1
|
Kaufman MJ, Meloni EG. Xenon gas as a potential treatment for opioid use disorder, alcohol use disorder, and related disorders. Med Gas Res 2025; 15:234-253. [PMID: 39812023 PMCID: PMC11918480 DOI: 10.4103/mgr.medgasres-d-24-00063] [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: 08/02/2024] [Revised: 08/23/2024] [Accepted: 09/26/2024] [Indexed: 01/16/2025] Open
Abstract
Xenon gas is considered to be a safe anesthetic and imaging agent. Research on its other potentially beneficial effects suggests that xenon may have broad efficacy for treating health disorders. A number of reviews on xenon applications have been published, but none have focused on substance use disorders. Accordingly, we review xenon effects and targets relevant to the treatment of substance use disorders, with a focus on opioid use disorder and alcohol use disorder. We report that xenon inhaled at subsedative concentrations inhibits conditioned memory reconsolidation and opioid withdrawal symptoms. We review work by others reporting on the antidepressant, anxiolytic, and analgesic properties of xenon, which could diminish negative affective states and pain. We discuss research supporting the possibility that xenon could prevent analgesic- or stress-induced opioid tolerance and, by so doing could reduce the risk of developing opioid use disorder. The rapid kinetics, favorable safety and side effect profiles, and multitargeting capability of xenon suggest that it could be used as an ambulatory on-demand treatment to rapidly attenuate maladaptive memory, physical and affective withdrawal symptoms, and pain drivers of substance use disorders when they occur. Xenon may also have human immunodeficiency virus and oncology applications because its effects relevant to substance use disorders could be exploited to target human immunodeficiency virus reservoirs, human immunodeficiency virus protein-induced abnormalities, and cancers. Although xenon is expensive, low concentrations exert beneficial effects, and gas separation, recovery, and recycling advancements will lower xenon costs, increasing the economic feasibility of its therapeutic use. More research is needed to better understand the remarkable repertoire of effects of xenon and its potential therapeutic applications.
Collapse
|
2
|
Sun Y, Zou Q, Yu H, Yi X, Dou X, Yang Y, Liu Z, Yang H, Jia J, Chen Y, Sun SK, Zhang L. Melanin-like nanoparticles slow cyst growth in ADPKD by dual inhibition of oxidative stress and CREB. EMBO Mol Med 2025; 17:169-192. [PMID: 39567834 PMCID: PMC11730739 DOI: 10.1038/s44321-024-00167-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 10/18/2024] [Accepted: 10/18/2024] [Indexed: 11/22/2024] Open
Abstract
Melanin-like nanoparticles (MNPs) have recently emerged as valuable agents in antioxidant therapy due to their excellent biocompatibility and potent capacity to scavenge various reactive oxygen species (ROS). However, previous studies have mainly focused on acute ROS-related diseases, leaving a knowledge gap regarding their potential in chronic conditions. Furthermore, apart from their well-established antioxidant effects, it remains unclear whether MNPs target other intracellular molecular pathways. In this study, we synthesized ultra-small polyethylene glycol-incorporated Mn2+-chelated MNP (MMPP). We found that MMPP traversed the glomerular filtration barrier and specifically accumulated in renal tubules. Autosomal dominant polycystic kidney disease (ADPKD) is a chronic genetic disorder closely associated with increased oxidative stress and featured by the progressive enlargement of cysts originating from various segments of the renal tubules. Treatment with MMPP markedly attenuated oxidative stress levels, inhibited cyst growth, thereby improving renal function. Interestingly, we found that MMPP effectively inhibits a cyst-promoting gene program downstream of the cAMP-CREB pathway, a crucial signaling pathway implicated in ADPKD progression. Mechanistically, we observed that MMPP directly binds to the bZIP DNA-binding domain of CREB, leading to competitive inhibition of CREB's DNA binding ability and subsequent reduction in CREB target gene expression. In summary, our findings identify an intracellular target of MMPP and demonstrate its potential for treating ADPKD by simultaneously targeting oxidative stress and CREB transcriptional activity.
Collapse
Affiliation(s)
- Yongzhan Sun
- The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), State Key Laboratory of Experimental Hematology, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
- School of Biomedical Engineering and Technology, Tianjin Medical University, Tianjin, China
| | - Quan Zou
- Department of Radiology, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Huizheng Yu
- The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), State Key Laboratory of Experimental Hematology, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Xiaoping Yi
- The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), State Key Laboratory of Experimental Hematology, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Xudan Dou
- The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), State Key Laboratory of Experimental Hematology, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Yu Yang
- Department of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Zhiheng Liu
- The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), State Key Laboratory of Experimental Hematology, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Tianjin Medical University, Tianjin, China
| | - Hong Yang
- Department of Pharmacology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Junya Jia
- Department of Nephrology, Tianjin Medical University General Hospital, Tianjin, China
| | - Yupeng Chen
- The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), State Key Laboratory of Experimental Hematology, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China.
- Department of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China.
| | - Shao-Kai Sun
- School of Medical Imaging, Tianjin Medical University, Tianjin, China.
| | - Lirong Zhang
- The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), State Key Laboratory of Experimental Hematology, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China.
| |
Collapse
|
3
|
Chowdhury MAR, Haq MM, Lee JH, Jeong S. Multi-faceted regulation of CREB family transcription factors. Front Mol Neurosci 2024; 17:1408949. [PMID: 39165717 PMCID: PMC11333461 DOI: 10.3389/fnmol.2024.1408949] [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: 04/04/2024] [Accepted: 07/12/2024] [Indexed: 08/22/2024] Open
Abstract
cAMP response element-binding protein (CREB) is a ubiquitously expressed nuclear transcription factor, which can be constitutively activated regardless of external stimuli or be inducibly activated by external factors such as stressors, hormones, neurotransmitters, and growth factors. However, CREB controls diverse biological processes including cell growth, differentiation, proliferation, survival, apoptosis in a cell-type-specific manner. The diverse functions of CREB appear to be due to CREB-mediated differential gene expression that depends on cAMP response elements and multi-faceted regulation of CREB activity. Indeed, the transcriptional activity of CREB is controlled at several levels including alternative splicing, post-translational modification, dimerization, specific transcriptional co-activators, non-coding small RNAs, and epigenetic regulation. In this review, we present versatile regulatory modes of CREB family transcription factors and discuss their functional consequences.
Collapse
Affiliation(s)
- Md Arifur Rahman Chowdhury
- Department of Bioactive Material Sciences, Jeonbuk National University, Jeonju, Republic of Korea
- Department of Molecular Biology, and Research Center of Bioactive Materials, Jeonbuk National University, Jeonju, Republic of Korea
| | - Md Mazedul Haq
- Department of Bioactive Material Sciences, Jeonbuk National University, Jeonju, Republic of Korea
- Department of Molecular Biology, and Research Center of Bioactive Materials, Jeonbuk National University, Jeonju, Republic of Korea
| | - Jeong Hwan Lee
- Division of Life Sciences, Jeonbuk National University, Jeonju, Republic of Korea
| | - Sangyun Jeong
- Department of Bioactive Material Sciences, Jeonbuk National University, Jeonju, Republic of Korea
- Department of Molecular Biology, and Research Center of Bioactive Materials, Jeonbuk National University, Jeonju, Republic of Korea
| |
Collapse
|
4
|
Bosch M, Kallin N, Donakonda S, Zhang JD, Wintersteller H, Hegenbarth S, Heim K, Ramirez C, Fürst A, Lattouf EI, Feuerherd M, Chattopadhyay S, Kumpesa N, Griesser V, Hoflack JC, Siebourg-Polster J, Mogler C, Swadling L, Pallett LJ, Meiser P, Manske K, de Almeida GP, Kosinska AD, Sandu I, Schneider A, Steinbacher V, Teng Y, Schnabel J, Theis F, Gehring AJ, Boonstra A, Janssen HLA, Vandenbosch M, Cuypers E, Öllinger R, Engleitner T, Rad R, Steiger K, Oxenius A, Lo WL, Klepsch V, Baier G, Holzmann B, Maini MK, Heeren R, Murray PJ, Thimme R, Herrmann C, Protzer U, Böttcher JP, Zehn D, Wohlleber D, Lauer GM, Hofmann M, Luangsay S, Knolle PA. A liver immune rheostat regulates CD8 T cell immunity in chronic HBV infection. Nature 2024; 631:867-875. [PMID: 38987588 PMCID: PMC11269190 DOI: 10.1038/s41586-024-07630-7] [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: 09/19/2022] [Accepted: 05/30/2024] [Indexed: 07/12/2024]
Abstract
Chronic hepatitis B virus (HBV) infection affects 300 million patients worldwide1,2, in whom virus-specific CD8 T cells by still ill-defined mechanisms lose their function and cannot eliminate HBV-infected hepatocytes3-7. Here we demonstrate that a liver immune rheostat renders virus-specific CD8 T cells refractory to activation and leads to their loss of effector functions. In preclinical models of persistent infection with hepatotropic viruses such as HBV, dysfunctional virus-specific CXCR6+ CD8 T cells accumulated in the liver and, as a characteristic hallmark, showed enhanced transcriptional activity of cAMP-responsive element modulator (CREM) distinct from T cell exhaustion. In patients with chronic hepatitis B, circulating and intrahepatic HBV-specific CXCR6+ CD8 T cells with enhanced CREM expression and transcriptional activity were detected at a frequency of 12-22% of HBV-specific CD8 T cells. Knocking out the inhibitory CREM/ICER isoform in T cells, however, failed to rescue T cell immunity. This indicates that CREM activity was a consequence, rather than the cause, of loss in T cell function, further supported by the observation of enhanced phosphorylation of protein kinase A (PKA) which is upstream of CREM. Indeed, we found that enhanced cAMP-PKA-signalling from increased T cell adenylyl cyclase activity augmented CREM activity and curbed T cell activation and effector function in persistent hepatic infection. Mechanistically, CD8 T cells recognizing their antigen on hepatocytes established close and extensive contact with liver sinusoidal endothelial cells, thereby enhancing adenylyl cyclase-cAMP-PKA signalling in T cells. In these hepatic CD8 T cells, which recognize their antigen on hepatocytes, phosphorylation of key signalling kinases of the T cell receptor signalling pathway was impaired, which rendered them refractory to activation. Thus, close contact with liver sinusoidal endothelial cells curbs the activation and effector function of HBV-specific CD8 T cells that target hepatocytes expressing viral antigens by means of the adenylyl cyclase-cAMP-PKA axis in an immune rheostat-like fashion.
