1
|
Ishikawa H, Kimura S, Takase H, Borlongan M, Fukuda N, Hoshino T, Hamanaka G, Park JH, Shindo A, Kim KW, Gelman IH, Lok J, Lo EH, Arai K. Sex differences in the role of AKAP12 in behavioral function of middle-aged mice. Biol Sex Differ 2024; 15:93. [PMID: 39574214 PMCID: PMC11580627 DOI: 10.1186/s13293-024-00670-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Accepted: 11/04/2024] [Indexed: 11/25/2024] Open
Abstract
A-kinase anchoring protein 12 (AKAP12) is a key scaffolding protein that regulates cellular signaling by anchoring protein kinase A (PKA) and other signaling molecules. While recent studies suggest an important role for AKAP12 in the brain, including cognitive functions, its role in middle-aged mice and potential sex differences are not fully understood. Therefore, this study investigated the effects of AKAP12 on cognitive and exploratory behavior in middle-aged mice, focusing on sex differences. Cognitive function was assessed using the spontaneous Y-maze test and the novel object recognition test (NORT). No significant sex differences in cognitive function were found in middle-aged C57BL/6J mice; however, female mice showed greater exploratory behavior during the NORT. In addition, both middle-aged male and female Akap12 knockout (KO) mice performed similarly to wild-type (WT) mice in the Y-maze test, but had lower discrimination indices in the NORT, suggesting a potential role for AKAP12 in short-term memory. Notably, exploratory behavior was suppressed in female Akap12 KO mice compared to WT mice, whereas male Akap12 KO mice did not show this effect. There were no significant differences in movement distance and velocity during the Y-maze test and NORT between WT and KO mice of either sex. These results indicate that AKAP12 affects cognitive function and exploratory behavior in middle-aged mice and that these effects differ between sexes.
Collapse
Affiliation(s)
- Hidehiro Ishikawa
- Neuroprotection Research Laboratories, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, 149 Thirteenth Street, Room 2401, Charlestown, MA, 02129-2000, USA
- Department of Neurology, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu, Mie, Japan
| | - Shintaro Kimura
- Neuroprotection Research Laboratories, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, 149 Thirteenth Street, Room 2401, Charlestown, MA, 02129-2000, USA
- Life Science Research Center, Gifu University, Gifu, Japan
| | - Hajime Takase
- Neuroprotection Research Laboratories, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, 149 Thirteenth Street, Room 2401, Charlestown, MA, 02129-2000, USA
- YCU Center for Novel and Exploratory Clinical Trials (Y-NEXT), Yokohama City University Hospital, Yokohama, Japan
| | - Maximillian Borlongan
- Neuroprotection Research Laboratories, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, 149 Thirteenth Street, Room 2401, Charlestown, MA, 02129-2000, USA
| | - Norito Fukuda
- Neuroprotection Research Laboratories, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, 149 Thirteenth Street, Room 2401, Charlestown, MA, 02129-2000, USA
| | - Tomonori Hoshino
- Neuroprotection Research Laboratories, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, 149 Thirteenth Street, Room 2401, Charlestown, MA, 02129-2000, USA
| | - Gen Hamanaka
- Neuroprotection Research Laboratories, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, 149 Thirteenth Street, Room 2401, Charlestown, MA, 02129-2000, USA
| | - Ji Hyun Park
- Neuroprotection Research Laboratories, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, 149 Thirteenth Street, Room 2401, Charlestown, MA, 02129-2000, USA
| | - Akihiro Shindo
- Department of Neurology, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu, Mie, Japan
| | - Kyu-Won Kim
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, 08826, Republic of Korea
| | - Irwin H Gelman
- Department of Cancer Genetics and Genomics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Josephine Lok
- Neuroprotection Research Laboratories, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, 149 Thirteenth Street, Room 2401, Charlestown, MA, 02129-2000, USA
- Pediatric Critical Care Medicine, Department of Pediatrics, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Eng H Lo
- Neuroprotection Research Laboratories, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, 149 Thirteenth Street, Room 2401, Charlestown, MA, 02129-2000, USA
| | - Ken Arai
- Neuroprotection Research Laboratories, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, 149 Thirteenth Street, Room 2401, Charlestown, MA, 02129-2000, USA.
| |
Collapse
|
2
|
Rosenthal KJ, Gordan JD, Scott JD. Protein kinase A and local signaling in cancer. Biochem J 2024; 481:1659-1677. [PMID: 39540434 PMCID: PMC11975432 DOI: 10.1042/bcj20230352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 10/22/2024] [Accepted: 11/04/2024] [Indexed: 11/16/2024]
Abstract
Protein kinase A (PKA) is a basophilic kinase implicated in the modulation of many cell-signaling and physiological processes. PKA also contributes to cancer-relevant events such as growth factor action, cell cycle control, cell migration and tumor metabolism. Germline and somatic mutations in PKA, gene amplifications, and chromosome rearrangements that encode kinase fusions, are linked to a growing number of malignant neoplasms. Mislocalization of PKA by exclusion from A-Kinase Anchoring Protein (AKAP) signaling islands further underlies cancer progression. This article highlights the influence of AKAP signaling and local kinase action in selected hallmarks of cancer. We also feature the utility of kinase inhibitor drugs as frontline and future anti-cancer therapies.
Collapse
Affiliation(s)
- Kacey J. Rosenthal
- Department of Pharmacology, University of Washington School of Medicine, 1959 NE Pacific St., Box 357750, Seattle, WA 98195, U.S.A
| | - John D. Gordan
- Department of Medicine (Hematology/Oncology), Quantitative Biosciences Institute, UCSF Helen Diller Family Cancer Center, 1700 4th St., San Francisco, CA 94143, U.S.A
| | - John D. Scott
- Department of Pharmacology, University of Washington School of Medicine, 1959 NE Pacific St., Box 357750, Seattle, WA 98195, U.S.A
| |
Collapse
|
3
|
Acuña-Catalán D, Shah S, Wehrfritz C, Nomura M, Acevedo A, Olmos C, Quiroz G, Huerta H, Bons J, Ampuero E, Wyneken U, Sanhueza M, Arancibia F, Contreras D, Cárdenas JC, Morales B, Schilling B, Newman JC, González-Billault C. Ketogenic diet administration later in life improves memory by modifying the synaptic cortical proteome via the PKA signaling pathway in aging mice. Cell Rep Med 2024; 5:101593. [PMID: 38843842 PMCID: PMC11228662 DOI: 10.1016/j.xcrm.2024.101593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 02/26/2024] [Accepted: 05/14/2024] [Indexed: 06/21/2024]
Abstract
Aging compromises brain function leading to cognitive decline. A cyclic ketogenic diet (KD) improves memory in aged mice after long-term administration; however, short-term effects later in life and the molecular mechanisms that govern such changes remain unclear. Here, we explore the impact of a short-term KD treatment starting at elderly stage on brain function of aged mice. Behavioral testing and long-term potentiation (LTP) recordings reveal that KD improves working memory and hippocampal LTP. Furthermore, the synaptosome proteome of aged mice fed a KD long-term evidence changes predominantly at the presynaptic compartment associated to the protein kinase A (PKA) signaling pathway. These findings were corroborated in vivo by western blot analysis, with high BDNF abundance and PKA substrate phosphorylation. Overall, we show that a KD modifies brain function even when it is administered later in life and recapitulates molecular features of long-term administration, including the PKA signaling pathway, thus promoting synaptic plasticity at advanced age.
Collapse
Affiliation(s)
- Diego Acuña-Catalán
- Center for Geroscience, Brain Health, and Metabolism (GERO), Santiago, Chile; Department of Biology, Faculty of Sciences, Universidad de Chile, Santiago, Chile
| | - Samah Shah
- The Buck Institute for Research on Aging, Novato, CA, USA
| | | | | | - Alejandro Acevedo
- Institute of Nutrition and Food Technology (INTA), Universidad de Chile, Santiago, Chile
| | - Cristina Olmos
- Center for Geroscience, Brain Health, and Metabolism (GERO), Santiago, Chile; Department of Biology, Faculty of Sciences, Universidad de Chile, Santiago, Chile
| | - Gabriel Quiroz
- Center for Geroscience, Brain Health, and Metabolism (GERO), Santiago, Chile; Department of Biology, Faculty of Sciences, Universidad de Chile, Santiago, Chile
| | - Hernán Huerta
- Center for Geroscience, Brain Health, and Metabolism (GERO), Santiago, Chile; Center for Integrative Biology, Faculty of Sciences, Universidad Mayor, Santiago, Chile
| | - Joanna Bons
- The Buck Institute for Research on Aging, Novato, CA, USA
| | - Estibaliz Ampuero
- Neurobiology of Behavior Laboratory, Department of Biology, Universidad de Santiago de Chile, Santiago, Chile
| | - Ursula Wyneken
- IMPACT, Center for Interventional Medicine for Precision and Advanced Cellular Therapy, and Faculty of Medicine, Universidad de Los Andes, Santiago, Chile
| | - Magdalena Sanhueza
- Department of Biology, Faculty of Sciences, Universidad de Chile, Santiago, Chile
| | - Felipe Arancibia
- Department of Biology, Faculty of Sciences, Universidad de Chile, Santiago, Chile
| | - Darwin Contreras
- Laboratory of Neuroscience, Faculty of Chemistry and Biology, Universidad de Santiago de Chile, Santiago, Chile
| | - Julio César Cárdenas
- Center for Geroscience, Brain Health, and Metabolism (GERO), Santiago, Chile; Department of Biology, Faculty of Sciences, Universidad de Chile, Santiago, Chile; The Buck Institute for Research on Aging, Novato, CA, USA; Center for Integrative Biology, Faculty of Sciences, Universidad Mayor, Santiago, Chile; Department of Chemistry and Biochemistry and Center for Aging and Longevity Studies University of California, Santa Barbara, CA, USA
| | - Bernardo Morales
- Laboratory of Neuroscience, Faculty of Chemistry and Biology, Universidad de Santiago de Chile, Santiago, Chile
| | | | - John C Newman
- The Buck Institute for Research on Aging, Novato, CA, USA
| | - Christian González-Billault
- Center for Geroscience, Brain Health, and Metabolism (GERO), Santiago, Chile; Department of Biology, Faculty of Sciences, Universidad de Chile, Santiago, Chile; The Buck Institute for Research on Aging, Novato, CA, USA; Institute of Nutrition and Food Technology (INTA), Universidad de Chile, Santiago, Chile; Department of Neurosciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile.
| |
Collapse
|
4
|
He Z, Xie L, Liu J, Wei X, Zhang W, Mei Z. Novel insight into the role of A-kinase anchoring proteins (AKAPs) in ischemic stroke and therapeutic potentials. Biomed Pharmacother 2024; 175:116715. [PMID: 38739993 DOI: 10.1016/j.biopha.2024.116715] [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: 02/25/2024] [Revised: 05/03/2024] [Accepted: 05/06/2024] [Indexed: 05/16/2024] Open
Abstract
Ischemic stroke, a devastating disease associated with high mortality and disability worldwide, has emerged as an urgent public health issue. A-kinase anchoring proteins (AKAPs) are a group of signal-organizing molecules that compartmentalize and anchor a wide range of receptors and effector proteins and have a major role in stabilizing mitochondrial function and promoting neurodevelopmental development in the central nervous system (CNS). Growing evidence suggests that dysregulation of AKAPs expression and activity is closely associated with oxidative stress, ion disorder, mitochondrial dysfunction, and blood-brain barrier (BBB) impairment in ischemic stroke. However, the underlying mechanisms remain inadequately understood. This review provides a comprehensive overview of the composition and structure of A-kinase anchoring protein (AKAP) family members, emphasizing their physiological functions in the CNS. We explored in depth the molecular and cellular mechanisms of AKAP complexes in the pathological progression and risk factors of ischemic stroke, including hypertension, hyperglycemia, lipid metabolism disorders, and atrial fibrillation. Herein, we highlight the potential of AKAP complexes as a pharmacological target against ischemic stroke in the hope of inspiring translational research and innovative clinical approaches.
Collapse
Affiliation(s)
- Ziyu He
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese Medicine and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan 410208, China
| | - Letian Xie
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese Medicine and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan 410208, China
| | - Jiyong Liu
- Hunan Provincial Key Laboratory of Traditional Chinese Medicine Diagnostics, Hunan University of Chinese Medicine, Changsha, Hunan 410208, China
| | - Xuan Wei
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese Medicine and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan 410208, China
| | - Wenli Zhang
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha, Hunan 410208, China.
| | - Zhigang Mei
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese Medicine and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan 410208, China; Third-Grade Pharmacological Laboratory on Chinese Medicine Approved by State Administration of Traditional Chinese Medicine, College of Medicine and Health Sciences, China Three Gorges University, Yichang, Hubei 443002, China.
| |
Collapse
|
5
|
Qasim H, Rajaei M, Xu Y, Reyes-Alcaraz A, Abdelnasser HY, Stewart MD, Lahiri SK, Wehrens XHT, McConnell BK. AKAP12 Upregulation Associates With PDE8A to Accelerate Cardiac Dysfunction. Circ Res 2024; 134:1006-1022. [PMID: 38506047 DOI: 10.1161/circresaha.123.323655] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Accepted: 03/07/2024] [Indexed: 03/21/2024]
Abstract
BACKGROUND In heart failure, signaling downstream the β2-adrenergic receptor is critical. Sympathetic stimulation of β2-adrenergic receptor alters cAMP (cyclic adenosine 3',5'-monophosphate) and triggers PKA (protein kinase A)-dependent phosphorylation of proteins that regulate cardiac function. cAMP levels are regulated in part by PDEs (phosphodiesterases). Several AKAPs (A kinase anchoring proteins) regulate cardiac function and are proposed as targets for precise pharmacology. AKAP12 is expressed in the heart and has been reported to directly bind β2-adrenergic receptor, PKA, and PDE4D. However, its roles in cardiac function are unclear. METHODS cAMP accumulation in real time downstream of the β2-adrenergic receptor was detected for 60 minutes in live cells using the luciferase-based biosensor (GloSensor) in AC16 human-derived cardiomyocyte cell lines overexpressing AKAP12 versus controls. Cardiomyocyte intracellular calcium and contractility were studied in adult primary cardiomyocytes from male and female mice overexpressing cardiac AKAP12 (AKAP12OX) and wild-type littermates post acute treatment with 100-nM isoproterenol (ISO). Systolic cardiac function was assessed in mice after 14 days of subcutaneous ISO administration (60 mg/kg per day). AKAP12 gene and protein expression levels were evaluated in left ventricular samples from patients with end-stage heart failure. RESULTS AKAP12 upregulation significantly reduced total intracellular cAMP levels in AC16 cells through PDE8. Adult primary cardiomyocytes from AKAP12OX mice had significantly reduced contractility and impaired calcium handling in response to ISO, which was reversed in the presence of the selective PDE8 inhibitor (PF-04957325). AKAP12OX mice had deteriorated systolic cardiac function and enlarged left ventricles. Patients with end-stage heart failure had upregulated gene and protein levels of AKAP12. CONCLUSIONS AKAP12 upregulation in cardiac tissue is associated with accelerated cardiac dysfunction through the AKAP12-PDE8 axis.
