1
|
Zhao K, Zhang L, Lei M, Jin Z, Du T, Zhang H, Sheng Y, Hu Z, Wang S, Ma C. A specific negatively charged sequence confers intramolecular regulation on Munc13-1 function in synaptic exocytosis. Proc Natl Acad Sci U S A 2025; 122:e2508915122. [PMID: 40489622 DOI: 10.1073/pnas.2508915122] [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: 04/15/2025] [Accepted: 04/30/2025] [Indexed: 06/11/2025] Open
Abstract
Munc13 family proteins are crucial for the secretion of neurotransmitters and hormones necessary for cell communication. They share a conserved C-terminal region that includes C2 and the MUN domains, which facilitate membrane interactions and the assembly of soluble N-ethylmaleimide sensitive factor attachment protein receptor (SNARE) complexes. Neuronal isoforms of Munc13 possess a variable N-terminal region that is essential for neurotransmitter release and short-term plasticity, although the precise functions of this region remain not fully understood. Here, we identified a negatively charged sequence within the N terminus of Munc13-1, termed polyE, which is specific to Munc13-1 among all Munc13 isoforms and potentially derived from a common ancestor of homeotherms. We found that polyE binds significantly to the MUN domain through charge-charge interactions, inhibiting MUN activity in promoting SNARE complex assembly. Disrupting the polyE-MUN interaction by introducing pseudophosphorylated mutations in the MUN domain alleviates this inhibition, thereby enhancing neurotransmitter release. Strikingly, Ca2+ ions exhibit significant binding to polyE. We found that 40 μM of Ca2+ adequately competes with the polyE-MUN interaction to reduce polyE inhibition. This concentration is comparable to presynaptic local [Ca2+]i triggered by a single action potential. Taken together, these results indicate an autoinhibition conformation of Munc13-1 mediated by the polyE-MUN interaction. In addition, the relief of this autoinhibition conformation of Munc13-1 by presynaptic Ca2+ influx and/or posttranslational modifications in the MUN domain may underlie Munc13-1 function in neurotransmitter release and short-term plasticity.
Collapse
Affiliation(s)
- Kexu Zhao
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Li Zhang
- Key Clinical Laboratory of Henan Province, Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Mengshi Lei
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Ziqi Jin
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Tianxin Du
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Hong Zhang
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Yin Sheng
- School of Artificial Intelligence and Automation, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Zhitao Hu
- Department of Neuroscience, City University of Hong Kong, Kowloon 999077, China
| | - Shen Wang
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
- Hubei Bioinformatics and Molecular Imaging Key Laboratory, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Cong Ma
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
- Hubei Key Laboratory of Brain-inspired Intelligent Systems, School of Artificial Intelligence and Automation, Huazhong University of Science and Technology, Wuhan 430074, China
- Key Laboratory of Neurogenetics and Channelopathies of the Ministry of Education, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou 510260, China
| |
Collapse
|
2
|
Brenna A, Borsa M, Saro G, Ripperger JA, Glauser DA, Yang Z, Adamantidis A, Albrecht U. Cyclin-dependent kinase 5 (Cdk5) activity is modulated by light and gates rapid phase shifts of the circadian clock. eLife 2025; 13:RP97029. [PMID: 39937180 PMCID: PMC11820109 DOI: 10.7554/elife.97029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/13/2025] Open
Abstract
The circadian clock enables organisms to synchronize biochemical and physiological processes over a 24 hr period. Natural changes in lighting conditions, as well as artificial disruptions like jet lag or shift work, can advance or delay the clock phase to align physiology with the environment. Within the suprachiasmatic nucleus (SCN) of the hypothalamus, circadian timekeeping and resetting rely on both membrane depolarization and intracellular second-messenger signaling. Voltage-gated calcium channels (VGCCs) facilitate calcium influx in both processes, activating intracellular signaling pathways that trigger Period (Per) gene expression. However, the precise mechanism by which these processes are concertedly gated remains unknown. Our study in mice demonstrates that cyclin-dependent kinase 5 (Cdk5) activity is modulated by light and regulates phase shifts of the circadian clock. We observed that knocking down Cdk5 in the SCN of mice affects phase delays but not phase advances. This is linked to uncontrolled calcium influx into SCN neurons and an unregulated protein kinase A (PKA)-calcium/calmodulin-dependent kinase (CaMK)-cAMP response element-binding protein (CREB) signaling pathway. Consequently, genes such as Per1 are not induced by light in the SCN of Cdk5 knock-down mice. Our experiments identified Cdk5 as a crucial light-modulated kinase that influences rapid clock phase adaptation. This finding elucidates how light responsiveness and clock phase coordination adapt activity onset to seasonal changes, jet lag, and shift work.
Collapse
Affiliation(s)
- Andrea Brenna
- Department of Biology, University of FribourgFribourgSwitzerland
- Department of Endocrinology, Metabolism, and Cardiovascular System, Section of Medicine, University of FribourgFribourgSwitzerland
| | - Micaela Borsa
- Zentrum für Experimentelle Neurologie, Department of Neurology, Inselspital, Bern University Hospital, University of BernBernSwitzerland
- Department of Biomedical Research, University of BernBernSwitzerland
| | - Gabriella Saro
- Department of Biology, University of FribourgFribourgSwitzerland
| | | | | | - Zhihong Yang
- Department of Endocrinology, Metabolism, and Cardiovascular System, Section of Medicine, University of FribourgFribourgSwitzerland
| | - Antoine Adamantidis
- Zentrum für Experimentelle Neurologie, Department of Neurology, Inselspital, Bern University Hospital, University of BernBernSwitzerland
- Department of Biomedical Research, University of BernBernSwitzerland
| | - Urs Albrecht
- Department of Biology, University of FribourgFribourgSwitzerland
| |
Collapse
|
3
|
Basurto‐Islas G, Diaz MC, Ocampo LMZ, Martínez‐Herrera M, López‐Camacho PY. Natural products against tau hyperphosphorylation-induced aggregates: Potential therapies for Alzheimer's disease. Arch Pharm (Weinheim) 2025; 358:e2400721. [PMID: 39888017 PMCID: PMC11781347 DOI: 10.1002/ardp.202400721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 12/17/2024] [Accepted: 12/18/2024] [Indexed: 02/01/2025]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder characterized by cognitive decline and memory impairments and is considered the most prevalent form of dementia. Among the contributing factors to AD lies the hyperphosphorylation of the microtubule-associated protein tau. Phosphorylated tau reduces its affinity for microtubules and triggers other posttranslational modifications that result in its aggregation and assembly into filaments. These structures progressively accumulate within neurons leading to neurodegeneration. While current AD medications often involve undesirable side effects, the exploration of natural products as a potential therapeutic alternative has gained considerable attention. Numerous compounds have shown potential capacity for reducing tau pathology through different mechanisms, such as inhibiting kinases to reduce tau hyperphosphorylation, enhancing phosphatase activity, and blocking fibril formation. Since tau hyperphosphorylation-induced aggregation is pivotal in AD onset, this review aims to elucidate the potential of natural products in modulating this crucial molecular mechanism.
Collapse
Affiliation(s)
| | | | | | - Melchor Martínez‐Herrera
- Departamento de Ciencias NaturalesUniversidad Autónoma Metropolitana CuajimalpaCiudad de MéxicoMexico
| | - Perla Y. López‐Camacho
- Departamento de Ciencias NaturalesUniversidad Autónoma Metropolitana CuajimalpaCiudad de MéxicoMexico
| |
Collapse
|
4
|
Hernández-Echeagaray E, Miranda-Barrientos JA, Nieto-Mendoza E, Torres-Cruz FM. Exploring the role of Cdk5 on striatal synaptic plasticity in a 3-NP-induced model of early stages of Huntington's disease. Front Mol Neurosci 2024; 17:1362365. [PMID: 39569019 PMCID: PMC11576431 DOI: 10.3389/fnmol.2024.1362365] [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: 12/28/2023] [Accepted: 10/09/2024] [Indexed: 11/22/2024] Open
Abstract
Impaired mitochondrial function has been associated with the onset of neurodegenerative diseases. Specifically, certain mitochondrial toxins, such as 3-nitropropionic acid (3-NP), initiate cellular changes within the striatum that closely resemble the pathology observed in Huntington's disease (HD). Among the pivotal signaling molecules contributing to neurodegeneration, cyclin-dependent kinase 5 (Cdk5) stands out. In particular, Cdk5 has been implicated not only in cellular pathology but also in the modulation of synaptic plasticity. Given its widespread presence in the striatum, this study seeks to elucidate the potential role of Cdk5 in the induction of corticostriatal synaptic plasticity in murine striatal cells subjected to subchronic doses of 3-NP in vivo, aiming to mimic the early stages of HD. Immunostaining analyses revealed an increase in Cdk5 in tissues from animals treated with 3-NP, without a significant change in protein levels. Regarding striatal plasticity, long-term depression (LTD) was induced in both control and 3-NP cells when recorded in voltage clamp mode. The Cdk5 inhibitor roscovitine-reduced LTD in most cells. A minority subset of cells exhibited long-term potentiation (LTP) generation in the presence of roscovitine. The inhibitor of D1 receptors SCH23390 prevented LTP in three of nine cells, implying that MSN cells lacking D1/PKA activation were capable of LTP induction when Cdk5 was also blocked. Nevertheless, the co-administration of H89, a PKA inhibitor, along with roscovitine, prevented the generation of any type of plasticity in all recorded cells. These findings show the impact of 3-NP treatment on striatal plasticity and suggest that Cdk5 during early neurodegeneration may attenuate signaling pathways that lead neurons to increase their activity.
Collapse
Affiliation(s)
- Elizabeth Hernández-Echeagaray
- Laboratorio de Neurofisiología del Desarrollo y la Neurodegeneración, Unidad de Biomedicina, FES-I, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | | | - Elizabeth Nieto-Mendoza
- Laboratorio de Neurofisiología del Desarrollo y la Neurodegeneración, Unidad de Biomedicina, FES-I, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Francisco Miguel Torres-Cruz
- Laboratorio de Neurofisiología del Desarrollo y la Neurodegeneración, Unidad de Biomedicina, FES-I, Universidad Nacional Autónoma de México, Mexico City, Mexico
| |
Collapse
|
5
|
Ai H, Li M, Fang W, Wang X, Liu X, Wu L, Zhang B, Lu W. Disruption of Cdk5-GluN2B complex by a small interfering peptide attenuates social isolation-induced escalated intermale attack behavior and hippocampal oxidative stress in mice. Free Radic Biol Med 2024; 210:54-64. [PMID: 37979890 DOI: 10.1016/j.freeradbiomed.2023.11.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2023] [Revised: 09/04/2023] [Accepted: 11/11/2023] [Indexed: 11/20/2023]
Abstract
Social isolation has emerged as a significant issue during the COVID-19 pandemic that can adversely impact human mental health and potentially lead to pathological aggression. Given the lack of effective therapeutic interventions for aggressive behavior, alternative approaches are necessary. In this study, we utilized a genetic method combined with a pharmacological approach to identify and demonstrate the crucial role of Cdk5 in escalated intermale attack behavior induced by 2-week social isolation. Moreover, we developed a small peptide that effectively disrupts the interaction between Cdk5 and GluN2B, given the known involvement of this complex in various neuropsychiatric disorders. Administration of the peptide, either systemically or via intrahippocampal injection, significantly reduced oxidative stress in the hippocampus and attenuated intermale attack behavior induced by 2-week social isolation. These findings highlight the previously unknown role of the hippocampal Cdk5-GluN2B complex in social isolation-induced aggressive behavior in mice and propose the peptide as a promising therapeutic strategy for regulating attack behavior and oxidative stress.
Collapse
Affiliation(s)
- Heng Ai
- School of Basic Medical Sciences and Forensic Medicine, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Minghao Li
- School of Basic Medical Sciences and Forensic Medicine, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Weiqing Fang
- Department of Pharmacy, Women's Hospital, School of Medicine, Zhejiang University, Zhejiang, China
| | - Xuemeng Wang
- Department of the First Clinical Medicine, Hainan Medical University, Haikou, China; Key Laboratory of Molecular Biology, School of Basic Medicine and Life Sciences, Hainan Medical University, Haikou, Hainan, China
| | - Xinxin Liu
- Department of the First Clinical Medicine, Hainan Medical University, Haikou, China; Key Laboratory of Molecular Biology, School of Basic Medicine and Life Sciences, Hainan Medical University, Haikou, Hainan, China
| | - Lihui Wu
- School of Basic Medical Sciences and Forensic Medicine, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Bin Zhang
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, China.
| | - Wen Lu
- Department of Biochemistry and Molecular Biology, School of Basic Medicine and Life Sciences, Hainan Medical University, Haikou, Hainan, China; Key Laboratory of Molecular Biology, School of Basic Medicine and Life Sciences, Hainan Medical University, Haikou, Hainan, China.
| |
Collapse
|
6
|
Saidy B, Vasan R, Durant R, Greener MR, Immanuel A, Green AR, Rakha E, Ellis I, Ball G, Martin SG, Storr SJ. Unravelling transcriptomic complexity in breast cancer through modulation of DARPP-32 expression and signalling pathways. Sci Rep 2023; 13:21163. [PMID: 38036593 PMCID: PMC10689788 DOI: 10.1038/s41598-023-48198-y] [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: 06/06/2023] [Accepted: 11/23/2023] [Indexed: 12/02/2023] Open
Abstract
DARPP-32 is a key regulator of protein-phosphatase-1 (PP-1) and protein kinase A (PKA), with its function dependent upon its phosphorylation state. We previously identified DKK1 and GRB7 as genes with linked expression using Artificial Neural Network (ANN) analysis; here, we determine protein expression in a large cohort of early-stage breast cancer patients. Low levels of DARPP-32 Threonine-34 phosphorylation and DKK1 expression were significantly associated with poor patient prognosis, while low levels of GRB7 expression were linked to better survival outcomes. To gain insight into mechanisms underlying these associations, we analysed the transcriptome of T47D breast cancer cells following DARPP-32 knockdown. We identified 202 differentially expressed transcripts and observed that some overlapped with genes implicated in the ANN analysis, including PTK7, TRAF5, and KLK6, amongst others. Furthermore, we found that treatment of DARPP-32 knockdown cells with 17β-estradiol or PKA inhibitor fragment (6-22) amide led to the differential expression of 193 and 181 transcripts respectively. These results underscore the importance of DARPP-32, a central molecular switch, and its downstream targets, DKK1 and GRB7 in breast cancer. The discovery of common genes identified by a combined patient/cell line transcriptomic approach provides insights into the molecular mechanisms underlying differential breast cancer prognosis and highlights potential targets for therapeutic intervention.
Collapse
Affiliation(s)
- Behnaz Saidy
- Nottingham Breast Cancer Research Centre, Biodiscovery Institute, School of Medicine, University of Nottingham, University Park, Nottingham, NG7 2RD, UK
| | - Richa Vasan
- Nottingham Breast Cancer Research Centre, Biodiscovery Institute, School of Medicine, University of Nottingham, University Park, Nottingham, NG7 2RD, UK
| | - Rosie Durant
- Nottingham Breast Cancer Research Centre, Biodiscovery Institute, School of Medicine, University of Nottingham, University Park, Nottingham, NG7 2RD, UK
| | - Megan-Rose Greener
- Nottingham Breast Cancer Research Centre, Biodiscovery Institute, School of Medicine, University of Nottingham, University Park, Nottingham, NG7 2RD, UK
| | - Adelynn Immanuel
- Nottingham Breast Cancer Research Centre, Biodiscovery Institute, School of Medicine, University of Nottingham, University Park, Nottingham, NG7 2RD, UK
| | - Andrew R Green
- Nottingham Breast Cancer Research Centre, Biodiscovery Institute, School of Medicine, University of Nottingham, University Park, Nottingham, NG7 2RD, UK
| | - Emad Rakha
- Nottingham Breast Cancer Research Centre, Biodiscovery Institute, School of Medicine, University of Nottingham, University Park, Nottingham, NG7 2RD, UK
| | - Ian Ellis
- Nottingham Breast Cancer Research Centre, Biodiscovery Institute, School of Medicine, University of Nottingham, University Park, Nottingham, NG7 2RD, UK
| | - Graham Ball
- Medical Technology Research Centre, Anglia Ruskin University, Bishop Hall Lane, Chelmsford, CM1 1SQ, UK
| | - Stewart G Martin
- Nottingham Breast Cancer Research Centre, Biodiscovery Institute, School of Medicine, University of Nottingham, University Park, Nottingham, NG7 2RD, UK
| | - Sarah J Storr
- Nottingham Breast Cancer Research Centre, Biodiscovery Institute, School of Medicine, University of Nottingham, University Park, Nottingham, NG7 2RD, UK.
| |
Collapse
|
7
|
He K, Xie CZ, Li Y, Chen ZZ, Xu SH, Huang SQ, Yang JG, Wei ZQ, Peng XD. Dopamine and cyclic adenosine monophosphate-regulated phosphoprotein with an apparent Mr of 32000 promotes colorectal cancer growth. World J Gastrointest Oncol 2023; 15:1936-1950. [DOI: 10.4251/wjgo.v15.i11.1936] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 06/29/2023] [Accepted: 07/29/2023] [Indexed: 11/15/2023] Open
Abstract
BACKGROUND Dopamine and cyclic adenosine monophosphate (cAMP)-regulated phosphoprotein with an apparent Mr of 32000 (DARPP-32) is a protein that is involved in regulating dopamine and cAMP signaling pathways in the brain. However, recent studies have shown that DARPP-32 is also expressed in other tissues, including colorectal cancer (CRC), where its function is not well understood.