Collapse
Affiliation(s)
- Miriam Bosch
- Institute of Molecular Immunology, School of Medicine and Health, Technical University of Munich (TUM), Munich, Germany
| | - Nina Kallin
- Institute of Molecular Immunology, School of Medicine and Health, Technical University of Munich (TUM), Munich, Germany
| | - Sainitin Donakonda
- Institute of Molecular Immunology, School of Medicine and Health, Technical University of Munich (TUM), Munich, Germany
| | - Jitao David Zhang
- Roche Pharmaceutical Research and Early Development (pRED), Roche Innovation Center Basel, Basel, Switzerland
| | - Hannah Wintersteller
- Institute of Molecular Immunology, School of Medicine and Health, Technical University of Munich (TUM), Munich, Germany
| | - Silke Hegenbarth
- Institute of Molecular Immunology, School of Medicine and Health, Technical University of Munich (TUM), Munich, Germany
| | - Kathrin Heim
- Third Department of Medicine, University Hospital Freiburg, Freiburg, Germany
| | - Carlos Ramirez
- Health Data Science Unit, Biomedical Genomics Group, Bioquant, Faculty of Medicine Heidelberg, Heidelberg, Germany
| | - Anna Fürst
- Institute of Molecular Immunology, School of Medicine and Health, Technical University of Munich (TUM), Munich, Germany
| | - Elias Isaac Lattouf
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Martin Feuerherd
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Sutirtha Chattopadhyay
- Institute of Molecular Immunology, School of Medicine and Health, Technical University of Munich (TUM), Munich, Germany
| | - Nadine Kumpesa
- Roche Pharmaceutical Research and Early Development (pRED), Roche Innovation Center Basel, Basel, Switzerland
| | - Vera Griesser
- Roche Pharmaceutical Research and Early Development (pRED), Roche Innovation Center Basel, Basel, Switzerland
| | - Jean-Christophe Hoflack
- Roche Pharmaceutical Research and Early Development (pRED), Roche Innovation Center Basel, Basel, Switzerland
| | - Juliane Siebourg-Polster
- Roche Pharmaceutical Research and Early Development (pRED), Roche Innovation Center Basel, Basel, Switzerland
| | - Carolin Mogler
- Institute of Pathology, School of Medicine and Health, TUM, Munich, Germany
| | - Leo Swadling
- Division of Infection and Immunity, Institute of Immunity and Transplantation, University College London, London, UK
| | - Laura J Pallett
- Division of Infection and Immunity, Institute of Immunity and Transplantation, University College London, London, UK
| | - Philippa Meiser
- Institute of Molecular Immunology, School of Medicine and Health, Technical University of Munich (TUM), Munich, Germany
| | - Katrin Manske
- Institute of Molecular Immunology, School of Medicine and Health, Technical University of Munich (TUM), Munich, Germany
| | - Gustavo P de Almeida
- Institute of Immunology and Animal Physiology, School of Life Science, TUM, Munich, Germany
| | - Anna D Kosinska
- Institute of Virology, School of Medicine and Health, TUM, Munich, Germany
- Helmholtz Zentrum München, Munich, Germany
- German Center for Infection Research, Munich site, Munich, Germany
| | - Ioana Sandu
- Institute of Microbiology, ETH Zürich, Zürich, Switzerland
| | - Annika Schneider
- Institute of Molecular Immunology, School of Medicine and Health, Technical University of Munich (TUM), Munich, Germany
| | - Vincent Steinbacher
- Institute of Molecular Immunology, School of Medicine and Health, Technical University of Munich (TUM), Munich, Germany
| | - Yan Teng
- Institute of Virology, School of Medicine and Health, TUM, Munich, Germany
| | - Julia Schnabel
- Institute of Machine Learning and Biomedical Imaging, Helmholtz Zentrum Munich, Munich, Germany
| | - Fabian Theis
- Institute of Computational Biology, TUM, Munich, Germany
| | - Adam J Gehring
- Toronto Centre for Liver Disease and Toronto General Hospital Research Institute, Toronto, Ontario, Canada
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | - Andre Boonstra
- Department of Gastroenterology and Hepatology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Harry L A Janssen
- Department of Gastroenterology and Hepatology, Erasmus University Medical Center, Rotterdam, The Netherlands
- Toronto General Hospital, University of Toronto, Toronto, Ontario, Canada
| | - Michiel Vandenbosch
- Institute of Multimodal Imaging, University of Maastricht, Maastricht, The Netherlands
| | - Eva Cuypers
- Institute of Multimodal Imaging, University of Maastricht, Maastricht, The Netherlands
| | - Rupert Öllinger
- Institute of Molecular Oncology and Functional Genomics, School of Medicine and Health, TUM, Munich, Germany
| | - Thomas Engleitner
- Institute of Molecular Oncology and Functional Genomics, School of Medicine and Health, TUM, Munich, Germany
| | - Roland Rad
- Institute of Molecular Oncology and Functional Genomics, School of Medicine and Health, TUM, Munich, Germany
| | - Katja Steiger
- Comparative Experimental Pathology, School of Medicine and Health, TUM, Munich, Germany
| | | | - Wan-Lin Lo
- Department of Pathology, University of Utah, Salt Lake City, UT, USA
| | - Victoria Klepsch
- Institute of Cell Genetics, Medical University of Innsbruck, Innsbruck, Austria
| | - Gottfried Baier
- Institute of Cell Genetics, Medical University of Innsbruck, Innsbruck, Austria
| | - Bernhard Holzmann
- Department of Surgery, School of Medicine and Health, TUM, Munich, Germany
| | - Mala K Maini
- Institute of Pathology, School of Medicine and Health, TUM, Munich, Germany
| | - Ron Heeren
- Institute of Multimodal Imaging, University of Maastricht, Maastricht, The Netherlands
| | - Peter J Murray
- Max Planck Institute of Biochemistry, Martinsried, Munich, Germany
| | - Robert Thimme
- Third Department of Medicine, University Hospital Freiburg, Freiburg, Germany
| | - Carl Herrmann
- Health Data Science Unit, Biomedical Genomics Group, Bioquant, Faculty of Medicine Heidelberg, Heidelberg, Germany
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Ulrike Protzer
- Institute of Immunology and Animal Physiology, School of Life Science, TUM, Munich, Germany
- Institute of Virology, School of Medicine and Health, TUM, Munich, Germany
- Helmholtz Zentrum München, Munich, Germany
| | - Jan P Böttcher
- Institute of Molecular Immunology, School of Medicine and Health, Technical University of Munich (TUM), Munich, Germany
| | - Dietmar Zehn
- Institute of Immunology and Animal Physiology, School of Life Science, TUM, Munich, Germany
| | - Dirk Wohlleber
- Institute of Molecular Immunology, School of Medicine and Health, Technical University of Munich (TUM), Munich, Germany
| | - Georg M Lauer
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Maike Hofmann
- Third Department of Medicine, University Hospital Freiburg, Freiburg, Germany
| | - Souphalone Luangsay
- Roche Pharmaceutical Research and Early Development (pRED), Roche Innovation Center Basel, Basel, Switzerland
| | - Percy A Knolle
- Institute of Molecular Immunology, School of Medicine and Health, Technical University of Munich (TUM), Munich, Germany.
- German Center for Infection Research, Munich site, Munich, Germany.
- Institute of Molecular Immunology, School of Life Science, TUM, Munich, Germany.
| |
Collapse
|
5
|
Abstract
The brain is designed not only with molecules and cellular processes that help to form memories but also with molecules and cellular processes that suppress the formation and retention of memory. The latter processes are critical for an efficient memory management system, given the vast amount of information that each person experiences in their daily activities and that most of this information becomes irrelevant with time. Thus, efficiency dictates that the brain should have processes for selecting the most critical information for storage and suppressing the irrelevant or forgetting it later should it escape the initial filters. Such memory suppressor molecules and processes are revealed by genetic or pharmacologic insults that lead to enhanced memory expression. We review here the predominant memory suppressor molecules and processes that have recently been discovered. They are diverse, as expected, because the brain is complex and employs many different strategies and mechanisms to form memories. They include the gene-repressive actions of small noncoding RNAs, repressors of protein synthesis, cAMP-mediated gene expression pathways, inter- and intracellular signaling pathways for normal forgetting, and others. A deep understanding of memory suppressor molecules and processes is necessary to fully comprehend how the brain forms, stabilizes, and retrieves memories and to reveal how brain disorders disrupt memory.
Collapse
Affiliation(s)
- Nathaniel C. Noyes
- Department of Neuroscience, University of Florida Scripps Biomedical Research, Jupiter, FL, USA
| | - Ronald L. Davis
- Department of Neuroscience, University of Florida Scripps Biomedical Research, Jupiter, FL, USA
| |
Collapse
|
6
|
Zhang L, Shi W, Liu J, Chen K, Zhang G, Zhang S, Cong B, Li Y. Interleukin 6 (IL-6) Regulates GABAA Receptors in the Dorsomedial Hypothalamus Nucleus (DMH) through Activation of the JAK/STAT Pathway to Affect Heart Rate Variability in Stressed Rats. Int J Mol Sci 2023; 24:12985. [PMID: 37629166 PMCID: PMC10455568 DOI: 10.3390/ijms241612985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 08/09/2023] [Accepted: 08/17/2023] [Indexed: 08/27/2023] Open
Abstract
The dorsomedial hypothalamus nucleus (DMH) is an important component of the autonomic nervous system and plays a critical role in regulating the sympathetic outputs of the heart. Stress alters the neuronal activity of the DMH, affecting sympathetic outputs and triggering heart rate variability. However, the specific molecular mechanisms behind stress leading to abnormal DMH neuronal activity have still not been fully elucidated. Therefore, in the present study, we successfully constructed a stressed rat model and used it to investigate the potential molecular mechanisms by which IL-6 regulates GABAA receptors in the DMH through activation of the JAK/STAT pathway and thus affects heart rate variability in rats. By detecting the c-Fos expression of neurons in the DMH and electrocardiogram (ECG) changes in rats, we clarified the relationship between abnormal DMH neuronal activity and heart rate variability in stressed rats. Then, using ELISA, immunohistochemical staining, Western blotting, RT-qPCR, and RNAscope, we further explored the correlation between the IL-6/JAK/STAT signaling pathway and GABAA receptors. The data showed that an increase in IL-6 induced by stress inhibited GABAA receptors in DMH neurons by activating the JAK/STAT signaling pathway, while specific inhibition of the JAK/STAT signaling pathway using AG490 obviously reduced DMH neuronal activity and improved heart rate variability in rats. These findings suggest that IL-6 regulates the expression of GABAA receptors via the activation of the JAK/STAT pathway in the DMH, which may be an important cause of heart rate variability in stressed rats.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Bin Cong
- Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Department of Forensic Medicine, Hebei Medical University, Shijiazhuang 050017, China; (L.Z.); (W.S.); (J.L.); (K.C.); (G.Z.); (S.Z.)
| | - Yingmin Li
- Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Department of Forensic Medicine, Hebei Medical University, Shijiazhuang 050017, China; (L.Z.); (W.S.); (J.L.); (K.C.); (G.Z.); (S.Z.)
| |
Collapse
|
7
|
Gómez LCG, Medina NB, Blasco SS, Gravielle MC. Diazepam-Induced Down-Regulation of The Gaba a Receptor α1 Subunit, as Mediated by the Activation of L-Type Voltage-Gated Calcium Channel/Ca 2+/Protein Kinase A Signaling Cascade. Neurosci Lett 2023:137358. [PMID: 37356564 DOI: 10.1016/j.neulet.2023.137358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 06/12/2023] [Accepted: 06/20/2023] [Indexed: 06/27/2023]
Abstract
Benzodiazepines are among the most prescribed drug class worldwide to treat disorders such as anxiety, insomnia, muscle spasticity, and convulsive disorders, and to induce presurgical sedation. Although benzodiazepines exhibit a high therapeutic index and low toxicity in short-term treatments, prolonged administration induces tolerance to most of their therapeutic actions. The mechanism of this tolerance remains unclear. The central actions of benzodiazepines are mediated by binding to GABAA receptors, which mediate most fast inhibitory transmission in the brain. The majority of GABAA receptors are composed of two α-(1-6), two β-(1-3) and one γ-subunits (1-3). In a previous report, we demonstrated that the prolonged exposure of cerebrocortical neurons to diazepam produces a transcriptional repression of the GABAA receptor α1 subunit gene via a mechanism dependent on the activation of L-type voltage-gated calcium channels (L-VGCCs). The results reported here confirm that the diazepam-induced downregulation of the α1 subunit is contingent upon calcium influx from extracellular space. In addition, this regulatory mechanism involves the activation of protein kinase A (PKA) and is accompanied by the activation of two transcription factors, the cAMP-response element-binding protein (CREB) and the inducible cAMP early repressor (ICER). Together, our results suggest that diazepam's activation of an L-VGCC/Ca2+/PKA/CREB-ICER signaling pathway is responsible for the regulation of GABAA receptors. This elucidation of the intracellular signaling cascade activated by a prolonged benzodiazepine exposure, itself potentially involved in the development of tolerance, may contribute to locating molecular targets for future therapeutic interventions.
Collapse
Affiliation(s)
- Leydi Carolina González Gómez
- Instituto de Investigaciones Farmacológicas (ININFA). Facultad de Farmacia y Bioquímica. Universidad de Buenos Aires. CONICET. Buenos Aires, Argentina
| | - Nelsy Beatriz Medina
- Instituto de Investigaciones Farmacológicas (ININFA). Facultad de Farmacia y Bioquímica. Universidad de Buenos Aires. CONICET. Buenos Aires, Argentina
| | - Sara Sanz Blasco
- Instituto de Investigaciones Farmacológicas (ININFA). Facultad de Farmacia y Bioquímica. Universidad de Buenos Aires. CONICET. Buenos Aires, Argentina
| | - María Clara Gravielle
- Instituto de Investigaciones Farmacológicas (ININFA). Facultad de Farmacia y Bioquímica. Universidad de Buenos Aires. CONICET. Buenos Aires, Argentina.
| |
Collapse
|
8
|
Polygonatum sibiricum polysaccharides improve cognitive function in D-galactose-induced aging mice by regulating the microbiota-gut-brain axis. J Funct Foods 2023. [DOI: 10.1016/j.jff.2023.105476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/06/2023] Open
|
9
|
Yanai S, Tago T, Toyohara J, Arasaki T, Endo S. Reversal of spatial memory impairment by phosphodiesterase 3 inhibitor cilostazol is associated with reduced neuroinflammation and increased cerebral glucose uptake in aged male mice. Front Pharmacol 2022; 13:1031637. [PMID: 36618932 PMCID: PMC9810637 DOI: 10.3389/fphar.2022.1031637] [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: 08/30/2022] [Accepted: 11/30/2022] [Indexed: 12/24/2022] Open
Abstract
The nucleotide second messenger 3', 5'-cyclic adenosine monophosphate (cAMP) and 3', 5'-cyclic guanosine monophosphate (cGMP) mediate fundamental functions of the brain, including learning and memory. Phosphodiesterase 3 (PDE3) can hydrolyze both cAMP and cGMP and appears to be involved in the regulation of their contents in cells. We previously demonstrated that long-term administration of cilostazol, a PDE3 inhibitor, maintained good memory performance in aging mice. Here, we report on studies aimed at determining whether cilostazol also reverses already-impaired memory in aged male mice. One month of oral 1.5% cilostazol administration in 22-month-old mice reversed age-related declines in hippocampus-dependent memory tasks, including the object recognition and the Morris water maze. Furthermore, cilostazol reduced neuroinflammation, as evidenced by immunohistochemical staining, and increased glucose uptake in the brain, as evidence by positron emission tomography (PET) with 2-deoxy-2-[18F]fluoro-d-glucose ([18F]FDG). These results suggest that already-expressed memory impairment in aged male mice that depend on cyclic nucleotide signaling can be reversed by inhibition of PDE3. The reversal of age-related memory impairments may occur in the central nervous system, either through cilostazol-enhanced recall or strengthening of weak memories that otherwise may be resistant to recall.
