1
|
Liang D, Wang D, Zheng X, Xiang H, Liu S, Yu C, Tian J, Ma J, Niu Y. Aerobic plus resistance exercise attenuates skeletal muscle atrophy induced by dexamethasone through the HDAC4/FoxO3a pathway. Cell Signal 2025; 127:111581. [PMID: 39732306 DOI: 10.1016/j.cellsig.2024.111581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 12/04/2024] [Accepted: 12/24/2024] [Indexed: 12/30/2024]
Abstract
This study aimed to investigate the underlying mechanisms by which physical exercise mitigates muscle atrophy induced by Dexamethasone (Dex). A muscle atrophy model was established in the mouse C2C12 cell line and 8-week-old mice treated with Dex, with subsequent verification of phenotype and atrogene expression. The potential benefits of combined aerobic and resistance exercise in mitigating muscle atrophy were then examined. To elucidate the involvement of Histone deacetylase 4 (HDAC4) in the protective effects of exercise against muscle loss, a combination of RT-PCR, Western blotting, immunoprecipitation, and immunofluorescence staining techniques were employed. The upregulation of HDAC4 was observed following Dex-induced muscle atrophy in vitro and in vivo. Inhibition of HDAC4 in C2C12 cells resulted in an increase in myotube diameter and fusion index, along with a decrease in the expression of Atrogin-1 and MuRF1. Treatment with Tasquinimod, an HDAC4 inhibitor, effectively prevented muscle wasting and dysfunction in mice induced by Dex. After a 6-week exercise intervention, the Dex-Exercise group exhibited significant improvements in body fat level, hyperinsulinemia, muscle mass and function in comparison to the Dex-Sedentary group. Mechanistically, we discovered that HDAC4 bound to and deacetylated Forkhead box protein O 3a (FoxO3a) within the nucleus, leading to decreased phosphorylation of FoxO3a at Ser 253. This interaction subsequently facilitated the expression of downstream atrogene Atrogin-1 and MuRF1, resulting in muscle atrophy. Conversely, exercise was found to potentially mitigate muscle atrophy by inhibiting the HDAC4/FoxO3a pathway. These findings suggest that HDAC4 may be a potential therapeutic target for exercise to combat Dex-induced muscle atrophy.
Collapse
Affiliation(s)
- Dehuan Liang
- Department of Rehabilitation, School of Medical Technology, Tianjin Medical University, Tianjin 300070, China
| | - Danni Wang
- Department of Rehabilitation, School of Medical Technology, Tianjin Medical University, Tianjin 300070, China
| | - Xinyue Zheng
- Department of Rehabilitation, School of Medical Technology, Tianjin Medical University, Tianjin 300070, China
| | - Heng Xiang
- Department of Rehabilitation, School of Medical Technology, Tianjin Medical University, Tianjin 300070, China
| | - Sujuan Liu
- Department of Anatomy and Histology, School of Basic Medical Science, Tianjin Medical University, Tianjin 300070, China
| | - Chunxia Yu
- School of Medical Technology, Tianjin Medical University, Tianjin 300070, China
| | - Jiatong Tian
- Department of Rehabilitation, School of Medical Technology, Tianjin Medical University, Tianjin 300070, China
| | - Jianxiong Ma
- Tianjin Hospital, Tianjin University, No. 406 Jiefang South Road, Tianjin 300211, China
| | - Yanmei Niu
- Department of Rehabilitation, School of Medical Technology, Tianjin Medical University, Tianjin 300070, China.
| |
Collapse
|
2
|
Lambertus M, Geiseler S, Morland C. High-intensity interval exercise is more efficient than medium intensity exercise at inducing neurogenesis. J Physiol 2024; 602:7027-7042. [PMID: 39580614 DOI: 10.1113/jp287328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 10/18/2024] [Indexed: 11/26/2024] Open
Abstract
The neurogenic potential of the brain decreases during ageing, whereas the risk of neurodegenerative diseases and stroke rises. This creates a mismatch between the rate of neuron loss and the brain's capacity for replacement. Adult neurogenesis primarily occurs in the subgranular zone (SGZ) and the ventricular-subventricular zone (V-SVZ). Exercise enhances SGZ neurogenesis, and we previously showed that V-SVZ neurogenesis is induced by exercise via activation of the lactate receptor HCA1. Here, we investigated how high-intensity interval training (HIIT) and medium-intensity interval training (MIIT) affect neurogenesis in these niches. Wild-type (WT) and HCA1 knockout (KO) mice were randomized to sedentary, HIIT or MIIT (n = 5-8 per group) for 3 weeks. In the SGZ, HIIT increased the density of doublecortin (DCX)-positive cells in WT mice by 85% (5.77±1.76 vs. 3.12±1.54 cells/100 µm, P = 0.013) and KO mice (67% increase; 7.91±2.92 vs. 4.73±1.63 cells/100 µm, P = 0.004). MIIT did not alter the density of DCX-positive cells in either genotype. HIIT increased the density of Ki-67-positive cells only in KO mice (P = 0.038), whereas no differences in nestin-positive cells were observed. In the V-SVZ, HIIT increased the density of DCX-positive cells in WT mice by 155% (117.79±39.72 vs. 46.25±19.96 cells/100 µm, P < 0.001) and MIIT increased the density of DCX-positive cells by 80% (83.26±39.48 vs. 46.25±19.96 cells/100µm, P = 0.027). No exercise-induced changes were observed in KO mice. Similar patterns were noted for Ki-67 positive and DCX/Ki-67 double-positive cells in the V-SVZ. These findings suggest that HIIT enhances neurogenesis more robustly than MIIT in both niches, with HCA1 playing a crucial role in V-SVZ neurogenesis. KEY POINTS: The neurogenic potential of the brain decreases with age, whereas the risk of neurodegenerative diseases and stroke increases, highlighting a mismatch between neuronal loss and replacement capacity. Exercise enhances neurogenesis in both the subgranular zone and the ventricular-subventricular zone. High-intensity interval exercise is more effective than medium-intensity interval exercise at promoting neurogenesis in both the subgranular zone and the ventricular-subventricular zone of wild-type mice. The enhancement of neurogenesis in the ventricular-subventricular zone is dependent on the activation of the HCA1 receptor, as evidenced by the ability of medium- and high-intensity interval exercise to induce neurogenesis in wild-type mice and the lack of this effect in HCA1 knockout mice. By contrast, neurogenesis in the subgranular zone is independent on the activation of the HCA1 receptor, highlighting that neurogenesis in the two major neurogenic niches are regulated differently.
Collapse
Affiliation(s)
- Marvin Lambertus
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, University of Oslo, Oslo, Norway
| | - Samuel Geiseler
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, University of Oslo, Oslo, Norway
| | - Cecilie Morland
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, University of Oslo, Oslo, Norway
| |
Collapse
|
3
|
Khawagi WY, Al-Kuraishy HM, Hussein NR, Al-Gareeb AI, Atef E, Elhussieny O, Alexiou A, Papadakis M, Jabir MS, Alshehri AA, Saad HM, Batiha GES. Depression and type 2 diabetes: A causal relationship and mechanistic pathway. Diabetes Obes Metab 2024; 26:3031-3044. [PMID: 38802993 DOI: 10.1111/dom.15630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2024] [Revised: 04/16/2024] [Accepted: 04/16/2024] [Indexed: 05/29/2024]
Abstract
Depression is a mood disorder that may increase risk for the development of insulin resistance (IR) and type 2 diabetes (T2D), and vice versa. However, the mechanistic pathway linking depression and T2D is not fully elucidated. The aim of this narrative review, therefore, was to discuss the possible link between depression and T2D. The coexistence of T2D and depression is twice as great compared to the occurrence of either condition independently. Hyperglycaemia and dyslipidaemia promote the incidence of depression by enhancing inflammation and reducing brain serotonin (5-hydroxytryptamine [5HT]). Dysregulation of insulin signalling in T2D impairs brain 5HT signalling, leading to the development of depression. Furthermore, depression is associated with the development of hyperglycaemia and poor glycaemic control. Psychological stress and depression promote the development of T2D. In conclusion, T2D could be a potential risk factor for the development of depression through the induction of inflammatory reactions and oxidative stress that affect brain neurotransmission. In addition, chronic stress in depression may induce the development of T2D through dysregulation of the hypothalamic-pituitary-adrenal axis and increase circulating cortisol levels, which triggers IR and T2D.
Collapse
Affiliation(s)
- Wael Y Khawagi
- Department of Clinical Pharmacy, College of Pharmacy, Taif University, Taif, Saudi Arabia
| | - Hayder M Al-Kuraishy
- Department of Clinical Pharmacology and Medicine, College of Medicine, Mustansiriyah University, Baghdad, Iraq
| | - Nawar R Hussein
- College of Pharmacy, Pharmacology Department, Al-Farahidi University, Baghdad, Iraq
| | - Ali I Al-Gareeb
- Department of Clinical Pharmacology and Medicine, College of Medicine, Mustansiriyah University, Baghdad, Iraq
| | - Esraa Atef
- Respiratory Therapy Department, Mohammed Al-Mana College for Medical Sciences, Dammam, Saudi Arabia
| | - Omnya Elhussieny
- Department of Histology and Cytology, Faculty of Veterinary Medicine, Matrouh University, Marsa Matruh, Egypt
| | - Athanasios Alexiou
- University Centre for Research & Development, Chandigarh University Chandigarh-Ludhiana Highway, Mohali, India
- Department of Research and Development, Funogen, Athens, Greece
- Department of Research and Development, AFNP Med, Wien, Austria
- Department of Science and Engineering, Novel Global Community Educational Foundation, Hebersham, New South Wales, Australia
| | - Marios Papadakis
- Department of Surgery II, University Hospital Witten-Herdecke, University of Witten-Herdecke, Wuppertal, Germany
| | - Majid S Jabir
- Applied Science Department, University of Technology, Baghdad, Iraq
| | - Abdullah A Alshehri
- Department of Clinical Pharmacy, College of Pharmacy, Taif University, Taif, Saudi Arabia
| | - Hebatallah M Saad
- Department of Pathology, Faculty of Veterinary Medicine, Matrouh University, Marsa Matruh, Egypt
| | - Gaber El-Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhour University, Damanhour, Egypt
| |
Collapse
|
4
|
Eachus H, Ryu S. Glucocorticoid effects on the brain: from adaptive developmental plasticity to allostatic overload. J Exp Biol 2024; 227:jeb246128. [PMID: 38449327 PMCID: PMC10949071 DOI: 10.1242/jeb.246128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/08/2024]
Abstract
Exposure to stress during early life may alter the developmental trajectory of an animal by a mechanism known as adaptive plasticity. For example, to enhance reproductive success in an adverse environment, it is known that animals accelerate their growth during development. However, these short-term fitness benefits are often associated with reduced longevity, a phenomenon known as the growth rate-lifespan trade-off. In humans, early life stress exposure compromises health later in life and increases disease susceptibility. Glucocorticoids (GCs) are major stress hormones implicated in these processes. This Review discusses the evidence for GC-mediated adaptive plasticity in development, leading to allostatic overload in later life. We focus on GC-induced effects on brain structure and function, including neurogenesis; highlight the need for longitudinal studies; and discuss approaches to identify molecular mechanisms mediating GC-induced alteration of the brain developmental trajectory leading to adult dysfunctions. Further understanding of how stress and GC exposure can alter developmental trajectories at the molecular and cellular level is of critical importance to reduce the burden of mental and physical ill health across the life course.