AIM To explore the effect of DARPP-32 on CRC progression.
METHODS The expression levels of DARPP-32 were assessed in CRC tissues using both quantitative polymerase chain reaction and immunohistochemistry assays. The proliferative capacity of CRC cell lines was evaluated with Cell Counting Kit-8 and 5-ethynyl-2’-deoxyuridine assays, while apoptosis was measured by flow cytometry. The migratory and invasive potential of CRC cell lines were determined using wound healing and transwell chamber assays. In vivo studies involved monitoring the growth rate of xenograft tumors. Finally, the underlying molecular mechanism of DARPP-32 was investigated through RNA-sequencing and western blot analyses.
RESULTS DARPP-32 was frequently upregulated in CRC and associated with abnormal clinicopathological features in CRC. Overexpression of DARPP-32 was shown to promote cancer cell proliferation, migration, and invasion and reduce apoptosis. DARPP-32 knockdown resulted in the opposite functional effects. Mechanistically, DARPP-32 may regulate the phosphoinositide 3-kinase (PI3K)/AKT signaling pathway in order to carry out its biological function.
CONCLUSION DARPP-32 promotes CRC progression via the PI3K/AKT signaling pathway.
Collapse
Affiliation(s)
- Kuan He
- Gastrointestinal Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400000, China
| | - Chao-Zheng Xie
- Gastrointestinal Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400000, China
| | - Ya Li
- Gastrointestinal Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400000, China
| | - Zhen-Zhou Chen
- Gastrointestinal Surgery, The Third Affiliated Hospital of Chongqing Medical University, Chongqing 400000, China
| | - Shi-Hao Xu
- Gastrointestinal Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400000, China
| | - Si-Qi Huang
- Gastrointestinal Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400000, China
| | - Jian-Guo Yang
- Gastrointestinal Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400000, China
| | - Zheng-Qiang Wei
- Gastrointestinal Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400000, China
| | - Xu-Dong Peng
- Gastrointestinal Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400000, China
| |
Collapse
|
8
|
Jeong J, Han W, Hong E, Pandey S, Li Y, Lu W, Roche KW. Regulation of NLGN3 and the Synaptic Rho-GEF Signaling Pathway by CDK5. J Neurosci 2023; 43:7264-7275. [PMID: 37699715 PMCID: PMC10621767 DOI: 10.1523/jneurosci.2309-22.2023] [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: 12/19/2022] [Revised: 08/16/2023] [Accepted: 08/18/2023] [Indexed: 09/14/2023] Open
Abstract
Neuroligins (NLGNs) are postsynaptic cell adhesion molecules that are involved in synapse assembly and function. The NLGN gene family consists of 5 genes (NLGN1-3, 4X, and 4Y). NLGN3 forms heterodimers with other NLGNs and is expressed at both excitatory and inhibitory synapses, although the distinct role at different synapses is not fully understood. Cyclin-dependent kinase 5 (Cdk5) is a proline-directed serine/threonine kinase that targets various neuronal substrates to impact neuronal migration, neurite outgrowth, synaptic transmission, and plasticity. Both NLGNs and their presynaptic binding partners neurexins are highly associated with neurodevelopmental disorders. The NLGN3 gene is on the X chromosome and variants in NLGN3 have been linked to the pathophysiology in neurodevelopmental disorders. To better understand the endogenous modulation of NLGN3, we generated an HA-tagged knock-in mouse. We found that Cdk5 associates with NLGN3 in vivo and phosphorylates NLGN3 on serine 725 (S725) in the knock-in mouse of either sex. The phosphorylation affects the NLGN3 association with Kalirin-7, a postsynaptic guanine nucleotide exchange factors for Rho GTPase family proteins. We further observed that the phosphorylation modulates NLGN3 surface expression and NLGN3-mediated synaptic currents in cultured rat neurons. Thus, we characterized NLGN3 as a novel Cdk5 substrate and revealed the functional consequences of NLGN3 S725 phosphorylation in neurons. Our study provides a novel molecular mechanism underlying Cdk5-mediated regulation of postsynaptic cell adhesion molecules.SIGNIFICANCE STATEMENT NLGN3 is involved in synapse assembly and function at both excitatory and inhibitory synapses and has been associated with the pathophysiology of neurodevelopmental disorders. Cdk5 has brain-specific activity and is involved in neuronal transmission, synapse function, and plasticity. Here, we characterize NLGN3 as a Cdk5 substrate for the first time and show that Cdk5-mediated phosphorylation regulates NLGN3 function. We demonstrate that NLGN3 S725 is a Cdk5 phosphorylation site, and reveal that the site is important for NLGN3 association with Kalirin-7, NLGN3 surface expression, and NLGN3-mediated synaptic transmission.
Collapse
Affiliation(s)
- Jaehoon Jeong
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland 20892
| | - Wenyan Han
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland 20892
| | - Eunhye Hong
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland 20892
| | - Saurabh Pandey
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland 20892
| | - Yan Li
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland 20892
| | - Wei Lu
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland 20892
| | - Katherine W Roche
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland 20892
| |
Collapse
|
9
|
Roy D, Balasubramanian S, Krishnamurthy PT, Sola P, Rymbai E. Phosphodiesterase-4 Inhibition in Parkinson's Disease: Molecular Insights and Therapeutic Potential. Cell Mol Neurobiol 2023; 43:2713-2741. [PMID: 37074485 PMCID: PMC11410141 DOI: 10.1007/s10571-023-01349-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 04/09/2023] [Indexed: 04/20/2023]
Abstract
Clinicians and researchers are exploring safer and novel treatment strategies for treating the ever-prevalent Parkinson's disease (PD) across the globe. Several therapeutic strategies are used clinically for PD, including dopamine replacement therapy, DA agonists, MAO-B blockers, COMT blockers, and anticholinergics. Surgical interventions such as pallidotomy, particularly deep brain stimulation (DBS), are also employed. However, they only provide temporal and symptomatic relief. Cyclic adenosine monophosphate (cAMP) is one of the secondary messengers involved in dopaminergic neurotransmission. Phosphodiesterase (PDE) regulates cAMP and cGMP intracellular levels. PDE enzymes are subdivided into families and subtypes which are expressed throughout the human body. PDE4 isoenzyme- PDE4B subtype is overexpressed in the substantia nigra of the brain. Various studies have implicated multiple cAMP-mediated signaling cascades in PD, and PDE4 is a common link that can emerge as a neuroprotective and/or disease-modifying target. Furthermore, a mechanistic understanding of the PDE4 subtypes has provided perceptivity into the molecular mechanisms underlying the adverse effects of phosphodiesterase-4 inhibitors (PDE4Is). The repositioning and development of efficacious PDE4Is for PD have gained much attention. This review critically assesses the existing literature on PDE4 and its expression. Specifically, this review provides insights into the interrelated neurological cAMP-mediated signaling cascades involving PDE4s and the potential role of PDE4Is in PD. In addition, we discuss existing challenges and possible strategies for overcoming them.
Collapse
Affiliation(s)
- Dhritiman Roy
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, The Nilgiris, Ooty, 643001, Tamil Nadu, India
| | - Shivaramakrishnan Balasubramanian
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, The Nilgiris, Ooty, 643001, Tamil Nadu, India.
| | - Praveen Thaggikuppe Krishnamurthy
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, The Nilgiris, Ooty, 643001, Tamil Nadu, India
| | - Piyong Sola
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, The Nilgiris, Ooty, 643001, Tamil Nadu, India
| | - Emdormi Rymbai
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, The Nilgiris, Ooty, 643001, Tamil Nadu, India
| |
Collapse
|
10
|
Fjodorova M, Noakes Z, De La Fuente DC, Errington AC, Li M. Dysfunction of cAMP-Protein Kinase A-Calcium Signaling Axis in Striatal Medium Spiny Neurons: A Role in Schizophrenia and Huntington's Disease Neuropathology. BIOLOGICAL PSYCHIATRY GLOBAL OPEN SCIENCE 2023; 3:418-429. [PMID: 37519464 PMCID: PMC10382711 DOI: 10.1016/j.bpsgos.2022.03.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 03/15/2022] [Accepted: 03/16/2022] [Indexed: 12/12/2022] Open
Abstract
Background Striatal medium spiny neurons (MSNs) are preferentially lost in Huntington's disease. Genomic studies also implicate a direct role for MSNs in schizophrenia, a psychiatric disorder known to involve cortical neuron dysfunction. It remains unknown whether the two diseases share similar MSN pathogenesis or if neuronal deficits can be attributed to cell type-dependent biological pathways. Transcription factor BCL11B, which is expressed by all MSNs and deep layer cortical neurons, was recently proposed to drive selective neurodegeneration in Huntington's disease and identified as a candidate risk gene in schizophrenia. Methods Using human stem cell-derived neurons lacking BCL11B as a model, we investigated cellular pathology in MSNs and cortical neurons in the context of these disorders. Integrative analyses between differentially expressed transcripts and published genome-wide association study datasets identified cell type-specific disease-related phenotypes. Results We uncover a role for BCL11B in calcium homeostasis in both neuronal types, while deficits in mitochondrial function and PKA (protein kinase A)-dependent calcium transients are detected only in MSNs. Moreover, BCL11B-deficient MSNs display abnormal responses to glutamate and fail to integrate dopaminergic and glutamatergic stimulation, a key feature of striatal neurons in vivo. Gene enrichment analysis reveals overrepresentation of disorder risk genes among BCL11B-regulated pathways, primarily relating to cAMP-PKA-calcium signaling axis and synaptic signaling. Conclusions Our study indicates that Huntington's disease and schizophrenia are likely to share neuronal pathophysiology where dysregulation of intracellular calcium homeostasis is found in both striatal and cortical neurons. In contrast, reduction in PKA signaling and abnormal dopamine/glutamate receptor signaling is largely specific to MSNs.
Collapse
Affiliation(s)
- Marija Fjodorova
- Neuroscience and Mental Health Research Institute, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Zoe Noakes
- Neuroscience and Mental Health Research Institute, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Daniel C. De La Fuente
- Neuroscience and Mental Health Research Institute, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Adam C. Errington
- Neuroscience and Mental Health Research Institute, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Meng Li
- Neuroscience and Mental Health Research Institute, School of Medicine, Cardiff University, Cardiff, United Kingdom
- Division of Neuroscience, School of Bioscience, Cardiff University, Cardiff, United Kingdom
| |
Collapse
|
11
|
Kim JE, Lee DS, Kim TH, Park H, Kim MJ, Kang TC. PLPP/CIN inhibits dopamine D1 receptor-mediated seizure activity via DARPP-32 serine 97 dephosphorylation in the mouse hippocampus. Neuropharmacology 2023; 228:109462. [PMID: 36792029 DOI: 10.1016/j.neuropharm.2023.109462] [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: 11/22/2022] [Revised: 01/31/2023] [Accepted: 02/08/2023] [Indexed: 02/16/2023]
Abstract
Dopamine plays a central role in the regulation of psychomotor functions in the brain. Furthermore, the dopaminergic system is involved in the ictogenesis in human patients and animal models of epilepsy. Dopamine and cAMP-regulated phosphoprotein, 32 kDa (DARPP-32) plays an important role in the regulation of interactions between dopamine and glutamate receptors in neurons. Indeed, SKF 83822 (a specific D1 receptor agonist) facilitates DARPP-32-mediated protein phosphatase 1 (PP1) inhibition leading to the increase in phosphorylation of α-amino-3-hydroxy-5-methyl-4-isoxazolepropionate receptor (AMPAR), which potentiates channel activities and currents and thereby generates seizure activity. In the present study, we found that pyridoxal-5'-phosphate phosphatase/chronophin (PLPP/CIN), a selective phosphatase for serine (S) residues, attenuated seizure susceptibility in response to SKF 83822 by dephosphorylating DARPP-32 S97 site. Similarly, inhibition of DARPP-32 S97 phosphorylation by 2-[4,5,6,7-Tetrabromo-2-(dimethylamino)-1H-benzo[d]imidazole-1-yl]acetic acid (TMCB; a selective casein kinase 2 inhibitor) attenuated SKF 83822-induced seizure activity. These inhibitory effects of PLPP/CIN and TMCB were relevant to the regulations of DARPP-32-PP1-AMPAR signaling pathway. Therefore, our findings suggest that PLPP/CIN may be a modulator in dopaminergic neurotransmission as well as glutamatergic systems, and that the PLPP/CIN-mediated DARPP-32 regulation may be one of the potential therapeutic targets for medication of seizure or epilepsy induced by D1 receptor hyperactivation.
Collapse
Affiliation(s)
- Ji-Eun Kim
- Department of Anatomy and Neurobiology, Institute of Epilepsy Research, College of Medicine, Hallym University, Chuncheon, 24252, South Korea
| | - Duk-Shin Lee
- Department of Anatomy and Neurobiology, Institute of Epilepsy Research, College of Medicine, Hallym University, Chuncheon, 24252, South Korea
| | - Tae-Hyun Kim
- Department of Anatomy and Neurobiology, Institute of Epilepsy Research, College of Medicine, Hallym University, Chuncheon, 24252, South Korea
| | - Hana Park
- Department of Anatomy and Neurobiology, Institute of Epilepsy Research, College of Medicine, Hallym University, Chuncheon, 24252, South Korea
| | - Min-Ju Kim
- Department of Anatomy and Neurobiology, Institute of Epilepsy Research, College of Medicine, Hallym University, Chuncheon, 24252, South Korea
| | - Tae-Cheon Kang
- Department of Anatomy and Neurobiology, Institute of Epilepsy Research, College of Medicine, Hallym University, Chuncheon, 24252, South Korea.
| |
Collapse
|
12
|
Inouye MO, Colameo D, Ammann I, Winterer J, Schratt G. miR-329- and miR-495-mediated Prr7 down-regulation is required for homeostatic synaptic depression in rat hippocampal neurons. Life Sci Alliance 2022; 5:5/12/e202201520. [PMID: 36150742 PMCID: PMC9510147 DOI: 10.26508/lsa.202201520] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 09/13/2022] [Accepted: 09/13/2022] [Indexed: 12/03/2022] Open
Abstract
In rat hippocampal neurons, miRNA-dependent regulation of the synaptic Prr7 protein is required for the homeostatic synaptic depression of excitatory synapses upstream of the CDK5-SPAR pathway. Homeostatic synaptic depression (HSD) in excitatory neurons is a cell-autonomous mechanism which protects excitatory neurons from over-excitation as a consequence of chronic increases in network activity. In this process, excitatory synapses are weakened and eventually eliminated, as evidenced by a reduction in synaptic AMPA receptor expression and dendritic spine loss. Originally considered a global, cell-wide mechanism, local forms of regulation, such as the local control of mRNA translation in dendrites, are being increasingly recognized in HSD. Yet, identification of excitatory proteins whose local regulation is required for HSD is still limited. Here, we show that proline-rich protein 7/transmembrane adapter protein 3 (Prr7) down-regulation in dendrites of rat hippocampal neurons is necessary for HSD induced by chronic increase in network activity resulting from a blockade of inhibitory synaptic transmission by picrotoxin (PTX). We further identify two activity-regulated miRNAs, miR-329-3p and miR-495-3p, which inhibit Prr7 mRNA translation and are required for HSD. Moreover, we found that Prr7 knockdown reduces expression of the synaptic scaffolding protein SPAR, which is rescued by pharmacological inhibition of CDK5, indicating a role of Prr7 protein in the maintenance of excitatory synapses via protection of SPAR from degradation. Together, our findings highlight a novel HSD mechanism in which chronic activity leads to miR-329– and miR-495–mediated Prr7 reduction upstream of the CDK5-SPAR pathway.
Collapse
Affiliation(s)
- Michiko O Inouye
- Department of Health Science and Technology, Laboratory of Systems Neuroscience, Institute for Neuroscience, Swiss Federal Institute of Technology ETH, Zürich, Switzerland
| | - David Colameo
- Department of Health Science and Technology, Laboratory of Systems Neuroscience, Institute for Neuroscience, Swiss Federal Institute of Technology ETH, Zürich, Switzerland
| | - Irina Ammann
- Department of Health Science and Technology, Laboratory of Systems Neuroscience, Institute for Neuroscience, Swiss Federal Institute of Technology ETH, Zürich, Switzerland
| | - Jochen Winterer
- Department of Health Science and Technology, Laboratory of Systems Neuroscience, Institute for Neuroscience, Swiss Federal Institute of Technology ETH, Zürich, Switzerland
| | - Gerhard Schratt
- Department of Health Science and Technology, Laboratory of Systems Neuroscience, Institute for Neuroscience, Swiss Federal Institute of Technology ETH, Zürich, Switzerland
| |
Collapse
|
13
|
Greener MR, Storr SJ. Exploring the Role of DARPP-32 in Addiction: A Review of the Current Limitations of Addiction Treatment Pathways and the Role of DARPP-32 to Improve Them. NEUROSCI 2022; 3:494-509. [PMID: 39483434 PMCID: PMC11523713 DOI: 10.3390/neurosci3030035] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Accepted: 08/23/2022] [Indexed: 11/03/2024] Open
Abstract
We are amidst a global addiction crisis, yet stigmas surrounding addiction counterintuitively prevail. Understanding and appreciating the neurobiology of addiction is essential to dissolve this stigma and for the development of new pharmacological agents to improve upon currently narrow therapeutic options. This review highlights this and evaluates dopamine-and-cAMP-regulated phosphoprotein, Mr 32 kDa (DARPP-32) as a potential target to treat various forms of substance abuse. Despite the proven involvement of DARPP-32 in addiction pathophysiology, no robust investigations into compounds that could pharmacologically modulate it have been carried out. Agents capable of altering DARPP-32 signalling in this way could prevent or reverse drug abuse and improve upon currently substandard treatment options.