Collapse
Affiliation(s)
- Shuichi Yanai
- Aging Neuroscience Research Team, Tokyo Metropolitan Institute of Gerontology, Tokyo, Japan
| | - Tetsuro Tago
- Research Team for Neuroimaging, Tokyo Metropolitan Institute of Gerontology, Tokyo, Japan
| | - Jun Toyohara
- Research Team for Neuroimaging, Tokyo Metropolitan Institute of Gerontology, Tokyo, Japan
| | - Tomoko Arasaki
- Aging Neuroscience Research Team, Tokyo Metropolitan Institute of Gerontology, Tokyo, Japan
| | - Shogo Endo
- Aging Neuroscience Research Team, Tokyo Metropolitan Institute of Gerontology, Tokyo, Japan,*Correspondence: Shogo Endo,
| |
Collapse
|
10
|
Fu P, Zhao Y, Dong C, Cai Z, Li R, Yung KKL. An integrative analysis of miRNA and mRNA expression in the brains of Alzheimer's disease transgenic mice after real-world PM 2.5 exposure. J Environ Sci (China) 2022; 122:25-40. [PMID: 35717088 DOI: 10.1016/j.jes.2021.10.007] [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: 07/05/2021] [Revised: 09/27/2021] [Accepted: 10/06/2021] [Indexed: 06/15/2023]
Abstract
Fine particulate matter (PM2.5) is associated with increased risks of Alzheimer's disease (AD), yet the toxicological mechanisms of PM2.5 promoting AD remain unclear. In this study, wild-type and APP/PS1 transgenic mice (AD mice) were exposed to either filtered air (FA) or PM2.5 for eight weeks with a real-world exposure system in Taiyuan, China (mean PM2.5 concentration in the cage was 61 µg/m3). We found that PM2.5 exposure could remarkably aggravate AD mice's ethological and brain ultrastructural damage, along with the elevation of the pro-inflammatory cytokines (IL-6 and TNF-α), Aβ-42 and AChE levels and the decline of ChAT levels in the brains. Based on high-throughput sequencing results, some differentially expressed (DE) mRNAs and DE miRNAs in the brains of AD mice after PM2.5 exposure were screened. Using RT-qPCR, seven DE miRNAs (mmu-miR-193b-5p, 122b-5p, 466h-3p, 10b-5p, 1895, 384-5p, and 6412) and six genes (Pcdhgb8, Unc13b, Robo3, Prph, Pter, and Tbata) were evidenced the and verified. Two miRNA-target gene pairs (miR-125b-Pcdhgb8 pair and miR-466h-3p-IL-17Rα/TGF-βR2/Aβ-42/AChE pairs) were demonstrated that they were more related to PM2.5-induced brain injury. Results of Gene Ontology (GO) pathways and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways predicted that synaptic and postsynaptic regulation, axon guidance, Wnt, MAPK, and mTOR pathways might be the possible regulatory mechanisms associated with pathological response. These revealed that PM2.5-elevated pro-inflammatory cytokine levels and PM2.5-altered neurotransmitter levels in AD mice could be the important causes of brain damage and proposed the promising miRNA and mRNA biomarkers and potential miRNA-mRNA interaction networks of PM2.5-promoted AD.
Collapse
Affiliation(s)
- Pengfei Fu
- Department of Biology, Hong Kong Baptist University, Hong Kong SAR, China; Golden Meditech Center for NeuroRegeneration Sciences, Hong Kong Baptist University, Hong Kong SAR, China
| | - Yufei Zhao
- Institute of Environmental Science, Shanxi University, Taiyuan 237016, China
| | - Chuan Dong
- Institute of Environmental Science, Shanxi University, Taiyuan 237016, China
| | - Zongwei Cai
- State Key Laboratory of Environmental and Biological Analysis, Hong Kong Baptist University, Hong Kong SAR, China
| | - Ruijin Li
- Institute of Environmental Science, Shanxi University, Taiyuan 237016, China.
| | - Ken Kin Lam Yung
- Department of Biology, Hong Kong Baptist University, Hong Kong SAR, China; Golden Meditech Center for NeuroRegeneration Sciences, Hong Kong Baptist University, Hong Kong SAR, China.
| |
Collapse
|
11
|
Allen CNS, Arjona SP, Santerre M, De Lucia C, Koch WJ, Sawaya BE. Metabolic Reprogramming in HIV-Associated Neurocognitive Disorders. Front Cell Neurosci 2022; 16:812887. [PMID: 35418836 PMCID: PMC8997587 DOI: 10.3389/fncel.2022.812887] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 02/21/2022] [Indexed: 12/20/2022] Open
Abstract
A significant number of patients infected with HIV-1 suffer from HIV-associated neurocognitive disorders (HAND) such as spatial memory impairments and learning disabilities (SMI-LD). SMI-LD is also observed in patients using combination antiretroviral therapy (cART). Our lab has demonstrated that the HIV-1 protein, gp120, promotes SMI-LD by altering mitochondrial functions and energy production. We have investigated cellular processes upstream of the mitochondrial functions and discovered that gp120 causes metabolic reprogramming. Effectively, the addition of gp120 protein to neuronal cells disrupted the glycolysis pathway at the pyruvate level. Looking for the players involved, we found that gp120 promotes increased expression of polypyrimidine tract binding protein 1 (PTBP1), causing the splicing of pyruvate kinase M (PKM) into PKM1 and PKM2. We have also shown that these events lead to the accumulation of advanced glycation end products (AGEs) and prevent the cleavage of pro-brain-derived neurotrophic factor (pro-BDNF) protein into mature brain-derived neurotrophic factor (BDNF). The accumulation of proBDNF results in signaling that increases the expression of the inducible cAMP early repressor (ICER) protein which then occupies the cAMP response element (CRE)-binding sites within the BDNF promoters II and IV, thus altering normal synaptic plasticity. We reversed these events by adding Tepp-46, which stabilizes the tetrameric form of PKM2. Therefore, we concluded that gp120 reprograms cellular metabolism, causing changes linked to disrupted memory in HIV-infected patients and that preventing the disruption of the metabolism presents a potential cure against HAND progression.
Collapse
Affiliation(s)
- Charles N. S. Allen
- Molecular Studies of Neurodegenerative Diseases Lab, Fels Cancer Institute for Personalized Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| | - Sterling P. Arjona
- Molecular Studies of Neurodegenerative Diseases Lab, Fels Cancer Institute for Personalized Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| | - Maryline Santerre
- Molecular Studies of Neurodegenerative Diseases Lab, Fels Cancer Institute for Personalized Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| | - Claudio De Lucia
- Department of Cardiovascular Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| | - Walter J. Koch
- Department of Cardiovascular Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| | - Bassel E. Sawaya
- Molecular Studies of Neurodegenerative Diseases Lab, Fels Cancer Institute for Personalized Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
- Department of Cancer and Cellular Biology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
- Department of Neural Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
- *Correspondence: Bassel E. Sawaya,
| |
Collapse
|
12
|
Hakim M, Beecher K, Jacques A, Chaaya N, Belmer A, Battle AR, Johnson LR, Bartlett SE, Chehrehasa F. Retrieval of olfactory fear memory alters cell proliferation and expression of pCREB and pMAPK in the corticomedial amygdala and piriform cortex. Chem Senses 2022; 47:6673813. [PMID: 35997758 PMCID: PMC9397123 DOI: 10.1093/chemse/bjac021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The brain forms robust associations between odors and emotionally salient memories, making odors especially effective at triggering fearful or traumatic memories. Using Pavlovian olfactory fear conditioning (OFC), a variant of the traditional tone-shock paradigm, this study explored the changes involved in its processing. We assessed the expression of neuronal plasticity markers phosphorylated cyclic adenosine monophosphate response element binding protein (pCREB) and phosphorylated mitogen-activated protein kinase (pMAPK) 24 h and 14 days following OFC, in newborn neurons (EdU+) and in brain regions associated with olfactory memory processing; the olfactory bulb, piriform cortex, amygdale, and hippocampus. Here, we show that all proliferating neurons in the dentate gyrus of the hippocampus and glomerular layer of the olfactory bulb were colocalized with pCREB at 24 h and 14 days post-conditioning, and the number of proliferating neurons at both time points were statistically similar. This suggests the occurrence of long-term potentiation within the neurons of this pathway. Finally, OFC significantly increased the density of pCREB- and pMAPK-positive immunoreactive neurons in the medial and cortical subnuclei of the amygdala and the posterior piriform cortex, suggesting their key involvement in its processing. Together, our investigation identifies changes in neuroplasticity within critical neural circuits responsible for olfactory fear memory.
Collapse
Affiliation(s)
- Marziah Hakim
- Addiction Neuroscience and Obesity Laboratory, School of Biomedical Sciences, Faculty of Health, Translational Research Institute, Queensland University of Technology, Brisbane, QLD, Australia
| | - Kate Beecher
- Addiction Neuroscience and Obesity Laboratory, School of Clinical Sciences, Faculty of Health, Translational Research Institute, Queensland University of Technology, Brisbane, QLD, Australia
| | - Angela Jacques
- Addiction Neuroscience and Obesity Laboratory, School of Clinical Sciences, Faculty of Health, Translational Research Institute, Queensland University of Technology, Brisbane, QLD, Australia
| | - Nicholas Chaaya
- Addiction Neuroscience and Obesity Laboratory, School of Clinical Sciences, Faculty of Health, Translational Research Institute, Queensland University of Technology, Brisbane, QLD, Australia
| | - Arnauld Belmer
- Addiction Neuroscience and Obesity Laboratory, School of Clinical Sciences, Faculty of Health, Translational Research Institute, Queensland University of Technology, Brisbane, QLD, Australia
| | - Andrew R Battle
- Addiction Neuroscience and Obesity Laboratory, School of Biomedical Sciences, Faculty of Health, Translational Research Institute, Queensland University of Technology, Brisbane, QLD, Australia
| | - Luke R Johnson
- Addiction Neuroscience and Obesity Laboratory, School of Clinical Sciences, Faculty of Health, Translational Research Institute, Queensland University of Technology, Brisbane, QLD, Australia.,School of Medicine. Division of Psychology, University of Tasmania, Launceston, TAS, Australia
| | - Selena E Bartlett
- Addiction Neuroscience and Obesity Laboratory, School of Clinical Sciences, Faculty of Health, Translational Research Institute, Queensland University of Technology, Brisbane, QLD, Australia
| | - Fatemeh Chehrehasa
- Addiction Neuroscience and Obesity Laboratory, School of Biomedical Sciences, Faculty of Health, Translational Research Institute, Queensland University of Technology, Brisbane, QLD, Australia
| |
Collapse
|
13
|
Seidl MD, Fels B, Kranick D, Sternberg A, Grimm K, Stümpel FT, Pluteanu F, Schulte JS, Heinick A, Kojima N, Endo S, Huge A, Stoll M, Müller FU. Induction of ICER is superseded by smICER, challenging the impact of ICER under chronic beta-adrenergic stimulation. FASEB J 2020; 34:11272-11291. [PMID: 32602979 DOI: 10.1096/fj.201902301rr] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Revised: 03/30/2020] [Accepted: 04/21/2020] [Indexed: 11/11/2022]
Abstract
ICER (inducible cAMP early repressor) isoforms are transcriptional repressors encoded by the Crem (cAMP responsive element modulator) gene. They were linked to the regulation of a multitude of cellular processes and pathophysiological mechanisms. Here, we show for the first time that two independent induction patterns for CREM repressor isoforms exist in the heart, namely for ICER and smICER (small ICER), which are induced in response to β-adrenergic stimulation in a transient- and saturation-like manner, respectively. This time-shifted induction pattern, driven by two internal promoters in the Crem gene, leads to the predominant transcription of smIcer after prolonged β-adrenergic stimulation. Using an ICER knockout mouse model with preserved smICER induction, we show that the transient-like induction of Icer itself has minor effects on gene regulation, cardiac hypertrophy or contractile function in the heart. We conclude that the functions previously linked to ICER may be rather attributed to smICER, also beyond the cardiac background.
Collapse
Affiliation(s)
- Matthias D Seidl
- Institute of Pharmacology and Toxicology, University of Münster, Münster, Germany
| | - Benedikt Fels
- Institute of Pharmacology and Toxicology, University of Münster, Münster, Germany
| | - Daniel Kranick
- Institute of Pharmacology and Toxicology, University of Münster, Münster, Germany
| | - Alexandra Sternberg
- Institute of Pharmacology and Toxicology, University of Münster, Münster, Germany
| | - Kristina Grimm
- Institute of Pharmacology and Toxicology, University of Münster, Münster, Germany
| | - Frank T Stümpel
- Institute of Pharmacology and Toxicology, University of Münster, Münster, Germany
| | - Florentina Pluteanu
- Institute of Pharmacology and Toxicology, University of Münster, Münster, Germany
| | - Jan S Schulte
- Institute of Pharmacology and Toxicology, University of Münster, Münster, Germany
| | - Alexander Heinick
- Institute of Pharmacology and Toxicology, University of Münster, Münster, Germany
| | | | - Shogo Endo
- Aging Neuroscience Research Team, Tokyo Metropolitan Institute of Gerontology, Tokyo, Japan
| | - Andreas Huge
- Core Facility Genomik, Medical Faculty, University of Muenster, Germany
| | - Monika Stoll
- Core Facility Genomik, Medical Faculty, University of Muenster, Germany
| | - Frank U Müller
- Institute of Pharmacology and Toxicology, University of Münster, Münster, Germany
| |
Collapse
|
14
|
P311, a novel intrinsically disordered protein, regulates adipocyte development. Biochem Biophys Res Commun 2019; 515:234-240. [PMID: 31146912 PMCID: PMC6561663 DOI: 10.1016/j.bbrc.2019.05.105] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Accepted: 05/14/2019] [Indexed: 12/12/2022]
Abstract
Adipocyte development and adipose tissue expansion have many implications for human diseases, including obesity. Obesity is a debilitating disorder and a risk factor for metabolic disorders including insulin resistance and diabetes mellitus, due in part to an overabundance of adipocytes and adipocyte dysfunction. In recent years, obesity has become a global pandemic with approximately one-third of US adults classified as obese. Adipose tissue has recently been identified as a major metabolic organ, classified into white adipose tissue (WAT) and brown adipose tissue (BAT). Other than lifestyle modifications and invasive surgeries, only a very limited number of drugs are available to treat obesity and overweight. P311 has been shown to play a key role in blood pressure regulation, vascular contractility and tissue remodeling. Here we present a role for P311 in adipogenesis using a 3T3-L1 cell culture model. P311 expression is initiated with the induction of adipogenesis and increased during adipogenesis. This increase correlates with an increase in the expression of the key adipogenic transcriptional factors PPARγ2 and C/EBPα. In addition, siRNA-mediated P311 knockdown inhibits adipogenic differentiation in 3T3-L1 cells. Finally, P311 binds to the PPARγ2 promoter, implicating P311 mediates adipogenesis partly through PPARγ activation.
