1
|
Kachemov M, Vaibhav V, Smith C, Sundararaman N, Heath M, Pendlebury DF, Matlock A, Lau A, Morozko E, Lim RG, Reidling J, Steffan JS, Van Eyk JE, Thompson LM. Dysregulation of protein SUMOylation networks in Huntington's disease R6/2 mouse striatum. Brain 2025; 148:1212-1227. [PMID: 39391934 PMCID: PMC11969464 DOI: 10.1093/brain/awae319] [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: 05/06/2024] [Revised: 08/13/2024] [Accepted: 09/21/2024] [Indexed: 10/12/2024] Open
Abstract
Huntington's disease is a neurodegenerative disorder caused by an expanded CAG repeat mutation in the Huntingtin (HTT) gene. The mutation impacts neuronal protein homeostasis and cortical/striatal circuitry. SUMOylation is a post-translational modification with broad cellular effects including via modification of synaptic proteins. Here, we used an optimized SUMO protein-enrichment and mass spectrometry method to identify the protein SUMOylation/SUMO interaction proteome in the context of Huntington's disease using R6/2 transgenic and non-transgenic mice. Significant changes in the enrichment of SUMOylated and SUMO-interacting proteins were observed, including those involved in presynaptic function, cytomatrix at the active zone, cytoskeleton organization and glutamatergic signalling. Mitochondrial and RNA-binding proteins also showed altered enrichment. Modified SUMO-associated pathways in Huntington's disease tissue include clathrin-mediated endocytosis signalling, synaptogenesis signalling, synaptic long-term potentiation and SNARE signalling. To evaluate how modulation of SUMOylation might influence functional measures of neuronal activity in Huntington's disease cells in vitro, we used primary neuronal cultures from R6/2 and non-transgenic mice. A receptor internalization assay for the metabotropic glutamate receptor 7 (mGLUR7), a SUMO-enriched protein in the mass spectrometry, showed decreased internalization in R6/2 neurons compared to non-transgenic neurons. SiRNA-mediated knockdown of the E3 SUMO ligase protein inhibitor of activated STAT1 (Pias1), which can SUMO modify mGLUR7, reduced this Huntington's disease phenotype. In addition, microelectrode array analysis of primary neuronal cultures indicated early hyperactivity in Huntington's disease cells, while later time points demonstrated deficits in several measurements of neuronal activity within cortical neurons. Huntington's disease phenotypes were rescued at selected time points following knockdown of Pias1. Collectively, our results provide a mouse brain SUMOome resource and show that significant alterations occur within the post-translational landscape of SUMO-protein interactions of synaptic proteins in Huntington's disease mice, suggesting that targeting of synaptic SUMO networks may provide a proteostatic systems-based therapeutic approach for Huntington's disease and other neurological disorders.