Collapse
Affiliation(s)
- Hanan Qasim
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy (H.Q., M.R., Y.X., A.R.-A., H.Y.A., B.K.M.), University of Houston, TX
| | - Mehrdad Rajaei
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy (H.Q., M.R., Y.X., A.R.-A., H.Y.A., B.K.M.), University of Houston, TX
| | - Ying Xu
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy (H.Q., M.R., Y.X., A.R.-A., H.Y.A., B.K.M.), University of Houston, TX
| | - Arfaxad Reyes-Alcaraz
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy (H.Q., M.R., Y.X., A.R.-A., H.Y.A., B.K.M.), University of Houston, TX
| | - Hala Y Abdelnasser
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy (H.Q., M.R., Y.X., A.R.-A., H.Y.A., B.K.M.), University of Houston, TX
| | - M David Stewart
- Department of Biology and Biochemistry (M.D.S.), University of Houston, TX
| | - Satadru K Lahiri
- Cardiovascular Research Institute, Departments of Integrative Physiology, Medicine, Neuroscience, Pediatrics, and Center for Space Medicine, Baylor College of Medicine, Houston, TX (S.K.L., X.H.T.W.)
| | - Xander H T Wehrens
- Cardiovascular Research Institute, Departments of Integrative Physiology, Medicine, Neuroscience, Pediatrics, and Center for Space Medicine, Baylor College of Medicine, Houston, TX (S.K.L., X.H.T.W.)
| | - Bradley K McConnell
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy (H.Q., M.R., Y.X., A.R.-A., H.Y.A., B.K.M.), University of Houston, TX
| |
Collapse
|
6
|
Raven F, Riemersma IW, Olthuis MF, Rybakovaite I, Meijer EL, Meerlo P, Van der Zee EA, Havekes R. Cofilin overactivation improves hippocampus-dependent short-term memory. Front Behav Neurosci 2023; 17:1243524. [PMID: 37638111 PMCID: PMC10448394 DOI: 10.3389/fnbeh.2023.1243524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Accepted: 07/28/2023] [Indexed: 08/29/2023] Open
Abstract
Many living organisms of the animal kingdom have the fundamental ability to form and retrieve memories. Most information is initially stored as short-term memory, which is then converted to a more stable long-term memory through a process called memory consolidation. At the neuronal level, synaptic plasticity is crucial for memory storage. It includes the formation of new spines, as well as the modification of existing spines, thereby tuning and shaping synaptic efficacy. Cofilin critically contributes to memory processes as upon activation, it regulates the shape of dendritic spines by targeting actin filaments. We previously found that prolonged activation of cofilin in hippocampal neurons attenuated the formation of long-term object-location memories. Because the modification of spine shape and structure is also essential for short-term memory formation, we determined whether overactivation of hippocampal cofilin also influences the formation of short-term memories. To this end, mice were either injected with an adeno-associated virus expressing catalytically active cofilin, or an eGFP control, in the hippocampus. We show for the first time that cofilin overactivation improves short-term memory formation in the object-location memory task, without affecting anxiety-like behavior. Surprisingly, we found no effect of cofilin overactivation on AMPA receptor expression levels. Altogether, while cofilin overactivation might negatively impact the formation of long-lasting memories, it may benefit short-term plasticity.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Robbert Havekes
- Neurobiology Expertise Group, Groningen Institute for Evolutionary Life Sciences (GELIFES), University of Groningen, Groningen, Netherlands
| |
Collapse
|
7
|
Kimura S, Lok J, Gelman IH, Lo EH, Arai K. Role of A-Kinase Anchoring Protein 12 in the Central Nervous System. J Clin Neurol 2023; 19:329-337. [PMID: 37417430 DOI: 10.3988/jcn.2023.0095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 04/09/2023] [Accepted: 04/10/2023] [Indexed: 07/08/2023] Open
Abstract
A-kinase anchoring protein (AKAP) 12 is a scaffolding protein that anchors various signaling proteins to the plasma membrane. These signaling proteins include protein kinase A, protein kinase C, protein phosphatase 2B, Src-family kinases, cyclins, and calmodulin, which regulate their respective signaling pathways. AKAP12 expression is observed in the neurons, astrocytes, endothelial cells, pericytes, and oligodendrocytes of the central nervous system (CNS). Its physiological roles include promoting the development of the blood-brain barrier, maintaining white-matter homeostasis, and even regulating complex cognitive functions such as long-term memory formation. Under pathological conditions, dysregulation of AKAP12 expression levels may be involved in the pathology of neurological diseases such as ischemic brain injury and Alzheimer's disease. This minireview aimed to summarize the current literature on the role of AKAP12 in the CNS.
Collapse
Affiliation(s)
- Shintaro Kimura
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Life Science Research Center, Gifu University, Gifu, Japan
| | - Josephine Lok
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Pediatric Critical Care Medicine, Department of Pediatrics, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Irwin H Gelman
- Department of Cancer Genetics and Genomics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Eng H Lo
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Ken Arai
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
8
|
Collins KB, Scott JD. Phosphorylation, compartmentalization, and cardiac function. IUBMB Life 2023; 75:353-369. [PMID: 36177749 PMCID: PMC10049969 DOI: 10.1002/iub.2677] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Accepted: 09/15/2022] [Indexed: 11/08/2022]
Abstract
Protein phosphorylation is a fundamental element of cell signaling. First discovered as a biochemical switch in glycogen metabolism, we now know that this posttranslational modification permeates all aspects of cellular behavior. In humans, over 540 protein kinases attach phosphate to acceptor amino acids, whereas around 160 phosphoprotein phosphatases remove phosphate to terminate signaling. Aberrant phosphorylation underlies disease, and kinase inhibitor drugs are increasingly used clinically as targeted therapies. Specificity in protein phosphorylation is achieved in part because kinases and phosphatases are spatially organized inside cells. A prototypic example is compartmentalization of the cyclic adenosine 3',5'-monophosphate (cAMP)-dependent protein kinase A through association with A-kinase anchoring proteins. This configuration creates autonomous signaling islands where the anchored kinase is constrained in proximity to activators, effectors, and selected substates. This article primarily focuses on A kinase anchoring protein (AKAP) signaling in the heart with an emphasis on anchoring proteins that spatiotemporally coordinate excitation-contraction coupling and hypertrophic responses.
Collapse
Affiliation(s)
- Kerrie B. Collins
- Department of Pharmacology, University of Washington, School of Medicine, 1959 NE Pacific Ave, Seattle WA, 98195
| | - John D. Scott
- Department of Pharmacology, University of Washington, School of Medicine, 1959 NE Pacific Ave, Seattle WA, 98195
| |
Collapse
|
9
|
Zhou R, Cao Y, Xiang Y, Fang P, Shang W. Emerging roles of histone deacetylases in adaptive thermogenesis. Front Endocrinol (Lausanne) 2023; 14:1124408. [PMID: 36875455 PMCID: PMC9978507 DOI: 10.3389/fendo.2023.1124408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 02/07/2023] [Indexed: 02/18/2023] Open
Abstract
Brown and beige adipose tissues regulate body energy expenditure through adaptive thermogenesis, which converts energy into heat by oxidative phosphorylation uncoupling. Although promoting adaptive thermogenesis has been demonstrated to be a prospective strategy for obesity control, there are few methods for increasing adipose tissue thermogenesis in a safe and effective way. Histone deacetylase (HDAC) is a category of epigenetic modifying enzymes that catalyzes deacetylation on both histone and non-histone proteins. Recent studies illustrated that HDACs play an important role in adipose tissue thermogenesis through modulating gene transcription and chromatin structure as well as cellular signals transduction in both deacetylation dependent or independent manners. Given that different classes and subtypes of HDACs show diversity in the mechanisms of adaptive thermogenesis regulation, we systematically summarized the effects of different HDACs on adaptive thermogenesis and their underlying mechanisms in this review. We also emphasized the differences among HDACs in thermogenesis regulation, which will help to find new efficient anti-obesity drugs targeting specific HDAC subtypes.
Collapse
Affiliation(s)
- Ruonan Zhou
- Department of Endocrinology, Jiangsu Province Hospital of Chinese Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- Key Laboratory for Metabolic Diseases in Chinese Medicine, First College of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yue Cao
- Department of Endocrinology, Jiangsu Province Hospital of Chinese Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- Key Laboratory for Metabolic Diseases in Chinese Medicine, First College of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yingying Xiang
- Department of Endocrinology, Jiangsu Province Hospital of Chinese Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- Key Laboratory for Metabolic Diseases in Chinese Medicine, First College of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Penghua Fang
- Key Laboratory for Metabolic Diseases in Chinese Medicine, First College of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Wenbin Shang
- Department of Endocrinology, Jiangsu Province Hospital of Chinese Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- Key Laboratory for Metabolic Diseases in Chinese Medicine, First College of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
10
|
Reversible lysine fatty acylation of an anchoring protein mediates adipocyte adrenergic signaling. Proc Natl Acad Sci U S A 2022; 119:2119678119. [PMID: 35149557 PMCID: PMC8851525 DOI: 10.1073/pnas.2119678119] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/08/2021] [Indexed: 01/05/2023] Open
Abstract
N-myristoylation on glycine is an irreversible modification that has long been recognized to govern protein localization and function. In contrast, the biological roles of lysine myristoylation remain ill-defined. We demonstrate that the cytoplasmic scaffolding protein, gravin-α/A kinase-anchoring protein 12, is myristoylated on two lysine residues embedded in its carboxyl-terminal protein kinase A (PKA) binding domain. Histone deacetylase 11 (HDAC11) docks to an adjacent region of gravin-α and demyristoylates these sites. In brown and white adipocytes, lysine myristoylation of gravin-α is required for signaling via β2- and β3-adrenergic receptors (β-ARs), which are G protein-coupled receptors (GPCRs). Lysine myristoylation of gravin-α drives β-ARs to lipid raft membrane microdomains, which results in PKA activation and downstream signaling that culminates in protective thermogenic gene expression. These findings define reversible lysine myristoylation as a mechanism for controlling GPCR signaling and highlight the potential of inhibiting HDAC11 to manipulate adipocyte phenotypes for therapeutic purposes.
Collapse
|
11
|
Wang X, Zhao Y, Shi X, Gong M, Hao Y, Fu Y, Velez de-la-Paz OI, Wang X, Du Y, Guo X, Song L, Meng L, Gao Y, Yin X, Wang S, Shi Y, Shi H. Sulfur dioxide derivatives attenuates consolidation of contextual fear memory in mice. Eur J Pharmacol 2022; 914:174658. [PMID: 34861211 DOI: 10.1016/j.ejphar.2021.174658] [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: 02/19/2021] [Revised: 11/20/2021] [Accepted: 11/29/2021] [Indexed: 11/28/2022]
Abstract
Post-traumatic stress disorder (PTSD) is characterized by an enhancement of traumatic memory. Intervention strategies based on the different stages of memory have been shown to be effective in the prevention and control of PTSD. The endogenous gaseous molecule, sulfur dioxide (SO2), has been reported to significantly exert neuromodulatory effects; however, its regulation of learning and memory remains unestablished. This study aimed to investigate the effects of exogenous SO2 derivatives administration on the formation, consolidation, reconsolidation, retention, and expression of contextual fear memory. Behavioral results showed that both intraperitoneal injection (50 mg/kg, ip) and hippocampal infusion (5 μg/side) of SO2 derivatives (a mixture of sodium sulfite and sodium bisulfite, Na2SO3/NaHSO3, 3:1 M/M) significantly impaired consolidation but had no effect on reconsolidation and retention of contextual fear memory. These findings suggest that the attenuating effects of SO2 on the consolidation of fear memory involves, at least partially, the region of the hippocampus. The findings of this study provide direct evidence for the development of new strategies for PTSD prevention and treatment involving the use of gaseous SO2.
