast University, Nanjing 210009, China
| | - Conghui Wang
- Department of Physiology, School of Medicine, Southeast University, Nanjing 210009, China
| | - Xiaoming Kang
- School of Life Science and Technology, Southeast University, Nanjing 210009, China
| | - Chenxi Yang
- Department of Physiology, School of Medicine, Southeast University, Nanjing 210009, China
| | - Jingyi Zhao
- School of Life Science and Technology, Southeast University, Nanjing 210009, China
| | - Wenhao Chen
- School of Medicine, Southeast University, Nanjing 210009, China
| | - Jiatang Wang
- School of Medicine, Southeast University, Nanjing 210009, China
| | - Jinyu Li
- School of Medicine, Southeast University, Nanjing 210009, China
| | - Caichen Luo
- School of Medicine, Southeast University, Nanjing 210009, China
| | - Jie Wang
- School of Medicine, Southeast University, Nanjing 210009, China
| | - Xirui Jia
- School of Life Science and Technology, Southeast University, Nanjing 210009, China
| | - Zhehao Yu
- Department of Physiology, School of Medicine, Southeast University, Nanjing 210009, China
| | - Lijie Liu
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Department of Physiology, School of Medicine, Southeast University, Nanjing 210009, China.
| |
Collapse
|
13
|
Sinks MR, Morrison DE, Ramdev RA, Lentzou S, Spritzer MD. Cell proliferation and cell death levels in the dentate gyrus correlate with home range size among adult male meadow voles. Neuroscience 2023:S0306-4522(23)00231-2. [PMID: 37245693 DOI: 10.1016/j.neuroscience.2023.05.018] [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: 11/21/2022] [Revised: 05/07/2023] [Accepted: 05/20/2023] [Indexed: 05/30/2023]
Abstract
Neurogenesis occurs throughout adulthood within the dentate gyrus, and evidence indicates that these new neurons play a critical role in both spatial and social memory. However, a vast majority of past research on adult neurogenesis has involved experiments with captive mice and rats, making the generalizability of results to natural settings questionable. We assessed the connection between adult neurogenesis and memory by measuring the home range size of wild-caught, free-ranging meadow voles (Microtus pennsylvanicus). Adult male voles (n = 18) were captured, fitted with radio collars, and released back into their natural habitat, where each vole's home range was assessed using 40 radio-telemetry fixes over the course of 5 evenings. Voles were then recaptured, and brain tissue was collected. Cellular markers of cell proliferation (pHisH3, Ki67), neurogenesis (DCX), and pyknosis were labeled on histological sections and then quantified using either fluorescent or light microscopy. Voles with larger home ranges had significantly higher pHisH3+ cell densities within the granule cell layer and subgranular zone (GCL+SGZ) of the dentate gyrus and higher Ki67+ cell densities in the dorsal GCL+SGZ. Voles with larger ranges also had significantly higher pyknotic cell densities in the entire GCL+SGZ and in the dorsal GCL+SGZ. These results support the hypothesis that cell proliferation and cell death within the hippocampus are involved with spatial memory formation. However, a marker of neurogenesis (DCX+) was not correlated with range size, suggesting that there may be selective cellular turnover in the dentate gyrus when a vole is ranging through its environment.
Collapse
Affiliation(s)
- Mark R Sinks
- Department of Biology, Middlebury College, McCardell Bicentennial Hall, Middlebury, VT 05753, U.S.A.
| | - Daryl E Morrison
- Department of Biology, Middlebury College, McCardell Bicentennial Hall, Middlebury, VT 05753, U.S.A.
| | - Rajan A Ramdev
- Program in Neuroscience, Middlebury College, McCardell Bicentennial Hall, Middlebury, VT 05753, U.S.A.
| | - Stergiani Lentzou
- Program in Neuroscience, Middlebury College, McCardell Bicentennial Hall, Middlebury, VT 05753, U.S.A.
| | - Mark D Spritzer
- Department of Biology, Middlebury College, McCardell Bicentennial Hall, Middlebury, VT 05753, U.S.A; Program in Neuroscience, Middlebury College, McCardell Bicentennial Hall, Middlebury, VT 05753, U.S.A.
| |
Collapse
|
14
|
Huang R, Lin B, Tian H, Luo Q, Li Y. Prenatal Exposure to General Anesthesia Drug Esketamine Impaired Neurobehavior in Offspring. Cell Mol Neurobiol 2023:10.1007/s10571-023-01354-4. [PMID: 37119312 DOI: 10.1007/s10571-023-01354-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Accepted: 04/19/2023] [Indexed: 05/01/2023]
Abstract
Prenatal exposure to anesthetics has raised increasing attention about the neuronal development in offspring. Animal models are usually used for investigation. As a new drug, esketamine is the s-isoform of ketamine and is twice as potent as the racemic ketamine with less reported adverse effects. Esketamine is currently being used and become more favorable in clinical anesthesia work, including surgeries during pregnancy, yet the effect on the offspring is unknown. The present study aimed to elucidate the effects of gestational administration of esketamine on neuronal development in offspring, using a rat model. Gestational day 14.5 pregnant rats received intravenous injections of esketamine. The postnatal day 0 (P0) hippocampus was digested and cultured in vitro to display the neuronal growth morphology. On Day 4 the in vitro experiments revealed a shorter axon length and fewer dendrite branches in the esketamine group. The results from the EdU- imaging kit showed decreased proliferative capacity in the subventricular zone (SVZ) and dentate gyrus (DG) in both P0 and P30 offspring brains in the esketamine group. Moreover, neurogenesis, neuron maturity and spine density were impaired, resulting in attenuated long-term potentiation (LTP). Compromised hippocampal function accounted for the deficits in neuronal cognition, memory and emotion. The evidence obtained suggests that the neurobehavioral deficit due to prenatal exposure to esketamine may be related to the decrease phosphorylation of CREB and abnormalities in N-methyl-D-aspartic acid receptor subunits. Taken together, these results demonstrate the negative effect of prenatal esketamine exposure on neuronal development in offspring rats. G14.5 esketamine administration influenced the neurobehavior of the offspring in adolescence. Poorer neuronal growth and reduced brain proliferative capacity in late gestation and juvenile pups resulted in impaired P30 neuronal plasticity and synaptic spines as well as abnormalities in NMDAR subunits. Attenuated LTP reflected compromised hippocampal function, as confirmed by behavioral tests of cognition, memory and emotions. This figure was completed on the website of Figdraw.
Collapse
Affiliation(s)
- Ronghua Huang
- Department of Anesthesiology, The First Affiliated Hospital of Jinan University, Number 613, The West of Huangpu Avenue, Tianhe Region, Guangzhou, 510630, Guangdong Province, China
| | - Bingbiao Lin
- Department of Urology, Kidney and Urology Center, Pelvic Floor Disorders Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, 518000, Guangdong, China
| | - Hongyan Tian
- Department of Anesthesiology, The First Affiliated Hospital of Jinan University, Number 613, The West of Huangpu Avenue, Tianhe Region, Guangzhou, 510630, Guangdong Province, China
| | - Qichen Luo
- Department of Anesthesiology, The First Affiliated Hospital of Jinan University, Number 613, The West of Huangpu Avenue, Tianhe Region, Guangzhou, 510630, Guangdong Province, China
| | - Yalan Li
- Department of Anesthesiology, The First Affiliated Hospital of Jinan University, Number 613, The West of Huangpu Avenue, Tianhe Region, Guangzhou, 510630, Guangdong Province, China.
| |
Collapse
|
15
|
Ehlers JS, Bracke K, von Bohlen Und Halbach V, Siegerist F, Endlich N, von Bohlen Und Halbach O. Morphological and behavioral analysis of Slc35f1-deficient mice revealed no neurodevelopmental phenotype. Brain Struct Funct 2023; 228:895-906. [PMID: 36951990 PMCID: PMC10147817 DOI: 10.1007/s00429-023-02629-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 03/09/2023] [Indexed: 03/24/2023]
Abstract
SLC35F1 is a member of the sugar-like carrier (SLC) superfamily that is expressed in the mammalian brain. Malfunction of SLC35F1 in humans is associated with neurodevelopmental disorders. To get insight into the possible roles of Slc35f1 in the brain, we generated Slc35f1-deficient mice. The Slc35f1-deficient mice are viable and survive into adulthood, which allowed examining adult Slc35f1-deficient mice on the anatomical as well as behavioral level. In humans, mutation in the SLC35F1 gene can induce a Rett syndrome-like phenotype accompanied by intellectual disability (Fede et al. Am J Med Genet A 185:2238-2240, 2021). The Slc35f1-deficient mice, however, display only a very mild phenotype and no obvious deficits in learning and memory as, e.g., monitored with the novel object recognition test or the Morris water maze test. Moreover, neuroanatomical parameters of neuronal plasticity (as dendritic spines and adult hippocampal neurogenesis) are also unaltered. Thus, Slc35f1-deficient mice display no major alterations that resemble a neurodevelopmental phenotype.
Collapse
Affiliation(s)
- Julia Sophie Ehlers
- Institute for Anatomy and Cell Biology, Universitätsmedizin Greifswald, Friedrich Loeffler Str. 23C, 17487, Greifswald, Germany
| | - Katharina Bracke
- Institute for Anatomy and Cell Biology, Universitätsmedizin Greifswald, Friedrich Loeffler Str. 23C, 17487, Greifswald, Germany
| | - Viola von Bohlen Und Halbach
- Institute for Anatomy and Cell Biology, Universitätsmedizin Greifswald, Friedrich Loeffler Str. 23C, 17487, Greifswald, Germany
| | - Florian Siegerist
- Institute for Anatomy and Cell Biology, Universitätsmedizin Greifswald, Friedrich Loeffler Str. 23C, 17487, Greifswald, Germany
| | - Nicole Endlich
- Institute for Anatomy and Cell Biology, Universitätsmedizin Greifswald, Friedrich Loeffler Str. 23C, 17487, Greifswald, Germany
| | - Oliver von Bohlen Und Halbach
- Institute for Anatomy and Cell Biology, Universitätsmedizin Greifswald, Friedrich Loeffler Str. 23C, 17487, Greifswald, Germany.
| |
Collapse
|
16
|
Paronett EM, Bryan CA, Maynard TM, LaMantia AS. Identity, lineage and fates of a temporally distinct progenitor population in the embryonic olfactory epithelium. Dev Biol 2023; 495:76-91. [PMID: 36627077 PMCID: PMC9926479 DOI: 10.1016/j.ydbio.2023.01.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 12/29/2022] [Accepted: 01/01/2023] [Indexed: 01/09/2023]
Abstract
We defined a temporally and transcriptionally divergent precursor cohort in the medial olfactory epithelium (OE) shortly after it differentiates as a distinct tissue at mid-gestation in the mouse. This temporally distinct population of Ascl1+ cells in the dorsomedial OE is segregated from Meis1+/Pax7+ progenitors in the lateral OE, and does not appear to be generated by Pax7+ lateral OE precursors. The medial Ascl1+ precursors do not yield a substantial number of early-generated ORNs. Instead, they first generate additional proliferative precursors as well as a distinct population of frontonasal mesenchymal cells associated with the migratory mass that surrounds the nascent olfactory nerve. Parallel to these in vivo distinctions, isolated medial versus lateral OE precursors in vitro retain distinct proliferative capacities and modes of division that reflect their in vivo identities. At later fetal stages, these early dorsomedial Ascl1+ precursors cells generate spatially restricted subsets of ORNs as well as other non-neuronal cell classes. Accordingly, the initial compliment of ORNs and other OE cell types is derived from at least two distinct early precursor populations: lateral Meis1/Pax7+ precursors that generate primarily early ORNs, and a temporally, spatially, and transcriptionally distinct subset of medial Ascl1+ precursors that initially generate additional OE progenitors and apparent migratory mass cells before yielding a subset of ORNs and likely supporting cell classes.
Collapse
Affiliation(s)
- Elizabeth M Paronett
- Department of Pharmacology and Physiology, George Washington University School of Medicine, Washington, DC, 20037, USA
| | - Corey A Bryan
- Laboratory of Developmental Disorders and Genetics, The Fralin Biomedical Research Institute, Virginia Tech-Carilion School of Medicine, Roanoke, VA, USA
| | - Thomas M Maynard
- Center for Neurobiology Research, The Fralin Biomedical Research Institute, Virginia Tech-Carilion School of Medicine, Roanoke, VA, USA
| | - Anthony-S LaMantia
- Center for Neurobiology Research, The Fralin Biomedical Research Institute, Virginia Tech-Carilion School of Medicine, Roanoke, VA, USA; Department of Biological Sciences Virginia Tech, Blacksburg, VA, USA.
| |
Collapse
|
17
|
Pathological Nuclear Hallmarks in Dentate Granule Cells of Alzheimer’s Patients: A Biphasic Regulation of Neurogenesis. Int J Mol Sci 2022; 23:ijms232112873. [PMID: 36361662 PMCID: PMC9654738 DOI: 10.3390/ijms232112873] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2022] [Revised: 10/14/2022] [Accepted: 10/18/2022] [Indexed: 11/29/2022] Open
Abstract
The dentate gyrus (DG) of the human hippocampus is a complex and dynamic structure harboring mature and immature granular neurons in diverse proliferative states. While most mammals show persistent neurogenesis through adulthood, human neurogenesis is still under debate. We found nuclear alterations in granular cells in autopsied human brains, detected by immunohistochemistry. These alterations differ from those reported in pyramidal neurons of the hippocampal circuit. Aging and early AD chromatin were clearly differentiated by the increased epigenetic markers H3K9me3 (heterochromatin suppressive mark) and H3K4me3 (transcriptional euchromatin mark). At early AD stages, lamin B2 was redistributed to the nucleoplasm, indicating cell-cycle reactivation, probably induced by hippocampal nuclear pathology. At intermediate and late AD stages, higher lamin B2 immunopositivity in the perinucleus suggests fewer immature neurons, less neurogenesis, and fewer adaptation resources to environmental factors. In addition, senile samples showed increased nuclear Tau interacting with aged chromatin, likely favoring DNA repair and maintaining genomic stability. However, at late AD stages, the progressive disappearance of phosphorylated Tau forms in the nucleus, increased chromatin disorganization, and increased nuclear autophagy support a model of biphasic neurogenesis in AD. Therefore, designing therapies to alleviate the neuronal nuclear pathology might be the only pathway to a true rejuvenation of brain circuits.