Collapse
Affiliation(s)
- Megan R. Greener
- Biodiscovery Institute Phase 3, Entrance 2, Building 43, University of Nottingham, University Park, Nottingham NG7 2RD, UK
| | | |
Collapse
|
14
|
Kim JE, Lee DS, Kim TH, Park H, Kim MJ, Kang TC. PLPP/CIN-mediated DARPP-32 serine 97 dephosphorylation delays the seizure onset in response to kainic acid in the mouse hippocampus. Neuropharmacology 2022; 219:109238. [PMID: 36055413 DOI: 10.1016/j.neuropharm.2022.109238] [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: 04/21/2022] [Revised: 08/22/2022] [Accepted: 08/24/2022] [Indexed: 11/26/2022]
Abstract
Dopamine and cAMP-regulated phosphoprotein, 32 kDa (DARPP-32)-mediated protein phosphatase 1 (PP1) inhibition leads to the increase in phosphorylation of α-amino-3-hydroxy-5-methyl-4-isoxazolepropionate receptor (AMPAR), which potentiates channel activity and current and thereby may facilitate seizure activity. In the present study, we found that pyridoxal-5'-phosphate phosphatase/chronophin (PLPP/CIN) transiently dephosphorylated DARPP-32 serine (S) 97 site in the early time window, and casein kinase 2 (CK2) subsequently phosphorylated this site in the later time points after kainic acid (KA) injection, which increased the latency of seizure onset in response to KA, but exacerbated the intensity (severity), duration and progression of seizures. TMCB (a CK2 inhibitor) delayed the seizure onset in response to KA, concomitant with the reduced DARPP-32 S97 phosphorylation. Therefore, our findings suggest that PLPP/CIN may play an important role in the latency of seizure onset via DARPP-32-PP1-AMPAR signaling pathway, and may be one of the potential therapeutic targets for medication of seizure or epilepsy.
Collapse
Affiliation(s)
- Ji-Eun Kim
- Department of Anatomy and Neurobiology, Institute of Epilepsy Research, College of Medicine, Hallym University, Chuncheon, 24252, South Korea
| | - Duk-Shin Lee
- Department of Anatomy and Neurobiology, Institute of Epilepsy Research, College of Medicine, Hallym University, Chuncheon, 24252, South Korea
| | - Tae-Hyun Kim
- Department of Anatomy and Neurobiology, Institute of Epilepsy Research, College of Medicine, Hallym University, Chuncheon, 24252, South Korea
| | - Hana Park
- Department of Anatomy and Neurobiology, Institute of Epilepsy Research, College of Medicine, Hallym University, Chuncheon, 24252, South Korea
| | - Min-Ju Kim
- Department of Anatomy and Neurobiology, Institute of Epilepsy Research, College of Medicine, Hallym University, Chuncheon, 24252, South Korea
| | - Tae-Cheon Kang
- Department of Anatomy and Neurobiology, Institute of Epilepsy Research, College of Medicine, Hallym University, Chuncheon, 24252, South Korea.
| |
Collapse
|
15
|
Gupta P, Strange K, Telange R, Guo A, Hatch H, Sobh A, Elie J, Carter AM, Totenhagen J, Tan C, Sonawane YA, Neuzil J, Natarajan A, Ovens AJ, Oakhill JS, Wiederhold T, Pacak K, Ghayee HK, Meijer L, Reddy S, Bibb JA. Genetic impairment of succinate metabolism disrupts bioenergetic sensing in adrenal neuroendocrine cancer. Cell Rep 2022; 40:111218. [PMID: 35977518 DOI: 10.1016/j.celrep.2022.111218] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 05/24/2022] [Accepted: 07/19/2022] [Indexed: 01/11/2023] Open
Abstract
Metabolic dysfunction mutations can impair energy sensing and cause cancer. Loss of function of the mitochondrial tricarboxylic acid (TCA) cycle enzyme subunit succinate dehydrogenase B (SDHB) results in various forms of cancer typified by pheochromocytoma (PC). Here we delineate a signaling cascade where the loss of SDHB induces the Warburg effect, triggers dysregulation of [Ca2+]i, and aberrantly activates calpain and protein kinase Cdk5, through conversion of its cofactor from p35 to p25. Consequently, aberrant Cdk5 initiates a phospho-signaling cascade where GSK3 inhibition inactivates energy sensing by AMP kinase through dephosphorylation of the AMP kinase γ subunit, PRKAG2. Overexpression of p25-GFP in mouse adrenal chromaffin cells also elicits this phosphorylation signaling and causes PC. A potent Cdk5 inhibitor, MRT3-007, reverses this phospho-cascade, invoking a senescence-like phenotype. This therapeutic approach halted tumor progression in vivo. Thus, we reveal an important mechanistic feature of metabolic sensing and demonstrate that its dysregulation underlies tumor progression in PC and likely other cancers.
Collapse
Affiliation(s)
- Priyanka Gupta
- Department of Surgery, University of Alabama at Birmingham Heersink School of Medicine, Birmingham, AL 35233, USA
| | - Keehn Strange
- Department of Surgery, University of Alabama at Birmingham Heersink School of Medicine, Birmingham, AL 35233, USA
| | - Rahul Telange
- Department of Hematology, St Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Ailan Guo
- Cell Signaling Technology, Danvers, MA 01923, USA
| | - Heather Hatch
- Department of Physiological Sciences, College of Veterinary Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Amin Sobh
- Department of Medicine, Division of Hematology and Oncology, University of Florida, Gainesville, FL 32608, USA
| | - Jonathan Elie
- Perha Pharmaceuticals, Hôtel de Recherche, Perharidy Peninsula, 29680 Roscoff, France
| | - Angela M Carter
- Department of Surgery, University of Alabama at Birmingham Heersink School of Medicine, Birmingham, AL 35233, USA
| | - John Totenhagen
- Department of Radiology, University of Alabama at Birmingham Heersink School of Medicine, Birmingham, AL 35233, USA
| | - Chunfeng Tan
- UT Health Science Center at Houston, Department of Neurology, University of Texas McGovern Medical School, Houston, TX 77030, USA
| | - Yogesh A Sonawane
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Jiri Neuzil
- Institute of Biotechnology, Czech Academy of Sciences, Prague-West 252 50, Czech Republic; School of Pharmacy Medical Science, Griffith University, Southport, QLD 4222, Australia
| | - Amarnath Natarajan
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Ashley J Ovens
- Metabolic Signalling Laboratory, St Vincent's Institute of Medical Research, Fitzroy, VIC, Australia; Mary MacKillop Institute for Health Research, Australian Catholic University, Melbourne, VIC, Australia
| | - Jonathan S Oakhill
- Metabolic Signalling Laboratory, St Vincent's Institute of Medical Research, Fitzroy, VIC, Australia; Mary MacKillop Institute for Health Research, Australian Catholic University, Melbourne, VIC, Australia
| | | | - Karel Pacak
- Section on Medical Neuroendocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Hans K Ghayee
- Department of Internal Medicine, Division of Endocrinology, University of Florida College of Medicine and Malcom Randall VA Medical Center, Gainesville, FL 32608, USA
| | - Laurent Meijer
- Perha Pharmaceuticals, Hôtel de Recherche, Perharidy Peninsula, 29680 Roscoff, France
| | - Sushanth Reddy
- Department of Surgery, University of Alabama at Birmingham Heersink School of Medicine, Birmingham, AL 35233, USA
| | - James A Bibb
- Department of Surgery, University of Alabama at Birmingham Heersink School of Medicine, Birmingham, AL 35233, USA; O'Neal Comprehensive Cancer Center and the Department of Neurobiology, University of Alabama at Birmingham Heersink School of Medicine, Birmingham, AL 35233, USA.
| |
Collapse
|
16
|
Thomas R, Hernandez A, Benavides DR, Li W, Tan C, Umfress A, Plattner F, Chakraborti A, Pozzo-Miller L, Taylor SS, Bibb JA. Integrated regulation of PKA by fast and slow neurotransmission in the nucleus accumbens controls plasticity and stress responses. J Biol Chem 2022; 298:102245. [PMID: 35835216 PMCID: PMC9386499 DOI: 10.1016/j.jbc.2022.102245] [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] [Received: 04/19/2022] [Revised: 06/27/2022] [Accepted: 06/30/2022] [Indexed: 11/30/2022] Open
Abstract
Cortical glutamate and midbrain dopamine neurotransmission converge to mediate striatum-dependent behaviors, while maladaptations in striatal circuitry contribute to mental disorders. However, the crosstalk between glutamate and dopamine signaling has not been entirely elucidated. Here we uncover a molecular mechanism by which glutamatergic and dopaminergic signaling integrate to regulate cAMP-dependent protein kinase (PKA) via phosphorylation of the PKA regulatory subunit, RIIβ. Using a combination of biochemical, pharmacological, neurophysiological, and behavioral approaches, we find that glutamate-dependent reduction in cyclin-dependent kinase 5 (Cdk5)-dependent RIIβ phosphorylation alters the PKA holoenzyme autoinhibitory state to increase PKA signaling in response to dopamine. Furthermore, we show that disruption of RIIβ phosphorylation by Cdk5 enhances cortico-ventral striatal synaptic plasticity. In addition, we demonstrate that acute and chronic stress in rats inversely modulate RIIβ phosphorylation and ventral striatal infusion of a small interfering peptide that selectively targets RIIβ regulation by Cdk5 improves behavioral response to stress. We propose this new signaling mechanism integrating ventral striatal glutamate and dopamine neurotransmission is important to brain function, may contribute to neuropsychiatric conditions, and serves as a possible target for the development of novel therapeutics for stress-related disorders.
Collapse
Affiliation(s)
- Rachel Thomas
- Department of Neurology, University of Pennsylvania, Philadelphia, PA 19104 USA; Department of Psychiatry, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Adan Hernandez
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Juriquilla 76230, Santiago de Querétaro, Querétaro, México; Department of Psychiatry, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - David R Benavides
- Department of Neurology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; Department of Psychiatry, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Wei Li
- Department of Neurobiology, Civitan International Research Center, The University of Alabama Birmingham Medical Center, Birmingham, AL 35233, USA
| | - Chunfeng Tan
- Department of Neurology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030; Department of Psychiatry, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Alan Umfress
- Department of Surgery, The University of Alabama Birmingham Medical Center, Birmingham, AL 35233, USA
| | - Florian Plattner
- Department of Psychiatry, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Ayanabha Chakraborti
- Department of Surgery, The University of Alabama Birmingham Medical Center, Birmingham, AL 35233, USA
| | - Lucas Pozzo-Miller
- Department of Neurobiology, Civitan International Research Center, The University of Alabama Birmingham Medical Center, Birmingham, AL 35233, USA
| | - Susan S Taylor
- Department of Chemistry and Biochemistry, University of California, San Diego, La Jolla, CA 92093, USA
| | - James A Bibb
- Department of Neurobiology, Civitan International Research Center, The University of Alabama Birmingham Medical Center, Birmingham, AL 35233, USA; Department of Surgery, The University of Alabama Birmingham Medical Center, Birmingham, AL 35233, USA; Department of Psychiatry, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| |
Collapse
|
17
|
Kim J, Jung H, Choi JY, Lee JW, Yoon M. Plasma concentration of dopamine varies depending on breed, sex, and
the genotype of DRD4 in horses. JOURNAL OF ANIMAL SCIENCE AND TECHNOLOGY 2022; 64:792-799. [PMID: 35969706 PMCID: PMC9353348 DOI: 10.5187/jast.2022.e44] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 03/29/2022] [Accepted: 05/30/2022] [Indexed: 11/20/2022]
Abstract
Dopamine (DA) is known to be a key modulator of animal behaviors. Thus, the
plasma concentration of DA might be used as a biomarker for the behavioral
characteristics of horses. The behavioral characteristics of horses vary
depending on the breed, age, and sex. Moreover, the DA receptor genotypes are
also related to horse behaviors. Thus, the aim of this study was to investigate
the DA concentration variations of horse plasma by breed, age, sex, or genotype
of its receptor. The horses were divided by breed into Thoroughbred (n = 13),
Pony (n = 9), Warmblood (n = 4), and Haflinger (n = 5). The age variable was
divided into three different groups: post-pubertal (2–5 years, n = 6),
adult (6–13 years, n = 19), and aged horses (15–24 years, n = 6).
The sex variable was divided into geldings (n = 8) and mares (n = 23).
Approximately 10 mL of blood was collected, and an ELISA kit was used to measure
the plasma concentration of DA. Polymerase chain reaction analysis was performed
to identify the genetic variation in the DA D4 receptor gene
(DRD4). SPSS statistical software was used for statistical
analysis. The DA concentrations in geldings were significantly lower than those
in mares. There was no significant difference in DA concentrations among breed
and age groups. Horses with the GG and GA genotypes had significantly higher
plasma concentrations of DA compared to horses with the AA genotype for the
G292A gene. Briefly, the plasma concentration of DA varied depending on the sex
and genotype of G292A. These factors should be considered when the concentration
of DA is used as a biomarker for the behavioral characteristics of horses. In
conclusion, the DA concentration or DRD4 genotype of horse
plasma has the potential to be used as a biomarker that can predict the
behavioral characteristics of horses.
Collapse
Affiliation(s)
- Junyoung Kim
- Department of Animal Science and
Biotechnology, Kyungpook National University, Sangju 37224,
Korea
| | - Heejun Jung
- Department of Animal Science and
Biotechnology, Kyungpook National University, Sangju 37224,
Korea
| | - Jae-Young Choi
- Subtropical Livestock Research Institute,
National Institute of Animal Science, Rural Development
Administration, Jeju 63242, Korea
| | - Jang-Won Lee
- Department of Integrative Biological
Sciences and Industry, Sejong University, Seoul 05006,
Korea
- Corresponding author: Jang-Won Lee,
Department of Integrative Biological Sciences and Industry, Sejong University,
Seoul 05006, Korea. Tel: +82-2-3408-3375, E-mail:
| | - Minjung Yoon
- Department of Animal Science and
Biotechnology, Kyungpook National University, Sangju 37224,
Korea
- Department of Horse, Companion and Wild
Animal Science, Kyungpook National University, Sangju 37224,
Korea
- Research Center for Horse Industry,
Kyungpook National University, Sangju 37224, Korea
- Corresponding author: Minjung Yoon,
Department of Animal Science and Biotechnology, Kyungpook National University,
Sangju 37224, Korea. Tel: +82-54-530-1233, E-mail:
| |
Collapse
|
18
|
Umfress A, Singh S, Ryan KJ, Chakraborti A, Plattner F, Sonawane Y, Mallareddy JR, Acosta EP, Natarajan A, Bibb JA. Systemic Administration of a Brain Permeable Cdk5 Inhibitor Alters Neurobehavior. Front Pharmacol 2022; 13:863762. [PMID: 35645825 PMCID: PMC9134315 DOI: 10.3389/fphar.2022.863762] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Accepted: 04/01/2022] [Indexed: 01/09/2023] Open
Abstract
Cyclin-dependent kinase 5 (Cdk5) is a crucial regulator of neuronal signal transduction. Cdk5 activity is implicated in various neuropsychiatric and neurodegenerative conditions such as stress, anxiety, depression, addiction, Alzheimer's disease, and Parkinson's disease. While constitutive Cdk5 knockout is perinatally lethal, conditional knockout mice display resilience to stress-induction, enhanced cognition, neuroprotection from stroke and head trauma, and ameliorated neurodegeneration. Thus, Cdk5 represents a prime target for treatment in a spectrum of neurological and neuropsychiatric conditions. While intracranial infusions or treatment of acutely dissected brain tissue with compounds that inhibit Cdk5 have allowed the study of kinase function and corroborated conditional knockout findings, potent brain-penetrant systemically deliverable Cdk5 inhibitors are extremely limited, and no Cdk5 inhibitor has been approved to treat any neuropsychiatric or degenerative diseases to date. Here, we screened aminopyrazole-based analogs as potential Cdk5 inhibitors and identified a novel analog, 25-106, as a uniquely brain-penetrant anti-Cdk5 drug. We characterize the pharmacokinetic and dynamic responses of 25-106 in mice and functionally validate the effects of Cdk5 inhibition on open field and tail-suspension behaviors. Altogether, 25-106 represents a promising preclinical Cdk5 inhibitor that can be systemically administered with significant potential as a neurological/neuropsychiatric therapeutic.