Collapse
Affiliation(s)
- Helen Eachus
- Living Systems Institute & Department of Clinical and Biomedical Sciences, University of Exeter, Stocker Road, Exeter EX4 4QD, UK
| | - Soojin Ryu
- Living Systems Institute & Department of Clinical and Biomedical Sciences, University of Exeter, Stocker Road, Exeter EX4 4QD, UK
| |
Collapse
|
5
|
Liu M, Fan Y, Ni N, Yu T, Mao Z, Huang H, Zhang J, Tang Y, He H, Meng F, You Y, Zhou Q. TERT mediates the U-shape of glucocorticoids effects in modulation of hippocampal neural stem cells and associated brain function. CNS Neurosci Ther 2024; 30:e14577. [PMID: 38421107 PMCID: PMC10850922 DOI: 10.1111/cns.14577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 10/17/2023] [Accepted: 12/07/2023] [Indexed: 03/02/2024] Open
Abstract
BACKGROUND Glucocorticoids (GCs) are steroidal hormones produced by the adrenal cortex. A physiological-level GCs have a crucial function in maintaining many cognitive processes, like cognition, memory, and mood, however, both insufficient and excessive GCs impair these functions. Although this phenomenon could be explained by the U-shape of GC effects, the underlying mechanisms are still not clear. Therefore, understanding the underlying mechanisms of GCs may provide insight into the treatments for cognitive and mood-related disorders. METHODS Consecutive administration of corticosterone (CORT, 10 mg/kg, i.g.) proceeded for 28 days to mimic excessive GCs condition. Adrenalectomy (ADX) surgery was performed to ablate endogenous GCs in mice. Microinjection of 1 μL of Ad-mTERT-GFP virus into mouse hippocampus dentate gyrus (DG) and behavioral alterations in mice were observed 4 weeks later. RESULTS Different concentrations of GCs were shown to affect the cell growth and development of neural stem cells (NSCs) in a U-shaped manner. The physiological level of GCs (0.01 μM) promoted NSC proliferation in vitro, while the stress level of GCs (10 μM) inhibited it. The glucocorticoid synthesis blocker metyrapone (100 mg/kg, i.p.) and ADX surgery both decreased the quantity and morphological development of doublecortin (DCX)-positive immature cells in the DG. The physiological level of GCs activated mineralocorticoid receptor and then promoted the production of telomerase reverse transcriptase (TERT); in contrast, the stress level of GCs activated glucocorticoid receptor and then reduced the expression of TERT. Overexpression of TERT by AD-mTERT-GFP reversed both chronic stresses- and ADX-induced deficiency of TERT and the proliferation and development of NSCs, chronic stresses-associated depressive symptoms, and ADX-associated learning and memory impairment. CONCLUSION The bidirectional regulation of TERT by different GCs concentrations is a key mechanism mediating the U-shape of GC effects in modulation of hippocampal NSCs and associated brain function. Replenishment of TERT could be a common treatment strategy for GC dysfunction-associated diseases.
Collapse
Affiliation(s)
- Meng‐Ying Liu
- Department of Pharmacy, Nanjing Drum Tower HospitalAffiliated Hospital of Medical School, Nanjing UniversityNanjingChina
- State Key Laboratory of Reproductive Medicine, Department of Clinical Pharmacology, School of PharmacyNanjing Medical UniversityNanjingChina
| | - Yixin Fan
- State Key Laboratory of Reproductive Medicine, Department of Clinical Pharmacology, School of PharmacyNanjing Medical UniversityNanjingChina
- Department of Pharmacy, Sir Run Run HospitalNanjing Medical UniversityNanjingChina
| | - Ningjie Ni
- State Key Laboratory of Reproductive Medicine, Department of Clinical Pharmacology, School of PharmacyNanjing Medical UniversityNanjingChina
- Department of NeurosurgeryThe First Affiliated Hospital of Nanjing Medical UniversityNanjingChina
| | - Tao Yu
- School of PharmacyNanjing Medical UniversityNanjingChina
| | - Zhiyuan Mao
- Key Laboratory for Aging & Disease, The State Key Laboratory of Reproductive Medicine, Department of Human Anatomy, Research Centre for Bone and Stem CellsNanjing Medical UniversityNanjingChina
| | - Hanyu Huang
- State Key Laboratory of Reproductive Medicine, Department of Clinical Pharmacology, School of PharmacyNanjing Medical UniversityNanjingChina
- Department of NeurosurgeryThe First Affiliated Hospital of Nanjing Medical UniversityNanjingChina
| | - Jing Zhang
- Department of Clinical Pharmacology, School of PharmacyNanjing Medical UniversityNanjingChina
| | - Yulin Tang
- Department of Pharmacy, Sir Run Run HospitalNanjing Medical UniversityNanjingChina
| | - Hongliang He
- Department of Pharmacy, Sir Run Run HospitalNanjing Medical UniversityNanjingChina
| | - Fan Meng
- Department of Clinical Pharmacology, School of PharmacyNanjing Medical UniversityNanjingChina
| | - Yongping You
- Department of NeurosurgeryThe First Affiliated Hospital of Nanjing Medical UniversityNanjingChina
| | - Qi‐Gang Zhou
- State Key Laboratory of Reproductive Medicine, Department of Clinical Pharmacology, School of PharmacyNanjing Medical UniversityNanjingChina
- Department of Pharmacy, Sir Run Run HospitalNanjing Medical UniversityNanjingChina
- Department of Clinical Pharmacology, School of PharmacyNanjing Medical UniversityNanjingChina
| |
Collapse
|
6
|
Viho EMG, Punt AM, Distel B, Houtman R, Kroon J, Elgersma Y, Meijer OC. The Hippocampal Response to Acute Corticosterone Elevation Is Altered in a Mouse Model for Angelman Syndrome. Int J Mol Sci 2022; 24:ijms24010303. [PMID: 36613751 PMCID: PMC9820460 DOI: 10.3390/ijms24010303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 12/18/2022] [Accepted: 12/21/2022] [Indexed: 12/28/2022] Open
Abstract
Angelman Syndrome (AS) is a severe neurodevelopmental disorder, caused by the neuronal absence of the ubiquitin protein ligase E3A (UBE3A). UBE3A promotes ubiquitin-mediated protein degradation and functions as a transcriptional coregulator of nuclear hormone receptors, including the glucocorticoid receptor (GR). Previous studies showed anxiety-like behavior and hippocampal-dependent memory disturbances in AS mouse models. Hippocampal GR is an important regulator of the stress response and memory formation, and we therefore investigated whether the absence of UBE3A in AS mice disrupted GR signaling in the hippocampus. We first established a strong cortisol-dependent interaction between the GR ligand binding domain and a UBE3A nuclear receptor box in a high-throughput interaction screen. In vivo, we found that UBE3A-deficient AS mice displayed significantly more variation in circulating corticosterone levels throughout the day compared to wildtypes (WT), with low to undetectable levels of corticosterone at the trough of the circadian cycle. Additionally, we observed an enhanced transcriptomic response in the AS hippocampus following acute corticosterone treatment. Surprisingly, chronic corticosterone treatment showed less contrast between AS and WT mice in the hippocampus and liver transcriptomic responses. This suggests that UBE3A limits the acute stimulation of GR signaling, likely as a member of the GR transcriptional complex. Altogether, these data indicate that AS mice are more sensitive to acute glucocorticoid exposure in the brain compared to WT mice. This suggests that stress responsiveness is altered in AS which could lead to anxiety symptoms.
Collapse
Affiliation(s)
- Eva M. G. Viho
- Department of Medicine, Division of Endocrinology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
- Correspondence:
| | - A. Mattijs Punt
- Department of Clinical Genetics, Erasmus MC, 3015 GD Rotterdam, The Netherlands
- ENCORE Expertise Center for Neurodevelopmental Disorders, Erasmus MC, 3015 GD Rotterdam, The Netherlands
| | - Ben Distel
- Department of Clinical Genetics, Erasmus MC, 3015 GD Rotterdam, The Netherlands
- ENCORE Expertise Center for Neurodevelopmental Disorders, Erasmus MC, 3015 GD Rotterdam, The Netherlands
| | - René Houtman
- Precision Medicine Lab, 5349 AB Oss, The Netherlands
| | - Jan Kroon
- Department of Medicine, Division of Endocrinology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Ype Elgersma
- Department of Clinical Genetics, Erasmus MC, 3015 GD Rotterdam, The Netherlands
- ENCORE Expertise Center for Neurodevelopmental Disorders, Erasmus MC, 3015 GD Rotterdam, The Netherlands
| | - Onno C. Meijer
- Department of Medicine, Division of Endocrinology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| |
Collapse
|
7
|
Merabet N, Lucassen PJ, Crielaard L, Stronks K, Quax R, Sloot PMA, la Fleur SE, Nicolaou M. How exposure to chronic stress contributes to the development of type 2 diabetes: A complexity science approach. Front Neuroendocrinol 2022; 65:100972. [PMID: 34929260 DOI: 10.1016/j.yfrne.2021.100972] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 11/24/2021] [Accepted: 12/12/2021] [Indexed: 11/18/2022]
Abstract
Chronic stress contributes to the onset of type 2 diabetes (T2D), yet the underlying etiological mechanisms are not fully understood. Responses to stress are influenced by earlier experiences, sex, emotions and cognition, and involve a complex network of neurotransmitters and hormones, that affect multiple biological systems. In addition, the systems activated by stress can be altered by behavioral, metabolic and environmental factors. The impact of stress on metabolic health can thus be considered an emergent process, involving different types of interactions between multiple variables, that are driven by non-linear dynamics at different spatiotemporal scales. To obtain a more comprehensive picture of the links between chronic stress and T2D, we followed a complexity science approach to build a causal loop diagram (CLD) connecting the various mediators and processes involved in stress responses relevant for T2D pathogenesis. This CLD could help develop novel computational models and formulate new hypotheses regarding disease etiology.
Collapse
Affiliation(s)
- Nadège Merabet
- Department of Public and Occupational Health, Amsterdam UMC, University of Amsterdam, Amsterdam Public Health Research Institute, Meibergdreef 9, Amsterdam, the Netherlands; Institute for Advanced Study, University of Amsterdam, Amsterdam 1012 GC, the Netherlands; Centre for Urban Mental Health, University of Amsterdam, Amsterdam 1012 GC, the Netherlands
| | - Paul J Lucassen
- Centre for Urban Mental Health, University of Amsterdam, Amsterdam 1012 GC, the Netherlands; Brain Plasticity Group, Swammerdam Institute for Life Sciences, Faculty of Science, University of Amsterdam, Amsterdam 1098 XH, the Netherlands
| | - Loes Crielaard
- Department of Public and Occupational Health, Amsterdam UMC, University of Amsterdam, Amsterdam Public Health Research Institute, Meibergdreef 9, Amsterdam, the Netherlands; Institute for Advanced Study, University of Amsterdam, Amsterdam 1012 GC, the Netherlands
| | - Karien Stronks
- Department of Public and Occupational Health, Amsterdam UMC, University of Amsterdam, Amsterdam Public Health Research Institute, Meibergdreef 9, Amsterdam, the Netherlands; Institute for Advanced Study, University of Amsterdam, Amsterdam 1012 GC, the Netherlands; Centre for Urban Mental Health, University of Amsterdam, Amsterdam 1012 GC, the Netherlands
| | - Rick Quax
- Institute for Advanced Study, University of Amsterdam, Amsterdam 1012 GC, the Netherlands; Computational Science Lab, University of Amsterdam, Amsterdam 1098 XH, the Netherlands
| | - Peter M A Sloot
- Institute for Advanced Study, University of Amsterdam, Amsterdam 1012 GC, the Netherlands; Centre for Urban Mental Health, University of Amsterdam, Amsterdam 1012 GC, the Netherlands; Computational Science Lab, University of Amsterdam, Amsterdam 1098 XH, the Netherlands; National Centre of Cognitive Research, ITMO University, St. Petersburg, Russian Federation
| | - Susanne E la Fleur
- Department of Endocrinology and Metabolism & Laboratory of Endocrinology, Department of Clinical Chemistry, Amsterdam Neuroscience, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, Amsterdam, the Netherlands; Metabolism and Reward Group, Netherlands Institute for Neuroscience, an Institute of the Royal Netherlands Academy of Arts and Sciences, Meibergdreef 47, Amsterdam, the Netherlands.
| | - Mary Nicolaou
- Department of Public and Occupational Health, Amsterdam UMC, University of Amsterdam, Amsterdam Public Health Research Institute, Meibergdreef 9, Amsterdam, the Netherlands; Institute for Advanced Study, University of Amsterdam, Amsterdam 1012 GC, the Netherlands; Centre for Urban Mental Health, University of Amsterdam, Amsterdam 1012 GC, the Netherlands.
| |
Collapse
|
8
|
Paul SN, Wingenfeld K, Otte C, Meijer OC. Brain Mineralocorticoid receptor in health and disease: from molecular signaling to cognitive and emotional function. Br J Pharmacol 2022; 179:3205-3219. [PMID: 35297038 PMCID: PMC9323486 DOI: 10.1111/bph.15835] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 02/22/2022] [Accepted: 03/08/2022] [Indexed: 11/27/2022] Open
Abstract
Brain mineralocorticoid receptors (MR) mediate effects of glucocorticoid hormones in stress adaptation, as well as the effects of aldosterone itself in relation to salt homeostasis. Brain stem MRs respond to aldosterone, whereas forebrain MRs mediate rapid and delayed glucocorticoid effects in conjunction with the glucocorticoid receptor (GR). MR‐mediated effects depend on age, gender, genetic variations, and environmental influences. Disturbed MR activity through chronic stress, certain (endocrine) diseases or during glucocorticoid therapy can cause deleterious effects on affective state, cognitive and behavioural function in susceptible individuals. Considering the important role MR plays in cognition and emotional function in health and disease, MR modulation by pharmacological intervention could relieve stress‐ and endocrine‐related symptoms. Here, we discuss recent pharmacological interventions in the clinic and genetic developments in the molecular underpinnings of MR signalling. Further understanding of MR‐dependent pathways may help to improve psychiatric symptoms in a diversity of settings.