Collapse
|
15
|
PDE3 Inhibitors Repurposed as Treatments for Age-Related Cognitive Impairment. Mol Neurobiol 2018; 56:4306-4316. [PMID: 30311144 DOI: 10.1007/s12035-018-1374-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Accepted: 09/27/2018] [Indexed: 12/21/2022]
Abstract
As the population of older individuals grows worldwide, researchers have increasingly focused their attention on identifying key molecular targets of age-related cognitive impairments, with the aim of developing possible therapeutic interventions. Two such molecules are the intracellular cyclic nucleotides, cAMP and cGMP. These second messengers mediate fundamental aspects of brain function relevant to memory, learning, and cognitive function. Consequently, phosphodiesterases (PDEs), which hydrolyze cAMP and cGMP, are promising targets for the development of cognition-enhancing drugs. Inhibitors that target PDEs work by elevating intracellular cAMP. In this review, we provide an overview of different PDE inhibitors, and then we focus on pharmacological and physiological effects of PDE3 inhibitors in the CNS and peripheral tissues. Finally, we discuss findings from experimental and preliminary clinical studies and the potential beneficial effects of the PDE3 inhibitor cilostazol on age-related cognitive impairments. In the innovation pipeline of pharmaceutical development, the antiplatelet agent cilostazol has come into the spotlight as a novel treatment for mild cognitive impairment. Overall, the repurposing of cilostazol may represent a potentially promising way to treat mild cognitive impairment, Alzheimer's disease, and vascular dementia. In this review, we present a brief summary of cAMP signaling and different PDE inhibitors, followed by a discussion of the pharmacological and physiological role of PDE3 inhibitors. In this context, we discuss the repurposing of a PDE3 inhibitor, cilostazol, as a potential treatment for age-related cognitive impairment based on recent research.
Collapse
|
16
|
Zygmunt M, Piechota M, Rodriguez Parkitna J, Korostyński M. Decoding the transcriptional programs activated by psychotropic drugs in the brain. GENES BRAIN AND BEHAVIOR 2018; 18:e12511. [PMID: 30084543 DOI: 10.1111/gbb.12511] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Revised: 07/25/2018] [Accepted: 08/03/2018] [Indexed: 01/01/2023]
Abstract
Analysis of drug-induced gene expression in the brain has long held the promise of revealing the molecular mechanisms of drug actions as well as predicting their long-term clinical efficacy. However, despite some successes, this promise has yet to be fulfilled. Here, we present an overview of the current state of understanding of drug-induced gene expression in the brain and consider the obstacles to achieving a robust prediction of the properties of psychoactive compounds based on gene expression profiles. We begin with a comprehensive overview of the mechanisms controlling drug-inducible transcription and the complexity resulting from expression of noncoding RNAs and alternative gene isoforms. Particular interest is placed on studies that examine the associations within drug classes with regard to the effects on gene transcription, alterations in cell signaling and neuropharmacological drug properties. While the ability of gene expression signatures to distinguish specific clinical classes of psychotropic and addictive drugs remains unclear, some reports show that under specific constraints, drug properties can be predicted based on gene expression. Such signatures offer a simple and effective way to classify psychotropic drugs and screen novel psychoactive compounds. Finally, we note that the amount of data regarding molecular programs activated in the brain by drug treatment has grown exponentially in recent years and that future advances may therefore come in large part from integrating the currently available high-throughput data sets.
Collapse
Affiliation(s)
- Magdalena Zygmunt
- Department of Molecular Neuropharmacology, Institute of Pharmacology of the Polish Academy of Sciences, Krakow, Poland
| | - Marcin Piechota
- Department of Molecular Neuropharmacology, Institute of Pharmacology of the Polish Academy of Sciences, Krakow, Poland
| | - Jan Rodriguez Parkitna
- Department of Molecular Neuropharmacology, Institute of Pharmacology of the Polish Academy of Sciences, Krakow, Poland
| | - Michał Korostyński
- Department of Molecular Neuropharmacology, Institute of Pharmacology of the Polish Academy of Sciences, Krakow, Poland
| |
Collapse
|
17
|
Differential Gene Expression Profile Induced by Valproic Acid (VPA) in Pediatric Epileptic Patients. Genes (Basel) 2018; 9:genes9070328. [PMID: 29958461 PMCID: PMC6070821 DOI: 10.3390/genes9070328] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Revised: 06/20/2018] [Accepted: 06/26/2018] [Indexed: 12/31/2022] Open
Abstract
Epilepsy is a neuronal disease that affects up to 70 million people worldwide. The development of effective therapies to combat childhood epilepsy requires early biomarkers. Here, we performed a whole-genome microarray analysis in blood cells to identify genes differentially expressed between epileptic and epileptic valproic acid (VPA)-treated children versus normal children to obtain information about the gene expression to help us to understand genetic aspects of this disease. We found that the most significant differentially expressed genes were related to the transcriptional factor cAMP-response element binding protein (CREB) that is overexpressed in children with epilepsy compared with normal children, and 6 and 12 months of VPA treatment reversed several of these changes. Interestingly, leukocyte-associated immunoglobulin-like receptor 1 (LAIR1), a type I transmembrane glycoprotein that binds collagen proteins and contains CREB binding sites, was one of the more up-regulated genes in epileptic patients, and treatment with VPA strongly reversed its up-regulation. CREB up-regulates genes related to epilepsy; here, we suggest that LAIR1 could activate CREB, and together, they trigger epilepsy. After VPA treatment, LAIR1 repressed genes by disrupting the functional LAIR1–CREB complex, resulting in successful treatment. A functional microarray analysis offers new information that could open novel avenues of research in biomarker discovery, which may be useful for the early identification of children with a predisposition to epilepsy.
Collapse
|
18
|
Feng H, Khalil S, Neubig RR, Sidiropoulos C. A mechanistic review on GNAO1-associated movement disorder. Neurobiol Dis 2018; 116:131-141. [PMID: 29758257 DOI: 10.1016/j.nbd.2018.05.005] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Revised: 04/28/2018] [Accepted: 05/10/2018] [Indexed: 02/07/2023] Open
Abstract
Mutations in the GNAO1 gene cause a complex constellation of neurological disorders including epilepsy, developmental delay, and movement disorders. GNAO1 encodes Gαo, the α subunit of Go, a member of the Gi/o family of heterotrimeric G protein signal transducers. Go is the most abundant membrane protein in the mammalian central nervous system and plays major roles in synaptic neurotransmission and neurodevelopment. GNAO1 mutations were first reported in early infantile epileptic encephalopathy 17 (EIEE17) but are also associated with a more common syndrome termed neurodevelopmental disorder with involuntary movements (NEDIM). Here we review a mechanistic model in which loss-of-function (LOF) GNAO1 alleles cause epilepsy and gain-of-function (GOF) alleles are primarily associated with movement disorders. We also develop a signaling framework related to cyclic AMP (cAMP), synaptic vesicle release, and neural development and discuss gene mutations perturbing those mechanisms in a range of genetic movement disorders. Finally, we analyze clinical reports of patients carrying GNAO1 mutations with respect to their symptom onset and discuss pharmacological/surgical treatments in the context of our mechanistic model.
Collapse
Affiliation(s)
- Huijie Feng
- Department of Pharmacology & Toxicology, Michigan State University, East Lansing, MI 48824, USA
| | - Suad Khalil
- Department of Neurology & Ophthalmology, Michigan State University, East Lansing, MI 48824, USA
| | - Richard R Neubig
- Department of Pharmacology & Toxicology, Michigan State University, East Lansing, MI 48824, USA.
| | - Christos Sidiropoulos
- Department of Neurology & Ophthalmology, Michigan State University, East Lansing, MI 48824, USA.
| |
Collapse
|
19
|
Yanai S, Ito H, Endo S. Long-term cilostazol administration prevents age-related decline of hippocampus-dependent memory in mice. Neuropharmacology 2017; 129:57-68. [PMID: 29122629 DOI: 10.1016/j.neuropharm.2017.11.008] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Revised: 10/30/2017] [Accepted: 11/04/2017] [Indexed: 12/17/2022]
Abstract
Phosphodiesterases (PDEs) are enzymes that hydrolyze and inactivate 3', 5'-cyclic adenosine monophosphate (cAMP) and/or 3', 5'-cyclic guanosine monophosphate (cGMP). The regulation of intracellular signaling pathways mediated by cyclic nucleotides is imperative to synaptic plasticity and memory in animals. Because PDEs play an important role in this regulation, PDE inhibitors are considered as candidate compounds for treating cognitive and memory disorders. In the present study, we tested whether cilostazol, a selective PDE3 inhibitor, prevents the cognitive deterioration that occurs during the course of normal aging in mice. Ten months of cilostazol administration (1.5%) in 13-month-old mice improved spatial memory when tested at 23 months of age. First, it prevented the decline in the ability of these aged mice to recognize a change in an object's location in the object recognition task. Second, spatial memory of these cilostazol-treated aged mice in the Morris water maze was comparable to that of untreated middle-aged mice (13 months old). Cilostazol administration had no effect on the emotional states and physical ability of aged mice. Thus, long-term cilostazol administration prevented hippocampus-dependent memory decline in aged mice, allowing them to achieve a level of cognitive performance similar to middle-aged mice and without negative behavioral side effects. Considering its well-established safety in other medical contexts, cilostazol may be a potential therapeutic candidate drug for staving off cognitive decline in the aging human population.
Collapse
Affiliation(s)
- Shuichi Yanai
- Aging Neuroscience Research Team, Tokyo Metropolitan Institute of Gerontology, Itabashi, Tokyo 173-0015, Japan
| | - Hideki Ito
- Department of CNS Research, Otsuka Pharmaceutical Co., Ltd., Tokushima, 771-0192, Japan
| | - Shogo Endo
- Aging Neuroscience Research Team, Tokyo Metropolitan Institute of Gerontology, Itabashi, Tokyo 173-0015, Japan.
| |
Collapse
|
20
|
Yanai S, Toyohara J, Ishiwata K, Ito H, Endo S. Long-term cilostazol administration ameliorates memory decline in senescence-accelerated mouse prone 8 (SAMP8) through a dual effect on cAMP and blood-brain barrier. Neuropharmacology 2016; 116:247-259. [PMID: 27979612 DOI: 10.1016/j.neuropharm.2016.12.006] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Revised: 11/18/2016] [Accepted: 12/09/2016] [Indexed: 12/12/2022]
Abstract
Phosphodiesterases (PDEs), which hydrolyze and inactivate 3', 5'-cyclic adenosine monophosphate (cAMP) and 3', 5'-cyclic guanosine monophosphate (cGMP), play an important role in synaptic plasticity that underlies memory. Recently, several PDE inhibitors were assessed for their possible therapeutic efficacy in treating cognitive disorders. Here, we examined how cilostazol, a selective PDE3 inhibitor, affects brain functions in senescence-accelerated mouse prone 8 (SAMP8), an animal model of age-related cognitive impairment. Long-term administration of cilostazol restored the impaired context-dependent conditioned fear memory of SAMP8 to match that in normal aging control substrain SAMR1. Cilostazol also increased the number of cells containing phosphorylated cAMP-responsive element binding protein (CREB), a downstream component of the cAMP pathway. Finally, cilostazol improves blood-brain barrier (BBB) integrity, demonstrated by reduced extravasation of 2-deoxy-2-18F-fluoro-d-glucose and Evans Blue dye in the brains of SAMP8. This improvement in BBB integrity was associated with an increased amount of zona occludens protein 1 (ZO-1) and occludin proteins, components of tight junctions integral to the BBB. The results suggest that long-term administration of cilostazol exerts its beneficial effects on age-related cognitive impairment through a dual mechanism: by enhancing the cAMP system in the brain and by maintaining or improving BBB integrity.
Collapse
Affiliation(s)
- Shuichi Yanai
- Aging Neuroscience Research Team, Tokyo Metropolitan Institute of Gerontology, Itabashi, Tokyo 173-0015, Japan
| | - Jun Toyohara
- Research Team for Neuroimaging, Tokyo Metropolitan Institute of Gerontology, Itabashi, Tokyo 173-0015, Japan
| | - Kiichi Ishiwata
- Research Team for Neuroimaging, Tokyo Metropolitan Institute of Gerontology, Itabashi, Tokyo 173-0015, Japan; Institute of Cyclotron and Drug Discovery Research, Southern TOHOKU Research Institute for Neuroscience, Koriyama, Fukushima 963-8052, Japan; Department of Biofunctional Imaging, Fukushima Medical University, Fukushima 960-1295, Japan
| | - Hideki Ito
- Department of CNS Research, Otsuka Pharmaceutical Co., Ltd., Tokushima 771-0192, Japan
| | - Shogo Endo
- Aging Neuroscience Research Team, Tokyo Metropolitan Institute of Gerontology, Itabashi, Tokyo 173-0015, Japan.
| |
Collapse
|
21
|
Yanai S, Endo S. Data on the optimization of behavioral tasks for senescence-accelerated mouse prone 8 (SAMP8). Data Brief 2016; 8:262-6. [PMID: 27331099 PMCID: PMC4906132 DOI: 10.1016/j.dib.2016.05.044] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Revised: 05/12/2016] [Accepted: 05/19/2016] [Indexed: 01/30/2023] Open
Abstract
This data article contains the supporting information for the research article entitled "Early onset of behavioral alterations in senescence-accelerated mouse prone 8 (SAMP8)" [1]. Senescence-accelerated mouse prone 8 (SAMP8), which originally developed from AKR/J mice, shows learning and memory impairments at the age of 8-12 months. However, little information is still available on phenotypical characteristics of younger SAMP8. To fully understand the phenotype of younger SAMP8, we optimized two behavioral tasks for SAMP8. In the object recognition task, 4-month-old SAMP8 made significantly more contacts with the familiar objects compared to age-matched SAMR1, however, distance traveled for both strains of mice were comparable. In the fear conditioning task, conventionally-used CS-US combination failed to induce robust conditioned fear in both strains of mice.
