1
|
Dvorzhak A, Brecht M, Schmitz D. Social play behavior is driven by glycine-dependent mechanisms. Curr Biol 2024; 34:3654-3664.e6. [PMID: 39053464 DOI: 10.1016/j.cub.2024.06.073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 06/19/2024] [Accepted: 06/27/2024] [Indexed: 07/27/2024]
Abstract
Social play is pervasive in juvenile mammals, yet it is poorly understood in terms of its underlying brain mechanisms. Specifically, we do not know why young animals are most playful and why most adults cease to social play. Here, we analyze the synaptic mechanisms underlying social play. We found that blocking the rat periaqueductal gray (PAG) interfered with social play. Furthermore, an age-related decrease of neural firing in the PAG is associated with a decrease in synaptic release of glycine. Most importantly, modulation of glycine concentration-apparently acting on the glycinergic binding site of the N-methyl-D-aspartate (NMDA) receptor-not only strongly modulates social play but can also reverse the age-related decline in social play. In conclusion, we demonstrate that social play critically depends on the neurotransmitter glycine within the PAG.
Collapse
Affiliation(s)
- Anton Dvorzhak
- Charité-Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Neuroscience Research Center, 10117 Berlin, Germany
| | - Michael Brecht
- Humboldt-Universität zu Berlin, Bernstein Center for Computational Neuroscience, Philippstr. 13, 10115 Berlin, Germany; Charité-Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, NeuroCure Cluster of Excellence, 10117 Berlin, Germany
| | - Dietmar Schmitz
- Charité-Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Neuroscience Research Center, 10117 Berlin, Germany; Humboldt-Universität zu Berlin, Bernstein Center for Computational Neuroscience, Philippstr. 13, 10115 Berlin, Germany; Charité-Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, NeuroCure Cluster of Excellence, 10117 Berlin, Germany; German Center for Neurodegenerative Diseases (DZNE), 10117 Berlin, Germany; Charité-Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Einstein Center for Neuroscience, 10117 Berlin, Germany; Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Robert-Rössle-Straße 10, 13125 Berlin, Germany.
| |
Collapse
|
2
|
Alzu'bi A, Abu-El-Rub E, Almahasneh F, Tahat L, Athamneh RY, Khasawneh R, Alzoubi H, Ghorab DS, Almazari R, Zoubi MSA, Al-Zoubi RM. Delineating the molecular mechanisms of hippocampal neurotoxicity induced by chronic administration of synthetic cannabinoid AB-FUBINACA in mice. Neurotoxicology 2024; 103:50-59. [PMID: 38823587 DOI: 10.1016/j.neuro.2024.05.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Revised: 05/28/2024] [Accepted: 05/28/2024] [Indexed: 06/03/2024]
Abstract
Chronic use of synthetic cannabinoids (SCs) has been associated with cognitive and behavioural deficits and an increased risk of neuropsychiatric disorders. The underlying molecular and cellular mechanisms of the neurotoxic effects of long-term use of SCs have not been well investigated in the literature. Herein, we evaluated the in vivo effects of chronic administration of AB-FUBINACA on the hippocampus in mice. Our results revealed that the administration of AB-FUBINACA induced a significant impairment in recognition memory associated with histopathological changes in the hippocampus. These findings were found to be correlated with increased level of oxidative stress, neuroinflammation, and apoptosis markers, and reduced expression of brain-derived neurotrophic factor (BDNF), which plays an essential role in modulating synaptic plasticity integral for promoting learning and memory in the hippocampus. Additionally, we showed that AB-FUBINACA significantly decreased the expression of NR1, an important functional subunit of glutamate/NMDA receptors and closely implicated in the development of toxic psychosis. These findings shed light on the long-term neurotoxic effects of SCs on hippocampus and the underlying mechanisms of these effects. This study provided new targets for possible medical interventions to improve the treatment guidelines for SCs addiction.
Collapse
Affiliation(s)
- Ayman Alzu'bi
- Department of Basic Medical Sciences, Faculty of Medicine, Yarmouk University, Irbid 211-63, Jordan.
| | - Ejlal Abu-El-Rub
- Department of Basic Medical Sciences, Faculty of Medicine, Yarmouk University, Irbid 211-63, Jordan
| | - Fatimah Almahasneh
- Department of Basic Medical Sciences, Faculty of Medicine, Yarmouk University, Irbid 211-63, Jordan
| | - Lena Tahat
- Department of Biological Sciences, Faculty of Science, Yarmouk University, Irbid 211-63, Jordan
| | - Rabaa Y Athamneh
- Department of Medical Laboratory Sciences, Faculty of Allied Science, Zarqa University, Zarqa 13110, Jordan
| | - Ramada Khasawneh
- Department of Basic Medical Sciences, Faculty of Medicine, Yarmouk University, Irbid 211-63, Jordan
| | - Hiba Alzoubi
- Department of Basic Medical Sciences, Faculty of Medicine, Yarmouk University, Irbid 211-63, Jordan
| | - Doaa S Ghorab
- Department of Basic Medical Sciences, Faculty of Medicine, Yarmouk University, Irbid 211-63, Jordan; Pathology Department, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Rawan Almazari
- Department of Basic Medical Sciences, Faculty of Medicine, Yarmouk University, Irbid 211-63, Jordan
| | - Mazhar Salim Al Zoubi
- Department of Basic Medical Sciences, Faculty of Medicine, Yarmouk University, Irbid 211-63, Jordan
| | - Raed M Al-Zoubi
- Surgical Research Section, Department of Surgery, Hamad Medical Corporation, Doha, Qatar; Department of Biomedical Sciences, QU-Health, College of Health Sciences, Qatar University, Doha 2713, Qatar; Department of Chemistry, Jordan University of Science and Technology, P.O.Box 3030, Irbid 22110, Jordan.
| |
Collapse
|
3
|
Aleksandrova S, Alexova R, Dragomanova S, Kalfin R, Nicoletti F, Fagone P, Petralia MC, Mangano K, Tancheva L. Preventive and Therapeutic Effects of Punica granatum L. Polyphenols in Neurological Conditions. Int J Mol Sci 2023; 24:ijms24031856. [PMID: 36768185 PMCID: PMC9916020 DOI: 10.3390/ijms24031856] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 01/05/2023] [Accepted: 01/11/2023] [Indexed: 01/19/2023] Open
Abstract
Pomegranate (Punica granatum L.) is a polyphenol-rich food and medicinal plant containing flavonols, anthocyanins, and tannins. Ellagitannins (ETs) are the most abundant polyphenols in pomegranate. A growing body of research shows that polyphenol-rich pomegranate extracts and their metabolites target multiple types of brain cell and support their redox balance, proliferation and survival, as well as cell signaling. Independent studies have demonstrated that the significant neuroprotective effects of ETs are mediated by their antioxidant and anti-inflammatory effects, their chelating properties, by their ability to activate various signaling pathways, as well as the ability to influence mitochondrial damage, thus regulating autophagy, apoptosis and neurotransmitter signaling. The multitude of in vitro and in vivo studies summarized in the present review suggest that pomegranate polyphenols act on both neuronal and glial cells directly, and also affect blood-brain barrier function, restoring redox balance in the blood and brain and increasing blood flow to the brain.
Collapse
Affiliation(s)
- Simona Aleksandrova
- Department of Biological Activity of Natural and Synthetic Substances, Institute of Neurobiology, Bulgarian Academy of Sciences, 1113 Sofia, Bulgaria
| | - Ralitza Alexova
- Department of Medical Chemistry and Biochemistry, Medical Faculty, Medical University—Sofia, 2 Zdrave St., 1431 Sofia, Bulgaria
| | - Stela Dragomanova
- Department of Pharmacology, Toxicology and Pharmacotherapy, Faculty of Pharmacy, Medical University, 9002 Varna, Bulgaria
| | - Reni Kalfin
- Department of Biological Activity of Natural and Synthetic Substances, Institute of Neurobiology, Bulgarian Academy of Sciences, 1113 Sofia, Bulgaria
- Department of Health Care, South-West University “Neofit Rilski”, Ivan Mihailov St. 66, 2700 Blagoevgrad, Bulgaria
| | - Ferdinando Nicoletti
- Department of Biomedical and Biotechnological Sciences, University of Catania, Via S. Sofia 89, 95123 Catania, Italy
- Correspondence:
| | - Paolo Fagone
- Department of Biomedical and Biotechnological Sciences, University of Catania, Via S. Sofia 89, 95123 Catania, Italy
| | - Maria Cristina Petralia
- Department of Clinical and Experimental Medicine, University of Messina, 98122 Messina, Italy
| | - Katia Mangano
- Department of Biomedical and Biotechnological Sciences, University of Catania, Via S. Sofia 89, 95123 Catania, Italy
| | - Lyubka Tancheva
- Department of Biological Activity of Natural and Synthetic Substances, Institute of Neurobiology, Bulgarian Academy of Sciences, 1113 Sofia, Bulgaria
| |
Collapse
|
4
|
Wong-Riley MTT. The critical period: neurochemical and synaptic mechanisms shared by the visual cortex and the brain stem respiratory system. Proc Biol Sci 2021; 288:20211025. [PMID: 34493083 DOI: 10.1098/rspb.2021.1025] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
The landmark studies of Wiesel and Hubel in the 1960's initiated a surge of investigations into the critical period of visual cortical development, when abnormal visual experience can alter cortical structures and functions. Most studies focused on the visual cortex, with relatively little attention to subcortical structures. The goal of the present review is to elucidate neurochemical and synaptic mechanisms common to the critical periods of the visual cortex and the brain stem respiratory system in the normal rat. In both regions, the critical period is a time of (i) heightened inhibition; (ii) reduced expression of brain-derived neurotrophic factor (BDNF); and (iii) synaptic imbalance, with heightened inhibition and suppressed excitation. The last two mechanisms are contrary to the conventional premise. Synaptic imbalance renders developing neurons more vulnerable to external stressors. However, the critical period is necessary to enable each system to strengthen its circuitry, adapt to its environment, and transition from immaturity to maturity, when a state of relative synaptic balance is attained. Failure to achieve such a balance leads to neurological disorders.
Collapse
Affiliation(s)
- Margaret T T Wong-Riley
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| |
Collapse
|
5
|
Lau CG, Zhang H, Murthy VN. Deletion of TrkB in parvalbumin interneurons alters cortical neural dynamics. J Cell Physiol 2021; 237:949-964. [PMID: 34491578 PMCID: PMC8810709 DOI: 10.1002/jcp.30571] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 08/20/2021] [Accepted: 08/23/2021] [Indexed: 11/25/2022]
Abstract
Signaling by neurotrophins such as the brain‐derived neurotrophic factor (BDNF) is known to modulate development of interneurons, but the circuit effects of this modulation remain unclear. Here, we examined the impact of deleting TrkB, a BDNF receptor, in parvalbumin‐expressing (PV) interneurons on the balance of excitation and inhibition (E‐I) in cortical circuits. In the mouse olfactory cortex, TrkB deletion impairs multiple aspects of PV neuronal function including synaptic excitation, intrinsic excitability, and the innervation pattern of principal neurons. Impaired PV cell function resulted in aberrant spiking patterns in principal neurons in response to stimulation of sensory inputs. Surprisingly, dampened PV neuronal function leads to a paradoxical decrease in overall excitability in cortical circuits. Our study demonstrates that, by modulating PV circuit plasticity and development, TrkB plays a critical role in shaping the evoked pattern of activity in a cortical network.
Collapse
Affiliation(s)
- Chunyue Geoffrey Lau
- Department of Neuroscience, City University of Hong Kong, Hong Kong, China.,Department of Molecular and Cellular Biology and Center for Brain Science, Harvard University, Cambridge, Massachusetts, USA
| | - Huiqi Zhang
- Department of Neuroscience, City University of Hong Kong, Hong Kong, China
| | - Venkatesh N Murthy
- Department of Molecular and Cellular Biology and Center for Brain Science, Harvard University, Cambridge, Massachusetts, USA
| |
Collapse
|
6
|
Robinson JL, Yanes JA, Reid MA, Murphy JE, Busler JN, Mumford PW, Young KC, Pietrzkowski ZJ, Nemzer BV, Hunter JM, Beck DT. Neurophysiological Effects of Whole Coffee Cherry Extract in Older Adults with Subjective Cognitive Impairment: A Randomized, Double-Blind, Placebo-Controlled, Cross-Over Pilot Study. Antioxidants (Basel) 2021; 10:144. [PMID: 33498314 PMCID: PMC7909261 DOI: 10.3390/antiox10020144] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 01/10/2021] [Accepted: 01/13/2021] [Indexed: 12/11/2022] Open
Abstract
Bioactive plant-based compounds have shown promise as protective agents across multiple domains including improvements in neurological and psychological measures. Methodological challenges have limited our understanding of the neurophysiological changes associated with polyphenol-rich supplements such as whole coffee cherry extract (WCCE). In the current study, we (1) compared 100 mg of WCCE to a placebo using an acute, randomized, double-blind, within-subject, cross-over design, and we (2) conducted a phytochemical analysis of WCCE. The primary objective of the study was to determine the neurophysiological and behavioral changes that resulted from the acute administration of WCCE. We hypothesized that WCCE would increase brain-derived neurotrophic factor (BDNF) and glutamate levels while also increasing neurofunctional measures in cognitive brain regions. Furthermore, we expected there to be increased behavioral performance associated with WCCE, as measured by reaction time and accuracy. Participants underwent four neuroimaging scans (pre- and post-WCCE and placebo) to assess neurofunctional/metabolic outcomes using functional magnetic resonance imaging and magnetic resonance spectroscopy. The results suggest that polyphenol-rich WCCE is associated with decreased reaction time and may protect against cognitive errors on tasks of working memory and response inhibition. Behavioral findings were concomitant with neurofunctional changes in structures involved in decision-making and attention. Specifically, we found increased functional connectivity between the anterior cingulate and regions involved in sensory and decision-making networks. Additionally, we observed increased BDNF and an increased glutamate/gamma-aminobutyric acid (GABA) ratio following WCCE administration. These results suggest that WCCE is associated with acute neurophysiological changes supportive of faster reaction times and increased, sustained attention.