Collapse
Affiliation(s)
- Susana N Paul
- Department of Medicine, Division of Endocrinology, Leiden University Medical Center, Leiden, The Netherlands.,Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Katja Wingenfeld
- Klinik für Psychiatrie und Psychotherapie, Charité Universitätsmedizin Campus Benjamin Franklin, Berlin, Germany
| | - Christian Otte
- Klinik für Psychiatrie und Psychotherapie, Charité Universitätsmedizin Campus Benjamin Franklin, Berlin, Germany.,NeuroCure Cluster of Excellence, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Onno C Meijer
- Department of Medicine, Division of Endocrinology, Leiden University Medical Center, Leiden, The Netherlands.,Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
9
|
Buijs RM, Soto Tinoco EC, Hurtado Alvarado G, Escobar C. The circadian system: From clocks to physiology. HANDBOOK OF CLINICAL NEUROLOGY 2021; 179:233-247. [PMID: 34225965 DOI: 10.1016/b978-0-12-819975-6.00013-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
The circadian system, composed of the central autonomous clock, the suprachiasmatic nucleus (SCN), and systems of the body that follow the signals of the SCN, continuously change the homeostatic set points of the body over the day-night cycle. Changes in the body's physiological state that do not agree with the time of the day feedback to the hypothalamus, and provide input to the SCN to adjust the condition, thus reaching another set point required by the changed conditions. This allows the adjustment of the set points to another level when environmental conditions change, which is thought to promote adaptation and survival. In fasting, the body temperature drops to a lower level only at the beginning of the sleep phase. Stressful conditions raise blood pressure relatively more during the active period than during the rest phase. Extensive, mostly reciprocal SCN interactions, with hypothalamic networks, induce these physiological adjustments by hormonal and autonomic control of the body's organs. More importantly, in addition to SCN's hormonal and autonomic influences, SCN induced behavior, such as rhythmic food intake, induces the oscillation of many genes in all tissues, including the so-called clock genes, which have an essential role as a transcriptional driving force for numerous cellular processes. Consequently, the light-dark cycle, the rhythm of the SCN, and the resulting rhythm in behavior need to be perfectly synchronized, especially where it involves synchronizing food intake with the activity phase. If these rhythms are not synchronous for extended periods of times, such as during shift work, light exposure at night, or frequent night eating, disease may develop. As such, our circadian system is a perfect illustration of how hypothalamic-driven processes depend on and interact with each other and need to be in seamless synchrony with the body's physiology.
Collapse
Affiliation(s)
- Ruud M Buijs
- Hypothalamic Integration Mechanisms Laboratory, Department of Cellular Biology and Physiology, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México (UNAM), Ciudad de México, Mexico.
| | - Eva C Soto Tinoco
- Hypothalamic Integration Mechanisms Laboratory, Department of Cellular Biology and Physiology, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México (UNAM), Ciudad de México, Mexico
| | - Gabriela Hurtado Alvarado
- Hypothalamic Integration Mechanisms Laboratory, Department of Cellular Biology and Physiology, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México (UNAM), Ciudad de México, Mexico
| | - Carolina Escobar
- Faculty of Medicine, Universidad Nacional Autónoma de México (UNAM), Ciudad de México, Mexico
| |
Collapse
|
10
|
Ruby CL, Major RJ, Hinrichsen RD. Regulation of tissue regeneration by the circadian clock. Eur J Neurosci 2021; 53:3576-3597. [PMID: 33893679 DOI: 10.1111/ejn.15244] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 03/31/2021] [Accepted: 04/16/2021] [Indexed: 12/12/2022]
Abstract
Circadian rhythms are regulated by a highly conserved transcriptional/translational feedback loop that maintains approximately 24-hr periodicity from cellular to organismal levels. Much research effort is being devoted to understanding how the outputs of the master clock affect peripheral oscillators, and in turn, numerous biological processes. Recent studies have revealed roles for circadian timing in the regulation of numerous cellular behaviours in support of complex tissue regeneration. One such role involves the interaction between the circadian clockwork and the cell cycle. The molecular mechanisms that control the cell cycle create a system of regulation that allows for high fidelity DNA synthesis, mitosis and apoptosis. In recent years, it has become clear that clock gene products are required for proper DNA synthesis and cell cycle progression, and conversely, elements of the cell cycle cascade feedback to influence molecular circadian timing mechanisms. It is through this crosstalk that the circadian system orchestrates stem cell proliferation, niche exit and control of the signalling pathways that govern differentiation and self-renewal. In this review, we discuss the evidence for circadian control of tissue homeostasis and repair and suggest new avenues for research.
Collapse
Affiliation(s)
- Christina L Ruby
- Department of Biology, Indiana University of Pennsylvania, Indiana, PA, USA
| | - Robert J Major
- Department of Biology, Indiana University of Pennsylvania, Indiana, PA, USA
| | | |
Collapse
|
11
|
Pakdeepak K, Chokchaisiri R, Govitrapong P, Tocharus C, Suksamrarn A, Tocharus J. 5,6,7,4'-Tetramethoxyflavanone alleviates neurodegeneration in a dexamethasone-induced neurodegenerative mouse model through promotion of neurogenesis via the Raf/ERK1/2 pathway. Phytother Res 2021; 35:2536-2544. [PMID: 33319436 DOI: 10.1002/ptr.6983] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 11/17/2020] [Accepted: 12/03/2020] [Indexed: 12/27/2022]
Abstract
Adult neurogenesis plays an important role in improving cognitive functions. Neurogenesis generates new neurons, a process mediated by neural stem cell proliferation, migration, and differentiation. Long-term exposure to high levels of glucocorticoid results in the suppression of neurogenesis pathways and leads to the onset of cognitive impairment. The induction of neurogenesis by a potent bioactive compound is considered the most promising treatment for neurodegenerative disorders. 5,6,7,4'-Tetramethoxyflavanone (TMF) is a flavonoid compound isolated from Chromolaena odorata (L.) R. M. King & H. Rob. Previous study showed that TMF improved cognitive impairment by attenuating Aβ production and pTau expression, thereby increased cell survival and promoted synaptic plasticity. The aim of this study was to investigate the effect of TMF on dexamethasone (DEX)-suppressed neurogenesis in mice. Mice received DEX for 28 days before being treated with TMF for additional 30 days. Mice were randomly divided into four groups: control, TMF, DEX, and DEX + TMF. TMF promoted neurogenesis by increasing BrdU-positive cells, Prox1, doublecortin, and Nestin expression. TMF also upregulated the expression of Raf and extracellular-signal-regulated kinase (ERK)1/2, which are pivotal for neurogenesis signaling. In conclusion, TMF promoted neurogenesis-related protein expression in the proliferation, differentiation, and maturation phases via Raf/ERK1/2 signaling pathway.
Collapse
Affiliation(s)
- Kanet Pakdeepak
- Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
- Graduate School, Chiang Mai University, Chiang Mai, Thailand
| | | | | | - Chainarong Tocharus
- Department of Anatomy, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Apichart Suksamrarn
- Department of Chemistry and Center of Excellence for Innovation in Chemistry, Faculty of Science, Ramkhamhaeng University, Bangkok, Thailand
| | - Jiraporn Tocharus
- Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
- Functional Food Research Center for Well-being, Chiang Mai University, Chiang Mai, Thailand
| |
Collapse
|
12
|
Luo OD, Kwiecien-Delaney B, Martin P, Foster JA, Sidor MM. The effect of early life immune challenge on adult forced swim test performance and hippocampal neurogenesis. J Neuroimmunol 2021; 354:577530. [PMID: 33744708 DOI: 10.1016/j.jneuroim.2021.577530] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 02/22/2021] [Accepted: 02/22/2021] [Indexed: 10/22/2022]
Abstract
Many psychiatric diseases can be considered neurodevelopmental in nature and accumulating evidence links immune system dysfunction to disease etiology. Yet, it is currently unknown how the immune system alters brain function through development to increase susceptibility to psychiatric illness. Neonatal immune challenge in rodents is a neurodevelopmental model that has been associated with long-term molecular and behavioural changes in stress-reactivity. As enhanced stress-reactivity is associated with the emergence of depressive-like behaviours concurrent with hippocampal pathology, we measured depressive-like behaviour in the forced swim test and hippocampal neurogenesis in adult mice neonatally exposed to lipopolysaccharide LPS; 0.05 mg/kg, i.p. on postnatal days 3 and 5. As there are important functional differences along the ventral-dorsal hippocampus axis, ventral and dorsal hippocampal neurogenesis were measured separately. Our findings reveal a sexually-dimorphic response to early-life LPS challenge. Male LPS-mice spent less time immobile in the forced swim test, suggesting altered reactivity to swim stress. This was accompanied by an increase in doublecortin-positive cells in the dorsal hippocampus of female mice. These findings demonstrate that exposure to an immune challenge during critical developmental time periods leads to long-term sexually-dimorphic alterations in stress-reactivity that are accompanied by changes to adult hippocampal neurogenesis.
Collapse
Affiliation(s)
- Owen D Luo
- Psychiatry and Behavioural Neuroscience, McMaster University, Hamilton, Ontario, Canada
| | | | - Patrick Martin
- Psychiatry and Behavioural Neuroscience, McMaster University, Hamilton, Ontario, Canada
| | - Jane A Foster
- Psychiatry and Behavioural Neuroscience, McMaster University, Hamilton, Ontario, Canada; Department of Psychiatry, St. Michael's Hospital, Toronto, Ontario, Canada.
| | - Michelle M Sidor
- Psychiatry and Behavioural Neuroscience, McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|
13
|
Gomes PRL, Motta-Teixeira LC, Gallo CC, Carmo Buonfiglio DD, Camargo LSD, Quintela T, Reiter RJ, Amaral FGD, Cipolla-Neto J. Maternal pineal melatonin in gestation and lactation physiology, and in fetal development and programming. Gen Comp Endocrinol 2021; 300:113633. [PMID: 33031801 DOI: 10.1016/j.ygcen.2020.113633] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Accepted: 09/20/2020] [Indexed: 12/21/2022]
Abstract
Pregnancy and lactation are reproductive processes that rely on physiological adaptations that should be timely and adequately triggered to guarantee both maternal and fetal health. Pineal melatonin is a hormone that presents daily and seasonal variations that synchronizes the organism's physiology to the different demands across time through its specific mechanisms and ways of action. The reproductive system is a notable target for melatonin as it actively participates on reproductive physiology and regulates the hypothalamus-pituitary-gonads axis, influencing gonadotropins and sexual hormones synthesis and release. For its antioxidant properties, melatonin is also vital for the oocytes and spermatozoa quality and viability, and for blastocyst development. Maternal pineal melatonin blood levels increase during pregnancy and triggers the maternal physiological alterations in energy metabolism both during pregnancy and lactation to cope with the energy demands of both periods and to promote adequate mammary gland development. Moreover, maternal melatonin freely crosses the placenta and is the only source of this hormone to the fetus. It importantly times the conceptus physiology and influences its development and programing of several functions that depend on neural and brain development, ultimately priming adult behavior and energy and glucose metabolism. The present review aims to explain the above listed melatonin functions, including the potential alterations observed in the progeny gestated under maternal chronodisruption and/or hypomelatoninemia.