Collapse
Affiliation(s)
- Xinhao Wang
- Neuroscience Research Center, Institute of Medical and Health Science of HeBMU, Hebei Medical University, Shijiazhuang, 050017, China; Hebei Key Laboratory of Neurophysiology, Hebei Medicinal University, Shijiazhuang, 050017, China
| | - Yize Zhao
- Neuroscience Research Center, Institute of Medical and Health Science of HeBMU, Hebei Medical University, Shijiazhuang, 050017, China; Hebei Key Laboratory of Neurophysiology, Hebei Medicinal University, Shijiazhuang, 050017, China
| | - Xiaorui Shi
- Neuroscience Research Center, Institute of Medical and Health Science of HeBMU, Hebei Medical University, Shijiazhuang, 050017, China; Hebei Key Laboratory of Neurophysiology, Hebei Medicinal University, Shijiazhuang, 050017, China
| | - Miao Gong
- Neuroscience Research Center, Institute of Medical and Health Science of HeBMU, Hebei Medical University, Shijiazhuang, 050017, China; Hebei Key Laboratory of Neurophysiology, Hebei Medicinal University, Shijiazhuang, 050017, China
| | - Ying Hao
- Neuroscience Research Center, Institute of Medical and Health Science of HeBMU, Hebei Medical University, Shijiazhuang, 050017, China; Hebei Key Laboratory of Neurophysiology, Hebei Medicinal University, Shijiazhuang, 050017, China
| | - Yaling Fu
- Neuroscience Research Center, Institute of Medical and Health Science of HeBMU, Hebei Medical University, Shijiazhuang, 050017, China; Hebei Key Laboratory of Neurophysiology, Hebei Medicinal University, Shijiazhuang, 050017, China
| | - Omar Israel Velez de-la-Paz
- Neuroscience Research Center, Institute of Medical and Health Science of HeBMU, Hebei Medical University, Shijiazhuang, 050017, China; Hebei Key Laboratory of Neurophysiology, Hebei Medicinal University, Shijiazhuang, 050017, China
| | - Xi Wang
- Neuroscience Research Center, Institute of Medical and Health Science of HeBMU, Hebei Medical University, Shijiazhuang, 050017, China; Hebei Key Laboratory of Neurophysiology, Hebei Medicinal University, Shijiazhuang, 050017, China
| | - Yuru Du
- Neuroscience Research Center, Institute of Medical and Health Science of HeBMU, Hebei Medical University, Shijiazhuang, 050017, China; Hebei Key Laboratory of Neurophysiology, Hebei Medicinal University, Shijiazhuang, 050017, China
| | - Xiangfei Guo
- Neuroscience Research Center, Institute of Medical and Health Science of HeBMU, Hebei Medical University, Shijiazhuang, 050017, China; Hebei Key Laboratory of Neurophysiology, Hebei Medicinal University, Shijiazhuang, 050017, China
| | - Li Song
- Neuroscience Research Center, Institute of Medical and Health Science of HeBMU, Hebei Medical University, Shijiazhuang, 050017, China; Hebei Key Laboratory of Neurophysiology, Hebei Medicinal University, Shijiazhuang, 050017, China; Department of Biochemistry and Molecular Biology, College of Basic Medicine, Hebei Medical University, Shijiazhuang, Hebei, 050017, China
| | - Li Meng
- Neuroscience Research Center, Institute of Medical and Health Science of HeBMU, Hebei Medical University, Shijiazhuang, 050017, China; Hebei Key Laboratory of Neurophysiology, Hebei Medicinal University, Shijiazhuang, 050017, China
| | - Yuan Gao
- Neuroscience Research Center, Institute of Medical and Health Science of HeBMU, Hebei Medical University, Shijiazhuang, 050017, China; Hebei Key Laboratory of Neurophysiology, Hebei Medicinal University, Shijiazhuang, 050017, China; Department of Biochemistry and Molecular Biology, College of Basic Medicine, Hebei Medical University, Shijiazhuang, Hebei, 050017, China
| | - Xi Yin
- Neuroscience Research Center, Institute of Medical and Health Science of HeBMU, Hebei Medical University, Shijiazhuang, 050017, China; Department of Functional Region of Diagnosis, Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, China
| | - Sheng Wang
- Neuroscience Research Center, Institute of Medical and Health Science of HeBMU, Hebei Medical University, Shijiazhuang, 050017, China; Hebei Key Laboratory of Neurophysiology, Hebei Medicinal University, Shijiazhuang, 050017, China
| | - Yun Shi
- Neuroscience Research Center, Institute of Medical and Health Science of HeBMU, Hebei Medical University, Shijiazhuang, 050017, China; Department of Biochemistry and Molecular Biology, College of Basic Medicine, Hebei Medical University, Shijiazhuang, Hebei, 050017, China.
| | - Haishui Shi
- Neuroscience Research Center, Institute of Medical and Health Science of HeBMU, Hebei Medical University, Shijiazhuang, 050017, China; Hebei Key Laboratory of Neurophysiology, Hebei Medicinal University, Shijiazhuang, 050017, China.
| |
Collapse
|
12
|
Liu S, Xiu J, Zhu C, Meng K, Li C, Han R, Du T, Li L, Xu L, Liu R, Zhu W, Shen Y, Xu Q. Fat mass and obesity-associated protein regulates RNA methylation associated with depression-like behavior in mice. Nat Commun 2021; 12:6937. [PMID: 34836959 PMCID: PMC8626436 DOI: 10.1038/s41467-021-27044-7] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Accepted: 10/28/2021] [Indexed: 11/30/2022] Open
Abstract
Post-transcriptional modifications of RNA, such as RNA methylation, can epigenetically regulate behavior, for instance learning and memory. However, it is unclear whether RNA methylation plays a critical role in the pathophysiology of major depression disorder (MDD). Here, we report that expression of the fat mass and obesity associated gene (FTO), an RNA demethylase, is downregulated in the hippocampus of patients with MDD and mouse models of depression. Suppressing Fto expression in the mouse hippocampus results in depression-like behaviors in adult mice, whereas overexpression of FTO expression leads to rescue of the depression-like phenotype. Epitranscriptomic profiling of N6-methyladenosine (m6A) RNA methylation in the hippocampus of Fto knockdown (KD), Fto knockout (cKO), and FTO-overexpressing (OE) mice allows us to identify adrenoceptor beta 2 (Adrb2) mRNA as a target of FTO. ADRB2 stimulation rescues the depression-like behaviors in mice and spine loss induced by hippocampal Fto deficiency, possibly via the modulation of hippocampal SIRT1 expression by c-MYC. Our findings suggest that FTO is a regulator of a mechanism underlying depression-like behavior in mice.
Collapse
Affiliation(s)
- Shu Liu
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, 100005, China
- Neuroscience Center, Chinese Academy of Medical Sciences, Beijing, 100005, China
| | - Jianbo Xiu
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, 100005, China
- Neuroscience Center, Chinese Academy of Medical Sciences, Beijing, 100005, China
| | - Caiyun Zhu
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, 100005, China
- Neuroscience Center, Chinese Academy of Medical Sciences, Beijing, 100005, China
| | - Kexin Meng
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, 100005, China
- Neuroscience Center, Chinese Academy of Medical Sciences, Beijing, 100005, China
| | - Chen Li
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, 100005, China
- Neuroscience Center, Chinese Academy of Medical Sciences, Beijing, 100005, China
| | - Rongrong Han
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, 100005, China
- Neuroscience Center, Chinese Academy of Medical Sciences, Beijing, 100005, China
| | - Tingfu Du
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, 100005, China
- Neuroscience Center, Chinese Academy of Medical Sciences, Beijing, 100005, China
| | - Lanlan Li
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, 100005, China
- Neuroscience Center, Chinese Academy of Medical Sciences, Beijing, 100005, China
| | - Lingdan Xu
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, 100005, China
- Neuroscience Center, Chinese Academy of Medical Sciences, Beijing, 100005, China
| | - Renjie Liu
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, 100005, China
- Neuroscience Center, Chinese Academy of Medical Sciences, Beijing, 100005, China
| | - Wanwan Zhu
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, 100005, China
- Neuroscience Center, Chinese Academy of Medical Sciences, Beijing, 100005, China
| | - Yan Shen
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, 100005, China
- Neuroscience Center, Chinese Academy of Medical Sciences, Beijing, 100005, China
| | - Qi Xu
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, 100005, China.
- Neuroscience Center, Chinese Academy of Medical Sciences, Beijing, 100005, China.
| |
Collapse
|
13
|
Abstract
The field of cAMP signaling is witnessing exciting developments with the recognition that cAMP is compartmentalized and that spatial regulation of cAMP is critical for faithful signal coding. This realization has changed our understanding of cAMP signaling from a model in which cAMP connects a receptor at the plasma membrane to an intracellular effector in a linear pathway to a model in which cAMP signals propagate within a complex network of alternative branches and the specific functional outcome strictly depends on local regulation of cAMP levels and on selective activation of a limited number of branches within the network. In this review, we cover some of the early studies and summarize more recent evidence supporting the model of compartmentalized cAMP signaling, and we discuss how this knowledge is starting to provide original mechanistic insight into cell physiology and a novel framework for the identification of disease mechanisms that potentially opens new avenues for therapeutic interventions. SIGNIFICANCE STATEMENT: cAMP mediates the intracellular response to multiple hormones and neurotransmitters. Signal fidelity and accurate coordination of a plethora of different cellular functions is achieved via organization of multiprotein signalosomes and cAMP compartmentalization in subcellular nanodomains. Defining the organization and regulation of subcellular cAMP nanocompartments is necessary if we want to understand the complex functional ramifications of pharmacological treatments that target G protein-coupled receptors and for generating a blueprint that can be used to develop precision medicine interventions.
Collapse
Affiliation(s)
- Manuela Zaccolo
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| | - Anna Zerio
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| | - Miguel J Lobo
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
14
|
Heckman PR, Roig Kuhn F, Meerlo P, Havekes R. A brief period of sleep deprivation negatively impacts the acquisition, consolidation, and retrieval of object-location memories. Neurobiol Learn Mem 2020; 175:107326. [DOI: 10.1016/j.nlm.2020.107326] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 08/24/2020] [Accepted: 10/08/2020] [Indexed: 01/06/2023]
|
15
|
Peixoto RT, Chantranupong L, Hakim R, Levasseur J, Wang W, Merchant T, Gorman K, Budnik B, Sabatini BL. Abnormal Striatal Development Underlies the Early Onset of Behavioral Deficits in Shank3B -/- Mice. Cell Rep 2020; 29:2016-2027.e4. [PMID: 31722214 PMCID: PMC6889826 DOI: 10.1016/j.celrep.2019.10.021] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Revised: 07/12/2019] [Accepted: 10/04/2019] [Indexed: 11/17/2022] Open
Abstract
The neural substrates and pathophysiological mechanisms underlying the onset of cognitive and motor deficits in autism spectrum disorders (ASDs) remain unclear. Mutations in ASD-associated SHANK3 in mice (Shank3B−/−) result in the accelerated maturation of corticostriatal circuits during the second and third postnatal weeks. Here, we show that during this period, there is extensive remodeling of the striatal synaptic proteome and a developmental switch in glutamatergic synaptic plasticity induced by cortical hyperactivity in striatal spiny projection neurons (SPNs). Behavioral abnormalities in Shank3B−/− mice emerge during this stage and are ameliorated by normalizing excitatory synapse connectivity in medial striatal regions by the downregulation of PKA activity. These results suggest that the abnormal postnatal development of striatal circuits is implicated in the onset of behavioral deficits in Shank3B−/− mice and that modulation of postsynaptic PKA activity can be used to regulate corticostriatal drive in developing SPNs of mouse models of ASDs and other neurodevelopmental disorders. Peixoto et al. show that the onset of behavioral deficits in Shank3B−/− mice occurs during early postnatal development and that these can be ameliorated by reducing the glutamatergic synaptic drive in medial regions of the striatum by the downregulation of PKA activity.
Collapse
Affiliation(s)
- Rui Tiago Peixoto
- Department of Psychiatry, University of Pittsburgh, 450 Technology Dr, Pittsburgh, PA 15219, USA; Howard Hughes Medical Institute, Department of Neurobiology, Harvard Medical School, 220 Longwood Ave., Boston, MA 02115, USA.
| | - Lynne Chantranupong
- Howard Hughes Medical Institute, Department of Neurobiology, Harvard Medical School, 220 Longwood Ave., Boston, MA 02115, USA
| | - Richard Hakim
- Howard Hughes Medical Institute, Department of Neurobiology, Harvard Medical School, 220 Longwood Ave., Boston, MA 02115, USA
| | - James Levasseur
- Howard Hughes Medical Institute, Department of Neurobiology, Harvard Medical School, 220 Longwood Ave., Boston, MA 02115, USA
| | - Wengang Wang
- Howard Hughes Medical Institute, Department of Neurobiology, Harvard Medical School, 220 Longwood Ave., Boston, MA 02115, USA
| | - Tasha Merchant
- Department of Psychiatry, University of Pittsburgh, 450 Technology Dr, Pittsburgh, PA 15219, USA; Howard Hughes Medical Institute, Department of Neurobiology, Harvard Medical School, 220 Longwood Ave., Boston, MA 02115, USA
| | - Kelly Gorman
- Howard Hughes Medical Institute, Department of Neurobiology, Harvard Medical School, 220 Longwood Ave., Boston, MA 02115, USA
| | - Bogdan Budnik
- Mass Spectrometry and Proteomic Laboratory, FAS Division of Science, Harvard University, 52 Oxford Street, Cambridge, MA 02138, USA
| | - Bernardo Luis Sabatini
- Howard Hughes Medical Institute, Department of Neurobiology, Harvard Medical School, 220 Longwood Ave., Boston, MA 02115, USA
| |
Collapse
|
16
|
Kelly MP, Heckman PRA, Havekes R. Genetic manipulation of cyclic nucleotide signaling during hippocampal neuroplasticity and memory formation. Prog Neurobiol 2020; 190:101799. [PMID: 32360536 DOI: 10.1016/j.pneurobio.2020.101799] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 03/14/2020] [Accepted: 03/26/2020] [Indexed: 12/12/2022]
Abstract
Decades of research have underscored the importance of cyclic nucleotide signaling in memory formation and synaptic plasticity. In recent years, several new genetic techniques have expanded the neuroscience toolbox, allowing researchers to measure and modulate cyclic nucleotide gradients with high spatiotemporal resolution. Here, we will provide an overview of studies using genetic approaches to interrogate the role cyclic nucleotide signaling plays in hippocampus-dependent memory processes and synaptic plasticity. Particular attention is given to genetic techniques that measure real-time changes in cyclic nucleotide levels as well as newly-developed genetic strategies to transiently manipulate cyclic nucleotide signaling in a subcellular compartment-specific manner with high temporal resolution.
Collapse
Affiliation(s)
- Michy P Kelly
- Department of Pharmacology, Physiology & Neuroscience, University of South Carolina School of Medicine, 6439 Garners Ferry Rd, VA Bldg1, 3(rd) Fl, D-12, Columbia, 29209, SC, USA.
| | - Pim R A Heckman
- Neurobiology Expertise Group, Groningen Institute for Evolutionary Life Sciences (GELIFES), University of Groningen, Nijenborgh 7, 9747 AG Groningen, the Netherlands.
| | - Robbert Havekes
- Neurobiology Expertise Group, Groningen Institute for Evolutionary Life Sciences (GELIFES), University of Groningen, Nijenborgh 7, 9747 AG Groningen, the Netherlands.
| |
Collapse
|
17
|
Nguyen PV, Connor SA. Noradrenergic Regulation of Hippocampus-Dependent Memory. Cent Nerv Syst Agents Med Chem 2020; 19:187-196. [PMID: 31749419 DOI: 10.2174/1871524919666190719163632] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Revised: 06/24/2019] [Accepted: 06/28/2019] [Indexed: 12/24/2022]
Abstract
Neuromodulation regulates critical functions of CNS synapses, ranging from neural circuit development to high-order cognitive processes, including learning and memory. This broad scope of action is generally mediated through alterations of the strength of synaptic transmission (i.e. synaptic plasticity). Changes in synaptic strength are widely considered to be a cellular representation of learned information. Noradrenaline is a neuromodulator that is secreted throughout the brain in response to novelty or increased arousal. Once released, noradrenaline activates metabotropic receptors, initiating intracellular signaling cascades that promote enduring changes in synaptic strength and facilitate memory storage. Here, we provide an overview of noradrenergic modulation of synaptic plasticity and memory formation within mammalian neural circuits, which has broad applicability within the neurotherapeutics community. Advances in our understanding of noradrenaline in the context of these processes may provide a foundation for refining treatment strategies for multiple brain diseases, ranging from post-traumatic stress disorder to Alzheimer's Disease.