Collapse
Affiliation(s)
- Jennifer L. Robinson
- Department of Psychology, Auburn University, Auburn, AL 36849, USA; (J.A.Y.); (J.E.M.); (J.N.B.)
- Auburn University MRI Research Center, Auburn University, Auburn, AL 36849, USA;
- Alabama Advanced Imaging Consortium, Auburn University, Auburn, AL 36849, USA
- Initiative for the Center for Neuroscience, Auburn University, Auburn, AL 36849, USA;
- Department of Electrical and Computer Engineering, Auburn University, Auburn, AL 36849, USA
| | - Julio A. Yanes
- Department of Psychology, Auburn University, Auburn, AL 36849, USA; (J.A.Y.); (J.E.M.); (J.N.B.)
- Auburn University MRI Research Center, Auburn University, Auburn, AL 36849, USA;
- Alabama Advanced Imaging Consortium, Auburn University, Auburn, AL 36849, USA
- Initiative for the Center for Neuroscience, Auburn University, Auburn, AL 36849, USA;
| | - Meredith A. Reid
- Auburn University MRI Research Center, Auburn University, Auburn, AL 36849, USA;
- Alabama Advanced Imaging Consortium, Auburn University, Auburn, AL 36849, USA
- Initiative for the Center for Neuroscience, Auburn University, Auburn, AL 36849, USA;
- Department of Electrical and Computer Engineering, Auburn University, Auburn, AL 36849, USA
| | - Jerry E. Murphy
- Department of Psychology, Auburn University, Auburn, AL 36849, USA; (J.A.Y.); (J.E.M.); (J.N.B.)
| | - Jessica N. Busler
- Department of Psychology, Auburn University, Auburn, AL 36849, USA; (J.A.Y.); (J.E.M.); (J.N.B.)
- Auburn University MRI Research Center, Auburn University, Auburn, AL 36849, USA;
- Alabama Advanced Imaging Consortium, Auburn University, Auburn, AL 36849, USA
- Initiative for the Center for Neuroscience, Auburn University, Auburn, AL 36849, USA;
| | - Petey W. Mumford
- School of Kinesiology, Auburn University, Auburn, AL 36849, USA; (P.W.M.); (K.C.Y.)
| | - Kaelin C. Young
- School of Kinesiology, Auburn University, Auburn, AL 36849, USA; (P.W.M.); (K.C.Y.)
- Edward Via College of Osteopathic Medicine, Auburn, AL 36830, USA
| | | | - Boris V. Nemzer
- VDF FutureCeuticals, Inc., 2692 N. State Route 1-17, Momence, IL 60954, USA; (B.V.N.); (J.M.H.)
| | - John M. Hunter
- VDF FutureCeuticals, Inc., 2692 N. State Route 1-17, Momence, IL 60954, USA; (B.V.N.); (J.M.H.)
| | - Darren T. Beck
- Initiative for the Center for Neuroscience, Auburn University, Auburn, AL 36849, USA;
- Edward Via College of Osteopathic Medicine, Auburn, AL 36830, USA
| |
Collapse
|
7
|
Minutillo A, Panza G, Mauri MC. Musical practice and BDNF plasma levels as a potential marker of synaptic plasticity: an instrument of rehabilitative processes. Neurol Sci 2020; 42:1861-1867. [PMID: 32940801 PMCID: PMC8043880 DOI: 10.1007/s10072-020-04715-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 09/08/2020] [Indexed: 01/06/2023]
Abstract
Background and objectives The aim of the study was to investigate the influence of musical practice on brain plasticity. BDNF (brain-derived neurotrophic factor) is a neurotrophin involved in neuroplasticity and synaptic function. Materials and methods We recruited 48 healthy subjects of equal age and sex (21 musicians and 27 non-musicians). All subjects were administered the AQ (Autism-Spectrum Questionnaire) and plasma levels (PLs) of BDNF, oxytocin (OT), and vasopressin (VP) were measured in the blood sample of every participant. Results. The difference between BDNF PLs in the two groups was found to be statistically significant (t = − 2.214, p = 0.03). Furthermore, oxytocin (OT) PLs and musical practice were found to be independent positive predictors of BDNF PLs (p < 0.04). We also found a negative correlation between BDNF PLs and AD (attention to detail) sub-scale score of AQ throughout the whole sample. Assuming BDNF PLs to be a marker of synaptic plasticity, higher PLs could be associated with the activation of alternative neural pathways: a lower score in the “attention to detail” sub-scale could imply greater flexibility of higher cerebral functions among musicians. Further researches should be conducted to assess the rehabilitative usefulness of these findings among patients affected by psychiatric disorders.
Collapse
Affiliation(s)
- Alessandro Minutillo
- Department of Neurosciences and Mental Health, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy. .,Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy.
| | - Gabriele Panza
- Department of Neurosciences and Mental Health, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Massimo Carlo Mauri
- Department of Neurosciences and Mental Health, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| |
Collapse
|
8
|
Balikci A, Ilbay G, Ates N. Neonatal Tactile Stimulations Affect Genetic Generalized Epilepsy and Comorbid Depression-Like Behaviors. Front Behav Neurosci 2020; 14:132. [PMID: 32792925 PMCID: PMC7390910 DOI: 10.3389/fnbeh.2020.00132] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Accepted: 07/02/2020] [Indexed: 12/18/2022] Open
Abstract
Recent studies suggest that development of absence epilepsy and comorbid depression might be prevented by increased maternal care of the offspring, in which tactile stimulation induced by licking/grooming and non-nutritive contact seem to be crucial. In this study, we aimed to evaluate the effect of neonatal tactile stimulations (NTS) on absence epilepsy and depression-like behaviors in adulthood. Wistar Albino Glaxo from Rijswijk (WAG/Rij) rat pups with a genetic predisposition to absence epilepsy were divided into tactile stimulation (TS) group, deep touch pressure (DTP) group, maternal separation (MS) group or control group. Between postnatal day 3 and 21, manipulations (TS, DTP, and MS) were carried out for 15 min and three times a day. Animals were submitted to locomotor activity, sucrose consumption test (SCT) and forced swimming test (FST) at five months of age. At the age of six months, the electroencephalogram (EEG) recordings were conducted in order to quantify the spike-wave discharges (SWDs), which is the hallmark of absence epilepsy. The TS and DTP groups showed less and shorter SWDs in later life in comparison to maternally separated and control rats. SWDs’ number and total duration were significantly reduced in TS and DTP groups whereas mean duration of SWDs was reduced only in DTP group (p < 0.05). TS and DTP also decreased depression-like behaviors measured by SCT and FST in adult animals. In the SCT, number of approaches was significantly higher in TS and DTP groups than the maternally separated and control rats. In the FST, while the immobility latency of TS and DTP groups was significantly higher, only TS group showed significantly decreased immobility and increased swimming time. The results showed that NTS decreases both the number and length of SWDs and the depression-like behaviors in WAG/Rij rats probably by increasing arousal level and causing alterations in the level of some neurotrophic factors as well as in functions of the neural plasticity in the developing rat’s brain.
Collapse
Affiliation(s)
- Aymen Balikci
- Department of Physiology, Faculty of Medicine, Kocaeli University, Kocaeli, Turkey
| | - Gul Ilbay
- Department of Physiology, Faculty of Medicine, Kocaeli University, Kocaeli, Turkey
| | - Nurbay Ates
- Department of Physiology, Faculty of Medicine, Kocaeli University, Kocaeli, Turkey
| |
Collapse
|
9
|
Kim OY, Song J. The importance of BDNF and RAGE in diabetes-induced dementia. Pharmacol Res 2020; 160:105083. [PMID: 32679182 DOI: 10.1016/j.phrs.2020.105083] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 06/30/2020] [Accepted: 07/12/2020] [Indexed: 01/11/2023]
Abstract
Diabetes-induced dementia is an emerging neurodisorder all over the world. The prevalence rates of dementia and diabetes have been gradually increasing worldwide. Diabetes has been known to lead to oxidative stress, inflammation aggravation, and hyperglycemia conditions in the brain. Various diabetic implications cause the lower secretion of brain-derived neurotrophic factor (BDNF) and the increase of receptor for advanced glycation end products (RAGE), ultimately leading to both cerebrovascular dysfunction and cognitive decline. Here, we summarized the significant evidences highlighting the specific mechanisms between BDNF and RAGE and cerebrovascular dysfunction and memory function and how these relate to diabetes-induced dementia. Especially, we review that the association between BDFN and RAGE in neuroinflammation, the reduction of long-term potentiation, and the vascular implications in brain.
Collapse
Affiliation(s)
- Oh Yoen Kim
- The Department of Food Science and Nutrition, Dong-A University, Busan 49315, Republic of Korea; The Center for Silver-Targeted Biomaterials, Brain Busan 21 Plus Program, Graduate School, Dong-A University, Busan 49315, Republic of Korea.
| | - Juhyun Song
- The Department of Anatomy, Chonnam National University, Chonnam National University Medical School, Hwasun 58128, Jeollanam-do, Republic of Korea.
| |
Collapse
|
10
|
Almeida FB, Nin MS, Barros HMT. The role of allopregnanolone in depressive-like behaviors: Focus on neurotrophic proteins. Neurobiol Stress 2020; 12:100218. [PMID: 32435667 PMCID: PMC7231971 DOI: 10.1016/j.ynstr.2020.100218] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 03/16/2020] [Accepted: 03/30/2020] [Indexed: 12/18/2022] Open
Abstract
Allopregnanolone (3α,5α-tetrahydroprogesterone; pharmaceutical formulation: brexanolone) is a neurosteroid that has recently been approved for the treatment of postpartum depression, promising to fill part of a long-lasting gap in the effectiveness of pharmacotherapies for depressive disorders. In this review, we explore the experimental research that characterized the antidepressant-like effects of allopregnanolone, with a particular focus on the neurotrophic adaptations induced by this neurosteroid in preclinical studies. We demonstrate that there is a consistent decrease in allopregnanolone levels in limbic brain areas in rodents submitted to stress-induced models of depression, such as social isolation and chronic unpredictable stress. Further, both the drug-induced upregulation of allopregnanolone or its direct administration reduce depressive-like behaviors in models such as the forced swim test. The main drugs of interest that upregulate allopregnanolone levels are selective serotonin reuptake inhibitors (SSRIs), which present the neurosteroidogenic property even in lower, non-SSRI doses. Finally, we explore how these antidepressant-like behaviors are related to neurogenesis, particularly in the hippocampus. The protagonist in this mechanism is likely the brain-derived neurotrophic factor (BFNF), which is decreased in animal models of depression and may be restored by the normalization of allopregnanolone levels. The role of an interaction between GABA and the neurotrophic mechanisms needs to be further investigated.
Collapse
Key Words
- 3α,5α-tetrahydroprogesterone
- BDNF
- BDNF, brain-derived neurotrophic factor
- Brexanolone
- CSF, cerebrospinal fluid
- CUS, chronic unpredictable stress
- Depression
- EKR, extracellular signal-regulated kinase
- FST, forced swim test
- GABA, γ-aminobutyric acid
- GABAAR, GABA type A receptor
- HSD, hydroxysteroid dehydrogenase
- NGF, nerve growth factor
- Neurosteroid
- PTSD, post-traumatic stress disorder
- PXR, pregnane xenobiotic receptor
- SBSS, selective brain steroidogenic stimulant
- SSRI, selective serotonin reuptake inhibitor
- Selective brain steroidogenic stimulant
- THP, tetrahydroprogesterone
- TSPO, 18 kDa translocator protein
- TrkB, tropomyosin receptor kinase B
- USV, ultrasonic vocalization
Collapse
Affiliation(s)
- Felipe Borges Almeida
- Graduate Program in Health Sciences: Pharmacology and Toxicology, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), 90050-170, Porto Alegre, RS, Brazil
| | - Maurício Schüler Nin
- Graduate Program in Health Sciences: Pharmacology and Toxicology, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), 90050-170, Porto Alegre, RS, Brazil.,Centro Universitário Metodista do IPA, 90420-060, Porto Alegre, RS, Brazil.,Graduate Program in Biological Sciences: Pharmacology and Therapeutics, Universidade Federal do Rio Grande do Sul (UFRGS), 90040-060, Porto Alegre, RS, Brazil
| | - Helena Maria Tannhauser Barros
- Graduate Program in Health Sciences: Pharmacology and Toxicology, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), 90050-170, Porto Alegre, RS, Brazil
| |
Collapse
|
11
|
Benedetti B, Dannehl D, König R, Coviello S, Kreutzer C, Zaunmair P, Jakubecova D, Weiger TM, Aigner L, Nacher J, Engelhardt M, Couillard-Després S. Functional Integration of Neuronal Precursors in the Adult Murine Piriform Cortex. Cereb Cortex 2020; 30:1499-1515. [PMID: 31647533 PMCID: PMC7132906 DOI: 10.1093/cercor/bhz181] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Revised: 06/05/2019] [Accepted: 07/09/2019] [Indexed: 11/20/2022] Open
Abstract
The extent of functional maturation and integration of nonproliferative neuronal precursors, becoming neurons in the adult murine piriform cortex, is largely unexplored. We thus questioned whether precursors eventually become equivalent to neighboring principal neurons or whether they represent a novel functional network element. Adult brain neuronal precursors and immature neurons (complex cells) were labeled in transgenic mice (DCX-DsRed and DCX-CreERT2 /flox-EGFP), and their cell fate was characterized with patch clamp experiments and morphometric analysis of axon initial segments. Young (DCX+) complex cells in the piriform cortex of 2- to 4-month-old mice received sparse synaptic input and fired action potentials at low maximal frequency, resembling neonatal principal neurons. Following maturation, the synaptic input detected on older (DCX-) complex cells was larger, but predominantly GABAergic, despite evidence of glutamatergic synaptic contacts. Furthermore, the rheobase current of old complex cells was larger and the maximal firing frequency was lower than those measured in neighboring age-matched principal neurons. The striking differences between principal neurons and complex cells suggest that the latter are a novel type of neuron and new coding element in the adult brain rather than simple addition or replacement for preexisting network components.