Collapse
Affiliation(s)
- Patrícia Rodrigues Lourenço Gomes
- Neurobiology Lab, Department of Physiology and Biophysics, 1524 Prof. Lineu Prestes Ave., Institute of Biomedical Sciences, Bldg 1, Lab 118, University of São Paulo, São Paulo 05508-000, Brazil
| | - Lívia Clemente Motta-Teixeira
- Neurobiology Lab, Department of Physiology and Biophysics, 1524 Prof. Lineu Prestes Ave., Institute of Biomedical Sciences, Bldg 1, Lab 118, University of São Paulo, São Paulo 05508-000, Brazil
| | - Camila Congentino Gallo
- Pineal Neurobiology Lab, Department of Physiology, 862 Botucatu St., 5th floor, Federal University of São Paulo, São Paulo 04023-901, Brazil.
| | - Daniella do Carmo Buonfiglio
- Neurobiology Lab, Department of Physiology and Biophysics, 1524 Prof. Lineu Prestes Ave., Institute of Biomedical Sciences, Bldg 1, Lab 118, University of São Paulo, São Paulo 05508-000, Brazil
| | - Ludmilla Scodeler de Camargo
- Pineal Neurobiology Lab, Department of Physiology, 862 Botucatu St., 5th floor, Federal University of São Paulo, São Paulo 04023-901, Brazil.
| | - Telma Quintela
- CICS-UBI - Health Sciences Research Center, Infante D. Henrique Ave, University of Beira Interior, Covilhã 6200-506, Portugal.
| | - Russel J Reiter
- Department of Cell Systems and Anatomy, Long School of Medicine, 7703 Floyd Curl Drive, UT Health San Antonio, San Antonio, TX 78229, USA.
| | - Fernanda Gaspar do Amaral
- Pineal Neurobiology Lab, Department of Physiology, 862 Botucatu St., 5th floor, Federal University of São Paulo, São Paulo 04023-901, Brazil.
| | - José Cipolla-Neto
- Neurobiology Lab, Department of Physiology and Biophysics, 1524 Prof. Lineu Prestes Ave., Institute of Biomedical Sciences, Bldg 1, Lab 118, University of São Paulo, São Paulo 05508-000, Brazil.
| |
Collapse
|
14
|
Sjörs Dahlman A, Jonsdottir IH, Hansson C. The hypothalamo-pituitary-adrenal axis and the autonomic nervous system in burnout. HANDBOOK OF CLINICAL NEUROLOGY 2021; 182:83-94. [PMID: 34266613 DOI: 10.1016/b978-0-12-819973-2.00006-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Burnout constitutes a serious health concern in the modern working environment. It is a stress-related condition that has developed as a result of a prolonged psychosocial stress exposure causing a persistent mismatch between demands and resources. The main symptom is emotional exhaustion, but physical fatigue, diminished professional efficacy, cynicism, and cognitive impairments are also associated with this condition. Burnout has been used both as a psychologic term in occupational settings and as a clinical diagnosis in patient populations, and there is currently no universally accepted definition and diagnostic criteria of burnout. It has been hypothesized that the two main stress response systems, the autonomic nervous system (ANS) and the hypothalamus-pituitary-adrenal axis (HPA axis), are involved in the pathogenesis of burnout. A common hypothesis is that in the early stages of chronic stress, the HPA axis and sympathetic ANS activity tend to be higher, while it will decrease with a longer duration of chronic stress to ultimately reach a state of hypoactivity in clinical burnout. The current research in this field shows many contradictory results. Thus there is no compelling evidence of either ANS or HPA dysfunction in burnout. However, there is partial support for the hypothesis of HPA and sympathetic hyperactivity in early stages, and HPA hyporeactivity and low vagal activity in more severe burnout cases, but high-quality studies investigating the causal links are still lacking.
Collapse
Affiliation(s)
- Anna Sjörs Dahlman
- Institute of Stress Medicine, Region Västra Götaland, Gothenburg, Sweden; Human Factors Department, Swedish National Road and Transport Research Institute, Gothenburg, Sweden.
| | - Ingibjörg H Jonsdottir
- Institute of Stress Medicine, Region Västra Götaland, Gothenburg, Sweden; School of Public Health and Community Medicine, Institute of Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Caroline Hansson
- Institute of Stress Medicine, Region Västra Götaland, Gothenburg, Sweden; Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
15
|
Podgorny OV, Gulyaeva NV. Glucocorticoid-mediated mechanisms of hippocampal damage: Contribution of subgranular neurogenesis. J Neurochem 2020; 157:370-392. [PMID: 33301616 DOI: 10.1111/jnc.15265] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 11/09/2020] [Accepted: 11/30/2020] [Indexed: 12/19/2022]
Abstract
A comprehensive overview of the interplay between glucocorticoids (GCs) and adult hippocampal neurogenesis (AHN) is presented, particularly, in the context of a diseased brain. The effectors of GCs in the dentate gyrus neurogenic niche of the hippocampal are reviewed, and the consequences of the GC signaling on the generation and integration of new neurons are discussed. Recent findings demonstrating how GC signaling mediates impairments of the AHN in various brain pathologies are overviewed. GC-mediated effects on the generation and integration of adult-born neurons in the hippocampal dentate gyrus depend on the nature, severity, and duration of the acting stress factor. GCs realize their effects on the AHN primarily via specific glucocorticoid and mineralocorticoid receptors. Disruption of the reciprocal regulation between the hypothalamic-pituitary-adrenal (HPA) axis and the generation of the adult-born granular neurons is currently considered to be a key mechanism implicating the AHN into the pathogenesis of numerous brain diseases, including those without a direct hippocampal damage. These alterations vary from reduced proliferation of stem and progenitor cells to increased cell death and abnormalities in morphology, connectivity, and localization of young neurons. Although the involvement of the mutual regulation between the HPA axis and the AHN in the pathogenesis of cognitive deficits and mood impairments is evident, several unresolved critical issues are stated. Understanding the details of GC-mediated mechanisms involved in the alterations in AHN could enable the identification of molecular targets for ameliorating pathology-induced imbalance in the HPA axis/AHN mutual regulation to conquer cognitive and psychiatric disturbances.
Collapse
Affiliation(s)
- Oleg V Podgorny
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia.,Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, Moscow, Russia.,Koltzov Institute of Developmental Biology, Russian Academy of Sciences, Moscow, Russia
| | - Natalia V Gulyaeva
- Institute of Higher Nervous Activity and Neurophysiology, Russian Academy of Sciences, Moscow, Russia.,Research and Clinical Center for Neuropsychiatry of Moscow Healthcare Department, Moscow, Russia
| |
Collapse
|
16
|
Cognitive and emotional empathy after stimulation of brain mineralocorticoid and NMDA receptors in patients with major depression and healthy controls. Neuropsychopharmacology 2020; 45:2155-2161. [PMID: 32722659 PMCID: PMC7785026 DOI: 10.1038/s41386-020-0777-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 06/19/2020] [Accepted: 07/10/2020] [Indexed: 02/06/2023]
Abstract
Mineralocorticoid receptors (MR) are predominantly expressed in the hippocampus and prefrontal cortex. Both brain areas are associated with social cognition, which includes cognitive empathy (ability to understand others' emotions) and emotional empathy (ability to empathize with another person). MR stimulation improves memory and executive functioning in patients with major depressive disorder (MDD) and healthy controls, and leads to glutamate-mediated N-methyl-D-aspartate receptor (NMDA-R) signaling. We examined whether the beneficial effects of MR stimulation can be extended to social cognition (empathy), and whether DCS would have additional beneficial effects. In this double-blind placebo-controlled single-dose study, we randomized 116 unmedicated MDD patients (mean age 34 years, 78% women) and 116 age-, sex-, and education years-matched healthy controls to four conditions: MR stimulation (fludrocortisone (0.4 mg) + placebo), NMDA-R stimulation (placebo + D-cycloserine (250 mg)), MR and NMDA-R stimulation (both drugs), or placebo. Cognitive and emotional empathy were assessed by the Multifaceted Empathy Test. The study was registered on clinicaltrials.gov (NCT03062150). MR stimulation increased cognitive empathy across groups, whereas NMDA-R stimulation decreased cognitive empathy in MDD patients only. Independent of receptor stimulation, cognitive empathy did not differ between groups. Emotional empathy was not affected by MR or NMDA-R stimulation. However, MDD patients showed decreased emotional empathy compared with controls but, according to exploratory analyses, only for positive emotions. We conclude that MR stimulation has beneficial effects on cognitive empathy in MDD patients and healthy controls, whereas NMDA-R stimulation decreased cognitive empathy in MDD patients. It appears that MR rather than NMDA-R are potential treatment targets to modulate cognitive empathy in MDD.
Collapse
|
17
|
Schouten M, Bielefeld P, Garcia-Corzo L, Passchier EMJ, Gradari S, Jungenitz T, Pons-Espinal M, Gebara E, Martín-Suárez S, Lucassen PJ, De Vries HE, Trejo JL, Schwarzacher SW, De Pietri Tonelli D, Toni N, Mira H, Encinas JM, Fitzsimons CP. Circadian glucocorticoid oscillations preserve a population of adult hippocampal neural stem cells in the aging brain. Mol Psychiatry 2020; 25:1382-1405. [PMID: 31222184 PMCID: PMC7303016 DOI: 10.1038/s41380-019-0440-2] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Revised: 04/09/2019] [Accepted: 04/29/2019] [Indexed: 12/17/2022]
Abstract
A decrease in adult hippocampal neurogenesis has been linked to age-related cognitive impairment. However, the mechanisms involved in this age-related reduction remain elusive. Glucocorticoid hormones (GC) are important regulators of neural stem/precursor cells (NSPC) proliferation. GC are released from the adrenal glands in ultradian secretory pulses that generate characteristic circadian oscillations. Here, we investigated the hypothesis that GC oscillations prevent NSPC activation and preserve a quiescent NSPC pool in the aging hippocampus. We found that hippocampal NSPC populations lacking expression of the glucocorticoid receptor (GR) decayed exponentially with age, while GR-positive populations decayed linearly and predominated in the hippocampus from middle age onwards. Importantly, GC oscillations controlled NSPC activation and GR knockdown reactivated NSPC proliferation in aged mice. When modeled in primary hippocampal NSPC cultures, GC oscillations control cell cycle progression and induce specific genome-wide DNA methylation profiles. GC oscillations induced lasting changes in the methylation state of a group of gene promoters associated with cell cycle regulation and the canonical Wnt signaling pathway. Finally, in a mouse model of accelerated aging, we show that disruption of GC oscillations induces lasting changes in dendritic complexity, spine numbers and morphology of newborn granule neurons. Together, these results indicate that GC oscillations preserve a population of GR-expressing NSPC during aging, preventing their activation possibly by epigenetic programming through methylation of specific gene promoters. Our observations suggest a novel mechanism mediated by GC that controls NSPC proliferation and preserves a dormant NSPC pool, possibly contributing to a neuroplasticity reserve in the aging brain.
Collapse
Affiliation(s)
- M Schouten
- Neuroscience Collaboration, Swammerdam Institute for Life Sciences, Faculty of Sciences, Amsterdam Neuroscience, University of Amsterdam, Amsterdam, The Netherlands
- Department of Molecular Cell Biology and Immunology, VU University Medical Center, Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - P Bielefeld
- Neuroscience Collaboration, Swammerdam Institute for Life Sciences, Faculty of Sciences, Amsterdam Neuroscience, University of Amsterdam, Amsterdam, The Netherlands
| | - L Garcia-Corzo
- Biomedicine Institute of Valencia (IBV), Consejo Superior de Investigaciones Científicas (CSIC), Valencia, Spain
| | - E M J Passchier
- Neuroscience Collaboration, Swammerdam Institute for Life Sciences, Faculty of Sciences, Amsterdam Neuroscience, University of Amsterdam, Amsterdam, The Netherlands
| | - S Gradari
- Cajal Institute, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - T Jungenitz
- Institute of Clinical Neuroanatomy, Neuroscience Center, Goethe-University Frankfurt, Frankfurt am Main, Germany
| | - M Pons-Espinal
- Neurobiology of miRNA Lab, Neuroscience and Brain Technologies Department, Istituto Italiano di Tecnologia, Genoa, Italy
| | - E Gebara
- Center for Psychiatric Neuroscience, Department of Psychiatry, Lausanne University Hospital (CHUV), Lausanne, Switzerland
| | | | - P J Lucassen
- Neuroscience Collaboration, Swammerdam Institute for Life Sciences, Faculty of Sciences, Amsterdam Neuroscience, University of Amsterdam, Amsterdam, The Netherlands
| | - H E De Vries
- Department of Molecular Cell Biology and Immunology, VU University Medical Center, Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - J L Trejo
- Cajal Institute, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - S W Schwarzacher
- Institute of Clinical Neuroanatomy, Neuroscience Center, Goethe-University Frankfurt, Frankfurt am Main, Germany
| | - D De Pietri Tonelli
- Neurobiology of miRNA Lab, Neuroscience and Brain Technologies Department, Istituto Italiano di Tecnologia, Genoa, Italy
| | - N Toni
- Center for Psychiatric Neuroscience, Department of Psychiatry, Lausanne University Hospital (CHUV), Lausanne, Switzerland
| | - H Mira
- Biomedicine Institute of Valencia (IBV), Consejo Superior de Investigaciones Científicas (CSIC), Valencia, Spain
| | - J M Encinas
- Achucarro Basque Center for Neuroscience, Leioa, Spain
- Ikerbasque, The Basque Foundation for Science, Bilbao, Spain
- University of the Basque Country (UPV/EHU), Leioa, Spain
| | - C P Fitzsimons
- Neuroscience Collaboration, Swammerdam Institute for Life Sciences, Faculty of Sciences, Amsterdam Neuroscience, University of Amsterdam, Amsterdam, The Netherlands.
