1
|
Hernández-Hernández E, Ledesma-Corvi S, Jornet-Plaza J, García-Fuster MJ. Fast-acting antidepressant-like effects of ketamine in aged male rats. Pharmacol Rep 2024; 76:991-1000. [PMID: 39158787 PMCID: PMC11387441 DOI: 10.1007/s43440-024-00636-y] [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: 05/29/2024] [Revised: 07/25/2024] [Accepted: 08/06/2024] [Indexed: 08/20/2024]
Abstract
BACKGROUND The aging process causes anatomical and physiological changes that predispose to the development of late-life depression while reduces the efficacy of classical antidepressants. Novel fast-acting antidepressants such as ketamine might be good candidates to be explored in the context of aging, especially given the lack of previous research on its efficacy for this age period. Thus, the aim of the present study was to characterize ketamine's effects in older rats. METHODS The fast-acting (30 min) and repeated (7 days) antidepressant-like effects of ketamine (5 mg/kg, ip) were evaluated in 14-month-old single-housed rats through the forced-swim and novelty-suppressed feeding tests. In parallel, the modulation of neurotrophic-related proteins (i.e., mBDNF, mTOR, GSK3) was assessed in brain regions affected by the aging process, prefrontal cortex and hippocampus, as well as possible changes in hippocampal cell proliferation. RESULTS Acute ketamine induced a fast-acting antidepressant-like response in male aged rats, as observed by a reduced immobility in the forced-swim test, in parallel with a region-specific increase in mBDNF protein content in prefrontal cortex. However, repeated ketamine failed to induce antidepressant-like efficacy, but decreased mBDNF protein content in prefrontal cortex. The rate of hippocampal cell proliferation and/or other markers evaluated was not modulated by either paradigm of ketamine. CONCLUSIONS These results complement prior data supporting a fast-acting antidepressant-like effect of ketamine in rats, to further extend its efficacy to older ages. Future studies are needed to further clarify the lack of response after the repeated treatment as well as its potential adverse effects in aging.
Collapse
Affiliation(s)
- Elena Hernández-Hernández
- IUNICS, University of the Balearic Islands, Cra. de Valldemossa, Km 7.5, Palma, E-07122, Spain
- Health Research Institute of the Balearic Islands (IdISBa), Palma, Spain
- Present address: Department of Pharmacology, University of the Basque Country (EHU/UPV), Leioa, Spain
| | - Sandra Ledesma-Corvi
- IUNICS, University of the Balearic Islands, Cra. de Valldemossa, Km 7.5, Palma, E-07122, Spain
- Health Research Institute of the Balearic Islands (IdISBa), Palma, Spain
| | - Jordi Jornet-Plaza
- IUNICS, University of the Balearic Islands, Cra. de Valldemossa, Km 7.5, Palma, E-07122, Spain
- Health Research Institute of the Balearic Islands (IdISBa), Palma, Spain
| | - M Julia García-Fuster
- IUNICS, University of the Balearic Islands, Cra. de Valldemossa, Km 7.5, Palma, E-07122, Spain.
- Health Research Institute of the Balearic Islands (IdISBa), Palma, Spain.
- Department of Medicine, University of the Balearic Islands, Palma, Spain.
| |
Collapse
|
2
|
Qian X, Zhong ZD, Zhang Y, Qiu LQ, Tan HJ. Fluoxetine mitigates depressive-like behaviors in mice via anti-inflammation and enhancing neuroplasticity. Brain Res 2024; 1825:148723. [PMID: 38101693 DOI: 10.1016/j.brainres.2023.148723] [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: 10/09/2023] [Revised: 11/22/2023] [Accepted: 12/12/2023] [Indexed: 12/17/2023]
Abstract
Neuroplasticity and inflammation represent a common final pathway for effective antidepressant treatment. SSRIs are the most commonly prescribed medications for depression and have demonstrated efficacy in reducing depressive symptoms. However, the precise impact of SSRIs on neuroplasticity and inflammation remains unclear. In this study, we aimed to investigate the influence of long-term treatment with SSRIs on hippocampal neuron, inflammation, synaptic function and morphology. Our findings revealed that fluoxetine treatment significantly alleviated behavioral despair, anhedonia, and anxiety in reserpine-treated mice. Moreover, fluoxetine mitigated hippocampal neuron impairment, inhibited inflammatory release, and increased the expression of synaptic proteins markers (SYP and PSD95) in mice. Notably, fluoxetine also suppressed reserpine-induced synapse loss in the hippocampus. Based on these results, fluoxetine has been demonstrated effectively to ameliorate depressive mood and cognitive dysfunction, possibly through the enhancement of synaptic plasticity. Overall, our study contributes to a further understanding of the mechanisms underlying the therapeutic effects of fluoxetine and its potential role in improving depressive symptoms and cognitive impairments.
Collapse
Affiliation(s)
- Xu Qian
- School of Chemistry, Guangdong Key Lab of Chiral Molecules and Drug Discovery, Sun Yat-Sen University, Guangzhou 510275, China.
| | - Zuo-Dong Zhong
- School of Pharmacy, Guangzhou Medical University, Guangzhou 510275, China
| | - Yao Zhang
- Department of Respiratory and Critical Medicine, General Hospital of Eastern Theater Command, Nanjing 210016, China
| | - Li-Qin Qiu
- School of Chemistry, Guangdong Key Lab of Chiral Molecules and Drug Discovery, Sun Yat-Sen University, Guangzhou 510275, China
| | - Hui-Jun Tan
- School of Chemistry, Guangdong Key Lab of Chiral Molecules and Drug Discovery, Sun Yat-Sen University, Guangzhou 510275, China.
| |
Collapse
|
3
|
Micheli L, D'Andrea G, Creanza TM, Volpe D, Ancona N, Scardigli R, Tirone F. Transcriptome analysis reveals genes associated with stem cell activation by physical exercise in the dentate gyrus of aged p16Ink4a knockout mice. Front Cell Dev Biol 2023; 11:1270892. [PMID: 37928906 PMCID: PMC10621069 DOI: 10.3389/fcell.2023.1270892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 10/06/2023] [Indexed: 11/07/2023] Open
Abstract
Throughout adulthood neural stem cells divide in neurogenic niches-the dentate gyrus of the hippocampus and the subventricular zone-producing progenitor cells and new neurons. Stem cells self-renew, thus preserving their pool. Furthermore, the number of stem/progenitor cells in the neurogenic niches decreases with age. We have previously demonstrated that the cyclin-dependent kinase inhibitor p16Ink4a maintains, in aged mice, the pool of dentate gyrus stem cells by preventing their activation after a neurogenic stimulus such as exercise (running). We showed that, although p16Ink4a ablation by itself does not activate stem/progenitor cells, exercise strongly induced stem cell proliferation in p16Ink4a knockout dentate gyrus, but not in wild-type. As p16Ink4a regulates stem cell self-renewal during aging, we sought to profile the dentate gyrus transcriptome from p16Ink4a wild-type and knockout aged mice, either sedentary or running for 12 days. By pairwise comparisons of differentially expressed genes and by correlative analyses through the DESeq2 software, we identified genes regulated by p16Ink4a deletion, either without stimulus (running) added, or following running. The p16Ink4a knockout basic gene signature, i.e., in sedentary mice, involves upregulation of apoptotic, neuroinflammation- and synaptic activity-associated genes, suggesting a reactive cellular state. Conversely, another set of 106 genes we identified, whose differential expression specifically reflects the pattern of proliferative response of p16 knockout stem cells to running, are involved in processes that regulate stem cell activation, such as synaptic function, neurotransmitter metabolism, stem cell proliferation control, and reactive oxygen species level regulation. Moreover, we analyzed the regulation of these stem cell-specific genes after a second running stimulus. Surprisingly, the second running neither activated stem cell proliferation in the p16Ink4a knockout dentate gyrus nor changed the expression of these genes, confirming that they are correlated to the stem cell reactivity to stimulus, a process where they may play a role regulating stem cell activation.
Collapse
Affiliation(s)
- Laura Micheli
- Institute of Biochemistry and Cell Biology, National Research Council, Rome, Italy
| | - Giorgio D'Andrea
- Institute of Biochemistry and Cell Biology, National Research Council, Rome, Italy
| | - Teresa Maria Creanza
- CNR-Institute of Intelligent Industrial Technologies and Systems for Advanced Manufacturing, Bari, Italy
| | - Daniel Volpe
- Institute of Biochemistry and Cell Biology, National Research Council, Rome, Italy
| | - Nicola Ancona
- CNR-Institute of Intelligent Industrial Technologies and Systems for Advanced Manufacturing, Bari, Italy
| | - Raffaella Scardigli
- Institute of Translational Pharmacology, National Research Council, Rome, Italy
- European Brain Research Institute (EBRI), Rome, Italy
| | - Felice Tirone
- Institute of Biochemistry and Cell Biology, National Research Council, Rome, Italy
| |
Collapse
|
4
|
Urmann C, Bieler L, Hackl M, Chia-Leeson O, Couillard-Despres S, Riepl H. Semi-Synthesis of Different Pyranoflavonoid Backbones and the Neurogenic Potential. Molecules 2023; 28:molecules28104023. [PMID: 37241764 DOI: 10.3390/molecules28104023] [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: 03/30/2023] [Revised: 05/04/2023] [Accepted: 05/06/2023] [Indexed: 05/28/2023] Open
Abstract
Flavonoids and chalcones are known for their manifold biological activities, of which many affect the central nervous system. Pyranochalcones were recently shown to have a great neurogenic potential, which is partly due to a specific structural motif-the pyran ring. Accordingly, we questioned if other flavonoid backbones with a pyran ring as structural moiety would also show neurogenic potential. Different semi-synthetic approaches starting with the prenylated chalcone xanthohumol, isolated from hops, led to pyranoflavanoids with different backbones. We identified the chalcone backbone as the most active backbone with pyran ring using a reporter gene assay based on the promoter activity of doublecortin, an early neuronal marker. Pyranochalcones therefore appear to be promising compounds for further development as a treatment strategy for neurodegenerative diseases.
Collapse
Affiliation(s)
- Corinna Urmann
- Organic-Analytical Chemistry, Weihenstephan-Triesdorf University of Applied Sciences, 94315 Straubing, Germany
- TUM Campus Straubing for Biotechnology and Sustainability, Technical University of Munich, 94315 Straubing, Germany
| | - Lara Bieler
- Institute of Experimental Neuroregeneration, Spinal Cord Injury and Tissue Regeneration Center Salzburg, Paracelsus Medical University Salzburg, 5020 Salzburg, Austria
- Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria
| | - Michael Hackl
- TUM Campus Straubing for Biotechnology and Sustainability, Technical University of Munich, 94315 Straubing, Germany
| | - Olivia Chia-Leeson
- TUM Campus Straubing for Biotechnology and Sustainability, Technical University of Munich, 94315 Straubing, Germany
| | - Sebastien Couillard-Despres
- Institute of Experimental Neuroregeneration, Spinal Cord Injury and Tissue Regeneration Center Salzburg, Paracelsus Medical University Salzburg, 5020 Salzburg, Austria
- Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria
| | - Herbert Riepl
- Organic-Analytical Chemistry, Weihenstephan-Triesdorf University of Applied Sciences, 94315 Straubing, Germany
- TUM Campus Straubing for Biotechnology and Sustainability, Technical University of Munich, 94315 Straubing, Germany
| |
Collapse
|
5
|
Jaggar M, Ghosh S, Janakiraman B, Chatterjee A, Maheshwari M, Dewan V, Hare B, Deb S, Figueiredo D, Duman RS, Vaidya VA. Influence of Chronic Electroconvulsive Seizures on Plasticity-Associated Gene Expression and Perineuronal Nets Within the Hippocampi of Young Adult and Middle-Aged Sprague-Dawley Rats. Int J Neuropsychopharmacol 2023; 26:294-306. [PMID: 36879414 PMCID: PMC10109107 DOI: 10.1093/ijnp/pyad008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Accepted: 03/01/2023] [Indexed: 03/08/2023] Open
Abstract
BACKGROUND Electroconvulsive seizure therapy is often used in both treatment-resistant and geriatric depression. However, preclinical studies identifying targets of chronic electroconvulsive seizure (ECS) are predominantly focused on animal models in young adulthood. Given that putative transcriptional, neurogenic, and neuroplastic mechanisms implicated in the behavioral effects of chronic ECS themselves exhibit age-dependent modulation, it remains unknown whether the molecular and cellular targets of chronic ECS vary with age. METHODS We subjected young adult (2-3 months) and middle-aged (12-13 months), male Sprague Dawley rats to sham or chronic ECS and assessed for despair-like behavior, hippocampal gene expression, hippocampal neurogenesis, and neuroplastic changes in the extracellular matrix, reelin, and perineuronal net numbers. RESULTS Chronic ECS reduced despair-like behavior at both ages, accompanied by overlapping and unique changes in activity-dependent and trophic factor gene expression. Although chronic ECS had a similar impact on quiescent neural progenitor numbers at both ages, the eventual increase in hippocampal progenitor proliferation was substantially higher in young adulthood. We noted a decline in reelin⁺ cell numbers following chronic ECS only in young adulthood. In contrast, an age-invariant, robust dissolution of perineuronal net numbers that encapsulate parvalbumin⁺ neurons in the hippocampus were observed following chronic ECS. CONCLUSION Our findings indicate that age is a key variable in determining the nature of chronic ECS-evoked molecular and cellular changes in the hippocampus. This raises the intriguing possibility that chronic ECS may recruit distinct, as well as overlapping, mechanisms to drive antidepressant-like behavioral changes in an age-dependent manner.
Collapse
Affiliation(s)
- Minal Jaggar
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai, India
| | - Shreya Ghosh
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai, India
| | - Balaganesh Janakiraman
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai, India
| | - Ashmita Chatterjee
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai, India
| | - Megha Maheshwari
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai, India
| | - Vani Dewan
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai, India
| | - Brendan Hare
- Division of Molecular Psychiatry, Department of Psychiatry and Pharmacology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Sukrita Deb
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai, India
| | - Dwight Figueiredo
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai, India
| | - Ronald S Duman
- Division of Molecular Psychiatry, Department of Psychiatry and Pharmacology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Vidita A Vaidya
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai, India
| |
Collapse
|
6
|
Micheli L, Bertini L, Bonato A, Villanova N, Caruso C, Caruso M, Bernini R, Tirone F. Role of Hydroxytyrosol and Oleuropein in the Prevention of Aging and Related Disorders: Focus on Neurodegeneration, Skeletal Muscle Dysfunction and Gut Microbiota. Nutrients 2023; 15:1767. [PMID: 37049607 PMCID: PMC10096778 DOI: 10.3390/nu15071767] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Revised: 03/16/2023] [Accepted: 03/24/2023] [Indexed: 04/09/2023] Open
Abstract
Aging is a multi-faceted process caused by the accumulation of cellular damage over time, associated with a gradual reduction of physiological activities in cells and organs. This degeneration results in a reduced ability to adapt to homeostasis perturbations and an increased incidence of illnesses such as cognitive decline, neurodegenerative and cardiovascular diseases, cancer, diabetes, and skeletal muscle pathologies. Key features of aging include a chronic low-grade inflammation state and a decrease of the autophagic process. The Mediterranean diet has been associated with longevity and ability to counteract the onset of age-related disorders. Extra virgin olive oil, a fundamental component of this diet, contains bioactive polyphenolic compounds as hydroxytyrosol (HTyr) and oleuropein (OLE), known for their antioxidant, anti-inflammatory, and neuroprotective properties. This review is focused on brain, skeletal muscle, and gut microbiota, as these systems are known to interact at several levels. After the description of the chemistry and pharmacokinetics of HTyr and OLE, we summarize studies reporting their effects in in vivo and in vitro models of neurodegenerative diseases of the central/peripheral nervous system, adult neurogenesis and depression, senescence and lifespan, and age-related skeletal muscle disorders, as well as their impact on the composition of the gut microbiota.