Collapse
Affiliation(s)
- Bruno Benedetti
- Spinal Cord Injury and Tissue Regeneration Center Salzburg, 5020 Salzburg, Austria
- Institute of Experimental Neuroregeneration, Paracelsus Medical University, 5020 Salzburg, Austria
- Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Dominik Dannehl
- Spinal Cord Injury and Tissue Regeneration Center Salzburg, 5020 Salzburg, Austria
- Institute of Experimental Neuroregeneration, Paracelsus Medical University, 5020 Salzburg, Austria
- Institute of Neuroanatomy, CBTM, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
| | - Richard König
- Spinal Cord Injury and Tissue Regeneration Center Salzburg, 5020 Salzburg, Austria
- Institute of Molecular Regenerative Medicine, Paracelsus Medical University, 5020 Salzburg, Austria
| | - Simona Coviello
- BIOTECMED, Universitat de València and Center for Collaborative Research on Mental Health CIBERSAM, 46100 València, Spain
| | - Christina Kreutzer
- Spinal Cord Injury and Tissue Regeneration Center Salzburg, 5020 Salzburg, Austria
- Institute of Experimental Neuroregeneration, Paracelsus Medical University, 5020 Salzburg, Austria
- Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Pia Zaunmair
- Spinal Cord Injury and Tissue Regeneration Center Salzburg, 5020 Salzburg, Austria
- Institute of Experimental Neuroregeneration, Paracelsus Medical University, 5020 Salzburg, Austria
- Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Dominika Jakubecova
- Spinal Cord Injury and Tissue Regeneration Center Salzburg, 5020 Salzburg, Austria
- Institute of Experimental Neuroregeneration, Paracelsus Medical University, 5020 Salzburg, Austria
- Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Thomas M Weiger
- Department of Biosciences, University of Salzburg, 5020 Salzburg, Austria
| | - Ludwig Aigner
- Spinal Cord Injury and Tissue Regeneration Center Salzburg, 5020 Salzburg, Austria
- Austrian Cluster for Tissue Regeneration, Vienna, Austria
- Institute of Molecular Regenerative Medicine, Paracelsus Medical University, 5020 Salzburg, Austria
| | - Juan Nacher
- BIOTECMED, Universitat de València and Center for Collaborative Research on Mental Health CIBERSAM, 46100 València, Spain
| | - Maren Engelhardt
- Institute of Neuroanatomy, CBTM, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
| | - Sébastien Couillard-Després
- Spinal Cord Injury and Tissue Regeneration Center Salzburg, 5020 Salzburg, Austria
- Institute of Experimental Neuroregeneration, Paracelsus Medical University, 5020 Salzburg, Austria
- Austrian Cluster for Tissue Regeneration, Vienna, Austria
| |
Collapse
|
12
|
Tomás FJB, Turko P, Heilmann H, Trimbuch T, Yanagawa Y, Vida I, Münster-Wandowski A. BDNF Expression in Cortical GABAergic Interneurons. Int J Mol Sci 2020; 21:E1567. [PMID: 32106593 PMCID: PMC7084226 DOI: 10.3390/ijms21051567] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 02/07/2020] [Accepted: 02/21/2020] [Indexed: 12/14/2022] Open
Abstract
Brain-derived neurotrophic factor (BDNF) is a major neuronal growth factor that is widely expressed in the central nervous system. It is synthesized as a glycosylated precursor protein, (pro)BDNF and post-translationally converted to the mature form, (m)BDNF. BDNF is known to be produced and secreted by cortical glutamatergic principal cells (PCs); however, it remains a question whether it can also be synthesized by other neuron types, in particular, GABAergic interneurons (INs). Therefore, we utilized immunocytochemical labeling and reverse transcription quantitative PCR (RT-qPCR) to investigate the cellular distribution of proBDNF and its RNA in glutamatergic and GABAergic neurons of the mouse cortex. Immunofluorescence labeling revealed that mBDNF, as well as proBDNF, localized to both the neuronal populations in the hippocampus. The precursor proBDNF protein showed a perinuclear distribution pattern, overlapping with the rough endoplasmic reticulum (ER), the site of protein synthesis. RT-qPCR of samples obtained using laser capture microdissection (LCM) or fluorescence-activated cell sorting (FACS) of hippocampal and cortical neurons further demonstrated the abundance of BDNF transcripts in both glutamatergic and GABAergic cells. Thus, our data provide compelling evidence that BDNF can be synthesized by both principal cells and INs of the cortex.
Collapse
Affiliation(s)
- Federico José Barreda Tomás
- Institute of Integrative Neuroanatomy, Charité-Universitätsmedizin Berlin, Campus Mitte, 10117 Berlin, Germany; (F.J.B.T.); (P.T.); (H.H.); (I.V.)
- Bernstein Center for Computational Neuroscience (BCCN) Berlin, 10115 Berlin, Germany
| | - Paul Turko
- Institute of Integrative Neuroanatomy, Charité-Universitätsmedizin Berlin, Campus Mitte, 10117 Berlin, Germany; (F.J.B.T.); (P.T.); (H.H.); (I.V.)
| | - Heike Heilmann
- Institute of Integrative Neuroanatomy, Charité-Universitätsmedizin Berlin, Campus Mitte, 10117 Berlin, Germany; (F.J.B.T.); (P.T.); (H.H.); (I.V.)
| | - Thorsten Trimbuch
- Institute of Neurophysiology, Charité - Universitätsmedizin Berlin, Campus Mitte, 10117 Berlin, Germany;
| | - Yuchio Yanagawa
- Departments of Genetic and Behavioral Neuroscience, Graduate School of Medicine, Gunma University, Graduate School of Medicine, Maebashi City 371-8511, Japan;
| | - Imre Vida
- Institute of Integrative Neuroanatomy, Charité-Universitätsmedizin Berlin, Campus Mitte, 10117 Berlin, Germany; (F.J.B.T.); (P.T.); (H.H.); (I.V.)
- Bernstein Center for Computational Neuroscience (BCCN) Berlin, 10115 Berlin, Germany
| | - Agnieszka Münster-Wandowski
- Institute of Integrative Neuroanatomy, Charité-Universitätsmedizin Berlin, Campus Mitte, 10117 Berlin, Germany; (F.J.B.T.); (P.T.); (H.H.); (I.V.)
| |
Collapse
|
13
|
Meis S, Endres T, Munsch T, Lessmann V. Impact of Chronic BDNF Depletion on GABAergic Synaptic Transmission in the Lateral Amygdala. Int J Mol Sci 2019; 20:ijms20174310. [PMID: 31484392 PMCID: PMC6747405 DOI: 10.3390/ijms20174310] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 08/30/2019] [Accepted: 09/01/2019] [Indexed: 01/14/2023] Open
Abstract
Brain-derived neurotrophic factor (BDNF) has previously been shown to play an important role in glutamatergic synaptic plasticity in the amygdala, correlating with cued fear learning. While glutamatergic neurotransmission is facilitated by BDNF signaling in the amygdala, its mechanism of action at inhibitory synapses in this nucleus is far less understood. We therefore analyzed the impact of chronic BDNF depletion on GABAA-mediated synaptic transmission in BDNF heterozygous knockout mice (BDNF+/−). Analysis of miniature and evoked inhibitory postsynaptic currents (IPSCs) in the lateral amygdala (LA) revealed neither pre- nor postsynaptic differences in BDNF+/− mice compared to wild-type littermates. In addition, long-term potentiation (LTP) of IPSCs was similar in both genotypes. In contrast, facilitation of spontaneous IPSCs (sIPSCs) by norepinephrine (NE) was significantly reduced in BDNF+/− mice. These results argue against a generally impaired efficacy and plasticity at GABAergic synapses due to a chronic BDNF deficit. Importantly, the increase in GABAergic tone mediated by NE is reduced in BDNF+/− mice. As release of NE is elevated during aversive behavioral states in the amygdala, effects of a chronic BDNF deficit on GABAergic inhibition may become evident in response to states of high arousal, leading to amygdala hyper-excitability and impaired amygdala function.
Collapse
Affiliation(s)
- Susanne Meis
- Institut für Physiologie, Otto-von-Guericke-Universität, D-39120 Magdeburg, Germany.
- Center for Behavioral Brain Sciences, D-39106 Magdeburg, Germany.
| | - Thomas Endres
- Institut für Physiologie, Otto-von-Guericke-Universität, D-39120 Magdeburg, Germany.
| | - Thomas Munsch
- Institut für Physiologie, Otto-von-Guericke-Universität, D-39120 Magdeburg, Germany.
- Center for Behavioral Brain Sciences, D-39106 Magdeburg, Germany.
| | - Volkmar Lessmann
- Institut für Physiologie, Otto-von-Guericke-Universität, D-39120 Magdeburg, Germany.
- Center for Behavioral Brain Sciences, D-39106 Magdeburg, Germany.
| |
Collapse
|
14
|
Off-label Antidepressant Use for Treatment and Management of Chronic Pain: Evolving Understanding and Comprehensive Review. Curr Pain Headache Rep 2019; 23:66. [DOI: 10.1007/s11916-019-0803-z] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
15
|
Li RS, Fukumori R, Takeda T, Song Y, Morimoto S, Kikura-Hanajiri R, Yamaguchi T, Watanabe K, Aritake K, Tanaka Y, Yamada H, Yamamoto T, Ishii Y. Elevation of endocannabinoids in the brain by synthetic cannabinoid JWH-018: mechanism and effect on learning and memory. Sci Rep 2019; 9:9621. [PMID: 31270353 PMCID: PMC6610139 DOI: 10.1038/s41598-019-45969-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Accepted: 06/19/2019] [Indexed: 02/06/2023] Open
Abstract
The impairment of learning and memory is a well-documented effect of both natural and synthetic cannabinoids. In the present study, we aimed to investigate the effect of acute administration of JWH-018, a synthetic cannabinoid, on the hippocampal metabolome to assess biochemical changes in vivo. JWH-018 elevated levels of the endocannabinoids, anandamide (AEA) and 2-arachidonoylglycerol (2-AG). The increase of endocannabinoid levels in response to JWH-018 could be inhibited by co-administration of AM251, a CB1 receptor antagonist. Biochemical analyses revealed that this was the result of suppression of two hydrolases involved in endocannabinoid degradation (fatty acid amide hydrolase [FAAH] and monoacylglycerol lipase [MAGL]). Additionally, we showed that JWH-018 causes a reduction in the levels of brain-derived neurotrophic factor (BDNF), which is known to modulate synaptic plasticity and adaptive processes underlying learning and memory. The decrease of BDNF following JWH-018 treatment was also rescued by co-administration of AM251. As both endocannabinoids and BDNF have been shown to modulate learning and memory in the hippocampus, the alteration of their levels in response to JWH-018 may explain the contribution of synthetic cannabinoids to impairment of memory.
Collapse
Affiliation(s)
- Ren-Shi Li
- Laboratory of Molecular Life Sciences, Graduate School of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan.,Research Department of Pharmacognosy, China Pharmaceutical University, Nanjing, 211198, People's Republic of China
| | - Ryo Fukumori
- Department of Pharmacotherapeutics and Neuropsychopharmacology, Faculty of Pharmaceutical Sciences, Nagasaki International University, Sasebo, Nagasaki, Japan
| | - Tomoki Takeda
- Laboratory of Molecular Life Sciences, Graduate School of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Yingxia Song
- Laboratory of Molecular Life Sciences, Graduate School of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Satoshi Morimoto
- Division of Pharmacognosy, Graduate School of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Ruri Kikura-Hanajiri
- Division of Pharmacognosy, Phytochemistry and Narcotics, National Institute of Health Sciences (NIHS), 3-25-26 Tonomachi, Kawasaki-ku, Kawasaki-city, Kanagawa, 210-9501, Japan
| | - Taku Yamaguchi
- Department of Pharmacotherapeutics and Neuropsychopharmacology, Faculty of Pharmaceutical Sciences, Nagasaki International University, Sasebo, Nagasaki, Japan
| | - Kazuhito Watanabe
- Daiichi University of Pharmacy, 22-1 Tamagawa-cho, Minami-ku Fukuoka, 815-8511, Japan
| | - Kousuke Aritake
- Daiichi University of Pharmacy, 22-1 Tamagawa-cho, Minami-ku Fukuoka, 815-8511, Japan
| | - Yoshitaka Tanaka
- Division of Pharmaceutical Cell Biology, Graduate School of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Hideyuki Yamada
- Laboratory of Molecular Life Sciences, Graduate School of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Tsuneyuki Yamamoto
- Department of Pharmacotherapeutics and Neuropsychopharmacology, Faculty of Pharmaceutical Sciences, Nagasaki International University, Sasebo, Nagasaki, Japan
| | - Yuji Ishii
- Laboratory of Molecular Life Sciences, Graduate School of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan.
| |
Collapse
|
16
|
Telias M. Molecular Mechanisms of Synaptic Dysregulation in Fragile X Syndrome and Autism Spectrum Disorders. Front Mol Neurosci 2019; 12:51. [PMID: 30899214 PMCID: PMC6417395 DOI: 10.3389/fnmol.2019.00051] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Accepted: 02/12/2019] [Indexed: 12/21/2022] Open
Abstract
Fragile X syndrome (FXS) is the most common form of monogenic hereditary cognitive impairment. FXS patient exhibit a high comorbidity rate with autism spectrum disorders (ASDs). This makes FXS a model disease for understanding how synaptic dysregulation alters neuronal excitability, learning and memory, social behavior, and more. Since 1991, with the discovery of fragile X mental retardation 1 (FMR1) as the sole gene that is mutated in FXS, thousands of studies into the function of the gene and its encoded protein FMR1 protein (FMRP), have been conducted, yielding important information regarding the pathophysiology of the disease, as well as insight into basic synaptic mechanisms that control neuronal networking and circuitry. Among the most important, are molecular mechanisms directly involved in plasticity, including glutamate and γ-aminobutyric acid (GABA) receptors, which can control synaptic transmission and signal transduction, including short- and long-term plasticity. More recently, several novel mechanisms involving growth factors, enzymatic cascades and transcription factors (TFs), have been proposed to have the potential of explaining some of the synaptic dysregulation in FXS. In this review article, I summarize the main mechanisms proposed to underlie synaptic disruption in FXS and ASDs. I focus on studies conducted on the Fmr1 knock-out (KO) mouse model and on FXS-human pluripotent stem cells (hPSCs), emphasizing the differences and even contradictions between mouse and human, whenever possible. As FXS and ASDs are both neurodevelopmental disorders that follow a specific time-course of disease progression, I highlight those studies focusing on the differential developmental regulation of synaptic abnormalities in these diseases.