Collapse
Affiliation(s)
- Alan Umfress
- Department of Surgery, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Sarbjit Singh
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE, United States
| | - Kevin J. Ryan
- Department of Pharmacology and Toxicology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Ayanabha Chakraborti
- Department of Surgery, University of Alabama at Birmingham, Birmingham, AL, United States
| | | | - Yogesh Sonawane
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE, United States
| | - Jayapal Reddy Mallareddy
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE, United States
| | - Edward P. Acosta
- Department of Pharmacology and Toxicology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Amarnath Natarajan
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE, United States
| | - James A. Bibb
- Department of Surgery, University of Alabama at Birmingham, Birmingham, AL, United States
- Departments of Neurobiology and Neurology, University of Alabama at Birmingham, Birmingham, AL, United States
- O’Neil Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, United States
| |
Collapse
|
19
|
Sciolino N, Liu A, Breindel L, Burz DS, Sulchek T, Shekhtman A. Microfluidics delivery of DARPP-32 into HeLa cells maintains viability for in-cell NMR spectroscopy. Commun Biol 2022; 5:451. [PMID: 35551287 PMCID: PMC9098904 DOI: 10.1038/s42003-022-03412-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Accepted: 04/26/2022] [Indexed: 11/09/2022] Open
Abstract
High-resolution structural studies of proteins and protein complexes in a native eukaryotic environment present a challenge to structural biology. In-cell NMR can characterize atomic resolution structures but requires high concentrations of labeled proteins in intact cells. Most exogenous delivery techniques are limited to specific cell types or are too destructive to preserve cellular physiology. The feasibility of microfluidics transfection or volume exchange for convective transfer, VECT, as a means to deliver labeled target proteins to HeLa cells for in-cell NMR experiments is demonstrated. VECT delivery does not require optimization or impede cell viability; cells are immediately available for long-term eukaryotic in-cell NMR experiments. In-cell NMR-based drug screening using VECT was demonstrated by collecting spectra of the sensor molecule DARPP32, in response to exogenous administration of Forskolin.
Collapse
Affiliation(s)
- Nicholas Sciolino
- University at Albany, Department of Chemistry, Albany, NY, 12222, USA
| | - Anna Liu
- Georgia Tech, School of Mechanical Engineering, Atlanta, GA, 30332, USA
| | - Leonard Breindel
- University at Albany, Department of Chemistry, Albany, NY, 12222, USA
| | - David S Burz
- University at Albany, Department of Chemistry, Albany, NY, 12222, USA
| | - Todd Sulchek
- Georgia Tech, School of Mechanical Engineering, Atlanta, GA, 30332, USA
| | | |
Collapse
|
20
|
kumar Bhardwaj V, Das P, Purohit R. Identification and comparison of plant-derived scaffolds as selective CDK5 inhibitors against standard molecules: Insights from umbrella sampling simulations. J Mol Liq 2022. [DOI: 10.1016/j.molliq.2021.118015] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
21
|
Jones-Tabah J, Mohammad H, Paulus EG, Clarke PBS, Hébert TE. The Signaling and Pharmacology of the Dopamine D1 Receptor. Front Cell Neurosci 2022; 15:806618. [PMID: 35110997 PMCID: PMC8801442 DOI: 10.3389/fncel.2021.806618] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 12/23/2021] [Indexed: 12/30/2022] Open
Abstract
The dopamine D1 receptor (D1R) is a Gαs/olf-coupled GPCR that is expressed in the midbrain and forebrain, regulating motor behavior, reward, motivational states, and cognitive processes. Although the D1R was initially identified as a promising drug target almost 40 years ago, the development of clinically useful ligands has until recently been hampered by a lack of suitable candidate molecules. The emergence of new non-catechol D1R agonists, biased agonists, and allosteric modulators has renewed clinical interest in drugs targeting this receptor, specifically for the treatment of motor impairment in Parkinson's Disease, and cognitive impairment in neuropsychiatric disorders. To develop better therapeutics, advances in ligand chemistry must be matched by an expanded understanding of D1R signaling across cell populations in the brain, and in disease states. Depending on the brain region, the D1R couples primarily to either Gαs or Gαolf through which it activates a cAMP/PKA-dependent signaling cascade that can regulate neuronal excitability, stimulate gene expression, and facilitate synaptic plasticity. However, like many GPCRs, the D1R can signal through multiple downstream pathways, and specific signaling signatures may differ between cell types or be altered in disease. To guide development of improved D1R ligands, it is important to understand how signaling unfolds in specific target cells, and how this signaling affects circuit function and behavior. In this review, we provide a summary of D1R-directed signaling in various neuronal populations and describe how specific pathways have been linked to physiological and behavioral outcomes. In addition, we address the current state of D1R drug development, including the pharmacology of newly developed non-catecholamine ligands, and discuss the potential utility of D1R-agonists in Parkinson's Disease and cognitive impairment.
Collapse
|
22
|
Parsegian A, García-Fuster MJ, Hebda-Bauer E, Watson SJ, Flagel SB, Akil H. Adolescent cocaine differentially impacts psychomotor sensitization and epigenetic profiles in adult male rats with divergent affective phenotypes. Front Psychiatry 2022; 13:1024617. [PMID: 36311521 PMCID: PMC9599748 DOI: 10.3389/fpsyt.2022.1024617] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Accepted: 09/26/2022] [Indexed: 11/24/2022] Open
Abstract
Adolescent drug use reliably predicts increased addiction liability in adulthood, but not all individuals are equally impacted. To explore the biological bases of this differential reactivity to early life drug experience, we used a genetic rat model of temperament and evaluated the impact of adolescent cocaine exposure on adult psychomotor sensitization. Relative to adult bred low-responder (bLR) rats, bred high-responders (bHR) are more sensitive to the psychomotor-activating effects of cocaine and reinstate drug-seeking behavior more readily following prolonged cocaine exposure and/or abstinence. We found that a 7-day sensitizing cocaine regimen (15 mg/kg/day) during either adolescence or adulthood produced psychomotor sensitization in bHRs only, while a dual cocaine exposure prevented further sensitization, suggesting limits on neuroplasticity. By contrast, adolescent cocaine in bLRs shifted their resilient phenotype, rendering them more responsive to cocaine in adulthood following adolescent cocaine. To begin to explore the neural correlates of these behavioral phenotypes, we assessed two functionally opposite epigenetic chromatin modifications implicated in addiction liability, permissive acetylation (ac) and repressive tri-methylation (me3) on Histone 3 Lysine 9 (H3K9), in four striatal sub-regions. In bHRs, decreased H3K9me3 and increased acH3K9 in the nucleus accumbens (NAc) core associated with cocaine sensitization. In bLRs, the combination of cocaine exposure in adolescence and adulthood, which lead to an increased response to a cocaine challenge, also increased acH3K9 in the core. Thus, adolescent cocaine experience interacts with genetic background to elicit different behavioral profiles relevant to addiction in adulthood, with concurrent modifications in the epigenetic histone profiles in the NAc that associate with cocaine sensitization and with metaplasticity.
Collapse
Affiliation(s)
- Aram Parsegian
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI, United States
| | - M Julia García-Fuster
- IUNICS, University of the Balearic Islands, Palma, Spain.,Health Research Institute of the Balearic Islands (IdISBa), Palma, Spain
| | - Elaine Hebda-Bauer
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI, United States
| | - Stanley J Watson
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI, United States.,Department of Psychiatry, University of Michigan, Ann Arbor, MI, United States
| | - Shelly B Flagel
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI, United States.,Department of Psychiatry, University of Michigan, Ann Arbor, MI, United States
| | - Huda Akil
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI, United States.,Department of Psychiatry, University of Michigan, Ann Arbor, MI, United States
| |
Collapse
|
23
|
DARPP-32 promotes ERBB3-mediated resistance to molecular targeted therapy in EGFR-mutated lung adenocarcinoma. Oncogene 2022; 41:83-98. [PMID: 34675407 PMCID: PMC8529229 DOI: 10.1038/s41388-021-02028-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 09/03/2021] [Accepted: 09/17/2021] [Indexed: 12/11/2022]
Abstract
Epidermal growth factor receptor (EGFR) tyrosine kinase inhibitor (TKI)-refractory lung adenocarcinoma (LUAD) progression is a major clinical problem. New approaches to predict and prevent acquired resistance to EGFR TKIs are urgently needed. Here, we show that dopamine and cyclic AMP-regulated phosphoprotein, Mr 32000 (DARPP-32) physically recruits ERBB3 (HER3) to EGFR to mediate switching from EGFR homodimers to EGFR:ERBB3 heterodimers to bypass EGFR TKI-mediated inhibition by potentiating ERBB3-dependent activation of oncogenic signaling. In paired LUAD patient-derived specimens before and after EGFR TKI-refractory disease progression, we reveal that DARPP-32 and kinase-activated EGFR and ERBB3 proteins are overexpressed upon acquired resistance. In mice, DARPP-32 ablation sensitizes gefitinib-resistant xenografts to EGFR TKIs, while DARPP-32 overexpression increases gefitinib-refractory LUAD progression in gefitinib-sensitive lung tumors. We introduce a DARPP-32-mediated, ERBB3-dependent mechanism the LUAD cells use to evade EGFR TKI-induced cell death, potentially paving the way for the development of therapies to better combat therapy-refractory LUAD progression.
Collapse
|
24
|
Khan A, Molitor A, Mayeur S, Zhang G, Rinaldi B, Lannes B, Lhermitte B, Umair M, Arold ST, Friant S, Rastegar S, Anheim M, Bahram S, Carapito R. A Homozygous Missense Variant in PPP1R1B/DARPP-32 Is Associated With Generalized Complex Dystonia. Mov Disord 2021; 37:365-374. [PMID: 34820905 DOI: 10.1002/mds.28861] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 10/16/2021] [Accepted: 10/18/2021] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND The dystonias are a heterogeneous group of hyperkinetic disorders characterized by sustained or intermittent muscle contractions that cause abnormal movements and/or postures. Although more than 200 causal genes are known, many cases of primary dystonia have no clear genetic cause. OBJECTIVES To identify the causal gene in a consanguineous family with three siblings affected by a complex persistent generalized dystonia, generalized epilepsy, and mild intellectual disability. METHODS We performed exome sequencing in the parents and two affected siblings and characterized the expression of the identified gene by immunohistochemistry in control human and zebrafish brains. RESULTS We identified a novel missense variant (c.142G>A (NM_032192); p.Glu48Lys) in the protein phosphatase 1 regulatory inhibitor subunit 1B gene (PPP1R1B) that was homozygous in all three siblings and heterozygous in the parents. This gene is also known as dopamine and cAMP-regulated neuronal phosphoprotein 32 (DARPP-32) and has been involved in the pathophysiology of abnormal movements. The uncovered variant is absent in public databases and modifies the conserved glutamate 48 localized close to the serine 45 phosphorylation site. The PPP1R1B protein was shown to be expressed in cells and regions involved in movement control, including projection neurons of the caudate-putamen, substantia nigra neuropil, and cerebellar Purkinje cells. The latter cells were also confirmed to be positive for PPP1R1B expression in the zebrafish brain. CONCLUSIONS We report the association of a PPP1R1B/DARPP-32 variant with generalized dystonia in man. It might be relevant to include the sequencing of this new gene in the diagnosis of patients with otherwise unexplained movement disorders. © 2021 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Amjad Khan
- Laboratoire d'ImmunoRhumatologie Moléculaire, Plateforme GENOMAX, INSERM UMR_S 1109, Faculté de Médecine, Fédération Hospitalo-Universitaire OMICARE, ITI TRANSPLANTEX NG, Université de Strasbourg, Strasbourg, France.,Strasbourg Federation of Translational Medicine (FMTS), Strasbourg University, Strasbourg, France.,Faculty of Science, Department of Biological Sciences (Zoology), University of Lakki Marwat, Lakki Marwat, Khyber Pakhtunkhwa, Pakistan
| | - Anne Molitor
- Laboratoire d'ImmunoRhumatologie Moléculaire, Plateforme GENOMAX, INSERM UMR_S 1109, Faculté de Médecine, Fédération Hospitalo-Universitaire OMICARE, ITI TRANSPLANTEX NG, Université de Strasbourg, Strasbourg, France.,Strasbourg Federation of Translational Medicine (FMTS), Strasbourg University, Strasbourg, France
| | - Sylvain Mayeur
- Laboratoire d'ImmunoRhumatologie Moléculaire, Plateforme GENOMAX, INSERM UMR_S 1109, Faculté de Médecine, Fédération Hospitalo-Universitaire OMICARE, ITI TRANSPLANTEX NG, Université de Strasbourg, Strasbourg, France.,Strasbourg Federation of Translational Medicine (FMTS), Strasbourg University, Strasbourg, France.,Department of Pathology, Strasbourg University Hospitals, Strasbourg, France
| | - Gaoqun Zhang
- Institute of Biological and Chemical Systems-Biological Information Processing (IBCS-BIP), Karlsruhe Institute of Technology (KIT), Karlsruhe, Germany
| | - Bruno Rinaldi
- Laboratoire de Génétique Moléculaire, Génomique, Microbiologie, GMGM UMR7156 CNRS/Université de Strasbourg, IPCB, Strasbourg, France
| | - Béatrice Lannes
- Strasbourg Federation of Translational Medicine (FMTS), Strasbourg University, Strasbourg, France.,Department of Pathology, Strasbourg University Hospitals, Strasbourg, France
| | - Benoît Lhermitte
- Strasbourg Federation of Translational Medicine (FMTS), Strasbourg University, Strasbourg, France.,Department of Pathology, Strasbourg University Hospitals, Strasbourg, France
| | - Muhammad Umair
- Medical Genomics Research Department, King Abdullah International Research Center (KAIMRC), King Saud Bin Abdulaziz University for Health Sciences, King Abdulaziz Medical City, Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia.,Department of Life Sciences, School of Science, University of Management and Technology (UMT), Lahore, Pakistan
| | - Stefan T Arold
- King Abdullah University of Science and Technology (KAUST), Computational Bioscience Research Center (CBRC), Division of Biological and Environmental Sciences and Engineering (BESE), Thuwal, Saudi Arabia.,Centre de Biologie Structurale, CNRS, INSERM, Université de Montpellier, Montpellier, France
| | - Sylvie Friant
- Laboratoire de Génétique Moléculaire, Génomique, Microbiologie, GMGM UMR7156 CNRS/Université de Strasbourg, IPCB, Strasbourg, France
| | - Sepand Rastegar
- Institute of Biological and Chemical Systems-Biological Information Processing (IBCS-BIP), Karlsruhe Institute of Technology (KIT), Karlsruhe, Germany
| | - Mathieu Anheim
- Strasbourg Federation of Translational Medicine (FMTS), Strasbourg University, Strasbourg, France.,Department of Neurology, Strasbourg University Hospitals, Strasbourg, France.,INSERM UMR_S 964; CNRS UMR 7104, University of Strasbourg, Illkirch-Graffenstaden, France
| | - Seiamak Bahram
- Laboratoire d'ImmunoRhumatologie Moléculaire, Plateforme GENOMAX, INSERM UMR_S 1109, Faculté de Médecine, Fédération Hospitalo-Universitaire OMICARE, ITI TRANSPLANTEX NG, Université de Strasbourg, Strasbourg, France.,Strasbourg Federation of Translational Medicine (FMTS), Strasbourg University, Strasbourg, France.,Service d'Immunologie Biologique, Plateau Technique de Biologie, Pôle de Biologie, Nouvel Hôpital Civil, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - Raphael Carapito
- Laboratoire d'ImmunoRhumatologie Moléculaire, Plateforme GENOMAX, INSERM UMR_S 1109, Faculté de Médecine, Fédération Hospitalo-Universitaire OMICARE, ITI TRANSPLANTEX NG, Université de Strasbourg, Strasbourg, France.,Strasbourg Federation of Translational Medicine (FMTS), Strasbourg University, Strasbourg, France.,Service d'Immunologie Biologique, Plateau Technique de Biologie, Pôle de Biologie, Nouvel Hôpital Civil, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| |
Collapse
|
25
|
Satake SI, Konishi S. Topographical distance between presynaptic Ca 2+ channels and exocytotic Ca 2+ sensors contributes to differential facilitatory actions of roscovitine on neurotransmitter release at cerebellar glutamatergic and GABAergic synapses. Eur J Neurosci 2021; 54:7048-7062. [PMID: 34622493 DOI: 10.1111/ejn.15487] [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: 01/23/2021] [Revised: 09/27/2021] [Accepted: 09/29/2021] [Indexed: 11/29/2022]
Abstract
Calcium influx into presynaptic terminals through voltage-gated Ca2+ channels triggers univesicular or multivesicular release of neurotransmitters depending on the characteristics of the release machinery. However, the mechanisms underlying multivesicular release (MVR) and its regulation remain unclear. Previous studies showed that in rat cerebellum, the cyclin-dependent kinase inhibitor roscovitine profoundly increases excitatory postsynaptic current (EPSC) amplitudes at granule cell (GC)-Purkinje cell (PC) synapses by enhancing the MVR of glutamate. This compound can also moderately augment the amplitude and prolong the decay time of inhibitory postsynaptic currents (IPSCs) at molecular layer interneuron (MLI)-PC synapses via MVR enhancement and GABA spillover, thus allowing for persistent activation of perisynaptic GABA receptors. The enhanced MVR may depend on the driving force for Cav 2.1 channel-mediated Ca2+ influx. To determine whether the distinct spatiotemporal dynamics of presynaptic Ca2+ influence MVR, we compared the effects of slow and fast Ca2+ chelators, that is, EGTA and BAPTA, respectively, on roscovitine-induced actions at GC-PC and MLI-PC synapses. Membrane-permeable EGTA-AM decreased GC-PC EPSC and MLI-PC IPSC amplitudes to a similar extent but suppressed the roscovitine-induced enhancement of EPSCs. In contrast, BAPTA-AM attenuated the effects of roscovitine on IPSCs. These results suggest that roscovitine augmented glutamate release by activating the release machinery located distally from the Cav 2.1 channel clusters, while it enhanced GABA release in a manner less dependent on those at distal sites. Therefore, the spatial relationships among Ca2+ channels, buffers, and sensors are critical determinants of the differential facilitatory actions of roscovitine on glutamatergic and GABAergic synapses in the cerebellar cortex.