Collapse
Affiliation(s)
- Laura Micheli
- Institute of Biochemistry and Cell Biology, National Research Council (IBBC-CNR), Via E. Ramarini 32, Monterotondo, 00015 Rome, Italy
| | - Laura Bertini
- Department of Ecological and Biological Sciences (DEB), University of Tuscia, Largo dell’Università, 01100 Viterbo, Italy
| | - Agnese Bonato
- Institute of Biochemistry and Cell Biology, National Research Council (IBBC-CNR), Via E. Ramarini 32, Monterotondo, 00015 Rome, Italy
| | - Noemi Villanova
- Department of Agriculture and Forest Sciences (DAFNE), University of Tuscia, Via San Camillo de Lellis, 01100 Viterbo, Italy
| | - Carla Caruso
- Department of Ecological and Biological Sciences (DEB), University of Tuscia, Largo dell’Università, 01100 Viterbo, Italy
| | - Maurizia Caruso
- Institute of Biochemistry and Cell Biology, National Research Council (IBBC-CNR), Via E. Ramarini 32, Monterotondo, 00015 Rome, Italy
| | - Roberta Bernini
- Department of Agriculture and Forest Sciences (DAFNE), University of Tuscia, Via San Camillo de Lellis, 01100 Viterbo, Italy
| | - Felice Tirone
- Institute of Biochemistry and Cell Biology, National Research Council (IBBC-CNR), Via E. Ramarini 32, Monterotondo, 00015 Rome, Italy
| |
Collapse
|
7
|
Surya K, Manickam N, Jayachandran KS, Kandasamy M, Anusuyadevi M. Resveratrol Mediated Regulation of Hippocampal Neuroregenerative Plasticity via SIRT1 Pathway in Synergy with Wnt Signaling: Neurotherapeutic Implications to Mitigate Memory Loss in Alzheimer's Disease. J Alzheimers Dis 2023; 94:S125-S140. [PMID: 36463442 PMCID: PMC10473144 DOI: 10.3233/jad-220559] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Alzheimer's disease (AD) is a major form of dementia. Abnormal amyloidogenic event-mediated degeneration of cholinergic neurons in the cognitive centers of the brain has been attributed to neuropathological sequelae and behavioral deficits in AD. Besides, impaired adult neurogenesis in the hippocampus has experimentally been realized as an underlying cause of dementia regardless of neurodegeneration. Therefore, nourishing the neurogenic process in the hippocampus has been considered an effective therapeutic strategy to mitigate memory loss. In the physiological state, the Wnt pathway has been identified as a potent mitogenic generator in the hippocampal stem cell niche. However, downstream components of Wnt signaling have been noticed to be downregulated in AD brains. Resveratrol (RSV) is a potent Sirtuin1 (SIRT1) enhancer that facilitates neuroprotection and promotes neurogenesis in the hippocampus of the adult brain. While SIRT1 is an important positive regulator of Wnt signaling, ample reports indicate that RSV treatment strongly mediates the fate determination of stem cells through Wnt signaling. However, the possible therapeutic roles of RSV-mediated SIRT1 enhancement on the regulation of hippocampal neurogenesis and reversal of memory loss through the Wnt signaling pathway have not been addressed yet. Taken together, this review describes RSV-mediated effects on the regulation of hippocampal neurogenesis via the activation of SIRT1 in synergy with the Wnt signaling. Further, the article emphasizes a hypothesis that RSV treatment can provoke the activation of quiescent neural stem cells and prime their neurogenic capacity in the hippocampus via Wnt signaling in AD.
Collapse
Affiliation(s)
- Kumar Surya
- Department of Biochemistry, Molecular Neuro-gerontology Laboratory, School of Life Sciences, Bharathidasan University, Tiruchirappalli, Tamil Nadu, India
| | - Nivethitha Manickam
- Department of Animal Science, Laboratory of Stem Cells and Neuroregeneration, School of Life Sciences, Bharathidasan University, Tiruchirappalli, Tamil Nadu, India
| | - Kesavan Swaminathan Jayachandran
- Department of Bioinformatics, Molecular Cardiology and Drug Discovery Laboratory, School of Life Sciences, Bharathidasan University, Tiruchirappalli, Tamil Nadu, India
| | - Mahesh Kandasamy
- Department of Animal Science, Laboratory of Stem Cells and Neuroregeneration, School of Life Sciences, Bharathidasan University, Tiruchirappalli, Tamil Nadu, India
- University Grants Commission-Faculty Recharge Programme (UGC-FRP), New Delhi, India
| | - Muthuswamy Anusuyadevi
- Department of Biochemistry, Molecular Neuro-gerontology Laboratory, School of Life Sciences, Bharathidasan University, Tiruchirappalli, Tamil Nadu, India
| |
Collapse
|
8
|
McNerlin C, Guan F, Bronk L, Lei K, Grosshans D, Young DW, Gaber MW, Maletic-Savatic M. Targeting hippocampal neurogenesis to protect astronauts' cognition and mood from decline due to space radiation effects. LIFE SCIENCES IN SPACE RESEARCH 2022; 35:170-179. [PMID: 36336363 DOI: 10.1016/j.lssr.2022.07.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 06/30/2022] [Accepted: 07/26/2022] [Indexed: 06/16/2023]
Abstract
Neurogenesis is an essential, lifelong process during which neural stem cells generate new neurons within the hippocampus, a center for learning, memory, and mood control. Neural stem cells are vulnerable to environmental insults spanning from chronic stress to radiation. These insults reduce their numbers and diminish neurogenesis, leading to memory decline, anxiety, and depression. Preserving neural stem cells could thus help prevent these neurogenesis-associated pathologies, an outcome particularly important for long-term space missions where environmental exposure to radiation is significantly higher than on Earth. Multiple developments, from mechanistic discoveries of radiation injury on hippocampal neurogenesis to new platforms for the development of selective, specific, effective, and safe small molecules as neurogenesis-protective agents hold great promise to minimize radiation damage on neurogenesis. In this review, we summarize the effects of space-like radiation on hippocampal neurogenesis. We then focus on current advances in drug discovery and development and discuss the nuclear receptor TLX/NR2E1 (oleic acid receptor) as an example of a neurogenic target that might rescue neurogenesis following radiation.
Collapse
Affiliation(s)
- Clare McNerlin
- Georgetown University School of Medicine, 3900 Reservoir Rd NW, Washington D.C. 20007, United States of America
| | - Fada Guan
- Department of Therapeutic Radiology, Yale University School of Medicine, New Haven, CT, 06510, United States of America
| | - Lawrence Bronk
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, United States of America
| | - Kevin Lei
- Graduate School for Biomedical Sciences, Baylor College of Medicine, Houston, Texas, 77030, United States of America; Jan and Dan Duncan Neurological Research Institute, 1250 Moursund St. Houston, TX 77030, United States of America
| | - David Grosshans
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, United States of America
| | - Damian W Young
- Jan and Dan Duncan Neurological Research Institute, 1250 Moursund St. Houston, TX 77030, United States of America; Center for Drug Discovery, Department of Pathology and Immunology Baylor College of Medicine, Houston, Texas, 77030, United States of America; Department of Pharmacology and Chemical Biology, Baylor College of Medicine, Houston, Texas 77030, United States of America
| | - M Waleed Gaber
- Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, United States of America.
| | - Mirjana Maletic-Savatic
- Jan and Dan Duncan Neurological Research Institute, 1250 Moursund St. Houston, TX 77030, United States of America; Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, United States of America; Department of Neuroscience, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, United States of America.
| |
Collapse
|
9
|
Piwowarczyk-Nowak A, Pałasz A, Suszka-Świtek A, Błaszczyk I, Bogus K, Łasut-Szyszka B, Krzystanek M, Worthington JJ. Effect of Escitalopram on the Number of DCX-Positive Cells and NMUR2 Receptor Expression in the Rat Hippocampus under the Condition of NPSR Receptor Blockade. Pharmaceuticals (Basel) 2022; 15:631. [PMID: 35631458 PMCID: PMC9143903 DOI: 10.3390/ph15050631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 05/18/2022] [Accepted: 05/19/2022] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Neuropeptide S (NPS) is a multifunctional regulatory factor that exhibits a potent anxiolytic activity in animal models. However, there are no reports dealing with the potential molecular interactions between the activity of selective serotonin reuptake inhibitors (SSRIs) and NPS signaling, especially in the context of adult neurogenesis and the expression of noncanonical stress-related neuropeptides such as neuromedin U (NMU). The present work therefore focused on immunoexpression of neuromedin U receptor 2 (NMUR2) and doublecortin (DCX) in the rat hippocampus after acute treatment with escitalopram and in combination with selective neuropeptide S receptor (NPSR) blockade. METHODS Studies were carried out on adult, male Sprague-Dawley rats that were divided into five groups: animals injected with saline (control) and experimental individuals treated with escitalopram (at single dose 10 mg/kg daily), escitalopram + SHA-68, a selective NPSR antagonist (at single dose 40 mg/kg), SHA-68 alone, and corresponding vehicle control. All animals were sacrificed under halothane anaesthesia. The whole hippocampi were quickly excised, fixed, and finally sliced for general qualitative immunohistochemical assessment of the NPSR and NMUR2 expression. The number of immature neurons was enumerated using immunofluorescent detection of doublecortin (DCX) expression within the subgranular zone (SGZ). RESULTS Acute escitalopram administration affects the number of DCX and NMUR2-expressing cells in the adult rat hippocampus. A decreased number of DCX-expressing neuroblasts after treatment with escitalopram was augmented by SHA-68 coadministration. CONCLUSIONS Early pharmacological effects of escitalopram may be at least partly connected with local NPSR-related alterations of neuroblast maturation in the rat hippocampus. Escitalopram may affect neuropeptide and DCX-expression starting even from the first dose. Adult neurogenesis may be regulated via paracrine neuropeptide S and NMU-related signaling.
Collapse
Affiliation(s)
- Aneta Piwowarczyk-Nowak
- Department of Anatomy, Faculty of Medical Sciences in Katowice, Medical University of Silesia, ul. Medyków 18, 40-752 Katowice, Poland;
| | - Artur Pałasz
- Department of Histology, Faculty of Medical Sciences in Katowice, Medical University of Silesia, ul. Medyków 18, 40-752 Katowice, Poland; (A.S.-Ś.); (I.B.); (K.B.)
| | - Aleksandra Suszka-Świtek
- Department of Histology, Faculty of Medical Sciences in Katowice, Medical University of Silesia, ul. Medyków 18, 40-752 Katowice, Poland; (A.S.-Ś.); (I.B.); (K.B.)
| | - Iwona Błaszczyk
- Department of Histology, Faculty of Medical Sciences in Katowice, Medical University of Silesia, ul. Medyków 18, 40-752 Katowice, Poland; (A.S.-Ś.); (I.B.); (K.B.)
| | - Katarzyna Bogus
- Department of Histology, Faculty of Medical Sciences in Katowice, Medical University of Silesia, ul. Medyków 18, 40-752 Katowice, Poland; (A.S.-Ś.); (I.B.); (K.B.)
| | - Barbara Łasut-Szyszka
- Center for Translational Research and Molecular Biology of Cancer, Maria Skłodowska-Curie National Research Institute of Oncology, Gliwice Branch, 44-102 Gliwice, Poland;
| | - Marek Krzystanek
- Clinic of Psychiatric Rehabilitation, Department of Psychiatry and Psychotherapy, Faculty of Medical Sciences in Katowice, Medical University of Silesia, ul. Ziolowa 45/47, 40-635 Katowice, Poland;
| | - John J. Worthington
- Division of Biomedical and Life Sciences, Faculty of Health and Medicine, Lancaster University, Lancaster LA1 4YG, UK;
| |
Collapse
|
10
|
Identification and functional characterization of CD133+GFAP+CD117+Sca1+ neural stem cells. Mol Cell Biochem 2022; 477:897-914. [DOI: 10.1007/s11010-021-04339-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2021] [Accepted: 12/15/2021] [Indexed: 02/03/2023]
|
11
|
Micheli L, Creanza TM, Ceccarelli M, D'Andrea G, Giacovazzo G, Ancona N, Coccurello R, Scardigli R, Tirone F. Transcriptome Analysis in a Mouse Model of Premature Aging of Dentate Gyrus: Rescue of Alpha-Synuclein Deficit by Virus-Driven Expression or by Running Restores the Defective Neurogenesis. Front Cell Dev Biol 2021; 9:696684. [PMID: 34485283 PMCID: PMC8415876 DOI: 10.3389/fcell.2021.696684] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2021] [Accepted: 07/21/2021] [Indexed: 02/05/2023] Open
Abstract
The dentate gyrus of the hippocampus and the subventricular zone are neurogenic niches where neural stem and progenitor cells replicate throughout life to generate new neurons. The Btg1 gene maintains the stem cells of the neurogenic niches in quiescence. The deletion of Btg1 leads to an early transient increase of stem/progenitor cells division, followed, however, by a decrease during adulthood of their proliferative capability, accompanied by apoptosis. Since a physiological decrease of neurogenesis occurs during aging, the Btg1 knockout mouse may represent a model of neural aging. We have previously observed that the defective neurogenesis of the Btg1 knockout model is rescued by the powerful neurogenic stimulus of physical exercise (running). To identify genes responsible for stem and progenitor cells maintenance, we sought here to find genes underlying this premature neural aging, and whose deregulated expression could be rescued by running. Through RNA sequencing we analyzed the transcriptomic profiles of the dentate gyrus isolated from Btg1 wild-type or Btg1 knockout adult (2-month-old) mice submitted to physical exercise or sedentary. In Btg1 knockout mice, 545 genes were deregulated, relative to wild-type, while 2081 genes were deregulated by running. We identified 42 genes whose expression was not only down-regulated in the dentate gyrus of Btg1 knockout, but was also counter-regulated to control levels by running in Btg1 knockout mice, vs. sedentary. Among these 42 counter-regulated genes, alpha-synuclein (Snca), Fos, Arc and Npas4 showed significantly greater differential regulation. These genes control neural proliferation, apoptosis, plasticity and memory and are involved in aging. In particular, Snca expression decreases during aging. We tested, therefore, whether an Snca-expressing lentivirus, by rescuing the defective Snca levels in the dentate gyrus of Btg1 knockout mice, could also reverse the aging phenotype, in particular the defective neurogenesis. We found that the exogenous expression of Snca reversed the Btg1 knockout-dependent decrease of stem cell proliferation as well as the increase of progenitor cell apoptosis. This indicates that Snca has a functional role in the process of neural aging observed in this model, and also suggests that Snca acts as a positive regulator of stem cell maintenance.
Collapse
Affiliation(s)
- Laura Micheli
- Institute of Biochemistry and Cell Biology, National Research Council, Rome, Italy
| | - Teresa Maria Creanza
- Institute of Intelligent Industrial Technologies and Systems for Advanced Manufacturing, National Research Council, Bari, Italy
| | - Manuela Ceccarelli
- Institute of Biochemistry and Cell Biology, National Research Council, Rome, Italy
| | - Giorgio D'Andrea
- Institute of Biochemistry and Cell Biology, National Research Council, Rome, Italy
| | - Giacomo Giacovazzo
- Preclinical Neuroscience, European Center for Brain Research (CERC)/IRCCS Santa Lucia Foundation, Rome, Italy
| | - Nicola Ancona
- Institute of Intelligent Industrial Technologies and Systems for Advanced Manufacturing, National Research Council, Bari, Italy
| | - Roberto Coccurello
- Preclinical Neuroscience, European Center for Brain Research (CERC)/IRCCS Santa Lucia Foundation, Rome, Italy.,Institute for Complex Systems, National Research Council, Rome, Italy
| | - Raffaella Scardigli
- Institute of Translational Pharmacology, National Research Council, Rome, Italy
| | - Felice Tirone
- Institute of Biochemistry and Cell Biology, National Research Council, Rome, Italy
| |
Collapse
|
12
|
Sumien N, Wells MS, Sidhu A, Wong JM, Forster MJ, Zheng QX, Kelleher-Andersson JA. Novel pharmacotherapy: NNI-362, an allosteric p70S6 kinase stimulator, reverses cognitive and neural regenerative deficits in models of aging and disease. Stem Cell Res Ther 2021; 12:59. [PMID: 33436007 PMCID: PMC7805132 DOI: 10.1186/s13287-020-02126-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Accepted: 12/27/2020] [Indexed: 12/21/2022] Open
Abstract
Aging is known to slow the neurogenic capacity of the hippocampus, one of only two mammalian adult neurogenic niches. The reduction of adult-born neurons with age may initiate cognitive decline progression which is exacerbated in chronic neurodegenerative disorders, e.g., Alzheimer's disease (AD). With physiologic neurogenesis diminished, but still viable in aging, non-invasive therapeutic modulation of this neuron regeneration process remains possible. The discovery of truly novel neuron regenerative therapies could be identified through phenotypic screening of small molecules that promote adult-born neurons from human neural progenitor cells (hNPCs). By identifying neuron-generating therapeutics and potentially novel mechanism of actions, therapeutic benefit could be confirmed through in vivo proof-of-concept studies. The key aging and longevity mTOR/p70S6 kinase axis, a commonly targeted pathway, is substrate for potential selective kinase modulators to promote new hippocampal neurons from NPCs. The highly regulated downstream substrate of mTOR, p70S6 kinase, directly controls pleiotropic cellular activities, including translation and cell growth. Stimulating this kinase, selectively in an adult neurogenic niche, should promote NPC proliferation, and cell growth and survival in the hippocampus. Studies of kinase profiling and immunocytochemistry of human progenitor neurogenesis suggest that the novel small molecule NNI-362 stimulates p70S6 kinase phosphorylation, which, in turn, promotes proliferation and differentiation of NPCs to neurons. NNI-362 promoted the associative reversal of age- and disease-related cognitive deficits in aged mice and Down syndrome-modeled mice. This oral, allosteric modulator may ultimately be beneficial for age-related neurodegenerative disorders involving hippocampal-dependent cognitive impairment, specifically AD, by promoting endogenous hippocampal regeneration.