Collapse
|
18
|
Crespo I, Pignatelli J, Kinare V, Méndez-Gómez HR, Esgleas M, Román MJ, Canals JM, Tole S, Vicario C. Tbr1 Misexpression Alters Neuronal Development in the Cerebral Cortex. Mol Neurobiol 2022; 59:5750-5765. [PMID: 35781633 PMCID: PMC9395452 DOI: 10.1007/s12035-022-02936-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 06/10/2022] [Indexed: 11/26/2022]
Abstract
Changes in the transcription factor (TF) expression are critical for brain development, and they may also underlie neurodevelopmental disorders. Indeed, T-box brain1 (Tbr1) is a TF crucial for the formation of neocortical layer VI, and mutations and microdeletions in that gene are associated with malformations in the human cerebral cortex, alterations that accompany autism spectrum disorder (ASD). Interestingly, Tbr1 upregulation has also been related to the occurrence of ASD-like symptoms, although limited studies have addressed the effect of increased Tbr1 levels during neocortical development. Here, we analysed the impact of Tbr1 misexpression in mouse neural progenitor cells (NPCs) at embryonic day 14.5 (E14.5), when they mainly generate neuronal layers II-IV. By E18.5, cells accumulated in the intermediate zone and in the deep cortical layers, whereas they became less abundant in the upper cortical layers. In accordance with this, the proportion of Sox5+ cells in layers V-VI increased, while that of Cux1+ cells in layers II-IV decreased. On postnatal day 7, fewer defects in migration were evident, although a higher proportion of Sox5+ cells were seen in the upper and deep layers. The abnormal neuronal migration could be partially due to the altered multipolar-bipolar neuron morphologies induced by Tbr1 misexpression, which also reduced dendrite growth and branching, and disrupted the corpus callosum. Our results indicate that Tbr1 misexpression in cortical NPCs delays or disrupts neuronal migration, neuronal specification, dendrite development and the formation of the callosal tract. Hence, genetic changes that provoke ectopic Tbr1 upregulation during development could provoke cortical brain malformations.
Collapse
Affiliation(s)
- Inmaculada Crespo
- Instituto Cajal-Consejo Superior de Investigaciones Científicas (CSIC), Avenida Doctor Arce 37, 28002, Madrid, Spain
- CIBERNED-Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- CES Cardenal Cisneros, Madrid, Spain
| | - Jaime Pignatelli
- Instituto Cajal-Consejo Superior de Investigaciones Científicas (CSIC), Avenida Doctor Arce 37, 28002, Madrid, Spain
- CIBERNED-Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Veena Kinare
- Department of Life Sciences, Sophia College for Women, Mumbai, 400026, India
| | - Héctor R Méndez-Gómez
- Instituto Cajal-Consejo Superior de Investigaciones Científicas (CSIC), Avenida Doctor Arce 37, 28002, Madrid, Spain
- CIBERNED-Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Miriam Esgleas
- CIBERNED-Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Laboratory of Stem Cells and Regenerative Medicine, Department of Biomedical Sciences, Creatio, Production and Validation Center of Advanced Therapies, Faculty of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona, Barcelona, Spain
- August Pi I Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
| | - María José Román
- Instituto Cajal-Consejo Superior de Investigaciones Científicas (CSIC), Avenida Doctor Arce 37, 28002, Madrid, Spain
- CIBERNED-Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Josep M Canals
- CIBERNED-Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Laboratory of Stem Cells and Regenerative Medicine, Department of Biomedical Sciences, Creatio, Production and Validation Center of Advanced Therapies, Faculty of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona, Barcelona, Spain
- August Pi I Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
| | - Shubha Tole
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai, 400005, India
| | - Carlos Vicario
- Instituto Cajal-Consejo Superior de Investigaciones Científicas (CSIC), Avenida Doctor Arce 37, 28002, Madrid, Spain.
- CIBERNED-Instituto de Salud Carlos III (ISCIII), Madrid, Spain.
| |
Collapse
|
19
|
Zhang S, Kong DW, Ma GD, Liu CD, Yang YJ, Liu S, Jiang N, Pan ZR, Zhang W, Kong LL, Du GH. Long-term administration of salvianolic acid A promotes endogenous neurogenesis in ischemic stroke rats through activating Wnt3a/GSK3β/β-catenin signaling pathway. Acta Pharmacol Sin 2022; 43:2212-2225. [PMID: 35217812 PMCID: PMC9433393 DOI: 10.1038/s41401-021-00844-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Accepted: 12/15/2021] [Indexed: 12/20/2022] Open
Abstract
Stroke is the major cause of death and disability worldwide. Most stroke patients who survive in the acute phase of ischemia display various extents of neurological deficits. In order to improve the prognosis of ischemic stroke, promoting endogenous neurogenesis has attracted great attention. Salvianolic acid A (SAA) has shown neuroprotective effects against ischemic diseases. In the present study, we investigated the neurogenesis effects of SAA in ischemic stroke rats, and explored the underlying mechanisms. An autologous thrombus stroke model was established by electrocoagulation. The rats were administered SAA (10 mg/kg, ig) or a positive drug edaravone (5 mg/kg, iv) once a day for 14 days. We showed that SAA administration significantly decreased infarction volume and vascular embolism, and ameliorated pathological injury in the hippocampus and striatum as well as the neurological deficits as compared with the model rats. Furthermore, we found that SAA administration significantly promoted neural stem/progenitor cells (NSPCs) proliferation, migration and differentiation into neurons, enhanced axonal regeneration and diminished neuronal apoptosis around the ipsilateral subventricular zone (SVZ), resulting in restored neural density and reconstructed neural circuits in the ischemic striatum. Moreover, we revealed that SAA-induced neurogenesis was associated to activating Wnt3a/GSK3β/β-catenin signaling pathway and downstream target genes in the hippocampus and striatum. Edaravone exerted equivalent inhibition on neuronal apoptosis in the SVZ, as SAA, but edaravone-induced neurogenesis was weaker than that of SAA. Taken together, our results demonstrate that long-term administration of SAA improves neurological function through enhancing endogenous neurogenesis and inhibiting neuronal apoptosis in ischemic stroke rats via activating Wnt3a/GSK3β/β-catenin signaling pathway. SAA may be a potential therapeutic drug to promote neurogenesis after stroke.
Collapse
Affiliation(s)
- Sen Zhang
- Beijing Key Laboratory of Drug Targets Identification and Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, 100050, China
| | - De-Wen Kong
- Beijing Key Laboratory of Drug Targets Identification and Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, 100050, China
| | - Guo-Dong Ma
- Beijing Key Laboratory of Drug Targets Identification and Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, 100050, China
| | - Cheng-di Liu
- Beijing Key Laboratory of Drug Targets Identification and Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, 100050, China
| | - Yu-Jiao Yang
- Beijing Key Laboratory of Drug Targets Identification and Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, 100050, China
- School of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Shan Liu
- Beijing Key Laboratory of Drug Targets Identification and Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, 100050, China
- College of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Nan Jiang
- Beijing Key Laboratory of Drug Targets Identification and Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, 100050, China
- School of Pharmacy, Henan University, Zhengzhou, 475004, China
| | - Zi-Rong Pan
- Beijing Key Laboratory of Drug Targets Identification and Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, 100050, China
| | - Wen Zhang
- Beijing Key Laboratory of Drug Targets Identification and Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, 100050, China
| | - Ling-Lei Kong
- Beijing Key Laboratory of Drug Targets Identification and Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, 100050, China.
| | - Guan-Hua Du
- Beijing Key Laboratory of Drug Targets Identification and Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, 100050, China.
| |
Collapse
|
20
|
Longterm Increased S100B Enhances Hippocampal Progenitor Cell Proliferation in a Transgenic Mouse Model. Int J Mol Sci 2022; 23:ijms23179600. [PMID: 36076994 PMCID: PMC9455494 DOI: 10.3390/ijms23179600] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Revised: 07/13/2022] [Accepted: 08/19/2022] [Indexed: 12/11/2022] Open
Abstract
(1) The neurotrophic protein S100B is a marker of brain injury and has been associated with neuroregeneration. In S100Btg mice rendering 12 copies of the murine S100B gene we evaluated whether S100B may serve as a treatment option. (2) In juvenile, adult, and one-year-old S100Btg mice (female and male; n = 8 per group), progenitor cell proliferation was quantified in the subgranular zone (SGZ) and the granular cell layer (GCL) of the dentate gyrus with the proliferative marker Ki67 and BrdU (50 mg/kg). Concomitant signaling was quantified utilizing glial fibrillary acidic protein (GFAP), apolipoprotein E (ApoE), brain-derived neurotrophic factor (BDNF), and the receptor for advanced glycation end products (RAGE) immunohistochemistry. (3) Progenitor cell proliferation in the SGZ and migration to the GCL was enhanced. Hippocampal GFAP was reduced in one-year-old S100Btg mice. ApoE in the hippocampus and frontal cortex of male and BDNF in the frontal cortex of female S100Btg mice was reduced. RAGE was not affected. (4) Enhanced hippocampal neurogenesis in S100Btg mice was not accompanied by reactive astrogliosis. Sex- and brain region-specific variations of ApoE and BDNF require further elucidations. Our data reinforce the importance of this S100Btg model in evaluating the role of S100B in neuroregenerative medicine.
Collapse
|
21
|
Aronica E, Binder DK, Drexel M, Ikonomidou C, Kadam SD, Sperk G, Steinhäuser C. A companion to the preclinical common data elements and case report forms for neuropathology studies in epilepsy research. A report of the TASK3 WG2 Neuropathology Working Group of the ILAE/AES Joint Translational Task Force. Epilepsia Open 2022. [PMID: 35938285 DOI: 10.1002/epi4.12638] [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: 11/27/2021] [Accepted: 01/28/2022] [Indexed: 11/06/2022] Open
Abstract
The International League Against Epilepsy/American Epilepsy Society (ILAE/AES) Joint Translational Task Force initiated the TASK3 working group to create common data elements (CDEs) for various aspects of preclinical epilepsy research studies, which could help improve the standardization of experimental designs. This article addresses neuropathological changes associated with seizures and epilepsy in rodent models of epilepsy. We discuss CDEs for histopathological parameters for neurodegeneration, changes in astrocyte morphology and function, mechanisms of inflammation, and changes in the blood-brain barrier and myelin/oligodendrocytes resulting from recurrent seizures in rats and mice. We provide detailed CDE tables and case report forms (CRFs), and with this companion manuscript, we discuss the rationale and methodological aspects of individual neuropathological examinations. The CDEs, CRFs, and companion paper are available to all researchers, and their use will benefit the harmonization and comparability of translational preclinical epilepsy research. The ultimate hope is to facilitate the development of rational therapy concepts for treating epilepsies, seizures, and comorbidities and the development of biomarkers assessing the pathological state of the disease.
Collapse
Affiliation(s)
- Eleonora Aronica
- Amsterdam UMC, University of Amsterdam, Department of (Neuro) Pathology, Amsterdam Neuroscience, Amsterdam, The Netherlands
- Stichting Epilepsie Instellingen Nederland (SEIN), Heemstede, The Netherlands
| | - Devin K Binder
- Center for Glial-Neuronal Interactions, Division of Biomedical Sciences, School of Medicine, University of California, Riverside, California, USA
| | - Meinrad Drexel
- Department of Genetics and Pharmacology, Institute of Molecular and Cellular Pharmacology, Medical University Innsbruck, Innsbruck, Austria
| | | | - Shilpa D Kadam
- The Hugo Moser Research Institute at Kennedy Krieger, Baltimore, Maryland, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Guenther Sperk
- Department of Pharmacology, Medical University Innsbruck, Innsbruck, Austria
| | - Christian Steinhäuser
- Institute of Cellular Neurosciences, Medical School, University of Bonn, Bonn, Germany
| |
Collapse
|
22
|
Jones KL, Zhou M, Jhaveri DJ. Dissecting the role of adult hippocampal neurogenesis towards resilience versus susceptibility to stress-related mood disorders. NPJ SCIENCE OF LEARNING 2022; 7:16. [PMID: 35842419 PMCID: PMC9288448 DOI: 10.1038/s41539-022-00133-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 07/01/2022] [Indexed: 05/13/2023]
Abstract
Adult hippocampal neurogenesis in the developmental process of generating and integrating new neurons in the hippocampus during adulthood and is a unique form of structural plasticity with enormous potential to modulate neural circuit function and behaviour. Dysregulation of this process is strongly linked to stress-related neuropsychiatric conditions such as anxiety and depression, and efforts have focused on unravelling the contribution of adult-born neurons in regulating stress response and recovery. Chronic stress has been shown to impair this process, whereas treatment with clinical antidepressants was found to enhance the production of new neurons in the hippocampus. However, the precise role of adult hippocampal neurogenesis in mediating the behavioural response to chronic stress is not clear and whether these adult-born neurons buffer or increase susceptibility to stress-induced mood-related maladaptation remains one of the controversial issues. In this review, we appraise evidence probing the causal role of adult hippocampal neurogenesis in the regulation of emotional behaviour in rodents. We find that the relationship between adult-born hippocampal neurons and stress-related mood disorders is not linear, and that simple subtraction or addition of these neurons alone is not sufficient to lead to anxiety/depression or have antidepressant-like effects. We propose that future studies examining how stress affects unique properties of adult-born neurons, such as the excitability and the pattern of connectivity during their critical period of maturation will provide a deeper understanding of the mechanisms by which these neurons contribute to functional outcomes in stress-related mood disorders.