Collapse
Affiliation(s)
- Shin' Ichiro Satake
- Brain Research Support Center, National Institute for Physiological Sciences (NIPS), Okazaki, Japan.,School of Life Science, The Graduate University for Advanced Studies (SOKENDAI), Okazaki, Japan
| | - Shiro Konishi
- Department of Neurophysiology, Kagawa School of Pharmaceutical Sciences, Tokushima Bunri University, Sanuki, Japan
| |
Collapse
|
26
|
Umfress A, Speed HE, Tan C, Ramezani S, Birnbaum S, Brekken RA, Sun X, Plattner F, Powell CM, Bibb JA. Neuropathological Effects of Chemotherapeutic Drugs. ACS Chem Neurosci 2021; 12:3038-3048. [PMID: 34370453 DOI: 10.1021/acschemneuro.1c00338] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Novel treatments, screening, and detection methods have prolonged the lives of numerous cancer patients worldwide. Unfortunately, existing and many promising new chemotherapeutics can cause deleterious, off-target side effects in normal tissue and organ systems. The central and peripheral nervous systems are widely recognized as frequent off-target effectors of anticancer drugs which can produce persistent neurological and neuropsychiatric symptoms collectively termed "chemobrain". Following chemotherapy, patients report several forms of cognitive impairment occurring acutely and sometimes persisting years after treatment. There are no effective treatments for cognitive decline induced by chemotherapeutics, and the underlying molecular mechanisms are poorly characterized and understood. In this study, we find that chronic treatment with two common chemotherapeutic agents, cisplatin and gemcitabine, impairs brain region-specific metabolism, hippocampus-dependent memory formation, and stress response behavior. This corresponds to reduced hippocampal synaptic excitability, altered neuronal signal transduction, and neuroinflammation. These findings underline that a better understanding of the basic pathological consequences of chemotherapy-induced cognitive impairment is the first step toward improving cancer treatment survivorship.
Collapse
Affiliation(s)
- Alan Umfress
- Department of Surgery, University of Alabama at Birmingham, Birmingham, Alabama 35233, United States
| | | | | | | | | | | | | | | | | | - James A. Bibb
- Department of Surgery, University of Alabama at Birmingham, Birmingham, Alabama 35233, United States
| |
Collapse
|
27
|
Brattås PL, Hersbach BA, Madsen S, Petri R, Jakobsson J, Pircs K. Impact of differential and time-dependent autophagy activation on therapeutic efficacy in a model of Huntington disease. Autophagy 2021; 17:1316-1329. [PMID: 32374203 PMCID: PMC8204969 DOI: 10.1080/15548627.2020.1760014] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Revised: 04/08/2020] [Accepted: 04/12/2020] [Indexed: 12/20/2022] Open
Abstract
Activation of macroautophagy/autophagy, a key mechanism involved in the degradation and removal of aggregated proteins, can successfully reverse Huntington disease phenotypes in various model systems. How neuronal autophagy impairments need to be considered in Huntington disease progression to achieve a therapeutic effect is currently not known. In this study, we used a mouse model of HTT (huntingtin) protein aggregation to investigate how different methods and timing of autophagy activation influence the efficacy of autophagy-activating treatment in vivo. We found that overexpression of human TFEB, a master regulator of autophagy, did not decrease mutant HTT aggregation. On the other hand, Becn1 overexpression, an autophagic regulator that plays a key role in autophagosome formation, partially cleared mutant HTT aggregates and restored neuronal pathology, but only when administered early in the disease progression. When Becn1 was administered at a later stage, when prominent mutant HTT accumulation and autophagy impairments have occurred, Becn1 overexpression did not rescue the mutant HTT-associated phenotypes. Together, these results demonstrate that the targets used to activate autophagy, as well as the timing of autophagy activation, are crucial for achieving efficient therapeutic effects.Abbreviations: AAV: adeno-associated viral vectors; ACTB: actin beta; BECN1: beclin 1, autophagy related; DAPI: 4',6-diamidino-2-phenylindole; GO: gene ontology; HD: Huntington disease; HTT: huntingtin; ICQ: Li's intensity correlation quotient; IHC: immunohistochemistry; LAMP1: lysosomal-associated membrane protein 1; MAP1LC3B/LC3B: microtubule-associated protein 1 light chain 3 beta; mHTT: mutant huntingtin; PCA: principal component analysis; PPP1R1B/DARPP-32: protein phosphatase 1 regulatory inhibitor subunit 1B; SQSTM1: sequestosome 1; TFEB: transcription factor EB; WB: western blot; WT: wild-type.
Collapse
Affiliation(s)
- Per Ludvik Brattås
- Laboratory of Molecular Neurogenetics, Department of Experimental Medical Science, Wallenberg Neuroscience Center and Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Bob A. Hersbach
- Laboratory of Molecular Neurogenetics, Department of Experimental Medical Science, Wallenberg Neuroscience Center and Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Sofia Madsen
- Laboratory of Molecular Neurogenetics, Department of Experimental Medical Science, Wallenberg Neuroscience Center and Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Rebecca Petri
- Laboratory of Molecular Neurogenetics, Department of Experimental Medical Science, Wallenberg Neuroscience Center and Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Johan Jakobsson
- Laboratory of Molecular Neurogenetics, Department of Experimental Medical Science, Wallenberg Neuroscience Center and Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Karolina Pircs
- Laboratory of Molecular Neurogenetics, Department of Experimental Medical Science, Wallenberg Neuroscience Center and Lund Stem Cell Center, Lund University, Lund, Sweden
| |
Collapse
|
28
|
Chakraborti A, Graham C, Chehade S, Vashi B, Umfress A, Kurup P, Vickers B, Chen HA, Telange R, Berryhill T, Van Der Pol W, Powell M, Barnes S, Morrow C, Smith DL, Mukhtar MS, Watts S, Kennedy G, Bibb J. High Fructose Corn Syrup-Moderate Fat Diet Potentiates Anxio-Depressive Behavior and Alters Ventral Striatal Neuronal Signaling. Front Neurosci 2021; 15:669410. [PMID: 34121997 PMCID: PMC8187874 DOI: 10.3389/fnins.2021.669410] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Accepted: 04/15/2021] [Indexed: 12/12/2022] Open
Abstract
The neurobiological mechanisms that mediate psychiatric comorbidities associated with metabolic disorders such as obesity, metabolic syndrome and diabetes remain obscure. High fructose corn syrup (HFCS) is widely used in beverages and is often included in food products with moderate or high fat content that have been linked to many serious health issues including diabetes and obesity. However, the impact of such foods on the brain has not been fully characterized. Here, we evaluated the effects of long-term consumption of a HFCS-Moderate Fat diet (HFCS-MFD) on behavior, neuronal signal transduction, gut microbiota, and serum metabolomic profile in mice to better understand how its consumption and resulting obesity and metabolic alterations relate to behavioral dysfunction. Mice fed HFCS-MFD for 16 weeks displayed enhanced anxiogenesis, increased behavioral despair, and impaired social interactions. Furthermore, the HFCS-MFD induced gut microbiota dysbiosis and lowered serum levels of serotonin and its tryptophan-based precursors. Importantly, the HFCS-MFD altered neuronal signaling in the ventral striatum including reduced inhibitory phosphorylation of glycogen synthase kinase 3β (GSK3β), increased expression of ΔFosB, increased Cdk5-dependent phosphorylation of DARPP-32, and reduced PKA-dependent phosphorylation of the GluR1 subunit of the AMPA receptor. These findings suggest that HFCS-MFD-induced changes in the gut microbiota and neuroactive metabolites may contribute to maladaptive alterations in ventral striatal function that underlie neurobehavioral impairment. While future studies are essential to further evaluate the interplay between these factors in obesity and metabolic syndrome-associated behavioral comorbidities, these data underscore the important role of peripheral-CNS interactions in diet-induced behavioral and brain function. This study also highlights the clinical need to address neurobehavioral comorbidities associated with obesity and metabolic syndrome.
Collapse
Affiliation(s)
- Ayanabha Chakraborti
- Department of Surgery, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Christopher Graham
- Department of Surgery, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Sophie Chehade
- Department of Biology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Bijal Vashi
- Department of Surgery, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Alan Umfress
- Department of Surgery, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Pradeep Kurup
- Department of Surgery, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Benjamin Vickers
- Department of Surgery, University of Alabama at Birmingham, Birmingham, AL, United States
| | - H. Alexander Chen
- Department of Surgery, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Rahul Telange
- Department of Surgery, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Taylor Berryhill
- Department of Pharmacology, University of Alabama at Birmingham Medical Center, Birmingham, AL, United States
| | - William Van Der Pol
- Center for Clinical and Translational Science, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Mickie Powell
- Department of Biology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Stephen Barnes
- Department of Pharmacology, University of Alabama at Birmingham Medical Center, Birmingham, AL, United States
| | - Casey Morrow
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Daniel L. Smith
- Department of Nutrition Sciences, University of Alabama at Birmingham, Birmingham, AL, United States
| | - M. Shahid Mukhtar
- Department of Biology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Stephen Watts
- Department of Biology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Gregory Kennedy
- Department of Surgery, University of Alabama at Birmingham, Birmingham, AL, United States
| | - James Bibb
- Department of Surgery, University of Alabama at Birmingham, Birmingham, AL, United States
| |
Collapse
|
29
|
Saidy B, Kotecha S, Butler A, Rakha EA, Ellis IO, Green AR, Martin SG, Storr SJ. PP1, PKA and DARPP-32 in breast cancer: A retrospective assessment of protein and mRNA expression. J Cell Mol Med 2021; 25:5015-5024. [PMID: 33991172 PMCID: PMC8178272 DOI: 10.1111/jcmm.16447] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 02/22/2021] [Accepted: 02/24/2021] [Indexed: 12/28/2022] Open
Abstract
Cyclic AMP–dependent protein kinase A (PKA) and protein phosphatase 1 (PP1) are proteins involved in numerous essential signalling pathways that modulate physiological and pathological functions. Both PP1 and PKA can be inhibited by dopamine‐ and cAMP‐regulated phosphoprotein 32 kD (DARPP‐32). Using immunohistochemistry, PKA and PP1 expression was determined in a large primary breast tumour cohort to evaluate associations between clinical outcome and clinicopathological criteria (n > 1100). In addition, mRNA expression of PKA and PP1 subunits was assessed in the METABRIC data set (n = 1980). Low protein expression of PKA was significantly associated with adverse survival of breast cancer patients; interestingly, this relationship was stronger in ER‐positive breast cancer patients. PP1 protein expression was not associated with patient survival. PKA and PP1 subunit mRNA was also assessed; PPP1CA, PRKACG and PRKAR1B were associated with breast cancer–specific survival. In patients with high expression of DARPP‐32, low expression of PP1 was associated with adverse survival when compared to high expression in the same group. PKA expression and PP1 expression are of significant interest in cancer as they are involved in a wide array of cellular processes, and these data indicate PKA and PP1 may play an important role in patient outcome.
Collapse
Affiliation(s)
- Behnaz Saidy
- Division of Cancer and Stem Cells, Nottingham Breast Cancer Research Centre, School of Medicine, University of Nottingham Biodiscovery Institute, Nottingham, UK
| | - Shreeya Kotecha
- Division of Cancer and Stem Cells, Nottingham Breast Cancer Research Centre, School of Medicine, University of Nottingham Biodiscovery Institute, Nottingham, UK
| | - Anna Butler
- Division of Cancer and Stem Cells, Nottingham Breast Cancer Research Centre, School of Medicine, University of Nottingham Biodiscovery Institute, Nottingham, UK
| | - Emad A Rakha
- Division of Cancer and Stem Cells, Nottingham Breast Cancer Research Centre, School of Medicine, University of Nottingham Biodiscovery Institute, Nottingham, UK
| | - Ian O Ellis
- Division of Cancer and Stem Cells, Nottingham Breast Cancer Research Centre, School of Medicine, University of Nottingham Biodiscovery Institute, Nottingham, UK
| | - Andrew R Green
- Division of Cancer and Stem Cells, Nottingham Breast Cancer Research Centre, School of Medicine, University of Nottingham Biodiscovery Institute, Nottingham, UK
| | - Stewart G Martin
- Division of Cancer and Stem Cells, Nottingham Breast Cancer Research Centre, School of Medicine, University of Nottingham Biodiscovery Institute, Nottingham, UK
| | - Sarah J Storr
- Division of Cancer and Stem Cells, Nottingham Breast Cancer Research Centre, School of Medicine, University of Nottingham Biodiscovery Institute, Nottingham, UK
| |
Collapse
|
30
|
Lemonnier T, Daldello EM, Poulhe R, Le T, Miot M, Lignières L, Jessus C, Dupré A. The M-phase regulatory phosphatase PP2A-B55δ opposes protein kinase A on Arpp19 to initiate meiotic division. Nat Commun 2021; 12:1837. [PMID: 33758202 PMCID: PMC7988065 DOI: 10.1038/s41467-021-22124-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Accepted: 02/25/2021] [Indexed: 02/06/2023] Open
Abstract
Oocytes are held in meiotic prophase for prolonged periods until hormonal signals trigger meiotic divisions. Key players of M-phase entry are the opposing Cdk1 kinase and PP2A-B55δ phosphatase. In Xenopus, the protein Arpp19, phosphorylated at serine 67 by Greatwall, plays an essential role in inhibiting PP2A-B55δ, promoting Cdk1 activation. Furthermore, Arpp19 has an earlier role in maintaining the prophase arrest through a second serine (S109) phosphorylated by PKA. Prophase release, induced by progesterone, relies on Arpp19 dephosphorylation at S109, owing to an unknown phosphatase. Here, we identified this phosphatase as PP2A-B55δ. In prophase, PKA and PP2A-B55δ are simultaneously active, suggesting the presence of other important targets for both enzymes. The drop in PKA activity induced by progesterone enables PP2A-B55δ to dephosphorylate S109, unlocking the prophase block. Hence, PP2A-B55δ acts critically on Arpp19 on two distinct sites, opposing PKA and Greatwall to orchestrate the prophase release and M-phase entry.
Collapse
Affiliation(s)
- Tom Lemonnier
- Sorbonne Université, CNRS, Laboratoire de Biologie du Développement-Institut de Biologie Paris Seine, LBD-IBPS, Paris, France
| | - Enrico Maria Daldello
- Sorbonne Université, CNRS, Laboratoire de Biologie du Développement-Institut de Biologie Paris Seine, LBD-IBPS, Paris, France
| | - Robert Poulhe
- Sorbonne Université, CNRS, Laboratoire de Biologie du Développement-Institut de Biologie Paris Seine, LBD-IBPS, Paris, France
| | - Tran Le
- Sorbonne Université, CNRS, Laboratoire de Biologie du Développement-Institut de Biologie Paris Seine, LBD-IBPS, Paris, France
| | - Marika Miot
- Sorbonne Université, CNRS, Laboratoire de Biologie du Développement-Institut de Biologie Paris Seine, LBD-IBPS, Paris, France
| | | | - Catherine Jessus
- Sorbonne Université, CNRS, Laboratoire de Biologie du Développement-Institut de Biologie Paris Seine, LBD-IBPS, Paris, France
| | - Aude Dupré
- Sorbonne Université, CNRS, Laboratoire de Biologie du Développement-Institut de Biologie Paris Seine, LBD-IBPS, Paris, France.
| |
Collapse
|
31
|
Shifts in the neurobiological mechanisms motivating cocaine use with the development of an addiction-like phenotype in male rats. Psychopharmacology (Berl) 2021; 238:811-823. [PMID: 33241478 PMCID: PMC8290931 DOI: 10.1007/s00213-020-05732-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Accepted: 11/18/2020] [Indexed: 10/22/2022]
Abstract
RATIONALE The development of addiction is accompanied by a shift in the mechanisms motivating cocaine use from nucleus accumbens (NAc) dopamine D1 receptor (D1R) signaling to glutamate AMPA-kainate receptor (AMPA-R) signaling. OBJECTIVE Here, we determined whether similar shifts occur for NAc-D2R signaling and following systemic manipulation of D1R, D2R, and AMPA-R signaling. METHODS Male rats were given short-access (20 infusions/day) or extended-access to cocaine (24 h/day, 96 infusions/day, 10 days). Motivation for cocaine was assessed following 14 days of abstinence using a progressive-ratio schedule. Once responding stabilized, the effects of NAc-D2R antagonism (eticlopride; 0-10.0 μg/side) and systemic D1R (SCH-23390; 0-1.0 mg/kg), D2R (eticlopride; 0-0.1 mg/kg), and AMPA-R (CNQX; 0-1.5 mg/kg) antagonism, and NAc-dopamine-R gene expression (Drd1/2/3) were examined. RESULTS Motivation for cocaine was markedly higher in the extended- versus short-access group confirming the development of an addiction-like phenotype in the extended-access group. NAc-infused eticlopride decreased motivation for cocaine in both the short- and extended-access groups although low doses (0.1-0.3 μg) were more effective in the short-access group and high doses (3-10 μg/side) tended to be more effective in the extended-access group. Systemic administration of eticlopride (0.1 mg/kg) was more effective in the extended-access group, and systemic administration of CNQX was effective in the extended- but not short-access group. NAc-Drd2 expression was decreased in both the short- and extended-access groups. CONCLUSION These findings indicate that in contrast to NAc-D1R, D2R remain critical for motivating cocaine use with the development of an addiction-like phenotype. These findings also indicate that shifts in the mechanisms motivating cocaine use impact the response to both site-specific and systemic pharmacological treatment.