Collapse
Affiliation(s)
- Nathalie Sumien
- Department of Pharmacology & Neuroscience, University of North Texas Health Science Center, 3500 Camp Bowie Blvd., Fort Worth, TX, USA
| | - Matthew S Wells
- Neuronascent, Inc., 15601 Crabbs Branch Way, Rockville, MD, 20855, USA
- Novavax, 21 Firstfield Rd., Gaithersburg, MD, 20878, USA
| | - Akram Sidhu
- Department of Pharmacology & Neuroscience, University of North Texas Health Science Center, 3500 Camp Bowie Blvd., Fort Worth, TX, USA
| | - Jessica M Wong
- Department of Pharmacology & Neuroscience, University of North Texas Health Science Center, 3500 Camp Bowie Blvd., Fort Worth, TX, USA
| | - Michael J Forster
- Department of Pharmacology & Neuroscience, University of North Texas Health Science Center, 3500 Camp Bowie Blvd., Fort Worth, TX, USA
| | - Qiao-Xi Zheng
- Neuronascent, Inc., 15601 Crabbs Branch Way, Rockville, MD, 20855, USA
| | | |
Collapse
|
13
|
Damiani AP, Strapazzon G, de Oliveira Sardinha TT, Rohr P, Gajski G, de Pinho RA, de Andrade VM. Melatonin supplementation over different time periods until ageing modulates genotoxic parameters in mice. Mutagenesis 2020; 35:465-478. [PMID: 32720686 DOI: 10.1093/mutage/geaa017] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Accepted: 06/19/2020] [Indexed: 12/17/2022] Open
Abstract
The ageing process is a multifactorial phenomenon, associated with decreased physiological and cellular functions and an increased propensity for various degenerative diseases. Studies on melatonin (N-acetyl-5-methoxytryptamine), a potent antioxidant, are gaining attention since melatonin production declines with advancing age. Hence, the aim of this study was to evaluate the effects of chronic melatonin consumption on genotoxic and mutagenic parameters of old Swiss mice. Herein, 3-month-old Swiss albino male mice (n = 240) were divided into eight groups and subdivided into two experiments: first (three groups): natural ageing experiment; second (five groups): animals that started water or melatonin supplementation at different ages (3, 6, 12 and 18 months) until 21 months. After 21 months, the animals from the second experiment were euthanized to perform the comet assay, micronucleus test and western blot analysis. The results demonstrated that melatonin prolonged the life span of the animals. Relative to genomic instability, melatonin was effective in reducing DNA damage caused by ageing, presenting antigenotoxic and antimutagenic activities, independently of initiation age. The group receiving melatonin for 18 months had high levels of APE1 and OGG1 repair enzymes. Conclusively, melatonin presents an efficient antioxidant mechanism aiding modulating genetic and physiological alterations due to ageing.
Collapse
Affiliation(s)
- Adriani Paganini Damiani
- Translational Biomedicine Laboratory, Graduate Programme of Health Sciences, Health Sciences Unit, University of Southern Santa Catarina, Criciúma, SC, Brazil
| | - Giulia Strapazzon
- Translational Biomedicine Laboratory, Graduate Programme of Health Sciences, Health Sciences Unit, University of Southern Santa Catarina, Criciúma, SC, Brazil
| | - Thanielly Thais de Oliveira Sardinha
- Translational Biomedicine Laboratory, Graduate Programme of Health Sciences, Health Sciences Unit, University of Southern Santa Catarina, Criciúma, SC, Brazil
| | - Paula Rohr
- Translational Biomedicine Laboratory, Graduate Programme of Health Sciences, Health Sciences Unit, University of Southern Santa Catarina, Criciúma, SC, Brazil
| | - Goran Gajski
- Mutagenesis Unit, Institute for Medical Research and Occupational Health, Zagreb, Croatia, Croatia
| | - Ricardo Aurino de Pinho
- Laboratory of Exercise Biochemistry in Health, Graduate Program in Health Sciences, School of Medicine, Pontifícia Universidade Católica do Paraná, R. Imac. Conceição - Curitiba - PR, Brazil
| | - Vanessa Moraes de Andrade
- Translational Biomedicine Laboratory, Graduate Programme of Health Sciences, Health Sciences Unit, University of Southern Santa Catarina, Criciúma, SC, Brazil
| |
Collapse
|
14
|
Jorgensen C, Wang Z. Hormonal Regulation of Mammalian Adult Neurogenesis: A Multifaceted Mechanism. Biomolecules 2020; 10:biom10081151. [PMID: 32781670 PMCID: PMC7465680 DOI: 10.3390/biom10081151] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Revised: 07/27/2020] [Accepted: 08/01/2020] [Indexed: 02/07/2023] Open
Abstract
Adult neurogenesis—resulting in adult-generated functioning, integrated neurons—is still one of the most captivating research areas of neuroplasticity. The addition of new neurons in adulthood follows a seemingly consistent multi-step process. These neurogenic stages include proliferation, differentiation, migration, maturation/survival, and integration of new neurons into the existing neuronal network. Most studies assessing the impact of exogenous (e.g., restraint stress) or endogenous (e.g., neurotrophins) factors on adult neurogenesis have focused on proliferation, survival, and neuronal differentiation. This review will discuss the multifaceted impact of hormones on these various stages of adult neurogenesis. Specifically, we will review the evidence for hormonal facilitation (via gonadal hormones), inhibition (via glucocorticoids), and neuroprotection (via recruitment of other neurochemicals such as neurotrophin and neuromodulators) on newly adult-generated neurons in the mammalian brain.
Collapse
Affiliation(s)
- Claudia Jorgensen
- Behavioral Science Department, Utah Valley University, Orem, UT 84058, USA
- Correspondence:
| | - Zuoxin Wang
- Psychology Department and Program in Neuroscience, Florida State University, Tallahassee, FL 32306, USA;
| |
Collapse
|
15
|
Ceccarelli M, D’Andrea G, Micheli L, Tirone F. Interaction Between Neurogenic Stimuli and the Gene Network Controlling the Activation of Stem Cells of the Adult Neurogenic Niches, in Physiological and Pathological Conditions. Front Cell Dev Biol 2020; 8:211. [PMID: 32318568 PMCID: PMC7154047 DOI: 10.3389/fcell.2020.00211] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Accepted: 03/11/2020] [Indexed: 12/26/2022] Open
Abstract
In the adult mammalian brain new neurons are continuously generated throughout life in two niches, the dentate gyrus of the hippocampus and the subventricular zone. This process, called adult neurogenesis, starts from stem cells, which are activated and enter the cell cycle. The proliferative capability of stem cells progressively decreases during aging. The population of stem cells is generally quiescent, and it is not clear whether the potential for stem cells to expand is limited, or whether they can expand and then return to quiescence, remaining available for further activation. Certain conditions may deregulate stem cells quiescence and self-renewal. In fact we discuss the possibility of activation of stem cells by neurogenic stimuli as a function of the intensity of the stimulus (i.e., whether this is physiological or pathological), and of the deregulation of the system (i.e., whether the model is aged or carrying genetic mutations in the gene network controlling quiescence). It appears that when the system is aged and/or carrying mutations of quiescence-maintaining genes, preservation of the quiescent state of stem cells is more critical and stem cells can be activated by a neurogenic stimulus which is ineffective in normal conditions. Moreover, when a neurogenic stimulus is in itself a cause of brain damage (e.g., kainic acid treatment) the activation of stem cells occurs bypassing any inhibitory control. Plausibly, with strong neurogenic stimuli, such as kainic acid injected into the dentate gyrus, the self-renewal capacity of stem cells may undergo rapid exhaustion. However, the self-renewal capability of stem cells persists when normal stimuli are elicited in the presence of a mutation of one of the quiescence-maintaining genes, such as p16Ink4a, p21Cip1 or Btg1. In this case, stem cells become promptly activated by a neurogenic stimulus even during aging. This indicates that stem cells retain a high proliferative capability and plasticity, and suggests that stem cells are protected against the response to stimulus and are resilient to exhaustion. It will be interesting to assess at which functional degree of deregulation of the quiescence-maintaining system, stem cells will remain responsive to repeated neurogenic stimuli without undergoing exhaustion of their pool.
Collapse
Affiliation(s)
| | | | | | - Felice Tirone
- Institute of Biochemistry and Cell Biology, National Research Council (IBBC-CNR), Rome, Italy
| |
Collapse
|
16
|
Ramírez-Rodríguez GB, Palacios-Cabriales DM, Ortiz-López L, Estrada-Camarena EM, Vega-Rivera NM. Melatonin Modulates Dendrite Maturation and Complexity in the Dorsal- and Ventral- Dentate Gyrus Concomitantly with Its Antidepressant-Like Effect in Male Balb/C Mice. Int J Mol Sci 2020; 21:ijms21051724. [PMID: 32138332 PMCID: PMC7084558 DOI: 10.3390/ijms21051724] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 01/17/2020] [Accepted: 01/20/2020] [Indexed: 12/13/2022] Open
Abstract
Adult neurogenesis occurs in the dentate gyrus (DG) of the hippocampus. New neurons help to counteract the effects of stress and several interventions including antidepressant drugs, environmental modifications and internal factors act pro-neurogenic with consequences in the dorsal and ventral DG. Melatonin, the main product synthesized by the pineal gland, induces antidepressant-like effects and modulates several events of the neurogenic process. However, the information related to the capability of melatonin to modulate dendrite maturation and complexity in the dorsal and ventral regions of the DG and their correlation with its antidepressant-like effect is absent. Thus, in this study, we analyzed the impact of melatonin (0, 0.5, 1, 2.5, 5 or 10 mg/kg) administered daily for fourteen days on the number, dendrite complexity and distribution of doublecortin (DCX)-cells in the dorsal-ventral regions of the DG in male Balb/C mice. Doublecortin is a microtubule-associated protein that is expressed during the course of dendritic maturation of newborn neurons. Also, we analyzed the impact of melatonin on despair-like behavior in the forced swim test. We first found a significant increase in the number and higher dendrite complexity, mainly with the doses of 2.5, 5 and 10 mg/kg of melatonin (81%, 122%, 78%). These cells showed more complex dendritic trees in the ventral- and the dorsal- DG. Concomitantly, the doses of 5 and 10 mg/kg of melatonin decreased depressant-like behavior (76%, 82%). Finally, the data corroborate the antidepressant-like effect of melatonin and the increasing number of doublecortin-associated cells. Besides, the data indicate that melatonin favors the number and dendrite complexity of DCX-cells in the dorsal- and ventral- region of the DG, which may explain part of the antidepressant-like effect of melatonin.
Collapse
Affiliation(s)
- Gerardo Bernabé Ramírez-Rodríguez
- Laboratory of Neurogenesis, Division of Clinical Investigations, National Institute of Psychiatry “Ramón de la Fuente Muñiz”, Calzada Mexico-Xochimilco No. 101, Mexico City C.P. 14370, Mexico; (D.M.P.-C.); (L.O.-L.)
- Correspondence: (G.B.R.-R.); (N.M.V.-R.)
| | - Diana Montserrat Palacios-Cabriales
- Laboratory of Neurogenesis, Division of Clinical Investigations, National Institute of Psychiatry “Ramón de la Fuente Muñiz”, Calzada Mexico-Xochimilco No. 101, Mexico City C.P. 14370, Mexico; (D.M.P.-C.); (L.O.-L.)
| | - Leonardo Ortiz-López
- Laboratory of Neurogenesis, Division of Clinical Investigations, National Institute of Psychiatry “Ramón de la Fuente Muñiz”, Calzada Mexico-Xochimilco No. 101, Mexico City C.P. 14370, Mexico; (D.M.P.-C.); (L.O.-L.)
| | - Erika Montserrat Estrada-Camarena
- Laboratory of Neuropsychopharmacology, Division of Neurosciences, National Institute of Psychiatry “Ramón de la Fuente Muñiz”, Calzada Mexico-Xochimilco No. 101, Mexico City C.P. 14370, Mexico;
| | - Nelly Maritza Vega-Rivera
- Laboratory of Neuropsychopharmacology, Division of Neurosciences, National Institute of Psychiatry “Ramón de la Fuente Muñiz”, Calzada Mexico-Xochimilco No. 101, Mexico City C.P. 14370, Mexico;
- Correspondence: (G.B.R.-R.); (N.M.V.-R.)
| |
Collapse
|
17
|
D’Andrea G, Ceccarelli M, Bernini R, Clemente M, Santi L, Caruso C, Micheli L, Tirone F. Hydroxytyrosol stimulates neurogenesis in aged dentate gyrus by enhancing stem and progenitor cell proliferation and neuron survival. FASEB J 2020; 34:4512-4526. [DOI: 10.1096/fj.201902643r] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2019] [Revised: 12/31/2019] [Accepted: 01/16/2020] [Indexed: 12/18/2022]
Affiliation(s)
- Giorgio D’Andrea
- Institute of Biochemistry and Cell Biology National Research Council (IBBC‐CNR) Monterotondo, Rome Italy
- Department of Ecological and Biological Sciences University of Tuscia Viterbo Italy
| | - Manuela Ceccarelli
- Institute of Biochemistry and Cell Biology National Research Council (IBBC‐CNR) Monterotondo, Rome Italy
| | - Roberta Bernini
- Department of Agriculture and Forest Sciences (DAFNE) University of Tuscia Viterbo Italy
| | - Mariangela Clemente
- Department of Agriculture and Forest Sciences (DAFNE) University of Tuscia Viterbo Italy
| | - Luca Santi
- Department of Agriculture and Forest Sciences (DAFNE) University of Tuscia Viterbo Italy
| | - Carla Caruso
- Department of Ecological and Biological Sciences University of Tuscia Viterbo Italy
| | - Laura Micheli
- Institute of Biochemistry and Cell Biology National Research Council (IBBC‐CNR) Monterotondo, Rome Italy
| | - Felice Tirone
- Institute of Biochemistry and Cell Biology National Research Council (IBBC‐CNR) Monterotondo, Rome Italy
| |
Collapse
|
18
|
Umschweif G, Greengard P, Sagi Y. The dentate gyrus in depression. Eur J Neurosci 2019; 53:39-64. [DOI: 10.1111/ejn.14640] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2019] [Revised: 11/05/2019] [Accepted: 11/28/2019] [Indexed: 12/19/2022]
Affiliation(s)
- Gali Umschweif
- Laboratory for Molecular and Cellular Neuroscience Rockefeller University New York NY USA
| | - Paul Greengard
- Laboratory for Molecular and Cellular Neuroscience Rockefeller University New York NY USA
| | - Yotam Sagi
- Laboratory for Molecular and Cellular Neuroscience Rockefeller University New York NY USA
| |
Collapse
|
19
|
Deficiency in Androgen Receptor Aggravates the Depressive-Like Behaviors in Chronic Mild Stress Model of Depression. Cells 2019; 8:cells8091021. [PMID: 31480771 PMCID: PMC6769639 DOI: 10.3390/cells8091021] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 08/22/2019] [Accepted: 08/28/2019] [Indexed: 02/06/2023] Open
Abstract
While androgen receptor (AR) and stress may influence the development of the major depressive disorder (MDD), the detailed relationship, however, remains unclear. Here we found loss of AR accelerated development of depressive-like behaviors in mice under chronic mild stress (CMS). Mechanism dissection indicated that AR might function via altering the expression of miR-204-5p to modulate the brain-derived neurotrophic factor (BDNF) expression to influence the depressive-like behaviors in the mice under the CMS. Adding the antiandrogen flutamide with the stress hormone corticosterone can additively decrease BDNF mRNA in mouse hippocampus mHippoE-14 cells, which can then be reversed via down-regulating the miR-204-5p expression. Importantly, targeting this newly identified AR-mediated miR-204-5p/BDNF/AKT/MAPK signaling with small molecules including 7,8-DHF and fluoxetine, all led to alter the depressive-like behavior in AR knockout mice under CMS exposure. Together, results from these preclinical studies conclude that decreased AR may accelerate the stress-induced MDD via altering miR-204-5p/BDNF/AKT/MAPK signaling, and targeting this newly identified signaling may help in the development of better therapeutic approaches to reduce the development of MDD.