Collapse
Affiliation(s)
- Marketta Kachemov
- Neurobiology and Behavior, University of California Irvine, Irvine, CA 92697, USA
| | - Vineet Vaibhav
- Advanced Clinical Biosystems Research Institute, The Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Charlene Smith
- Department of Psychiatry and Human Behavior, University of California Irvine, Irvine, CA 92868, USA
| | - Niveda Sundararaman
- Advanced Clinical Biosystems Research Institute, The Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Marie Heath
- Neurobiology and Behavior, University of California Irvine, Irvine, CA 92697, USA
| | - Devon F Pendlebury
- Neurobiology and Behavior, University of California Irvine, Irvine, CA 92697, USA
| | - Andrea Matlock
- Advanced Clinical Biosystems Research Institute, The Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Alice Lau
- Sue and Bill Gross Stem Cell Center, University of California Irvine, Irvine, CA 92697, USA
| | - Eva Morozko
- Neurobiology and Behavior, University of California Irvine, Irvine, CA 92697, USA
| | - Ryan G Lim
- Institute of Memory Impairments and Neurological Disorders, University of California Irvine, Irvine, CA 92697, USA
| | - Jack Reidling
- Institute of Memory Impairments and Neurological Disorders, University of California Irvine, Irvine, CA 92697, USA
| | - Joan S Steffan
- Department of Psychiatry and Human Behavior, University of California Irvine, Irvine, CA 92868, USA
| | - Jennifer E Van Eyk
- Advanced Clinical Biosystems Research Institute, The Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Leslie M Thompson
- Neurobiology and Behavior, University of California Irvine, Irvine, CA 92697, USA
- Department of Psychiatry and Human Behavior, University of California Irvine, Irvine, CA 92868, USA
- Sue and Bill Gross Stem Cell Center, University of California Irvine, Irvine, CA 92697, USA
- Institute of Memory Impairments and Neurological Disorders, University of California Irvine, Irvine, CA 92697, USA
| |
Collapse
|
2
|
Huang Y, Huang W, Liang G, Liu D, He C, Xiao L, Gao X. PD98059 inhibit the proliferation and differentiation of osteoblasts in the formation of tympanosclerosis via ERK1/2-MAPK signaling pathway. Exp Cell Res 2025; 447:114437. [PMID: 39986597 DOI: 10.1016/j.yexcr.2025.114437] [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: 01/03/2025] [Revised: 02/07/2025] [Accepted: 02/08/2025] [Indexed: 02/24/2025]
Abstract
Tympanosclerosis (TS) has become a common pathological condition of the middle ear, but the underlying mechanism of it is still ambiguous. It was found that osteoprotegerin/receptor activator of nuclear factor kappa B ligand (OPG/RANKL) axis played an important role in the development of TS and was regulated by the extracellular signal regulated kinase 1/2-mitogen activated protein kinase (ERK1/2-MAPK) pathway. However, whether ERK1/2-MAPK pathway mediates the occurrence of TS by regulating OPG/RANKL axis has not been reported. In this study, MAPK and calcium pathway were found significantly activated in TS model. In vivo, the expression of p-ERK1/2 in TS model was significantly increased. In vitro, osteoblasts were isolated from auditory vesicles of neonatal rats for the first time, and then cultured with ERK1/2-MAPK pathway inhibitor PD98059. As results, PD98059 showed inhibitory effects on the phosphorylation of ERK1/2 and proliferation of osteoblasts. Besides, different concentrations of PD98059 showed different inhibitory effects on mRNA expression of osteocalcin (Ocn), bone sialoprotein (Bsp), runt-related transcription factor 2 (Runx2), bone morphogenetic protein 2 (Bmp2) and OPG. Therefore, it was speculated that the ERK1/2-MAPK pathway may affect the formation of TS by regulating the proliferation and differentiation of osteoblasts, which may be helpful for the study of drug target for tympanosclerosis.
Collapse
Affiliation(s)
- Yu Huang
- Department of Otolaryngology-Head and Neck Surgery, Tongren Hospital, Wuhan University, Wuhan, Hubei, China.
| | - Wenxia Huang
- Department of Otolaryngology-Head and Neck Surgery, Tongren Hospital, Wuhan University, Wuhan, Hubei, China.
| | - Gengtian Liang
- Department of Otolaryngology-Head and Neck Surgery, Tongren Hospital, Wuhan University, Wuhan, Hubei, China.
| | - Dou Liu
- Department of Otolaryngology-Head and Neck Surgery, Tongren Hospital, Wuhan University, Wuhan, Hubei, China.
| | - Chenhui He
- Department of Otolaryngology-Head and Neck Surgery, Tongren Hospital, Wuhan University, Wuhan, Hubei, China.
| | - Longkai Xiao
- Department of Otolaryngology-Head and Neck Surgery, Tongren Hospital, Wuhan University, Wuhan, Hubei, China.
| | - Xianting Gao
- Department of Otolaryngology-Head and Neck Surgery, Tongren Hospital, Wuhan University, Wuhan, Hubei, China.