Collapse
Affiliation(s)
- Katherine L Jones
- Queensland Brain Institute, University of Queensland, Brisbane, QLD 4072, Australia
| | - Mei Zhou
- Queensland Brain Institute, University of Queensland, Brisbane, QLD 4072, Australia
- Mater Research Institute - University of Queensland, Translational Research Institute, Woolloongabba, QLD 4102, Australia
| | - Dhanisha J Jhaveri
- Queensland Brain Institute, University of Queensland, Brisbane, QLD 4072, Australia.
- Mater Research Institute - University of Queensland, Translational Research Institute, Woolloongabba, QLD 4102, Australia.
| |
Collapse
|
23
|
Osacka J, Kiss A, Bacova Z, Tillinger A. Effect of Haloperidol and Olanzapine on Hippocampal Cells’ Proliferation in Animal Model of Schizophrenia. Int J Mol Sci 2022; 23:ijms23147711. [PMID: 35887056 PMCID: PMC9323809 DOI: 10.3390/ijms23147711] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 07/07/2022] [Accepted: 07/12/2022] [Indexed: 12/07/2022] Open
Abstract
Aberrant neurogenesis in the subventricular zone (SVZ) and hippocampus (HIP) contributes to schizophrenia pathogenesis. Haloperidol (HAL) and olanzapine (OLA), commonly prescribed antipsychotics for schizophrenia treatment, affect neurogenesis too. The effect of HAL and OLA on an mHippoE-2 cell line was studied in vitro where we measured the cell number and projection length. In vivo, we studied the gene expression of DCX, Sox2, BDNF, and NeuN in the SVZ and HIP in an MK-801-induced animal schizophrenia model. Cells were incubated with HAL, OLA, and MK-801 for 24, 48, and 72 h. Animals were injected for 6 days with saline or MK801 (0.5 mg/kg), and from the 7th day with either vehicle HAL (1 mg/kg) or OLA (2 mg/kg), for the next 7 days. In vitro, HAL and OLA dose/time-dependently suppressed cells’ proliferation and shortened their projection length. HAL/OLA co-treatment with MK-801 for 24 h reversed HAL’s/OLA’s inhibitory effect. In vivo, HAL and OLA suppressed DCX and NeuN genes’ expression in the HIP and SVZ. MK-801 decreased DCX and NeuN genes’ expression in the HIP and OLA prevented this effect. The data suggest that subchronic HAL/OLA treatment can inhibit DCX and NeuN expression. In an MK-801 schizophrenia model, OLA reversed the MK-801 inhibitory effect on DCX and NeuN and HAL reversed the effect on DCX expression; however, only in the HIP.
Collapse
|
24
|
Gao J, Liu J, Yao M, Zhang W, Yang B, Wang G. Panax notoginseng Saponins Stimulates Neurogenesis and Neurological Restoration After Microsphere-Induced Cerebral Embolism in Rats Partially Via mTOR Signaling. Front Pharmacol 2022; 13:889404. [PMID: 35770087 PMCID: PMC9236302 DOI: 10.3389/fphar.2022.889404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 05/23/2022] [Indexed: 11/30/2022] Open
Abstract
P. Notoginseng Saponins (PNS), the main active component of herbal medicine Panax notoginseng, has been widely used to treat cerebrovascular diseases. It has been acknowledged that PNS exerted protection on nerve injuries induced by ischemic stroke, however, the long-term impacts of PNS on the restoration of neurological defects and neuroregeneration after stroke have not been thoroughly studied and the underlying molecular mechanism of stimulating neurogenesis is difficult to precisely clarify, much more in-depth researches are badly needed. In the present study, cerebral ischemia injury was induced by microsphere embolism (ME) in rats. After 14 days, PNS administration relieved cerebral ischemia injury as evidenced by alleviating neurological deficits and reducing hippocampal pathological damage. What’s more, PNS stimulated hippocampal neurogenesis by promoting cell proliferation, migration and differentiation activity and modulated synaptic plasticity. Increased number of BrdU/Nestin, BrdU/DCX and NeuroD1-positive cells and upregulated synapse-related GAP43, SYP, and PSD95 expression were observed in the hippocampus. We hypothesized that upregulation of brain-derived neurotrophic factor (BDNF) expression and activation of Akt/mTOR/p70S6K signaling after ME could partially underlie the neuroprotective effects of PNS against cerebral ischemia injury. Our findings offer some new viewpoints into the beneficial roles of PNS against ischemic stroke.
Collapse
Affiliation(s)
- Jiale Gao
- Beijing Key Laboratory of Pharmacology of Chinese Materia Medica, Institute of Basic Medical Sciences of Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jianxun Liu
- Beijing Key Laboratory of Pharmacology of Chinese Materia Medica, Institute of Basic Medical Sciences of Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- *Correspondence: Jianxun Liu,
| | - Mingjiang Yao
- Beijing Key Laboratory of Pharmacology of Chinese Materia Medica, Institute of Basic Medical Sciences of Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Wei Zhang
- Department of Pathology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Bin Yang
- Department of Pathology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Guangrui Wang
- Beijing Key Laboratory of Pharmacology of Chinese Materia Medica, Institute of Basic Medical Sciences of Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
25
|
Ivanova A, Gruzova O, Ermolaeva E, Astakhova O, Itaman S, Enikolopov G, Lazutkin A. Synthetic Thymidine Analog Labeling without Misconceptions. Cells 2022; 11:cells11121888. [PMID: 35741018 PMCID: PMC9220989 DOI: 10.3390/cells11121888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 06/08/2022] [Accepted: 06/08/2022] [Indexed: 11/16/2022] Open
Abstract
Tagging proliferating cells with thymidine analogs is an indispensable research tool; however, the issue of the potential in vivo cytotoxicity of these compounds remains unresolved. Here, we address these concerns by examining the effects of BrdU and EdU on adult hippocampal neurogenesis and EdU on the perinatal somatic development of mice. We show that, in a wide range of doses, EdU and BrdU label similar numbers of cells in the dentate gyrus shortly after administration. Furthermore, whereas the administration of EdU does not affect the division and survival of neural progenitor within 48 h after injection, it does affect cell survival, as evaluated 6 weeks later. We also show that a single injection of various doses of EdU on the first postnatal day does not lead to noticeable changes in a panel of morphometric criteria within the first week; however, higher doses of EdU adversely affect the subsequent somatic maturation and brain growth of the mouse pups. Our results indicate the potential caveats in labeling the replicating DNA using thymidine analogs and suggest guidelines for applying this approach.
Collapse
Affiliation(s)
- Anna Ivanova
- Institute of Higher Nervous Activity and Neurophysiology, Russian Academy of Science, Moscow 117485, Russia; (A.I.); (O.G.); (E.E.); (O.A.)
- Institute for Advanced Brain Studies, Lomonosov Moscow State University, Moscow 119991, Russia
| | - Olesya Gruzova
- Institute of Higher Nervous Activity and Neurophysiology, Russian Academy of Science, Moscow 117485, Russia; (A.I.); (O.G.); (E.E.); (O.A.)
| | - Elizaveta Ermolaeva
- Institute of Higher Nervous Activity and Neurophysiology, Russian Academy of Science, Moscow 117485, Russia; (A.I.); (O.G.); (E.E.); (O.A.)
| | - Olga Astakhova
- Institute of Higher Nervous Activity and Neurophysiology, Russian Academy of Science, Moscow 117485, Russia; (A.I.); (O.G.); (E.E.); (O.A.)
- Institute for Advanced Brain Studies, Lomonosov Moscow State University, Moscow 119991, Russia
| | - Sheed Itaman
- Center for Developmental Genetics and Department of Anesthesiology, Stony Brook University, Stony Brook, NY 11794, USA;
- Graduate Program in Neurobiology, Stony Brook University, Stony Brook, NY 11794, USA
| | - Grigori Enikolopov
- Center for Developmental Genetics and Department of Anesthesiology, Stony Brook University, Stony Brook, NY 11794, USA;
- Correspondence: (G.E.); (A.L.)
| | - Alexander Lazutkin
- Institute of Higher Nervous Activity and Neurophysiology, Russian Academy of Science, Moscow 117485, Russia; (A.I.); (O.G.); (E.E.); (O.A.)
- Institute for Advanced Brain Studies, Lomonosov Moscow State University, Moscow 119991, Russia
- Center for Developmental Genetics and Department of Anesthesiology, Stony Brook University, Stony Brook, NY 11794, USA;
- Correspondence: (G.E.); (A.L.)
| |
Collapse
|
26
|
Xia Z, Gao M, Sheng P, Shen M, Zhao L, Gao L, Yan B. Fe 3O 4 Nanozymes Improve Neuroblast Differentiation and Blood-Brain Barrier Integrity of the Hippocampal Dentate Gyrus in D-Galactose-Induced Aged Mice. Int J Mol Sci 2022; 23:ijms23126463. [PMID: 35742908 PMCID: PMC9224281 DOI: 10.3390/ijms23126463] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 06/05/2022] [Accepted: 06/08/2022] [Indexed: 02/01/2023] Open
Abstract
Aging is a process associated with blood-brain barrier (BBB) damage and the reduction in neurogenesis, and is the greatest known risk factor for neurodegenerative disorders. However, the effects of Fe3O4 nanozymes on neurogenesis have rarely been studied. This study examined the effects of Fe3O4 nanozymes on neuronal differentiation in the dentate gyrus (DG) and BBB integrity of D-galactose-induced aged mice. Long-term treatment with Fe3O4 nanozymes (10 μg/mL diluted in ddH2O daily) markedly increased the doublecortin (DCX) immunoreactivity and decreased BBB injury induced by D-galactose treatment. In addition, the decreases in the levels of antioxidant proteins including superoxide dismutase (SOD) and catalase as well as autophagy-related proteins such as Becin-1, LC3II/I, and Atg7 induced by D-galactose treatment were significantly ameliorated by Fe3O4 nanozymes in the DG of the mouse hippocampus. Furthermore, Fe3O4 nanozyme treatment showed an inhibitory effect against apoptosis in the hippocampus. In conclusion, Fe3O4 nanozymes can relieve neuroblast damage and promote neuroblast differentiation in the hippocampal DG by regulating oxidative stress, apoptosis, and autophagy.
Collapse
Affiliation(s)
- Zihao Xia
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Medical College, Yangzhou University, Yangzhou 225001, China; (Z.X.); (M.G.); (P.S.); (M.S.); (L.Z.)
| | - Manman Gao
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Medical College, Yangzhou University, Yangzhou 225001, China; (Z.X.); (M.G.); (P.S.); (M.S.); (L.Z.)
| | - Peng Sheng
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Medical College, Yangzhou University, Yangzhou 225001, China; (Z.X.); (M.G.); (P.S.); (M.S.); (L.Z.)
| | - Mengmeng Shen
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Medical College, Yangzhou University, Yangzhou 225001, China; (Z.X.); (M.G.); (P.S.); (M.S.); (L.Z.)
| | - Lin Zhao
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Medical College, Yangzhou University, Yangzhou 225001, China; (Z.X.); (M.G.); (P.S.); (M.S.); (L.Z.)
| | - Lizeng Gao
- CAS Engineering Laboratory for Nanozyme, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China;
| | - Bingchun Yan
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Medical College, Yangzhou University, Yangzhou 225001, China; (Z.X.); (M.G.); (P.S.); (M.S.); (L.Z.)