Collapse
|
20
|
Kandasamy M, Yesudhas A, Poornimai Abirami GP, Radhakrishnan RK, Roshan SA, Johnson E, Ravichandran VR, Biswas A, Shanmugaapriya S, Anusuyadevi M, Aigner L. Genetic reprogramming of somatic cells into neuroblasts through a co-induction of the doublecortin gene along the Yamanaka factors: A promising approach to model neuroregenerative disorders. Med Hypotheses 2019; 127:105-111. [PMID: 31088631 DOI: 10.1016/j.mehy.2019.04.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 03/28/2019] [Accepted: 04/11/2019] [Indexed: 12/25/2022]
Abstract
Neural stem cell (NSC) mediated adult neurogenesis represents the regenerative plasticity of the brain. The functionality of the neurogenic process appears to be operated by neuroblasts, the multipotent immature neuronal population of the adult brain. While neuroblasts have been realized to play a major role in synaptic remodeling and immunogenicity, neurodegenerative disorders have been characterized by failure in the terminal differentiation, maturation, integration and survival of newborn neuroblasts. Advancement in understanding the impaired neuroregenerative process along the neuropathological conditions has currently been limited by lack of an appropriate experimental model of neuroblasts. The genetic reprogramming of somatic cells into pluripotent state offers a potential strategy for the experimental modeling of brain disorders. Thus, the induced pluripotent stem cell (iPSC) based direct reprogramming of somatic cells into neuroblasts would represent a potential tool to understand the regenerative biology of the adult brain. Therefore, this concise article discusses the significance of iPSCs, the functional roles of neuroblasts in the adult brain and provides a research hypothesis for the direct reprogramming of somatic cells into neuroblasts through the co-induction of a potential proneurogenic marker, the doublecortin (DCX) gene along with the Yamanaka factors. The proposed cellular model of adult neurogenesis may provide us with further insights into neuropathogenesis of many neurodegenerative disorders and will provide a potential experimental platform for diagnostic, drug discovery and regenerative therapeutic strategies.
Collapse
Affiliation(s)
- Mahesh Kandasamy
- Laboratory of Stem Cells and Neuroregeneration, Department of Animal Science, School of Life Sciences, Bharathidasan University, Tiruchirappalli, Tamil Nadu, India; School of Life Sciences, Bharathidasan University, Tiruchirappalli, Tamil Nadu, India; UGC-Faculty Recharge Programme (UGC-FRP), University Grants Commission, New Delhi, India.
| | - Ajisha Yesudhas
- Laboratory of Stem Cells and Neuroregeneration, Department of Animal Science, School of Life Sciences, Bharathidasan University, Tiruchirappalli, Tamil Nadu, India
| | - G P Poornimai Abirami
- School of Life Sciences, Bharathidasan University, Tiruchirappalli, Tamil Nadu, India
| | - Risna Kanjirassery Radhakrishnan
- Laboratory of Stem Cells and Neuroregeneration, Department of Animal Science, School of Life Sciences, Bharathidasan University, Tiruchirappalli, Tamil Nadu, India
| | - Syed Aasish Roshan
- Molecular Gerontology Laboratory, Department of Biochemistry, School of Life Sciences, Bharathidasan University, Tiruchirappalli, Tamil Nadu, India
| | - Esther Johnson
- Laboratory of Stem Cells and Neuroregeneration, Department of Animal Science, School of Life Sciences, Bharathidasan University, Tiruchirappalli, Tamil Nadu, India
| | - Vijaya Roobini Ravichandran
- Laboratory of Stem Cells and Neuroregeneration, Department of Animal Science, School of Life Sciences, Bharathidasan University, Tiruchirappalli, Tamil Nadu, India
| | - Abir Biswas
- Molecular Gerontology Laboratory, Department of Biochemistry, School of Life Sciences, Bharathidasan University, Tiruchirappalli, Tamil Nadu, India
| | | | - Muthuswamy Anusuyadevi
- School of Life Sciences, Bharathidasan University, Tiruchirappalli, Tamil Nadu, India; Molecular Gerontology Laboratory, Department of Biochemistry, School of Life Sciences, Bharathidasan University, Tiruchirappalli, Tamil Nadu, India
| | - Ludwig Aigner
- Institute of Molecular Regenerative Medicine, Paracelsus Medical University, Salzburg, Austria; Spinal Cord Injury and Tissue Regeneration Center, Salzburg, Paracelsus Medical University, Salzburg, Austria.
| |
Collapse
|
21
|
Kirschen GW, Ge S. Young at heart: Insights into hippocampal neurogenesis in the aged brain. Behav Brain Res 2019; 369:111934. [PMID: 31054278 DOI: 10.1016/j.bbr.2019.111934] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Revised: 04/09/2019] [Accepted: 04/30/2019] [Indexed: 12/24/2022]
Abstract
While the existence and importance of adult hippocampal neurogenesis in young adult rodents has been well-established, such qualities in aged animals and humans have remained poorly understood. Most evidence in humans has come from hippocampal volumetric changes that provide no direct proof of new neurons in adulthood. Here, we review the basic neurobiological evidence for adult hippocampal neurogenesis in the aged brain of experimental animals with short and long lifespans, and humans. The rate of cell cycling and addition of new hippocampal neurons to the existing hippocampal circuit undoubtedly decreases with age. Yet, neural stem/progenitor cells that persist into senescence may activate and produce a substantial number of functional new neurons that exhibit enhanced survival and integration given the right set of conditions. There thus exists remarkable potential for newly-generated neurons in the senescent hippocampus to make important circuit- and behavioral-level contributions, which may serve as a target for future therapeutics.
Collapse
Affiliation(s)
- Gregory W Kirschen
- Medical Scientist Training Program (MSTP), Renaissance School of Medicine at Stony Brook University, 101 Nicolls Rd, Stony Brook, NY 11794, United States.
| | - Shaoyu Ge
- Department of Neurobiology and Behavior, Stony Brook University, Stony Brook, NY 11794, United States
| |
Collapse
|
22
|
Villas Boas GR, Boerngen de Lacerda R, Paes MM, Gubert P, Almeida WLDC, Rescia VC, de Carvalho PMG, de Carvalho AAV, Oesterreich SA. Molecular aspects of depression: A review from neurobiology to treatment. Eur J Pharmacol 2019; 851:99-121. [PMID: 30776369 DOI: 10.1016/j.ejphar.2019.02.024] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Revised: 02/14/2019] [Accepted: 02/14/2019] [Indexed: 12/14/2022]
Abstract
Major depressive disorder (MDD), also known as unipolar depression, is one of the leading causes of disability and disease worldwide. The signs and symptoms are low self‑esteem, anhedonia, feeling of worthlessness, sense of rejection and guilt, suicidal thoughts, among others. This review focuses on studies with molecular-based approaches involving MDD to obtain an integrated, more detailed and comprehensive view of the brain changes produced by this disorder and its treatment and how the Central Nervous System (CNS) produces neuroplasticity to orchestrate adaptive defensive behaviors. This article integrates affective neuroscience, psychopharmacology, neuroanatomy and molecular biology data. In addition, there are two problems with current MDD treatments, namely: 1) Low rates of responsiveness to antidepressants and too slow onset of therapeutic effect; 2) Increased stress vulnerability and autonomy, which reduces the responses of currently available treatments. In the present review, we encourage the prospection of new bioactive agents for the development of treatments with post-transduction mechanisms, neurogenesis and pharmacogenetics inducers that bring greater benefits, with reduced risks and maximized access to patients, stimulating the field of research on mood disorders in order to use the potential of preclinical studies. For this purpose, improved animal models that incorporate the molecular and anatomical tools currently available can be applied. Besides, we encourage the study of drugs that do not present "classical application" as antidepressants, (e.g., the dissociative anesthetic ketamine and dextromethorphan) and drugs that have dual action mechanisms since they represent potential targets for novel drug development more useful for the treatment of MDD.
Collapse
Affiliation(s)
- Gustavo Roberto Villas Boas
- Research Group on Development of Pharmaceutical Products (P&DProFar), Center for Biological and Health Sciences, Federal University of Western Bahia, Rua Bertioga, 892, Morada Nobre II, CEP 47810-059, Barreiras, Bahia, Brazil; Faculty of Health Sciences, Federal University of Grande Dourados, Dourados Rodovia Dourados, Itahum Km 12, Cidade Universitaria, Caixa. postal 364, CEP 79804-970, Dourados, Mato Grosso do Sul, Brazil.
| | - Roseli Boerngen de Lacerda
- Department of Pharmacology of the Biological Sciences Center, Federal University of Paraná, Jardim das Américas, Caixa. postal 19031, CEP 81531-990, Curitiba, Paraná, Brazil.
| | - Marina Meirelles Paes
- Research Group on Development of Pharmaceutical Products (P&DProFar), Center for Biological and Health Sciences, Federal University of Western Bahia, Rua Bertioga, 892, Morada Nobre II, CEP 47810-059, Barreiras, Bahia, Brazil.
| | - Priscila Gubert
- Center for Biological and Health Sciences, Federal University of Western Bahia, Rua Bertioga, 892, Morada Nobre II, CEP 47810-059, Barreiras, Bahia, Brazil.
| | - Wagner Luis da Cruz Almeida
- Research Group on Development of Pharmaceutical Products (P&DProFar), Center for Biological and Health Sciences, Federal University of Western Bahia, Rua Bertioga, 892, Morada Nobre II, CEP 47810-059, Barreiras, Bahia, Brazil.
| | - Vanessa Cristina Rescia
- Research Group on Development of Pharmaceutical Products (P&DProFar), Center for Biological and Health Sciences, Federal University of Western Bahia, Rua Bertioga, 892, Morada Nobre II, CEP 47810-059, Barreiras, Bahia, Brazil.
| | - Pablinny Moreira Galdino de Carvalho
- Center for Biological and Health Sciences, Federal University of Western Bahia, Rua Bertioga, 892, Morada Nobre II, CEP 47810-059, Barreiras, Bahia, Brazil.
| | - Adryano Augustto Valladao de Carvalho
- Center for Biological and Health Sciences, Federal University of Western Bahia, Rua Bertioga, 892, Morada Nobre II, CEP 47810-059, Barreiras, Bahia, Brazil.
| | - Silvia Aparecida Oesterreich
- Faculty of Health Sciences, Federal University of Grande Dourados, Dourados Rodovia Dourados, Itahum Km 12, Cidade Universitaria, Caixa. postal 364, CEP 79804-970, Dourados, Mato Grosso do Sul, Brazil.
| |
Collapse
|
23
|
Micheli L, D'Andrea G, Ceccarelli M, Ferri A, Scardigli R, Tirone F. p16Ink4a Prevents the Activation of Aged Quiescent Dentate Gyrus Stem Cells by Physical Exercise. Front Cell Neurosci 2019; 13:10. [PMID: 30792628 PMCID: PMC6374340 DOI: 10.3389/fncel.2019.00010] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Accepted: 01/11/2019] [Indexed: 12/19/2022] Open
Abstract
In the neurogenic niches—the dentate gyrus of the hippocampus and the subventricular zone (SVZ) adjacent to lateral ventricles—stem cells continue to divide during adulthood, generating progenitor cells and new neurons, and to self-renew, thus maintaining the stem cell pool. During aging, the numbers of stem/progenitor cells in the neurogenic niches are reduced. The preservation of the neurogenic pool is committed to a number of antiproliferative genes, with the role of maintaining the quiescence of neural cells. The cyclin-dependent kinase inhibitor p16Ink4a, whose expression increases with age, controls the expansion of SVZ aging stem cells, since in mice its deficiency prevents the decline of neurogenesis in SVZ. No change of neurogenesis is however observed in the p16Ink4a-null dentate gyrus. Here, we hypothesized that p16Ink4a plays a role as a regulator of the self-renewal of the stem cell pool also in the dentate gyrus, and to test this possibility we stimulated the dentate gyrus neural cells of p16Ink4a-null aging mice with physical exercise, a powerful neurogenic activator. We observed that running highly induced the generation of new stem cells in the p16Ink4a-null dentate gyrus, forcing them to exit from quiescence. Stem cells, notably, are not induced to proliferate by running in wild-type (WT) mice. Moreover, p16Ink4a-null progenitor cells were increased by running significantly above the number observed in WT mice. The new stem and progenitor cells generated new neurons, and continued to actively proliferate in p16Ink4a-null mice longer than in the WT after cessation of exercise. Thus, p16Ink4a prevents aging dentate gyrus stem cells from being activated by exercise. Therefore, p16Ink4a may play a role in the maintenance of dentate gyrus stem cells after stimulus, by keeping a reserve of their self-renewal capacity during aging.
Collapse
Affiliation(s)
- Laura Micheli
- Institute of Cell Biology and Neurobiology, National Research Council, Foundation Santa Lucia, Rome, Italy
| | - Giorgio D'Andrea
- Institute of Cell Biology and Neurobiology, National Research Council, Foundation Santa Lucia, Rome, Italy.,Department of Ecological and Biological Sciences, University of Tuscia, Viterbo, Italy
| | - Manuela Ceccarelli
- Institute of Cell Biology and Neurobiology, National Research Council, Foundation Santa Lucia, Rome, Italy
| | - Alessandra Ferri
- Institute of Cell Biology and Neurobiology, National Research Council, Foundation Santa Lucia, Rome, Italy
| | - Raffaella Scardigli
- Institute of Translational Pharmacology (IFT), National Research Council, Rome, Italy
| | - Felice Tirone
- Institute of Cell Biology and Neurobiology, National Research Council, Foundation Santa Lucia, Rome, Italy
| |
Collapse
|
24
|
Ramírez-Rodríguez GB, Olvera-Hernández S, Vega-Rivera NM, Ortiz-López L. Melatonin Influences Structural Plasticity in the Axons of Granule Cells in the Dentate Gyrus of Balb/C Mice. Int J Mol Sci 2018; 20:ijms20010073. [PMID: 30585191 PMCID: PMC6337618 DOI: 10.3390/ijms20010073] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Revised: 12/19/2018] [Accepted: 12/20/2018] [Indexed: 02/07/2023] Open
Abstract
Melatonin, the main product synthesized by the pineal gland, acts as a regulator of the generation of new neurons in the dentate gyrus (DG). Newborn neurons buffer the deleterious effects of stress and are involved in learning and memory processes. Furthermore, melatonin, through the regulation of the cytoskeleton, favors dendrite maturation of newborn neurons. Moreover, newborn neurons send their axons via the mossy fiber tract to Cornu Ammonis 3 (CA3) region to form synapses with pyramidal neurons. Thus, axons of newborn cells contribute to the mossy fiber projection and their plasticity correlates with better performance in several behavioral tasks. Thus, in this study, we analyzed the impact of exogenous melatonin (8 mg/kg) administered daily for one- or six-months on the structural plasticity of infrapyramidal- and suprapyramidal mossy fiber projection of granule cells in the DG in male Balb/C mice. We analyzed the mossy fiber projection through the staining of calbindin, that is a calcium-binding protein localized in dendrites and axons. We first found an increase in the number of calbindin-positive cells in the granular cell layer in the DG (11%, 33%) after treatment. Futhermore, we found an increase in the volume of suprapyramidal (>135%, 59%) and infrapyramidal (>128%, 36%) mossy fiber projection of granule neurons in the DG after treatment. We also found an increase in the volume of CA3 region (>146%, 33%) after treatment, suggesting that melatonin modulates the structural plasticity of the mossy fiber projection to establish functional synapses in the hippocampus. Together, the data suggest that, in addition to the previously reported effects of melatonin on the generation of new neurons and its antidepressant like effects, melatonin also modulates the structural plasticity of axons in granule cells in the DG.