Collapse
Affiliation(s)
- Michael Telias
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, United States
| |
Collapse
|
17
|
Concomitant abuse of methadone and methamphetamine could impair spatial learning and memory in male rats. LEARNING AND MOTIVATION 2019. [DOI: 10.1016/j.lmot.2019.01.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
18
|
The role of agrin, Lrp4 and MuSK during dendritic arborization and synaptogenesis in cultured embryonic CNS neurons. Dev Biol 2019; 445:54-67. [DOI: 10.1016/j.ydbio.2018.10.017] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Revised: 10/22/2018] [Accepted: 10/22/2018] [Indexed: 01/06/2023]
|
19
|
Nguyen LT, Reverter A, Cánovas A, Venus B, Anderson ST, Islas-Trejo A, Dias MM, Crawford NF, Lehnert SA, Medrano JF, Thomas MG, Moore SS, Fortes MRS. STAT6, PBX2, and PBRM1 Emerge as Predicted Regulators of 452 Differentially Expressed Genes Associated With Puberty in Brahman Heifers. Front Genet 2018; 9:87. [PMID: 29616079 PMCID: PMC5869259 DOI: 10.3389/fgene.2018.00087] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Accepted: 03/02/2018] [Indexed: 12/17/2022] Open
Abstract
The liver plays a central role in metabolism and produces important hormones. Hepatic estrogen receptors and the release of insulin-like growth factor 1 (IGF1) are critical links between liver function and the reproductive system. However, the role of liver in pubertal development is not fully understood. To explore this question, we applied transcriptomic analyses to liver samples of pre- and post-pubertal Brahman heifers and identified differentially expressed (DE) genes and genes encoding transcription factors (TFs). Differential expression of genes suggests potential biological mechanisms and pathways linking liver function to puberty. The analyses identified 452 DE genes and 82 TF with significant contribution to differential gene expression by using a regulatory impact factor metric. Brain-derived neurotrophic factor was observed as the most down-regulated gene (P = 0.003) in post-pubertal heifers and we propose this gene influences pubertal development in Brahman heifers. Additionally, co-expression network analysis provided evidence for three TF as key regulators of liver function during pubertal development: the signal transducer and activator of transcription 6, PBX homeobox 2, and polybromo 1. Pathway enrichment analysis identified transforming growth factor-beta and Wnt signaling pathways as significant annotation terms for the list of DE genes and TF in the co-expression network. Molecular information regarding genes and pathways described in this work are important to further our understanding of puberty onset in Brahman heifers.
Collapse
Affiliation(s)
- Loan T Nguyen
- School of Chemistry and Molecular Biosciences, The University of Queensland, St. Lucia, QLD, Australia.,Faculty of Biotechnology, Vietnam National University of Agriculture, Hanoi, Vietnam
| | - Antonio Reverter
- CSIRO Agriculture and Food, Queensland Bioscience Precinct, St. Lucia, QLD, Australia
| | - Angela Cánovas
- Centre for Genetic Improvement of Livestock, Department of Animal Biosciences, University of Guelph, Guelph, ON, Canada
| | - Bronwyn Venus
- Queensland Alliance for Agriculture and Food Innovation, The University of Queensland, St. Lucia, QLD, Australia
| | - Stephen T Anderson
- School of Biomedical Sciences, The University of Queensland, Brisbane, QLD, Australia
| | - Alma Islas-Trejo
- Department of Animal Science, University of California, Davis, Davis, CA, United States
| | - Marina M Dias
- Departamento de Zootecnia, Faculdade de Ciências Agráìrias e Veterináìrias, Universidade Estadual Paulista Júlio de Mesquita Filho, São Paulo, Brazil
| | - Natalie F Crawford
- Department of Animal Science, Colorado State University, Fort Collins, CO, United States
| | - Sigrid A Lehnert
- CSIRO Agriculture and Food, Queensland Bioscience Precinct, St. Lucia, QLD, Australia
| | - Juan F Medrano
- Department of Animal Science, University of California, Davis, Davis, CA, United States
| | - Milt G Thomas
- Department of Animal Science, Colorado State University, Fort Collins, CO, United States
| | - Stephen S Moore
- Queensland Alliance for Agriculture and Food Innovation, The University of Queensland, St. Lucia, QLD, Australia
| | - Marina R S Fortes
- School of Chemistry and Molecular Biosciences, The University of Queensland, St. Lucia, QLD, Australia.,Queensland Alliance for Agriculture and Food Innovation, The University of Queensland, St. Lucia, QLD, Australia
| |
Collapse
|
20
|
Yiannakas A, Rosenblum K. The Insula and Taste Learning. Front Mol Neurosci 2017; 10:335. [PMID: 29163022 PMCID: PMC5676397 DOI: 10.3389/fnmol.2017.00335] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Accepted: 10/03/2017] [Indexed: 12/29/2022] Open
Abstract
The sense of taste is a key component of the sensory machinery, enabling the evaluation of both the safety as well as forming associations regarding the nutritional value of ingestible substances. Indicative of the salience of the modality, taste conditioning can be achieved in rodents upon a single pairing of a tastant with a chemical stimulus inducing malaise. This robust associative learning paradigm has been heavily linked with activity within the insular cortex (IC), among other regions, such as the amygdala and medial prefrontal cortex. A number of studies have demonstrated taste memory formation to be dependent on protein synthesis at the IC and to correlate with the induction of signaling cascades involved in synaptic plasticity. Taste learning has been shown to require the differential involvement of dopaminergic GABAergic, glutamatergic, muscarinic neurotransmission across an extended taste learning circuit. The subsequent activation of downstream protein kinases (ERK, CaMKII), transcription factors (CREB, Elk-1) and immediate early genes (c-fos, Arc), has been implicated in the regulation of the different phases of taste learning. This review discusses the relevant neurotransmission, molecular signaling pathways and genetic markers involved in novel and aversive taste learning, with a particular focus on the IC. Imaging and other studies in humans have implicated the IC in the pathophysiology of a number of cognitive disorders. We conclude that the IC participates in circuit-wide computations that modulate the interception and encoding of sensory information, as well as the formation of subjective internal representations that control the expression of motivated behaviors.
Collapse
Affiliation(s)
- Adonis Yiannakas
- Sagol Department of Neuroscience, University of Haifa, Haifa, Israel
| | - Kobi Rosenblum
- Sagol Department of Neuroscience, University of Haifa, Haifa, Israel
- Center for Gene Manipulation in the Brain, University of Haifa, Haifa, Israel
| |
Collapse
|
21
|
Yang Y, Gao L. Celecoxib Alleviates Memory Deficits by Downregulation of COX-2 Expression and Upregulation of the BDNF-TrkB Signaling Pathway in a Diabetic Rat Model. J Mol Neurosci 2017; 62:188-198. [PMID: 28466254 PMCID: PMC5486519 DOI: 10.1007/s12031-017-0922-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Accepted: 04/13/2017] [Indexed: 01/09/2023]
Abstract
Previous studies conveyed that diabetes causes learning and memory deficits. Data also suggest that celecoxib exerts an anti-hyperalgesic, anti-allodynic, and a plethora of other beneficial effects in diabetic rats. However, whether celecoxib could alleviate memory deficit in diabetic rat is unknown. In the present study, we aimed to examine the potential of celecoxib to counter memory deficits in diabetes. Experimental diabetes was induced by streptozotocin (STZ, 60 mg/kg) in male SD rats. Rats were divided into three groups (n = 16/group): normal control group injected with normal saline, diabetes group injected with STZ, and diabetes + celecoxib group in which diabetic rats were administered with celecoxib by gavage in drinking water (10 mg/kg) for 10 days in terms of which memory performance in animals was measured, hippocampal tissue harvested, and long-term potentiation assessed. Western blotting and immunohistochemical staining were performed to determine cyclooxygenase 2 (COX-2) expression in hippocampus. The results showed that a rat model of STZ-induced diabetes was successfully established and that celecoxib treatment significantly improved the associated nephropathy and inflammation. Moreover, spatial memory and hippocampal long-term potentiation (LTP) were impaired in diabetic model (P < 0.05). Interestingly, our data revealed that oral application of celecoxib reversed the memory deficit and hippocampal LTP in the diabetic rats. To understand the underlying mechanisms, the expression of some important pathways involved in memory impairment was determined. We found that brain-derived neurotrophic factor (BDNF) and phosphorylated tropomyosin-related kinase (p-TrkB) were decreased in diabetic rats but were effectively reversed by celecoxib treatment. As evidenced by western blotting and immunohistochemical staining, the expression of COX-2 in hippocampus was significantly upregulated in diabetic rat (P < 0.05) but inhibited by celecoxib treatment. The present findings provide novel data that celecoxib reverses memory deficits via probable downregulation of hippocampal COX-2 expression and upregulation of the BDNF-TrkB signaling pathway in a diabetic rat.
Collapse
Affiliation(s)
- Ying Yang
- Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, No. 136, Jingzhou Street, Xiangcheng District, Xiangyang City, Hubei, 441021, China
| | - Ling Gao
- Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, No. 136, Jingzhou Street, Xiangcheng District, Xiangyang City, Hubei, 441021, China.
| |
Collapse
|
22
|
Aoki C, Chowdhury TG, Wable GS, Chen YW. Synaptic changes in the hippocampus of adolescent female rodents associated with resilience to anxiety and suppression of food restriction-evoked hyperactivity in an animal model for anorexia nervosa. Brain Res 2017; 1654:102-115. [PMID: 26779892 PMCID: PMC4947030 DOI: 10.1016/j.brainres.2016.01.019] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Accepted: 01/10/2016] [Indexed: 12/30/2022]
Abstract
Anorexia nervosa is a mental illness that emerges primarily during early adolescence, with mortality rate that is 200 times higher than that of suicide. The illness is characterized by intense fear of gaining weight, heightened anxiety, obstinate food restriction, often accompanied by excessive exercise, in spite of mounting hunger. The illness affects females nine times more often than males, suggesting an endocrine role in its etiology. Its relapse rate exceeds 25%, yet there are no accepted pharmacological treatments to prevent this. Here, we summarize studies from this laboratory that have used adolescent female rodents in activity-based anorexia (ABA), an animal model of anorexia nervosa, with the goal of identifying neurobiological underpinnings of this disease. We put forth a hypothesis that a GABAergic mechanism within the hippocampus is central to regulating an individual׳s anxiety which, in turn, strongly influences the individual׳s resilience/vulnerability to ABA. In particular, we propose that ionotropic GABAA receptors containing the subunits alpha4 and delta, are at play for exerting shunting inhibition upon hippocampal pyramidal neurons that become more excitable during ABA. Since these receptors confer insensitivity to benzodiazepines, this pharmacological profile of ABA fits with lack of report indicating efficacy of benzodiazepines in reducing the anxiety experienced by individuals with anorexia nervosa. The idea that the GABAergic system of the hippocampus regulates resilience/vulnerability to anorexia nervosa complements current opinions about the important roles of the prefrontal cortex, amygdala, striatum, gustatory pathways and feeding centers of the hypothalamus and of the neuromodulators, serotonin and dopamine, in the etiology of the disease. This article is part of a Special Issue entitled SI: Adolescent plasticity.