Collapse
|
32
|
Shumilov AV, Gotovtsev PM. Modeling the activity of the dopamine signaling pathway by combination of analog electrical circuit and mathematical approaches. Heliyon 2021; 7:e05879. [PMID: 33553717 PMCID: PMC7855338 DOI: 10.1016/j.heliyon.2020.e05879] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Revised: 12/22/2019] [Accepted: 12/24/2020] [Indexed: 11/24/2022] Open
Abstract
This paper demonstrates the application of the system biology principles on the example of the dopamine signaling pathway in neurons. Presented model is based on two approaches - cytomorphic electronic circuits and mathematical modeling. Transcription and phosphorylation of DARPP-32 was modeled by analog circuit, based on well-known approaches presented in [1]. It was shown that application of circuit helps to receive signal oscillations that close to described ones in real biological systems. This combination on the one hand gives possibility to simplify calculations, on another to show this signaling pathway dynamics. The expected effect of changes in the functioning of calcium channels is considered, and the mathematical model of the interaction of system components is proposed. The average frequency of calcium current oscillations due to the presence of dopamine was 30 Hz in presented model, that is consistent with the literature, where the frequency of such oscillations is up to several tens of Hz. All presented results shows good correlation with known data, which already published today.
Collapse
Affiliation(s)
- A V Shumilov
- National Research Centre "Kurchatov Institute", Biotechnology and Bioenergy Department, Russia.,Skolkovo Institute of Science and Technology, Informational Science and Technology, Russia
| | - P M Gotovtsev
- National Research Centre "Kurchatov Institute", Biotechnology and Bioenergy Department, Russia.,Moscow Institute of Physics and Technology, Dolgoprudny, Moscow Region, Russia
| |
Collapse
|
33
|
de Almeida GRL, Szczepanik JC, Selhorst I, Schmitz AE, Dos Santos B, Cunha MP, Heinrich IA, de Paula GC, De Bem AF, Leal RB, Dafre AL. Methylglyoxal-Mediated Dopamine Depletion, Working Memory Deficit, and Depression-Like Behavior Are Prevented by a Dopamine/Noradrenaline Reuptake Inhibitor. Mol Neurobiol 2021; 58:735-749. [PMID: 33011857 DOI: 10.1007/s12035-020-02146-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Accepted: 09/22/2020] [Indexed: 01/17/2023]
Abstract
Methylglyoxal (MGO) is an endogenous toxin, mainly produced as a by-product of glycolysis that has been associated to aging, Alzheimer's disease, and inflammation. Cell culture studies reported that MGO could impair the glyoxalase, thioredoxin, and glutathione systems. Thus, we investigated the effect of in vivo MGO administration on these systems, but no major changes were observed in the glyoxalase, thioredoxin, and glutathione systems, as evaluated in the prefrontal cortex and the hippocampus of mice. A previous study from our group indicated that MGO administration produced learning/memory deficits and depression-like behavior. Confirming these findings, the tail suspension test indicated that MGO treatment for 7 days leads to depression-like behavior in three different mice strains. MGO treatment for 12 days induced working memory impairment, as evaluated in the Y maze spontaneous alternation test, which was paralleled by low dopamine and serotonin levels in the cerebral cortex. Increased DARPP32 Thr75/Thr34 phosphorylation ratio was observed, suggesting a suppression of phosphatase 1 inhibition, which may be involved in behavioral responses to MGO. Co-treatment with a dopamine/noradrenaline reuptake inhibitor (bupropion, 10 mg/kg, p.o.) reversed the depression-like behavior and working memory impairment and restored the serotonin and dopamine levels in the cerebral cortex. Overall, the cerebral cortex monoaminergic system appears to be a preferential target of MGO toxicity, a new potential therapeutic target that remains to be addressed.
Collapse
Affiliation(s)
| | - Jozimar Carlos Szczepanik
- Neurosciences Post-Graduation Program, Federal University of Santa Catarina, Florianópolis, SC, 88040-900, Brazil
| | - Ingrid Selhorst
- Department of Biochemistry, Biological Sciences Center, Federal University of Santa Catarina, Florianópolis, SC, 88040-900, Brazil
| | - Ariana Ern Schmitz
- Department of Biochemistry, Biological Sciences Center, Federal University of Santa Catarina, Florianópolis, SC, 88040-900, Brazil
| | - Bárbara Dos Santos
- Department of Biochemistry, Biological Sciences Center, Federal University of Santa Catarina, Florianópolis, SC, 88040-900, Brazil
| | - Maurício Peña Cunha
- Biochemistry Post-Graduation Program, Federal University of Santa Catarina, Florianópolis, SC, 88040-900, Brazil
| | - Isabella Aparecida Heinrich
- Neurosciences Post-Graduation Program, Federal University of Santa Catarina, Florianópolis, SC, 88040-900, Brazil
| | - Gabriela Cristina de Paula
- Department of Biochemistry, Biological Sciences Center, Federal University of Santa Catarina, Florianópolis, SC, 88040-900, Brazil
| | - Andreza Fabro De Bem
- Biochemistry Post-Graduation Program, Federal University of Santa Catarina, Florianópolis, SC, 88040-900, Brazil
- Department of Physiological Science, Institute for Biological Sciences, University of Brasília, Brasília, Brazil
| | - Rodrigo Bainy Leal
- Biochemistry Post-Graduation Program, Federal University of Santa Catarina, Florianópolis, SC, 88040-900, Brazil
- Neurosciences Post-Graduation Program, Federal University of Santa Catarina, Florianópolis, SC, 88040-900, Brazil
- Department of Biochemistry, Biological Sciences Center, Federal University of Santa Catarina, Florianópolis, SC, 88040-900, Brazil
| | - Alcir Luiz Dafre
- Biochemistry Post-Graduation Program, Federal University of Santa Catarina, Florianópolis, SC, 88040-900, Brazil.
- Neurosciences Post-Graduation Program, Federal University of Santa Catarina, Florianópolis, SC, 88040-900, Brazil.
- Department of Biochemistry, Biological Sciences Center, Federal University of Santa Catarina, Florianópolis, SC, 88040-900, Brazil.
| |
Collapse
|
34
|
Amin HS, Parikh PK, Ghate MD. Medicinal chemistry strategies for the development of phosphodiesterase 10A (PDE10A) inhibitors - An update of recent progress. Eur J Med Chem 2021; 214:113155. [PMID: 33581555 DOI: 10.1016/j.ejmech.2021.113155] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 12/27/2020] [Accepted: 01/03/2021] [Indexed: 11/26/2022]
Abstract
Phosphodiesterase 10A is a member of Phosphodiesterase (PDE)-superfamily of the enzyme which is responsible for hydrolysis of cAMP and cGMP to their inactive forms 5'-AMP and 5'-GMP, respectively. PDE10A is highly expressed in the brain, particularly in the putamen and caudate nucleus. PDE10A plays an important role in the regulation of localization, duration, and amplitude of the cyclic nucleotide signalling within the subcellular domain of these regions, and thereby modulation of PDE10A enzyme can give rise to a new therapeutic approach in the treatment of schizophrenia and other neurodegenerative disorders. Limitation of the conventional therapy of schizophrenia forced the pharmaceutical industry to move their efforts to develop a novel treatment approach with reduced side effects. In the past decade, considerable developments have been made in pursuit of PDE10A centric antipsychotic agents by several pharmaceutical industries due to the distribution of PDE10A in the brain and the ability of PDE10A inhibitors to mimic the effect of D2 antagonists and D1 agonists. However, no selective PDE10A inhibitor is currently available in the market for the treatment of schizophrenia. The present compilation concisely describes the role of PDE10A inhibitors in the therapy of neurodegenerative disorders mainly in psychosis, the structure of PDE10A enzyme, key interaction of different PDE10A inhibitors with human PDE10A enzyme and recent medicinal chemistry developments in designing of safe and effective PDE10A inhibitors for the treatment of schizophrenia. The present compilation also provides useful information and future direction to bring further improvements in the discovery of PDE10A inhibitors.
Collapse
Affiliation(s)
- Harsh S Amin
- Department of Pharmaceutical Chemistry, Institute of Pharmacy, Nirma University, Ahmedabad, 382 481, Gujarat, India
| | - Palak K Parikh
- Department of Pharmaceutical Chemistry, Institute of Pharmacy, Nirma University, Ahmedabad, 382 481, Gujarat, India; Department of Pharmaceutical Chemistry, L. M. College of Pharmacy, Navrangpura, Ahmedabad, 380 009, Gujarat, India.
| | - Manjunath D Ghate
- Department of Pharmaceutical Chemistry, Institute of Pharmacy, Nirma University, Ahmedabad, 382 481, Gujarat, India
| |
Collapse
|
35
|
Abstract
DARPP-32 (dopamine- and cAMP-regulated phosphoprotein with an apparent Mr of 32,000), now also known as phosphoprotein phosphatase 1 regulatory subunit 1B (PPP1R1B), is a potent inhibitor of protein phosphatase 1 (PP1, also known as PPP1) when phosphorylated at Thr34 by cAMP-dependent protein kinase (PKA). DARPP-32 exhibits a remarkable regional distribution in brain, roughly similar to that of dopamine innervation. Its discovery was a culmination of the long-standing effort of Paul Greengard to understand the mechanisms through which neurotransmitters such as dopamine exert their effects on target neurons. DARPP-32 is particularly enriched in striatal projection neurons where it is regulated by numerous signals through which it integrates and amplifies responses to many stimuli. Molecular studies of DARPP-32 have revealed that its regulation and function are more complex than anticipated. It is phosphorylated on multiple sites by several protein kinases that modulate DARPP-32 properties. Primarily, when phosphorylated at Thr34 DARPP-32 is a potent inhibitor of PP1, whereas when phosphorylated at Thr75 by Cdk5 it inhibits PKA. Phosphorylation at serine residues by CK1 and CK2 modulates its intracellular localization and its sensitivity to kinases or phosphatases. Modeling studies provide evidence that the signaling pathways including DARPP-32 are endowed of strong robustness and bistable properties favoring switch-like responses. Thus DARPP-32 combined with a set of other distinct signaling molecules enriched in striatal projection neurons plays a key role in the characteristic properties and physiological function of these neurons.
Collapse
|
36
|
Sobczuk P, Łomiak M, Cudnoch-Jędrzejewska A. Dopamine D1 Receptor in Cancer. Cancers (Basel) 2020; 12:cancers12113232. [PMID: 33147760 PMCID: PMC7693420 DOI: 10.3390/cancers12113232] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 10/18/2020] [Accepted: 10/29/2020] [Indexed: 12/18/2022] Open
Abstract
Simple Summary Circulating hormones and their specific receptors play a significant role in the development and progression of various cancers. This review aimed to summarize current knowledge about the dopamine D1 receptor’s biological role in different cancers, including breast cancer, central nervous system tumors, lymphoproliferative disorders, and other neoplasms. Treatment with dopamine D1 receptor agonists was proven to exert a major anti-cancer effect in many preclinical models. We highlight this receptor’s potential as a target for the adjunct therapy of tumors and discuss possibilities and necessities for further research in this area. Abstract Dopamine is a biologically active compound belonging to catecholamines. It plays its roles in the human body, acting both as a circulating hormone and neurotransmitter. It acts through G-protein-coupled receptors divided into two subgroups: D1-like receptors (D1R and D5R) and D2-like receptors (D2R, D3R, D4R). Physiologically, dopamine receptors are involved in central nervous system functions: motivation or cognition, and peripheral actions such as blood pressure and immune response modulation. Increasing evidence indicates that the dopamine D1 receptor may play a significant role in developing different human neoplasms. This receptor’s value was presented in the context of regulating various signaling pathways important in tumor development, including neoplastic cell proliferation, apoptosis, autophagy, migration, invasiveness, or the enrichment of cancer stem cells population. Recent studies proved that its activation by selective or non-selective agonists is associated with significant tumor growth suppression, metastases prevention, and tumor microvasculature maturation. It may also exert a synergistic anti-cancer effect when combined with tyrosine kinase inhibitors or temozolomide. This review provides a comprehensive insight into the heterogeneity of dopamine D1 receptor molecular roles and signaling pathways in human neoplasm development and discusses possible perspectives of its therapeutic targeting as an adjunct anti-cancer strategy of treatment. We highlight the priorities for further directions in this research area.
Collapse
Affiliation(s)
- Paweł Sobczuk
- Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, 02-097 Warsaw, Poland; (M.Ł.); (A.C.-J.)
- Department of Soft Tissue/Bone Sarcoma and Melanoma, Maria Sklodowska-Curie National Research Institute of Oncology, 02-097 Warsaw, Poland
- Correspondence: ; Tel.: +48-221166113
| | - Michał Łomiak
- Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, 02-097 Warsaw, Poland; (M.Ł.); (A.C.-J.)
| | - Agnieszka Cudnoch-Jędrzejewska
- Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, 02-097 Warsaw, Poland; (M.Ł.); (A.C.-J.)
| |
Collapse
|
37
|
Bukke VN, Archana M, Villani R, Romano AD, Wawrzyniak A, Balawender K, Orkisz S, Beggiato S, Serviddio G, Cassano T. The Dual Role of Glutamatergic Neurotransmission in Alzheimer's Disease: From Pathophysiology to Pharmacotherapy. Int J Mol Sci 2020; 21:ijms21207452. [PMID: 33050345 PMCID: PMC7589203 DOI: 10.3390/ijms21207452] [Citation(s) in RCA: 90] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 09/28/2020] [Accepted: 10/05/2020] [Indexed: 12/19/2022] Open
Abstract
Alzheimer's disease (AD) is an age-related dementia and neurodegenerative disorder, characterized by Aβ and tau protein deposition impairing learning, memory and suppressing synaptic plasticity of neurons. Increasing evidence suggests that there is a link between the glucose and glutamate alterations with age that down-regulates glucose utilization reducing glutamate levels in AD patients. Deviations in brain energy metabolism reinforce the development of AD by hampering glutamate levels in the brain. Glutamate is a nonessential amino acid and the major excitatory neurotransmitter synthesized from glucose. Alterations in cerebral glucose and glutamate levels precede the deposition of Aβ plaques. In the brain, over 40% of neuronal synapses are glutamatergic and disturbances in glutamatergic function have been implicated in pathophysiology of AD. Nevertheless, targeting the glutamatergic system seems to be a promising strategy to develop novel, improved therapeutics for AD. Here, we review data supporting the involvement of the glutamatergic system in AD pathophysiology as well as the efficacy of glutamatergic agents in this neurodegenerative disorder. We also discuss exciting new prospects for the development of improved therapeutics for this devastating disorder.