Collapse
Affiliation(s)
- Gerardo Bernabé Ramírez-Rodríguez
- Laboratorio de Neurogenesis, Subidrección de Investigaciones Clínicas, Instituto Nacional de Psiquiatría "Ramón de la Fuente Muñiz", Calzada México-Xochimilco 101, Col. San Lorenzo Huipulco, Tlalpan, México City C.P. 14370, México.
| | - Sandra Olvera-Hernández
- Laboratorio de Neurogenesis, Subidrección de Investigaciones Clínicas, Instituto Nacional de Psiquiatría "Ramón de la Fuente Muñiz", Calzada México-Xochimilco 101, Col. San Lorenzo Huipulco, Tlalpan, México City C.P. 14370, México.
| | - Nelly Maritza Vega-Rivera
- Laboratorio de Neuropsicofarmacología, Dirección de Neurociencias, Instituto Nacional de Psiquiatría "Ramón de la Fuente Muñiz", Calzada México-Xochimilco 101, Col. San Lorenzo Huipulco, Tlalpan, México City C.P. 14370, México.
| | - Leonardo Ortiz-López
- Laboratorio de Neurogenesis, Subidrección de Investigaciones Clínicas, Instituto Nacional de Psiquiatría "Ramón de la Fuente Muñiz", Calzada México-Xochimilco 101, Col. San Lorenzo Huipulco, Tlalpan, México City C.P. 14370, México.
| |
Collapse
|
25
|
Depression and adult neurogenesis: Positive effects of the antidepressant fluoxetine and of physical exercise. Brain Res Bull 2018; 143:181-193. [PMID: 30236533 DOI: 10.1016/j.brainresbull.2018.09.002] [Citation(s) in RCA: 178] [Impact Index Per Article: 25.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Revised: 09/03/2018] [Accepted: 09/11/2018] [Indexed: 12/11/2022]
Abstract
Of wide interest for health is the relation existing between depression, a very common psychological illness, accompanied by anxiety and reduced ability to concentrate, and adult neurogenesis. We will focus on two neurogenic stimuli, fluoxetine and physical exercise, both endowed with the ability to activate adult neurogenesis in the dentate gyrus of the hippocampus, known to be required for learning and memory, and both able to counteract depression. Fluoxetine belongs to the class of selective serotonin reuptake inhibitor (SSRI) antidepressants, which represent the most used pharmacological therapy; physical exercise has also been shown to effectively counteract depression symptoms in rodents as well as in humans. While there is evidence that the antidepressant effect of fluoxetine requires its pro-neurogenic action, exerted by promoting proliferation, differentiation and survival of progenitor cells of the hippocampus, on the other hand fluoxetine exerts also neurogenesis-independent antidepressant effects by influencing the plasticity of the new neurons generated. Similarly, the antidepressant action of running also correlates with an increase of hippocampal neurogenesis and plasticity, although the gene pathways involved are only partially coincident with those of fluoxetine, such as those involved in serotonin metabolism and synapse formation. We further discuss how extra-neurogenic actions are also suggested by the fact that, unlike running, fluoxetine is unable to stimulate neurogenesis during aging, but still displays antidepressant effects. Moreover, in specific conditions, fluoxetine or running activate not only progenitor but also stem cells, which normally are not stimulated; this fact reveals how stem cells have a long-term, hidden ability to self-renew and, more generally, that neurogenesis is subject to complex controls that may play a role in depression, such as the type of neurogenic stimulus or the state of the local niche. Finally, we discuss how fluoxetine or running are effective in counteracting depression originated from stress or neurodegenerative diseases.
Collapse
|
26
|
Micheli L, Ceccarelli M, D'Andrea G, Costanzi M, Giacovazzo G, Coccurello R, Caruso C, Tirone F. Fluoxetine or Sox2 reactivate proliferation-defective stem and progenitor cells of the adult and aged dentate gyrus. Neuropharmacology 2018; 141:316-330. [PMID: 30142401 DOI: 10.1016/j.neuropharm.2018.08.023] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2018] [Revised: 08/06/2018] [Accepted: 08/20/2018] [Indexed: 01/19/2023]
Abstract
The dentate gyrus of the hippocampus and the subventricular zone are neurogenic niches where the production of new neurons from glia-like stem cells continues throughout adult life. It is not clear whether the pool of stem cells is fated to be exhausted or is conserved until old age. We observed that the antiproliferative gene Btg1 maintains the quiescence of stem cells, and its ablation causes an increase of stem/progenitor cells proliferation in neonatal mice followed by progressive loss of proliferation during adulthood. Fluoxetine is an antidepressant, which exerts a powerful neurogenic effect on dentate gyrus progenitor cells, but is ineffective on stem cells. Here we show that adult dentate gyrus stem cells in the Btg1 knockout mice, with reduced self-renewal and proliferative capability, can be reactivated by fluoxetine, which increases their number greatly above the level of control or fluoxetine-treated wild-type mice. The increase of mitotic index above wild-type in Btg1 knockout fluoxetine-treated stem cells indicates that fluoxetine forces quiescent stem cells to enter the cycle. Stem cell proliferation undergoes continuous reactivation until fluoxetine is administered. Remarkably, fluoxetine reactivates proliferation-defective stem cells also in aged Btg1 knockout mice (15-month-old), an effect absent in wild-type aged mice. Moreover, overexpression of Sox2 retrovirally transduced in Btg1 knockout dentate gyrus cells significantly increases the number of neuroblasts, indicating that Sox2 is able to promote the self-renewal of proliferation-defective stem cells. Overall, the deletion of an antiproliferative gene, such as Btg1, reveals that dentate gyrus stem cells retain a hidden plasticity for self-renewal also in old age, in agreement with a model of permanent self-renewal.
Collapse
Affiliation(s)
- Laura Micheli
- Institute of Cell Biology and Neurobiology, National Research Council, Fondazione S.Lucia, Via Del Fosso di Fiorano 64, 00143, Rome, Italy.
| | - Manuela Ceccarelli
- Institute of Cell Biology and Neurobiology, National Research Council, Fondazione S.Lucia, Via Del Fosso di Fiorano 64, 00143, Rome, Italy.
| | - Giorgio D'Andrea
- Institute of Cell Biology and Neurobiology, National Research Council, Fondazione S.Lucia, Via Del Fosso di Fiorano 64, 00143, Rome, Italy; Department of Ecological and Biological Sciences, University of Tuscia, Largo Dell'Università S.n.c., 01100, Viterbo, Italy.
| | - Marco Costanzi
- Department of Human Sciences, LUMSA University, Piazza Delle Vaschette 101, 00193, Rome, Italy.
| | - Giacomo Giacovazzo
- Institute of Cell Biology and Neurobiology, National Research Council, Fondazione S.Lucia, Via Del Fosso di Fiorano 64, 00143, Rome, Italy.
| | - Roberto Coccurello
- Institute of Cell Biology and Neurobiology, National Research Council, Fondazione S.Lucia, Via Del Fosso di Fiorano 64, 00143, Rome, Italy.
| | - Carla Caruso
- Department of Ecological and Biological Sciences, University of Tuscia, Largo Dell'Università S.n.c., 01100, Viterbo, Italy.
| | - Felice Tirone
- Institute of Cell Biology and Neurobiology, National Research Council, Fondazione S.Lucia, Via Del Fosso di Fiorano 64, 00143, Rome, Italy.
| |
Collapse
|
27
|
Sun Y, Sun X, Qu H, Zhao S, Xiao T, Zhao C. Neuroplasticity and behavioral effects of fluoxetine after experimental stroke. Restor Neurol Neurosci 2018; 35:457-468. [PMID: 28854520 DOI: 10.3233/rnn-170725] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
The brain can undergo self-repair and has the ability to compensate for functions lost after a stroke. The plasticity of the ischemic brain is influenced by several factors including aging and pharmacotherapy. Fluoxetine is an antidepressant which enhances serotonergic neurotransmission through selective inhibition of neuronal reuptake of serotonin. In clinical practice, fluoxetine alleviates the symptoms of post-stroke depression (PSD), helps motor recovery in stroke patients. In animal experiments, chronic administration of fluoxetine induces increased excitability of mature granule cells (GCs), enhancing axonal and dendritic reorganization, as well as promoting neurogenesis or angiogenesis in the dentate gurus (DG), but the effect of fluoxetine in the subventricular zone (SVZ) remains controversial. Meanwhile, chronic treatment with fluoxetine did not reverse age-dependent suppression of proliferation cells in the DG. Interestingly, although fluoxetine has been found to enhance neurogenesis in the DG in stroke rats, this property is not consistent with the behavioral recovery. More studies into this issue will be required to reveal how to translate enhanced neuronal plasticity into behavioral benefits. This review provides an update of the current knowledge about the neurogenesis and the fate of the newly generated cells after the use of fluoxetine, as well as its ability to promote a behavioral recovery after stroke in clinical and experimental results and attempts to define the therapeutic properties of fluoxetine in regenerative neuroscience.
Collapse
Affiliation(s)
- Yefei Sun
- Gastrointestinal Surgery, The First Hospital of China Medical University, Shenyang, China
| | - Xiaoyu Sun
- Department of Neurology, The People's Hospital of Liaoning Province, Shenyang, China
| | - Huiling Qu
- Department of Neurology, The People's Hospital of Liaoning Province, Shenyang, China
| | - Shanshan Zhao
- Neurology, The First Hospital of China Medical University, Shenyang, China
| | - Ting Xiao
- Dermatology, The First Hospital of China Medical University, Shenyang, China.,Key Laboratory of Immunodermatology, Ministry of Health, Ministry of Education, Shenyang, China
| | - Chuansheng Zhao
- Neurology, The First Hospital of China Medical University, Shenyang, China
| |
Collapse
|
28
|
Dario MFR, Sara T, Estela CO, Margarita PM, Guillermo ET, Fernando RDF, Javier SL, Carmen P. Stress, Depression, Resilience and Ageing: A Role for the LPA-LPA1 Pathway. Curr Neuropharmacol 2018; 16:271-283. [PMID: 28699486 PMCID: PMC5843979 DOI: 10.2174/1570159x15666170710200352] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Revised: 05/26/2017] [Accepted: 06/30/2017] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND Chronic stress affects health and the quality of life, with its effects being particularly relevant in ageing due to the psychobiological characteristics of this population. However, while some people develop psychiatric disorders, especially depression, others seem very capable of dealing with adversity. There is no doubt that along with the identification of neurobiological mechanisms involved in developing depression, discovering which factors are involved in positive adaptation under circumstances of extreme difficulty will be crucial for promoting resilience. METHODS Here, we review recent work in our laboratory, using an animal model lacking the LPA1 receptor, together with pharmacological studies and clinical evidence for the possible participation of the LPA1 receptor in mood and resilience to stress. RESULTS Substantial evidence has shown that the LPA1 receptor is involved in emotional regulation and in coping responses to chronic stress, which, if dysfunctional, may induce vulnerability to stress and predisposition to the development of depression. Given that there is commonality of mechanisms between those involved in negative consequences of stress and in ageing, this is not surprising, considering that the LPA1 receptor may be involved in coping with adversity during ageing. CONCLUSION Alterations in this receptor may be a susceptibility factor for the presence of depression and cognitive deficits in the elderly population. However, because this is only a promising hypothesis based on previous data, future studies should focus on the involvement of the LPA-LPA1 pathway in coping with stress and resilience in ageing.
Collapse
Affiliation(s)
- Moreno-Fernández Román Dario
- Departamento de Psicobiología y Metodología de las CC, Instituto de Investigación Biomédica de Málaga (IBIMA), Universidad de Málaga; Málaga 29071, Spain
| | - Tabbai Sara
- Departamento de Psicobiología y Metodología de las CC, Instituto de Investigación Biomédica de Málaga (IBIMA), Universidad de Málaga; Málaga 29071, Spain
| | - Castilla-Ortega Estela
- Unidad de Gestión Clínica de Salud Mental, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de Málaga; Málaga 29010, Spain
| | - Pérez-Martín Margarita
- Departamento de Biología Celular, Genética y Fisiología, Instituto de Investigación Biomédica de Málaga (IBIMA), Universidad de
Málaga; Málaga 29071, Spain
| | - Estivill-Torrús Guillermo
- Unidad de Gestión Clínica de Neurociencias, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitarios de Málaga, Málaga, Spain
| | - Rodríguez de Fonseca Fernando
- Unidad de Gestión Clínica de Salud Mental, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de Málaga; Málaga 29010, Spain
| | - Santin Luis Javier
- Departamento de Psicobiología y Metodología de las CC, Instituto de Investigación Biomédica de Málaga (IBIMA), Universidad de Málaga; Málaga 29071, Spain
| | - Pedraza Carmen
- Departamento de Psicobiología y Metodología de las CC, Instituto de Investigación Biomédica de Málaga (IBIMA), Universidad de Málaga; Málaga 29071, Spain
| |
Collapse
|
29
|
Chronic Treatment with Fluoxetine or Clozapine of Socially Isolated Rats Prevents Subsector-Specific Reduction of Parvalbumin Immunoreactive Cells in the Hippocampus. Neuroscience 2018; 371:384-394. [DOI: 10.1016/j.neuroscience.2017.12.020] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Revised: 12/07/2017] [Accepted: 12/15/2017] [Indexed: 12/12/2022]
|
30
|
Sun DS, Gao LF, Jin L, Wu H, Wang Q, Zhou Y, Fan S, Jiang X, Ke D, Lei H, Wang JZ, Liu GP. Fluoxetine administration during adolescence attenuates cognitive and synaptic deficits in adult 3×TgAD mice. Neuropharmacology 2017; 126:200-212. [PMID: 28911966 DOI: 10.1016/j.neuropharm.2017.08.037] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Revised: 08/28/2017] [Accepted: 08/30/2017] [Indexed: 02/04/2023]
Abstract
Fluoxetine (FLX) has broad neurobiological functions and neuroprotective effects; however, the preventive effects of FLX on cognitive impairments in Alzheimer's disease (AD) have not been reported. Here, we studied whether adolescent administration of fluoxetine can prevent memory deficits in AD transgenic mice that harbour PS1m146v, APPswe and TauP301L mutations (3 × TgAD). FLX was applied through peritoneal injection to the mice at postnatal day 35 (p35) for 15 consecutive days, and the effects of FLX were observed at 6-month. We found that adolescent administration of FLX improved learning and memory abilities in 6-month-old 3 × TgAD mice. FLX exposure also increased the sizes of the hippocampal CA1, dentate gyrus (DG) and extensive cortex regions, with increased numbers of neurons and higher dendritic spine density. Meanwhile, the synaptic plasticity of neurons in the hippocampus was remodelled, and the expression levels of synaptic-related proteins were increased along with activation of the cyclic AMP response element-binding (CREB) protein/brain-derived neurotrophic factor (BDNF) signalling pathway. Finally, we found that FLX effectively prevented the increase of beta-amyloid (Aβ) levels. These data suggest that adolescent administration of the antidepressant drug FLX can efficiently preserve cognitive functions and improve pathologies in 3×Tg AD mice.
Collapse
Affiliation(s)
- Dong-Sheng Sun
- Department of Pathophysiology, School of Basic Medicine and the Collaborative Innovation Center for Brain Science, Key Laboratory of Ministry of Education of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Li-Feng Gao
- National Center for Magnetic Resonance in Wuhan, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Wuhan Institute of Physics and Mathematics, Chinese Academy of Sciences, Wuhan, China
| | - Li Jin
- Department of Pathophysiology, Henan Medical College, Zhengzhou 451191, China; Henan Key Laboratory of Degenerative Brain Disease, Henan Medical College, Zhengzhou 451191, China
| | - Hao Wu
- National Center for Magnetic Resonance in Wuhan, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Wuhan Institute of Physics and Mathematics, Chinese Academy of Sciences, Wuhan, China
| | - Qun Wang
- Department of Pathophysiology, School of Basic Medicine and the Collaborative Innovation Center for Brain Science, Key Laboratory of Ministry of Education of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - You Zhou
- Department of Neurosurgery, Tianyou Hospital, Wuhan University of Science and Technology, Wuhan, China
| | - Shuhao Fan
- Department of Neurosurgery, Tianyou Hospital, Wuhan University of Science and Technology, Wuhan, China
| | - Xia Jiang
- Department of Pathology, Hubei University of Chinese Medicine, Wuhan 430030, China
| | - Dan Ke
- Department of Pathophysiology, School of Basic Medicine and the Collaborative Innovation Center for Brain Science, Key Laboratory of Ministry of Education of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Hao Lei
- National Center for Magnetic Resonance in Wuhan, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Wuhan Institute of Physics and Mathematics, Chinese Academy of Sciences, Wuhan, China.
| | - Jian-Zhi Wang
- Department of Pathophysiology, School of Basic Medicine and the Collaborative Innovation Center for Brain Science, Key Laboratory of Ministry of Education of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Co-Innovation Center of Neuroregeneration, Nantong University, Nantong JS 226001, China.
| | - Gong-Ping Liu
- Department of Pathophysiology, School of Basic Medicine and the Collaborative Innovation Center for Brain Science, Key Laboratory of Ministry of Education of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Co-Innovation Center of Neuroregeneration, Nantong University, Nantong JS 226001, China.
| |
Collapse
|
31
|
Brandt MD, Krüger-Gerlach D, Hermann A, Meyer AK, Kim KS, Storch A. Early Postnatal but Not Late Adult Neurogenesis Is Impaired in the Pitx3-Mutant Animal Model of Parkinson's Disease. Front Neurosci 2017; 11:471. [PMID: 28883785 PMCID: PMC5573808 DOI: 10.3389/fnins.2017.00471] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2017] [Accepted: 08/09/2017] [Indexed: 01/10/2023] Open
Abstract
The generation of new neurons in the adult dentate gyrus has functional implications for hippocampal formation. Reduced hippocampal neurogenesis has been described in various animal models of hippocampal dysfunction such as dementia and depression, which are both common non-motor-symptoms of Parkinson's disease (PD). As dopamine plays an important role in regulating precursor cell proliferation, the loss of dopaminergic neurons in the substantia nigra (SN) in PD may be related to the reduced neurogenesis observed in the neurogenic regions of the adult brain: subventricular zone (SVZ) and dentate gyrus (DG). Here we examined adult hippocampal neurogenesis in the Pitx3-mutant mouse model of PD (aphakia mice), which phenotypically shows a selective embryonic degeneration of dopamine neurons within the SN and to a smaller extent in the ventral tegmental area (VTA). Proliferating cells were labeled with BrdU in aphakia mice and healthy controls from 3 to 42 weeks of age. Three weeks old mutant mice showed an 18% reduction of proliferating cells in the DG and of 26% in the SVZ. Not only proliferation but also the number of new neurons was impaired in young aphakia mice resulting in 33% less newborn cells 4 weeks after BrdU-labeling. Remarkably, however, the decline in the number of proliferating cells in the neurogenic regions vanished in older animals (8–42 weeks) indicating that aging masks the effect of dopamine depletion on adult neurogenesis. Region specific reduction in precursor cells proliferation correlated with the extent of dopaminergic degeneration in mesencephalic subregions (VTA and SN), which supports the theory of age- and region-dependent regulatory effects of dopaminergic projections. Physiological stimulation of adult neurogenesis by physical activity (wheel running) almost doubled the number of proliferating cells in the dentate gyrus of 8 weeks old aphakia mice to a number comparable to that of wild-type mice, abolishing the slight reduction of baseline neurogenesis at this age. The described age-dependent susceptibility of adult neurogenesis to PD-like dopaminergic degeneration and its responsiveness to physical activity might have implications for the understanding of the pathophysiology and treatment of non-motor symptoms in PD.