Collapse
Affiliation(s)
- Chiye Aoki
- Center for Neural Science, 4 Washington Place, Room 809, New York, NY 10003, United States.
| | - Tara G Chowdhury
- Center for Neural Science, 4 Washington Place, Room 809, New York, NY 10003, United States
| | - Gauri S Wable
- Center for Neural Science, 4 Washington Place, Room 809, New York, NY 10003, United States
| | - Yi-Wen Chen
- Center for Neural Science, 4 Washington Place, Room 809, New York, NY 10003, United States
| |
Collapse
|
23
|
Experience Affects Critical Period Plasticity in the Visual Cortex through an Epigenetic Regulation of Histone Post-Translational Modifications. J Neurosci 2016; 36:3430-40. [PMID: 27013673 DOI: 10.1523/jneurosci.1787-15.2016] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2015] [Accepted: 01/05/2016] [Indexed: 11/21/2022] Open
Abstract
UNLABELLED During an early phase of enhanced sensitivity called the critical period (CP), monocular deprivation causes a shift in the response of visual cortex binocular neurons in favor of the nondeprived eye, a process named ocular dominance (OD) plasticity. While the time course of the CP for OD plasticity can be modulated by genetic/pharmacological interventions targeting GABAergic inhibition, whether an increased sensory-motor experience can affect this major plastic phenomenon is not known. We report that exposure to environmental enrichment (EE) accelerated the closure of the CP for OD plasticity in the rat visual cortex. Histone H3 acetylation was developmentally regulated in primary visual cortex, with enhanced levels being detectable early in enriched pups, and chromatin immunoprecipitation revealed an increase at the level of the BDNF P3 promoter. Administration of the histone deacetylase inhibitor SAHA (suberoylanilide hydroxamic acid) to animals reared in a standard cage mimicked the increase in H3 acetylation observed in the visual cortex and resulted in an accelerated decay of OD plasticity. Finally, exposure to EE in adulthood upregulated H3 acetylation and was paralleled by a reopening of the CP. These findings demonstrate a critical involvement of the epigenetic machinery as a mediator of visual cortex developmental plasticity and of the impact of EE on OD plasticity. SIGNIFICANCE STATEMENT While it is known that an epigenetic remodeling of chromatin structure controls developmental plasticity in the visual cortex, three main questions have remained open. Which is the physiological time course of histone modifications? Is it possible, by manipulating the chromatin epigenetic state, to modulate plasticity levels during the critical period? How can we regulate histone acetylation in the adult brain in a noninvasive manner? We show that the early exposure of rat pups to enriching environmental conditions accelerates the critical period for plasticity in the primary visual cortex, linking this effect to increased histone acetylation, specifically at the BDNF gene level. Moreover, we report that the exposure of adult animals to environmental enrichment enhances histone acetylation and reopens juvenile-like plasticity.
Collapse
|
24
|
Yang C, Sun N, Liu Z, Li X, Xu Y, Zhang K. The interaction of combined effects of the BDNF and PRKCG genes and negative life events in major depressive disorder. Psychiatry Res 2016; 237:72-7. [PMID: 26921055 DOI: 10.1016/j.psychres.2016.01.076] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Revised: 11/20/2015] [Accepted: 01/31/2016] [Indexed: 02/07/2023]
Abstract
Major depressive disorder (MDD) is a mental disorder that results from complex interplay between multiple and partially overlapping sets of susceptibility genes and environmental factors. The brain derived neurotrophic factor (BDNF) and Protein kinase C gamma type (PRKCG) are logical candidate genes in MDD. Among diverse environmental factors, negative life events have been suggested to exert a crucial impact on brain development. In the present study, we hypothesized that interactions between genetic variants in BDNF and PRKCG and negative life events may play an important role in the development of MDD. We recruited a total of 406 patients with MDD and 391 age- and gender-matched control subjects. Gene-environment interactions were analyzed using generalized multifactor dimensionality reduction (GMDR). Under a dominant model, we observed a significant three-way interaction among BDNF rs6265, PRKCG rs3745406, and negative life events. The gene-environment combination of PRKCG rs3745406 C allele, BDNF rs6265 G allele and high level of negative life events (C-G-HN) was significantly associated with MDD (OR, 5.97; 95% CI, 2.71-13.15). To our knowledge, this is the first report of evidence that the BDNF-PRKCG interaction may modify the relationship between negative life events and MDD in the Chinese population.
Collapse
Affiliation(s)
- Chunxia Yang
- Department of Psychiatry, First Hospital of Shanxi Medical University, 85 Jiefang South Road, Taiyuan 030001, People's Republic of China
| | - Ning Sun
- Department of Psychiatry, First Hospital of Shanxi Medical University, 85 Jiefang South Road, Taiyuan 030001, People's Republic of China
| | - Zhifen Liu
- Department of Psychiatry, First Hospital of Shanxi Medical University, 85 Jiefang South Road, Taiyuan 030001, People's Republic of China
| | - Xinrong Li
- Department of Psychiatry, First Hospital of Shanxi Medical University, 85 Jiefang South Road, Taiyuan 030001, People's Republic of China
| | - Yong Xu
- Department of Psychiatry, First Hospital of Shanxi Medical University, 85 Jiefang South Road, Taiyuan 030001, People's Republic of China
| | - Kerang Zhang
- Department of Psychiatry, First Hospital of Shanxi Medical University, 85 Jiefang South Road, Taiyuan 030001, People's Republic of China.
| |
Collapse
|
25
|
Astrocytes Protect against Isoflurane Neurotoxicity by Buffering pro-brain-derived Neurotrophic Factor. Anesthesiology 2015; 123:810-9. [PMID: 26270940 DOI: 10.1097/aln.0000000000000824] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
BACKGROUND Isoflurane induces cell death in neurons undergoing synaptogenesis via increased production of pro-brain-derived neurotrophic factor (proBDNF) and activation of postsynaptic p75 neurotrophin receptor (p75). Astrocytes express p75, but their role in neuronal p75-mediated cell death remains unclear. The authors investigated whether astrocytes have the capacity to buffer increases in proBDNF and protect against isoflurane/p75 neurotoxicity. METHODS Cell death was assessed in day in vitro (DIV) 7 mouse primary neuronal cultures alone or in co-culture with age-matched or DIV 21 astrocytes with propidium iodide 24 h after 1 h exposure to 2% isoflurane or recombinant proBDNF. Astrocyte-targeted knockdown of p75 in co-culture was achieved with small-interfering RNA and astrocyte-specific transfection reagent and verified with immunofluorescence microscopy. proBDNF levels were assessed by enzyme-linked immunosorbent assay. Each experiment used six to eight replicate cultures/condition and was repeated at least three times. RESULTS Exposure to isoflurane significantly (P < 0.05) increased neuronal cell death in primary neuronal cultures (1.5 ± 0.7 fold, mean ± SD) but not in co-culture with DIV 7 (1.0 ± 0.5 fold) or DIV 21 astrocytes (1.2 ± 1.2 fold). Exogenous proBDNF dose dependently induced neuronal cell death in both primary neuronal and co-cultures, an effect enhanced by astrocyte p75 inhibition. Astrocyte-targeted p75 knockdown in co-cultures increased media proBDNF (1.2 ± 0.1 fold) and augmented isoflurane-induced neuronal cell death (3.8 ± 3.1 fold). CONCLUSIONS The presence of astrocytes provides protection to growing neurons by buffering increased levels of proBDNF induced by isoflurane. These findings may hold clinical significance for the neonatal and injured brain where increased levels of proBDNF impair neurogenesis.
Collapse
|
26
|
Nakamura Y, Darnieder LM, Deeb TZ, Moss SJ. Regulation of GABAARs by phosphorylation. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2015; 72:97-146. [PMID: 25600368 PMCID: PMC5337123 DOI: 10.1016/bs.apha.2014.11.008] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
γ-Aminobutyric acid type A receptors (GABAARs) are the principal mediators of fast synaptic inhibition in the brain as well as the low persistent extrasynaptic inhibition, both of which are fundamental to proper brain function. Thus unsurprisingly, deficits in GABAARs are implicated in a number of neurological disorders and diseases. The complexity of GABAAR regulation is determined not only by the heterogeneity of these receptors but also by its posttranslational modifications, the foremost, and best characterized of which is phosphorylation. This review will explore the details of this dynamic process, our understanding of which has barely scratched the surface. GABAARs are regulated by a number of kinases and phosphatases, and its phosphorylation plays an important role in governing its trafficking, expression, and interaction partners. Here, we summarize the progress in understanding the role phosphorylation plays in the regulation of GABAARs. This includes how phosphorylation can affect the allosteric modulation of GABAARs, as well as signaling pathways that affect GABAAR phosphorylation. Finally, we discuss the dysregulation of GABAAR phosphorylation and its implication in disease processes.
Collapse
|
27
|
Short-Term Plasticity of Glutamatergic and GABA-Ergic Synaptic Transmission between Co-Cultured Retinal Ganglion Cells and Superior Colliculus Neurons. NEUROPHYSIOLOGY+ 2014. [DOI: 10.1007/s11062-014-9449-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
28
|
Zhao L, Levine ES. BDNF-endocannabinoid interactions at neocortical inhibitory synapses require phospholipase C signaling. J Neurophysiol 2013; 111:1008-15. [PMID: 24335212 DOI: 10.1152/jn.00554.2013] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Endogenous cannabinoids (endocannabinoids) and neurotrophins, particularly brain-derived neurotrophic factor (BDNF), are potent synaptic modulators that are expressed throughout the forebrain and play critical roles in many behavioral processes. Although the effects of BDNF at excitatory synapses have been well characterized, the mechanisms of action of BDNF at inhibitory synapses are not well understood. Previously we have found that BDNF suppresses presynaptic GABA release in layer 2/3 of the neocortex via postsynaptic tropomyosin-related kinase receptor B (trkB) receptor-induced release of endocannabinoids. To examine the intracellular signaling pathways that underlie this effect, we used pharmacological approaches and whole cell patch-clamp techniques in layer 2/3 pyramidal neurons of somatosensory cortex in brain slices from juvenile Swiss CD1 mice. Our results indicated that phospholipase Cγ (PLCγ) is involved in the CB1 receptor-mediated synaptic effect of BDNF, because the BDNF effect was blocked in the presence of the broad-spectrum PLC inhibitors U-73122 and edelfosine, whereas the inactive analog U-73343 did not alter the suppressive effect of BDNF at inhibitory synapses. Endocannabinoid release can also be triggered by metabotropic glutamate receptor (mGluR)-mediated activation of PLCβ, and BDNF has been shown to enhance spontaneous glutamate release. An mGluR antagonist, E4CPG, however, did not block the BDNF effect. In addition, the effect of BDNF was independent of other signaling pathways downstream of trkB receptor activation, namely, mitogen-activated protein kinase and phosphoinositide 3-kinase pathways, as well as protein kinase C signaling.
Collapse
Affiliation(s)
- Liangfang Zhao
- Department of Neuroscience, University of Connecticut Health Center, Farmington, Connecticut
| | | |
Collapse
|
29
|
Nieto-Gonzalez JL, Jensen K. BDNF Depresses Excitability of Parvalbumin-Positive Interneurons through an M-Like Current in Rat Dentate Gyrus. PLoS One 2013; 8:e67318. [PMID: 23840662 PMCID: PMC3686736 DOI: 10.1371/journal.pone.0067318] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2013] [Accepted: 05/16/2013] [Indexed: 11/19/2022] Open
Abstract
In addition to their classical roles in neuronal growth, survival and differentiation, neurotrophins are also rapid regulators of excitability, synaptic transmission and activity-dependent synaptic plasticity. We have recently shown that mature BDNF (Brain Derived Neurotrophic Factor), but not proBDNF, modulates the excitability of interneurons in dentate gyrus within minutes. Here, we used brain slice patch-clamp recordings to study the mechanisms through which BDNF modulates the firing of interneurons in rat dentate gyrus by binding to TrkB receptors. Bath application of BDNF (15 ng/ml) under current-clamp decreased the firing frequency (by 80%) and input resistance, blocking the delayed firing observed at near-threshold voltage ranges, with no changes in resting membrane potential or action potential waveform. Using TEA (tetraethylammonium), or XE991(a Kv7/KCNQ channel antagonist), the effect of BDNF was abolished, whereas application of retigabine (a Kv7/KCNQ channel opener) mimicked the effect of BDNF, suggesting that the M-current could be implicated in the modulation of the firing. In voltage-clamp experiments, BDNF increased the M-like current amplitude with no change in holding current. This effect was again blocked by XE991 and mimicked by retigabine, the latter accompanied with a change in holding current. In agreement with the electrophysiology, parvalbumin-positive interneurons co-expressed TrkB receptors and Kv7.2/KCNQ2 channels. In conclusion, BDNF depresses the excitability of interneurons by activating an M-like current and possibly blocking Kv1 channels, thereby controlling interneuron resting membrane potential and excitability.
Collapse
Affiliation(s)
| | - Kimmo Jensen
- Department of Biomedicine, Aarhus University, Aarhus C, Denmark
- Lundbeck Foundation Research Center MIND, Aarhus University, Aarhus C, Denmark
| |
Collapse
|
30
|
Mechanisms Underlying Tolerance after Long-Term Benzodiazepine Use: A Future for Subtype-Selective GABA(A) Receptor Modulators? Adv Pharmacol Sci 2012; 2012:416864. [PMID: 22536226 PMCID: PMC3321276 DOI: 10.1155/2012/416864] [Citation(s) in RCA: 87] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2011] [Revised: 10/10/2011] [Accepted: 11/02/2011] [Indexed: 01/01/2023] Open
Abstract
Despite decades of basic and clinical research, our understanding of how benzodiazepines tend to lose their efficacy over time (tolerance) is at least incomplete. In appears that tolerance develops relatively quickly for the sedative and anticonvulsant actions of benzodiazepines, whereas tolerance to anxiolytic and amnesic effects probably does not develop at all. In light of this evidence, we review the current evidence for the neuroadaptive mechanisms underlying benzodiazepine tolerance, including changes of (i) the GABA(A) receptor (subunit expression and receptor coupling), (ii) intracellular changes stemming from transcriptional and neurotrophic factors, (iii) ionotropic glutamate receptors, (iv) other neurotransmitters (serotonin, dopamine, and acetylcholine systems), and (v) the neurosteroid system. From the large variance in the studies, it appears that either different (simultaneous) tolerance mechanisms occur depending on the benzodiazepine effect, or that the tolerance-inducing mechanism depends on the activated GABA(A) receptor subtypes. Importantly, there is no convincing evidence that tolerance occurs with α subunit subtype-selective compounds acting at the benzodiazepine site.