Collapse
Affiliation(s)
- Peter V Nguyen
- Department of Physiology, University of Alberta School of Medicine, Edmonton, AB, T6G 2H7, Canada
| | - Steven A Connor
- Department of Biology, York University, 4700 Keele Street, Toronto, ON, M3J 1P3, Canada
| |
Collapse
|
18
|
Purkey AM, Dell’Acqua ML. Phosphorylation-Dependent Regulation of Ca 2+-Permeable AMPA Receptors During Hippocampal Synaptic Plasticity. Front Synaptic Neurosci 2020; 12:8. [PMID: 32292336 PMCID: PMC7119613 DOI: 10.3389/fnsyn.2020.00008] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 02/18/2020] [Indexed: 01/28/2023] Open
Abstract
Experience-dependent learning and memory require multiple forms of plasticity at hippocampal and cortical synapses that are regulated by N-methyl-D-aspartate receptors (NMDA) and α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA)-type ionotropic glutamate receptors (NMDAR, AMPAR). These plasticity mechanisms include long-term potentiation (LTP) and depression (LTD), which are Hebbian input-specific mechanisms that rapidly increase or decrease AMPAR synaptic strength at specific inputs, and homeostatic plasticity that globally scales-up or -down AMPAR synaptic strength across many or even all inputs. Frequently, these changes in synaptic strength are also accompanied by a change in the subunit composition of AMPARs at the synapse due to the trafficking to and from the synapse of receptors lacking GluA2 subunits. These GluA2-lacking receptors are most often GluA1 homomeric receptors that exhibit higher single-channel conductance and are Ca2+-permeable (CP-AMPAR). This review article will focus on the role of protein phosphorylation in regulation of GluA1 CP-AMPAR recruitment and removal from hippocampal synapses during synaptic plasticity with an emphasis on the crucial role of local signaling by the cAMP-dependent protein kinase (PKA) and the Ca2+calmodulin-dependent protein phosphatase 2B/calcineurin (CaN) that is coordinated by the postsynaptic scaffold protein A-kinase anchoring protein 79/150 (AKAP79/150).
Collapse
Affiliation(s)
| | - Mark L. Dell’Acqua
- Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO, United States
| |
Collapse
|
19
|
Bridi M, Schoch H, Florian C, Poplawski SG, Banerjee A, Hawk JD, Porcari GS, Lejards C, Hahn CG, Giese KP, Havekes R, Spruston N, Abel T. Transcriptional corepressor SIN3A regulates hippocampal synaptic plasticity via Homer1/mGluR5 signaling. JCI Insight 2020; 5:92385. [PMID: 32069266 DOI: 10.1172/jci.insight.92385] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Accepted: 02/12/2020] [Indexed: 12/12/2022] Open
Abstract
Long-term memory depends on the control of activity-dependent neuronal gene expression, which is regulated by epigenetic modifications. The epigenetic modification of histones is orchestrated by the opposing activities of 2 classes of regulatory complexes: permissive coactivators and silencing corepressors. Much work has focused on coactivator complexes, but little is known about the corepressor complexes that suppress the expression of plasticity-related genes. Here, we define a critical role for the corepressor SIN3A in memory and synaptic plasticity, showing that postnatal neuronal deletion of Sin3a enhances hippocampal long-term potentiation and long-term contextual fear memory. SIN3A regulates the expression of genes encoding proteins in the postsynaptic density. Loss of SIN3A increases expression of the synaptic scaffold Homer1, alters the metabotropic glutamate receptor 1α (mGluR1α) and mGluR5 dependence of long-term potentiation, and increases activation of ERK in the hippocampus after learning. Our studies define a critical role for corepressors in modulating neural plasticity and memory consolidation and reveal that Homer1/mGluR signaling pathways may be central molecular mechanisms for memory enhancement.
Collapse
Affiliation(s)
| | | | | | | | - Anamika Banerjee
- Department of Psychiatry, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | | | | | | | - Chang-Gyu Hahn
- Department of Psychiatry, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | | | | | - Nelson Spruston
- Howard Hughes Medical Institute (HHMI) Janelia Research Campus, Ashburn, Virginia, USA
| | | |
Collapse
|
20
|
Raven F, Bolsius YG, Renssen LV, Meijer EL, Zee EA, Meerlo P, Havekes R. Elucidating the role of protein synthesis in hippocampus‐dependent memory consolidation across the day and night. Eur J Neurosci 2020; 54:6972-6981. [PMID: 31965655 PMCID: PMC8596627 DOI: 10.1111/ejn.14684] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Revised: 01/08/2020] [Accepted: 01/13/2020] [Indexed: 01/11/2023]
Abstract
It is widely acknowledged that de novo protein synthesis is crucial for the formation and consolidation of long‐term memories. While the basal activity of many signaling cascades that modulate protein synthesis fluctuates in a circadian fashion, it is unclear whether the temporal dynamics of protein synthesis‐dependent memory consolidation vary depending on the time of day. More specifically, it is unclear whether protein synthesis inhibition affects hippocampus‐dependent memory consolidation in rodents differentially across the day (i.e., the inactive phase with an abundance of sleep) and night (i.e., the active phase with little sleep). To address this question, male and female C57Bl6/J mice were trained in a contextual fear conditioning task at the beginning or the end of the light phase. Animals received a single systemic injection with the protein synthesis inhibitor anisomycin or vehicle directly, 4, 8 hr, or 11.5 hr following training, and memory was assessed after 24 hr. Here, we show that protein synthesis inhibition impaired the consolidation of context–fear memories selectively when the protein synthesis inhibitor was administered at the first three time points, irrespective of timing of training. Even though the basal activity of signaling pathways regulating de novo protein synthesis may fluctuate across the 24‐hr cycle, these results suggest that the temporal dynamics of protein synthesis‐dependent memory consolidation are similar for day‐time and night‐time learning.
Collapse
Affiliation(s)
- Frank Raven
- Groningen Institute for Evolutionary Life Sciences (GELIFES) University of Groningen Groningen The Netherlands
| | - Youri G. Bolsius
- Groningen Institute for Evolutionary Life Sciences (GELIFES) University of Groningen Groningen The Netherlands
| | - Lara V. Renssen
- Groningen Institute for Evolutionary Life Sciences (GELIFES) University of Groningen Groningen The Netherlands
| | - Elroy L. Meijer
- Groningen Institute for Evolutionary Life Sciences (GELIFES) University of Groningen Groningen The Netherlands
| | - Eddy A. Zee
- Groningen Institute for Evolutionary Life Sciences (GELIFES) University of Groningen Groningen The Netherlands
| | - Peter Meerlo
- Groningen Institute for Evolutionary Life Sciences (GELIFES) University of Groningen Groningen The Netherlands
| | - Robbert Havekes
- Groningen Institute for Evolutionary Life Sciences (GELIFES) University of Groningen Groningen The Netherlands
| |
Collapse
|
21
|
The Role of Cyclic AMP Signaling in Cardiac Fibrosis. Cells 2019; 9:cells9010069. [PMID: 31888098 PMCID: PMC7016856 DOI: 10.3390/cells9010069] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Revised: 12/23/2019] [Accepted: 12/24/2019] [Indexed: 12/18/2022] Open
Abstract
Myocardial stress and injury invariably promote remodeling of the cardiac tissue, which is associated with cardiomyocyte death and development of fibrosis. The fibrotic process is initially triggered by the differentiation of resident cardiac fibroblasts into myofibroblasts. These activated fibroblasts display increased proliferative capacity and secrete large amounts of extracellular matrix. Uncontrolled myofibroblast activation can thus promote heart stiffness, cardiac dysfunction, arrhythmias, and progression to heart failure. Despite the well-established role of myofibroblasts in mediating cardiac disease, our current knowledge on how signaling pathways promoting fibrosis are regulated and coordinated in this cell type is largely incomplete. In this respect, cyclic adenosine monophosphate (cAMP) signaling acts as a major modulator of fibrotic responses activated in fibroblasts of injured or stressed hearts. In particular, accumulating evidence now suggests that upstream cAMP modulators including G protein-coupled receptors, adenylyl cyclases (ACs), and phosphodiesterases (PDEs); downstream cAMP effectors such as protein kinase A (PKA) and the guanine nucleotide exchange factor Epac; and cAMP signaling organizers such as A-kinase anchoring proteins (AKAPs) modulate a variety of fundamental cellular processes involved in myocardial fibrosis including myofibroblast differentiation, proliferation, collagen secretion, and invasiveness. The current review will discuss recent advances highlighting the role of cAMP and AKAP-mediated signaling in regulating pathophysiological responses controlling cardiac fibrosis.
Collapse
|
22
|
Zhu Y, Jiang X, Zheng Y, Xiong J, Wei D, Zhang D. Cardiac function modulation depends on the A-kinase anchoring protein complex. J Cell Mol Med 2019; 23:7170-7179. [PMID: 31512389 PMCID: PMC6815827 DOI: 10.1111/jcmm.14659] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 06/27/2019] [Accepted: 08/06/2019] [Indexed: 12/26/2022] Open
Abstract
The A-kinase anchoring proteins (AKAPs) are a group of structurally diverse proteins identified in various species and tissues. These proteins are able to anchor protein kinase and other signalling proteins to regulate cardiac function. Acting as a scaffold protein, AKAPs ensure specificity in signal transduction by enzymes close to their appropriate effectors and substrates. Over the decades, more than 70 different AKAPs have been discovered. Accumulative evidence indicates that AKAPs play crucial roles in the functional regulation of cardiac diseases, including cardiac hypertrophy, myofibre contractility dysfunction and arrhythmias. By anchoring different partner proteins (PKA, PKC, PKD and LTCCs), AKAPs take part in different regulatory pathways to function as regulators in the heart, and a damaged structure can influence the activities of these complexes. In this review, we highlight recent advances in AKAP-associated protein complexes, focusing on local signalling events that are perturbed in cardiac diseases and their roles in interacting with ion channels and their regulatory molecules. These new findings suggest that AKAPs might have potential therapeutic value in patients with cardiac diseases, particularly malignant rhythm.
Collapse
Affiliation(s)
- Yan‐Rong Zhu
- Department of CardiologyNanjing First HospitalNanjing Medical UniversityNanjingChina
| | - Xiao‐Xin Jiang
- Department of CardiologyNanjing First HospitalNanjing Medical UniversityNanjingChina
| | - Yaguo Zheng
- Department of CardiologyNanjing First HospitalNanjing Medical UniversityNanjingChina
| | - Jing Xiong
- Department of PharmacologyNanjing Medical UniversityNanjingChina
| | - Dongping Wei
- Department of OncologyNanjing First HospitalNanjing Medical UniversityNanjingChina
| | - Dai‐Min Zhang
- Department of CardiologyNanjing First HospitalNanjing Medical UniversityNanjingChina
| |
Collapse
|
23
|
Neuromodulators and Long-Term Synaptic Plasticity in Learning and Memory: A Steered-Glutamatergic Perspective. Brain Sci 2019; 9:brainsci9110300. [PMID: 31683595 PMCID: PMC6896105 DOI: 10.3390/brainsci9110300] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 10/24/2019] [Accepted: 10/29/2019] [Indexed: 12/19/2022] Open
Abstract
The molecular pathways underlying the induction and maintenance of long-term synaptic plasticity have been extensively investigated revealing various mechanisms by which neurons control their synaptic strength. The dynamic nature of neuronal connections combined with plasticity-mediated long-lasting structural and functional alterations provide valuable insights into neuronal encoding processes as molecular substrates of not only learning and memory but potentially other sensory, motor and behavioural functions that reflect previous experience. However, one key element receiving little attention in the study of synaptic plasticity is the role of neuromodulators, which are known to orchestrate neuronal activity on brain-wide, network and synaptic scales. We aim to review current evidence on the mechanisms by which certain modulators, namely dopamine, acetylcholine, noradrenaline and serotonin, control synaptic plasticity induction through corresponding metabotropic receptors in a pathway-specific manner. Lastly, we propose that neuromodulators control plasticity outcomes through steering glutamatergic transmission, thereby gating its induction and maintenance.
Collapse
|
24
|
Hoffman JR, Brandwein NJ, Nguyen PV. Induction of β-adrenergic metaplasticity of LTP requires intact anchoring of PKA. ACTA ACUST UNITED AC 2019; 26:187-190. [PMID: 31109969 PMCID: PMC6529881 DOI: 10.1101/lm.049635.119] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2019] [Accepted: 04/23/2019] [Indexed: 11/24/2022]
Abstract
Beta-adrenergic receptors (β-ARs) prime hippocampal synapses to stabilize long-term potentiation (LTP). This “metaplasticity” can persist for 1–2 h after pharmacologic activation of β-ARs. It requires activation of PKA (cAMP-dependent protein kinase) during β-AR priming. A-kinase anchoring proteins (AKAPs) tether PKA to downstream signaling proteins. We hypothesized that induction of this metaplasticity requires intact functioning of AKAPs. Acute application of stearated ht31, a membrane-permeant inhibitor of AKAPs, either during β-AR activation 30 min prior to LTP induction or during LTP induction, attenuated the persistence of LTP. A control, inactive ht31 peptide did not affect β-AR-mediated metaplasticity. These findings implicate PKA anchoring in the induction of β-adrenergic metaplasticity of LTP.