Collapse
Affiliation(s)
- Moritz D Brandt
- Department of Neurology, Technische Universität DresdenDresden, Germany.,German Center for Neurodegenerative Diseases DresdenDresden, Germany
| | | | - Andreas Hermann
- Department of Neurology, Technische Universität DresdenDresden, Germany.,German Center for Neurodegenerative Diseases DresdenDresden, Germany.,Center for Regenerative Therapies Dresden, Technische Universität DresdenDresden, Germany
| | - Anne K Meyer
- Department of Neurology, Technische Universität DresdenDresden, Germany
| | - Kwang-Soo Kim
- Molecular Neurobiology Laboratory, McLean Hospital/Harvard Medical SchoolBelmont, MA, United States
| | - Alexander Storch
- German Center for Neurodegenerative Diseases RostockRostock, Germany.,Department of Neurology, University of RostockRostock, Germany
| |
Collapse
|
32
|
Wu H, Cottingham C, Chen L, Wang H, Che P, Liu K, Wang Q. Age-dependent differential regulation of anxiety- and depression-related behaviors by neurabin and spinophilin. PLoS One 2017; 12:e0180638. [PMID: 28700667 PMCID: PMC5503268 DOI: 10.1371/journal.pone.0180638] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Accepted: 06/19/2017] [Indexed: 12/28/2022] Open
Abstract
Affective disorders impact nearly 10% of the adult population in the United States in a given year. Synaptic dysfunction has recently emerged as a key neurobiological mechanism underlying affective disorders such as anxiety and depression. In this study, we investigate the potential role of two synaptic scaffolding proteins, neurabin and spinophilin, in regulating anxiety- and depression-related behaviors at different ages using genetically deficient mice. Loss of the neurabin gene reduces anxiety-like behavior in the elevated zero maze in young adult mice (3-5 months old), but not in middle aged mice (11-13 months old), whereas loss of spinophilin decreases anxiety in middle-aged mice, but not in young adult mice. Neurabin knockout (KO) mice also show reduced immobility in the repeated force swim test (FST) at 3-5 months, but not 11-3 months, of age, compared to age- and strain-matched wild type (WT) controls. Conversely, spinophilin KO mice display a lower level of this behavioral despair than matched WT controls after repeated FST trials at the middle age (11-13 months) but not the young age (3-5 months). Together, these data indicate that, despite their structural similarities and overlapping function in regulating synaptic cytoskeleton, the two homologs neurabin and spinophilin play important yet distinct roles in the regulation of anxiety- and depression-like behaviors in an age-dependent manner. Our studies provide new insights into the complex neurobiology of affective disorders.
Collapse
Affiliation(s)
- Huiying Wu
- Ultrasonic Diagnosis Department, The Second Hospital of Jilin University, Changchun, Jilin, China
- Departments of Cell, Molecular and Developmental Biology, University of Alabama at Birmingham, Birmingham, AL, United States of America
| | - Christopher Cottingham
- Departments of Cell, Molecular and Developmental Biology, University of Alabama at Birmingham, Birmingham, AL, United States of America
- Department of Biology and Chemistry, Morehead State University, Morehead, KY, United States of America
| | - Liping Chen
- Departments of Cell, Molecular and Developmental Biology, University of Alabama at Birmingham, Birmingham, AL, United States of America
| | - Hongxia Wang
- Departments of Cell, Molecular and Developmental Biology, University of Alabama at Birmingham, Birmingham, AL, United States of America
| | - Pulin Che
- Departments of Cell, Molecular and Developmental Biology, University of Alabama at Birmingham, Birmingham, AL, United States of America
| | - Kexiang Liu
- Department of Cardiovascular Surgery, The Second Hospital of Jilin University, Changchun, Jilin, China
| | - Qin Wang
- Departments of Cell, Molecular and Developmental Biology, University of Alabama at Birmingham, Birmingham, AL, United States of America
- * E-mail:
| |
Collapse
|
33
|
Song NN, Huang Y, Yu X, Lang B, Ding YQ, Zhang L. Divergent Roles of Central Serotonin in Adult Hippocampal Neurogenesis. Front Cell Neurosci 2017; 11:185. [PMID: 28713247 PMCID: PMC5492328 DOI: 10.3389/fncel.2017.00185] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Accepted: 06/16/2017] [Indexed: 11/13/2022] Open
Abstract
The central serotonin (5-HT) system is the main target of selective serotonin reuptake inhibitors (SSRIs), the first-line antidepressants widely used in current general practice. One of the prominent features of chronic SSRI treatment in rodents is the enhanced adult neurogenesis in the hippocampus, which has been proposed to contribute to antidepressant effects. Therefore, tremendous effort has been made to decipher how central 5-HT regulates adult hippocampal neurogenesis. In this paper, we review how changes in the central serotonergic system alter adult hippocampal neurogenesis. We focus on data obtained from three categories of genetically engineered mouse models: (1) mice with altered central 5-HT levels from embryonic stages, (2) mice with deletion of 5-HT receptors from embryonic stages, and (3) mice with altered central 5-HT system exclusively in adulthood. These recent findings provide unique insights to interpret the multifaceted roles of central 5-HT on adult hippocampal neurogenesis and its associated effects on depression.
Collapse
Affiliation(s)
- Ning-Ning Song
- Key Laboratory of Arrhythmias, Ministry of Education, East Hospital, Tongji University School of MedicineShanghai, China.,Department of Anatomy and Neurobiology, Tongji University School of MedicineShanghai, China
| | - Ying Huang
- Key Laboratory of Arrhythmias, Ministry of Education, East Hospital, Tongji University School of MedicineShanghai, China.,Department of Anatomy and Neurobiology, Tongji University School of MedicineShanghai, China
| | - Xin Yu
- Key Laboratory of Arrhythmias, Ministry of Education, East Hospital, Tongji University School of MedicineShanghai, China.,Department of Anatomy and Neurobiology, Tongji University School of MedicineShanghai, China
| | - Bing Lang
- Key Laboratory of Arrhythmias, Ministry of Education, East Hospital, Tongji University School of MedicineShanghai, China.,Department of Anatomy and Neurobiology, Tongji University School of MedicineShanghai, China.,Mental Health Institute of the Second Xiangya Hospital, National Clinical Research Center on Mental Disorders, National Technology Institute on Mental Disorders, Key Laboratory of Psychiatry and Mental Health of Hunan Province, Central South UniversityChangsha, China
| | - Yu-Qiang Ding
- Key Laboratory of Arrhythmias, Ministry of Education, East Hospital, Tongji University School of MedicineShanghai, China.,Department of Anatomy and Neurobiology, Tongji University School of MedicineShanghai, China
| | - Lei Zhang
- Key Laboratory of Arrhythmias, Ministry of Education, East Hospital, Tongji University School of MedicineShanghai, China.,Department of Anatomy and Neurobiology, Tongji University School of MedicineShanghai, China
| |
Collapse
|
34
|
Hippocampal bone morphogenetic protein signaling mediates behavioral effects of antidepressant treatment. Mol Psychiatry 2017; 22:910-919. [PMID: 27698430 PMCID: PMC5378681 DOI: 10.1038/mp.2016.160] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2016] [Revised: 07/20/2016] [Accepted: 08/11/2016] [Indexed: 12/28/2022]
Abstract
Many antidepressants stimulate adult hippocampal neurogenesis, but the mechanisms by which they increase neurogenesis and modulate behavior are incompletely understood. Here we show that hippocampal bone morphogenetic protein (BMP) signaling is modulated by antidepressant treatment, and that the changes in BMP signaling mediate effects of antidepressant treatment on neural progenitor cell proliferation and behavior. Treatment with the selective serotonin reuptake inhibitor fluoxetine suppressed BMP signaling in the adult mouse hippocampus both by decreasing levels of BMP4 ligand and increasing production of the BMP inhibitor noggin. Increasing BMP signaling in the hippocampus via viral overexpression of BMP4 blocked the effects of fluoxetine on proliferation in the dentate gyrus and on depressive behavior. Conversely, inhibiting BMP signaling via viral overexpression of noggin in the hippocampus or infusion of noggin into the ventricles exerted antidepressant and anxiolytic activity along with an increase in hippocampal neurogenesis. Similarly, conditional genetic deletion of the type II BMP receptor in Ascl1-expressing cells promoted neurogenesis and reduced anxiety- and depression-like behaviors, suggesting that neural progenitor cells contribute to the effects of BMP signaling on affective behavior. These observations indicate that BMP signaling in the hippocampus regulates depressive behavior, and that decreasing BMP signaling may be required for the effects of some antidepressants. Thus BMP signaling is a new and powerful potential target for the treatment of depression.
Collapse
|
35
|
Slattery DA, Cryan JF. Modelling depression in animals: at the interface of reward and stress pathways. Psychopharmacology (Berl) 2017; 234:1451-1465. [PMID: 28224183 DOI: 10.1007/s00213-017-4552-6] [Citation(s) in RCA: 83] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Accepted: 01/27/2017] [Indexed: 12/13/2022]
Abstract
RATIONALE Despite substantial research efforts the aetiology of major depressive disorder (MDD) remains poorly understood, which is due in part to the heterogeneity of the disorder and the complexity of designing appropriate animal models. However, in the last few decades, a focus on the development of novel stress-based paradigms and a focus on using hedonic/anhedonic behaviour have led to renewed optimism in the use of animal models to assess aspects of MDD. OBJECTIVES Therefore, in this review article, dedicated to Athina Markou, we summarise the use of stress-based animal models for studying MDD in rodents and how reward-related readouts can be used to validate/assess the model and/or treatment. RESULTS We reveal the use and limitations of chronic stress paradigms, which we split into non-social (i.e. chronic mild stress), social (i.e. chronic social defeat) and drug-withdrawal paradigms for studying MDD and detail numerous reward-related readouts that are employed in preclinical research. Finally, we finish with a section regarding important factors to consider when using animal models. CONCLUSIONS One of the most consistent findings following chronic stress exposure in rodents is a disruption of the brain reward system, which can be easily assessed using sucrose, social interaction, food, drug of abuse or intracranial self-stimulation as a readout. Probing the underlying causes of such alterations is providing a greater understanding of the potential systems and processes that are disrupted in MDD.
Collapse
Affiliation(s)
- D A Slattery
- Laboratory of Translational Psychiatry, Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital Frankfurt, Heinrich-Hoffmann-Str. 10, 60528, Frankfurt, Germany.
| | - J F Cryan
- APC Microbiome Institute, Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| |
Collapse
|
36
|
Buga AM, Ciobanu O, Bădescu GM, Bogdan C, Weston R, Slevin M, Di Napoli M, Popa-Wagner A. Up-regulation of serotonin receptor 2B mRNA and protein in the peri-infarcted area of aged rats and stroke patients. Oncotarget 2017; 7:17415-30. [PMID: 27013593 PMCID: PMC4951222 DOI: 10.18632/oncotarget.8277] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Accepted: 03/14/2016] [Indexed: 12/14/2022] Open
Abstract
Despite the fact that a high proportion of elderly stroke patients develop mood disorders, the mechanisms underlying late-onset neuropsychiatric and neurocognitive symptoms have so far received little attention in the field of neurobiology. In rodents, aged animals display depressive symptoms following stroke, whereas young animals recover fairly well. This finding has prompted us to investigate the expression of serotonin receptors 2A and 2B, which are directly linked to depression, in the brains of aged and young rats following stroke. Although the development of the infarct was more rapid in aged rats in the first 3 days after stroke, by day 14 the cortical infarcts were similar in size in both age groups i.e. 45% of total cortical volume in young rats and 55.7% in aged rats. We also found that the expression of serotonin receptor type B mRNA was markedly increased in the perilesional area of aged rats as compared to the younger counterparts. Furthermore, histologically, HTR2B protein expression in degenerating neurons was closely associated with activated microglia both in aged rats and human subjects. Treatment with fluoxetine attenuated the expression of Htr2B mRNA, stimulated post-stroke neurogenesis in the subventricular zone and was associated with an improved anhedonic behavior and an increased activity in the forced swim test in aged animals. We hypothesize that HTR2B expression in the infarcted territory may render degenerating neurons susceptible to attack by activated microglia and thus aggravate the consequences of stroke.
Collapse
Affiliation(s)
- Ana-Maria Buga
- Department of Psychiatry and Psychotheraphy, University of Medicine Rostock, Rostock, Germany.,Center of Clinical and Experimental Medicine, University of Medicine and Pharmacy Craiova, Craiova, Romania
| | - Ovidiu Ciobanu
- Center of Clinical and Experimental Medicine, University of Medicine and Pharmacy Craiova, Craiova, Romania.,Vivantes Humboldt-Klinikum, Center for Affective Disorders, Berlin, Germany
| | - George Mihai Bădescu
- Psychiatry Clinical Hospital, University of Medicine and Pharmacy of Craiova, Craiova, Romania
| | - Catalin Bogdan
- Center of Clinical and Experimental Medicine, University of Medicine and Pharmacy Craiova, Craiova, Romania
| | - Ria Weston
- Department of Healthcare Science, Manchester Metropolitan University, Manchester, UK
| | - Mark Slevin
- Department of Healthcare Science, Manchester Metropolitan University, Manchester, UK
| | - Mario Di Napoli
- Neurological Service, San Camillo de' Lellis General Hospital, Rieti, Italy.,Neurological Section, SMDN-Center for Cardiovascular Medicine and Cerebrovascular Disease Prevention, Sulmona, L'Aquila, Italy
| | - Aurel Popa-Wagner
- Department of Psychiatry and Psychotheraphy, University of Medicine Rostock, Rostock, Germany
| |
Collapse
|
37
|
Velusamy T, Panneerselvam AS, Purushottam M, Anusuyadevi M, Pal PK, Jain S, Essa MM, Guillemin GJ, Kandasamy M. Protective Effect of Antioxidants on Neuronal Dysfunction and Plasticity in Huntington's Disease. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:3279061. [PMID: 28168008 PMCID: PMC5266860 DOI: 10.1155/2017/3279061] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Revised: 11/09/2016] [Accepted: 12/05/2016] [Indexed: 01/08/2023]
Abstract
Huntington's disease (HD) is characterised by movement disorders, cognitive impairments, and psychiatric problems. The abnormal generation of reactive oxygen species and the resulting oxidative stress-induced mitochondrial damage in neurons upon CAG mutations in the HTT gene have been hypothesized as the contributing factors of neurodegeneration in HD. The potential use of antioxidants against free radical toxicity has been an emerging field in the management of ageing and many neurodegenerative disorders. Neural stem cells derived adult neurogenesis represents the regenerative capacity of the adult brain. The process of adult neurogenesis has been implicated in the cognitive functions of the brain and is highly modulated positively by different factors including antioxidants. The supportive role of antioxidants to reduce the severity of HD via promoting the functional neurogenesis and neuroprotection in the pathological adult brain has great promise. This review comprehends the recent studies describing the therapeutic roles of antioxidants in HD and other neurologic disorders and highlights the scope of using antioxidants to promote adult neurogenesis in HD. It also advocates a new line of research to delineate the mechanisms by which antioxidants promote adult neurogenesis in HD.