Collapse
|
31
|
Huang Y, Ko H, Cheung ZH, Yung KKL, Yao T, Wang JJ, Morozov A, Ke Y, Ip NY, Yung WH. Dual actions of brain-derived neurotrophic factor on GABAergic transmission in cerebellar Purkinje neurons. Exp Neurol 2012; 233:791-8. [PMID: 22178325 DOI: 10.1016/j.expneurol.2011.11.043] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2011] [Revised: 11/18/2011] [Accepted: 11/25/2011] [Indexed: 10/14/2022]
Abstract
The ability to regulate inhibitory synapses is a critical feature of the nervous system and a growing body of evidence indicates that brain-derived neurotrophic factor (BDNF) acutely modulates the efficacy of GABA synaptic transmission. Although the neuronal potassium-chloride cotransporter 2 (KCC2) has been implied in this BDNF-induced ionic plasticity, the reports about actions of BDNF on GABA signaling remain conflicting. Here we show dual effects of BDNF on GABAergic synaptic transmission in Purkinje neurons in rat cerebellar slices. BDNF decreased the amplitude of evoked outward IPSCs postsynaptically. It induced a depolarizing shift in the reversal potential (E(IPSC)), which reduced the driving force for outward IPSCs. However, in the absence of KCC2 activity, BDNF directly potentiated rather than inhibited GABA(A) receptor, which was reflected by an increase in the amplitude of outward IPSCs. This action of BDNF coincided with its effect in increasing the amplitude of inward IPSCs. Furthermore, an interaction between GABA(A) receptor and KCC2 was revealed by co-immunoprecipitation. The effects of BDNF on both GABA(A) receptor and KCC2 were dependent on TrkB and also activation of cyclin-dependent kinase 5 (Cdk5). However, only the effect of BDNF on KCC2 activity was dependent on a rise of intracellular calcium. Taken together, these data highlight distinct actions of BDNF on KCC2 and GABA(A) receptor in the regulation of GABAergic synaptic transmission.
Collapse
Affiliation(s)
- Ying Huang
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong, China.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Grantyn R, Henneberger C, Jüttner R, Meier JC, Kirischuk S. Functional hallmarks of GABAergic synapse maturation and the diverse roles of neurotrophins. Front Cell Neurosci 2011; 5:13. [PMID: 21772813 PMCID: PMC3131524 DOI: 10.3389/fncel.2011.00013] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2011] [Accepted: 06/17/2011] [Indexed: 12/03/2022] Open
Abstract
Functional impairment of the adult brain can result from deficits in the ontogeny of GABAergic synaptic transmission. Gene defects underlying autism spectrum disorders, Rett’s syndrome or some forms of epilepsy, but also a diverse set of syndromes accompanying perinatal trauma, hormonal imbalances, intake of sleep-inducing or mood-improving drugs or, quite common, alcohol intake during pregnancy can alter GABA signaling early in life. The search for therapeutically relevant endogenous molecules or exogenous compounds able to alleviate the consequences of dysfunction of GABAergic transmission in the embryonic or postnatal brain requires a clear understanding of its site- and state-dependent development. At the level of single synapses, it is necessary to discriminate between presynaptic and postsynaptic alterations, and to define parameters that can be regarded as both suitable and accessible for the quantification of developmental changes. Here we focus on the performance of GABAergic synapses in two brain structures, the hippocampus and the superior colliculus, describe some novel aspects of neurotrophin effects during the development of GABAergic synaptic transmission and examine the applicability of the following rules: (1) synaptic transmission starts with GABA, (2) nascent/immature GABAergic synapses operate in a ballistic mode (multivesicular release), (3) immature synaptic terminals release vesicles with higher probability than mature synapses, (4) immature GABAergic synapses are prone to paired pulse and tetanic depression, (5) synapse maturation is characterized by an increasing dominance of synchronous over asynchronous release, (6) in immature neurons GABA acts as a depolarizing transmitter, (7) synapse maturation implies inhibitory postsynaptic current shortening due to an increase in alpha1 subunit expression, (8) extrasynaptic (tonic) conductances can inhibit the development of synaptic (phasic) GABA actions.
Collapse
Affiliation(s)
- Rosemarie Grantyn
- Institute of Neurophysiology, University Medicine Charité Berlin, Germany
| | | | | | | | | |
Collapse
|
33
|
Ren P, Zhang H, Qiu F, Liu YQ, Gu H, O'Dowd DK, Zhou QY, Hu WP. Prokineticin 2 regulates the electrical activity of rat suprachiasmatic nuclei neurons. PLoS One 2011; 6:e20263. [PMID: 21687716 PMCID: PMC3110640 DOI: 10.1371/journal.pone.0020263] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2011] [Accepted: 04/21/2011] [Indexed: 12/11/2022] Open
Abstract
Neuropeptide signaling plays roles in coordinating cellular activities and maintaining robust oscillations within the mammalian suprachiasmatic nucleus (SCN). Prokineticin2 (PK2) is a signaling molecule from the SCN and involves in the generation of circadian locomotor activity. Prokineticin receptor 2 (PKR2), a receptor for PK2, has been shown to be expressed in the SCN. However, very little is known about the cellular action of PK2 within the SCN. In the present study, we investigated the effect of PK2 on spontaneous firing and miniature inhibitory postsynaptic currents (mIPSCs) using whole cell patch-clamp recording in the SCN slices. PK2 dose-dependently increased spontaneous firing rates in most neurons from the dorsal SCN. PK2 acted postsynaptically to reduce γ-aminobutyric acid (GABA)-ergic function within the SCN, and PK2 reduced the amplitude but not frequency of mIPSCs. Furthermore, PK2 also suppressed exogenous GABA-induced currents. And the inhibitory effect of PK2 required PKC activation in the postsynaptic cells. Our data suggest that PK2 could alter cellular activities within the SCN and may influence behavioral and physiological rhythms.
Collapse
Affiliation(s)
- Ping Ren
- Department of Pharmacology, Xianning College, Xianning, Hubei, People's Republic of China
| | - Huiping Zhang
- Family Planning Research Institute, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Fang Qiu
- Department of Pharmacology, Xianning College, Xianning, Hubei, People's Republic of China
| | - Yu-Qiang Liu
- Department of Pharmacology, Xianning College, Xianning, Hubei, People's Republic of China
| | - Huaiyu Gu
- Departments of Anatomy and Neurobiology, Developmental and Cell Biology, University of California Irvine, Irvine, California, United States of America
| | - Diane K. O'Dowd
- Departments of Anatomy and Neurobiology, Developmental and Cell Biology, University of California Irvine, Irvine, California, United States of America
| | - Qun-Yong Zhou
- Department of Pharmacology, University of California Irvine, Irvine, California, United States of America
| | - Wang-Ping Hu
- Department of Pharmacology, Xianning College, Xianning, Hubei, People's Republic of China
- * E-mail:
| |
Collapse
|
34
|
Abstract
Increasing evidence points to an association between major depressive disorders (MDDs) and diverse types of GABAergic deficits. In this review, we summarize clinical and preclinical evidence supporting a central and causal role of GABAergic deficits in the etiology of depressive disorders. Studies of depressed patients indicate that MDDs are accompanied by reduced brain concentration of the inhibitory neurotransmitter γ-aminobutyric acid (GABA) and by alterations in the subunit composition of the principal receptors (GABA(A) receptors) mediating GABAergic inhibition. In addition, there is abundant evidence that suggests that GABA has a prominent role in the brain control of stress, the most important vulnerability factor in mood disorders. Furthermore, preclinical evidence suggests that currently used antidepressant drugs (ADs) designed to alter monoaminergic transmission and nonpharmacological therapies may ultimately act to counteract GABAergic deficits. In particular, GABAergic transmission has an important role in the control of hippocampal neurogenesis and neural maturation, which are now established as cellular substrates of most if not all antidepressant therapies. Finally, comparatively modest deficits in GABAergic transmission in GABA(A) receptor-deficient mice are sufficient to cause behavioral, cognitive, neuroanatomical and neuroendocrine phenotypes, as well as AD response characteristics expected of an animal model of MDD. The GABAergic hypothesis of MDD suggests that alterations in GABAergic transmission represent fundamentally important aspects of the etiological sequelae of MDDs that are reversed by monoaminergic AD action.
Collapse
|
35
|
GABAA receptor trafficking is regulated by protein kinase C(epsilon) and the N-ethylmaleimide-sensitive factor. J Neurosci 2010; 30:13955-65. [PMID: 20962217 DOI: 10.1523/jneurosci.0270-10.2010] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Disturbances in GABA(A) receptor trafficking contribute to several neurological and psychiatric disorders by altering inhibitory neurotransmission. Identifying mechanisms that regulate GABA(A) receptor trafficking could lead to better understanding of disease pathogenesis and treatment. Here, we show that protein kinase Cε (PKCε) regulates the N-ethylmaleimide-sensitive factor (NSF), an ATPase critical for membrane fusion events, and thereby promotes the trafficking of GABA(A) receptors. Activation of PKCε decreased cell surface expression of GABA(A) receptors and attenuated GABA(A) currents. Activated PKCε associated with NSF, phosphorylated NSF at serine 460 and threonine 461, and increased NSF ATPase activity, which was required for GABA(A) receptor downregulation. These findings identify new roles for NSF and PKCε in regulating synaptic inhibition through downregulation of GABA(A) receptors. Reducing NSF activity by inhibiting PKCε could help restore synaptic inhibition in disease states in which it is impaired.
Collapse
|
36
|
Knipper M, Zimmermann U, Müller M. Molecular aspects of tinnitus. Hear Res 2010; 266:60-9. [DOI: 10.1016/j.heares.2009.07.013] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2009] [Revised: 07/28/2009] [Accepted: 07/28/2009] [Indexed: 01/18/2023]
|
37
|
Mature BDNF, but not proBDNF, reduces excitability of fast-spiking interneurons in mouse dentate gyrus. J Neurosci 2009; 29:12412-8. [PMID: 19812317 DOI: 10.1523/jneurosci.2978-09.2009] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Mature BDNF and its precursor proBDNF may both be secreted to exert opposite effects on synaptic plasticity in the hippocampus. However, it is unknown how proBDNF and mature BDNF affect the excitability of GABAergic interneurons and thereby regulate GABAergic inhibition. We made recordings of GABAergic spontaneous IPSCs (sIPSCs) in mouse dentate gyrus granule cells and found that chronic or acute BDNF reductions led to large increases in the sIPSC frequencies, which were TTX (tetrodotoxin) sensitive and therefore action-potential driven. Conversely, addition of mature BDNF, but not proBDNF, within minutes led to a decrease in the sIPSC frequency to 44%. Direct recordings from fast-spiking GABAergic interneurons revealed that mature BDNF reduced their excitability and depressed their action potential firing, whereas proBDNF had no effect. Using the TrkB inhibitor K-252a, or mice deficient for the common neurotrophin receptor p75(NTR), the regulation of GABAergic activity was shown specifically to be mediated by BDNF binding to the neurotrophin receptor TrkB. In agreement, immunohistochemistry demonstrated that TrkB, but not p75(NTR), was expressed in parvalbumin-positive interneurons. Our results suggest that mature BDNF decreases the excitability of GABAergic interneurons via activation of TrkB, while proBDNF does not impact on GABAergic activity. Thus, by affecting the firing of GABAergic interneurons, mature BDNF may play an important role in regulating network oscillations in the hippocampus.
Collapse
|
38
|
BDNF signaling in the formation, maturation and plasticity of glutamatergic and GABAergic synapses. Exp Brain Res 2009; 199:203-34. [PMID: 19777221 DOI: 10.1007/s00221-009-1994-z] [Citation(s) in RCA: 228] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2009] [Accepted: 08/12/2009] [Indexed: 01/17/2023]
Abstract
In the past 15 years numerous reports provided strong evidence that brain-derived neurotrophic factor (BDNF) is one of the most important modulators of glutamatergic and GABAergic synapses. Remarkable progress regarding localization, kinetics, and molecular mechanisms of BDNF secretion has been achieved, and a large number of studies provided evidence that continuous extracellular supply of BDNF is important for the proper formation and functional maturation of glutamatergic and GABAergic synapses. BDNF can play a permissive role in shaping synaptic networks, making them more susceptible for the occurrence of plastic changes. In addition, BDNF appears to be also an instructive factor for activity-dependent long-term synaptic plasticity. BDNF release just in response to synaptic stimulation might be a molecular trigger to convert high-frequency synaptic activity into long-term synaptic memories. This review attempts to summarize the current knowledge in synaptic secretion and synaptic action of BDNF, including both permissive and instructive effects of BDNF in synaptic plasticity.
Collapse
|
39
|
Tretter V, Moss SJ. GABA(A) Receptor Dynamics and Constructing GABAergic Synapses. Front Mol Neurosci 2008; 1:7. [PMID: 18946540 PMCID: PMC2526003 DOI: 10.3389/neuro.02.007.2008] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2008] [Accepted: 05/16/2008] [Indexed: 12/04/2022] Open
Abstract
GABAA receptors are located on the majority of neurons in the central and peripheral nervous system, where they mediate important actions of the neurotransmitter gamma-aminobutyric acid. Early in development the trophic properties of GABA allow a healthy development of the nervous system. Most neurons have a high intracellular Cl-concentration early in life due to the late functional expression of the Cl-pump KCC2, therefore GABA has excitatory effects at this stage. Upon higher expression and activation of KCC2 GABA takes on its inhibitory effects while glutamate functions as the major excitatory neurotransmitter. Like all multisubunit membrane proteins the GABAA receptor is assembled in the ER and travels through the Golgi and remaining secretory pathway to the cell surface, where it mediates GABA actions either directly at the synapses or at extrasynaptic sites responding to ambient GABA to provide a basal tonic inhibitory state. In order to adapt to changing needs and information states, the GABAergic system is highly dynamic. That includes subtype specific trafficking to different locations in the cell, regulation of mobility by interaction with scaffold molecules, posttranslational modifications, that either directly affect channel function or the interaction with other proteins and finally the dynamic exchange between surface and intracellular receptor pools, that either prepare receptors for recycling to the surface or degradation. Here we give an overview of the current understanding of GABAA receptor functional and molecular dynamics that play a major part in maintaining the balance between excitation and inhibition and in changes in network activity.