Collapse
Affiliation(s)
- Janlyn R Hoffman
- Department of Physiology, University of Alberta School of Medicine, Edmonton, Alberta T6G 2H7, Canada
| | - Nathan J Brandwein
- Department of Physiology, University of Alberta School of Medicine, Edmonton, Alberta T6G 2H7, Canada
| | - Peter V Nguyen
- Department of Physiology, University of Alberta School of Medicine, Edmonton, Alberta T6G 2H7, Canada
| |
Collapse
|
25
|
From membrane receptors to protein synthesis and actin cytoskeleton: Mechanisms underlying long lasting forms of synaptic plasticity. Semin Cell Dev Biol 2019; 95:120-129. [PMID: 30634048 DOI: 10.1016/j.semcdb.2019.01.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Revised: 01/07/2019] [Accepted: 01/08/2019] [Indexed: 12/13/2022]
Abstract
Synaptic plasticity, the activity dependent change in synaptic strength, forms the molecular foundation of learning and memory. Synaptic plasticity includes structural changes, with spines changing their size to accomodate insertion and removal of postynaptic receptors, which are correlated with functional changes. Of particular relevance for memory storage are the long lasting forms of synaptic plasticity which are protein synthesis dependent. Due to the importance of spine structural plasticity and protein synthesis, this review focuses on the signaling pathways that connect synaptic stimulation with regulation of protein synthesis and remodeling of the actin cytoskeleton. We also review computational models that implement novel aspects of molecular signaling in synaptic plasticity, such as the role of neuromodulators and spatial microdomains, as well as highlight the need for computational models that connect activation of memory kinases with spine actin dynamics.
Collapse
|
26
|
Patriarchi T, Buonarati OR, Hell JW. Postsynaptic localization and regulation of AMPA receptors and Cav1.2 by β2 adrenergic receptor/PKA and Ca 2+/CaMKII signaling. EMBO J 2018; 37:e99771. [PMID: 30249603 PMCID: PMC6187224 DOI: 10.15252/embj.201899771] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2018] [Revised: 07/25/2018] [Accepted: 08/17/2018] [Indexed: 11/09/2022] Open
Abstract
The synapse transmits, processes, and stores data within its tiny space. Effective and specific signaling requires precise alignment of the relevant components. This review examines current insights into mechanisms of AMPAR and NMDAR localization by PSD-95 and their spatial distribution at postsynaptic sites to illuminate the structural and functional framework of postsynaptic signaling. It subsequently delineates how β2 adrenergic receptor (β2 AR) signaling via adenylyl cyclase and the cAMP-dependent protein kinase PKA is organized within nanodomains. Here, we discuss targeting of β2 AR, adenylyl cyclase, and PKA to defined signaling complexes at postsynaptic sites, i.e., AMPARs and the L-type Ca2+ channel Cav1.2, and other subcellular surface localizations, the role of A kinase anchor proteins, the physiological relevance of the spatial restriction of corresponding signaling, and their interplay with signal transduction by the Ca2+- and calmodulin-dependent kinase CaMKII How localized and specific signaling by cAMP occurs is a central cellular question. The dendritic spine constitutes an ideal paradigm for elucidating the dimensions of spatially restricted signaling because of their small size and defined protein composition.
Collapse
MESH Headings
- Animals
- Calcium Channels, L-Type/genetics
- Calcium Channels, L-Type/metabolism
- Calcium Signaling/physiology
- Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics
- Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism
- Cyclic AMP-Dependent Protein Kinases/genetics
- Cyclic AMP-Dependent Protein Kinases/metabolism
- Humans
- Receptors, AMPA/genetics
- Receptors, AMPA/metabolism
- Receptors, Adrenergic, beta-2/genetics
- Receptors, Adrenergic, beta-2/metabolism
- Receptors, N-Methyl-D-Aspartate/genetics
- Receptors, N-Methyl-D-Aspartate/metabolism
- Synapses/genetics
- Synapses/metabolism
Collapse
Affiliation(s)
- Tommaso Patriarchi
- Department of Pharmacology, University of California, Davis, CA, USA
- Department of Biochemistry and Molecular Medicine, University of California, Davis, CA, USA
| | | | - Johannes W Hell
- Department of Pharmacology, University of California, Davis, CA, USA
| |
Collapse
|
27
|
Nguyen PV, Gelinas JN. Noradrenergic gating of long-lasting synaptic potentiation in the hippocampus: from neurobiology to translational biomedicine. J Neurogenet 2018; 32:171-182. [DOI: 10.1080/01677063.2018.1497630] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Affiliation(s)
- Peter V. Nguyen
- Department of Physiology and Institute of Neuroscience & Mental Health, University of Alberta School of Medicine, Edmonton, Canada
| | - Jennifer N. Gelinas
- Department of Neurology and Institute for Genomic Medicine, College of Physicians & Surgeons of Columbia University, New York, NY,USA
| |
Collapse
|
28
|
Clocking In Time to Gate Memory Processes: The Circadian Clock Is Part of the Ins and Outs of Memory. Neural Plast 2018; 2018:6238989. [PMID: 29849561 PMCID: PMC5925033 DOI: 10.1155/2018/6238989] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Revised: 01/22/2018] [Accepted: 02/05/2018] [Indexed: 01/11/2023] Open
Abstract
Learning, memory consolidation, and retrieval are processes known to be modulated by the circadian (circa: about; dies: day) system. The circadian regulation of memory performance is evolutionarily conserved, independent of the type and complexity of the learning paradigm tested, and not specific to crepuscular, nocturnal, or diurnal organisms. In mammals, long-term memory (LTM) formation is tightly coupled to de novo gene expression of plasticity-related proteins and posttranslational modifications and relies on intact cAMP/protein kinase A (PKA)/protein kinase C (PKC)/mitogen-activated protein kinase (MAPK)/cyclic adenosine monophosphate response element-binding protein (CREB) signaling. These memory-essential signaling components cycle rhythmically in the hippocampus across the day and night and are clearly molded by an intricate interplay between the circadian system and memory. Important components of the circadian timing mechanism and its plasticity are members of the Period clock gene family (Per1, Per2). Interestingly, Per1 is rhythmically expressed in mouse hippocampus. Observations suggest important and largely unexplored roles of the clock gene protein PER1 in synaptic plasticity and in the daytime-dependent modulation of learning and memory. Here, we review the latest findings on the role of the clock gene Period 1 (Per1) as a candidate molecular and mechanistic blueprint for gating the daytime dependency of memory processing.
Collapse
|
29
|
Wild AR, Dell'Acqua ML. Potential for therapeutic targeting of AKAP signaling complexes in nervous system disorders. Pharmacol Ther 2017; 185:99-121. [PMID: 29262295 DOI: 10.1016/j.pharmthera.2017.12.004] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
A common feature of neurological and neuropsychiatric disorders is a breakdown in the integrity of intracellular signal transduction pathways. Dysregulation of ion channels and receptors in the cell membrane and the enzymatic mediators that link them to intracellular effectors can lead to synaptic dysfunction and neuronal death. However, therapeutic targeting of these ubiquitous signaling elements can lead to off-target side effects due to their widespread expression in multiple systems of the body. A-kinase anchoring proteins (AKAPs) are multivalent scaffolding proteins that compartmentalize a diverse range of receptor and effector proteins to streamline signaling within nanodomain signalosomes. A number of essential neurological processes are known to critically depend on AKAP-directed signaling and an understanding of the role AKAPs play in nervous system disorders has emerged in recent years. Selective targeting of AKAP protein-protein interactions may be a means to uncouple pathologically active signaling pathways in neurological disorders with a greater degree of specificity. In this review we will discuss the role of AKAPs in both regulating normal nervous system function and dysfunction associated with disease, and the potential for therapeutic targeting of AKAP signaling complexes.
Collapse
Affiliation(s)
- Angela R Wild
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Mark L Dell'Acqua
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO 80045, USA.
| |
Collapse
|
30
|
Reggi E, Diviani D. The role of A-kinase anchoring proteins in cancer development. Cell Signal 2017; 40:143-155. [DOI: 10.1016/j.cellsig.2017.09.011] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Revised: 09/08/2017] [Accepted: 09/14/2017] [Indexed: 02/06/2023]
|
31
|
Park AJ, Havekes R, Fu X, Hansen R, Tudor JC, Peixoto L, Li Z, Wu YC, Poplawski SG, Baraban JM, Abel T. Learning induces the translin/trax RNase complex to express activin receptors for persistent memory. eLife 2017; 6. [PMID: 28927503 PMCID: PMC5606845 DOI: 10.7554/elife.27872] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Accepted: 09/01/2017] [Indexed: 12/16/2022] Open
Abstract
Long-lasting forms of synaptic plasticity and memory require de novo protein synthesis. Yet, how learning triggers this process to form memory is unclear. Translin/trax is a candidate to drive this learning-induced memory mechanism by suppressing microRNA-mediated translational silencing at activated synapses. We find that mice lacking translin/trax display defects in synaptic tagging, which requires protein synthesis at activated synapses, and long-term memory. Hippocampal samples harvested from these mice following learning show increases in several disease-related microRNAs targeting the activin A receptor type 1C (ACVR1C), a component of the transforming growth factor-β receptor superfamily. Furthermore, the absence of translin/trax abolishes synaptic upregulation of ACVR1C protein after learning. Finally, synaptic tagging and long-term memory deficits in mice lacking translin/trax are mimicked by ACVR1C inhibition. Thus, we define a new memory mechanism by which learning reverses microRNA-mediated silencing of the novel plasticity protein ACVR1C via translin/trax.
Collapse
Affiliation(s)
- Alan Jung Park
- Department of Biology, University of Pennsylvania, Philadelphia, United States
| | - Robbert Havekes
- Department of Biology, University of Pennsylvania, Philadelphia, United States
| | - Xiuping Fu
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, United States
| | - Rolf Hansen
- Department of Biology, University of Pennsylvania, Philadelphia, United States
| | - Jennifer C Tudor
- Department of Biology, University of Pennsylvania, Philadelphia, United States
| | - Lucia Peixoto
- Department of Biology, University of Pennsylvania, Philadelphia, United States
| | - Zhi Li
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, United States
| | - Yen-Ching Wu
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, United States
| | - Shane G Poplawski
- Department of Biology, University of Pennsylvania, Philadelphia, United States
| | - Jay M Baraban
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, United States
| | - Ted Abel
- Department of Biology, University of Pennsylvania, Philadelphia, United States.,Molecular Physiology and Biophysics, Iowa Neuroscience Institute, Carver College of Medicine, University of Iowa, Iowa City, Iowa, United States
| |
Collapse
|
32
|
Jȩdrzejewska-Szmek J, Luczak V, Abel T, Blackwell KT. β-adrenergic signaling broadly contributes to LTP induction. PLoS Comput Biol 2017; 13:e1005657. [PMID: 28742159 PMCID: PMC5546712 DOI: 10.1371/journal.pcbi.1005657] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Revised: 08/07/2017] [Accepted: 06/27/2017] [Indexed: 12/18/2022] Open
Abstract
Long-lasting forms of long-term potentiation (LTP) represent one of the major cellular mechanisms underlying learning and memory. One of the fundamental questions in the field of LTP is why different molecules are critical for long-lasting forms of LTP induced by diverse experimental protocols. Further complexity stems from spatial aspects of signaling networks, such that some molecules function in the dendrite and some are critical in the spine. We investigated whether the diverse experimental evidence can be unified by creating a spatial, mechanistic model of multiple signaling pathways in hippocampal CA1 neurons. Our results show that the combination of activity of several key kinases can predict the occurrence of long-lasting forms of LTP for multiple experimental protocols. Specifically Ca2+/calmodulin activated kinase II, protein kinase A and exchange protein activated by cAMP (Epac) together predict the occurrence of LTP in response to strong stimulation (multiple trains of 100 Hz) or weak stimulation augmented by isoproterenol. Furthermore, our analysis suggests that activation of the β-adrenergic receptor either via canonical (Gs-coupled) or non-canonical (Gi-coupled) pathways underpins most forms of long-lasting LTP. Simulations make the experimentally testable prediction that a complete antagonist of the β-adrenergic receptor will likely block long-lasting LTP in response to strong stimulation. Collectively these results suggest that converging molecular mechanisms allow CA1 neurons to flexibly utilize signaling mechanisms best tuned to temporal pattern of synaptic input to achieve long-lasting LTP and memory storage. Long-term potentiation of the strength of synaptic connections is a mechanism of learning and memory storage. One of the most confusing aspects of hippocampal synaptic potentiation is that numerous experiments have revealed the requirement for a plethora of signaling molecules. Furthermore the degree to which molecules activated by the stress response modify hippocampal synaptic potentiation and memory is still unclear. We used a computational model to demonstrate that this molecular diversity can be explained by considering a combination of several key molecules. We also show that activation of β-adrenergic receptors by the stress response appears to be involved in most forms of synaptic potentiation, though in some cases unconventional mechanisms are utilized. This suggests that novel treatments for stress-related disorders may have more success if they target unconventional mechanisms activated by β-adrenergic receptors.