- Correspondence: ; Tel.: +86-514-87992215
| |
Collapse
|
27
|
Radial Glia and Neuronal-like Ependymal Cells Are Present within the Spinal Cord of the Trunk (Body) in the Leopard Gecko (Eublepharis macularius). J Dev Biol 2022; 10:jdb10020021. [PMID: 35735912 PMCID: PMC9224675 DOI: 10.3390/jdb10020021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 05/12/2022] [Accepted: 05/20/2022] [Indexed: 11/28/2022] Open
Abstract
As is the case for many lizards, leopard geckos (Eublepharis macularius) can self-detach a portion of their tail to escape predation, and then regenerate a replacement complete with a spinal cord. Previous research has shown that endogenous populations of neural stem/progenitor cells (NSPCs) reside within the spinal cord of the original tail. In response to tail loss, these NSPCs are activated and contribute to regeneration. Here, we investigate whether similar populations of NSPCs are found within the spinal cord of the trunk (body). Using a long-duration 5-bromo-2′-deoxyuridine pulse-chase experiment, we determined that a population of cells within the ependymal layer are label-retaining following a 20-week chase. Tail loss does not significantly alter rates of ependymal cell proliferation within the trunk spinal cord. Ependymal cells of the trunk spinal cord express SOX2 and represent at least two distinct cell populations: radial glial-like (glial fibrillary acidic protein- and Vimentin-expressing) cells; and neuronal-like (HuCD-expressing) cells. Taken together, these data demonstrate that NSPCs of the trunk spinal cord closely resemble those of the tail and support the use of the tail spinal cord as a less invasive proxy for body spinal cord injury investigations.
Collapse
|
28
|
Shen X, Tang C, Kang Q, Zhu Y, Xu S, Jiang J, Xu R. Distribution Changes of Neural Precursor Cells in the Brain Stem of Tg(SOD1*G93A)1Gur Mice. NEURODEGENER DIS 2022; 21:132-145. [PMID: 35584655 DOI: 10.1159/000525124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2021] [Accepted: 04/19/2022] [Indexed: 11/19/2022] Open
Abstract
OBJECTIVES The alteration of vimentin-containing cells (VCCs) proliferation, differentiation and migration in the brain stem of amyotrophic lateral sclerosis (ALS)-like transgenic mice (Tg(SOD1*G93A)1Gur mice) (TG mice) and wild-type mice (WT mice) at the different disease stages of TG mice were studied in this study. The aim of this study was to investigate the change features of proliferation, differentiation and migration of endogenous neural precursor cells (NPCs) and to explore the potential effects of NPCs on restoring degenerated neurons in ALS. METHODS The proliferation, differentiation and migration of VCCs in both different anatomic regions and neural cells of brain stem at the different stages including pre-onset (60-70 days), onset (90-100 days) and progression (120-130 days) stages of TG mice and in WT mice (control) were examined using the immunofluorescence technology. RESULTS VCCs mainly distributed in the around (peripheral) central canal (CC) and the nuclei of brain stem in adult WT mice. VCCs proliferated and differentiated into astrocytes and directionally migrated from the around CC to the nuclei of brain stem, then to the ventral part of damaged regions in brain stem at the pre-onset, onset and progression stages of TG mice. CONCLUSIONS The data suggest that NPCs widely distribute in the brain stem of adult TG mice can differentiate into astrocytes and migrate into damaged brain regions. This response might be a potential mechanism to repair degenerated motor neurons and restore dysfunctional neural circuitry in ALS.
Collapse
Affiliation(s)
- Xiaoping Shen
- Department of Neurology, Jiangxi Provincial People's Hospital, Clinical College of Nanchang Medical College, First Affiliated Hospital of Nanchang Medical College, Nanchang, China
- Department of Neurology, Jiujiang First People's Hospital, Jiujiang, China
- Department of Neurology, Medical College of Nanchang University, Nanchang, China
| | - Chunyan Tang
- Department of Neurology, Jiangxi Provincial People's Hospital, Clinical College of Nanchang Medical College, First Affiliated Hospital of Nanchang Medical College, Nanchang, China
| | - Qin Kang
- Department of Neurology, Jiangxi Provincial People's Hospital, Clinical College of Nanchang Medical College, First Affiliated Hospital of Nanchang Medical College, Nanchang, China
| | - Yu Zhu
- Department of Neurology, Jiangxi Provincial People's Hospital, Clinical College of Nanchang Medical College, First Affiliated Hospital of Nanchang Medical College, Nanchang, China
| | - Shengyuan Xu
- Department of Neurology, Jiangxi Provincial People's Hospital, Clinical College of Nanchang Medical College, First Affiliated Hospital of Nanchang Medical College, Nanchang, China
| | - Jianxiang Jiang
- Department of Neurology, Jiangxi Provincial People's Hospital, Clinical College of Nanchang Medical College, First Affiliated Hospital of Nanchang Medical College, Nanchang, China
| | - Renshi Xu
- Department of Neurology, Jiangxi Provincial People's Hospital, Clinical College of Nanchang Medical College, First Affiliated Hospital of Nanchang Medical College, Nanchang, China
| |
Collapse
|
29
|
Xie WS, Shehzadi K, Ma HL, Liang JH. A Potential Strategy for Treatment of Neurodegenerative Disorders by Regulation of Adult Hippocampal Neurogenesis in Human Brain. Curr Med Chem 2022; 29:5315-5347. [DOI: 10.2174/0929867329666220509114232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 02/13/2022] [Accepted: 03/17/2022] [Indexed: 11/22/2022]
Abstract
Abstract:
Adult hippocampal neurogenesis is a multistage mechanism that continues throughout the lifespan of human and non-human mammals. These adult-born neurons in the central nervous system (CNS) play a significant role in various hippocampus-dependent processes, including learning, mood regulation, pattern recognition, etc. Reduction of adult hippocampal neurogenesis, caused by multiple factors such as neurological disorders and aging, would impair neuronal proliferation and differentiation and result in memory loss. Accumulating studies have indicated that functional neuron impairment could be restored by promoting adult hippocampal neurogenesis. In this review, we summarized the small molecules that could efficiently promote the process of adult neurogenesis, particularly the agents that have the capacity of crossing the blood-brain barrier (BBB), and showed in vivo efficacy in mammalian brains. This may pave the way for the rational design of drugs to treat humnan neurodegenerative disorders in the future.
Collapse
Affiliation(s)
- Wei-Song Xie
- Key Laboratory of Medical Molecule Science and Pharmaceutics Engineering, School of Chemistry and Chemical Engineering, Beijing Institute of Technology, Beijing 102488, China
| | - Kiran Shehzadi
- Key Laboratory of Medical Molecule Science and Pharmaceutics Engineering, School of Chemistry and Chemical Engineering, Beijing Institute of Technology, Beijing 102488, China
| | - Hong-Le Ma
- Key Laboratory of Medical Molecule Science and Pharmaceutics Engineering, School of Chemistry and Chemical Engineering, Beijing Institute of Technology, Beijing 102488, China
| | - Jian-Hua Liang
- Key Laboratory of Medical Molecule Science and Pharmaceutics Engineering, School of Chemistry and Chemical Engineering, Beijing Institute of Technology, Beijing 102488, China
- Yangtze Delta Region Academy of Beijing Institute of Technology, Jiaxing 314019, China
| |
Collapse
|
30
|
Disrupted in schizophrenia 1 regulates ectopic neurogenesis in the mouse hilus after pilocarpine-induced status epilepticus. Neuroscience 2022; 494:69-81. [DOI: 10.1016/j.neuroscience.2022.05.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 05/01/2022] [Accepted: 05/06/2022] [Indexed: 11/20/2022]
|
31
|
Mohammadi F, Bahrami N, Nazariyan M, Mohamadnia A, Hakimiha N, Nazariyan A. Effect of Photobiomodulation Therapy on Differentiation of Mesenchymal Stem Cells Derived from Impacted Third Molar Tooth into Neuron-like Cells. Photochem Photobiol 2022; 98:1434-1440. [PMID: 35363889 DOI: 10.1111/php.13627] [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: 09/28/2021] [Accepted: 03/28/2022] [Indexed: 11/29/2022]
Abstract
Peripheral nerve damages are among the most important consequences of dental and maxillofacial procedures. Tissue engineering using mesenchymal stem cells (MSCs) is a promising method to manage such injuries. Moreover, photobiomodulation therapy (PBMT) can enhance this treatment. The present study aimed to investigate the effect of PBMT on differentiation of MSCs derived from dental follicle (DF) into neurons. MSCs were isolated from an impacted tooth follicle by digestion method. The stem cells were cultured, and differentiated into neurons. The cells received two sessions of PBMT with 810 or 980nm diode laser (100 mW, 4 J/cm2 ) in either DMEM or neural inductive medium . Phenotypic characterization of the cells was determined using Flow cytometry. In addition, β-tubulin and MAP2 genes expression level changes were analyzed using RT-PCR and western blot technique. After 14 days, Flow cytometry analysis confirmed the mesenchymal nature of cells. RT-PCR and western blot affirmed the expression of β-tubulin and MAP2 genes and proteins, respectively. PBMT with both wavelengths significantly increased β-tubulin and MAP2 expression in neural inductive medium with highest expression mean in 980-nm group. PBMT with 810 and 980-nm lasers could be a promising adjunctive method in differentiation of DF-originated MSCs into neural cells.
Collapse
Affiliation(s)
- Farnoush Mohammadi
- Craniomaxillofacial Research center, Tehran University of Medical Sciences, Tehran, Iran.,Oral and Maxillofacial Surgery Department, School of Dentistry, Tehran University of Medical Sciences, Tehran, Iran
| | - Naghmeh Bahrami
- Craniomaxillofacial Research center, Tehran University of Medical Sciences, Tehran, Iran.,Department of Tissue Engineering, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahya Nazariyan
- School of Dentistry, Tehran University of Medical Sciences, Tehran, Iran
| | - Abdolreza Mohamadnia
- Chronic Respiratory Diseases Research Center, National Research Institute of Tuberculosis and Lung Diseases (NRITLD), Shahid Beheshti University of Medical Sciences, Tehran, Iran.,Department of Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Neda Hakimiha
- laser Application in Medical Sciences Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Abolfazl Nazariyan
- Clinical biochemistry Department, Faculty of Medicine, Zanjan University of Medical Sciences, Tehran, Iran
| |
Collapse
|
32
|
Lopez-Lozano AP, Arevalo-Niño K, Gutierrez-Puente Y, Montiel-Hernandez JL, Urrutia-Baca VH, Del Angel-Mosqueda C, De la Garza-Ramos MA. SSEA-4 positive dental pulp stem cells from deciduous teeth and their induction to neural precursor cells. Head Face Med 2022; 18:9. [PMID: 35236383 PMCID: PMC8889676 DOI: 10.1186/s13005-022-00313-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Accepted: 02/17/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Stage-specific embryonic antigen-4 (SSEA-4) is a marker for the identification of multipotent embryonic cells. It is also positive in neuroepithelial cells, precursor neural cells (NPC), and human dental pulp cells. The aim of this study was to evaluate the potential morphodifferentiation and histodifferentiation to NPC of SSEA-4 positive stem cells from human exfoliated deciduous teeth (SHED). METHODS A SHED population in culture, positive to SSEA-4, was obtained by magnetic cell separation. The cells were characterized by immunohistochemistry and flow cytometry. Subsequently, a neurosphere assay was performed in a medium supplemented with basic fibroblast growth factor (bFGF) and epidermal growth factor (EGF); afterward, cells were neurodifferenciated with a neurobasal medium. Finally, indirect immunohistochemistry was performed to identify neuronal markers. RESULTS The morphological and histological changes in the SSEA-4 positive SHEDs were observed after induction with epidermal and fibroblast growth factors in neurobasal culture medium. At the end of induction, the markers Nestin, TuJ-1, and GFAP were identified. CONCLUSIONS The findings show that SSEA-4 positive SHEDs have a behavior similar to neuronal precursor cells. Our findings indicate that the dental pulp of deciduous teeth is a promising source for regeneration therapies associated with neurodegenerative diseases or peripheral nerve alterations.