Collapse
Affiliation(s)
- Shuichi Yanai
- Aging Neuroscience Research Team, Tokyo Metropolitan Institute of Gerontology, Japan
| | - Shogo Endo
- Aging Neuroscience Research Team, Tokyo Metropolitan Institute of Gerontology, Japan
| |
Collapse
|
22
|
Early onset of behavioral alterations in senescence-accelerated mouse prone 8 (SAMP8). Behav Brain Res 2016; 308:187-95. [PMID: 27093926 DOI: 10.1016/j.bbr.2016.04.026] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Revised: 04/11/2016] [Accepted: 04/14/2016] [Indexed: 12/22/2022]
Abstract
Senescence-accelerated mouse (SAM) is inbred lines of mice originally developed from AKR/J mice. Among the six SAM prone (SAMP) substrains, 8- to 12-month-old SAMP8 have long been used as a model of age-related cognitive impairments. However, little is still known for younger SAMP8 mice. Here, we examined the phenotypical characteristics of 4-month-old SAMP8 using a battery of behavioral tests. Four-month-old SAMP8 mice failed to recognize spatially displaced object in an object recognition task and performed poorly in the probe test of the Morris water maze task compared to SAMR1, suggesting that SAMP8 have impaired spatial memory. In addition, young SAMP8 exhibited enhanced anxiety-like behavior in an open field test and showed depression-like behavior in the forced-swim test. Their circadian rhythm was also disrupted. These abnormal behaviors of young SAMP8 are similar to behavioral alterations also observed in aged mice. In summary, age-related behavioral alterations occur in SAMP8 as young as 4 months old.
Collapse
|
23
|
Yamamoto N, Agata K, Nakashima K, Imamura T. Bidirectional promoters link cAMP signaling with irreversible differentiation through promoter-associated non-coding RNA (pancRNA) expression in PC12 cells. Nucleic Acids Res 2016; 44:5105-22. [PMID: 26945044 PMCID: PMC4914083 DOI: 10.1093/nar/gkw113] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2015] [Accepted: 02/16/2016] [Indexed: 12/14/2022] Open
Abstract
Bidirectional promoters are the major source of gene activation-associated noncoding RNA (ncRNA). PC12 cells offer an interesting model for understanding the mechanism underlying bidirectional promoter-mediated cell cycle control. Nerve growth factor (NGF)-stimulated PC12 cells elongate neurites, and are in a reversible cell-cycle-arrested state. In contrast, these cells irreversibly differentiate and cannot re-enter the normal cell cycle after NGF plus cAMP treatment. In this study, using directional RNA-seq, we found that bidirectional promoters for protein-coding genes with promoter-associated ncRNA (pancRNA) were enriched for cAMP response element consensus sequences, and were preferred targets for transcriptional regulation by the transcription factors in the cAMP-dependent pathway. A spindle-formation-associated gene, Nusap1 and pancNusap1 were among the most strictly co-transcribed pancRNA–mRNA pairs. This pancRNA–mRNA pair was specifically repressed in irreversibly differentiated PC12 cells. Knockdown (KD) and overexpression experiments showed that pancNusap1 positively regulated the Nusap1 expression in a sequence-specific manner, which was accompanied by histone acetylation at the Nusap1 promoter. Furthermore, pancNusap1 KD recapitulated the effects of cAMP on cell cycle arrest. Thus, we conclude that pancRNA-mediated histone acetylation contributes to the establishment of the cAMP-induced transcription state of the Nusap1 locus and contributes to the irreversible cell cycle exit for terminal differentiation of PC12 cells.
Collapse
Affiliation(s)
- Naoki Yamamoto
- Department of Stem Cell Biology and Medicine, Graduate School of Medical Sciences, Kyushu University, Japan Department of Biophysics, Graduate School of Science, Kyoto University, Japan
| | - Kiyokazu Agata
- Department of Biophysics, Graduate School of Science, Kyoto University, Japan
| | - Kinichi Nakashima
- Department of Stem Cell Biology and Medicine, Graduate School of Medical Sciences, Kyushu University, Japan
| | - Takuya Imamura
- Department of Stem Cell Biology and Medicine, Graduate School of Medical Sciences, Kyushu University, Japan
| |
Collapse
|
24
|
Piechota M, Golda S, Ficek J, Jantas D, Przewlocki R, Korostynski M. Regulation of alternative gene transcription in the striatum in response to antidepressant drugs. Neuropharmacology 2015; 99:328-36. [PMID: 26254862 DOI: 10.1016/j.neuropharm.2015.08.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2015] [Revised: 05/21/2015] [Accepted: 08/03/2015] [Indexed: 12/13/2022]
Abstract
The mechanisms that control the selection of transcription initiation and termination sites in response to pharmacological stimulation of neuronal cells are poorly understood. We used next-generation sequencing and bioinformatics to construct a genome-wide inventory of protein-coding and non-coding transcripts altered by antidepressant treatment. We analyzed available ChIP-seq data to identify mechanisms that control drug-inducible expression of alternative gene variants in the brain. We identified 153 transcripts of various biotypes regulated in the mouse striatum in response to tranylcypromine or mianserin (at a 0.1% FDR threshold). Five drug-responsive gene patterns are enriched in protein-coding variants (77%), regulated by different sets of transcriptional factors (including SRF/CREB1 and GR/CTCF) and expressed in separate cellular compartments of the brain. We found that alterations mediated by proximal promoters in neurons are more specific in the selection of regulated transcriptional isoforms compared with enhancer-dependent alterations in glia. The change in transcriptional programs, from housekeeping to inducible, provides cells with the resource of functionally distinct proteins. We conclude that the regulation of drug-induced brain plasticity may occur at the level of transcripts rather than genes. The expression of specific isoforms in response to antidepressants may constitute a trigger that initiates the long-lasting effects of these drugs.
Collapse
Affiliation(s)
- Marcin Piechota
- Department of Molecular Neuropharmacology, Institute of Pharmacology PAS, Krakow, Poland
| | - Slawomir Golda
- Department of Molecular Neuropharmacology, Institute of Pharmacology PAS, Krakow, Poland
| | - Joanna Ficek
- Department of Molecular Neuropharmacology, Institute of Pharmacology PAS, Krakow, Poland
| | - Danuta Jantas
- Department of Experimental Neuroendocrinology, Institute of Pharmacology PAS, Krakow, Poland
| | - Ryszard Przewlocki
- Department of Molecular Neuropharmacology, Institute of Pharmacology PAS, Krakow, Poland; Department of Neurobiology and Neuropsychology, IPS, UJ, Krakow, Poland
| | - Michal Korostynski
- Department of Molecular Neuropharmacology, Institute of Pharmacology PAS, Krakow, Poland.
| |
Collapse
|
25
|
Zhang Y, Brownstein AJ, Buonora M, Niikura K, Ho A, Correa da Rosa J, Kreek MJ, Ott J. Self administration of oxycodone alters synaptic plasticity gene expression in the hippocampus differentially in male adolescent and adult mice. Neuroscience 2014; 285:34-46. [PMID: 25446355 DOI: 10.1016/j.neuroscience.2014.11.013] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2014] [Revised: 10/19/2014] [Accepted: 11/04/2014] [Indexed: 11/16/2022]
Abstract
Abuse and addiction to prescription opioids such as oxycodone (a short-acting Mu opioid receptor (MOP-r) agonist) in adolescence is a pressing public health issue. We have previously shown differences in oxycodone self-administration behaviors between adolescent and adult C57BL/6J mice and expression of striatal neurotransmitter receptor genes, in areas involved in reward. In this study, we aimed to determine whether oxycodone self-administration differentially affects genes regulating synaptic plasticity in the hippocampus of adolescent compared to adult mice, since the hippocampus may be involved in learning aspects associated with chronic drug self administration. Hippocampus was isolated for mRNA analysis from mice that had self administered oxycodone (0.25 mg/kg/infusion) 2h/day for 14 consecutive days or from yoked saline controls. Gene expression was analyzed with real-time polymerase chain reaction (PCR) using a commercially available "synaptic plasticity" PCR array containing 84 genes. We found that adolescent and adult control mice significantly differed in the expression of several genes in the absence of oxycodone exposure, including those coding for mitogen-activated protein kinase, calcium/calmodulin-dependent protein kinase II gamma subunit, glutamate receptor, ionotropic AMPA2 and metabotropic 5. Chronic oxycodone self administration increased proviral integration site 1 (Pim1) and thymoma viral proto-oncogene 1 mRNA levels compared to controls in both age groups. Both Pim1 and cadherin 2 mRNAs showed a significant combined effect of Drug Condition and Age × Drug Condition. Furthermore, the mRNA levels of both cadherin 2 and cAMP response element modulators showed an experiment-wise significant difference between oxycodone and saline control in adult but not in adolescent mice. Overall, this study demonstrates for the first time that chronic oxycodone self-administration differentially alters synaptic plasticity gene expression in the hippocampus of adolescent and adult mice.
Collapse
Affiliation(s)
- Y Zhang
- The Laboratory of the Biology of Addictive Diseases, The Rockefeller University, New York, NY 10065, USA.
| | - A J Brownstein
- The Laboratory of the Biology of Addictive Diseases, The Rockefeller University, New York, NY 10065, USA
| | - M Buonora
- The Laboratory of the Biology of Addictive Diseases, The Rockefeller University, New York, NY 10065, USA
| | - K Niikura
- The Laboratory of the Biology of Addictive Diseases, The Rockefeller University, New York, NY 10065, USA
| | - A Ho
- The Laboratory of the Biology of Addictive Diseases, The Rockefeller University, New York, NY 10065, USA
| | - J Correa da Rosa
- Laboratory of Investigative Dermatology, The Rockefeller University, New York, NY 10065, USA
| | - M J Kreek
- The Laboratory of the Biology of Addictive Diseases, The Rockefeller University, New York, NY 10065, USA
| | - J Ott
- Institute of Psychology, Chinese Academy of Sciences, Beijing, China; The Laboratory of Statistical Genetics, The Rockefeller University, New York, NY, USA
| |
Collapse
|
26
|
Bitar MS, Al-Mulla F. Upregulation of CREM/ICER suppresses wound endothelial CRE-HIF-1α-VEGF-dependent signaling and impairs angiogenesis in type 2 diabetes. Dis Model Mech 2014; 8:65-80. [PMID: 25381014 PMCID: PMC4283651 DOI: 10.1242/dmm.017145] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Impaired angiogenesis and endothelial dysfunction in type 2 diabetes constitute dominant risk factors for non-healing wounds and most forms of cardiovascular disease. We propose that diabetes shifts the ‘angiogenic balance’ in favor of an excessive anti-angiogenic phenotype. Herein, we report that diabetes impairs in vivo sponge angiogenic capacity by decreasing VEGF expression and fibrovascular invasion, and reciprocally enhances the formation of angiostatic molecules, such as thrombospondins, NFκB and FasL. Defective in vivo angiogenesis prompted cellular studies in cultured endothelial cells derived from subcutaneous sponge implants (SIECs) of control and Goto-Kakizaki rats. Ensuing data from diabetic SIECs demonstrated a marked upregulation in cAMP-PKA-CREB signaling, possibly stemming from increased expression of adenylyl cyclase isoforms 3 and 8, and decreased expression of PDE3. Mechanistically, we found that oxidative stress and PKA activation in diabetes enhanced CREM/ICER expression. This reduces IRS2 cellular content by inhibiting cAMP response element (CRE) transcriptional activity. Consequently, a decrease in the activity of Akt-mTOR ensued with a concomitant reduction in the total and nuclear protein levels of HIF-1α. Limiting HIF-1α availability for the specific hypoxia response elements in diabetic SIECs elicited a marked reduction in VEGF expression, both at the mRNA and protein levels. These molecular abnormalities were illustrated functionally by a defect in various pro-angiogenic properties, including cell proliferation, migration and tube formation. A genetic-based strategy in diabetic SIECs using siRNAs against CREM/ICER significantly augmented the PKA-dependent VEGF expression. To this end, the current data identify the importance of CREM/ICER as a negative regulator of endothelial function and establish a link between CREM/ICER overexpression and impaired angiogenesis during the course of diabetes. Moreover, it could also point to CREM/ICER as a potential therapeutic target in the treatment of pathological angiogenesis.