Collapse
Affiliation(s)
- Joanna Jȩdrzejewska-Szmek
- The Krasnow Institute for Advanced Studies, George Mason University, Fairfax, Virginia, United States of America
| | - Vincent Luczak
- Department of Biology, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Ted Abel
- Department of Biology, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Kim T Blackwell
- The Krasnow Institute for Advanced Studies, George Mason University, Fairfax, Virginia, United States of America
- * E-mail:
| |
Collapse
|
33
|
Compartmentalized PDE4A5 Signaling Impairs Hippocampal Synaptic Plasticity and Long-Term Memory. J Neurosci 2017; 36:8936-46. [PMID: 27559174 DOI: 10.1523/jneurosci.0248-16.2016] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2016] [Accepted: 06/29/2016] [Indexed: 12/21/2022] Open
Abstract
UNLABELLED Alterations in cAMP signaling are thought to contribute to neurocognitive and neuropsychiatric disorders. Members of the cAMP-specific phosphodiesterase 4 (PDE4) family, which contains >25 different isoforms, play a key role in determining spatial cAMP degradation so as to orchestrate compartmentalized cAMP signaling in cells. Each isoform binds to a different set of protein complexes through its unique N-terminal domain, thereby leading to targeted degradation of cAMP in specific intracellular compartments. However, the functional role of specific compartmentalized PDE4 isoforms has not been examined in vivo Here, we show that increasing protein levels of the PDE4A5 isoform in mouse hippocampal excitatory neurons impairs a long-lasting form of hippocampal synaptic plasticity and attenuates hippocampus-dependent long-term memories without affecting anxiety. In contrast, viral expression of a truncated version of PDE4A5, which lacks the unique N-terminal targeting domain, does not affect long-term memory. Further, overexpression of the PDE4A1 isoform, which targets a different subset of signalosomes, leaves memory undisturbed. Fluorescence resonance energy transfer sensor-based cAMP measurements reveal that the full-length PDE4A5, in contrast to the truncated form, hampers forskolin-mediated increases in neuronal cAMP levels. Our study indicates that the unique N-terminal localization domain of PDE4A5 is essential for the targeting of specific cAMP-dependent signaling underlying synaptic plasticity and memory. The development of compounds to disrupt the compartmentalization of individual PDE4 isoforms by targeting their unique N-terminal domains may provide a fruitful approach to prevent cognitive deficits in neuropsychiatric and neurocognitive disorders that are associated with alterations in cAMP signaling. SIGNIFICANCE STATEMENT Neurons exhibit localized signaling processes that enable biochemical cascades to be activated selectively in specific subcellular compartments. The phosphodiesterase 4 (PDE4) family coordinates the degradation of cAMP, leading to the local attenuation of cAMP-dependent signaling pathways. Sleep deprivation leads to increased hippocampal expression of the PDE4A5 isoform. Here, we explored whether PDE4A5 overexpression mimics behavioral and synaptic plasticity phenotypes associated with sleep deprivation. Viral expression of PDE4A5 in hippocampal neurons impairs long-term potentiation and attenuates the formation of hippocampus-dependent long-term memories. Our findings suggest that PDE4A5 is a molecular constraint on cognitive processes and may contribute to the development of novel therapeutic approaches to prevent cognitive deficits in neuropsychiatric and neurocognitive disorders that are associated with alterations in cAMP signaling.
Collapse
|
34
|
Li Z, Singh S, Suryavanshi SV, Ding W, Shen X, Wijaya CS, Gao WD, McConnell BK. Force development and intracellular Ca 2+ in intact cardiac muscles from gravin mutant mice. Eur J Pharmacol 2017; 807:117-126. [PMID: 28428008 DOI: 10.1016/j.ejphar.2017.04.020] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Revised: 04/13/2017] [Accepted: 04/13/2017] [Indexed: 02/08/2023]
Abstract
Gravin (AKAP12) is an A-kinase-anchoring-protein that scaffolds protein kinase A (PKA), β2-adrenergic receptor (β2-AR), protein phosphatase 2B and protein kinase C. Gravin facilitates β2-AR-dependent signal transduction through PKA to modulate cardiac excitation-contraction coupling and its removal positively affects cardiac contraction. Trabeculae from the right ventricles of gravin mutant (gravin-t/t) mice were employed for force determination. Simultaneously, corresponding intracellular Ca2+ transient ([Ca2+]i) were measured. Twitch force (Tf)-interval relationship, [Ca2+]i-interval relationship, and the rate of decay of post-extrasysolic potentiation (Rf) were also obtained. Western blot analysis were performed to correlate sarcomeric protein expression with alterations in calcium cycling between the WT and gravin-t/t hearts. Gravin-t/t muscles had similar developed force compared to WT muscles despite having lower [Ca2+]i at any given external Ca2+ concentration ([Ca2+]o). The time to peak force and peak [Ca2+]i were slower and the time to 75% relaxation was significantly prolonged in gravin-t/t muscles. Both Tf-interval and [Ca2+]i-interval relations were depressed in gravin-t/t muscles. Rf, however, did not change. Furthermore, Western blot analysis revealed decreased ryanodine receptor (RyR2) phosphorylation in gravin-t/t hearts. Gravin-t/t cardiac muscle exhibits increased force development in responsiveness to Ca2+. The Ca2+ cycling across the SR appears to be unaltered in gravin-t/t muscle. Our study suggests that gravin is an important component of cardiac contraction regulation via increasing myofilament sensitivity to calcium. Further elucidation of the mechanism can provide insights to role of gravin if any in the pathophysiology of impaired contractility.
Collapse
Affiliation(s)
- Zhitao Li
- Department of Pathophysiology, Harbin Medical University, Heilongjiang, China
| | - Sonal Singh
- Department of Pharmacotherapy and Translational Research, College of Pharmacy, University of Florida, Gainesville, FL, USA
| | - Santosh V Suryavanshi
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Texas Medical Center, Houston, TX, USA
| | - Wengang Ding
- Department of Anesthesiology of 2nd Affiliated Hospital, Harbin Medical University, Heilongjiang, China
| | - Xiaoxu Shen
- Cardiology Department, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
| | - Cori S Wijaya
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Texas Medical Center, Houston, TX, USA
| | - Wei Dong Gao
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, 1800 Orleans Street, Zaye Tower 6208, Baltimore, MD 21287, USA.
| | - Bradley K McConnell
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Texas Medical Center, Houston, TX, USA.
| |
Collapse
|
35
|
Bolger GB. The PDE4 cAMP-Specific Phosphodiesterases: Targets for Drugs with Antidepressant and Memory-Enhancing Action. ADVANCES IN NEUROBIOLOGY 2017; 17:63-102. [PMID: 28956330 DOI: 10.1007/978-3-319-58811-7_4] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The PDE4 cyclic nucleotide phosphodiesterases are essential regulators of cAMP abundance in the CNS through their ability to regulate PKA activity, the phosphorylation of CREB, and other important elements of signal transduction. In pre-clinical models and in early-stage clinical trials, PDE4 inhibitors have been shown to have antidepressant and memory-enhancing activity. However, the development of clinically-useful PDE4 inhibitors for CNS disorders has been limited by variable efficacy and significant side effects. Recent structural studies have greatly enhanced our understanding of the molecular configuration of PDE4 enzymes, especially the "long" PDE4 isoforms that are abundant in the CNS. The new structural data provide a rationale for the development of a new generation of PDE4 inhibitors that specifically act on long PDE4 isoforms. These next generation PDE4 inhibitors may also be capable of targeting the interactions of select long forms with their "partner" proteins, such as RACK1, β-arrestin, and DISC1. They would therefore have the ability to affect cAMP levels in specific cellular compartments and target localized cellular functions, such as synaptic plasticity. These new agents might also be able to target PDE4 populations in select regions of the CNS that are implicated in learning and memory, affect, and cognition. Potential therapeutic uses of these agents could include affective disorders, memory enhancement, and neurogenesis.
Collapse
Affiliation(s)
- Graeme B Bolger
- Departments of Medicine and Pharmacology, University of Alabama at Birmingham, 1720 2nd Avenue South, NP 2501, Birmingham, AL, 35294-3300, USA.
| |
Collapse
|
36
|
Jones BW, Deem J, Younts TJ, Weisenhaus M, Sanford CA, Slack MC, Chin J, Nachmanson D, McKennon A, Castillo PE, McKnight GS. Targeted deletion of AKAP7 in dentate granule cells impairs spatial discrimination. eLife 2016; 5. [PMID: 27911261 PMCID: PMC5135391 DOI: 10.7554/elife.20695] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Accepted: 11/23/2016] [Indexed: 01/26/2023] Open
Abstract
Protein Kinase A (PKA) mediates synaptic plasticity and is widely implicated in learning and memory. The hippocampal dentate gyrus (DG) is thought to be responsible for processing and encoding distinct contextual associations in response to highly similar inputs. The mossy fiber (MF) axons of the dentate granule cells convey strong excitatory drive to CA3 pyramidal neurons and express presynaptic, PKA-dependent forms of plasticity. Here, we demonstrate an essential role for the PKA anchoring protein, AKAP7, in mouse MF axons and terminals. Genetic ablation of AKAP7 specifically from dentate granule cells results in disruption of MF-CA3 LTP directly initiated by cAMP, and the AKAP7 mutant mice are selectively deficient in pattern separation behaviors. Our results suggest that the AKAP7/PKA complex in the MF projections plays an essential role in synaptic plasticity and contextual memory formation. DOI:http://dx.doi.org/10.7554/eLife.20695.001
Collapse
Affiliation(s)
- Brian W Jones
- Department of Pharmacology, University of Washington School of Medicine, Seattle, United States
| | - Jennifer Deem
- Department of Pharmacology, University of Washington School of Medicine, Seattle, United States
| | - Thomas J Younts
- Dominick P Purpura Department of Neuroscience, Albert Einstein College of Medicine, New York, United States
| | - Michael Weisenhaus
- Department of Pharmacology, University of Washington School of Medicine, Seattle, United States
| | - Christina A Sanford
- Department of Pharmacology, University of Washington School of Medicine, Seattle, United States
| | - Margaret C Slack
- Department of Pharmacology, University of Washington School of Medicine, Seattle, United States
| | - Jenesa Chin
- Department of Pharmacology, University of Washington School of Medicine, Seattle, United States
| | - Daniela Nachmanson
- Department of Pharmacology, University of Washington School of Medicine, Seattle, United States
| | - Alex McKennon
- Department of Pharmacology, University of Washington School of Medicine, Seattle, United States
| | - Pablo E Castillo
- Dominick P Purpura Department of Neuroscience, Albert Einstein College of Medicine, New York, United States
| | - G Stanley McKnight
- Department of Pharmacology, University of Washington School of Medicine, Seattle, United States
| |
Collapse
|
37
|
Havekes R, Park AJ, Tudor JC, Luczak VG, Hansen RT, Ferri SL, Bruinenberg VM, Poplawski SG, Day JP, Aton SJ, Radwańska K, Meerlo P, Houslay MD, Baillie GS, Abel T. Sleep deprivation causes memory deficits by negatively impacting neuronal connectivity in hippocampal area CA1. eLife 2016; 5. [PMID: 27549340 PMCID: PMC4996653 DOI: 10.7554/elife.13424] [Citation(s) in RCA: 189] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Accepted: 07/29/2016] [Indexed: 12/18/2022] Open
Abstract
Brief periods of sleep loss have long-lasting consequences such as impaired memory consolidation. Structural changes in synaptic connectivity have been proposed as a substrate of memory storage. Here, we examine the impact of brief periods of sleep deprivation on dendritic structure. In mice, we find that five hours of sleep deprivation decreases dendritic spine numbers selectively in hippocampal area CA1 and increased activity of the filamentous actin severing protein cofilin. Recovery sleep normalizes these structural alterations. Suppression of cofilin function prevents spine loss, deficits in hippocampal synaptic plasticity, and impairments in long-term memory caused by sleep deprivation. The elevated cofilin activity is caused by cAMP-degrading phosphodiesterase-4A5 (PDE4A5), which hampers cAMP-PKA-LIMK signaling. Attenuating PDE4A5 function prevents changes in cAMP-PKA-LIMK-cofilin signaling and cognitive deficits associated with sleep deprivation. Our work demonstrates the necessity of an intact cAMP-PDE4-PKA-LIMK-cofilin activation-signaling pathway for sleep deprivation-induced memory disruption and reduction in hippocampal spine density. DOI:http://dx.doi.org/10.7554/eLife.13424.001 The demands of modern society means that millions of people do not get sufficient sleep on a daily basis. Sleep deprivation, even if only for brief periods, can impair learning and memory. In many cases, this impairment appears to be related to changes in the activity of a brain region called the hippocampus. However, the exact processes responsible for producing the effects of sleep deprivation remain unclear. During learning or forming a new memory, the connections between the relevant neurons in the brain change. Havekes et al. found that depriving mice of sleep for just five hours dramatically reduced the connectivity between neurons in the hippocampus. This reduction is caused by the increased activity of cofilin, a protein that breaks down the actin filaments that shape the connections between neurons. Havekes et al. then used a virus to introduce an inactive version of cofilin into hippocampal neurons to suppress the activity of the naturally present cofilin. This manipulation prevented both the loss of the connections between neurons and the memory deficits normally associated with sleep deprivation. Havekes et al. also found that recovery sleep leads to the re-wiring of neurons in the hippocampus. Future studies are now needed to determine how the neurons are able to re-wire themselves during recovery sleep. DOI:http://dx.doi.org/10.7554/eLife.13424.002
Collapse
Affiliation(s)
- Robbert Havekes
- Department of Biology, University of Pennsylvania, Philadelphia, United States.,Groningen Institute for Evolutionary Life Sciences (GELIFES), University of Groningen, Groningen, The Netherlands
| | - Alan J Park
- Department of Biology, University of Pennsylvania, Philadelphia, United States
| | - Jennifer C Tudor
- Department of Biology, University of Pennsylvania, Philadelphia, United States
| | - Vincent G Luczak
- Department of Biology, University of Pennsylvania, Philadelphia, United States
| | - Rolf T Hansen
- Department of Biology, University of Pennsylvania, Philadelphia, United States
| | - Sarah L Ferri
- Department of Biology, University of Pennsylvania, Philadelphia, United States
| | - Vibeke M Bruinenberg
- Groningen Institute for Evolutionary Life Sciences (GELIFES), University of Groningen, Groningen, The Netherlands
| | - Shane G Poplawski
- Department of Biology, University of Pennsylvania, Philadelphia, United States
| | - Jonathan P Day
- Institute of Cardiovascular and Medical Science, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Sara J Aton
- LSA Molecular, Cellular, and Developmental Biology, University of Michigan-Ann Arbor, Ann Arbor, United States
| | - Kasia Radwańska
- Laboratory of Molecular Basis of Behavior, Head Nencki Institute of Experimental Biology, Warsaw, Poland
| | - Peter Meerlo
- Groningen Institute for Evolutionary Life Sciences (GELIFES), University of Groningen, Groningen, The Netherlands
| | - Miles D Houslay
- Institute of Pharmaceutical Science, King's College London, London, United Kingdom
| | - George S Baillie
- Institute of Cardiovascular and Medical Science, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Ted Abel
- Department of Biology, University of Pennsylvania, Philadelphia, United States
| |
Collapse
|
38
|
Yao G, Yun Y, Sang N. Differential effects between one week and four weeks exposure to same mass of SO2 on synaptic plasticity in rat hippocampus. ENVIRONMENTAL TOXICOLOGY 2016; 31:820-829. [PMID: 25534910 DOI: 10.1002/tox.22093] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/02/2014] [Revised: 12/02/2014] [Accepted: 12/07/2014] [Indexed: 06/04/2023]
Abstract
Sulfur dioxide (SO2 ) is a ubiquitous air pollutant. The previous studies have documented the adverse effects of SO2 on nervous system health, suggesting that acutely SO2 inhalation at high concentration may be associated with neurotoxicity and increase risk of hospitalization and mortality of many brain disorders. However, the remarkable features of air pollution exposure are lifelong duration and at low concentration; and it is rarely reported that whether there are different responses on synapse when rats inhaled same mass of SO2 at low concentration with a longer term. In this study, we evaluated the synaptic plasticity in rat hippocampus after exposure to same mass of SO2 at various concentrations and durations (3.5 and 7 mg/m(3) , 6 h/day, for 4 weeks; and 14 and 28 mg/m(3) , 6 h/day, for 1 week). The results showed that the mRNA level of synaptic plasticity marker Arc, glutamate receptors (GRIA1, GRIA2, GRIN1, GRIN2A, and GRIN2B) and the protein expression of memory related kinase p-CaMKпα were consistently inhibited by SO2 both in 1 week and 4 weeks exposure cases; the protein expression of presynaptic marker synaptophysin, postsynaptic density protein 95 (PSD-95), protein kinase A (PKA), and protein kinase C (PKC) were increased in 1 week exposure case, and decreased in 4 weeks exposure case. Our results indicated that SO2 inhalation caused differential synaptic injury in 1 week and 4 weeks exposure cases, and implied the differential effects might result from different PKA- and/or PKC-mediated signal pathway. © 2014 Wiley Periodicals, Inc. Environ Toxicol 31: 820-829, 2016.