| |
Collapse
|
18
|
Benitah SA, Welz PS. Circadian Regulation of Adult Stem Cell Homeostasis and Aging. Cell Stem Cell 2020; 26:817-831. [DOI: 10.1016/j.stem.2020.05.002] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
19
|
Bulin SE, Simmons SJ, Richardson DR, Latchney SE, Deutsch HM, Yun S, Eisch AJ. Indices of dentate gyrus neurogenesis are unaffected immediately after or following withdrawal from morphine self-administration compared to saline self-administering control male rats. Behav Brain Res 2020; 381:112448. [PMID: 31870778 PMCID: PMC7036141 DOI: 10.1016/j.bbr.2019.112448] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Revised: 12/01/2019] [Accepted: 12/19/2019] [Indexed: 12/16/2022]
Abstract
Opiates - including morphine - are powerful analgesics with high abuse potential. In rodents, chronic opiate exposure or self-administration negatively impacts hippocampal-dependent function, an effect perhaps due in part to the well-documented opiate-induced inhibition of dentate gyrus (DG) precursor proliferation and neurogenesis. Recently, however, intravenous (i.v.) morphine self-administration (MSA) was reported to enhance the survival of new rat DG neurons. To reconcile these disparate results, we used rat i.v. MSA to assess 1) whether a slightly-higher dose MSA paradigm also increases new DG neuron survival; 2) how MSA influences cells in different stages of DG neurogenesis, particularly maturation and survival; and 3) if MSA-induced changes in DG neurogenesis persist through a period of abstinence. To label basal levels of proliferation, rats received the S-phase marker bromodeoxyuridine (BrdU, i.p.) 24 -h prior to 21 days (D) of i.v. MSA or saline self-administration (SSA). Either immediately after SA (0-D) or after 4 weeks in the home cage (28-D withdrawal), stereology was used to quantify DG proliferating precursors (or cells in cell cycle; Ki67+ cells), neuroblast/immature neurons (DCX+ cells), and surviving DG granule cells (BrdU+ cells). Analysis revealed the number of DG cells immunopositive for these neurogenesis-relevant markers was similar between MSA and SSA rats at the 0-D or 28-D timepoints. These negative data highlight the impact experimental parameters, timepoint selection, and quantification approach have on neurogenesis results, and are discussed in the context of the large literature showing the negative impact of opiates on DG neurogenesis.
Collapse
Affiliation(s)
- Sarah E Bulin
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Steven J Simmons
- Department of Anesthesiology and Critical Care Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Devon R Richardson
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Sarah E Latchney
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, USA; Department of Neurobiology, St. Mary's College of Maryland, St. Mary's City, MD, 20686-3001
| | - Hannah M Deutsch
- Department of Anesthesiology and Critical Care Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Sanghee Yun
- Department of Anesthesiology and Critical Care Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Amelia J Eisch
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, USA; Department of Anesthesiology and Critical Care Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA; Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
20
|
Adult hippocampal neurogenesis and antidepressants effects. Curr Opin Pharmacol 2020; 50:88-95. [DOI: 10.1016/j.coph.2019.11.009] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Accepted: 11/21/2019] [Indexed: 12/11/2022]
|
21
|
Planchez B, Surget A, Belzung C. WITHDRAWN: Adult hippocampal neurogenesis and antidepressants effects. Curr Opin Pharmacol 2020; 50:17-24. [DOI: 10.1016/j.coph.2019.10.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Revised: 09/27/2019] [Accepted: 10/18/2019] [Indexed: 12/22/2022]
|
22
|
Salivary cortisol as a non-invasive window on the brain. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2020; 150:1-16. [PMID: 32204827 DOI: 10.1016/bs.irn.2019.12.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/08/2022]
Abstract
The validation of accurate and meaningful assessment of cortisol in saliva samples has proved revolutionary in stress research. Its many advantages have expanded the scope of investigation from traditional laboratory and clinical settings to include multidisciplinary and community-based research. These developments have given rise to a wealth insight into the links between stress and health. Here we highlight the potential of salivary cortisol as both a product and mediator of brain function, instrumental in disturbing brain health. However, the subtleties of salivary cortisol as a measure can be underestimated, leading to misinterpretation of findings. These issues are explored, with a particular emphasis on necessary methodological rigor. Notwithstanding great promise, there is undeniably more to learn so we conclude by making recommendations for future research including use of salivary cortisol in the development of integrative predictive models of stress-related risk factors and resilience across the life course.
Collapse
|
23
|
Unno K, Sumiyoshi A, Konishi T, Hayashi M, Taguchi K, Muguruma Y, Inoue K, Iguchi K, Nonaka H, Kawashima R, Hasegawa-Ishii S, Shimada A, Nakamura Y. Theanine, the Main Amino Acid in Tea, Prevents Stress-Induced Brain Atrophy by Modifying Early Stress Responses. Nutrients 2020; 12:nu12010174. [PMID: 31936294 PMCID: PMC7019546 DOI: 10.3390/nu12010174] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 12/30/2019] [Accepted: 01/04/2020] [Indexed: 11/16/2022] Open
Abstract
Chronic stress can impair the health of human brains. An important strategy that may prevent the accumulation of stress may be the consumption of functional foods. When senescence-accelerated mice prone 10 (SAMP10), a stress-sensitive strain, were loaded with stress using imposed male mouse territoriality, brain volume decreased. However, in mice that ingested theanine (6 mg/kg), the main amino acid in tea leaves, brain atrophy was suppressed, even under stress. On the other hand, brain atrophy was not clearly observed in a mouse strain that aged normally (Slc:ddY). The expression level of the transcription factor Npas4 (neuronal PAS domain protein 4), which regulates the formation and maintenance of inhibitory synapses in response to excitatory synaptic activity, decreased in the hippocampus and prefrontal cortex of stressed SAMP10 mice, but increased in mice that ingested theanine. Lipocalin 2 (Lcn2), the expression of which increased in response to stress, was significantly high in the hippocampus and prefrontal cortex of stressed SAMP10 mice, but not in mice that ingested theanine. These data suggest that Npas4 and Lcn2 are involved in the brain atrophy and stress vulnerability of SAMP10 mice, which are prevented by the consumption of theanine, causing changes in the expression of these genes.
Collapse
Affiliation(s)
- Keiko Unno
- Tea Science Center, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan; (M.H.); (K.T.); (Y.N.)
- School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan;
- Correspondence: ; Tel.: +81-54-264-5822
| | - Akira Sumiyoshi
- Institute of Development, Aging and Cancer, Tohoku University, 4-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Japan; (A.S.); (H.N.); (R.K.)
- National Institutes for Quantum and Radiological Science and Technology, 4-9-1, Anagawa, Inage-ku, Chiba 263-8555, Japan
| | - Tomokazu Konishi
- Faculty of Bioresource Sciences, Akita Prefectural University, Shimoshinjo Nakano, Akita 010-0195, Japan;
| | - Michiko Hayashi
- Tea Science Center, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan; (M.H.); (K.T.); (Y.N.)
| | - Kyoko Taguchi
- Tea Science Center, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan; (M.H.); (K.T.); (Y.N.)
| | - Yoshio Muguruma
- College of Pharmaceutical Sciences, Ritsumeikan University, 1-1-1 Nojihigashi, Kusatsu, Shiga 525-8577, Japan; (Y.M.); (K.I.)
| | - Koichi Inoue
- College of Pharmaceutical Sciences, Ritsumeikan University, 1-1-1 Nojihigashi, Kusatsu, Shiga 525-8577, Japan; (Y.M.); (K.I.)
| | - Kazuaki Iguchi
- School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan;
| | - Hiroi Nonaka
- Institute of Development, Aging and Cancer, Tohoku University, 4-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Japan; (A.S.); (H.N.); (R.K.)
| | - Ryuta Kawashima
- Institute of Development, Aging and Cancer, Tohoku University, 4-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Japan; (A.S.); (H.N.); (R.K.)
| | - Sanae Hasegawa-Ishii
- Faculty of Health Sciences, Kyorin University, 5-4-1 Shimorenjaku, Mitaka, Tokyo 181-8612, Japan; (S.H.-I.); (A.S.)
| | - Atsuyoshi Shimada
- Faculty of Health Sciences, Kyorin University, 5-4-1 Shimorenjaku, Mitaka, Tokyo 181-8612, Japan; (S.H.-I.); (A.S.)
| | - Yoriyuki Nakamura
- Tea Science Center, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan; (M.H.); (K.T.); (Y.N.)
| |
Collapse
|
24
|
Guest PC. Proteomic Analysis of Brain Tissue from a Chronic Model of Stress Using a Combined 2D Gel Electrophoresis and Mass Spectrometry Approach. Methods Mol Biol 2020; 2138:391-406. [PMID: 32219766 DOI: 10.1007/978-1-0716-0471-7_29] [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] [Indexed: 06/10/2023]
Abstract
Aging of the brain can result in excessive glucocorticoid secretion, potentially due to chronic stress and related situations. This can lead to dysfunction of brain areas involved in control of the hypothalamic-pituitary adrenal axis, growth, and metabolism, as well as areas associated with cognition and mood regulation. This chapter presents a protocol for two-dimensional differential in-gel electrophoresis (2D-DIGE) analysis of hypothalamus and hippocampus tissue obtained from mice following exposure to high levels of corticosterone for 14 days. The chapter also presents a method for identification of the affected proteins in these brain regions using matrix-assisted laser desorption/ionization time-of-flight (MALDI-TOF) mass spectrometry.
Collapse
Affiliation(s)
- Paul C Guest
- Laboratory of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, SP, Brazil.
| |
Collapse
|
25
|
Gulyaeva NV. Biochemical Mechanisms and Translational Relevance of Hippocampal Vulnerability to Distant Focal Brain Injury: The Price of Stress Response. BIOCHEMISTRY (MOSCOW) 2019; 84:1306-1328. [PMID: 31760920 DOI: 10.1134/s0006297919110087] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Focal brain injuries (in particular, stroke and traumatic brain injury) induce with high probability the development of delayed (months, years) cognitive and depressive disturbances which are frequently comorbid. The association of these complications with hippocampal alterations (in spite of the lack of a primary injury of this structure), as well as the lack of a clear dependence between the probability of depression and dementia development and primary damage severity and localization served as the basis for a new hypothesis on the distant hippocampal damage as a key link in the pathogenesis of cognitive and psychiatric disturbances. According to this hypothesis, the excess of corticosteroids secreted after a focal brain damage, in particular in patients with abnormal stress-response due to hypothalamic-pituitary-adrenal axis (HPAA) dysfunction, interacts with corticosteroid receptors in the hippocampus inducing signaling pathways which stimulate neuroinflammation and subsequent events including disturbances in neurogenesis and hippocampal neurodegeneration. In this article, the molecular and cellular mechanisms associated with the regulatory role of the HPAA and multiple functions of brain corticosteroid receptors in the hippocampus are analyzed. Functional and structural damage to the hippocampus, a brain region selectively vulnerable to external factors and responding to them by increased cytokine secretion, forms the basis for cognitive function disturbances and psychopathology development. This concept is confirmed by our own experimental data, results of other groups and by prospective clinical studies of post-stroke complications. Clinically relevant biochemical approaches to predict the risks and probability of post-stroke/post-trauma cognitive and depressive disturbances are suggested using the evaluation of biochemical markers of patients' individual stress-response. Pathogenetically justified ways for preventing these consequences of focal brain damage are proposed by targeting key molecular mechanisms underlying hippocampal dysfunction.
Collapse
Affiliation(s)
- N V Gulyaeva
- Institute of Higher Nervous Activity and Neurophysiology, Russian Academy of Sciences, Moscow, 117485, Russia. .,Moscow Research and Clinical Center for Neuropsychiatry, Healthcare Department of Moscow, Moscow, 115419, Russia
| |
Collapse
|
26
|
Magomedova L, Tiefenbach J, Zilberman E, Le Billan F, Voisin V, Saikali M, Boivin V, Robitaille M, Gueroussov S, Irimia M, Ray D, Patel R, Xu C, Jeyasuria P, Bader GD, Hughes TR, Morris QD, Scott MS, Krause H, Angers S, Blencowe BJ, Cummins CL. ARGLU1 is a transcriptional coactivator and splicing regulator important for stress hormone signaling and development. Nucleic Acids Res 2019; 47:2856-2870. [PMID: 30698747 PMCID: PMC6451108 DOI: 10.1093/nar/gkz010] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2017] [Revised: 12/21/2018] [Accepted: 01/04/2019] [Indexed: 12/17/2022] Open
Abstract
Stress hormones bind and activate the glucocorticoid receptor (GR) in many tissues including the brain. We identified arginine and glutamate rich 1 (ARGLU1) in a screen for new modulators of glucocorticoid signaling in the CNS. Biochemical studies show that the glutamate rich C-terminus of ARGLU1 coactivates multiple nuclear receptors including the glucocorticoid receptor (GR) and the arginine rich N-terminus interacts with splicing factors and binds to RNA. RNA-seq of neural cells depleted of ARGLU1 revealed significant changes in the expression and alternative splicing of distinct genes involved in neurogenesis. Loss of ARGLU1 is embryonic lethal in mice, and knockdown in zebrafish causes neurodevelopmental and heart defects. Treatment with dexamethasone, a GR activator, also induces changes in the pattern of alternatively spliced genes, many of which were lost when ARGLU1 was absent. Importantly, the genes found to be alternatively spliced in response to glucocorticoid treatment were distinct from those under transcriptional control by GR, suggesting an additional mechanism of glucocorticoid action is present in neural cells. Our results thus show that ARGLU1 is a novel factor for embryonic development that modulates basal transcription and alternative splicing in neural cells with consequences for glucocorticoid signaling.