Collapse
Affiliation(s)
- Milad S Bitar
- Department of Pharmacology and Toxicology, Kuwait University, Safat 13110, Kuwait.
| | - Fahd Al-Mulla
- Department of Pathology, Kuwait University, Safat 13110, Kuwait
| |
Collapse
|
27
|
Yanai S, Semba Y, Ito H, Endo S. Cilostazol improves hippocampus-dependent long-term memory in mice. Psychopharmacology (Berl) 2014; 231:2681-93. [PMID: 24464529 DOI: 10.1007/s00213-014-3442-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2013] [Accepted: 12/31/2013] [Indexed: 12/15/2022]
Abstract
RATIONALE Phosphodiesterases (PDEs) play an important role in the regulation of intracellular signaling mediated by cyclic adenosine monophosphate (cAMP). Recently, several PDE inhibitors were assessed for their possible cognitive enhancing properties. However, little is known about the effect of PDE3 inhibitors on memory function. OBJECTIVES We examined how the PDE3 inhibitor cilostazol affects C57BL/6 J mice as they perform various behavioral tasks. After behavioral assessment, brains of the mice were analyzed immunohistochemically to quantify the phosphorylation of cAMP-responsive element binding protein (CREB), a downstream component of the cAMP pathway. RESULTS Oral administration of cilostazol significantly enhanced recollection of the exact platform location in the Morris water maze probe test. Cilostazol also improved context-dependent long-term fear memory, without affecting short-term memory. No apparent effect was observed in cue-dependent fear memory. The results suggest that cilostazol selectively improves hippocampus-dependent long-term memory in these tasks. Cilostazol also significantly increased the number of phosphorylated-CREB-positive cells in hippocampal dentate gyrus. CONCLUSIONS These results suggest that cilostazol may exert its beneficial effects on learning and memory by enhancing the cAMP system in hippocampus, where it increases intracellular cAMP activity.
Collapse
Affiliation(s)
- Shuichi Yanai
- Aging Neuroscience Research Team, Tokyo Metropolitan Institute of Gerontology, Itabashi, Tokyo, 173-0015, Japan
| | | | | | | |
Collapse
|
28
|
Mikami N, Sakaguchi S. CD28 signals the differential control of regulatory T cells and effector T cells. Eur J Immunol 2014; 44:955-7. [PMID: 24652756 DOI: 10.1002/eji.201444513] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2014] [Revised: 02/27/2014] [Accepted: 03/17/2014] [Indexed: 01/08/2023]
Abstract
One possible means of driving antigen-specific immune suppression is to expand or induce antigen-specific FoxP3-expressing Treg cells. One way of activating and expanding these specialized cells, both in vitro and in vivo, is by strong costimulation via CD28 with an agonistic anti-CD28 monoclonal antibody, called anti-CD28 superagonist (CD28SA). However, CD28SA also strongly activates conventional T (Tconv) cells to secrete proinflammatory cytokines and, under certain conditions, causes serious cytokine release syndrome. In this issue of European Journal of Immunology, Tabares et al. [Eur. J. Immunol. 2014. 44: 1225-1236] address how CD28SA can be used for the differential control of human Treg and Tconv cells to suppress immune responses without serious adverse effects. They show that, depending on the dose of the antibody or by comedication of cortico-steroid, the selective expansion of Treg cells can be achieved without significantly activating Tconv cells to produce inflammatory cytokines. This difference in CD28 signal sensitivity between the two populations can be exploited for better control of immune responses.
Collapse
Affiliation(s)
- Norihisa Mikami
- Department of Experimental Immunology, Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | | |
Collapse
|
29
|
Guo J, Wang H, Wang Q, Chen Y, Chen S. Expression of p-CREB and activity-dependent miR-132 in temporal lobe epilepsy. Int J Clin Exp Med 2014; 7:1297-1306. [PMID: 24995086 PMCID: PMC4073747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2014] [Accepted: 04/23/2014] [Indexed: 06/03/2023]
Abstract
OBJECTIVE The mechanisms underlying the pathogenesis of intractable epilepsy (IE) are still unclear. This study aimed to investigate the role of phosphorylation of Cyclase Response Element Binding Protein (p-CREB)/microRNA-132 (miR-132) signal pathway in the epileptogenesis. METHODS Temporal lobe epilepsy (TLE) was induced in rodents, and the brain tissues were collected. P-CREB expression was detected by Western blot assay and immunochemistry at 6 h, 24 h, 3 d, 7 d, 14 d, 30 d and 60 d after induced status epilepsy (SE), respectively, and in patients with TLE. The expression of miR-132 was detected by RT-PCR. RESULTS The p-CREB and miR-132 were highly expressed in both rats and patients with TLE as compared to controls. CONCLUSIONS The expression of p-CREB and miR-132 increases in epilepsy rats and patients, suggesting a possible pathogenetic role of p-CREB and miR-132 in TLE via modulating the dendritic plasticity.
Collapse
Affiliation(s)
- Jing Guo
- Department of Neurology, The Affiliated Second Hospital of Chongqing Medical University76 Linjiang Road, Chongqing, 400010, China
| | - Heng Wang
- Department of Neurology, The Affiliated Second Hospital of Chongqing Medical University76 Linjiang Road, Chongqing, 400010, China
| | - Qian Wang
- Department of Neurology, The Affiliated Second Hospital of Chongqing Medical University76 Linjiang Road, Chongqing, 400010, China
| | - Yangmei Chen
- Department of Neurology, The Affiliated Second Hospital of Chongqing Medical University76 Linjiang Road, Chongqing, 400010, China
| | - Shengli Chen
- Chongqing Three Gorges Central HospitalChongqing, China
| |
Collapse
|
30
|
Hui Yin Y, Ahmad N, Makmor-Bakry M. Pathogenesis of epilepsy: challenges in animal models. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2013; 16:1119-32. [PMID: 24494063 PMCID: PMC3909622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/12/2013] [Accepted: 08/02/2013] [Indexed: 11/13/2022]
Abstract
Epilepsy is one of the most common chronic disorders affecting individuals of all ages. A greater understanding of pathogenesis in epilepsy will likely provide the basis fundamental for development of new antiepileptic therapies that aim to prevent the epileptogenesis process or modify the progression of epilepsy in addition to treatment of epilepsy symptomatically. Therefore, several investigations have embarked on advancing knowledge of the mechanism underlying epileptogenesis, understanding in mechanism of pharmacoresistance and discovering antiepileptogenic or disease-modifying therapy. Animal models play a crucial and significant role in providing additional insight into mechanism of epileptogenesis. With the help of these models, epileptogenesis process has been demonstrated to be involved in various molecular and biological pathways or processes. Hence, this article will discuss the known and postulated mechanisms of epileptogenesis and challenges in using the animal models.
Collapse
Affiliation(s)
- Yow Hui Yin
- Faculty of Pharmacy, University Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Nurulumi Ahmad
- Faculty of Pharmacy, University Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Mohd Makmor-Bakry
- Faculty of Pharmacy, University Kebangsaan Malaysia, Kuala Lumpur, Malaysia,Corresponding author: Mohd Makmor-Bakry. Faculty of Pharmacy, University Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, Kuala Lumpur 50300, Malaysia. Tel: +60392897244; Fax: +60326983271;
| |
Collapse
|
31
|
A mathematical model of pulse-coded hormone signal responses in pituitary gonadotroph cells. Math Biosci 2013; 246:38-46. [PMID: 24095971 DOI: 10.1016/j.mbs.2013.09.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2013] [Revised: 09/09/2013] [Accepted: 09/20/2013] [Indexed: 11/23/2022]
Abstract
Cells in the pituitary that synthesize luteinizing and follicle-stimulating hormones regulate the relative production of these two key reproductive hormones in response to signals from the hypothalamus. These signals are encoded in the frequency of gonadotrophin-releasing-hormone pulses. In vitro experiments with a murine-derived cell line have identified key elements of the processes that decode the signal to regulate transcription of the subunits encoding these hormones. The mathematical model described in this paper is based on the results of those experiments and advances quantitative understanding of the biochemical decoder. The model consists of non-linear differential equations for each of six processes that lead to the synthesis of follicle-stimulating hormone. Simulations of the model exhibit key characteristics found in the experiments, including a preference for follicle-stimulating hormone synthesis at low pulse frequencies and a loss of this characteristic when a mutation is introduced.
Collapse
|
32
|
Kádár E, Huguet G, Aldavert-Vera L, Morgado-Bernal I, Segura-Torres P. Intracranial self stimulation upregulates the expression of synaptic plasticity related genes and Arc protein expression in rat hippocampus. GENES BRAIN AND BEHAVIOR 2013; 12:771-9. [PMID: 23898803 DOI: 10.1111/gbb.12065] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/21/2013] [Revised: 05/31/2013] [Accepted: 07/25/2013] [Indexed: 11/30/2022]
Abstract
Post-training lateral hypothalamus (LH) intracranial self stimulation (ICSS) has a reliable enhancing effect on explicit memory formation evaluated in hippocampus-dependent tasks such as the Morris water maze. In this study, the effects of ICSS on gene expression in the hippocampus are examined 4.5 h post treatment by using oligonucleotide microarray and real-time PCR, and by measuring Arc protein levels in the different layers of hippocampal subfields through immunofluorescence. The microarray data analysis resulted in 65 significantly regulated genes in rat ICSS hippocampi compared to sham, including cAMP-mediated signaling as one of the most significantly enriched Database for Annotation, Visualization and Integrated Discovery (DAVID) functional categories. In particular, expression of CREB-dependent synaptic plasticity related genes (c-Fos, Arc, Bdnf, Ptgs-2 and Crem and Icer) was regulated in a time-dependent manner following treatment administration. Immunofluorescence results showed that ICSS treatment induced a significant increase in Arc protein expression in CA1 and DG hippocampal subfields. This empirical evidence supports our hypothesis that the effect of ICSS on improved or restored memory functions might be mediated by increased hippocampal expression of activity-dependent synaptic plasticity related genes, including Arc protein expression, as neural mechanisms related to memory consolidation.
Collapse
Affiliation(s)
- E Kádár
- Departament de Biologia, Universitat de Girona, Girona, Spain
| | | | | | | | | |
Collapse
|
33
|
Iacono G, Altafini C, Torre V. Early phase of plasticity-related gene regulation and SRF dependent transcription in the hippocampus. PLoS One 2013; 8:e68078. [PMID: 23935853 PMCID: PMC3720722 DOI: 10.1371/journal.pone.0068078] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2013] [Accepted: 05/25/2013] [Indexed: 02/07/2023] Open
Abstract
Hippocampal organotypic cultures are a highly reliable in vitro model for studying neuroplasticity: in this paper, we analyze the early phase of the transcriptional response induced by a 20 µM gabazine treatment (GabT), a GABA-Ar antagonist, by using Affymetrix oligonucleotide microarray, RT-PCR based time-course and chromatin-immuno-precipitation. The transcriptome profiling revealed that the pool of genes up-regulated by GabT, besides being strongly related to the regulation of growth and synaptic transmission, is also endowed with neuro-protective and pro-survival properties. By using RT-PCR, we quantified a time-course of the transient expression for 33 of the highest up-regulated genes, with an average sampling rate of 10 minutes and covering the time interval [10∶90] minutes. The cluster analysis of the time-course disclosed the existence of three different dynamical patterns, one of which proved, in a statistical analysis based on results from previous works, to be significantly related with SRF-dependent regulation (p-value<0.05). The chromatin immunoprecipitation (chip) assay confirmed the rich presence of working CArG boxes in the genes belonging to the latter dynamical pattern and therefore validated the statistical analysis. Furthermore, an in silico analysis of the promoters revealed the presence of additional conserved CArG boxes upstream of the genes Nr4a1 and Rgs2. The chip assay confirmed a significant SRF signal in the Nr4a1 CArG box but not in the Rgs2 CArG box.
Collapse
Affiliation(s)
- Giovanni Iacono
- Department of Functional Analysis, International School for Advanced Studies, Trieste, Italy
| | - Claudio Altafini
- Department of Functional Analysis, International School for Advanced Studies, Trieste, Italy
| | - Vincent Torre
- Department of Functional Analysis, International School for Advanced Studies, Trieste, Italy
- IIT Italian Institute of Technology, Genova, Italy
- * E-mail:
| |
Collapse
|
34
|
Senescent-induced dysregulation of cAMP/CREB signaling and correlations with cognitive decline. Brain Res 2013; 1516:93-109. [PMID: 23623816 DOI: 10.1016/j.brainres.2013.04.033] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2012] [Revised: 04/12/2013] [Accepted: 04/13/2013] [Indexed: 11/20/2022]
Abstract
It is well known that alongside senescence there is a gradual decline in cognitive ability, most noticeably certain kinds of memory such as working, episodic, spatial, and long term memory. However, until recently, not much has been known regarding the specific mechanisms responsible for the decline in cognitive ability with age. Over the past decades, researchers have become more interested in cAMP signaling, and its downstream transcription factor cAMP response element binding protein (CREB) in the context of senescence. However, there is still a lack of understanding on what ultimately causes the cognitive deficits observed with senescence. This review will focus on the changes in intracellular signaling in the brain, more specifically, alterations in cAMP/CREB signaling in aging. In addition, the downstream effects of altered cAMP signaling on cognitive ability with age will be further discussed. Overall, understanding the senescent-related changes that occur in cAMP/CREB signaling could be important for the development of novel drug targets for both healthy aging, and pathological aging such as Alzheimer's disease.
Collapse
|
35
|
Du B, Luo W, Li R, Tan B, Han H, Lu X, Li D, Qian M, Zhang D, Zhao Y, Liu M. Lgr4/Gpr48 negatively regulates TLR2/4-associated pattern recognition and innate immunity by targeting CD14 expression. J Biol Chem 2013; 288:15131-41. [PMID: 23589304 DOI: 10.1074/jbc.m113.455535] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The recognition of pathogen-associated molecular patterns by Toll-like receptors (TLRs) is pivotal in both innate and adaptive immune responses. Here we demonstrate that deletion of Lgr4/Gpr48 (G-protein-coupled receptor 48), a seven-transmembrane glycoprotein hormone receptor, potentiates TLR2/4-associated cytokine production and attenuates mouse resistance to septic shock. The expression of CD14, a co-receptor for TLR2/4-associated pathogen-associated molecular patterns, is increased significantly in Lgr4-deficient macrophages, which is consistent with the increased immune response, whereas the binding activity of cAMP-response element-binding protein is decreased significantly in Lgr4-deficient macrophages, which up-regulate the expression of CD14 at the transcriptional level. Together, our data demonstrate that Lgr4/Gpr48 plays a critical role in modulating the TLR2/4 signaling pathway and represents a useful therapeutic approach of targeting Lgr4/Gpr48 in TLR2/4-associated septic shock and autoimmune diseases.