Collapse
Affiliation(s)
- Thirunavukkarasu Velusamy
- Department of Biotechnology, Bharathiar University, Coimbatore, Tamil Nadu, India
- DBT Ramalingaswami Re-Entry Fellowship Programme, Department of Biotechnology (DBT), New Delhi, India
| | - Archana S. Panneerselvam
- Laboratory of Stem Cells and Neuroregeneration, Department of Animal Science, School of Life Sciences, Bharathidasan University, Tiruchirappalli, Tamil Nadu, India
| | - Meera Purushottam
- Department of Psychiatry, National Institute of Mental Health and Neurosciences (NIMHANS), Bangalore, Karnataka, India
| | - Muthuswamy Anusuyadevi
- Molecular Gerontology Laboratory, Department of Biochemistry, School of Life Sciences, Bharathidasan University, Tiruchirappalli, Tamil Nadu, India
| | - Pramod Kumar Pal
- Department of Neurology, National Institute of Mental Health and Neurosciences (NIMHANS), Bangalore, Karnataka, India
| | - Sanjeev Jain
- Department of Psychiatry, National Institute of Mental Health and Neurosciences (NIMHANS), Bangalore, Karnataka, India
| | - Musthafa Mohamed Essa
- Department of Food Science and Nutrition, CAMS, Sultan Qaboos University, Muscat, Oman
| | - Gilles J. Guillemin
- Neuroinflammation Group, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW, Australia
| | - Mahesh Kandasamy
- Laboratory of Stem Cells and Neuroregeneration, Department of Animal Science, School of Life Sciences, Bharathidasan University, Tiruchirappalli, Tamil Nadu, India
- UGC-Faculty Recharge Program (UGC-FRP), University Grant Commission, New Delhi, India
| |
Collapse
|
38
|
Adamczak J, Aswendt M, Kreutzer C, Rotheneichner P, Riou A, Selt M, Beyrau A, Uhlenküken U, Diedenhofen M, Nelles M, Aigner L, Couillard-Despres S, Hoehn M. Neurogenesis upregulation on the healthy hemisphere after stroke enhances compensation for age-dependent decrease of basal neurogenesis. Neurobiol Dis 2016; 99:47-57. [PMID: 28007584 DOI: 10.1016/j.nbd.2016.12.015] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Revised: 12/12/2016] [Accepted: 12/18/2016] [Indexed: 01/27/2023] Open
Abstract
Stroke is a leading cause of death and disability worldwide with no treatment for the chronic phase available. Interestingly, an endogenous repair program comprising inflammation and neurogenesis is known to modulate stroke outcome. Several studies have shown that neurogenesis decreases with age but the therapeutic importance of endogenous neurogenesis for recovery from cerebral diseases has been indicated as its ablation leads to stroke aggravation and worsened outcome. A detailed characterization of the neurogenic response after stroke related to ageing would help to develop novel and targeted therapies. In an innovative approach, we used the DCX-Luc mouse, a transgenic model expressing luciferase in doublecortin-positive neuroblasts, to monitor the neurogenic response following middle cerebral artery occlusion over three weeks in three age groups (2, 6, 12months) by optical imaging while the stroke lesion was monitored by quantitative MRI. The individual longitudinal and noninvasive time profiles provided exclusive insight into age-dependent decrease in basal neurogenesis and neurogenic upregulation in response to stroke which are not accessible by conventional BrdU-based measures of cell proliferation. For cortico-striatal strokes the maximal upregulation occurred at 4days post stroke followed by a continuous decrease to basal levels by three weeks post stroke. Older animals effectively compensated for reduced basal neurogenesis by an enhanced sensitivity to the cerebral lesion, resulting in upregulated neurogenesis levels approaching those measured in young mice. In middle aged and older mice, but not in the youngest ones, additional upregulation of neurogenesis was observed in the contralateral healthy hemisphere. This further substantiates the increased propensity of older brains to respond to lesion situation. Our results clearly support the therapeutic relevance of endogenous neurogenesis for stroke recovery and particularly in older brains.
Collapse
Affiliation(s)
- Joanna Adamczak
- In-vivo-NMR Laboratory, Max Planck Institute for Metabolism Research, Gleuelerstrasse 50, 50931 Cologne, Germany; Percuros B.V., Drienerlolaan 5-Zuidhorst, 7522 NB Enschede, The Netherlands
| | - Markus Aswendt
- In-vivo-NMR Laboratory, Max Planck Institute for Metabolism Research, Gleuelerstrasse 50, 50931 Cologne, Germany
| | - Christina Kreutzer
- Institute of Experimental Neuroregeneration, Spinal Cord Injury and Tissue Regeneration Center, Paracelsus Medical University Salzburg, Strubergasse 21, 5020 Salzburg, Austria; Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Austria
| | - Peter Rotheneichner
- Institute of Experimental Neuroregeneration, Spinal Cord Injury and Tissue Regeneration Center, Paracelsus Medical University Salzburg, Strubergasse 21, 5020 Salzburg, Austria; Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Austria
| | - Adrien Riou
- In-vivo-NMR Laboratory, Max Planck Institute for Metabolism Research, Gleuelerstrasse 50, 50931 Cologne, Germany
| | - Marion Selt
- In-vivo-NMR Laboratory, Max Planck Institute for Metabolism Research, Gleuelerstrasse 50, 50931 Cologne, Germany
| | - Andreas Beyrau
- In-vivo-NMR Laboratory, Max Planck Institute for Metabolism Research, Gleuelerstrasse 50, 50931 Cologne, Germany
| | - Ulla Uhlenküken
- In-vivo-NMR Laboratory, Max Planck Institute for Metabolism Research, Gleuelerstrasse 50, 50931 Cologne, Germany
| | - Michael Diedenhofen
- In-vivo-NMR Laboratory, Max Planck Institute for Metabolism Research, Gleuelerstrasse 50, 50931 Cologne, Germany
| | - Melanie Nelles
- In-vivo-NMR Laboratory, Max Planck Institute for Metabolism Research, Gleuelerstrasse 50, 50931 Cologne, Germany
| | - Ludwig Aigner
- Institute of Molecular Regenerative Medicine, Spinal Cord Injury and Tissue Regeneration Center, Paracelsus Medical University Salzburg, Strubergasse 21, 5020 Salzburg, Austria; Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Austria
| | - Sebastien Couillard-Despres
- Institute of Experimental Neuroregeneration, Spinal Cord Injury and Tissue Regeneration Center, Paracelsus Medical University Salzburg, Strubergasse 21, 5020 Salzburg, Austria; Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Austria
| | - Mathias Hoehn
- In-vivo-NMR Laboratory, Max Planck Institute for Metabolism Research, Gleuelerstrasse 50, 50931 Cologne, Germany; Department of Radiology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands; Percuros B.V., Drienerlolaan 5-Zuidhorst, 7522 NB Enschede, The Netherlands.
| |
Collapse
|
39
|
Mahmoud R, Wainwright SR, Chaiton JA, Lieblich SE, Galea LA. Ovarian hormones, but not fluoxetine, impart resilience within a chronic unpredictable stress model in middle-aged female rats. Neuropharmacology 2016; 107:278-293. [DOI: 10.1016/j.neuropharm.2016.01.033] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Revised: 01/18/2016] [Accepted: 01/22/2016] [Indexed: 01/30/2023]
|
40
|
Mitchell NC, Koek W, Daws LC. Antidepressant-like effects and basal immobility depend on age and serotonin transporter genotype. GENES BRAIN AND BEHAVIOR 2016; 14:543-549. [PMID: 26250357 DOI: 10.1111/gbb.12238] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/28/2015] [Revised: 07/28/2015] [Accepted: 07/29/2015] [Indexed: 01/31/2023]
Abstract
Monoamine uptake inhibitors are common treatments for depression; however, the therapeutic efficacy of these drugs varies widely. Two factors that are commonly linked to clinical outcome are age and serotonin transporter (SERT) genotype. Mouse models provide powerful tools to study consequences of age and genotype on antidepressant-like efficacy; however, to date, systematic studies of this nature are lacking. Here, we used the tail suspension test (TST), a preclinical assay for antidepressant efficacy, to gain insight into age and SERT genotype dependency of immobility time in the TST under control conditions (saline injection) and in response to the tricyclic antidepressant, desipramine (DMI). Immobility after saline injection in juvenile, adolescent, adult, mature adult and middle-aged mice (postnatal days 21, 28, 90, 210 and 300, respectively) significantly increased with age; however, the rate of increase was slower for SERT null (-/-) mice than for wild-type (+/+) or heterozygote (+/-) mice. Desipramine reduced immobility across ages and SERT genotypes. Middle-aged, but not adult, SERT(-/-) mice were significantly more sensitive to DMI than age-matched SERT(+/+) or SERT(+/-) mice. Desipramine was less potent in middle-aged SERT(+/+) and SERT(+/-) mice than in adult SERT(+/+) or SERT(+/-) mice. Regardless of age, DMI's maximal effects were greater in SERT(-/-) mice than in SERT(+/+) or SERT(+/-) mice. These results show that immobility time in the TST varies as a function of age and SERT genotype, underscoring the utility of the TST as a potential model to examine age- and SERT genotype-dependent influences on antidepressant response.
Collapse
Affiliation(s)
- Nathan C Mitchell
- Department of Physiology, University of Texas Health Science Center, San Antonio, TX, USA
| | - Wouter Koek
- Department of Psychiatry, University of Texas Health Science Center, San Antonio, TX, USA.,Department of Pharmacology, University of Texas Health Science Center, San Antonio, TX, USA
| | - Lynette C Daws
- Department of Physiology, University of Texas Health Science Center, San Antonio, TX, USA.,Department of Pharmacology, University of Texas Health Science Center, San Antonio, TX, USA
| |
Collapse
|
41
|
Tannenholz L, Hen R, Kheirbek MA. GluN2B-Containg NMDA Receptors on Adult-Born Granule Cells Contribute to the Antidepressant Action of Fluoxetine. Front Neurosci 2016; 10:242. [PMID: 27303260 PMCID: PMC4885883 DOI: 10.3389/fnins.2016.00242] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Accepted: 05/17/2016] [Indexed: 01/09/2023] Open
Abstract
Ablation of adult neurogenesis in mice has revealed that young adult-born granule cells (abGCs) are required for some of the behavioral responses to antidepressants (ADs), yet the mechanism by which abGCs contribute to AD action remains unknown. During their maturation process, these immature neurons exhibit unique properties that could underlie their ability to influence behavioral output. In particular, abGCs in the DG exhibit a period of heightened plasticity 4–6 weeks after birth that is mediated by GluN2B-expressing NMDA receptors. The functional contribution of this critical window to AD responsiveness is unclear. Here, we determined the behavioral and neurogenic responses to the AD fluoxetine (FLX) in mice lacking GluN2B-containing NMDA receptors in abGCs. We found that these mice exhibited an attenuated response to FLX in a neurogenesis-dependent behavioral assay of FLX action, while neurogenesis-independent behaviors were unaffected by GluN2B deletion. In addition, deletion of GluN2B attenuated FLX-induced increases in dendritic complexity of abGCs suggesting that the blunted behavioral efficacy of FLX may be caused by impaired differentiation of young abGCs.
Collapse
Affiliation(s)
- Lindsay Tannenholz
- Department of Pharmacology, Columbia UniversityNew York, NY, USA; Division of Integrative Neuroscience, New York State Psychiatric InstituteNew York, NY, USA
| | - René Hen
- Department of Pharmacology, Columbia UniversityNew York, NY, USA; Division of Integrative Neuroscience, New York State Psychiatric InstituteNew York, NY, USA; Department of Psychiatry, Columbia UniversityNew York, NY, USA; Department of Neuroscience, Columbia UniversityNew York, NY, USA
| | - Mazen A Kheirbek
- Division of Integrative Neuroscience, New York State Psychiatric InstituteNew York, NY, USA; Department of Psychiatry, Columbia UniversityNew York, NY, USA; Department of Psychiatry, University of CaliforniaSan Francisco, CA, USA
| |
Collapse
|
42
|
Dong Y, Zhou Y, Chu X, Chen S, Chen L, Yang B, Zhang X, Wang L, Wang S, Lou J, Deng Q, Wang L, Cao Z, Wang J, Xie J, Serdyuk T, Li S, He L, Chen X, Li W. Dental noise exposed mice display depressive-like phenotypes. Mol Brain 2016; 9:50. [PMID: 27160396 PMCID: PMC4894364 DOI: 10.1186/s13041-016-0229-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2016] [Accepted: 04/21/2016] [Indexed: 11/10/2022] Open
Abstract
Background Studies have indicated that depressive disorders are observed frequently in dentists. It’s suggested that dentists encounter numerous sources of stress in their professional career. We noticed that the noises in dental environments are very unpleasant. The animal modeling studies suggested that stressful noise could produce depressive-like phenotypes in rodent animals. We hypothesize that the dental noise may be one of the primary stressors causing depressive disorders in dentists. Results We treated C57BL/6 mice with programmatically played wide-spectrum dental noise for 8 h/day at 75 ± 10 dB SPL level for 30 days, and then tested the behaviors. After exposure to dental noise, animals displayed the depressive-like phenotypes, accompanied by inhibition of neurogenesis in hippocampus. These deficits were ameliorated by orally administered with antidepressant fluoxetine. Conclusions Our results suggested that dental noise could be one of the primary stressors for the pathogenesis of depressive disorders and the dental noise mouse model could be used in further depression studies. Electronic supplementary material The online version of this article (doi:10.1186/s13041-016-0229-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yujie Dong
- Bio-X Institutes, Key Laboratory for the Genetics of Development and Neuropsychiatric Disorders (Ministry of Education), Shanghai Key Laboratory of Psychotic Disorders, and Brain Science and Technology Research Center, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, China
| | - Ying Zhou
- Bio-X Institutes, Key Laboratory for the Genetics of Development and Neuropsychiatric Disorders (Ministry of Education), Shanghai Key Laboratory of Psychotic Disorders, and Brain Science and Technology Research Center, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, China
| | - Xixia Chu
- Bio-X Institutes, Key Laboratory for the Genetics of Development and Neuropsychiatric Disorders (Ministry of Education), Shanghai Key Laboratory of Psychotic Disorders, and Brain Science and Technology Research Center, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, China
| | - Shiqing Chen
- Bio-X Institutes, Key Laboratory for the Genetics of Development and Neuropsychiatric Disorders (Ministry of Education), Shanghai Key Laboratory of Psychotic Disorders, and Brain Science and Technology Research Center, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, China
| | - Lei Chen
- Bio-X Institutes, Key Laboratory for the Genetics of Development and Neuropsychiatric Disorders (Ministry of Education), Shanghai Key Laboratory of Psychotic Disorders, and Brain Science and Technology Research Center, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, China
| | - Beimeng Yang
- Bio-X Institutes, Key Laboratory for the Genetics of Development and Neuropsychiatric Disorders (Ministry of Education), Shanghai Key Laboratory of Psychotic Disorders, and Brain Science and Technology Research Center, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, China
| | - Xu Zhang
- Bio-X Institutes, Key Laboratory for the Genetics of Development and Neuropsychiatric Disorders (Ministry of Education), Shanghai Key Laboratory of Psychotic Disorders, and Brain Science and Technology Research Center, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, China
| | - Lin Wang
- Bio-X Institutes, Key Laboratory for the Genetics of Development and Neuropsychiatric Disorders (Ministry of Education), Shanghai Key Laboratory of Psychotic Disorders, and Brain Science and Technology Research Center, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, China
| | - Shuai Wang
- Bio-X Institutes, Key Laboratory for the Genetics of Development and Neuropsychiatric Disorders (Ministry of Education), Shanghai Key Laboratory of Psychotic Disorders, and Brain Science and Technology Research Center, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, China
| | - Jingyu Lou
- Bio-X Institutes, Key Laboratory for the Genetics of Development and Neuropsychiatric Disorders (Ministry of Education), Shanghai Key Laboratory of Psychotic Disorders, and Brain Science and Technology Research Center, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, China
| | - Qing Deng
- Shanghai Elli Dental Clinic, No.26 South Yili Road, Shanghai, China
| | - Li Wang
- Shanghai Elli Dental Clinic, No.26 South Yili Road, Shanghai, China
| | - Zheyi Cao
- Bio-X Institutes, Key Laboratory for the Genetics of Development and Neuropsychiatric Disorders (Ministry of Education), Shanghai Key Laboratory of Psychotic Disorders, and Brain Science and Technology Research Center, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, China
| | - Jianan Wang
- Bio-X Institutes, Key Laboratory for the Genetics of Development and Neuropsychiatric Disorders (Ministry of Education), Shanghai Key Laboratory of Psychotic Disorders, and Brain Science and Technology Research Center, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, China
| | - Jiaxin Xie
- Bio-X Institutes, Key Laboratory for the Genetics of Development and Neuropsychiatric Disorders (Ministry of Education), Shanghai Key Laboratory of Psychotic Disorders, and Brain Science and Technology Research Center, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, China
| | - Tatiana Serdyuk
- Bio-X Institutes, Key Laboratory for the Genetics of Development and Neuropsychiatric Disorders (Ministry of Education), Shanghai Key Laboratory of Psychotic Disorders, and Brain Science and Technology Research Center, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, China
| | - Shengtian Li
- Bio-X Institutes, Key Laboratory for the Genetics of Development and Neuropsychiatric Disorders (Ministry of Education), Shanghai Key Laboratory of Psychotic Disorders, and Brain Science and Technology Research Center, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, China
| | - Lin He
- Bio-X Institutes, Key Laboratory for the Genetics of Development and Neuropsychiatric Disorders (Ministry of Education), Shanghai Key Laboratory of Psychotic Disorders, and Brain Science and Technology Research Center, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, China
| | - Xiaoping Chen
- National Key Laboratory of Human Factors Engineering, China Astronaut Research and Training Center, Beijing, 100094, China.