Collapse
Affiliation(s)
- Gaoyi Yao
- College of Environment and Resource, Research Center of Environment and Health, Institute of Environmental Science, Shanxi University, Taiyuan, Shanxi, 030006, People's Republic of China
| | - Yang Yun
- College of Environment and Resource, Research Center of Environment and Health, Institute of Environmental Science, Shanxi University, Taiyuan, Shanxi, 030006, People's Republic of China
| | - Nan Sang
- College of Environment and Resource, Research Center of Environment and Health, Institute of Environmental Science, Shanxi University, Taiyuan, Shanxi, 030006, People's Republic of China
| |
Collapse
|
39
|
Persistent Electrical Activity in Primary Nociceptors after Spinal Cord Injury Is Maintained by Scaffolded Adenylyl Cyclase and Protein Kinase A and Is Associated with Altered Adenylyl Cyclase Regulation. J Neurosci 2016; 36:1660-8. [PMID: 26843647 DOI: 10.1523/jneurosci.0895-15.2016] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
UNLABELLED Little is known about intracellular signaling mechanisms that persistently excite neurons in pain pathways. Persistent spontaneous activity (SA) generated in the cell bodies of primary nociceptors within dorsal root ganglia (DRG) has been found to make major contributions to chronic pain in a rat model of spinal cord injury (SCI) (Bedi et al., 2010; Yang et al., 2014). The occurrence of SCI-induced SA in a large fraction of DRG neurons and the persistence of this SA long after dissociation of the neurons provide an opportunity to define intrinsic cell signaling mechanisms that chronically drive SA in pain pathways. The present study demonstrates that SCI-induced SA requires continuing activity of adenylyl cyclase (AC) and cAMP-dependent protein kinase (PKA), as well as a scaffolded complex containing AC5/6, A-kinase anchoring protein 150 (AKAP150), and PKA. SCI caused a small but significant increase in the expression of AKAP150 but not other AKAPs. DRG membranes isolated from SCI animals revealed a novel alteration in the regulation of AC. AC activity stimulated by Ca(2+)-calmodulin increased, while the inhibition of AC activity by Gαi showed an unexpected and dramatic decrease after SCI. Localized enhancement of the activity of AC within scaffolded complexes containing PKA is likely to contribute to chronic pathophysiological consequences of SCI, including pain, that are promoted by persistent hyperactivity in DRG neurons. SIGNIFICANCE STATEMENT Chronic neuropathic pain is a major clinical problem with poorly understood mechanisms and inadequate treatments. Recent findings indicate that chronic pain in a rat SCI model depends upon hyperactivity in dorsal root ganglia (DRG) neurons. Although cAMP signaling is involved in many forms of neural plasticity, including hypersensitivity of nociceptors in the presence of inflammatory mediators, our finding that continuing cAMP-PKA signaling is required for persistent SA months after SCI and long after isolation of nociceptors is surprising. The dependence of ongoing SA upon AKAP150 and AC5/6 was unknown. The discovery of a dramatic decrease in Gαi inhibition of AC activity after SCI is novel for any physiological system and potentially has broad implications for understanding chronic pain mechanisms.
Collapse
|
40
|
Tudor JC, Davis EJ, Peixoto L, Wimmer ME, van Tilborg E, Park AJ, Poplawski SG, Chung CW, Havekes R, Huang J, Gatti E, Pierre P, Abel T. Sleep deprivation impairs memory by attenuating mTORC1-dependent protein synthesis. Sci Signal 2016; 9:ra41. [PMID: 27117251 DOI: 10.1126/scisignal.aad4949] [Citation(s) in RCA: 107] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Sleep deprivation is a public health epidemic that causes wide-ranging deleterious consequences, including impaired memory and cognition. Protein synthesis in hippocampal neurons promotes memory and cognition. The kinase complex mammalian target of rapamycin complex 1 (mTORC1) stimulates protein synthesis by phosphorylating and inhibiting the eukaryotic translation initiation factor 4E-binding protein 2 (4EBP2). We investigated the involvement of the mTORC1-4EBP2 axis in the molecular mechanisms mediating the cognitive deficits caused by sleep deprivation in mice. Using an in vivo protein translation assay, we found that loss of sleep impaired protein synthesis in the hippocampus. Five hours of sleep loss attenuated both mTORC1-mediated phosphorylation of 4EBP2 and the interaction between eukaryotic initiation factor 4E (eIF4E) and eIF4G in the hippocampi of sleep-deprived mice. Increasing the abundance of 4EBP2 in hippocampal excitatory neurons before sleep deprivation increased the abundance of phosphorylated 4EBP2, restored the amount of eIF4E-eIF4G interaction and hippocampal protein synthesis to that seen in mice that were not sleep-deprived, and prevented the hippocampus-dependent memory deficits associated with sleep loss. These findings collectively demonstrate that 4EBP2-regulated protein synthesis is a critical mediator of the memory deficits caused by sleep deprivation.
Collapse
Affiliation(s)
- Jennifer C Tudor
- Department of Biology, School of Arts and Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Emily J Davis
- Department of Biology, School of Arts and Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Lucia Peixoto
- Department of Biology, School of Arts and Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Mathieu E Wimmer
- Department of Biology, School of Arts and Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Erik van Tilborg
- Department of Biology, School of Arts and Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Alan J Park
- Department of Biology, School of Arts and Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Shane G Poplawski
- Department of Biology, School of Arts and Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Caroline W Chung
- Department of Biology, School of Arts and Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Robbert Havekes
- Department of Biology, School of Arts and Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jiayan Huang
- Global Statistical Science, Eli Lilly and Company, Lilly Corporate Center, Indianapolis, IN 46285, USA
| | - Evelina Gatti
- Centre d'Immunologie de Marseille-Luminy, Aix Marseille Université UM2, INSERM U1104, CNRS UMR7280, 13288 Marseille, France. Institute for Research in Biomedicine (iBiMED) and Aveiro Health Sciences Program, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Philippe Pierre
- Centre d'Immunologie de Marseille-Luminy, Aix Marseille Université UM2, INSERM U1104, CNRS UMR7280, 13288 Marseille, France. Institute for Research in Biomedicine (iBiMED) and Aveiro Health Sciences Program, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Ted Abel
- Department of Biology, School of Arts and Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
41
|
Chay A, Zamparo I, Koschinski A, Zaccolo M, Blackwell KT. Control of βAR- and N-methyl-D-aspartate (NMDA) Receptor-Dependent cAMP Dynamics in Hippocampal Neurons. PLoS Comput Biol 2016; 12:e1004735. [PMID: 26901880 PMCID: PMC4763502 DOI: 10.1371/journal.pcbi.1004735] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2014] [Accepted: 01/05/2016] [Indexed: 11/18/2022] Open
Abstract
Norepinephrine, a neuromodulator that activates β-adrenergic receptors (βARs), facilitates learning and memory as well as the induction of synaptic plasticity in the hippocampus. Several forms of long-term potentiation (LTP) at the Schaffer collateral CA1 synapse require stimulation of both βARs and N-methyl-D-aspartate receptors (NMDARs). To understand the mechanisms mediating the interactions between βAR and NMDAR signaling pathways, we combined FRET imaging of cAMP in hippocampal neuron cultures with spatial mechanistic modeling of signaling pathways in the CA1 pyramidal neuron. Previous work implied that cAMP is synergistically produced in the presence of the βAR agonist isoproterenol and intracellular calcium. In contrast, we show that when application of isoproterenol precedes application of NMDA by several minutes, as is typical of βAR-facilitated LTP experiments, the average amplitude of the cAMP response to NMDA is attenuated compared with the response to NMDA alone. Models simulations suggest that, although the negative feedback loop formed by cAMP, cAMP-dependent protein kinase (PKA), and type 4 phosphodiesterase may be involved in attenuating the cAMP response to NMDA, it is insufficient to explain the range of experimental observations. Instead, attenuation of the cAMP response requires mechanisms upstream of adenylyl cyclase. Our model demonstrates that Gs-to-Gi switching due to PKA phosphorylation of βARs as well as Gi inhibition of type 1 adenylyl cyclase may underlie the experimental observations. This suggests that signaling by β-adrenergic receptors depends on temporal pattern of stimulation, and that switching may represent a novel mechanism for recruiting kinases involved in synaptic plasticity and memory. Noradrenaline is a stress related molecule that facilitates learning and memory when released in the hippocampus. The facilitation of memory is related to modulation of synaptic plasticity, but the mechanisms underlying this modulation are not well understood. We utilize a combination of live cell imaging and computational modeling to discover how noradrenergic receptor stimulation interacts with other molecules, such as calcium, required for synaptic plasticity and memory storage. Though prior work has shown that noradrenergic receptors and calcium interact synergistically to elevate intracellular second messengers when combined simultaneously, our results demonstrate that prior stimulation of noradrenergic receptors inhibits the elevation of intracellular second messengers. Our results further demonstrate that the inhibition may be caused by the noradrenergic receptor switching signaling pathways, thereby recruiting a different set of memory kinases. This switching represents a novel mechanism for recruiting molecules involved in synaptic plasticity and memory.
Collapse
Affiliation(s)
- Andrew Chay
- Molecular Neuroscience Department, Krasnow Institute, George Mason University, Fairfax, Virginia, United States of America
| | | | - Andreas Koschinski
- Department of Physiology, Anatomy and Genetics, Oxford University, Oxford, United Kingdom
| | - Manuela Zaccolo
- Department of Physiology, Anatomy and Genetics, Oxford University, Oxford, United Kingdom
| | - Kim T. Blackwell
- Molecular Neuroscience Department, Krasnow Institute, George Mason University, Fairfax, Virginia, United States of America
- * E-mail:
| |
Collapse
|
42
|
Salgado H, Treviño M, Atzori M. Layer- and area-specific actions of norepinephrine on cortical synaptic transmission. Brain Res 2016; 1641:163-76. [PMID: 26820639 DOI: 10.1016/j.brainres.2016.01.033] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2015] [Revised: 01/08/2016] [Accepted: 01/20/2016] [Indexed: 12/20/2022]
Abstract
The cerebral cortex is a critical target of the central noradrenergic system. The importance of norepinephrine (NE) in the regulation of cortical activity is underscored by clinical findings that involve this catecholamine and its receptor subtypes in the regulation of a large number of emotional and cognitive functions and illnesses. In this review, we highlight diverse effects of the LC/NE system in the mammalian cortex. Indeed, electrophysiological, pharmacological, and behavioral studies in the last few decades reveal that NE elicits a mixed repertoire of excitatory, inhibitory, and biphasic effects on the firing activity and transmitter release of cortical neurons. At the intrinsic cellular level, NE can produce a series of effects similar to those elicited by other monoamines or acetylcholine, associated with systemic arousal. At the synaptic level, NE induces numerous acute changes in synaptic function, and ׳gates' the induction of long-term plasticity of glutamatergic synapses, consisting in an enhancement of engaged and relevant cortical synapses and/or depression of unengaged synapses. Equally important in shaping cortical function, in many cortical areas NE promotes a characteristic, most often reversible, increase in the gain of local inhibitory synapses, whose extent and temporal properties vary between different areas and sometimes even between cortical layers of the same area. While we are still a long way from a comprehensive theory of the function of the LC/NE system, its cellular, synaptic, and plastic effects are consistent with the hypothesis that noradrenergic modulation is critical in coordinating the activity of cortical and subcortical circuits for the integration of sensory activity and working memory. This article is part of a Special Issue entitled SI: Noradrenergic System.