Collapse
Affiliation(s)
- Bing Du
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, 500 Dongchuan Road, Shanghai 200241, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Zmrzljak UP, Korenčič A, Košir R, Goličnik M, Sassone-Corsi P, Rozman D. Inducible cAMP early repressor regulates the Period 1 gene of the hepatic and adrenal clocks. J Biol Chem 2013; 288:10318-10327. [PMID: 23443664 PMCID: PMC3624415 DOI: 10.1074/jbc.m112.445692] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2012] [Revised: 02/23/2013] [Indexed: 08/27/2023] Open
Abstract
Light, restricted feeding, and hormonal inputs may operate as time givers (zeitgebers) for the circadian clock within peripheral organs through the activation of tissue-specific signaling cascades. cAMP signaling through CREM (cAMP-responsive element modulator) and its variant ICER (inducible cAMP early repressor) is linked to the circadian regulation of pineal melatonin synthesis, although little is known about its influence in other organs. We performed experiments in the absence of light and feeding-time cues to test which core clock genes are controlled by CREM/ICER in the liver and adrenal gland. In vivo, Crem loss-of-function mutation resulted in fine-tuning of all measured adrenal clock genes (Per1/2/3, Cry1/2, Bmal1, and Rev-erbα), whereas only Per1 and Cry1 were affected in the liver. Icer expression was circadian in the adrenal gland, with peak gene expression at zeitgeber 12 and the highest protein levels at zeitgeber ∼20. The expression of both Icer and Per1 genes responded to cAMP stimuli in an immediate-early fashion. In immortal cells, forskolin induced expression of Per1 after 2 h, and de novo protein synthesis led to Per1 attenuation. We show that the de novo synthesized protein responsible for Per1 attenuation is ICER. Indeed, Per1 expression is up-regulated in cells ectopically expressing antisense Icer, and mobility shift experiments identified ICER binding to cAMP-responsive elements of the Per1 promoter. We propose that ICER acts as a noise filter for different signals that could affect transcription in the adrenal gland. Because ICER is an immediate-early repressor, the circadian nature of adrenal Icer expression could serve a role in a time-dependent gating mechanism.
Collapse
Affiliation(s)
- Uršula Prosenc Zmrzljak
- From the Center for Functional Genomics and Bio-Chips, Institute of Biochemistry, Faculty of Medicine, University of Ljubljana, SI-1000 Ljubljana, Slovenia
- the Department of Laboratory Diagnostics, Institute of Oncology, SI-1000 Ljubljana, Slovenia
| | - Anja Korenčič
- From the Center for Functional Genomics and Bio-Chips, Institute of Biochemistry, Faculty of Medicine, University of Ljubljana, SI-1000 Ljubljana, Slovenia
| | - Rok Košir
- From the Center for Functional Genomics and Bio-Chips, Institute of Biochemistry, Faculty of Medicine, University of Ljubljana, SI-1000 Ljubljana, Slovenia
- DiaGenomi Limited, 1000 Ljubljana, Slovenia, and
| | - Marko Goličnik
- From the Center for Functional Genomics and Bio-Chips, Institute of Biochemistry, Faculty of Medicine, University of Ljubljana, SI-1000 Ljubljana, Slovenia
| | - Paolo Sassone-Corsi
- the Department of Biological Chemistry, University of California, Irvine, California 02607
| | - Damjana Rozman
- From the Center for Functional Genomics and Bio-Chips, Institute of Biochemistry, Faculty of Medicine, University of Ljubljana, SI-1000 Ljubljana, Slovenia
| |
Collapse
|
37
|
Bhowmik M, Khanam R, Vohora D. Histamine H3 receptor antagonists in relation to epilepsy and neurodegeneration: a systemic consideration of recent progress and perspectives. Br J Pharmacol 2012; 167:1398-1414. [PMID: 22758607 PMCID: PMC3514756 DOI: 10.1111/j.1476-5381.2012.02093.x] [Citation(s) in RCA: 95] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2012] [Revised: 05/03/2012] [Accepted: 06/12/2012] [Indexed: 12/22/2022] Open
Abstract
The central histaminergic actions are mediated by H(1) , H(2) , H(3) and H(4) receptors. The histamine H(3) receptor regulates the release of histamine and a number of other neurotransmitters and thereby plays a role in cognitive and homeostatic processes. Elevated histamine levels suppress seizure activities and appear to confer neuroprotection. The H(3) receptors have a number of enigmatic features like constitutive activity, interspecies variation, distinct ligand binding affinities and differential distribution of prototypic splice variants in the CNS. Furthermore, this Gi/Go-protein-coupled receptor modulates several intracellular signalling pathways whose involvement in epilepsy and neurotoxicity are yet to be ascertained and hence represent an attractive target in the search for new anti-epileptogenic drugs. So far, H(3) receptor antagonists/inverse agonists have garnered a great deal of interest in view of their promising therapeutic properties in various CNS disorders including epilepsy and related neurotoxicity. However, a number of experiments have yielded opposing effects. This article reviews recent works that have provided evidence for diverse mechanisms of antiepileptic and neuroprotective effects that were observed in various experimental models both in vitro and in vivo. The likely reasons for the apparent disparities arising from the literature are also discussed with the aim of establishing a more reliable basis for the future use of H(3) receptor antagonists, thus improving their utility in epilepsy and associated neurotoxicity.
Collapse
Affiliation(s)
- M Bhowmik
- Neurobehavioral Pharmacology Laboratory, Department of Pharmacology, Faculty of Pharmacy, Jamia Hamdard (Hamdard University), New Delhi, India
| | | | | |
Collapse
|
38
|
Johansson EM, Sanabra C, Mengod G. Sex-related differences of cAMP-specific PDE4B3 mRNA in oligodendrocytes following systemic inflammation. Glia 2012; 60:1815-25. [PMID: 22848007 DOI: 10.1002/glia.22399] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2012] [Accepted: 07/05/2012] [Indexed: 01/28/2023]
Abstract
Sex-related differences have been observed in the incidence and severity of several neurological diseases and in sepsis in humans. Cyclic adenosine monophosphate (cAMP) has been shown to play an important role in modulating the inflammatory environment during neuroinflammation and importantly in protecting myelin from excitotoxic cell death. Considering the sexual dimorphism in the functional properties of oligodendrocytes and the importance of a systemic inflammation in the progression of multiple sclerosis, we focused on identifying possible sex-related differences in the alterations previously reported for the two phosphodiesterase4B (PDE4B) splice-variants (PDE4B2 and PDE4B3) mRNA expression during innate neuroinflammation. PDE4A, PDE4B, and PDE4D are present in oligodendrocytes and we have previously reported that PDE4B3 mRNA is readily expressed in both oligodendrocytes and neurons. In this study, we analyzed the influence of an intraperitoneal lipopolysaccharide injection on the distribution pattern and expression levels of the PDE4B mRNA splicing variants in both male and female mice brains. Clear differences were observed in PDE4B2 and PDE4B3 mRNA expression levels in males compared with females in a time-dependent manner. Furthermore, we observed that the clear downregulation of PDE4B3 mRNA was reflected in a lower percentage of oligodendrocytes positive for this transcript which correlated with a decrease in inducible cAMP early repressor expression in female corpus callosum.
Collapse
Affiliation(s)
- Emily M Johansson
- Departament de Neuroquímica i Neurofarmacologia, Institut d'Investigacions Biomèdiques de Barcelona (CSIC), IDIBAPS, CIBERNED, Barcelona, Spain
| | | | | |
Collapse
|
39
|
Pruunsild P, Timmusk T. Subcellular localization and transcription regulatory potency of KCNIP/Calsenilin/DREAM/KChIP proteins in cultured primary cortical neurons do not provide support for their role in CRE-dependent gene expression. J Neurochem 2012; 123:29-43. [PMID: 22612322 DOI: 10.1111/j.1471-4159.2012.07796.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
KCNIP3/KChIP3 (voltage-dependent K+ channel interacting protein 3), alias Calsenilin and downstream regulatory element antagonist modulator (DREAM), is a multifunctional protein that modulates A-type potassium channels, affects processing of amyloid precursor protein and regulates transcription. KCNIP3 has been described to negatively influence the activity of CREB (cAMP/Ca(2+)-response element binding protein), an essential factor in neuronal activity-dependent gene expression regulation. However, reports on intracellular localization of KCNIP3 in neurons are diverse and necessitate additional analyses of distribution of KCNIPs in cells to clarify the potential of KCNIP3 to fulfill its functions in different cell compartments. Here, we examined localization of the entire family of highly similar KCNIP proteins in neuronal cells and show that over-expressed isoforms of KCNIP1/KChIP1, KCNIP2/KChIP2, KCNIP3/KChIP3, and KCNIP4/KChIP4 had varied, yet partially overlapping subcellular localization. In addition, although some of the over-expressed KCNIP isoforms localized to the nucleus, endogenous KCNIPs were not detected in nuclei of rat primary cortical neurons. Moreover, we analyzed the role of KCNIP proteins in cAMP/Ca(2+)-response element (CRE)-dependent transcription by luciferase reporter assay and electrophoretic mobility shift assay and report that our results do not support the role for KCNIPs, including DREAM/Calsenilin/KChIP3, in modulation of CREB-mediated transcription in neurons.
Collapse
Affiliation(s)
- Priit Pruunsild
- Department of Gene Technology, Tallinn University of Technology, Estonia.
| | | |
Collapse
|
40
|
Lantz CL, Wang W, Medina AE. Early alcohol exposure disrupts visual cortex plasticity in mice. Int J Dev Neurosci 2012; 30:351-7. [PMID: 22617459 DOI: 10.1016/j.ijdevneu.2012.05.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2012] [Revised: 04/20/2012] [Accepted: 05/05/2012] [Indexed: 12/30/2022] Open
Abstract
There is growing evidence that deficits in neuronal plasticity underlie the cognitive problems seen in fetal alcohol spectrum disorders (FASD). However, the mechanisms behind these deficits are not clear. Here we test the effects of early alcohol exposure on ocular dominance plasticity (ODP) in mice and the reversibility of these effects by phosphodiesterase (PDE) inhibitors. Mouse pups were exposed to 5 g/kg of 25% ethanol i.p. on postnatal days (P) 5, 7 and 9. This type of alcohol exposure mimics binge drinking during the third trimester equivalent of human gestation. To assess ocular dominance plasticity animals were monocularly deprived at P21 for 10 days, and tested using optical imaging of intrinsic signals. During the period of monocular deprivation animals were treated with vinpocetine (20mg/kg; PDE1 inhibitor), rolipram (1.25mg/kg; PDE4 inhibitor), vardenafil (3mg/kg; PDE5 inhibitor) or vehicle solution. Monocular deprivation resulted in the expected shift in ocular dominance of the binocular zone in saline controls but not in the ethanol group. While vinpocetine successfully restored ODP in the ethanol group, rolipram and vardenafil did not. However, when rolipram and vardenafil were given simultaneously ODP was restored. PDE4 and PDE5 are specific to cAMP and cGMP respectively, while PDE1 acts on both of these nucleotides. Our findings suggest that the combined activation of the cAMP and cGMP cascades may be a good approach to improve neuronal plasticity in FASD models.
Collapse
Affiliation(s)
- Crystal L Lantz
- Department of Anatomy and Neurobiology, School of Medicine, Virginia Commonwealth University, Richmond, VA 23298, United States
| | | | | |
Collapse
|
41
|
Košir R, Zmrzljak UP, Bele T, Acimovic J, Perse M, Majdic G, Prehn C, Adamski J, Rozman D. Circadian expression of steroidogenic cytochromes P450 in the mouse adrenal gland--involvement of cAMP-responsive element modulator in epigenetic regulation of Cyp17a1. FEBS J 2012; 279:1584-1593. [PMID: 21883931 DOI: 10.1111/j.1742-4658.2011.08317.x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The cytochrome P450 (CYP) genes Cyp51, Cyp11a1, Cyp17a1, Cyb11b1, Cyp11b2 and Cyp21a1 are involved in the adrenal production of corticosteroids, whose circulating levels are circadian. cAMP signaling plays an important role in adrenal steroidogenesis. By using cAMP responsive element modulator (Crem) knockout mice, we show that CREM isoforms contribute to circadian expression of steroidogenic CYPs in the mouse adrenal gland. Most striking was the CREM-dependent hypomethylation of the Cyp17a1 promoter at zeitgeber time 12, which resulted in higher Cyp17a1 mRNA and protein expression in the knockout adrenal glands. The data indicate that products of the Crem gene control the epigenetic repression of Cyp17 in mouse adrenal glands.