| | - Weidong Li
- Bio-X Institutes, Key Laboratory for the Genetics of Development and Neuropsychiatric Disorders (Ministry of Education), Shanghai Key Laboratory of Psychotic Disorders, and Brain Science and Technology Research Center, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, China.
| |
Collapse
|
43
|
Sun X, Zhou Z, Liu T, Zhao M, Zhao S, Xiao T, Jolkkonen J, Zhao C. Fluoxetine Enhances Neurogenesis in Aged Rats with Cortical Infarcts, but This is not Reflected in a Behavioral Recovery. J Mol Neurosci 2015; 58:233-42. [PMID: 26474565 DOI: 10.1007/s12031-015-0662-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Accepted: 10/05/2015] [Indexed: 01/28/2023]
Abstract
Age is associated with poor outcome and impaired functional recovery after stroke. Fluoxetine, which is widely used in clinical practice, can regulate hippocampal neurogenesis in young rodents. As the rate of neurogenesis is dramatically reduced during aging, we studied the effect of post-stroke fluoxetine treatment on neurogenesis in the subventricular zone (SVZ) and subgranular zone (SGZ) of dentate gyrus (DG) and whether this would be associated with any behavioral recovery after the cortical infarct in aged rats. Aged rats were randomly assigned to four groups: sham-operated rats, sham-operated rats treated with fluoxetine, rats subjected to cerebral ischemia, and rats with ischemia treated with fluoxetine. Focal cortical ischemia was induced by intracranial injection of vasoconstrictive peptide, endothelin-1 (ET-1). Fluoxetine was administered in the drinking water for 3 weeks starting 1 week after ischemia at a dose of 18 mg/kg/day. Behavioral recovery was evaluated on post-stroke days 29 to 31 after which the survival rate and fate of proliferating cells in the SVZ and DG were assessed by immunohistochemistry. Apoptosis was measured with the TUNEL assay. The results indicated that chronic fluoxetine treatment after stroke enhanced the proliferation of newborn neurons in the SVZ, but not in SGZ, and it suppressed perilesional apoptosis. Fluoxetine treatment did not affect the survival or differentiation of newly generated cells in the SVZ i.e., the enhanced neurogenesis was not translated into a behavioral outcome.
Collapse
Affiliation(s)
- Xiaoyu Sun
- Neurology, The First Hospital of China Medical University, No.155, North Nanjing Street, Heping District, Shenyang, Liaoning, 110001, China
| | - Zhike Zhou
- Neurology, The First Hospital of China Medical University, No.155, North Nanjing Street, Heping District, Shenyang, Liaoning, 110001, China
| | - Tingting Liu
- Neurology, The First Hospital of China Medical University, No.155, North Nanjing Street, Heping District, Shenyang, Liaoning, 110001, China
| | - Mei Zhao
- Cardiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Shanshan Zhao
- Neurology, The First Hospital of China Medical University, No.155, North Nanjing Street, Heping District, Shenyang, Liaoning, 110001, China
| | - Ting Xiao
- Dermatology, The First Hospital of China Medical University, Shenyang, China
- Key Laboratory of Immunodermatology, Ministry of Health, Ministry of Education, Shenyang, China
| | - Jukka Jolkkonen
- Institute of Clinical Medicine - Neurology, University of Eastern Finland, P. O. Box 1627, 70211, Kuopio, Finland
| | - Chuansheng Zhao
- Neurology, The First Hospital of China Medical University, No.155, North Nanjing Street, Heping District, Shenyang, Liaoning, 110001, China.
| |
Collapse
|
44
|
Marschallinger J, Sah A, Schmuckermair C, Unger M, Rotheneichner P, Kharitonova M, Waclawiczek A, Gerner P, Jaksch-Bogensperger H, Berger S, Striessnig J, Singewald N, Couillard-Despres S, Aigner L. The L-type calcium channel Cav1.3 is required for proper hippocampal neurogenesis and cognitive functions. Cell Calcium 2015; 58:606-16. [PMID: 26459417 DOI: 10.1016/j.ceca.2015.09.007] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2015] [Revised: 09/04/2015] [Accepted: 09/29/2015] [Indexed: 11/29/2022]
Abstract
L-type voltage gated Ca(2+) channels (LTCCs) are widely expressed within different brain regions including the hippocampus. The isoforms Cav1.2 and Cav1.3 have been shown to be involved in hippocampus-dependent learning and memory, cognitive functions that require proper hippocampal neurogenesis. In vitro, functional LTCCs are expressed on neuronal progenitor cells, where they promote neuronal differentiation. Expression of LTCCs on neural stem and progenitor cells within the neurogenic regions in the adult brain in vivo has not been examined so far, and a contribution of the individual isoforms Cav1.2 and Cav1.3 to adult neurogenesis remained to be clarified. To reveal the role of these channels we first evaluated the expression patterns of Cav1.2 and Cav1.3 in the hippocampal dentate gyrus and the subventricular zone (SVZ) in adult (2- and 3-month old) and middle-aged (15-month old) mice on mRNA and protein levels. We performed immunohistological analysis of hippocampal neurogenesis in adult and middle-aged Cav1.3(-/-) mice and finally addressed the importance of Cav1.3 for hippocampal function by evaluating spatial memory and depression-like behavior in adult Cav1.3(-/-) mice. Our results showed Cav1.2 and Cav1.3 expression at different stages of neuronal differentiation. While Cav1.2 was primarily restricted to mature NeuN(+) granular neurons, Cav1.3 was expressed in Nestin(+) neural stem cells and in mature NeuN(+) granular neurons. Adult and middle-aged Cav1.3(-/-) mice showed severe impairments in dentate gyrus neurogenesis, with significantly smaller dentate gyrus volume, reduced survival of newly generated cells, and reduced neuronal differentiation. Further, Cav1.3(-/-) mice showed impairment in the hippocampus dependent object location memory test, implicating Cav1.3 as an essential element for hippocampus-associated cognitive functions. Thus, modulation of LTCC activities may have a crucial impact on neurogenic responses and cognition, which should be considered for future therapeutic administration of LTCCs modulators.
Collapse
Affiliation(s)
- Julia Marschallinger
- Institute of Molecular Regenerative Medicine, Paracelsus Medical University, Salzburg, Austria; Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Paracelsus Medical University, Salzburg, Austria
| | - Anupam Sah
- Department of Pharmacology and Toxicology, Institute of Pharmacy and CMBI, Leopold-Franzens-University of Innsbruck, Innsbruck, Austria
| | - Claudia Schmuckermair
- Department of Pharmacology and Toxicology, Institute of Pharmacy and CMBI, Leopold-Franzens-University of Innsbruck, Innsbruck, Austria
| | - Michael Unger
- Institute of Molecular Regenerative Medicine, Paracelsus Medical University, Salzburg, Austria; Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Paracelsus Medical University, Salzburg, Austria
| | - Peter Rotheneichner
- Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Paracelsus Medical University, Salzburg, Austria; Institute of Experimental Neuroregeneration, Paracelsus Medical University, Salzburg, Austria
| | - Maria Kharitonova
- Department of Pharmacology and Toxicology, Institute of Pharmacy and CMBI, Leopold-Franzens-University of Innsbruck, Innsbruck, Austria
| | - Alexander Waclawiczek
- Institute of Molecular Regenerative Medicine, Paracelsus Medical University, Salzburg, Austria; Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Paracelsus Medical University, Salzburg, Austria
| | - Philipp Gerner
- Institute of Molecular Regenerative Medicine, Paracelsus Medical University, Salzburg, Austria; Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Paracelsus Medical University, Salzburg, Austria
| | - Heidi Jaksch-Bogensperger
- Institute of Molecular Regenerative Medicine, Paracelsus Medical University, Salzburg, Austria; Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Paracelsus Medical University, Salzburg, Austria
| | - Stefan Berger
- Department of Molecular Biology, Central Institute of Mental Health and Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Jörg Striessnig
- Department of Pharmacology and Toxicology, Institute of Pharmacy and CMBI, Leopold-Franzens-University of Innsbruck, Innsbruck, Austria
| | - Nicolas Singewald
- Department of Pharmacology and Toxicology, Institute of Pharmacy and CMBI, Leopold-Franzens-University of Innsbruck, Innsbruck, Austria
| | - Sebastien Couillard-Despres
- Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Paracelsus Medical University, Salzburg, Austria; Institute of Experimental Neuroregeneration, Paracelsus Medical University, Salzburg, Austria
| | - Ludwig Aigner
- Institute of Molecular Regenerative Medicine, Paracelsus Medical University, Salzburg, Austria; Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Paracelsus Medical University, Salzburg, Austria.
| |
Collapse
|
45
|
When ageing meets the blues: Are current antidepressants effective in depressed aged patients? Neurosci Biobehav Rev 2015; 55:478-97. [DOI: 10.1016/j.neubiorev.2015.06.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Accepted: 06/03/2015] [Indexed: 02/06/2023]
|
46
|
Reversal of age-associated cognitive deficits is accompanied by increased plasticity-related gene expression after chronic antidepressant administration in middle-aged mice. Pharmacol Biochem Behav 2015; 135:70-82. [DOI: 10.1016/j.pbb.2015.05.013] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2015] [Revised: 04/07/2015] [Accepted: 05/22/2015] [Indexed: 12/22/2022]
|
47
|
Abstract
The wide spectrum of disruptions that characterizes major depressive disorder (MDD) and bipolar disorder (BD) highlights the difficulties researchers are posed with as they try to mimic these disorders in the laboratory. Nonetheless, numerous attempts have been made to create rodent models of mood disorders or at least models of the symptoms of MDD and BD. Present antidepressants are all descendants of the serendipitous findings in the 1950s that the monoamine oxidase inhibitor iproniazid and the tricyclic antidepressant imipramine were effective antidepressants. Thus, the need for improved animal models to provide insights into the neuropathology underlying the disease is critical. Such information is in turn crucial for identifying new antidepressants and mood stabilisers. Currently, there is a shift away from traditional animal models to more focused research dealing with an endophenotype-style approach, genetic models, and incorporation of new findings from human neuroimaging and genetic studies. Such approaches are opening up more tractable avenues for understanding the neurobiological and genetic bases of these disorders. Further, such models promise to yield better translational animal models and hence more fruitful therapeutic targets. This overview focuses on such animal models and tests and how they can be used to assess MDD and BD in rodents.
Collapse
|
48
|
Po KT, Siu AMH, Lau BWM, Chan JNM, So KF, Chan CCH. Repeated, high-dose dextromethorphan treatment decreases neurogenesis and results in depression-like behavior in rats. Exp Brain Res 2015; 233:2205-14. [PMID: 25939533 DOI: 10.1007/s00221-015-4290-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2014] [Accepted: 04/15/2015] [Indexed: 01/28/2023]
Abstract
Abuse of cough mixture is increasingly prevalent worldwide. Clinical studies showed that chronic consumption of cough mixture at high dosages may lead to psychiatric symptoms, especially affective disturbances, with the underlying mechanisms remain elusive. The present study aims at exploring the effect of repeated, high-dose dextromethorphan (DXM, a common active component of cough mixture) treatment on adult hippocampal neurogenesis, which is associated with pathophysiology of mood disturbances. After treatment with a high-dose of DXM (40 mg/kg/day) for 2 weeks, Sprague-Dawley rats showed increased depression-like behavior when compared to the control animals. Neurogenesis in the hippocampus was suppressed by DXM treatment, which was indicated by decreases in number of proliferative cells and doublecortin (an immature neuron marker)-positive new neurons. Furthermore, the dendritic complexity of the immature neurons was suppressed by DXM treatment. These findings suggest that DXM induces depression- and anxiety-like behavior and suppresses neurogenesis in rats. The current experimental paradigm may serve as an animal model for study on affective effect of cough mixture abuse, rehabilitation treatment options for abusers and the related neurological mechanisms.
Collapse
Affiliation(s)
- Kai Ting Po
- ST 507, Department of Rehabilitation Sciences, The Hong Kong Polytechnic University, Hong Kong, China
| | | | | | | | | | | |
Collapse
|
49
|
McAvoy K, Russo C, Kim S, Rankin G, Sahay A. Fluoxetine induces input-specific hippocampal dendritic spine remodeling along the septotemporal axis in adulthood and middle age. Hippocampus 2015; 25:1429-46. [PMID: 25850664 DOI: 10.1002/hipo.22464] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/01/2015] [Indexed: 12/15/2022]
Abstract
Fluoxetine, a selective serotonin-reuptake inhibitor (SSRI), is known to induce structural rearrangements and changes in synaptic transmission in hippocampal circuitry. In the adult hippocampus, structural changes include neurogenesis, dendritic, and axonal plasticity of pyramidal and dentate granule neurons, and dedifferentiation of dentate granule neurons. However, much less is known about how chronic fluoxetine affects these processes along the septotemporal axis and during the aging process. Importantly, studies documenting the effects of fluoxetine on density and distribution of spines along different dendritic segments of dentate granule neurons and CA1 pyramidal neurons along the septotemporal axis of hippocampus in adulthood and during aging are conspicuously absent. Here, we use a transgenic mouse line in which mature dentate granule neurons and CA1 pyramidal neurons are genetically labeled with green fluorescent protein (GFP) to investigate the effects of chronic fluoxetine treatment (18 mg/kg/day) on input-specific spine remodeling and mossy fiber structural plasticity in the dorsal and ventral hippocampus in adulthood and middle age. In addition, we examine levels of adult hippocampal neurogenesis, maturation state of dentate granule neurons, neuronal activity, and glutamic acid decarboxylase-67 expression in response to chronic fluoxetine in adulthood and middle age. Our studies reveal that while chronic fluoxetine fails to augment adult hippocampal neurogenesis in middle age, the middle-aged hippocampus retains high sensitivity to changes in the dentate gyrus (DG) such as dematuration, hypoactivation, and increased glutamic acid decarboxylase 67 (GAD67) expression. Interestingly, the middle-aged hippocampus shows greater sensitivity to fluoxetine-induced input-specific synaptic remodeling than the hippocampus in adulthood with the stratum-oriens of CA1 exhibiting heightened structural plasticity. The input-specific changes and circuit-level modifications in middle-age were associated with modest enhancement in contextual fear memory precision, anxiety-like behavior and antidepressant-like behavioral responses.
Collapse
Affiliation(s)
- Kathleen McAvoy
- Center for Regenerative Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts.,Harvard Stem Cell Institute, Harvard University, Boston, Massachusetts.,Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Craig Russo
- Center for Regenerative Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts.,Harvard Stem Cell Institute, Harvard University, Boston, Massachusetts.,Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Shannen Kim
- Center for Regenerative Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts.,Harvard Stem Cell Institute, Harvard University, Boston, Massachusetts.,Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Genelle Rankin
- Center for Regenerative Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts.,Harvard Stem Cell Institute, Harvard University, Boston, Massachusetts.,Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Amar Sahay
- Center for Regenerative Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts.,Harvard Stem Cell Institute, Harvard University, Boston, Massachusetts.,Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
50
|
Aniol VA, Gulyaeva NV. Changes in adult neurogenesis in the hippocampus during depressive disorders in humans. NEUROCHEM J+ 2015. [DOI: 10.1134/s181971241501002x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|