Collapse
Affiliation(s)
- Ada Pricila Lopez-Lozano
- Facultad de Ciencias Biológicas, Instituto de Biotecnología, Universidad Autónoma de Nuevo León, Nuevo Leon, San Nicolas de los Garza, Mexico.,Unidad de Odontología Integral y Especialidades, Centro de Investigación y Desarrollo en Ciencias de la Salud, Universidad Autonoma de Nuevo Leon, Nuevo Leon, Monterrey, Mexico
| | - Katiushka Arevalo-Niño
- Facultad de Ciencias Biológicas, Instituto de Biotecnología, Universidad Autónoma de Nuevo León, Nuevo Leon, San Nicolas de los Garza, Mexico
| | - Yolanda Gutierrez-Puente
- Facultad de Ciencias Biológicas, Instituto de Biotecnología, Universidad Autónoma de Nuevo León, Nuevo Leon, San Nicolas de los Garza, Mexico
| | - Jose Luis Montiel-Hernandez
- Facultad De Farmacia, Coordinacion De Posgrado, Universidad Autonoma del Estado de Morelos, Morelos, Cuernavaca, Mexico
| | - Victor Hugo Urrutia-Baca
- Unidad de Odontología Integral y Especialidades, Centro de Investigación y Desarrollo en Ciencias de la Salud, Universidad Autonoma de Nuevo Leon, Nuevo Leon, Monterrey, Mexico
| | | | - Myriam Angelica De la Garza-Ramos
- Facultad de Ciencias Biológicas, Instituto de Biotecnología, Universidad Autónoma de Nuevo León, Nuevo Leon, San Nicolas de los Garza, Mexico. .,Unidad de Odontología Integral y Especialidades, Centro de Investigación y Desarrollo en Ciencias de la Salud, Universidad Autonoma de Nuevo Leon, Nuevo Leon, Monterrey, Mexico. .,Facultad de Odontología, Universidad Autonoma de Nuevo Leon, Nuevo Leon, Monterrey, Mexico. .,Facultad de Odontología/CIDICS, Universidad Autonoma de Nuevo Leon Monterrey, San Nicolás de los Garza, Mexico.
| |
Collapse
|
33
|
DeOliveira-Mello L, Vicente I, Gonzalez-Nunez V, Santos-Ledo A, Velasco A, Arévalo R, Lara JM, Mack AF. Doublecortin in the Fish Visual System, a Specific Protein of Maturing Neurons. BIOLOGY 2022; 11:biology11020248. [PMID: 35205114 PMCID: PMC8869232 DOI: 10.3390/biology11020248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/09/2022] [Revised: 01/31/2022] [Accepted: 02/04/2022] [Indexed: 11/30/2022]
Abstract
Simple Summary Doublecortin (DCX) is an essential protein in the development of the central nervous system and in lamination of the mammalian cortex. It is known that the expression of DCX is restricted to newborn neurons. The visual system of teleost fish has been postulated as an ideal model since it continuously grows throughout the animal’s life. Here, we report a comparative expression analysis of DCX between two teleost fish species as well as a bioinformatic analysis with other animal groups. Our results demonstrate that DCX is very useful for identifying new neurons in the visual systems of Astatotilapia burtoni, but is absent in Danio rerio. Abstract Doublecortin (DCX) is a microtubule associated protein, essential for correct central nervous system development and lamination in the mammalian cortex. It has been demonstrated to be expressed in developing—but not in mature—neurons. The teleost visual system is an ideal model to study mechanisms of adult neurogenesis due to its continuous life-long growth. Here, we report immunohistochemical, in silico, and western blot analysis to detect the DCX protein in the visual system of teleost fish. We clearly determined the expression of DCX in newly generated cells in the retina of the cichlid fish Astatotilapia burtoni, but not in the cyprinid fish Danio rerio. Here, we show that DCX is not associated with migrating cells but could be related to axonal growth. This work brings to light the high conservation of DCX sequences between different evolutionary groups, which make it an ideal marker for maturing neurons in various species. The results from different techniques corroborate the absence of DCX expression in zebrafish. In A. burtoni, DCX is very useful for identifying new neurons in the transition zone of the retina. In addition, this marker can be applied to follow axons from maturing neurons through the neural fiber layer, optic nerve head, and optic nerve.
Collapse
Affiliation(s)
- Laura DeOliveira-Mello
- Institute of Neurosciences of Castilla and León, University of Salamanca, 37007 Salamanca, Spain; (V.G.-N.); (A.S.-L.); (A.V.); (R.A.); (J.M.L.)
- Institute for Biomedical Research of Salamanca (IBSAL), University of Salamanca, 37007 Salamanca, Spain
- Correspondence:
| | - Isabel Vicente
- Department of Agriculture, Food and Environment, University of Pisa, 56124 Pisa, Italy;
| | - Veronica Gonzalez-Nunez
- Institute of Neurosciences of Castilla and León, University of Salamanca, 37007 Salamanca, Spain; (V.G.-N.); (A.S.-L.); (A.V.); (R.A.); (J.M.L.)
- Institute for Biomedical Research of Salamanca (IBSAL), University of Salamanca, 37007 Salamanca, Spain
| | - Adrian Santos-Ledo
- Institute of Neurosciences of Castilla and León, University of Salamanca, 37007 Salamanca, Spain; (V.G.-N.); (A.S.-L.); (A.V.); (R.A.); (J.M.L.)
- Institute for Biomedical Research of Salamanca (IBSAL), University of Salamanca, 37007 Salamanca, Spain
| | - Almudena Velasco
- Institute of Neurosciences of Castilla and León, University of Salamanca, 37007 Salamanca, Spain; (V.G.-N.); (A.S.-L.); (A.V.); (R.A.); (J.M.L.)
- Institute for Biomedical Research of Salamanca (IBSAL), University of Salamanca, 37007 Salamanca, Spain
| | - Rosario Arévalo
- Institute of Neurosciences of Castilla and León, University of Salamanca, 37007 Salamanca, Spain; (V.G.-N.); (A.S.-L.); (A.V.); (R.A.); (J.M.L.)
- Institute for Biomedical Research of Salamanca (IBSAL), University of Salamanca, 37007 Salamanca, Spain
| | - Juan M. Lara
- Institute of Neurosciences of Castilla and León, University of Salamanca, 37007 Salamanca, Spain; (V.G.-N.); (A.S.-L.); (A.V.); (R.A.); (J.M.L.)
- Institute for Biomedical Research of Salamanca (IBSAL), University of Salamanca, 37007 Salamanca, Spain
| | - Andreas F. Mack
- Institute of Clinical Anatomy and Cell Analysis, Eberhard-Karls Universität Tübingen, 72074 Tübingen, Germany;
| |
Collapse
|
34
|
Osawa K, Nakanishi Y, Noguchi M, Sugeno A, Goshima Y, Ohshima T. CRMP4 is required for the positioning and maturation of newly generated neurons in adult mouse hippocampus. Neurosci Lett 2022; 773:136503. [PMID: 35122931 DOI: 10.1016/j.neulet.2022.136503] [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: 08/31/2021] [Revised: 12/26/2021] [Accepted: 01/30/2022] [Indexed: 11/28/2022]
Abstract
Adult neurogenesis is a phenomenon in which neural stem cells differentiate and mature to generate new neurons in the adult brain. In mammals, the sites where adult neurogenesis occurs are limited to the subgranular zone (SGZ) of the hippocampal dentate gyrus and the subventricular zone. In the hippocampus, newly generated neurons migrate into the granule cell layer (GCL) and are integrated into neural circuits. Previous studies have revealed that CRMP4, a member of the CRMP family, is expressed in immature neurons in the hippocampal SGZ of the adult brain. However, the role of CRMP4 in adult neurogenesis is unknown. To study the role of CRMP4 in hippocampal adult neurogenesis, we compared adult neurogenesis between wild type and CRMP4-/- mice. In CRMP4-/- mice, the number of doublecortin (DCX)-positive cells was comparable to that in wild-type mice, and some DCX-positive cells were ectopically located in the granule cell layer, suggesting that CRMP4 is involved in the migration of adult neurogenesis. In addition, the number of calretinin-positive new neurons in the SGZ was significantly increased, whereas the number of EdU/NeuN-double positive neurons was decreased in CRMP4-/- mice, suggesting that CRMP4 plays an important role in neuronal maturation. Because CRMP4 is expressed in immature neurons, its expression may regulate the migration from the SGZ to the GCL during neuronal maturation in hippocampal adult neurogenesis.
Collapse
Affiliation(s)
- Koki Osawa
- Department of Life Science and Medical Bio-Science, Waseda University, Shinjuku-ku, Tokyo, 162-8480 Japan
| | - Yurika Nakanishi
- Department of Life Science and Medical Bio-Science, Waseda University, Shinjuku-ku, Tokyo, 162-8480 Japan
| | - Masahito Noguchi
- Department of Life Science and Medical Bio-Science, Waseda University, Shinjuku-ku, Tokyo, 162-8480 Japan
| | - Ayaka Sugeno
- Department of Life Science and Medical Bio-Science, Waseda University, Shinjuku-ku, Tokyo, 162-8480 Japan
| | - Yoshio Goshima
- Department of Molecular Pharmacology and Neurobiology, Yokohama City University Graduate School of Medicine, Yokohama 236-0004 Japan
| | - Toshio Ohshima
- Department of Life Science and Medical Bio-Science, Waseda University, Shinjuku-ku, Tokyo, 162-8480 Japan.
| |
Collapse
|
35
|
Machado-Santos AR, Loureiro-Campos E, Patrício P, Araújo B, Alves ND, Mateus-Pinheiro A, Correia JS, Morais M, Bessa JM, Sousa N, Rodrigues AJ, Oliveira JF, Pinto L. Beyond New Neurons in the Adult Hippocampus: Imipramine Acts as a Pro-Astrogliogenic Factor and Rescues Cognitive Impairments Induced by Stress Exposure. Cells 2022; 11:cells11030390. [PMID: 35159199 PMCID: PMC8834148 DOI: 10.3390/cells11030390] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 01/13/2022] [Accepted: 01/21/2022] [Indexed: 02/06/2023] Open
Abstract
Depression is a prevalent, socially burdensome disease. Different studies have demonstrated the important role of astrocytes in the pathophysiology of depression as modulators of neurotransmission and neurovascular coupling. This is evidenced by astrocyte impairments observed in brains of depressed patients and the appearance of depressive-like behaviors upon astrocytic dysfunctions in animal models. However, little is known about the importance of de novo generated astrocytes in the mammalian brain and in particular its possible involvement in the precipitation of depression and in the therapeutic actions of current antidepressants (ADs). Therefore, we studied the modulation of astrocytes and adult astrogliogenesis in the hippocampal dentate gyrus (DG) of rats exposed to an unpredictable chronic mild stress (uCMS) protocol, untreated and treated for two weeks with antidepressants—fluoxetine and imipramine. Our results show that adult astrogliogenesis in the DG is modulated by stress and imipramine. This study reveals that distinct classes of ADs impact differently in the astrogliogenic process, showing different cellular mechanisms relevant to the recovery from behavioral deficits induced by chronic stress exposure. As such, in addition to those resident, the newborn astrocytes in the hippocampal DG might also be promising therapeutic targets for future therapies in the neuropsychiatric field.
Collapse
Affiliation(s)
- Ana R Machado-Santos
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057 Braga, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Eduardo Loureiro-Campos
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057 Braga, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Patrícia Patrício
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057 Braga, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Bruna Araújo
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057 Braga, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Nuno Dinis Alves
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057 Braga, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - António Mateus-Pinheiro
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057 Braga, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Joana Sofia Correia
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057 Braga, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Mónica Morais
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057 Braga, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - João M Bessa
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057 Braga, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Nuno Sousa
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057 Braga, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Ana J Rodrigues
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057 Braga, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - João Filipe Oliveira
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057 Braga, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
- IPCA-EST-2Ai, Polytechnic Institute of Cávado and Ave, Applied Artificial Intelligence Laboratory, Campus of IPCA, 4750-810 Barcelos, Portugal
| | - Luísa Pinto
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057 Braga, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| |
Collapse
|
36
|
Malloul H, Bonzano S, Bennis M, De Marchis S, Ba-M'hamed S. Chronic thinner inhalation alters olfactory behaviors in adult mice. Behav Brain Res 2022; 417:113597. [PMID: 34563601 DOI: 10.1016/j.bbr.2021.113597] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 09/01/2021] [Accepted: 09/20/2021] [Indexed: 12/17/2022]
Abstract
Volatile solvents exposure can result in various behavioral impairments that have been partly associated to altered adult hippocampal neurogenesis. Despite recent evidence supporting this association, few studies have been devoted to examine the impact on olfactory functioning and olfactory bulb (OB) neurogenesis, although olfactory system is directly in contact with volatile molecules. Thus, this study was designed to evaluate in adult mice the potential modifications of the olfactory functioning after acute (1 day), subchronic (6 weeks) and chronic (12 weeks) exposure to thinner vapor at both behavioral and cellular levels. Firstly, behavioral evaluations showed that chronic thinner exposure impacts on odor detection ability of treated mice but does not affect mice ability to efficiently discriminate between two different odors. Moreover, chronic thinner exposure produces impairment in the olfactory-mediated associative memory. Secondly, analysis of the effects of thinner exposure in the subventricular zone (SVZ) of the lateral ventricle and in the OB revealed that thinner treatments do not induce apoptosis nor glial activation. Thirdly, immunohistochemical quantification of different markers of adult olfactory neurogenesis showed that inhalant treatments do not change the number of proliferating progenitors in the SVZ and the rostral migratory stream (RMS), as well as the number of newborn cells reaching and integrating in the OB circuitry. Altogether, our data highlight that the impaired olfactory performances in chronically-exposed mice are not associated to an alteration of adult neurogenesis in the SVZ-OB system.