Collapse
Affiliation(s)
- Verena Tretter
- Department of Neuroscience, University of Pennsylvania Philadelphia, PA, USA
| | | |
Collapse
|
40
|
Kron M, Zhang W, Dutschmann M. Developmental changes in the BDNF-induced modulation of inhibitory synaptic transmission in the Kölliker-Fuse nucleus of rat. Eur J Neurosci 2007; 26:3449-57. [PMID: 18052976 DOI: 10.1111/j.1460-9568.2007.05960.x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
The Kölliker-Fuse nucleus (KF), part of the pontine respiratory group, is involved in the control of respiratory phase duration, and receives both excitatory and inhibitory afferent input from various other brain regions. There is evidence for developmental changes in the modulation of excitatory inputs to the KF by the neurotrophin brain-derived neurotrophic factor (BDNF). In the present study we investigated if BDNF exerts developmental effects on inhibitory synaptic transmission in the KF. Recordings of inhibitory postsynaptic currents (IPSCs) in KF neurons in a pontine slice preparation revealed general developmental changes. Recording of spontaneous and evoked IPSCs (sIPSCs, eIPSCS) revealed that neonatally the gamma-aminobutyric acid (GABA)ergic fraction of IPSCs was predominant, while in later developmental stages glycinergic neurotransmission significantly increased. Bath-application of BDNF significantly reduced sIPSC frequency in all developmental stages, while BDNF-mediated modulation on eIPSCs showed developmental differences. The eIPSCs mean amplitude was uniformly and significantly reduced following BDNF application only in neurons from rats younger than postnatal day 10. At later postnatal stages the response pattern became heterogeneous, and both augmentations and reductions of eIPSC amplitudes occurred. All BDNF effects on eIPSCs and sIPSCs were reversed with the tyrosine kinase receptor-B inhibitor K252a. We conclude that developmental changes in inhibitory neurotransmission, including the BDNF-mediated modulation of eIPSCs, relate to the postnatal maturation of the KF. The changes in BDNF-mediated modulation of IPSCs in the KF may have strong implications for developmental changes in synaptic plasticity and the adaptation of the breathing pattern to afferent inputs.
Collapse
Affiliation(s)
- Miriam Kron
- Department of Neuro and Sensory Physiology, University Medicine Göttingen, Georg August University, Humboldtallee 23, 37073 Göttingen, Germany
| | | | | |
Collapse
|
41
|
Dergacheva O, Griffioen KJS, Wang X, Kamendi H, Gorini C, Mendelowitz D. 5-HT(2) receptor subtypes mediate different long-term changes in GABAergic activity to parasympathetic cardiac vagal neurons in the nucleus ambiguus. Neuroscience 2007; 149:696-705. [PMID: 17869437 PMCID: PMC2098105 DOI: 10.1016/j.neuroscience.2007.08.015] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2007] [Revised: 08/07/2007] [Accepted: 08/13/2007] [Indexed: 01/24/2023]
Abstract
Serotonin (5-HT), and in particular 5-HT(2) receptors, play an important role in cardiorespiratory function within the brainstem. In addition, abnormalities in the 5-HT system have been implicated in many cardiorespiratory disorders, including sudden infant death syndrome. However, little is known about the mechanisms of action of 5-HT(2) receptors in altering the activity of parasympathetic cardiac neurons in the brainstem. In this study we examined the effects of activation of different subtypes of 5-HT(2) receptors on spontaneous and respiratory-evoked GABAergic neurotransmission to cardioinhibitory vagal neurons within the nucleus ambiguus as well as rhythmic fictive inspiratory-related activity in rats. A single application of alpha-Me-5-hydroxytryptamine maleate (alpha-Me-5-HT), a 5-HT(2) receptor agonist, did not significantly alter the frequency of spontaneous or respiratory-evoked GABAergic inhibitory postsynaptic currents (IPSCs) in cardiac vagal neurons. However, repetitive successive applications of alpha-Me-5-HT elicited a long-lasting (>/=1 h) decrease in the frequency of spontaneous as well as inspiratory-related GABAergic IPSCs to cardiac vagal neurons. This study demonstrates multiple, but not single applications of the 5-HT(2) receptor agonist alpha-Me-5-HT caused a long-lasting inhibition of both spontaneous and fictive inspiratory-related GABAergic neurotransmission to CVNs, which can be prevented by the 5-HT(2B) receptor antagonist SB204741, but persisted with the 5-HT(2A/2C) receptor antagonist ketanserin. The 5-HT(2) receptor agonist alpha-Me-5-HT also reversibly and transiently excited central fictive inspiratory activity, which was abolished by ketanserin, but was unaffected by the 5-HT(2B) receptor antagonist SB204741.
Collapse
Affiliation(s)
- O Dergacheva
- Department of Pharmacology and Physiology, George Washington University, Washington, DC 20037, USA
| | | | | | | | | | | |
Collapse
|
42
|
Bardoni R, Ghirri A, Salio C, Prandini M, Merighi A. BDNF-mediated modulation of GABA and glycine release in dorsal horn lamina II from postnatal rats. Dev Neurobiol 2007; 67:960-75. [PMID: 17506495 DOI: 10.1002/dneu.20401] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Recent studies show that excitatory glutamatergic transmission is potentiated by BDNF in superficial dorsal horn, both at the pre- and the postsynaptic site. The role of BDNF in modulating GABA and glycine-mediated inhibitory transmission has not been fully investigated. To determine whether the neurotrophin is effective in regulating the spontaneous release of the two neurotransmitters, we have recorded miniature inhibitory postsynaptic currents (mIPSCs) in lamina II of post-natal rats. We show that application of BDNF enhanced the spontaneous release of GABA and glycine, in presence of tetrodotoxin. The effect was blocked by the trk-receptor inhibitor k-252a. Amplitude and kinetics of mIPSCs were not altered. Evoked GABA and glycine IPSCs (eIPSCs) were depressed by BDNF and the coefficient of variation of eIPSC amplitude was significantly increased. By recording glycine eIPSCs with the paired-pulse protocol, an increase of paired-pulse ratio during BDNF application was observed. We performed parallel ultrastructural studies to unveil the circuitry involved in the effects of BDNF. These studies show that synaptic interactions between full length functional trkB receptors and GABA-containing profiles only occur at non peptidergic synaptic glomeruli of types I and II. Expression of trkB in presynaptic vesicle-containing dendrites originating from GABAergic islet cells, indicates these profiles as key structures in the modulation of inhibitory neurotransmission by the neurotrophin. Our results thus describe a yet uncharacterized effect of BDNF in lamina II, giving further strength to the notion that the neurotrophin plays an important role in pain neurotransmission.
Collapse
Affiliation(s)
- Rita Bardoni
- Department of Biomedical Sciences, University of Modena and Reggio Emilia, Modena, Italy.
| | | | | | | | | |
Collapse
|
43
|
Wanaverbecq N, Semyanov A, Pavlov I, Walker MC, Kullmann DM. Cholinergic axons modulate GABAergic signaling among hippocampal interneurons via postsynaptic alpha 7 nicotinic receptors. J Neurosci 2007; 27:5683-93. [PMID: 17522313 PMCID: PMC2889598 DOI: 10.1523/jneurosci.1732-07.2007] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Homopentameric alpha7 nicotinic receptors have a high affinity for acetylcholine (ACh), are permeable to Ca2+ ions, and are abundant in hippocampal interneurons. Although nicotinic agonists evoke inward currents and Ca2+ transients in stratum radiatum interneurons, the role of endogenous ACh in modulating synaptic integration by interneurons is incompletely understood. Many cholinergic axonal varicosities do not have postsynaptic specializations, but alpha7 receptors frequently occur close to synaptic GABA(A) receptors. These observations raise the possibility that alpha7 nicotinic receptors activated by ACh released from cholinergic axons modulate GABAergic transmission in interneurons. We show that agonists of alpha7 receptors profoundly depress GABAergic IPSCs recorded in stratum radiatum interneurons in the CA1 region of the hippocampus. This depression is accompanied by a small increase in GABA release. Alpha7 nicotinic receptor agonists also depress GABA- or muscimol-evoked currents in interneurons, indicating that the major effect is a postsynaptic modulation of GABA(A) receptors. The depression of GABA-evoked currents is abolished by chelating Ca2+ in the recorded interneuron and attenuated by inhibitors of PKC. We also show that stimuli designed to release endogenous ACh from cholinergic axons evoke an alpha7 receptor-dependent heterosynaptic depression of GABAergic IPSCs in interneurons. This heterosynaptic modulation is amplified by blocking cholinesterases. These results reveal a novel mechanism by which cholinergic neurons modulate information processing in the hippocampus.
Collapse
Affiliation(s)
- Nicolas Wanaverbecq
- Institute of Neurology, University College London, London WC1N 3BG, United Kingdom, and
- Biozentrum, University of Basel, CH-4056 Basel, Switzerland
| | - Alexey Semyanov
- Institute of Neurology, University College London, London WC1N 3BG, United Kingdom, and
| | - Ivan Pavlov
- Institute of Neurology, University College London, London WC1N 3BG, United Kingdom, and
| | - Matthew C. Walker
- Institute of Neurology, University College London, London WC1N 3BG, United Kingdom, and
| | - Dimitri M. Kullmann
- Institute of Neurology, University College London, London WC1N 3BG, United Kingdom, and
| |
Collapse
|
44
|
Kron M, Mörschel M, Reuter J, Zhang W, Dutschmann M. Developmental changes in brain-derived neurotrophic factor-mediated modulations of synaptic activities in the pontine Kölliker-Fuse nucleus of the rat. J Physiol 2007; 583:315-27. [PMID: 17569735 PMCID: PMC2277243 DOI: 10.1113/jphysiol.2007.134726] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The Kölliker-Fuse nucleus (KF), part of the respiratory network, is involved in the modulation of respiratory phase durations in response to peripheral and central afferent inputs. The KF is immature at birth. Developmental changes in its physiological and anatomical properties have yet to be investigated. Since brain-derived neurotrophic factor (BDNF) is of major importance for the maturation of neuronal networks, we investigated its effects on developmental changes in the KF on different postnatal days (neonatal, P1-5; intermediate, P6-13; juvenile, P14-21) by analysing single neurones in the in vitro slice preparation and network activities in the perfused brainstem preparation in situ. The BDNF had only weak effects on the frequency of mixed excitatory and inhibitory spontaneous postsynaptic currents (sPSCs) in neonatal slice preparations. Postnatally, in the intermediate and juvenile age groups, a significant augmentation of the sPSC frequency was observed in the presence of 100 pm BDNF (+23.5+/-12.6 and +76.7+/-28.4%, respectively). Subsequent analyses of BDNF effects on evoked excitatory postsynaptic currents (eEPSCs) revealed significant enhancement of eEPSC amplitude of +20.8+/-7.0% only in juvenile stages (intermediates, -13.2+/-4.8%). On the network level, significant modulation of phrenic nerve activity following BDNF microinjection into the KF was also observed only in juveniles. The data suggest that KF neurones are subject to BDNF-mediated fast synaptic modulation after completion of postnatal maturation. After maturation, BDNF contributes to modulation of fast excitatory neurotransmission in respiratory-related KF neurones. This may be important for network plasticity associated with the processing of afferent information.
Collapse
Affiliation(s)
- Miriam Kron
- Department of Neuro and Sensory Physiology, University Medical Center Göttingen, Georg-August-University, Humboldtallee 23, 37073 Göttingen, Germany
| | | | | | | | | |
Collapse
|
45
|
Abstract
Neuronal inhibition is of paramount importance in maintaining the delicate and dynamic balance between excitatory and inhibitory influences in the central nervous system. GABA (gamma-aminobutyric acid), the primary inhibitory neurotransmitter in brain, exerts its fast inhibitory effects through ubiquitously expressed GABA(A) receptors. Activation of these heteropentameric receptors by GABA results in the gating of an integral chloride channel leading to membrane hyperpolarization and neuronal inhibition. To participate in neurotransmission, the receptor must reside on the cell surface. The trafficking of nascent receptors to the cell surface involves posttranslational modification and the interaction of the receptor with proteins that reside within the secretory pathway. The subsequent insertion of the receptor into specialized regions of the plasma membrane is dictated by receptor composition and other factors that guide insertion at synaptic or perisynaptic/extrasynaptic sites, where phasic and tonic inhibition are mediated, respectively. Once at the cell surface, the receptor is laterally mobile and subject to both constitutive and regulated endocytosis. Following endocytosis the receptor undergoes either recycling to the plasma membrane or degradation. These dynamic processes profoundly affect the strength of GABAergic signaling, neuronal inhibition, and presumably synaptic plasticity. Heritable channelopathies that affect receptor trafficking have been recently recognized and compelling evidence exists that mechanisms underlying acquired epilepsy involve GABA(A) receptor internalization. Additionally, GABA(A) receptor endocytosis has been identified as an early event in the ischemic response that leads to excitotoxicity and cell death. This chapter summarizes what is known regarding the regulation of receptor trafficking and cell surface expression and its impact on nervous system function from both cell biology and disease perspectives.