Collapse
Affiliation(s)
| | | | - Marco Atzori
- Universidad Autónoma de San Luis Potosí, México.
| |
Collapse
|
43
|
FRET biosensors reveal AKAP-mediated shaping of subcellular PKA activity and a novel mode of Ca(2+)/PKA crosstalk. Cell Signal 2016; 28:294-306. [PMID: 26772752 DOI: 10.1016/j.cellsig.2016.01.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2015] [Revised: 12/18/2015] [Accepted: 01/04/2016] [Indexed: 02/01/2023]
Abstract
Scaffold proteins play a critical role in cellular homeostasis by anchoring signaling enzymes in close proximity to downstream effectors. In addition to anchoring static enzyme complexes, some scaffold proteins also form dynamic signalosomes that can traffic to different subcellular compartments upon stimulation. Gravin (AKAP12), a multivalent scaffold, anchors PKA and other enzymes to the plasma membrane under basal conditions, but upon [Ca(2+)]i elevation, is rapidly redistributed to the cytosol. Because gravin redistribution also impacts PKA localization, we postulate that gravin acts as a calcium "switch" that modulates PKA-substrate interactions at the plasma membrane, thus facilitating a novel crosstalk mechanism between Ca(2+) and PKA-dependent pathways. To assess this, we measured the impact of gravin-V5/His expression on compartmentalized PKA activity using the FRET biosensor AKAR3 in cultured cells. Upon treatment with forskolin or isoproterenol, cells expressing gravin-V5/His showed elevated levels of plasma membrane PKA activity, but cytosolic PKA activity levels were reduced compared with control cells lacking gravin. This effect required both gravin interaction with PKA and localization at the plasma membrane. Pretreatment with calcium-elevating agents thapsigargin or ATP caused gravin redistribution away from the plasma membrane and prevented gravin from elevating PKA activity levels at the membrane. Importantly, this mode of Ca(2+)/PKA crosstalk was not observed in cells expressing a gravin mutant that resisted calcium-mediated redistribution from the cell periphery. These results reveal that gravin impacts subcellular PKA activity levels through the spatial targeting of PKA, and that calcium elevation modulates downstream β-adrenergic/PKA signaling through gravin redistribution, thus supporting the hypothesis that gravin mediates crosstalk between Ca(2+) and PKA-dependent signaling pathways. Based on these results, AKAP localization dynamics may represent an important paradigm for the regulation of cellular signaling networks.
Collapse
|
44
|
Diviani D, Reggi E, Arambasic M, Caso S, Maric D. Emerging roles of A-kinase anchoring proteins in cardiovascular pathophysiology. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2015; 1863:1926-36. [PMID: 26643253 DOI: 10.1016/j.bbamcr.2015.11.024] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Revised: 11/20/2015] [Accepted: 11/23/2015] [Indexed: 01/08/2023]
Abstract
Heart and blood vessels ensure adequate perfusion of peripheral organs with blood and nutrients. Alteration of the homeostatic functions of the cardiovascular system can cause hypertension, atherosclerosis, and coronary artery disease leading to heart injury and failure. A-kinase anchoring proteins (AKAPs) constitute a family of scaffolding proteins that are crucially involved in modulating the function of the cardiovascular system both under physiological and pathological conditions. AKAPs assemble multifunctional signaling complexes that ensure correct targeting of the cAMP-dependent protein kinase (PKA) as well as other signaling enzymes to precise subcellular compartments. This allows local regulation of specific effector proteins that control the function of vascular and cardiac cells. This review will focus on recent advances illustrating the role of AKAPs in cardiovascular pathophysiology. The accent will be mainly placed on the molecular events linked to the control of vascular integrity and blood pressure as well as on the cardiac remodeling process associated with heart failure. This article is part of a Special Issue entitled: Cardiomyocyte Biology: Integration of Developmental and Environmental Cues in the Heart edited by Marcus Schaub and Hughes Abriel.
Collapse
Affiliation(s)
- Dario Diviani
- Département de Pharmacologie et de Toxicologie, Faculté de Biologie et de Médecine, Lausanne 1005, Switzerland.
| | - Erica Reggi
- Département de Pharmacologie et de Toxicologie, Faculté de Biologie et de Médecine, Lausanne 1005, Switzerland
| | - Miroslav Arambasic
- Département de Pharmacologie et de Toxicologie, Faculté de Biologie et de Médecine, Lausanne 1005, Switzerland
| | - Stefania Caso
- Département de Pharmacologie et de Toxicologie, Faculté de Biologie et de Médecine, Lausanne 1005, Switzerland
| | - Darko Maric
- Département de Pharmacologie et de Toxicologie, Faculté de Biologie et de Médecine, Lausanne 1005, Switzerland
| |
Collapse
|
45
|
Woolfrey KM, Dell'Acqua ML. Coordination of Protein Phosphorylation and Dephosphorylation in Synaptic Plasticity. J Biol Chem 2015; 290:28604-12. [PMID: 26453308 DOI: 10.1074/jbc.r115.657262] [Citation(s) in RCA: 98] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
A central theme in nervous system function is equilibrium: synaptic strengths wax and wane, neuronal firing rates adjust up and down, and neural circuits balance excitation with inhibition. This push/pull regulatory theme carries through to the molecular level at excitatory synapses, where protein function is controlled through phosphorylation and dephosphorylation by kinases and phosphatases. However, these opposing enzymatic activities are only part of the equation as scaffolding interactions and assembly of multi-protein complexes are further required for efficient, localized synaptic signaling. This review will focus on coordination of postsynaptic serine/threonine kinase and phosphatase signaling by scaffold proteins during synaptic plasticity.
Collapse
Affiliation(s)
- Kevin M Woolfrey
- From the Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, Colorado 80045
| | - Mark L Dell'Acqua
- From the Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, Colorado 80045
| |
Collapse
|
46
|
Briggs CE, Wang Y, Kong B, Woo TUW, Iyer LK, Sonntag KC. Midbrain dopamine neurons in Parkinson's disease exhibit a dysregulated miRNA and target-gene network. Brain Res 2015; 1618:111-121. [PMID: 26047984 PMCID: PMC4522231 DOI: 10.1016/j.brainres.2015.05.021] [Citation(s) in RCA: 86] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2015] [Revised: 05/09/2015] [Accepted: 05/15/2015] [Indexed: 11/21/2022]
Abstract
The degeneration of substantia nigra (SN) dopamine (DA) neurons in sporadic Parkinson׳s disease (PD) is characterized by disturbed gene expression networks. Micro(mi)RNAs are post-transcriptional regulators of gene expression and we recently provided evidence that these molecules may play a functional role in the pathogenesis of PD. Here, we document a comprehensive analysis of miRNAs in SN DA neurons and PD, including sex differences. Our data show that miRNAs are dysregulated in disease-affected neurons and differentially expressed between male and female samples with a trend of more up-regulated miRNAs in males and more down-regulated miRNAs in females. Unbiased Ingenuity Pathway Analysis (IPA) revealed a network of miRNA/target-gene associations that is consistent with dysfunctional gene and signaling pathways in PD pathology. Our study provides evidence for a general association of miRNAs with the cellular function and identity of SN DA neurons, and with deregulated gene expression networks and signaling pathways related to PD pathogenesis that may be sex-specific.
Collapse
Affiliation(s)
- Christine E Briggs
- The Center of Cancer Systems Biology, Tufts University School of Medicine, Boston, MA 02135, USA
| | - Yulei Wang
- Life Technologies, Foster City, CA 94404, USA
| | | | - Tsung-Ung W Woo
- Department of Psychiatry, McLean Hospital, Harvard Medical School, MRC 223, McLean Hospital, 115 Mill Street, Belmont, MA 02478, USA
| | - Lakshmanan K Iyer
- Department of Neuroscience, Tufts University School of Medicine, Boston, MA 02135, USA
| | - Kai C Sonntag
- Department of Psychiatry, McLean Hospital, Harvard Medical School, MRC 223, McLean Hospital, 115 Mill Street, Belmont, MA 02478, USA.
| |
Collapse
|
47
|
O'Dell TJ, Connor SA, Guglietta R, Nguyen PV. β-Adrenergic receptor signaling and modulation of long-term potentiation in the mammalian hippocampus. ACTA ACUST UNITED AC 2015; 22:461-71. [PMID: 26286656 PMCID: PMC4561407 DOI: 10.1101/lm.031088.113] [Citation(s) in RCA: 101] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Accepted: 06/02/2015] [Indexed: 02/06/2023]
Abstract
Encoding new information in the brain requires changes in synaptic strength. Neuromodulatory transmitters can facilitate synaptic plasticity by modifying the actions and expression of specific signaling cascades, transmitter receptors and their associated signaling complexes, genes, and effector proteins. One critical neuromodulator in the mammalian brain is norepinephrine (NE), which regulates multiple brain functions such as attention, perception, arousal, sleep, learning, and memory. The mammalian hippocampus receives noradrenergic innervation and hippocampal neurons express β-adrenergic receptors, which are known to play important roles in gating the induction of long-lasting forms of synaptic potentiation. These forms of long-term potentiation (LTP) are believed to importantly contribute to long-term storage of spatial and contextual memories in the brain. In this review, we highlight the contributions of noradrenergic signaling in general and β-adrenergic receptors in particular, toward modulating hippocampal LTP. We focus on the roles of NE and β-adrenergic receptors in altering the efficacies of specific signaling molecules such as NMDA and AMPA receptors, protein phosphatases, and translation initiation factors. Also, the roles of β-adrenergic receptors in regulating synaptic "tagging" and "capture" of LTP within synaptic networks of the hippocampus are reviewed. Understanding the molecular and cellular bases of noradrenergic signaling will enrich our grasp of how the brain makes new, enduring memories, and may shed light on credible strategies for improving mental health through treatment of specific disorders linked to perturbed memory processing and dysfunctional noradrenergic synaptic transmission.
Collapse
Affiliation(s)
- Thomas J O'Dell
- Department of Physiology, David Geffen School of Medicine and Integrative Center for Learning and Memory, Brain Research Institute, University of California, Los Angeles, Los Angeles, California 90095, USA
| | - Steven A Connor
- Department of Physiology, University of Alberta School of Medicine, Edmonton, Alberta T6G 2H7, Canada
| | - Ryan Guglietta
- Interdepartmental Ph.D. Program for Neuroscience, University of California, Los Angeles, Los Angeles, California 90095, USA
| | - Peter V Nguyen
- Department of Physiology, University of Alberta School of Medicine, Edmonton, Alberta T6G 2H7, Canada Department of Psychiatry, University of Alberta School of Medicine, Edmonton, Alberta T6G 2H7, Canada Department of Neuroscience & Mental Health Institute, University of Alberta School of Medicine, Edmonton, Alberta T6G 2H7, Canada
| |
Collapse
|
48
|
Sáez-Briones P, Soto-Moyano R, Burgos H, Castillo A, Valladares L, Morgan C, Pérez H, Barra R, Constandil L, Laurido C, Hernández A. β2-Adrenoceptor stimulation restores frontal cortex plasticity and improves visuospatial performance in hidden-prenatally-malnourished young-adult rats. Neurobiol Learn Mem 2015; 119:1-9. [DOI: 10.1016/j.nlm.2014.11.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2014] [Revised: 10/11/2014] [Accepted: 11/04/2014] [Indexed: 10/24/2022]
|
49
|
Yao G, Yue H, Yun Y, Sang N. Chronic SO2 inhalation above environmental standard impairs neuronal behavior and represses glutamate receptor gene expression and memory-related kinase activation via neuroinflammation in rats. ENVIRONMENTAL RESEARCH 2015; 137:85-93. [PMID: 25498917 DOI: 10.1016/j.envres.2014.11.012] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/26/2014] [Revised: 11/19/2014] [Accepted: 11/25/2014] [Indexed: 06/04/2023]
Abstract
Sulfur dioxide (SO2), as a ubiquitous air pollutant implicated in the genesis of pulmonary disease, is now being considered to be involved in neurotoxicity and increased risk for hospitalization of brain disorders. However, comparatively little is known about the impact of chronically SO2 inhalation on neuronal function. In the present study, by exposing male Wistar rats to SO2 at 3.50 and 7.00 mg/m(3) (approximately 1225 and 2450 ppb, 4.08-8.16 (24h average concentration) times higher than the EPA standard for environmental air concentrations) or filtered air for 90 days, we investigated the impact of chronic SO2 inhalation on performance in Morris water maze, and probed the accompanying neurobiological effects, including activity-regulated cytoskeletal associated gene (Arc) and glutamate receptor gene expression, memory-related kinase level and inflammatory cytokine release in the hippocampus. Here, we found that SO2 exposure reduced the number of target zone crossings and time spent in the target quadrant during the test session in the spatial memory retention of the Morris water maze. Following the neuro-functional abnormality, we detected that SO2 inhalation reduced the expression of Arc and glutamate receptor subunits (GluR1, GluR2, NR1, NR2A, and NR2B) with a concentration-dependent property in comparison to controls. Additionally, the expression of memory kinases was attenuated statistically in the animals receiving the higher concentration, including protein kinase A (PKA), protein kinase C (PKC) and calcium/calmodulin-dependent protein kinaseIIα (CaMKIIα). And the inflammatory cytokine release was increased in rats exposed to SO2. Taken together, our results suggest that long-term exposure to SO2 air pollution at concentrations above the environmental standard in rats impaired spatial learning and memory, and indicate a close link between the neurobiological changes highlighted in the brain and the behavioral disturbances.
Collapse
Affiliation(s)
- Gaoyi Yao
- College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan, Shanxi 030006, PR China
| | - Huifeng Yue
- College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan, Shanxi 030006, PR China
| | - Yang Yun
- College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan, Shanxi 030006, PR China
| | - Nan Sang
- College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan, Shanxi 030006, PR China.
| |
Collapse
|
50
|
Transiently increasing cAMP levels selectively in hippocampal excitatory neurons during sleep deprivation prevents memory deficits caused by sleep loss. J Neurosci 2015; 34:15715-21. [PMID: 25411499 DOI: 10.1523/jneurosci.2403-14.2014] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The hippocampus is particularly sensitive to sleep loss. Although previous work has indicated that sleep deprivation impairs hippocampal cAMP signaling, it remains to be determined whether the cognitive deficits associated with sleep deprivation are caused by attenuated cAMP signaling in the hippocampus. Further, it is unclear which cell types are responsible for the memory impairments associated with sleep deprivation. Transgenic approaches lack the spatial resolution to manipulate specific signaling pathways selectively in the hippocampus, while pharmacological strategies are limited in terms of cell-type specificity. Therefore, we used a pharmacogenetic approach based on a virus-mediated expression of a Gαs-coupled Drosophila octopamine receptor selectively in mouse hippocampal excitatory neurons in vivo. With this approach, a systemic injection with the receptor ligand octopamine leads to increased cAMP levels in this specific set of hippocampal neurons. We assessed whether transiently increasing cAMP levels during sleep deprivation prevents memory consolidation deficits associated with sleep loss in an object-location task. Five hours of total sleep deprivation directly following training impaired the formation of object-location memories. Transiently increasing cAMP levels in hippocampal neurons during the course of sleep deprivation prevented these memory consolidation deficits. These findings demonstrate that attenuated cAMP signaling in hippocampal excitatory neurons is a critical component underlying the memory deficits in hippocampus-dependent learning tasks associated with sleep deprivation.
Collapse
|