Collapse
Affiliation(s)
- Vidyasagar Naik Bukke
- Department of Clinical and Experimental Medicine, University of Foggia, 71122 Foggia, Italy;
| | - Moola Archana
- Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy; (M.A.); (R.V.); (A.D.R.); (G.S.)
| | - Rosanna Villani
- Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy; (M.A.); (R.V.); (A.D.R.); (G.S.)
| | - Antonino Davide Romano
- Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy; (M.A.); (R.V.); (A.D.R.); (G.S.)
| | - Agata Wawrzyniak
- Morphological Science Department of Human Anatomy, Medical Faculty University of Rzeszów, 35-310 Rzeszów, Poland; (A.W.); (K.B.); (S.O.)
| | - Krzysztof Balawender
- Morphological Science Department of Human Anatomy, Medical Faculty University of Rzeszów, 35-310 Rzeszów, Poland; (A.W.); (K.B.); (S.O.)
| | - Stanislaw Orkisz
- Morphological Science Department of Human Anatomy, Medical Faculty University of Rzeszów, 35-310 Rzeszów, Poland; (A.W.); (K.B.); (S.O.)
| | - Sarah Beggiato
- Department of Medical, Oral and Biotechnological Sciences, University of Chieti-Pescara, 66100 Chieti, Italy;
| | - Gaetano Serviddio
- Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy; (M.A.); (R.V.); (A.D.R.); (G.S.)
| | - Tommaso Cassano
- Department of Clinical and Experimental Medicine, University of Foggia, 71122 Foggia, Italy;
- Correspondence:
| |
Collapse
|
38
|
Lee AM, Picciotto MR. Effects of nicotine on DARPP-32 and CaMKII signaling relevant to addiction. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2020; 90:89-115. [PMID: 33706940 PMCID: PMC8008986 DOI: 10.1016/bs.apha.2020.09.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/29/2023]
Abstract
Paul Greengard brought to neuroscience the idea of, and evidence for, the role of second messenger systems in neuronal signaling. The fundamental nature of his contributions is evident in the far reach of his work, relevant to various subfields and topics in neuroscience. In this review, we discuss some of Greengard's work from the perspective of nicotinic acetylcholine receptors and their relevance to nicotine addiction. Specifically, we review the roles of dopamine- and cAMP-regulated phospho-protein of 32kDa (DARPP-32) and Ca2+/calmodulin-dependent kinase II (CaMKII) in nicotine-dependent behaviors. For each protein, we discuss the historical context of their discovery and initial characterization, focusing on the extensive biochemical and immunohistochemical work conducted by Greengard and colleagues. We then briefly summarize contemporary understanding of each protein in key intracellular signaling cascades and evidence for the role of each protein with respect to systems and behaviors relevant to nicotine addiction.
Collapse
Affiliation(s)
- Angela M Lee
- Department of Psychiatry, Yale University, New Haven, CT, United States; Yale Interdepartmental Neuroscience Program, New Haven, CT, United States
| | - Marina R Picciotto
- Department of Psychiatry, Yale University, New Haven, CT, United States; Yale Interdepartmental Neuroscience Program, New Haven, CT, United States.
| |
Collapse
|
39
|
Christensen KR, Nairn AC. cAMP-regulated phosphoproteins DARPP-32, ARPP16/19, and RCS modulate striatal signal transduction through protein kinases and phosphatases. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2020; 90:39-65. [PMID: 33706938 DOI: 10.1016/bs.apha.2020.09.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Decades of research led by Paul Greengard identified protein phosphorylation as a ubiquitous and vital post-translational modification involved in many neuronal signaling pathways. In particular, his discovery that second messenger-regulated protein phosphorylation plays a central role in the propagation and transduction of signals in the nervous system has been essential in understanding the molecular mechanisms of neuronal communication. The establishment of dopamine (DA) as an essential neurotransmitter in the central nervous system, combined with observations that DA activates G-protein-coupled receptors to control the production of cyclic adenosine monophosphate (cAMP) in postsynaptic neurons, has provided fundamental insight into the regulation of neurotransmission. Notably, DA signaling in the striatum is involved in many neurological functions such as control of locomotion, reward, addiction, and learning, among others. This review focuses on the history, characterization, and function of cAMP-mediated regulation of serine/threonine protein phosphatases and their role in DA-mediated signaling in striatal neurons. Several small, heat- and acid-stable proteins, including DARPP-32, RCS, and ARPP-16/19, were discovered by the Greengard laboratory to be regulated by DA- and cAMP signaling, and found to undergo a complex but coordinated sequence of phosphorylation and dephosphorylation events. These studies have contributed significantly to the establishment of protein phosphorylation as a ubiquitous and vital process in signal propagation in neurons, paradigm shifting discoveries at the time. Understanding DA-mediated signaling in the context of signal propagation has led to numerous insights into human conditions and the development of treatments and therapies.
Collapse
Affiliation(s)
- Kyle R Christensen
- Department of Psychiatry, Yale University, School of Medicine, New Haven, CT, United States
| | - Angus C Nairn
- Department of Psychiatry, Yale University, School of Medicine, New Haven, CT, United States.
| |
Collapse
|
40
|
Boloc D, Rodríguez N, Torres T, García-Cerro S, Parellada M, Saiz-Ruiz J, Cuesta MJ, Bernardo M, Gassó P, Lafuente A, Mas S, Arnaiz JA. Identifying key transcription factors for pharmacogenetic studies of antipsychotics induced extrapyramidal symptoms. Psychopharmacology (Berl) 2020; 237:2151-2159. [PMID: 32382784 DOI: 10.1007/s00213-020-05526-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Accepted: 04/13/2020] [Indexed: 12/14/2022]
Abstract
INTRODUCTION We explore the transcription factors involved in the molecular mechanism of antipsychotic (AP)-induced acute extrapyramidalsymptoms (EPS) in order to identify new candidate genes for pharmacogenetic studies. METHODS Protein-protein interaction (PPI) networks previously created from three pharmacogenomic models (in vitro, animal, and peripheral blood inhumans) were used to, by means of several bioinformatic tools; identify key transcription factors (TFs) that regulate each network. Once the TFs wereidentified, SNPs disrupting the binding sites (TFBS) of these TFs in the genes of each network were selected for genotyping. Finally, SNP-basedassociations with EPS were analyzed in a sample of 356 psychiatric patients receiving AP. RESULTS Our analysis identified 33 TFs expressed in the striatum, and 125 SNPs disrupting TFBS in 50 genes of our initial networks. Two SNPs (rs938112,rs2987902) in two genes (LSMAP and ABL1) were significantly associated with AP induced EPS (p < 0.001). These SNPs disrupt TFBS regulated byPOU2F1. CONCLUSION Our results highlight the possible role of the disruption of TFBS by SNPs in the pharmacological response to AP.
Collapse
Affiliation(s)
- Daniel Boloc
- Department of Medicine, University of Barcelona, Barcelona, Spain
| | | | - Teresa Torres
- Dept. Clinical Foundations, Pharmacology Unit, University of Barcelona, Barcelona, Spain
| | - Susana García-Cerro
- Dept. Clinical Foundations, Pharmacology Unit, University of Barcelona, Barcelona, Spain
| | - Mara Parellada
- Child and Adolescent Psychiatry Department, Hospital General Universitario Gregorio Marañón, School of Medicine, Universidad Complutense, IiSGM, Madrid, Spain
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Carlos III Health Institute, Madrid, Spain
| | - Jeronimo Saiz-Ruiz
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Carlos III Health Institute, Madrid, Spain
- Hospital Ramon y Cajal, Universidad de Alcala, IRYCIS, Madrid, Spain
| | - Manuel J Cuesta
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Carlos III Health Institute, Madrid, Spain
- Department of Psychiatry, Complejo Hospitalario de Navarra. Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Miquel Bernardo
- Department of Medicine, University of Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Carlos III Health Institute, Madrid, Spain
- Barcelona Clínic Schizophrenia Unit, Hospital Clínic de Barcelona, Barcelona, Spain
- Spain The August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
| | - Patricia Gassó
- Dept. Clinical Foundations, Pharmacology Unit, University of Barcelona, Barcelona, Spain
- Spain The August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
| | - Amalia Lafuente
- Dept. Clinical Foundations, Pharmacology Unit, University of Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Carlos III Health Institute, Madrid, Spain
- Spain The August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
| | - Sergi Mas
- Dept. Clinical Foundations, Pharmacology Unit, University of Barcelona, Barcelona, Spain.
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Carlos III Health Institute, Madrid, Spain.
- Spain The August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain.
| | - Joan Albert Arnaiz
- Dept. Clinical Foundations, Pharmacology Unit, University of Barcelona, Barcelona, Spain.
| |
Collapse
|
41
|
Sharma PK, Wells L, Rizzo G, Elson JL, Passchier J, Rabiner EA, Gunn RN, Dexter DT, Pienaar IS. DREADD Activation of Pedunculopontine Cholinergic Neurons Reverses Motor Deficits and Restores Striatal Dopamine Signaling in Parkinsonian Rats. Neurotherapeutics 2020; 17:1120-1141. [PMID: 31965550 PMCID: PMC7609798 DOI: 10.1007/s13311-019-00830-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
The brainstem-based pedunculopontine nucleus (PPN) traditionally associates with motor function, but undergoes extensive degeneration during Parkinson's disease (PD), which correlates with axial motor deficits. PPN-deep brain stimulation (DBS) can alleviate certain symptoms, but its mechanism(s) of action remains unknown. We previously characterized rats hemi-intranigrally injected with the proteasomal inhibitor lactacystin, as an accurate preclinical model of PD. Here we used a combination of chemogenetics with positron emission tomography imaging for in vivo interrogation of discrete neural networks in this rat model of PD. Stimulation of excitatory designer receptors exclusively activated by designer drugs expressed within PPN cholinergic neurons activated residual nigrostriatal dopaminergic neurons to produce profound motor recovery, which correlated with striatal dopamine efflux as well as restored dopamine receptor 1- and dopamine receptor 2-based medium spiny neuron activity, as was ascertained with c-Fos-based immunohistochemistry and stereological cell counts. By revealing that the improved axial-related motor functions seen in PD patients receiving PPN-DBS may be due to stimulation of remaining PPN cholinergic neurons interacting with dopaminergic ones in both the substantia nigra pars compacta and the striatum, our data strongly favor the PPN cholinergic-midbrain dopaminergic connectome as mechanism for PPN-DBS's therapeutic effects. These findings have implications for refining PPN-DBS as a promising treatment modality available to PD patients.
Collapse
Affiliation(s)
- Puneet K Sharma
- Centre for Neuroinflammation and Neurodegeneration, Division of Brain Sciences, Faculty of Medicine, Imperial College London, London, W12 0NN, UK
| | - Lisa Wells
- Invicro, Hammersmith Hospital Campus, Imperial College London, London, W12 0NN, UK
| | - Gaia Rizzo
- Invicro, Hammersmith Hospital Campus, Imperial College London, London, W12 0NN, UK
| | - Joanna L Elson
- Institute of Genetic Medicine, Newcastle University, Newcastle upon Tyne, NE1 3BZ, UK
| | - Jan Passchier
- Invicro, Hammersmith Hospital Campus, Imperial College London, London, W12 0NN, UK
| | - Eugenii A Rabiner
- Invicro, Hammersmith Hospital Campus, Imperial College London, London, W12 0NN, UK
| | - Roger N Gunn
- Centre for Neuroinflammation and Neurodegeneration, Division of Brain Sciences, Faculty of Medicine, Imperial College London, London, W12 0NN, UK
- Invicro, Hammersmith Hospital Campus, Imperial College London, London, W12 0NN, UK
| | - David T Dexter
- Centre for Neuroinflammation and Neurodegeneration, Division of Brain Sciences, Faculty of Medicine, Imperial College London, London, W12 0NN, UK
| | - Ilse S Pienaar
- Centre for Neuroinflammation and Neurodegeneration, Division of Brain Sciences, Faculty of Medicine, Imperial College London, London, W12 0NN, UK.
- School of Life Sciences, University of Sussex, Falmer, BN1 9PH, UK.
| |
Collapse
|
42
|
Martin SG, Zhang S, Yang S, Saidy B, Deen S, Storr SJ. Dopamine and cAMP-regulated phosphoprotein 32kDa (DARPP-32), protein phosphatase-1 and cyclin-dependent kinase 5 expression in ovarian cancer. J Cell Mol Med 2020; 24:9165-9175. [PMID: 32588513 PMCID: PMC7417681 DOI: 10.1111/jcmm.15553] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 04/06/2020] [Accepted: 06/07/2020] [Indexed: 12/14/2022] Open
Abstract
Dopamine and cyclic‐AMP activated phosphoprotein Mr32kDa (DARPP‐32) is a central signalling protein in neurotransmission. Following DARPP‐32 phosphorylation by protein kinase A (PKA), DARPP‐32 becomes a potent protein phosphatase 1 (PP1) inhibitor. DARPP‐32 can itself inhibit PKA following DARPP‐32 phosphorylation by cyclin‐dependent kinase 5 (Cdk5). Increasing evidence indicates a role for DARPP‐32 and its associated signalling pathways in cancer; however, its role in ovarian cancer remains unclear. Using immunohistochemistry, expression of DARPP‐32, PP1 and Cdk5 was determined in a large cohort of primary tumours from ovarian cancer patients (n = 428, 445 and 434 respectively) to evaluate associations between clinical outcome and clinicopathological criteria. Low cytoplasmic and nuclear DARPP‐32 expression was associated with shorter patient overall survival and progression‐free survival (P = .001, .001, .004 and .037 respectively). Low nuclear and cytoplasmic DARPP‐32 expression remained significantly associated with overall survival in multivariate Cox regression (P = .045, hazard ratio (HR) = 0.734, 95% confidence interval (CI) = 0.542‐0.993 and P = .001, HR = 0.494, 95% CI = 0.325‐0.749, respectively). High cytoplasmic and nuclear PP1 expression was associated with shorter patient overall survival and high cytoplasmic PP1 expression with shorter progression‐free survival (P = .005, .033, and .037, respectively). High Cdk5 expression was associated with shorter progression‐free survival (P = .006). These data suggest a role for DARPP‐32 and associated signalling kinases as prognostic markers with clinical utility in ovarian cancer.
Collapse
Affiliation(s)
- Stewart G Martin
- Nottingham Breast Cancer Research Centre, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham Biodiscovery Institute, Nottingham, UK
| | - Siwei Zhang
- Nottingham Breast Cancer Research Centre, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham Biodiscovery Institute, Nottingham, UK
| | - Song Yang
- Nottingham Breast Cancer Research Centre, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham Biodiscovery Institute, Nottingham, UK
| | - Behnaz Saidy
- Nottingham Breast Cancer Research Centre, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham Biodiscovery Institute, Nottingham, UK
| | | | - Sarah J Storr
- Nottingham Breast Cancer Research Centre, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham Biodiscovery Institute, Nottingham, UK
| |
Collapse
|
43
|
Wang Y, Zhao J, Guo FL, Gao X, Xie X, Liu S, Yang X, Yang X, Zhang L, Ye Y, Fan L, Wang J. Metformin Ameliorates Synaptic Defects in a Mouse Model of AD by Inhibiting Cdk5 Activity. Front Cell Neurosci 2020; 14:170. [PMID: 32670025 PMCID: PMC7327108 DOI: 10.3389/fncel.2020.00170] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Accepted: 05/20/2020] [Indexed: 12/13/2022] Open
Abstract
Cyclin-dependent kinase 5 (Cdk5) is a serine/threonine kinase that is activated by the neuron-specific activators p35/p39 and plays important roles in neuronal development, synaptic plasticity, and cognitive behavior. However, the proteolytic cleavage of p35 to p25 leads to prolonged and aberrant Cdk5 activation and results in synaptic depression, highly mimicking the early pathology of Alzheimer’s disease (AD). Therefore, Cdk5 inhibition is a potential promising strategy for AD drug development. Here in the present study, we showed that metformin, the most widely used drug for type 2 diabetes, suppressed Cdk5 hyper-activation and Cdk5-dependent tau hyper-phosphorylation in the APP/PS1 mouse hippocampus. We also identified the underlying molecular and cellular mechanism that metformin prevented Cdk5 hyper-activation by inhibiting the calpain-dependent cleavage of p35 into p25. Moreover, chronic metformin treatment rescued the core phenotypes in APP/PS1 mice as evidenced by restored spine density, surface GluA1 trafficking, Long-term potentiation (LTP) expression, and spatial memory. Altogether our study discovered an unidentified role of metformin in suppressing Cdk5 hyper-activation and thus preventing AD pathogenesis and suggested that metformin is a potential promising AD therapeutic drug.