Collapse
Affiliation(s)
- Lilia Magomedova
- Department of Pharmaceutical Sciences, University of Toronto, Toronto, ON M5S 3M2, Canada
| | - Jens Tiefenbach
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON M5S 3E1, Canada
| | - Emma Zilberman
- Department of Pharmaceutical Sciences, University of Toronto, Toronto, ON M5S 3M2, Canada
| | - Florian Le Billan
- Department of Pharmaceutical Sciences, University of Toronto, Toronto, ON M5S 3M2, Canada
| | - Veronique Voisin
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON M5S 3E1, Canada
| | - Michael Saikali
- Department of Pharmaceutical Sciences, University of Toronto, Toronto, ON M5S 3M2, Canada
| | - Vincent Boivin
- Département de biochimie, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Sherbrooke, QC J1E 4K8, Canada
| | - Melanie Robitaille
- Department of Pharmaceutical Sciences, University of Toronto, Toronto, ON M5S 3M2, Canada
| | - Serge Gueroussov
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON M5S 3E1, Canada.,Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Manuel Irimia
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON M5S 3E1, Canada
| | - Debashish Ray
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON M5S 3E1, Canada
| | - Rucha Patel
- Department of Pharmaceutical Sciences, University of Toronto, Toronto, ON M5S 3M2, Canada
| | - ChangJiang Xu
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON M5S 3E1, Canada
| | - Pancharatnam Jeyasuria
- Department of Obstetrics and Gynecology, Wayne State University Perinatal Initiative, School of Medicine, Wayne State University, Detroit, MI, USA
| | - Gary D Bader
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON M5S 3E1, Canada.,Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Timothy R Hughes
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON M5S 3E1, Canada.,Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Quaid D Morris
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON M5S 3E1, Canada.,Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Michelle S Scott
- Département de biochimie, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Sherbrooke, QC J1E 4K8, Canada
| | - Henry Krause
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON M5S 3E1, Canada
| | - Stephane Angers
- Department of Pharmaceutical Sciences, University of Toronto, Toronto, ON M5S 3M2, Canada.,Department of Biochemistry,University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Benjamin J Blencowe
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON M5S 3E1, Canada.,Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Carolyn L Cummins
- Department of Pharmaceutical Sciences, University of Toronto, Toronto, ON M5S 3M2, Canada
| |
Collapse
|
27
|
Kozareva DA, Cryan JF, Nolan YM. Born this way: Hippocampal neurogenesis across the lifespan. Aging Cell 2019; 18:e13007. [PMID: 31298475 PMCID: PMC6718573 DOI: 10.1111/acel.13007] [Citation(s) in RCA: 89] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Revised: 05/31/2019] [Accepted: 06/30/2019] [Indexed: 12/30/2022] Open
Abstract
The capability of the mammalian brain to generate new neurons through the lifespan has gained much attention for the promise of new therapeutic possibilities especially for the aging brain. One of the brain regions that maintains a neurogenesis-permissive environment is the dentate gyrus of the hippocampus. Here, new neurons are generated from a pool of multipotent neural progenitor cells to become fully functional neurons that are integrated into the brain circuitry. A growing body of evidence points to the fact that neurogenesis in the adult hippocampus is necessary for certain memory processes, and in mood regulation, while alterations in hippocampal neurogenesis have been associated with a myriad of neurological and psychiatric disorders. More recently, evidence has come to light that new neurons may differ in their vulnerability to environmental and disease-related influences depending on the time during the life course at which they are exposed. Thus, it has been the topic of intense research in recent years. In this review, we will discuss the complex process and associated functional relevance of hippocampal neurogenesis during the embryonic/postnatal period and in adulthood. We consider the implications of hippocampal neurogenesis during the developmentally critical periods of adolescence and older age. We will further consider the literature surrounding hippocampal neurogenesis and its functional role during these critical periods with a view to providing insight into the potential of harnessing neurogenesis for health and therapeutic benefit.
Collapse
Affiliation(s)
- Danka A. Kozareva
- Department of Anatomy & NeuroscienceUniversity College CorkCorkIreland
| | - John F. Cryan
- Department of Anatomy & NeuroscienceUniversity College CorkCorkIreland
- APC Microbiome IrelandUniversity College CorkCorkIreland
| | - Yvonne M. Nolan
- Department of Anatomy & NeuroscienceUniversity College CorkCorkIreland
| |
Collapse
|
28
|
Heck SO, Zborowski VA, Pinton S, Nogueira CW. Pro-apoptotic cell signaling in the prefrontal cortex contributes to depressive-/anxiogenic-like behavioral phenotype of mice subchronically exposed to dexamethasone. J Chem Neuroanat 2019; 100:101663. [PMID: 31374258 DOI: 10.1016/j.jchemneu.2019.101663] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2019] [Revised: 07/19/2019] [Accepted: 07/29/2019] [Indexed: 12/19/2022]
Affiliation(s)
- Suélen Osório Heck
- Laboratory of Synthesis, Reactivity, Pharmacological and Toxicological Evaluation of Organochalcogen Compounds, Department of Biochemistry and Molecular Biology, Center of Natural and Exact Sciences, Federal University of Santa Maria, Santa Maria, RS, 97105-900, Brazil
| | - Vanessa Angonesi Zborowski
- Laboratory of Synthesis, Reactivity, Pharmacological and Toxicological Evaluation of Organochalcogen Compounds, Department of Biochemistry and Molecular Biology, Center of Natural and Exact Sciences, Federal University of Santa Maria, Santa Maria, RS, 97105-900, Brazil
| | - Simone Pinton
- Federal University of Pampa, Campus Uruguaiana, RS, 97500-701, Brazil
| | - Cristina Wayne Nogueira
- Laboratory of Synthesis, Reactivity, Pharmacological and Toxicological Evaluation of Organochalcogen Compounds, Department of Biochemistry and Molecular Biology, Center of Natural and Exact Sciences, Federal University of Santa Maria, Santa Maria, RS, 97105-900, Brazil.
| |
Collapse
|
29
|
Montenegro YHA, de Queiroga Nascimento D, de Assis TO, Santos-Lopes SSD. The epigenetics of the hypothalamic-pituitary-adrenal axis in fetal development. Ann Hum Genet 2019; 83:195-213. [PMID: 30843189 DOI: 10.1111/ahg.12306] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Revised: 02/13/2019] [Accepted: 02/14/2019] [Indexed: 01/16/2023]
Abstract
The hypothalamic-pituitary-adrenal (HPA) axis is an important hormonal mechanism of the human body and is extremely programmable during embryonic and fetal development. Analyzing its development in this period is the key to understanding in fact how vulnerabilities of congenital diseases occur and any other changes in the phenotypic and histophysiological aspects of the fetus. The environment in which the mother is exposed during the gestational period can influence this axis. Knowing this, our objective was to analyze in recent research the possible impact of epigenetic programming on the HPA axis and its consequences for fetal development. This review brought together articles from two databases: ScienceDirect and PUBMED researched based on key words such as "epigenetics, HPA axis, cardiovascular disease, and circulatory problems" where it demonstrated full relevance in experimental and scientific settings. A total of 101 articles were selected following the criteria established by the researchers. Thus, it was possible to verify that the development of the HPA axis is directly related to changes that occur in the cardiovascular system, to the cerebral growth and other systems depending on the influence that it receives in the period of fetal formation.
Collapse
|
30
|
REM sleep's unique associations with corticosterone regulation, apoptotic pathways, and behavior in chronic stress in mice. Proc Natl Acad Sci U S A 2019; 116:2733-2742. [PMID: 30683720 PMCID: PMC6377491 DOI: 10.1073/pnas.1816456116] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Sleep disturbances are common in stress-related disorders but the nature of these sleep disturbances and how they relate to changes in the stress hormone corticosterone and changes in gene expression remained unknown. Here we demonstrate that in response to chronic mild stress, rapid–eye-movement sleep (REMS), a sleep state involved in emotion regulation and fear conditioning, changed first and more so than any other measured sleep characteristic. Transcriptomic profiles related to REMS continuity and theta oscillations overlapped with those for corticosterone, as well as with predictors for anhedonia, and were enriched for apoptotic pathways. These data highlight the central role of REMS in response to stress and warrant further investigation into REMS’s involvement in stress-related mental health disorders. One of sleep’s putative functions is mediation of adaptation to waking experiences. Chronic stress is a common waking experience; however, which specific aspect of sleep is most responsive, and how sleep changes relate to behavioral disturbances and molecular correlates remain unknown. We quantified sleep, physical, endocrine, and behavioral variables, as well as the brain and blood transcriptome in mice exposed to 9 weeks of unpredictable chronic mild stress (UCMS). Comparing 46 phenotypic variables revealed that rapid–eye-movement sleep (REMS), corticosterone regulation, and coat state were most responsive to UCMS. REMS theta oscillations were enhanced, whereas delta oscillations in non-REMS were unaffected. Transcripts affected by UCMS in the prefrontal cortex, hippocampus, hypothalamus, and blood were associated with inflammatory and immune responses. A machine-learning approach controlling for unspecific UCMS effects identified transcriptomic predictor sets for REMS parameters that were enriched in 193 pathways, including some involved in stem cells, immune response, and apoptosis and survival. Only three pathways were enriched in predictor sets for non-REMS. Transcriptomic predictor sets for variation in REMS continuity and theta activity shared many pathways with corticosterone regulation, in particular pathways implicated in apoptosis and survival, including mitochondrial apoptotic machinery. Predictor sets for REMS and anhedonia shared pathways involved in oxidative stress, cell proliferation, and apoptosis. These data identify REMS as a core and early element of the response to chronic stress, and identify apoptosis and survival pathways as a putative mechanism by which REMS may mediate the response to stressful waking experiences.
Collapse
|
31
|
Stress-inducible-stem cells: a new view on endocrine, metabolic and mental disease? Mol Psychiatry 2019; 24:2-9. [PMID: 30242231 PMCID: PMC6755998 DOI: 10.1038/s41380-018-0244-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Accepted: 07/25/2018] [Indexed: 02/08/2023]
|
32
|
Functional Neurochemistry of the Ventral and Dorsal Hippocampus: Stress, Depression, Dementia and Remote Hippocampal Damage. Neurochem Res 2018; 44:1306-1322. [PMID: 30357653 DOI: 10.1007/s11064-018-2662-0] [Citation(s) in RCA: 83] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Revised: 09/15/2018] [Accepted: 10/15/2018] [Indexed: 12/15/2022]
Abstract
The hippocampus is not a homogeneous brain area, and the complex organization of this structure underlies its relevance and functional pleiotropism. The new data related to the involvement of the ventral hippocampus in the cognitive function, behavior, stress response and its association with brain pathology, in particular, depression, are analyzed with a focus on neuroplasticity, specializations of the intrinsic neuronal network, corticosteroid signaling through mineralocorticoid and glucocorticoid receptors and neuroinflammation in the hippocampus. The data on the septo-temporal hippicampal gradient are analyzed with particular emphasis on the ventral hippocampus, a region where most important alteration underlying depressive disorders occur. According to the recent data, the existing simple paradigm "learning (dorsal hippocampus) versus emotions (ventral hippocampus)" should be substantially revised and specified. A new hypothesis is suggested on the principal involvement of stress response mechanisms (including interaction of released glucocorticoids with hippocampal receptors and subsequent inflammatory events) in the remote hippocampal damage underlying delayed dementia and depression induced by focal brain damage (e.g. post-stroke and post-traumatic). The translational validity of this hypothesis comprising new approaches in preventing post-stroke and post-trauma depression and dementia can be confirmed in experimental and clinical studies.