Collapse
Affiliation(s)
- Rok Košir
- Center for Functional Genomics and Bio-Chips, Institute of Biochemistry, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Oncogenic K-ras expression is associated with derangement of the cAMP/PKA pathway and forskolin-reversible alterations of mitochondrial dynamics and respiration. Oncogene 2012; 32:352-62. [PMID: 22410778 DOI: 10.1038/onc.2012.50] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The Warburg effect in cancer cells has been proposed to involve several mechanisms, including adaptation to hypoxia, oncogenes activation or loss of oncosuppressors and impaired mitochondrial function. In previous papers, it has been shown that K-ras transformed mouse cells are much more sensitive as compared with normal cells to glucose withdrawal (undergoing apoptosis) and present a high glycolytic rate and a strong reduction of mitochondrial complex I. Recent observations suggest that transformed cells have a derangement in the cyclic adenosine monophosphate/cAMP-dependent protein kinase (cAMP/PKA) pathway, which is known to regulate several mitochondrial functions. Herein, the derangement of the cAMP/PKA pathway and its impact on transformation-linked changes of mitochondrial functions is investigated. Exogenous stimulation of PKA activity, achieved by forskolin treatment, protected K-ras-transformed cells from apoptosis induced by glucose deprivation, enhanced complex I activity, intracellular adenosine triphosphate (ATP) levels, mitochondrial fusion and decreased intracellular reactive oxygen species (ROS) levels. Several of these effects were almost completely prevented by inhibiting the PKA activity. Short-time treatment with compounds favoring mitochondrial fusion strongly decreased the cellular ROS levels especially in transformed cells. These findings support the notion that glucose shortage-induced apoptosis, specific of K-ras-transformed cells, is associated to a derangement of PKA signaling that leads to mitochondrial complex I decrease, reduction of ATP formation, prevalence of mitochondrial fission over fusion, and thereby opening new approaches for development of anticancer drugs.
Collapse
|
43
|
cAMP response element-binding protein is a primary hub of activity-driven neuronal gene expression. J Neurosci 2012; 31:18237-50. [PMID: 22171029 DOI: 10.1523/jneurosci.4554-11.2011] [Citation(s) in RCA: 93] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Long-lasting forms of neuronal plasticity require de novo gene expression, but relatively little is known about the events that occur genome-wide in response to activity in a neuronal network. Here, we unveil the gene expression programs initiated in mouse hippocampal neurons in response to different stimuli and explore the contribution of four prominent plasticity-related transcription factors (CREB, SRF, EGR1, and FOS) to these programs. Our study provides a comprehensive view of the intricate genetic networks and interactions elicited by neuronal stimulation identifying hundreds of novel downstream targets, including novel stimulus-associated miRNAs and candidate genes that may be differentially regulated at the exon/promoter level. Our analyses indicate that these four transcription factors impinge on similar biological processes through primarily non-overlapping gene-expression programs. Meta-analysis of the datasets generated in our study and comparison with publicly available transcriptomics data revealed the individual and collective contribution of these transcription factors to different activity-driven genetic programs. In addition, both gain- and loss-of-function experiments support a pivotal role for CREB in membrane-to-nucleus signal transduction in neurons. Our data provide a novel resource for researchers wanting to explore the genetic pathways associated with activity-regulated neuronal functions.
Collapse
|
44
|
Bieganska K, Figiel I, Gierej D, Kaczmarek L, Klejman A. Silencing of ICERs (Inducible cAMP Early Repressors) results in partial protection of neurons from programmed cell death. Neurobiol Dis 2012; 45:701-10. [DOI: 10.1016/j.nbd.2011.10.015] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2011] [Revised: 09/21/2011] [Accepted: 10/16/2011] [Indexed: 11/30/2022] Open
|
45
|
Patel N, Klassert TE, Greco SJ, Patel SA, Munoz JL, Reddy BY, Bryan M, Campbell N, Kokorina N, Sabaawy HE, Rameshwar P. Developmental regulation of TAC1 in peptidergic-induced human mesenchymal stem cells: implication for spinal cord injury in zebrafish. Stem Cells Dev 2012; 21:308-20. [PMID: 21671725 PMCID: PMC3258436 DOI: 10.1089/scd.2011.0179] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2011] [Accepted: 06/10/2011] [Indexed: 02/06/2023] Open
Abstract
Human mesenchymal stem cells (MSCs) are easy to expand, are relatively safe, and can be transplanted in allogeneic recipients as off-the-shelf cells. MSCs can be induced to form functional peptidergic neurons and express the neurotransmitter gene, TAC1. Expression of TAC1 requires that the repressor gene, RE-1 silencing transcription factor (REST), is decreased. This study investigated the molecular pathway in TAC1 induction as MSCs differentiated into neurons and then applied the findings in a model of spinal cord injury (SCI) in zebrafish. We studied the developmental roles of the 2 cAMP response element (CRE) sites: CRE1 and CRE2. Activator protein-1 (AP-1) binding site overlaps with CRE2 (CRE2/AP-1). Reporter gene studies with the 5' regulatory region of TAC1 containing wild-type or mutant CRE sites and, parallel studies with ectopically expressed inhibitor of cAMP proteins (inducible cAMP early repressor) indicated that CRE1 and CRE2/AP-1 are activated at days 6 and 12, respectively. Studies with protein kinase-A (PKA) and Jun N-terminal kinase (JNK) inhibitors in the reporter gene studies, chromatin immunoprecipation assay, and ectopic expression of REST indicated the following pathways: Decrease of REST activated upstream c-Jun N-terminal kinase (JNK). In turn, JNK activated ATF-2 and AP-1 for interaction with CRE1 and CRE2/AP-1, respectively. To apply the finding to SCI, we transplanted 6-day-induced MSCs in transgenic HB9-GFP zebrafish larvae with SCI, in the presence or absence of JNK inhibitors. Imaging and functional studies showed significant improvement in the fish. The repair mechanism involved the activation of JNK. The findings have long-term implications for SCI repair with MSCs.
Collapse
Affiliation(s)
- Nitixa Patel
- UMDNJ–New Jersey Medical School, Newark, New Jersey
| | - Tilman E. Klassert
- Instituto de Enfermedades Tropicales y Salud Pública, Universito of La Laguna, Canary Islands, Spain
| | | | | | | | | | | | - Neil Campbell
- UMDNJ–Robert Wood Johnson Medical School and Cancer Institute of New Jersey, New Brunswick, New Jersey
| | - Natalia Kokorina
- UMDNJ–Robert Wood Johnson Medical School and Cancer Institute of New Jersey, New Brunswick, New Jersey
| | - Hatem E. Sabaawy
- UMDNJ–Robert Wood Johnson Medical School and Cancer Institute of New Jersey, New Brunswick, New Jersey
| | | |
Collapse
|
46
|
|
47
|
Yu JZ, Rasenick MM. Receptor signaling and the cell biology of synaptic transmission. HANDBOOK OF CLINICAL NEUROLOGY 2012; 106:9-35. [PMID: 22608613 DOI: 10.1016/b978-0-444-52002-9.00002-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
This volume describes a series of psychiatric and neuropsychiatric disorders, connects some aspects of somatic and psychiatric medicine, and describes various current and emerging therapies. The purpose of this chapter is to set the stage for the volume by developing the theoretical basis of synaptic transmission and introducing the various neurotransmitters and their receptors involved in the process. The intent is to provide not only a historical context through which to understand neurotransmitters, but a current contextual basis for understanding neuronal signal transduction and applying this knowledge to facilitate treatment of maladies of the brain and mind.
Collapse
Affiliation(s)
- Jiang-Zhou Yu
- Department of Physiology, University of Illinois, Chicago, IL, USA
| | | |
Collapse
|
48
|
Zhu X, Han X, Blendy JA, Porter BE. Decreased CREB levels suppress epilepsy. Neurobiol Dis 2012; 45:253-63. [PMID: 21867753 PMCID: PMC4011562 DOI: 10.1016/j.nbd.2011.08.009] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2011] [Revised: 07/27/2011] [Accepted: 08/03/2011] [Indexed: 01/14/2023] Open
Abstract
Epilepsy is a common neurologic disorder yet no treatments aimed at preventing epilepsy have been developed. Several molecules including genes containing cAMP response elements (CREs) in their promoters have been identified that contribute to the development of epilepsy, a process called epileptogenesis. When phosphorylated cAMP response element binding protein (CREB) increases transcription from CRE regulated promoters. CREB phosphorylation is increased in rodent epilepsy models, and in the seizure onset region of humans with medically intractable epilepsy (Rakhade et al., 2005; Lee et al., 2007; Lund et al., 2008). Here we show that mice with decreased CREB levels (CREB(α∆) mutants) have a ~50% reduction in spontaneous seizures following pilocarpine induced status epilepticus (SE) and require more stimulation to electrically kindle. Following SE, brain derived neurotrophic factor (BDNF) and inducible cAMP early repressor (ICER) mRNAs are differentially up-regulated in the hippocampus and cortex of the CREB(α∆) mutants compared to wild-type mice, which may be contributing to differences in the severity of epilepsy. In contrast, we found no difference in KCC2 mRNA levels between the CREB(α∆) and wild-type mice after SE. The mechanism by which BDNF and ICER mRNAs increase specifically in the CREB(α∆) compared to wild-type mice following SE is not known. We did, however, find an increase in specific cAMP response element modulator (CREM) mRNA transcripts in the CREB(α∆) mutants that might be responsible for the differential regulation of BDNF and ICER after SE. Altering CREB activity following a neurologic insult provides a therapeutic strategy for modifying epileptogenesis.
Collapse
Affiliation(s)
- Xinjian Zhu
- The Children’s Hospital of Philadelphia, Division of Neurology
| | - Xiao Han
- The Children’s Hospital of Philadelphia, Division of Neurology
| | - Julie A. Blendy
- University of Pennsylvania, Department of Pharmacology, TRL Building, 125 S. 31st Street, Philadelphia, PA 19104-3403,
| | - Brenda E. Porter
- The Children’s Hospital of Philadelphia, Division of Neurology
- University of Pennsylvania, Department of Neurology and Pediatrics
| |
Collapse
|
49
|
Kumar V, Fahey PG, Jong YJI, Ramanan N, O'Malley KL. Activation of intracellular metabotropic glutamate receptor 5 in striatal neurons leads to up-regulation of genes associated with sustained synaptic transmission including Arc/Arg3.1 protein. J Biol Chem 2011; 287:5412-25. [PMID: 22179607 DOI: 10.1074/jbc.m111.301366] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
The G-protein coupled receptor, metabotropic glutamate receptor 5 (mGluR5), is expressed on both cell surface and intracellular membranes in striatal neurons. Using pharmacological tools to differentiate membrane responses, we previously demonstrated that cell surface mGluR5 triggers rapid, transient cytoplasmic Ca(2+) rises, resulting in c-Jun N-terminal kinase, Ca(2+)/calmodulin-dependent protein kinase, and cyclic adenosine 3',5'-monophosphate-responsive element-binding protein (CREB) phosphorylation, whereas stimulation of intracellular mGluR5 induces long, sustained Ca(2+) responses leading to the phosphorylation of extracellular signal-regulated kinase (ERK1/2) and Elk-1 (Jong, Y. J., Kumar, V., and O'Malley, K. L. (2009) J. Biol. Chem. 284, 35827-35838). Using pharmacological, genetic, and bioinformatics approaches, the current findings show that both receptor populations up-regulate many immediate early genes involved in growth and differentiation. Activation of intracellular mGluR5 also up-regulates genes involved in synaptic plasticity including activity-regulated cytoskeletal-associated protein (Arc/Arg3.1). Mechanistically, intracellular mGluR5-mediated Arc induction is dependent upon extracellular and intracellular Ca(2+) and ERK1/2 as well as calmodulin-dependent kinases as known chelators, inhibitors, and a dominant negative Ca(2+)/calmodulin-dependent protein kinase II construct block Arc increases. Moreover, intracellular mGluR5-induced Arc expression requires the serum response transcription factor (SRF) as wild type but not SRF-deficient neurons show this response. Finally, increased Arc levels due to high K(+) depolarization is significantly reduced in response to a permeable but not an impermeable mGluR5 antagonist. Taken together, these data highlight the importance of intracellular mGluR5 in the cascade of events associated with sustained synaptic transmission.
Collapse
Affiliation(s)
- Vikas Kumar
- Department of Anatomy and Neurobiology, Washington University School of Medicine, St. Louis, Missouri 63110, USA
| | | | | | | | | |
Collapse
|
50
|
Lyons MR, West AE. Mechanisms of specificity in neuronal activity-regulated gene transcription. Prog Neurobiol 2011; 94:259-95. [PMID: 21620929 PMCID: PMC3134613 DOI: 10.1016/j.pneurobio.2011.05.003] [Citation(s) in RCA: 154] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2011] [Revised: 05/05/2011] [Accepted: 05/05/2011] [Indexed: 02/06/2023]
Abstract
The brain is a highly adaptable organ that is capable of converting sensory information into changes in neuronal function. This plasticity allows behavior to be accommodated to the environment, providing an important evolutionary advantage. Neurons convert environmental stimuli into long-lasting changes in their physiology in part through the synaptic activity-regulated transcription of new gene products. Since the neurotransmitter-dependent regulation of Fos transcription was first discovered nearly 25 years ago, a wealth of studies have enriched our understanding of the molecular pathways that mediate activity-regulated changes in gene transcription. These findings show that a broad range of signaling pathways and transcriptional regulators can be engaged by neuronal activity to sculpt complex programs of stimulus-regulated gene transcription. However, the shear scope of the transcriptional pathways engaged by neuronal activity raises the question of how specificity in the nature of the transcriptional response is achieved in order to encode physiologically relevant responses to divergent stimuli. Here we summarize the general paradigms by which neuronal activity regulates transcription while focusing on the molecular mechanisms that confer differential stimulus-, cell-type-, and developmental-specificity upon activity-regulated programs of neuronal gene transcription. In addition, we preview some of the new technologies that will advance our future understanding of the mechanisms and consequences of activity-regulated gene transcription in the brain.
Collapse
Affiliation(s)
- Michelle R Lyons
- Department of Neurobiology, Duke University Medical Center, Durham, NC 27710, USA
| | | |
Collapse
|