Collapse
Affiliation(s)
- Hanaa Malloul
- Laboratory of Pharmacology, Neurobiology, Anthropobiology and Environment, Faculty of Sciences Semlalia, Cadi Ayyad University, Marrakech, Morocco; Department of Life Sciences and Systems Biology, University of Turin, Turin, Italy; Neuroscience Institute Cavalieri Ottolenghi (NICO), Orbassano, Italy
| | - Sara Bonzano
- Department of Life Sciences and Systems Biology, University of Turin, Turin, Italy; Neuroscience Institute Cavalieri Ottolenghi (NICO), Orbassano, Italy
| | - Mohammed Bennis
- Laboratory of Pharmacology, Neurobiology, Anthropobiology and Environment, Faculty of Sciences Semlalia, Cadi Ayyad University, Marrakech, Morocco.
| | - Silvia De Marchis
- Department of Life Sciences and Systems Biology, University of Turin, Turin, Italy; Neuroscience Institute Cavalieri Ottolenghi (NICO), Orbassano, Italy
| | - Saadia Ba-M'hamed
- Laboratory of Pharmacology, Neurobiology, Anthropobiology and Environment, Faculty of Sciences Semlalia, Cadi Ayyad University, Marrakech, Morocco
| |
Collapse
|
37
|
Rudnitskaya EA, Burnyasheva AO, Kozlova TA, Peunov DA, Kolosova NG, Stefanova NA. Changes in Glial Support of the Hippocampus during the Development of an Alzheimer's Disease-like Pathology and Their Correction by Mitochondria-Targeted Antioxidant SkQ1. Int J Mol Sci 2022; 23:ijms23031134. [PMID: 35163053 PMCID: PMC8834695 DOI: 10.3390/ijms23031134] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 01/18/2022] [Accepted: 01/19/2022] [Indexed: 02/06/2023] Open
Abstract
Astrocytes and microglia are the first cells to react to neurodegeneration, e.g., in Alzheimer's disease (AD); however, the data on changes in glial support during the most common (sporadic) type of the disease are sparse. Using senescence-accelerated OXYS rats, which simulate key characteristics of sporadic AD, and Wistar rats (parental normal strain, control), we investigated hippocampal neurogenesis and glial changes during AD-like pathology. Using immunohistochemistry, we showed that the early stage of the pathology is accompanied by a lower intensity of neurogenesis and decreased astrocyte density in the dentate gyrus. The progressive stage is concurrent with reactive astrogliosis and microglia activation, as confirmed by increased cell densities and by the acquisition of cell-specific gene expression profiles, according to transcriptome sequencing data. Besides, here, we continued to analyze the anti-AD effects of prolonged supplementation with mitochondria-targeted antioxidant SkQ1. The antioxidant did not affect neurogenesis, partly normalized the gene expression profile of astrocytes and microglia, and shifted the resting/activated microglia ratio toward a decrease in the activated-cell density. In summary, both astrocytes and microglia are more vulnerable to AD-associated neurodegeneration in the CA3 area than in other hippocampal areas; SkQ1 had an anti-inflammatory effect and is a promising modality for AD prevention and treatment.
Collapse
Affiliation(s)
- Ekaterina A. Rudnitskaya
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences (ICG SB RAS), 10 Lavrentyeva Ave., 630090 Novosibirsk, Russia; (A.O.B.); (T.A.K.); (D.A.P.); (N.G.K.); (N.A.S.)
- Correspondence:
| | - Alena O. Burnyasheva
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences (ICG SB RAS), 10 Lavrentyeva Ave., 630090 Novosibirsk, Russia; (A.O.B.); (T.A.K.); (D.A.P.); (N.G.K.); (N.A.S.)
| | - Tatiana A. Kozlova
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences (ICG SB RAS), 10 Lavrentyeva Ave., 630090 Novosibirsk, Russia; (A.O.B.); (T.A.K.); (D.A.P.); (N.G.K.); (N.A.S.)
| | - Daniil A. Peunov
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences (ICG SB RAS), 10 Lavrentyeva Ave., 630090 Novosibirsk, Russia; (A.O.B.); (T.A.K.); (D.A.P.); (N.G.K.); (N.A.S.)
- Faculty of Natural Sciences, Novosibirsk State University, 2 Pirogova St., 630090 Novosibirsk, Russia
| | - Nataliya G. Kolosova
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences (ICG SB RAS), 10 Lavrentyeva Ave., 630090 Novosibirsk, Russia; (A.O.B.); (T.A.K.); (D.A.P.); (N.G.K.); (N.A.S.)
| | - Natalia A. Stefanova
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences (ICG SB RAS), 10 Lavrentyeva Ave., 630090 Novosibirsk, Russia; (A.O.B.); (T.A.K.); (D.A.P.); (N.G.K.); (N.A.S.)
| |
Collapse
|
38
|
Alonso-Alconada D, Gressens P, Golay X, Robertson NJ. Neurogenesis Is Reduced at 48 h in the Subventricular Zone Independent of Cell Death in a Piglet Model of Perinatal Hypoxia-Ischemia. Front Pediatr 2022; 10:793189. [PMID: 35573964 PMCID: PMC9106110 DOI: 10.3389/fped.2022.793189] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Accepted: 04/04/2022] [Indexed: 11/13/2022] Open
Abstract
Cellular and tissue damage triggered after hypoxia-ischemia (HI) can be generalized and affect the neurogenic niches present in the central nervous system. As neuroregeneration may be critical for optimizing functional recovery in neonatal encephalopathy, the goal of the present work was to investigate the neurogenic response to HI in the neurogenic niche of the subventricular zone (SVZ) in the neonatal piglet. A total of 13 large white male piglets aged <24 h were randomized into two groups: i) HI group (n = 7), animals submitted to transient cerebral HI and resuscitation; and ii) Control group (n = 6), non-HI animals. At 48 h, piglets were euthanized, and the SVZ and its surrounding regions, such as caudate and periventricular white matter, were analyzed for histology using hematoxylin-eosin staining and immunohistochemistry by evaluating the presence of cleaved caspase 3 and TUNEL positive cells, together with the cell proliferation/neurogenesis markers Ki67 (cell proliferation), GFAP (neural stem cells processes), Sox2 (neural stem/progenitor cells), and doublecortin (DCX, a marker of immature migrating neuroblasts). Hypoxic-ischemic piglets showed a decrease in cellularity in the SVZ independent of cell death, together with decreased length of neural stem cells processes, neuroblast chains area, DCX immunoreactivity, and lower number of Ki67 + and Ki67 + Sox2 + cells. These data suggest a reduction in both cell proliferation and neurogenesis in the SVZ of the neonatal piglet, which could in turn compromise the replacement of the lost neurons and the achievement of global repair.
Collapse
Affiliation(s)
- Daniel Alonso-Alconada
- Department of Cell Biology and Histology, School of Medicine and Nursing, University of the Basque Country (UPV/EHU), Leioa, Spain
| | | | - Xavier Golay
- Department of Brain Repair and Rehabilitation, Institute of Neurology, University College London, London, United Kingdom
| | - Nicola J Robertson
- Institute for Women's Health, University College London, London, United Kingdom.,Edinburgh Neuroscience, Centre for Clinical Brain Sciences, The University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
39
|
Blankers SA, Galea LA. Androgens and Adult Neurogenesis in the Hippocampus. ANDROGENS: CLINICAL RESEARCH AND THERAPEUTICS 2021; 2:203-215. [PMID: 35024692 PMCID: PMC8744005 DOI: 10.1089/andro.2021.0016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Accepted: 08/19/2021] [Indexed: 11/12/2022]
Abstract
Adult neurogenesis in the hippocampus is modulated by steroid hormones, including androgens, in male rodents. In this review, we summarize research showing that chronic exposure to androgens, such as testosterone and dihydrotestosterone, enhances the survival of new neurons in the dentate gyrus of male, but not female, rodents, via the androgen receptor. However, the neurogenesis promoting the effect of androgens in the dentate gyrus may be limited to younger adulthood as it is not evident in middle-aged male rodents. Although direct exposure to androgens in adult or middle age does not significantly influence neurogenesis in female rodents, the aromatase inhibitor letrozole enhances neurogenesis in the hippocampus of middle-aged female mice. Unlike other androgens, androgenic anabolic steroids reduce neurogenesis in the hippocampus of male rodents. Collectively, the research indicates that the ability of androgens to enhance hippocampal neurogenesis in adult rodents is dependent on dose, androgen type, sex, duration, and age. We discuss these findings and how androgens may be influencing neuroprotection, via neurogenesis in the hippocampus, in the context of health and disease.
Collapse
Affiliation(s)
- Samantha A. Blankers
- Graduate Program in Neuroscience, The University of British Columbia, Vancouver, Canada
- Djavad Mowafaghian Centre for Brain Health, The University of British Columbia, Vancouver, Canada
| | - Liisa A.M. Galea
- Graduate Program in Neuroscience, The University of British Columbia, Vancouver, Canada
- Djavad Mowafaghian Centre for Brain Health, The University of British Columbia, Vancouver, Canada
- Department of Psychology, The University of British Columbia, Vancouver, Canada
| |
Collapse
|
40
|
Kashirina A, Gavrina A, Kryukov E, Elagin V, Kolesova Y, Artyuhov A, Momotyuk E, Abdyyev V, Meshcheryakova N, Zagaynova E, Dashinimaev E, Kashina A. Energy Metabolism and Intracellular pH Alteration in Neural Spheroids Carrying Down Syndrome. Biomedicines 2021; 9:1741. [PMID: 34829971 PMCID: PMC8615730 DOI: 10.3390/biomedicines9111741] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 11/17/2021] [Accepted: 11/18/2021] [Indexed: 12/18/2022] Open
Abstract
Brain diseases including Down syndrome (DS/TS21) are known to be characterized by changes in cellular metabolism. To adequately assess such metabolic changes during pathological processes and to test drugs, methods are needed that allow monitoring of these changes in real time with minimally invasive effects. Thus, the aim of our work was to study the metabolic status and intracellular pH of spheroids carrying DS using fluorescence microscopy and FLIM. For metabolic analysis we measured the fluorescence intensities, fluorescence lifetimes and the contributions of the free and bound forms of NAD(P)H. For intracellular pH assay we measured the fluorescence intensities of SypHer-2 and BCECF. Data were processed with SPCImage and Fiji-ImageJ. We demonstrated the predominance of glycolysis in TS21 spheroids compared with normal karyotype (NK) spheroids. Assessment of the intracellular pH indicated a more alkaline intracellular pH in the TS21 spheroids compared to NK spheroids. Using fluorescence imaging, we performed a comprehensive comparative analysis of the metabolism and intracellular pH of TS21 spheroids and showed that fluorescence microscopy and FLIM make it possible to study living cells in 3D models in real time with minimally invasive effects.
Collapse
Affiliation(s)
- Alena Kashirina
- Institute of Experimental Oncology and Biomedical Technologies, Privolzhsky Research Medical University, 603005 Nizhny Novgorod, Russia; (A.G.); (E.K.); (V.E.); (E.Z.); (A.K.)
| | - Alena Gavrina
- Institute of Experimental Oncology and Biomedical Technologies, Privolzhsky Research Medical University, 603005 Nizhny Novgorod, Russia; (A.G.); (E.K.); (V.E.); (E.Z.); (A.K.)
| | - Emil Kryukov
- Institute of Experimental Oncology and Biomedical Technologies, Privolzhsky Research Medical University, 603005 Nizhny Novgorod, Russia; (A.G.); (E.K.); (V.E.); (E.Z.); (A.K.)
| | - Vadim Elagin
- Institute of Experimental Oncology and Biomedical Technologies, Privolzhsky Research Medical University, 603005 Nizhny Novgorod, Russia; (A.G.); (E.K.); (V.E.); (E.Z.); (A.K.)
| | - Yuliya Kolesova
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, 119991 Moscow, Russia;
- Koltzov Institute of Developmental Biology, Russian Academy of Sciences, 119334 Moscow, Russia; (V.A.); (E.D.)
| | - Alexander Artyuhov
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, Ostrovitianov Street, 117997 Moscow, Russia; (A.A.); (E.M.); (N.M.)
| | - Ekaterina Momotyuk
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, Ostrovitianov Street, 117997 Moscow, Russia; (A.A.); (E.M.); (N.M.)
| | - Vepa Abdyyev
- Koltzov Institute of Developmental Biology, Russian Academy of Sciences, 119334 Moscow, Russia; (V.A.); (E.D.)
| | - Natalia Meshcheryakova
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, Ostrovitianov Street, 117997 Moscow, Russia; (A.A.); (E.M.); (N.M.)
| | - Elena Zagaynova
- Institute of Experimental Oncology and Biomedical Technologies, Privolzhsky Research Medical University, 603005 Nizhny Novgorod, Russia; (A.G.); (E.K.); (V.E.); (E.Z.); (A.K.)
- Institute of Biology and Biomedicine, Lobachevsky State University of Nizhni Novgorod, 603022 Nizhny Novgorod, Russia
| | - Erdem Dashinimaev
- Koltzov Institute of Developmental Biology, Russian Academy of Sciences, 119334 Moscow, Russia; (V.A.); (E.D.)
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, Ostrovitianov Street, 117997 Moscow, Russia; (A.A.); (E.M.); (N.M.)
- Moscow Institute of Physics and Technology, 141701 Dolgoprudny, Russia
| | - Aleksandra Kashina
- Institute of Experimental Oncology and Biomedical Technologies, Privolzhsky Research Medical University, 603005 Nizhny Novgorod, Russia; (A.G.); (E.K.); (V.E.); (E.Z.); (A.K.)
| |
Collapse
|
41
|
Rieskamp JD, Sarchet P, Smith BM, Kirby ED. Estimation of the density of neural, glial, and endothelial lineage cells in the adult mouse dentate gyrus. Neural Regen Res 2021; 17:1286-1292. [PMID: 34782573 PMCID: PMC8643033 DOI: 10.4103/1673-5374.327354] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
The dentate gyrus subregion of the mammalian hippocampus is an adult neural stem cell niche and site of lifelong neurogenesis. Hypotheses regarding the role of adult-born neuron synaptic integration in hippocampal circuit function are framed by robust estimations of adult-born versus pre/perinatally-born neuron number. In contrast, the non-neurogenic functions of adult neural stem cells and their immediate progeny, such as secretion of bioactive growth factors and expression of extracellular matrix-modifying proteins, lack similar framing due to few estimates of their number versus other prominent secretory cells. Here, we apply immunohistochemical methods to estimate cell density of neural stem/progenitor cells versus other major classes of glial and endothelial cell types that are potentially secretory in the dentate gyrus of adult mice. Of the cell types quantified, we found that GFAP+SOX2+ stellate astrocytes were the most numerous, followed by CD31+ endothelia, GFAP–SOX2+ intermediate progenitors, Olig2+ oligodendrocytes, Iba1+ microglia, and GFAP+SOX2+ radial glia-like neural stem cells. We did not observe any significant sex differences in density of any cell population. Notably, neural stem/progenitor cells were present at a similar density as several cell types known to have potent functional roles via their secretome. These findings may be useful for refining hypotheses regarding the contributions of these cell types to regulating hippocampal function and their potential therapeutic uses. All experimental protocols were approved by the Ohio State University Institutional Animal Care and Use Committee (protocol# 2016A00000068) on July 14, 2016.