Collapse
|
33
|
Bedos M, Portillo W, Paredes RG. Neurogenesis and sexual behavior. Front Neuroendocrinol 2018; 51:68-79. [PMID: 29438737 DOI: 10.1016/j.yfrne.2018.02.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Revised: 02/06/2018] [Accepted: 02/07/2018] [Indexed: 12/16/2022]
Abstract
Different conditions induce proliferation, migration and integration of new neurons in the adult brain. This process of neurogenesis is a clear example of long lasting plastic changes in the brain of different species. Sexual behavior is a motivated behavior that is crucial for the survival of the species, but an individual can spend all his life without displaying sexual behavior. In the present review, we briefly describe some of the effects of pheromones on neurogenesis. We review in detail studies describing the effects of sexual behavior in both males and females on proliferation, migration and integration of new cells and neurons. It will become evident that most of the studies have been done in rodents, assessing the effects of this behavior on neurogenesis within the dentate gyrus of the hippocampus and in the subventricular zone - rostral migratory stream - olfactory bulb system.
Collapse
Affiliation(s)
- M Bedos
- CONACYT - Instituto de Neurobiología - Universidad Nacional Autónoma de México, Blvd Juriquilla 3001, Campus UNAM-Juriquilla, 76230 Querétaro, QRO, México
| | - W Portillo
- Instituto de Neurobiología - Universidad Nacional Autónoma de México, Blvd Juriquilla 3001, Campus UNAM-Juriquilla, 76230 Querétaro, QRO, México
| | - R G Paredes
- Instituto de Neurobiología - Universidad Nacional Autónoma de México, Blvd Juriquilla 3001, Campus UNAM-Juriquilla, 76230 Querétaro, QRO, México.
| |
Collapse
|
34
|
Salazar ER, Richter HG, Spichiger C, Mendez N, Halabi D, Vergara K, Alonso IP, Corvalán FA, Azpeleta C, Seron-Ferre M, Torres-Farfan C. Gestational chronodisruption leads to persistent changes in the rat fetal and adult adrenal clock and function. J Physiol 2018; 596:5839-5857. [PMID: 30118176 DOI: 10.1113/jp276083] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Accepted: 08/06/2018] [Indexed: 12/14/2022] Open
Abstract
KEY POINTS Light at night is essential to a 24/7 society, but it has negative consequences on health. Basically, light at night induces an alteration of our biological clocks, known as chronodisruption, with effects even when this occurs during pregnancy. Here we explored the developmental impact of gestational chronodisruption (chronic photoperiod shift, CPS) on adult and fetal adrenal biorhythms and function. We found that gestational chronodisruption altered fetal and adult adrenal function, at the molecular, morphological and physiological levels. The differences between control and CPS offspring suggest desynchronization of the adrenal circadian clock and steroidogenic pathway, leading to abnormal stress responses and metabolic adaptation, potentially increasing the risk of developing chronic diseases. ABSTRACT Light at night is essential to a 24/7 society, but it has negative consequences on health. Basically, light at night induces an alteration of our biological clocks, known as chronodisruption, with effects even when this occurs during pregnancy. Indeed, an abnormal photoperiod during gestation alters fetal development, inducing long-term effects on the offspring. Accordingly, we carried out a longitudinal study in rats, exploring the impact of gestational chronodisruption on the adrenal biorhythms and function of the offspring. Adult rats (90 days old) gestated under chronic photoperiod shift (CPS) decrease the time spent in the open arm zone of an elevated plus maze to 62% and increase the rearing time to 170%. CPS adults maintained individual daily changes in corticosterone, but their acrophases were distributed from 12.00 h to 06.00 h. CPS offspring maintained clock gene expression and oscillation, nevertheless no daily rhythm was observed in genes involved in the regulation and synthesis of steroids. Consistent with adult adrenal gland being programmed during fetal life, blunted daily rhythms of corticosterone, core clock gene machinery, and steroidogenic genes were observed in CPS fetal adrenal glands. Comparisons of the global transcriptome of CPS versus control fetal adrenal gland revealed that 1078 genes were differentially expressed (641 down-regulated and 437 up-regulated). In silico analysis revealed significant changes in Lipid Metabolism, Small Molecule Biochemistry, Cellular Development and the Inflammatory Response pathway (z score: 48-20). Altogether, the present results demonstrate that gestational chronodisruption changed fetal and adult adrenal function. This could translate to long-term abnormal stress responses and metabolic adaptation, increasing the risk of developing chronic diseases.
Collapse
Affiliation(s)
- E R Salazar
- Laboratorio de Cronobiología del Desarrollo, Instituto de Anatomía, Histología y Patología, Facultad de Medicina, Universidad Austral de Chile, Valdivia, Chile
| | - H G Richter
- Laboratorio de Cronobiología del Desarrollo, Instituto de Anatomía, Histología y Patología, Facultad de Medicina, Universidad Austral de Chile, Valdivia, Chile
| | - C Spichiger
- Laboratorio de Cronobiología del Desarrollo, Instituto de Anatomía, Histología y Patología, Facultad de Medicina, Universidad Austral de Chile, Valdivia, Chile
| | - N Mendez
- Laboratorio de Cronobiología del Desarrollo, Instituto de Anatomía, Histología y Patología, Facultad de Medicina, Universidad Austral de Chile, Valdivia, Chile
| | - D Halabi
- Laboratorio de Cronobiología del Desarrollo, Instituto de Anatomía, Histología y Patología, Facultad de Medicina, Universidad Austral de Chile, Valdivia, Chile
| | - K Vergara
- Laboratorio de Cronobiología del Desarrollo, Instituto de Anatomía, Histología y Patología, Facultad de Medicina, Universidad Austral de Chile, Valdivia, Chile
| | - I P Alonso
- Laboratorio de Cronobiología del Desarrollo, Instituto de Anatomía, Histología y Patología, Facultad de Medicina, Universidad Austral de Chile, Valdivia, Chile
| | - F A Corvalán
- Laboratorio de Cronobiología del Desarrollo, Instituto de Anatomía, Histología y Patología, Facultad de Medicina, Universidad Austral de Chile, Valdivia, Chile
| | - C Azpeleta
- Department of Basic Biomedical Sciences, Faculty of Biomedical Sciences and Health, European University of Madrid, Villaviciosa de Odón, Spain
| | - M Seron-Ferre
- Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - C Torres-Farfan
- Laboratorio de Cronobiología del Desarrollo, Instituto de Anatomía, Histología y Patología, Facultad de Medicina, Universidad Austral de Chile, Valdivia, Chile.,Centro Interdisciplinario de Estudios del Sistema Nervioso (CISNe), Universidad Austral de Chile, Valdivia, Chile
| |
Collapse
|
35
|
Lou YX, Li J, Wang ZZ, Xia CY, Chen NH. Glucocorticoid receptor activation induces decrease of hippocampal astrocyte number in rats. Psychopharmacology (Berl) 2018; 235:2529-2540. [PMID: 30069586 DOI: 10.1007/s00213-018-4936-2] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Accepted: 05/30/2018] [Indexed: 11/24/2022]
Abstract
RATIONALE The decrease of astrocyte number and hypothalamic-pituitary-adrenal (HPA) axis overactivity are observed in individuals with major depressive disorder. Elevated levels of glucocorticoids induced by hyperactivation of the HPA axis may result in glucocorticoid receptor (GR) activation. However, it is unclear whether there is a direct link between GR activation and the decrease of astrocyte number. METHODS Animals were exposed to chronic unpredictable stress (CUS) for 28 days and treated with continuous subcutaneous injections of vehicle or corticosterone (CORT; 40 mg/kg/day) for 21 days. We then administered mifepristone on day 21 after CUS and on day 18 after the CORT treatment. We observed behavioral deficits in the sucrose preference test, open field test, and forced swim test. Protein expression was analyzed using immunofluorescence (IF) and western blot (WB). RESULTS Animals exposed to CUS exhibited behavioral deficits in tests measuring anhedonia, anxiety, and despair state. They also had decreases in glial fibrillary acidic protein (GFAP) expression and numbers of GFAP-positive cells in the hippocampus. The behavioral and cellular alterations induced by CUS were reversed by subchronic treatment with the GR antagonist mifepristone. We also found that the subcutaneous injection of glucocorticoids may induce depression-like behavior and reduce GFAP protein expression in rats, which was similarly reversed by mifepristone. CONCLUSIONS These findings provide experimental evidence that GR activation due to elevated CORT levels induces the decrease of hippocampal astrocyte number in rats.
Collapse
Affiliation(s)
- Yu-Xia Lou
- Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China
| | - Jing Li
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Zhen-Zhen Wang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Cong-Yuan Xia
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Nai-Hong Chen
- Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China. .,State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China.
| |
Collapse
|
36
|
van Olst L, Bielefeld P, Fitzsimons CP, de Vries HE, Schouten M. Glucocorticoid-mediated modulation of morphological changes associated with aging in microglia. Aging Cell 2018; 17:e12790. [PMID: 29882317 PMCID: PMC6052476 DOI: 10.1111/acel.12790] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/04/2018] [Indexed: 01/12/2023] Open
Abstract
Microglia dynamically adapt their morphology and function during increasing age. However, the mechanisms behind these changes are to date poorly understood. Glucocorticoids (GCs) are long known and utilized for their immunomodulatory actions and endogenous GC levels are described to alter with advancing age. We here tested the hypothesis that age‐associated elevations in GC levels implicate microglia function and morphology. Our data indicate a decrease in microglial complexity and a concomitant increase in GC levels during aging. Interestingly, enhancing GC levels in young mice enhanced microglial ramifications, while the knockdown of the glucocorticoid receptor expression in old mice aggravated age‐associated microglial amoebification. These data suggest that GCs increase ramification of hippocampal microglia and may modulate age‐associated changes in microglial morphology.
Collapse
Affiliation(s)
- Lynn van Olst
- Department of Molecular Cell Biology and Immunology; VU University Medical Center, Amsterdam Neuroscience; Amsterdam The Netherlands
| | - Pascal Bielefeld
- Neuroscience Program; Swammerdam Institute for Life Sciences; University of Amsterdam; Amsterdam The Netherlands
| | - Carlos P. Fitzsimons
- Neuroscience Program; Swammerdam Institute for Life Sciences; University of Amsterdam; Amsterdam The Netherlands
| | - Helga E. de Vries
- Department of Molecular Cell Biology and Immunology; VU University Medical Center, Amsterdam Neuroscience; Amsterdam The Netherlands
| | - Marijn Schouten
- Department of Molecular Cell Biology and Immunology; VU University Medical Center, Amsterdam Neuroscience; Amsterdam The Netherlands
| |
Collapse
|
37
|
Draijer S, Chaves I, Hoekman MFM. The circadian clock in adult neural stem cell maintenance. Prog Neurobiol 2018; 173:41-53. [PMID: 29886147 DOI: 10.1016/j.pneurobio.2018.05.007] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Revised: 05/14/2018] [Accepted: 05/29/2018] [Indexed: 12/25/2022]
Abstract
Neural stem cells persist in the adult central nervous system as a continuing source of astrocytes, oligodendrocytes and neurons. Various signalling pathways and transcription factors actively maintain this population by regulating cell cycle entry and exit. Similarly, the circadian clock is interconnected with the cell cycle and actively maintains stem cell populations in various tissues. Here, we discuss emerging evidence for an important role of the circadian clock in neural stem cell maintenance. We propose that the NAD+-dependent deacetylase SIRT1 exerts control over the circadian clock in adult neural stem cell function to limit exhaustion of their population. Conversely, disruption of the circadian clock may compromise neural stem cell quiescence resulting in a premature decline of the neural stem cell population. As such, energy metabolism and the circadian clock converge in adult neural stem cell maintenance.
Collapse
Affiliation(s)
- Swip Draijer
- Swammerdam Institute for Life Sciences, University of Amsterdam, 1098 XH Amsterdam, The Netherlands
| | - Inês Chaves
- Department of Molecular Genetics, Erasmus University Medical Center, 3000 CA Rotterdam, The Netherlands
| | - Marco F M Hoekman
- Swammerdam Institute for Life Sciences, University of Amsterdam, 1098 XH Amsterdam, The Netherlands.
| |
Collapse
|
38
|
Belle MDC, Diekman CO. Neuronal oscillations on an ultra-slow timescale: daily rhythms in electrical activity and gene expression in the mammalian master circadian clockwork. Eur J Neurosci 2018; 48:2696-2717. [PMID: 29396876 DOI: 10.1111/ejn.13856] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Revised: 01/16/2018] [Accepted: 01/28/2018] [Indexed: 12/17/2022]
Abstract
Neuronal oscillations of the brain, such as those observed in the cortices and hippocampi of behaving animals and humans, span across wide frequency bands, from slow delta waves (0.1 Hz) to ultra-fast ripples (600 Hz). Here, we focus on ultra-slow neuronal oscillators in the hypothalamic suprachiasmatic nuclei (SCN), the master daily clock that operates on interlocking transcription-translation feedback loops to produce circadian rhythms in clock gene expression with a period of near 24 h (< 0.001 Hz). This intracellular molecular clock interacts with the cell's membrane through poorly understood mechanisms to drive the daily pattern in the electrical excitability of SCN neurons, exhibiting an up-state during the day and a down-state at night. In turn, the membrane activity feeds back to regulate the oscillatory activity of clock gene programs. In this review, we emphasise the circadian processes that drive daily electrical oscillations in SCN neurons, and highlight how mathematical modelling contributes to our increasing understanding of circadian rhythm generation, synchronisation and communication within this hypothalamic region and across other brain circuits.