Collapse
Affiliation(s)
- Nancy J Leidenheimer
- Department of Biochemistry and Molecular Biology, Louisiana State University, Health Sciences Center, Shreveport, LA 71130, USA.
| |
Collapse
|
46
|
Hess DM, Scott MO, Potluri S, Pitts EV, Cisterni C, Balice-Gordon RJ. Localization of TrkC to Schwann cells and effects of neurotrophin-3 signaling at neuromuscular synapses. J Comp Neurol 2007; 501:465-82. [PMID: 17278135 DOI: 10.1002/cne.21163] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Neurotrophins and their receptors, the Trks, are differentially expressed among the cell types that make up neuromuscular and other synapses, but the function and directionality of neurotrophin signaling at synapses are poorly understood. Here we demonstrate, via immunostaining, Western blotting, and RT-PCR analyses, that TrkC, the receptor for neurotrophin-3 (NT3), is expressed by mouse perisynaptic and myelinating Schwann cells from birth through adulthood and is unaltered after denervation. Analyses of transgenic mice in which the NT3 coding sequence is replaced by lacZ showed that NT3 is expressed in motor neurons and Schwann cells during perinatal development, but not in adult mice. In muscle, NT3 is expressed by intrafusal muscle fibers within spindles, as has been previously reported. Surprisingly, NT3 is also expressed in extrafusal muscle fibers during perinatal life and in adults. Genetic approaches were used to explore the roles of NT3 and TrkC signaling at neuromuscular synapses. Overexpression of NT3 in muscle fibers during development resulted in an increased number of perisynaptic Schwann cells at neuromuscular synapses, without altering synaptic size, suggesting that muscle-derived NT3 might act as a mitogen or trophic factor for Schwann cells. Conditional deletion of NT3 from motor neurons did not alter the number of Schwann cells or other aspects of neuromuscular synaptic structure, suggesting that motor-neuron-derived NT3 is not required for normal development of perisynaptic Schwann cells or synapses. Together, these results demonstrate that NT3 expression is developmentally regulated in skeletal muscle and may modulate the number of Schwann cells at neuromuscular synapses.
Collapse
Affiliation(s)
- Darren M Hess
- Department of Neuroscience, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania 19104-6074, USA
| | | | | | | | | | | |
Collapse
|
47
|
Carrasco MA, Castro P, Sepulveda FJ, Tapia JC, Gatica K, Davis MI, Aguayo LG. Regulation of glycinergic and GABAergic synaptogenesis by brain-derived neurotrophic factor in developing spinal neurons. Neuroscience 2007; 145:484-94. [PMID: 17306467 DOI: 10.1016/j.neuroscience.2006.12.019] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2006] [Revised: 11/15/2006] [Accepted: 12/05/2006] [Indexed: 11/26/2022]
Abstract
Brain-derived neurotrophic factor (BDNF) effects on the establishment of glycinergic and GABAergic transmissions in mouse spinal neurons were examined using combined electrophysiological and calcium imaging techniques. BDNF (10 ng/ml) caused a significant acceleration in the onset of synaptogenesis without large effects on the survival of these neurons. Amplitude and frequency of spontaneous inhibitory postsynaptic currents (sIPSCs) and miniature inhibitory postsynaptic currents (mIPSCs) associated to activation of glycine and GABA(A) receptors were augmented in neurons cultured with BDNF. The neurotrophin effect was blocked by long term tetrodotoxin (TTX) addition suggesting a dependence on neuronal activity. In addition, BDNF caused a significant increase in glycine- and GABA-evoked current densities that partly explains the increase in synaptic transmission. Presynaptic mechanisms were also involved in BDNF effects since triethylammonium(propyl)-4-(2-(4-dibutylamino-phenyl)vinyl)pyridinium (FM1-43) destaining with high K(+) was augmented in neurons incubated with the neurotrophin. The effects of BDNF were mediated by receptor tyrosine kinase B (TrkB) and mitogen-activated protein kinase kinase (MEK) activation since culturing neurons with either (9S,10R,12R)-2,3,9,10,11,12-hexahydro-10-hydroxy-9-methyl-1-oxo-9,12-epoxy-1H-diindolo[1,2,3-fg:3',2',1'- kl]pyrrolo[3,4-i][1,6]benzodiazocine-10-carboxylic acid methyl ester (K252a) or 2-(2-amino-3-methoxyphenyl)-4H-1-benzopyran-4-one (PD98059) blocked the augmentation in synaptic activity induced by the neurotrophin.
Collapse
Affiliation(s)
- M A Carrasco
- Laboratory of Neurophysiology, Department of Physiology, University of Concepción, P.O. Box 160-C, Concepción, Chile
| | | | | | | | | | | | | |
Collapse
|
48
|
Kanematsu T, Mizokami A, Watanabe K, Hirata M. Regulation of GABAA-Receptor Surface Expression With Special Reference to the Involvement of GABARAP (GABAA Receptor-Associated Protein) and PRIP (Phospholipase C-Related, but Catalytically Inactive Protein). J Pharmacol Sci 2007; 104:285-92. [PMID: 17690529 DOI: 10.1254/jphs.cp0070063] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
Abstract
GABA(A) receptors are heteropentameric ligand-gated chloride channels composed of a variety of subunits, including alpha1 - 6, beta1 - 3, gamma1 - 3, delta, epsilon, theta, and pi, and play a key role in controlling inhibitory neuronal activity. Modification of the efficacy of the synaptic strength is produced by changes in both the number of neuronal surface receptors and pentameric molecular assembly, leading to differences of sensitivity to neurotransmitters and neuromimetic drugs. Therefore, it is important to understand the molecular mechanisms regulating the so-called "life cycle of GABA(A) receptors" including sequential pentameric assembly at the site synthesized, intracellular transport through the Golgi apparatus and the cytoplasm, insertion into the cell membrane, functional modulation at the cell surface, and finally internalization, followed by either recycling back to the surface membrane or lysosomal degradation. This review is focused on events related to the surface expression of the receptor containing the gamma2 subunit and clathrin/AP2 complex-mediated phospho-regulated endocytosis of the receptor, with special reference to the function of novel GABA(A) receptor modulators, GABARAP (GABA(A) receptor-associated protein) and PRIP (phospholipase C-related, but catalytically inactive protein).
Collapse
Affiliation(s)
- Takashi Kanematsu
- Laboratory of Molecular and Cellular Biochemistry, Faculty of Dental Science and Station for Collaborative Research, Kyushu University, Fukuoka, Japan.
| | | | | | | |
Collapse
|
49
|
Kanematsu T, Yasunaga A, Mizoguchi Y, Kuratani A, Kittler JT, Jovanovic JN, Takenaka K, Nakayama KI, Fukami K, Takenawa T, Moss SJ, Nabekura J, Hirata M. Modulation of GABAA Receptor Phosphorylation and Membrane Trafficking by Phospholipase C-related Inactive Protein/Protein Phosphatase 1 and 2A Signaling Complex Underlying Brain-derived Neurotrophic Factor-dependent Regulation of GABAergic Inhibition. J Biol Chem 2006; 281:22180-22189. [PMID: 16754670 DOI: 10.1074/jbc.m603118200] [Citation(s) in RCA: 76] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Brain-derived neurotrophic factor (BDNF) modulates several distinct aspects of synaptic transmission, including GABAergic transmission. Exposure to BDNF alters properties of GABA(A) receptors and induces changes in the expression level at the cell surface. Although phospholipase C-related inactive protein-1 (PRIP-1) plays an important role in GABA(A) receptor trafficking and function, its role in BDNF-dependent modulation of these receptors, together with the role of PRIP-2, was investigated using neurons cultured from PRIP double knock-out mice. The BDNF-dependent inhibition of whole cell GABA-evoked currents observed in wild type neurons was not detected in neurons cultured from knock-out mice. Instead, a gradual increase in GABA-evoked currents in these neurons correlated with a gradual increase in phosphorylation of GABA(A) receptor beta3 subunit in response to BDNF. To characterize the specific role(s) that PRIP plays as components of underlying molecular machinery, we examined the recruitment of protein phosphatase(s) to GABA(A) receptors. We demonstrate that PRIP associates with phosphatases as well as with beta subunits. PRIP was found to colocalize with GABA(A) receptor clusters in cultured neurons and with recombinant GABA(A) receptors when co-expressed in HEK293 cells. Importantly, a peptide mimicking a domain of PRIP involved in binding to beta subunits disrupted the co-localization of these proteins in HEK293 cells and potently inhibited the BDNF-mediated attenuation of GABA(A) receptor currents in wild type neurons. Together, the results suggest that PRIP plays an important role in BDNF-dependent regulation of GABA(A) receptors by mediating the specific association between beta subunits of these receptors with protein phosphatases.
Collapse
Affiliation(s)
- Takashi Kanematsu
- Laboratory of Molecular and Cellular Biochemistry, Faculty of Dental Science and Station for Collaborative Research, Kyushu University, Fukuoka 812-8582, Japan
| | - Atsushi Yasunaga
- Laboratory of Molecular and Cellular Biochemistry, Faculty of Dental Science and Station for Collaborative Research, Kyushu University, Fukuoka 812-8582, Japan
| | - Yoshito Mizoguchi
- Laboratory of Molecular and Cellular Biochemistry, Faculty of Dental Science and Station for Collaborative Research, Kyushu University, Fukuoka 812-8582, Japan; Department of Developmental Physiology, National Institute for Physiological Science, Okazaki 444-8585, Japan
| | - Akiko Kuratani
- Laboratory of Molecular and Cellular Biochemistry, Faculty of Dental Science and Station for Collaborative Research, Kyushu University, Fukuoka 812-8582, Japan
| | - Josef T Kittler
- Department of Physiology, University College London, London WC1E 6BT, United Kingdom
| | - Jasmina N Jovanovic
- Department of Pharmacology, School of Pharmacy, Brunswick Square, London, WC1N 1AX, United Kingdom
| | - Kei Takenaka
- Department of Biochemistry, Institute of Medical Science, University of Tokyo, Tokyo 108-8639, Japan
| | - Keiichi I Nakayama
- Department of Molecular and Cellular Biology, Medical Institute of Bioregulation, Kyushu University, Fukuoka 812-8582, Japan
| | - Kiyoko Fukami
- Laboratory of Genome and Biosignal, Tokyo University of Pharmacy and Life Science, Tokyo 192-0392, Japan
| | - Tadaomi Takenawa
- Department of Biochemistry, Institute of Medical Science, University of Tokyo, Tokyo 108-8639, Japan
| | - Stephen J Moss
- Department of Neuroscience, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania 19104
| | - Junichi Nabekura
- Department of Developmental Physiology, National Institute for Physiological Science, Okazaki 444-8585, Japan
| | - Masato Hirata
- Laboratory of Molecular and Cellular Biochemistry, Faculty of Dental Science and Station for Collaborative Research, Kyushu University, Fukuoka 812-8582, Japan.
| |
Collapse
|
50
|
Yamuy J, Ramos O, Torterolo P, Sampogna S, Chase MH. The role of tropomyosin-related kinase receptors in neurotrophin-induced rapid eye movement sleep in the cat. Neuroscience 2006; 135:357-69. [PMID: 16125858 DOI: 10.1016/j.neuroscience.2005.05.068] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2005] [Revised: 05/12/2005] [Accepted: 05/29/2005] [Indexed: 11/30/2022]
Abstract
The microinjection of nerve growth factor and neurotrophin-3 into the rostro-dorsal pontine tegmentum of the cat evokes a state that is comparable to naturally-occurring rapid eye movement sleep. Using two experimental paradigms, we tested the hypothesis that neurotrophin high-affinity receptors (trkA and trkC, tropomyosin-related kinase A and C, respectively) mediate this effect. First, trk and fos immunohistochemistry were combined to determine whether tyrosine kinase receptor-containing neurons in the dorsal pontine tegmentum are active in cats that exhibit long-lasting periods of rapid eye movement sleep following the local microinjection of nerve growth factor. During approximately two hours of recording, nerve growth factor-treated cats spent 59.8% of the time in a rapid eye movement sleep-like state; vehicle-injected (control) animals remained in quiet wakefulness and non-rapid eye movement sleep. Whereas control and nerve growth factor-treated cats exhibited a similar mean number of trkA- and trkC-immunoreactive neurons in the dorsal pontine tegmentum, the number of trkA- and trkC-immunoreactive neurons that expressed Fos, i.e. double-labeled cells that are presumably activated, was significantly larger in cats that were injected with nerve growth factor. Axon terminals contained tyrosine kinase receptor immunoreactivity in this region; many were apposed to Fos-immunoreactive neurons. In addition, patterns of tyrosine kinase receptor and Fos immunoreactivity similar to those observed in nerve growth factor-injected cats were present, in conjunction with long-lasting rapid eye movement sleep, following the microinjection of carbachol into the dorsal pons. In a second series of studies, nerve growth factor or neurotrophin-3 was injected alone or after K-252a, a blocker of tyrosine kinase receptors, into the rostro-dorsal pontine tegmentum. Nerve growth factor or neurotrophin-3 alone produced, with a mean latency of 4 min, a rapid eye movement sleep-like state. However, neurotrophin injections preceded by K-252a were not effective in inducing rapid eye movement sleep. These results indicate that the activation of trkA and trkC receptors in neurons in the pontine tegmentum is responsible, at least in part, for the rapid eye movement sleep-inducing effect of nerve growth factor and neurotrophin-3. Furthermore, the data suggest that these neurotrophins are capable of acting both pre- and postsynaptically to activate pontine neurons that are involved in the generation of rapid eye movement sleep.
Collapse
Affiliation(s)
- J Yamuy
- Department of Physiology, UCLA School of Medicine, Los Angeles, CA 90095, USA.
| | | | | | | | | |
Collapse
|