Collapse
Affiliation(s)
- Joshua D Rieskamp
- Neuroscience Graduate Program; Department of Psychology, The Ohio State University, Columbus, OH, USA
| | - Patricia Sarchet
- Department of Psychology, The Ohio State University, Columbus, OH, USA
| | - Bryon M Smith
- Department of Psychology, The Ohio State University, Columbus, OH, USA
| | - Elizabeth D Kirby
- Department of Psychology; Department of Neuroscience; Chronic Brain Injury Program, The Ohio State University, Columbus, OH, USA
| |
Collapse
|
42
|
Short high fat diet triggers reversible and region specific effects in DCX + hippocampal immature neurons of adolescent male mice. Sci Rep 2021; 11:21499. [PMID: 34728755 PMCID: PMC8563989 DOI: 10.1038/s41598-021-01059-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Accepted: 10/18/2021] [Indexed: 11/26/2022] Open
Abstract
Adolescence represents a crucial period for maturation of brain structures involved in cognition. Early in life unhealthy dietary patterns are associated with inferior cognitive outcomes at later ages; conversely, healthy diet is associated with better cognitive results. In this study we analyzed the effects of a short period of hypercaloric diet on newborn hippocampal doublecortin+ (DCX) immature neurons in adolescent mice. Male mice received high fat diet (HFD) or control low fat diet (LFD) from the 5th week of age for 1 or 2 weeks, or 1 week HFD followed by 1 week LFD. After diet supply, mice were either perfused for immunohistochemical (IHC) analysis or their hippocampi were dissected for biochemical assays. Detailed morphometric analysis was performed in DCX+ cells that displayed features of immature neurons. We report that 1 week-HFD was sufficient to dramatically reduce dendritic tree complexity of DCX+ cells. This effect occurred specifically in dorsal and not ventral hippocampus and correlated with reduced BDNF expression levels in dorsal hippocampus. Both structural and biochemical changes were reversed by a return to LFD. Altogether these studies increase our current knowledge on potential consequences of hypercaloric diet on brain and in particular on dorsal hippocampal neuroplasticity.
Collapse
|
43
|
Martins-Macedo J, Salgado AJ, Gomes ED, Pinto L. Adult brain cytogenesis in the context of mood disorders: From neurogenesis to the emergent role of gliogenesis. Neurosci Biobehav Rev 2021; 131:411-428. [PMID: 34555383 DOI: 10.1016/j.neubiorev.2021.09.030] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Revised: 09/06/2021] [Accepted: 09/16/2021] [Indexed: 12/18/2022]
Abstract
Psychiatric disorders severely impact patients' lives. Motivational, cognitive and emotional deficits are the most common symptoms observed in these patients and no effective treatment is still available, either due to the adverse side effects or the low rate of efficacy of currently available drugs. Neurogenesis recovery has been one important focus in the treatment of psychiatric disorders, which undeniably contributes to the therapeutic action of antidepressants. However, glial plasticity is emerging as a new strategy to explore the deficits observed in mood disorders and the efficacy of therapeutic interventions. Thus, it is crucial to understand the mechanisms behind glio- and neurogenesis to better define treatments and preventive therapies, once adult cytogenesis is of pivotal importance to cognitive and emotional components of behavior, both in healthy and pathological contexts, including in psychiatric disorders. Here, we review the concepts and history of neuro- and gliogenesis, providing as well a reflection on the functional importance of cytogenesis in the context of disease.
Collapse
Affiliation(s)
- Joana Martins-Macedo
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal.
| | - António J Salgado
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal.
| | - Eduardo D Gomes
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal.
| | - Luísa Pinto
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal.
| |
Collapse
|
44
|
Macht V, Vetreno R, Elchert N, Crews F. Galantamine prevents and reverses neuroimmune induction and loss of adult hippocampal neurogenesis following adolescent alcohol exposure. J Neuroinflammation 2021; 18:212. [PMID: 34530858 PMCID: PMC8447570 DOI: 10.1186/s12974-021-02243-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 08/18/2021] [Indexed: 01/17/2023] Open
Abstract
BACKGROUND Binge ethanol exposure during adolescence reduces hippocampal neurogenesis, a reduction which persists throughout adulthood despite abstinence. This loss of neurogenesis, indicated by reduced doublecortin+ immunoreactivity (DCX+IR), is paralleled by an increase in hippocampal proinflammatory signaling cascades. As galantamine, a cholinesterase inhibitor, has anti-inflammatory actions, we tested the hypothesis that galantamine would prevent (study 1) or restore (study 2) AIE induction of proinflammatory signals within the hippocampus as well as AIE-induced loss of hippocampal neurogenesis. METHODS Galantamine (4 mg/kg) or vehicle (saline) was administered to Wistar rats during adolescent intermittent ethanol (AIE; 5.0 g/kg ethanol, 2 days on/2 days off, postnatal day [P] 25-54) (study 1, prevention) or after AIE during abstinent maturation to adulthood (study 2, restoration). RESULTS Results indicate AIE reduced DCX+IR and induced cleaved caspase3 (Casp3) in DCX-expressing immature neurons. Excitingly, AIE induction of activated Casp3 in DCX-expressing neurons is both prevented and reversed by galantamine treatment, which also resulted in prevention and restoration of neurogenesis (DCX+IR). Similarly, galantamine prevented and/or reversed AIE induction of proinflammatory markers, including the chemokine (C-C motif) ligand 2 (CCL2), cyclooxygenase-2 (COX-2), and high mobility group box 1 (HMGB1) protein, suggesting that AIE induction of proinflammatory signaling mediates both cell death cascades and hippocampal neurogenesis. Interestingly, galantamine treatment increased Ki67+IR generally as well as increased pan-Trk expression specifically in AIE-treated rats but failed to reverse AIE induction of NADPH-oxidase (gp91phox). CONCLUSIONS Collectively, our studies suggest that (1) loss of neurogenesis after AIE is mediated by persistent induction of proinflammatory cascades which drive activation of cell death machinery in immature neurons, and (2) galantamine can prevent and restore AIE disruptions in the hippocampal environmental milieu to then prevent and restore AIE-mediated loss of neurogenesis.
Collapse
Affiliation(s)
- Victoria Macht
- Bowles Center for Alcohol Studies, School of Medicine, University of North Carolina at Chapel Hill, 104 Manning Drive, Chapel Hill, NC, 27599, USA.
| | - Ryan Vetreno
- Bowles Center for Alcohol Studies, School of Medicine, University of North Carolina at Chapel Hill, 104 Manning Drive, Chapel Hill, NC, 27599, USA
- Department of Psychiatry, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Natalie Elchert
- Bowles Center for Alcohol Studies, School of Medicine, University of North Carolina at Chapel Hill, 104 Manning Drive, Chapel Hill, NC, 27599, USA
| | - Fulton Crews
- Bowles Center for Alcohol Studies, School of Medicine, University of North Carolina at Chapel Hill, 104 Manning Drive, Chapel Hill, NC, 27599, USA
- Department of Psychiatry, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Pharmacology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| |
Collapse
|
45
|
Olson CA, Iñiguez AJ, Yang GE, Fang P, Pronovost GN, Jameson KG, Rendon TK, Paramo J, Barlow JT, Ismagilov RF, Hsiao EY. Alterations in the gut microbiota contribute to cognitive impairment induced by the ketogenic diet and hypoxia. Cell Host Microbe 2021; 29:1378-1392.e6. [PMID: 34358434 PMCID: PMC8429275 DOI: 10.1016/j.chom.2021.07.004] [Citation(s) in RCA: 57] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 05/17/2021] [Accepted: 07/12/2021] [Indexed: 01/16/2023]
Abstract
Many genetic and environmental factors increase susceptibility to cognitive impairment (CI), and the gut microbiome is increasingly implicated. However, the identity of gut microbes associated with CI risk, their effects on CI, and their mechanisms remain unclear. Here, we show that a carbohydrate-restricted (ketogenic) diet potentiates CI induced by intermittent hypoxia in mice and alters the gut microbiota. Depleting the microbiome reduces CI, whereas transplantation of the risk-associated microbiome or monocolonization with Bilophila wadsworthia confers CI in mice fed a standard diet. B. wadsworthia and the risk-associated microbiome disrupt hippocampal synaptic plasticity, neurogenesis, and gene expression. The CI is associated with microbiome-dependent increases in intestinal interferon-gamma (IFNg)-producing Th1 cells. Inhibiting Th1 cell development abrogates the adverse effects of both B. wadsworthia and environmental risk factors on CI. Together, these findings identify select gut bacteria that contribute to environmental risk for CI in mice by promoting inflammation and hippocampal dysfunction.
Collapse
Affiliation(s)
- Christine A. Olson
- Department of Integrative Biology & Physiology, University of California, Los Angeles, Los Angeles, CA 90095, USA,Correspondence to: ,
| | - Alonso J. Iñiguez
- Department of Integrative Biology & Physiology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Grace E. Yang
- Department of Integrative Biology & Physiology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Ping Fang
- Department of Integrative Biology & Physiology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Geoffrey N. Pronovost
- Department of Integrative Biology & Physiology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Kelly G. Jameson
- Department of Integrative Biology & Physiology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Tomiko K. Rendon
- Department of Integrative Biology & Physiology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Jorge Paramo
- Department of Integrative Biology & Physiology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Jacob T. Barlow
- Division of Chemistry & Chemical Engineering, California Institute of Technology, Pasadena, CA 91108, USA
| | - Rustem F. Ismagilov
- Division of Chemistry & Chemical Engineering, California Institute of Technology, Pasadena, CA 91108, USA
| | - Elaine Y. Hsiao
- Department of Integrative Biology & Physiology, University of California, Los Angeles, Los Angeles, CA 90095, USA,Correspondence to: ,
| |
Collapse
|
46
|
Cattani D, Struyf N, Steffensen V, Bergquist J, Zamoner A, Brittebo E, Andersson M. Perinatal exposure to a glyphosate-based herbicide causes dysregulation of dynorphins and an increase of neural precursor cells in the brain of adult male rats. Toxicology 2021; 461:152922. [PMID: 34474092 DOI: 10.1016/j.tox.2021.152922] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 08/05/2021] [Accepted: 08/27/2021] [Indexed: 01/01/2023]
Abstract
Glyphosate, the most used herbicide worldwide, has been suggested to induce neurotoxicity and behavioral changes in rats after developmental exposure. Studies of human glyphosate intoxication have reported adverse effects on the nervous system, particularly in substantia nigra (SN). Here we used matrix-assisted laser desorption ionization (MALDI) imaging mass spectrometry (IMS) to study persistent changes in peptide expression in the SN of 90-day-old adult male Wistar rats. The animals were perinatally exposed to 3 % GBH (glyphosate-based herbicide) in drinking water (corresponding to 0.36 % of glyphosate) starting at gestational day 5 and continued up to postnatal day 15 (PND15). Peptides are present in the central nervous system before birth and play a critical role in the development and survival of neurons, therefore, observed neuropeptide changes could provide better understanding of the GBH-induced long term effects on SN. The results revealed 188 significantly altered mass peaks in SN of animals perinatally exposed to GBH. A significant reduction of the peak intensity (P < 0.05) of several peptides from the opioid-related dynorphin family such as dynorphin B (57 %), alpha-neoendorphin (50 %), and its endogenous metabolite des-tyrosine alpha-neoendorphin (39 %) was detected in the GBH group. Immunohistochemical analysis confirmed a decreased dynorphin expression and showed a reduction of the total area of dynorphin immunoreactive fibers in the SN of the GBH group. In addition, a small reduction of dynorphin immunoreactivity associated with non-neuronal cells was seen in the hilus of the hippocampal dentate gyrus. Perinatal exposure to GBH also induced an increase in the number of nestin-positive cells in the subgranular zone of the dentate gyrus. In conclusion, the results demonstrate long-term changes in the adult male rat SN and hippocampus following a perinatal GBH exposure suggesting that this glyphosate-based formulation may perturb critical neurodevelopmental processes.