Collapse
Affiliation(s)
- Mino D C Belle
- Institute of Clinical and Biomedical Sciences, University of Exeter Medical School, University of Exeter, Exeter, EX4 4PS, UK
| | - Casey O Diekman
- Department of Mathematical Sciences, New Jersey Institute of Technology, Newark, NJ, USA.,Institute for Brain and Neuroscience Research, New Jersey Institute of Technology, Newark, NJ, USA
| |
Collapse
|
39
|
Numakawa T, Odaka H, Adachi N. Actions of Brain-Derived Neurotrophic Factor and Glucocorticoid Stress in Neurogenesis. Int J Mol Sci 2017; 18:ijms18112312. [PMID: 29099059 PMCID: PMC5713281 DOI: 10.3390/ijms18112312] [Citation(s) in RCA: 97] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Revised: 10/30/2017] [Accepted: 10/31/2017] [Indexed: 12/12/2022] Open
Abstract
Altered neurogenesis is suggested to be involved in the onset of brain diseases, including mental disorders and neurodegenerative diseases. Neurotrophic factors are well known for their positive effects on the proliferation/differentiation of both embryonic and adult neural stem/progenitor cells (NSCs/NPCs). Especially, brain-derived neurotrophic factor (BDNF) has been extensively investigated because of its roles in the differentiation/maturation of NSCs/NPCs. On the other hand, recent evidence indicates a negative impact of the stress hormone glucocorticoids (GCs) on the cell fate of NSCs/NPCs, which is also related to the pathophysiology of brain diseases, such as depression and autism spectrum disorder. Furthermore, studies including ours have demonstrated functional interactions between neurotrophic factors and GCs in neural events, including neurogenesis. In this review, we show and discuss relationships among the behaviors of NSCs/NPCs, BDNF, and GCs.
Collapse
Affiliation(s)
- Tadahiro Numakawa
- Department of Cell Modulation, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto 860-8555, Japan.
- Department of Mental Disorder Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry (NCNP), Tokyo 187-8551, Japan.
| | - Haruki Odaka
- Department of Cell Modulation, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto 860-8555, Japan.
- Department of Life Science and Medical Bioscience, School of Advanced Science and Engineering, Waseda University, Tokyo 169-8050, Japan.
| | - Naoki Adachi
- Department of Biomedical Chemistry, School of Science and Technology, Kwansei Gakuin University, Sanda City, Hyogo 662-8501, Japan.
| |
Collapse
|
40
|
A Protocol for Generation of a Corticosterone Model of Psychiatric Disorders. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017. [PMID: 28353245 DOI: 10.1007/978-3-319-52479-5_24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register]
Abstract
Some patients with psychiatric disorders show hyperactivity of the hypothalamic-pituitary-adrenal (HPA) axis. This may be due to an impaired feedback inhibition and can be seen through increased levels of circulating cortisol. Here a protocol is described to mimic this situation by subcutaneous implantation of corticosterone pellets in mice. We also present characterization of the model by looking at effects on neuronal proliferation in hippocampus, one of the main tissues known to be affected by HPA axis hyper-activation. Such tissues could be used in analyses by proteomic platforms.
Collapse
|
41
|
Khanzada NS, Butler MG, Manzardo AM. GeneAnalytics Pathway Analysis and Genetic Overlap among Autism Spectrum Disorder, Bipolar Disorder and Schizophrenia. Int J Mol Sci 2017; 18:ijms18030527. [PMID: 28264500 PMCID: PMC5372543 DOI: 10.3390/ijms18030527] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Revised: 02/15/2017] [Accepted: 02/23/2017] [Indexed: 12/18/2022] Open
Abstract
Bipolar disorder (BPD) and schizophrenia (SCH) show similar neuropsychiatric behavioral disturbances, including impaired social interaction and communication, seen in autism spectrum disorder (ASD) with multiple overlapping genetic and environmental influences implicated in risk and course of illness. GeneAnalytics software was used for pathway analysis and genetic profiling to characterize common susceptibility genes obtained from published lists for ASD (792 genes), BPD (290 genes) and SCH (560 genes). Rank scores were derived from the number and nature of overlapping genes, gene-disease association, tissue specificity and gene functions subdivided into categories (e.g., diseases, tissues or functional pathways). Twenty-three genes were common to all three disorders and mapped to nine biological Superpathways including Circadian entrainment (10 genes, score = 37.0), Amphetamine addiction (five genes, score = 24.2), and Sudden infant death syndrome (six genes, score = 24.1). Brain tissues included the medulla oblongata (11 genes, score = 2.1), thalamus (10 genes, score = 2.0) and hypothalamus (nine genes, score = 2.0) with six common genes (BDNF, DRD2, CHRNA7, HTR2A, SLC6A3, and TPH2). Overlapping genes impacted dopamine and serotonin homeostasis and signal transduction pathways, impacting mood, behavior and physical activity level. Converging effects on pathways governing circadian rhythms support a core etiological relationship between neuropsychiatric illnesses and sleep disruption with hypoxia and central brain stem dysfunction.
Collapse
Affiliation(s)
- Naveen S Khanzada
- Department of Psychiatry and Behavioral Sciences, University of Kansas Medical Center, Kansas City, KS 66160, USA.
| | - Merlin G Butler
- Department of Psychiatry and Behavioral Sciences, University of Kansas Medical Center, Kansas City, KS 66160, USA.
- Department of Pediatrics, University of Kansas Medical Center, Kansas City, KS 66160, USA.
| | - Ann M Manzardo
- Department of Psychiatry and Behavioral Sciences, University of Kansas Medical Center, Kansas City, KS 66160, USA.
| |
Collapse
|
42
|
Bielefeld P, Schouten M, Lucassen PJ, Fitzsimons CP. Transcription factor oscillations in neural stem cells: Implications for accurate control of gene expression. NEUROGENESIS 2017; 4:e1262934. [PMID: 28321433 PMCID: PMC5345753 DOI: 10.1080/23262133.2016.1262934] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/05/2016] [Revised: 11/14/2016] [Accepted: 11/15/2016] [Indexed: 01/14/2023]
Abstract
Naturally occurring oscillations in glucocorticoids induce a cyclic activation of the glucocorticoid receptor (GR), a well-characterized ligand-activated transcription factor. These cycles of GR activation/deactivation result in rapid GR exchange at genomic response elements and GR recycling through the chaperone machinery, ultimately generating pulses of GR-mediated transcriptional activity of target genes. In a recent article we have discussed the implications of circadian and high-frequency (ultradian) glucocorticoid oscillations for the dynamic control of gene expression in hippocampal neural stem/progenitor cells (NSPCs) (Fitzsimons et al., Front. Neuroendocrinol., 2016). Interestingly, this oscillatory transcriptional activity is common to other transcription factors, many of which regulate key biological functions in NSPCs, such as NF-kB, p53, Wnt and Notch. Here, we discuss the oscillatory behavior of these transcription factors, their role in a biologically accurate target regulation and the potential importance for a dynamic control of transcription activity and gene expression in NSPCs.
Collapse
Affiliation(s)
- Pascal Bielefeld
- Neuroscience Program, Swammerdam Institute for Life Sciences, Faculty of Sciences, University of Amsterdam
| | - Marijn Schouten
- Neuroscience Program, Swammerdam Institute for Life Sciences, Faculty of Sciences, University of Amsterdam
| | - Paul J Lucassen
- Neuroscience Program, Swammerdam Institute for Life Sciences, Faculty of Sciences, University of Amsterdam
| | - Carlos P Fitzsimons
- Neuroscience Program, Swammerdam Institute for Life Sciences, Faculty of Sciences, University of Amsterdam
| |
Collapse
|
43
|
Mufdi M, Núñez L, Ochoa JP, Mejía G. Relationship between overall child development and caries severity in Chilean three-year-old preschool children. JOURNAL OF ORAL RESEARCH 2016. [DOI: 10.17126/joralres.2016.058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|
44
|
Biedermann SV, Auer MK, Bindila L, Ende G, Lutz B, Weber-Fahr W, Gass P, Fuss J. Restricted vs. unrestricted wheel running in mice: Effects on brain, behavior and endocannabinoids. Horm Behav 2016; 86:45-54. [PMID: 27664019 DOI: 10.1016/j.yhbeh.2016.09.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2016] [Revised: 09/16/2016] [Accepted: 09/18/2016] [Indexed: 11/26/2022]
Abstract
Beneficial effects of voluntary wheel running on hippocampal neurogenesis, morphology and hippocampal-dependent behavior have widely been studied in rodents, but also serious side effects and similarities to stereotypy have been reported. Some mouse strains run excessively when equipped with running wheels, complicating the comparability to human exercise regimes. Here, we investigated how exercise restriction to 6h/day affects hippocampal morphology and metabolism, stereotypic and basal behaviors, as well as the endocannabinoid system in wheel running C57BL/6 mice; the strain most commonly used for behavioral analyses and psychiatric disease models. Restricted and unrestricted wheel running had similar effects on immature hippocampal neuron numbers, thermoregulatory nest building and basal home-cage behaviors. Surprisingly, hippocampal gray matter volume, assessed with magnetic resonance (MR) imaging at 9.4 Tesla, was only increased in unrestricted but not in restricted runners. Moreover, unrestricted runners showed less stereotypic behavior than restricted runners did. However, after blockage of running wheels for 24h stereotypic behavior also increased in unrestricted runners, arguing against a long-term effect of wheel running on stereotypic behavior. Stereotypic behaviors correlated with frontal glutamate and glucose levels assessed by 1H-MR spectroscopy. While acute running increased plasma levels of the endocannabinoid anandamide in former studies in mice and humans, we found an inverse correlation of anandamide with the daily running distance after long-term running. In conclusion, although there are some diverging effects of restricted and unrestricted running on brain and behavior, restricted running does not per se seem to be a better animal model for aerobic exercise in mice.
Collapse
Affiliation(s)
- Sarah V Biedermann
- Department of Neuroimaging, Central Institute of Mental Health, University Medicine Mannheim, University of Heidelberg, 68159 Mannheim, Germany; Department of Psychiatry and Psychotherapy, Center of Psychosocial Medicine, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Matthias K Auer
- Department of Clinical Neuroendocrinology, Max Planck Institute of Psychiatry, 80804 Munich, Germany
| | - Laura Bindila
- Institute of Physiological Chemistry, University Medical Center of the Johannes Gutenberg University Mainz, 55099 Mainz, Germany
| | - Gabriele Ende
- Department of Neuroimaging, Central Institute of Mental Health, University Medicine Mannheim, University of Heidelberg, 68159 Mannheim, Germany
| | - Beat Lutz
- Institute of Physiological Chemistry, University Medical Center of the Johannes Gutenberg University Mainz, 55099 Mainz, Germany
| | - Wolfgang Weber-Fahr
- Department of Neuroimaging, Central Institute of Mental Health, University Medicine Mannheim, University of Heidelberg, 68159 Mannheim, Germany; Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, University Medicine Mannheim, University of Heidelberg, 68159 Mannheim, Germany
| | - Peter Gass
- Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, University Medicine Mannheim, University of Heidelberg, 68159 Mannheim, Germany
| | - Johannes Fuss
- Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, University Medicine Mannheim, University of Heidelberg, 68159 Mannheim, Germany; Institute for Sex Research and Forensic Psychiatry, Center of Psychosocial Medicine, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany.
| |
Collapse
|
45
|
Galea LAM, Duarte-Guterman P. Hormones and the regulation of adult neurogenesis in the hippocampus and beyond: Where are we now? Introduction to the special issue on hormonal regulation of adult neurogenesis: Implications for disease. Front Neuroendocrinol 2016; 41:1-2. [PMID: 27343170 DOI: 10.1016/j.yfrne.2016.06.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Accepted: 06/20/2016] [Indexed: 12/18/2022]
Affiliation(s)
- Liisa A M Galea
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada; Department of Psychology, University of British Columbia, Vancouver, BC, Canada.
| | - Paula Duarte-Guterman
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada; Department of Psychology, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|