Collapse
Affiliation(s)
- YaLi Wang
- Key Laboratory for the Brain Research of Henan Province, Department of Physiology and Neurobiology, Xinxiang Medical University, Xinxiang, China
| | - JianHua Zhao
- Henan Key Laboratory of Neurorestoratology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Fang-Li Guo
- Department of Neurology, Anyang District Hospital of Puyang City, Anyang, China
| | - XiaHuan Gao
- Department of Pathology, People's Hospital of Tongchuan, Tongchuan, China
| | - Xine Xie
- Department of Neurology, The Second Hospital of Jinhua, Jinhua, China
| | - ShouQing Liu
- Department of Neurology, The Second Hospital of Jinhua, Jinhua, China
| | - Xin Yang
- Department of Physiology, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - XinFeng Yang
- Department of Pathophysiology, Xinxiang Medical University, Xinxiang, China
| | - LuYi Zhang
- Department of Pathophysiology, Xinxiang Medical University, Xinxiang, China
| | - YuXiao Ye
- Department of Pathophysiology, Xinxiang Medical University, Xinxiang, China
| | - LiBing Fan
- Department of Pathophysiology, Xinxiang Medical University, Xinxiang, China
| | - JianGang Wang
- Key Laboratory for the Brain Research of Henan Province, Department of Physiology and Neurobiology, Xinxiang Medical University, Xinxiang, China.,Henan Key Laboratory of Neurorestoratology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China.,Department of Pathophysiology, Xinxiang Medical University, Xinxiang, China.,Henan Key Laboratory of Biological Psychiatry, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang Medical University, Xinxiang, China
| |
Collapse
|
44
|
Alam SK, Wang L, Ren Y, Hernandez CE, Kosari F, Roden AC, Yang R, Hoeppner LH. ASCL1-regulated DARPP-32 and t-DARPP stimulate small cell lung cancer growth and neuroendocrine tumour cell proliferation. Br J Cancer 2020; 123:819-832. [PMID: 32499571 PMCID: PMC7463034 DOI: 10.1038/s41416-020-0923-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 04/21/2020] [Accepted: 05/13/2020] [Indexed: 01/09/2023] Open
Abstract
Background Small cell lung cancer (SCLC) is the most aggressive form of lung cancer, and new molecular insights are necessary for prognostic and therapeutic advances. Methods Dopamine and cAMP-regulated phosphoprotein, Mr 32000 (DARPP-32) and its N-terminally truncated splice variant, t-DARPP, were stably overexpressed or ablated in human DMS-53 and H1048 SCLC cells. Functional assays and immunoblotting were used to assess how DARPP-32 isoforms regulate SCLC cell growth, proliferation, and apoptosis. DARPP-32-modulated SCLC cells were orthotopically injected into the lungs of SCID mice to evaluate how DARPP-32 and t-DARPP regulate neuroendocrine tumour growth. Immunostaining for DARPP-32 proteins was performed in SCLC patient-derived specimens. Bioinformatics analysis and subsequent transcription assays were used to determine the mechanistic basis of DARPP-32-regulated SCLC growth. Results We demonstrate in mice that DARPP-32 and t-DARPP promote SCLC growth through increased Akt/Erk-mediated proliferation and anti-apoptotic signalling. DARPP-32 isoforms are overexpressed in SCLC patient-derived tumour tissue, but undetectable in physiologically normal lung. Achaete-scute homologue 1 (ASCL1) transcriptionally activates DARPP-32 isoforms in human SCLC cells. Conclusions We reveal new regulatory mechanisms of SCLC oncogenesis that suggest DARPP-32 isoforms may represent a negative prognostic indicator for SCLC and serve as a potential target for the development of new therapies.
Collapse
Affiliation(s)
- Sk Kayum Alam
- The Hormel Institute, University of Minnesota, Austin, MN, USA
| | - Li Wang
- The Hormel Institute, University of Minnesota, Austin, MN, USA
| | - Yanan Ren
- The Hormel Institute, University of Minnesota, Austin, MN, USA
| | | | - Farhad Kosari
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN, USA
| | - Anja C Roden
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Rendong Yang
- The Hormel Institute, University of Minnesota, Austin, MN, USA.,Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
| | - Luke H Hoeppner
- The Hormel Institute, University of Minnesota, Austin, MN, USA. .,Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA.
| |
Collapse
|
45
|
Peyressatre M, Laure A, Pellerano M, Boukhaddaoui H, Soussi I, Morris MC. Fluorescent Biosensor of CDK5 Kinase Activity in Glioblastoma Cell Extracts and Living Cells. Biotechnol J 2020; 15:e1900474. [DOI: 10.1002/biot.201900474] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Revised: 04/13/2020] [Indexed: 12/26/2022]
Affiliation(s)
- Marion Peyressatre
- Institut des Biomolécules Max Mousseron, CNRS, UMR 5247, Université de Montpellier Faculté de Pharmacie 15, Av. Charles Flahault Montpellier 34093 France
| | - Arthur Laure
- Institut des Biomolécules Max Mousseron, CNRS, UMR 5247, Université de Montpellier Faculté de Pharmacie 15, Av. Charles Flahault Montpellier 34093 France
| | - Morgan Pellerano
- Institut des Biomolécules Max Mousseron, CNRS, UMR 5247, Université de Montpellier Faculté de Pharmacie 15, Av. Charles Flahault Montpellier 34093 France
| | - Hassan Boukhaddaoui
- Plateau Imagerie Cellulaire MRI‐INM INM‐INSERM U 1051, Hôpital Saint Eloi 80 rue Augustin Fliche Montpellier 34091 France
| | - Ines Soussi
- Institut des Biomolécules Max Mousseron, CNRS, UMR 5247, Université de Montpellier Faculté de Pharmacie 15, Av. Charles Flahault Montpellier 34093 France
| | - May C. Morris
- Institut des Biomolécules Max Mousseron, CNRS, UMR 5247, Université de Montpellier Faculté de Pharmacie 15, Av. Charles Flahault Montpellier 34093 France
| |
Collapse
|
46
|
Luo W, Xu C, Phillips S, Gardenswartz A, Rosenblum JM, Ayello J, Lessnick SL, Hao HX, Cairo MS. Protein phosphatase 1 regulatory subunit 1A regulates cell cycle progression in Ewing sarcoma. Oncotarget 2020; 11:1691-1704. [PMID: 32477459 PMCID: PMC7233808 DOI: 10.18632/oncotarget.27571] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Accepted: 04/10/2020] [Indexed: 12/02/2022] Open
Abstract
Ewing sarcoma (ES) is a malignant pediatric bone and soft tissue tumor. Patients with metastatic ES have a dismal outcome which has not been improved in decades. The major challenge in the treatment of metastatic ES is the lack of specific targets and rational combinatorial therapy. We recently found that protein phosphatase 1 regulatory subunit 1A (PPP1R1A) is specifically highly expressed in ES and promotes tumor growth and metastasis in ES. In the current investigation, we show that PPP1R1A regulates ES cell cycle progression in G1/S phase by down-regulating cell cycle inhibitors p21Cip1 and p27Kip1, which leads to retinoblastoma (Rb) protein hyperphosphorylation. In addition, we show that PPP1R1A promotes normal transcription of histone genes during cell cycle progression. Importantly, we demonstrate a synergistic/additive effect of the combinatorial therapy of PPP1R1A and insulin-like growth factor 1 receptor (IGF-1R) inhibition on decreasing ES cell proliferation and migration in vitro and limiting xenograft tumor growth and metastasis in vivo. Taken together, our findings suggest a role of PPP1R1A as an ES specific cell cycle modulator and that simultaneous targeting of PPP1R1A and IGF-1R pathways is a promising specific and effective strategy to treat both primary and metastatic ES.
Collapse
Affiliation(s)
- Wen Luo
- Department of Pediatrics, New York Medical College, Valhalla, NY, USA.,Department of Pathology, New York Medical College, Valhalla, NY, USA
| | - Changxin Xu
- James J. Peters Veterans Affairs Medical Center, Bronx, NY, USA
| | - Sarah Phillips
- Department of Medicine, New York Medical College, Valhalla, NY, USA
| | | | | | - Janet Ayello
- Department of Pediatrics, New York Medical College, Valhalla, NY, USA
| | | | - Huai-Xiang Hao
- Novartis Institutes for BioMedical Research, Cambridge, MA, USA
| | - Mitchell S Cairo
- Department of Pediatrics, New York Medical College, Valhalla, NY, USA.,Department of Pathology, New York Medical College, Valhalla, NY, USA.,Department of Medicine, New York Medical College, Valhalla, NY, USA.,Department of Immunology and Microbiology, New York Medical College, Valhalla, NY, USA.,Department of Cell Biology and Anatomy, New York Medical College, Valhalla, NY, USA
| |
Collapse
|
47
|
Satake S, Konishi S. Roscovitine differentially facilitates cerebellar glutamatergic and GABAergic neurotransmission by enhancing Ca v 2.1 channel-mediated multivesicular release. Eur J Neurosci 2020; 52:3002-3021. [PMID: 32383214 DOI: 10.1111/ejn.14771] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2019] [Revised: 04/22/2020] [Accepted: 05/01/2020] [Indexed: 11/29/2022]
Abstract
Synaptic vesicle exocytosis is triggered by Ca2+ influx through several subtypes of voltage-gated calcium channels in the presynaptic terminal. We previously reported that paired-pulse stimulation at brief intervals increases Cav 2.1 (P/Q-type) channel-mediated multivesicular release (MVR) at glutamatergic synapses between granule cells (GCs) and molecular layer interneurons (MLIs) in rat cerebellar slices. However, it has yet to be determined how Cav 2 channel subtypes take part in MVR in single axon terminal. This study therefore aimed at examining the effects of roscovitine on different types of cerebellar synapses that make contacts with Purkinje cells (PCs), because this compound has been shown to enhance Cav 2.1 channel-mediated MVR at GC-MLI synapses. Bath application of roscovitine profoundly increased the amplitude of excitatory postsynaptic currents (EPSCs) at GC-PC synapses by a presynaptic mechanism as previously observed at GC-MLI synapses, whereas it caused a marginal effect on climbing fiber-mediated EPSCs in PCs. At MLI-PC synapses, roscovitine increased both the amplitude and decay time of inhibitory postsynaptic currents (IPSCs) by enhancing multivesicular GABA release. When extracellular Ca2+ concentration ([Ca2+ ]e ) decreased, roscovitine became less effective in increasing GC-PC EPSCs. By contrast, roscovitine was able to augment MLI-PC IPSCs in the low [Ca2+ ]e . The Cav 2.1 channel blocker ω-agatoxin IVA suppressed the roscovitine-induced facilitatory actions on both GC-PC EPSCs and MLI-PC IPSCs. These results demonstrate that roscovitine enhances MVR at the GC-PC excitatory synapses in a manner dependent on the driving force of Cav 2.1 channel-mediated Ca2+ influx into the nerve terminal, while it also facilitates MLI-PC inhibitory transmission via Ca2+ -insensitive mechanisms.
Collapse
Affiliation(s)
- Shin'Ichiro Satake
- Department of Fundamental Neuroscience, National Institute for Physiological Sciences (NIPS), Okazaki, Japan.,School of Life Science, The Graduate University for Advanced Studies (SOKENDAI), Okazaki, Japan
| | - Shiro Konishi
- Department of Neurophysiology, Kagawa School of Pharmaceutical Sciences, Tokushima Bunri University, Sanuki, Japan
| |
Collapse
|
48
|
Babenko VN, Galyamina AG, Rogozin IB, Smagin DA, Kudryavtseva NN. Dopamine response gene pathways in dorsal striatum MSNs from a gene expression viewpoint: cAMP-mediated gene networks. BMC Neurosci 2020; 21:12. [PMID: 32216748 PMCID: PMC7099774 DOI: 10.1186/s12868-020-00560-w] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Accepted: 03/18/2020] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Medium spiny neurons (MSNs) comprise the main body (95% in mouse) of the dorsal striatum neurons and represent dopaminoceptive GABAergic neurons. The cAMP (cyclic Adenosine MonoPhosphate)-mediated cascade of excitation and inhibition responses observed in MSN intracellular signal transduction is crucial for neuroscience research due to its involvement in the motor and behavioral functions. In particular, all types of addictions are related to MSNs. Shedding the light on the mechanics of the above-mentioned cascade is of primary importance for this research domain. RESULTS A mouse model of chronic social conflicts in daily agonistic interactions was used to analyze dorsal striatum neurons genes implicated in cAMP-mediated phosphorylation activation pathways specific for MSNs. Based on expression correlation analysis, we succeeded in dissecting Drd1- and Drd2-dopaminoceptive neurons (D1 and D2, correspondingly) gene pathways. We also found that D1 neurons genes clustering are split into two oppositely correlated states, passive and active ones, the latter apparently corresponding to D1 firing stage upon protein kinase A (PKA) activation. We observed that under defeat stress in chronic social conflicts the loser mice manifest overall depression of dopamine-mediated MSNs activity resulting in previously reported reduced motor activity, while the aggressive mice with positive fighting experience (aggressive mice) feature an increase in both D1-active phase and D2 MSNs genes expression leading to hyperactive behavior pattern corresponded by us before. Based on the alternative transcript isoforms expression analysis, it was assumed that many genes (Drd1, Adora1, Pde10, Ppp1r1b, Gnal), specifically those in D1 neurons, apparently remain transcriptionally repressed via the reversible mechanism of promoter CpG island silencing, resulting in alternative promoter usage following profound reduction in their expression rate. CONCLUSION Based on the animal stress model dorsal striatum pooled tissue RNA-Seq data restricted to cAMP related genes subset we elucidated MSNs steady states exhaustive projection for the first time. We correspond the existence of D1 active state not explicitly outlined before, and connected with dynamic dopamine neurotransmission cycles. Consequently, we were also able to indicate an oscillated postsynaptic dopamine vs glutamate action pattern in the course of the neurotransmission cycles.
Collapse
Affiliation(s)
- Vladimir N Babenko
- Institute of Cytology and Genetics SB RAS, Novosibirsk, Russia.
- Novosibirsk State University, Novosibirsk, Russia.
| | | | - Igor B Rogozin
- National Institutes of Health, Rockville Pike, Bethesda, MD, USA
| | - Dmitry A Smagin
- Institute of Cytology and Genetics SB RAS, Novosibirsk, Russia
| | | |
Collapse
|
49
|
Kang W, Jiang F, Wu YD, Wales DJ. Multifunnel Energy Landscapes for Phosphorylated Translation Repressor 4E-BP2 and Its Mutants. J Chem Theory Comput 2019; 16:800-810. [PMID: 31774674 PMCID: PMC7462351 DOI: 10.1021/acs.jctc.9b01042] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Upon phosphorylation of specific sites, eukaryotic translation initiation factor 4E (eIF4E) binding protein 2 (4E-BP2) undergoes a fundamental structural transformation from a disordered state to a four-stranded β-sheet, leading to decreased binding affinity for its partner. This change reflects the significant effects of phosphate groups on the underlying energy landscapes of proteins. In this study, we combine high-temperature molecular dynamics simulations and discrete path sampling to construct energy landscapes for a doubly phosphorylated 4E-BP218-62 and two mutants (a single site mutant D33K and a double mutant Y54A/L59A). The potential and free energy landscapes for these three systems are multifunneled with the folded state and several alternative states lying close in energy, suggesting perhaps a multifunneled and multifunctional protein. Hydrogen bonds between phosphate groups and other residues not only stabilize these low-lying conformations to different extents but also play an important role in interstate transitions. From the energy landscape perspective, our results explain some interesting experimental observations, including the low stability of doubly phosphorylated 4E-BP2 and its moderate binding to eIF4E and the inability of phosphorylated Y54A/L59A to fold.
Collapse
Affiliation(s)
- Wei Kang
- Laboratory of Computational Chemistry and Drug Design, Laboratory of Chemical Genomics , Peking University Shenzhen Graduate School , Shenzhen 518055 , China.,College of Chemistry and Molecular Engineering , Peking University , Beijing 100871 , China.,Department of Chemistry , University of Cambridge , Lensfield Road , Cambridge CB2 1EW , U.K
| | - Fan Jiang
- Laboratory of Computational Chemistry and Drug Design, Laboratory of Chemical Genomics , Peking University Shenzhen Graduate School , Shenzhen 518055 , China
| | - Yun-Dong Wu
- Laboratory of Computational Chemistry and Drug Design, Laboratory of Chemical Genomics , Peking University Shenzhen Graduate School , Shenzhen 518055 , China.,College of Chemistry and Molecular Engineering , Peking University , Beijing 100871 , China
| | - David J Wales
- Department of Chemistry , University of Cambridge , Lensfield Road , Cambridge CB2 1EW , U.K
| |
Collapse
|
50
|
Chen X, Gumina G, Virga KG. Recent Advances in Drug Repurposing for Parkinson's Disease. Curr Med Chem 2019; 26:5340-5362. [PMID: 30027839 DOI: 10.2174/0929867325666180719144850] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2017] [Revised: 04/27/2018] [Accepted: 05/02/2018] [Indexed: 12/25/2022]
Abstract
As a long-term degenerative disorder of the central nervous system that mostly affects older people, Parkinson's disease is a growing health threat to our ever-aging population. Despite remarkable advances in our understanding of this disease, all therapeutics currently available only act to improve symptoms but cannot stop the disease progression. Therefore, it is essential that more effective drug discovery methods and approaches are developed, validated, and used for the discovery of disease-modifying treatments for Parkinson's disease. Drug repurposing, also known as drug repositioning, or the process of finding new uses for existing or abandoned pharmaceuticals, has been recognized as a cost-effective and timeefficient way to develop new drugs, being equally promising as de novo drug discovery in the field of neurodegeneration and, more specifically for Parkinson's disease. The availability of several established libraries of clinical drugs and fast evolvement in disease biology, genomics and bioinformatics has stimulated the momentums of both in silico and activity-based drug repurposing. With the successful clinical introduction of several repurposed drugs for Parkinson's disease, drug repurposing has now become a robust alternative approach to the discovery and development of novel drugs for this disease. In this review, recent advances in drug repurposing for Parkinson's disease will be discussed.
Collapse
Affiliation(s)
- Xin Chen
- Department of Pharmaceutical and Administrative Sciences, Presbyterian College School of Pharmacy, Clinton, SC 29325, United States
| | - Giuseppe Gumina
- Department of Pharmaceutical and Administrative Sciences, Presbyterian College School of Pharmacy, Clinton, SC 29325, United States
| | - Kristopher G Virga
- Department of Pharmaceutical Sciences, William Carey University School of Pharmacy, Biloxi, MS 39532, United States
| |
Collapse
|