Collapse
Affiliation(s)
- Daiane Cattani
- Department of Pharmaceutical Biosciences - BMC, Uppsala University, Box 591, 75124, Uppsala, Sweden; Department of Biochemistry, Federal University of Santa Catarina, Florianopolis, 88040-970, Brazil.
| | - Nona Struyf
- Department of Pharmaceutical Biosciences - BMC, Uppsala University, Box 591, 75124, Uppsala, Sweden
| | - Vivien Steffensen
- Department of Pharmaceutical Biosciences - BMC, Uppsala University, Box 591, 75124, Uppsala, Sweden
| | - Jonas Bergquist
- Department of Chemistry - BMC, Analytical Chemistry and Neurochemistry, Uppsala University, Box 559, 75124, Uppsala, Sweden
| | - Ariane Zamoner
- Department of Biochemistry, Federal University of Santa Catarina, Florianopolis, 88040-970, Brazil
| | - Eva Brittebo
- Department of Pharmaceutical Biosciences - BMC, Uppsala University, Box 591, 75124, Uppsala, Sweden
| | - Malin Andersson
- Department of Pharmaceutical Biosciences - BMC, Uppsala University, Box 591, 75124, Uppsala, Sweden
| |
Collapse
|
47
|
Gustorff C, Scheuer T, Schmitz T, Bührer C, Endesfelder S. GABA B Receptor-Mediated Impairment of Intermediate Progenitor Maturation During Postnatal Hippocampal Neurogenesis of Newborn Rats. Front Cell Neurosci 2021; 15:651072. [PMID: 34421540 PMCID: PMC8377254 DOI: 10.3389/fncel.2021.651072] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Accepted: 07/12/2021] [Indexed: 12/19/2022] Open
Abstract
The neurotransmitter GABA and its receptors assume essential functions during fetal and postnatal brain development. The last trimester of a human pregnancy and early postnatal life involves a vulnerable period of brain development. In the second half of gestation, there is a developmental shift from depolarizing to hyperpolarizing in the GABAergic system, which might be disturbed by preterm birth. Alterations of the postnatal GABA shift are associated with several neurodevelopmental disorders. In this in vivo study, we investigated neurogenesis in the dentate gyrus (DG) in response to daily administration of pharmacological GABAA (DMCM) and GABAB (CGP 35348) receptor inhibitors to newborn rats. Six-day-old Wistar rats (P6) were daily injected (i.p.) to postnatal day 11 (P11) with DMCM, CGP 35348, or vehicle to determine the effects of both antagonists on postnatal neurogenesis. Due to GABAB receptor blockade by CGP 35348, immunohistochemistry revealed a decrease in the number of NeuroD1 positive intermediate progenitor cells and a reduction of proliferative Nestin-positive neuronal stem cells at the DG. The impairment of hippocampal neurogenesis at this stage of differentiation is in line with a significantly decreased RNA expression of the transcription factors Pax6, Ascl1, and NeuroD1. Interestingly, the number of NeuN-positive postmitotic neurons was not affected by GABAB receptor blockade, although strictly associated transcription factors for postmitotic neurons, Tbr1, Prox1, and NeuroD2, displayed reduced expression levels, suggesting impairment by GABAB receptor antagonization at this stage of neurogenesis. Antagonization of GABAB receptors decreased the expression of neurotrophins (BDNF, NT-3, and NGF). In contrast to the GABAB receptor blockade, the GABAA receptor antagonization revealed no significant changes in cell counts, but an increased transcriptional expression of Tbr1 and Tbr2. We conclude that GABAergic signaling via the metabotropic GABAB receptor is crucial for hippocampal neurogenesis at the time of rapid brain growth and of the postnatal GABA shift. Differentiation and proliferation of intermediate progenitor cells are dependent on GABA. These insights become more pertinent in preterm infants whose developing brains are prematurely exposed to spostnatal stress and predisposed to poor neurodevelopmental disorders, possibly as sequelae of early disruption in GABAergic signaling.
Collapse
Affiliation(s)
- Charlotte Gustorff
- Department of Neonatology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Till Scheuer
- Department of Neonatology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Thomas Schmitz
- Department of Neonatology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Christoph Bührer
- Department of Neonatology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | | |
Collapse
|
48
|
Defteralı Ç, Moreno-Estellés M, Crespo C, Díaz-Guerra E, Díaz-Moreno M, Vergaño-Vera E, Nieto-Estévez V, Hurtado-Chong A, Consiglio A, Mira H, Vicario C. Neural stem cells in the adult olfactory bulb core generate mature neurons in vivo. Stem Cells 2021; 39:1253-1269. [PMID: 33963799 DOI: 10.1002/stem.3393] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Accepted: 04/20/2021] [Indexed: 01/05/2023]
Abstract
Although previous studies suggest that neural stem cells (NSCs) exist in the adult olfactory bulb (OB), their location, identity, and capacity to generate mature neurons in vivo has been little explored. Here, we injected enhanced green fluorescent protein (EGFP)-expressing retroviral particles into the OB core of adult mice to label dividing cells and to track the differentiation/maturation of any neurons they might generate. EGFP-labeled cells initially expressed adult NSC markers on days 1 to 3 postinjection (dpi), including Nestin, GLAST, Sox2, Prominin-1, and GFAP. EGFP+ -doublecortin (DCX) cells with a migratory morphology were also detected and their abundance increased over a 7-day period. Furthermore, EGFP-labeled cells progressively became NeuN+ neurons, they acquired neuronal morphologies, and they became immunoreactive for OB neuron subtype markers, the most abundant representing calretinin expressing interneurons. OB-NSCs also generated glial cells, suggesting they could be multipotent in vivo. Significantly, the newly generated neurons established and received synaptic contacts, and they expressed presynaptic proteins and the transcription factor pCREB. By contrast, when the retroviral particles were injected into the subventricular zone (SVZ), nearly all (98%) EGFP+ -cells were postmitotic when they reached the OB core, implying that the vast majority of proliferating cells present in the OB are not derived from the SVZ. Furthermore, we detected slowly dividing label-retaining cells in this region that could correspond to the population of resident NSCs. This is the first time NSCs located in the adult OB core have been shown to generate neurons that incorporate into OB circuits in vivo.
Collapse
Affiliation(s)
- Çağla Defteralı
- Instituto Cajal-Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain.,CIBERNED-Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Mireia Moreno-Estellés
- Unidad de Neurobiología Molecular, Área de Biología Celular y del Desarrollo, CNM-ISCIII, Majadahonda, Spain.,Instituto de Biomedicina de Valencia-CSIC (IBV-CSIC), Valencia, Spain
| | - Carlos Crespo
- Departamento de Biología Celular, Estructura de Investigación Interdisciplinar en Biotecnología y Biomedicina (BIOTECMED), Universitat de Valencia, Valencia, Spain
| | - Eva Díaz-Guerra
- Instituto Cajal-Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain.,CIBERNED-Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - María Díaz-Moreno
- Unidad de Neurobiología Molecular, Área de Biología Celular y del Desarrollo, CNM-ISCIII, Majadahonda, Spain
| | - Eva Vergaño-Vera
- Instituto Cajal-Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain.,CIBERNED-Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Vanesa Nieto-Estévez
- Instituto Cajal-Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain.,CIBERNED-Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Anahí Hurtado-Chong
- Instituto Cajal-Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain.,CIBERNED-Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Antonella Consiglio
- Institute of Biomedicine, Department of Pathology and Experimental Therapeutics, Bellvitge University Hospital-IDIBELL, Barcelona, Spain.,Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Helena Mira
- Unidad de Neurobiología Molecular, Área de Biología Celular y del Desarrollo, CNM-ISCIII, Majadahonda, Spain.,Instituto de Biomedicina de Valencia-CSIC (IBV-CSIC), Valencia, Spain
| | - Carlos Vicario
- Instituto Cajal-Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain.,CIBERNED-Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| |
Collapse
|
49
|
Kim DS, Weber T, Straube U, Hellweg CE, Nasser M, Green DA, Fogtman A. The Potential of Physical Exercise to Mitigate Radiation Damage-A Systematic Review. Front Med (Lausanne) 2021; 8:585483. [PMID: 33996841 PMCID: PMC8117229 DOI: 10.3389/fmed.2021.585483] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Accepted: 03/10/2021] [Indexed: 12/12/2022] Open
Abstract
There is a need to investigate new countermeasures against the detrimental effects of ionizing radiation as deep space exploration missions are on the horizon. Objective: In this systematic review, the effects of physical exercise upon ionizing radiation-induced damage were evaluated. Methods: Systematic searches were performed in Medline, Embase, Cochrane library, and the databases from space agencies. Of 2,798 publications that were screened, 22 studies contained relevant data that were further extracted and analyzed. Risk of bias of included studies was assessed. Due to the high level of heterogeneity, meta-analysis was not performed. Five outcome groups were assessed by calculating Hedges' g effect sizes and visualized using effect size plots. Results: Exercise decreased radiation-induced DNA damage, oxidative stress, and inflammation, while increasing antioxidant activity. Although the results were highly heterogeneous, there was evidence for a beneficial effect of exercise in cellular, clinical, and functional outcomes. Conclusions: Out of 72 outcomes, 68 showed a beneficial effect of physical training when exposed to ionizing radiation. As the first study to investigate a potential protective mechanism of physical exercise against radiation effects in a systematic review, the current findings may help inform medical capabilities of human spaceflight and may also be relevant for terrestrial clinical care such as radiation oncology.
Collapse
Affiliation(s)
- David S. Kim
- Space Medicine Team (HRE-OM), European Astronaut Centre, European Space Agency, Cologne, Germany
- Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Tobias Weber
- Space Medicine Team (HRE-OM), European Astronaut Centre, European Space Agency, Cologne, Germany
- KBR GmbH, Cologne, Germany
| | - Ulrich Straube
- Space Medicine Team (HRE-OM), European Astronaut Centre, European Space Agency, Cologne, Germany
| | - Christine E. Hellweg
- Radiation Biology Department, Institute of Aerospace Medicine, German Aerospace Centre (DLR), Cologne, Germany
| | - Mona Nasser
- Peninsula Dental School, Plymouth University, Plymouth, United Kingdom
| | - David A. Green
- Space Medicine Team (HRE-OM), European Astronaut Centre, European Space Agency, Cologne, Germany
- KBR GmbH, Cologne, Germany
- Centre of Human & Applied Physiological Sciences (CHAPS), King's College London, London, United Kingdom
| | - Anna Fogtman
- Space Medicine Team (HRE-OM), European Astronaut Centre, European Space Agency, Cologne, Germany
| |
Collapse
|
50
|
van Groen T, Kadish I, Popović N, Caballero Bleda M, Baño-Otalora B, Rol MA, Madrid JA, Popović M. Widespread Doublecortin Expression in the Cerebral Cortex of the Octodon degus. Front Neuroanat 2021; 15:656882. [PMID: 33994960 PMCID: PMC8116662 DOI: 10.3389/fnana.2021.656882] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Accepted: 04/06/2021] [Indexed: 11/13/2022] Open
Abstract
It has been demonstrated that in adulthood rodents show newly born neurons in the subgranular layer (SGL) of the dentate gyrus (DG), and in the subventricular zone (SVZ). The neurons generated in the SVZ migrate through the rostral migratory stream (RMS) to the olfactory bulb. One of the markers of newly generated neurons is doublecortin (DCX). The degu similarly shows significant numbers of DCX-labeled neurons in the SGL, SVZ, and RMS. Further, most of the nuclei of these DCX-expressing neurons are also labeled by proliferating nuclear antigen (PCNA) and Ki67. Finally, whereas in rats and mice DCX-labeled neurons are predominantly present in the SGL and SVZ, with only a few DCX neurons present in piriform cortex, the degu also shows significant numbers of DCX expressing neurons in areas outside of SVZ, DG, and PC. Many areas of neocortex in degu demonstrate DCX-labeled neurons in layer II, and most of these neurons are found in the limbic cortices. The DCX-labeled cells do not stain with NeuN, indicating they are immature neurons.
Collapse
Affiliation(s)
- Thomas van Groen
- Department of Cell Biology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Inga Kadish
- Department of Cell Biology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Natalija Popović
- Department of Human Anatomy and Psychobiology, Faculty of Medicine, University of Murcia, Murcia, Spain.,Institute of Biomedical Research of Murcia, Virgen de la Arrixaca University Hospital, University of Murcia, Murcia, Spain.,CEIR Campus Mare Nostrum (CMN), University of Murcia, Murcia, Spain
| | - María Caballero Bleda
- Department of Human Anatomy and Psychobiology, Faculty of Medicine, University of Murcia, Murcia, Spain.,Institute of Biomedical Research of Murcia, Virgen de la Arrixaca University Hospital, University of Murcia, Murcia, Spain.,CEIR Campus Mare Nostrum (CMN), University of Murcia, Murcia, Spain
| | - Beatriz Baño-Otalora
- Division of Neuroscience and Experimental Psychology, Faculty of Biology Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - María Angeles Rol
- Institute of Biomedical Research of Murcia, Virgen de la Arrixaca University Hospital, University of Murcia, Murcia, Spain.,CEIR Campus Mare Nostrum (CMN), University of Murcia, Murcia, Spain.,Chronobiology Laboratory, Department of Physiology, Faculty of Biology, University of Murcia, Murcia, Spain
| | - Juan Antonio Madrid
- Institute of Biomedical Research of Murcia, Virgen de la Arrixaca University Hospital, University of Murcia, Murcia, Spain.,CEIR Campus Mare Nostrum (CMN), University of Murcia, Murcia, Spain.,Chronobiology Laboratory, Department of Physiology, Faculty of Biology, University of Murcia, Murcia, Spain
| | - Miroljub Popović
- Department of Human Anatomy and Psychobiology, Faculty of Medicine, University of Murcia, Murcia, Spain.,Institute of Biomedical Research of Murcia, Virgen de la Arrixaca University Hospital, University of Murcia, Murcia, Spain.,CEIR Campus Mare Nostrum (CMN), University of Murcia, Murcia, Spain
| |
Collapse
|