| |
Collapse
|
3
|
Roman KM, Dinasarapu AR, Cherian S, Fan X, Donsante Y, Aravind N, Chan CS, Jinnah H, Hess EJ. Striatal cell-type-specific molecular signatures reveal therapeutic targets in a model of dystonia. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.07.617010. [PMID: 39415987 PMCID: PMC11482807 DOI: 10.1101/2024.10.07.617010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
Striatal dysfunction is implicated in many forms of dystonia, including idiopathic, inherited and iatrogenic dystonias. The striatum is comprised largely of GABAergic spiny projection neurons (SPNs) that are defined by their long-range efferents. Direct SPNs (dSPNs) project to the internal globus pallidus/substantia nigra reticulata whereas indirect pathway SPNs (iSPNs) project to the external pallidum; the concerted activity of both SPN subtypes modulates movement. Convergent results from genetic, imaging and physiological studies in patients suggest that abnormalities of both dSPNs and iSPNs contribute to the expression of dystonia, but the molecular adaptations underlying these abnormalities are not known. Here we provide a comprehensive analysis of SPN cell-type-specific molecular signatures in a model of DOPA-responsive dystonia (DRD mice), which is caused by gene defects that reduce dopamine neurotransmission, resulting in dystonia that is specifically associated with striatal dysfunction. Individually profiling the translatome of dSPNs and iSPNs using translating ribosome affinity purification with RNA-seq revealed hundreds of differentially translating mRNAs in each SPN subtype in DRD mice, yet there was little overlap between the dysregulated genes in dSPNs and iSPNs. Despite the paucity of shared adaptations, a disruption in glutamatergic signaling was predicted for both dSPNs and iSPNs. Indeed, we found that both AMPA and NMDA receptor-mediated currents were enhanced in dSPNs but diminished in iSPNs in DRD mice. The pattern of mRNA dysregulation was specific to dystonia as the adaptations in DRD mice were distinct from those in parkinsonian mice where the dopamine deficit occurs in adults, suggesting that the phenotypic outcome is dependent on both the timing of the dopaminergic deficit and the SPN-specific adaptions. We leveraged the unique molecular signatures of dSPNs and iSPNs in DRD mice to identify biochemical mechanisms that may be targets for therapeutics, including LRRK2 inhibition. Administration of the LRRK2 inhibitor MLi-2 ameliorated the dystonia in DRD mice suggesting a novel target for therapeutics and demonstrating that the delineation of cell-type-specific molecular signatures provides a powerful approach to revealing both CNS dysfunction and therapeutic targets in dystonia.
Collapse
Affiliation(s)
- Kaitlyn M. Roman
- Department of Pharmacology and Chemical Biology, Emory University, Atlanta, GA, USA
| | | | - Suraj Cherian
- Department of Neuroscience, Northwestern University, Chicago, Illinois, USA
| | - Xueliang Fan
- Department of Pharmacology and Chemical Biology, Emory University, Atlanta, GA, USA
| | - Yuping Donsante
- Department of Pharmacology and Chemical Biology, Emory University, Atlanta, GA, USA
| | - Nivetha Aravind
- Department of Pharmacology and Chemical Biology, Emory University, Atlanta, GA, USA
| | - C. Savio Chan
- Department of Neuroscience, Northwestern University, Chicago, Illinois, USA
| | - H.A. Jinnah
- Department of Neurology, Emory University, Atlanta, Georgia, USA
- Department of Human Genetics, Emory University, Atlanta, Georgia, USA
- Department of Pediatrics, Emory University, Atlanta, Georgia, USA
| | - Ellen J. Hess
- Department of Pharmacology and Chemical Biology, Emory University, Atlanta, GA, USA
- Department of Neurology, Emory University, Atlanta, Georgia, USA
| |
Collapse
|
4
|
Zhou F, Wei L, Wang Y, Chen W. Aerobic exercise modulates the striatal Erk/MAPK signaling pathway and improves LID in a mouse model of Parkinson's disease. Brain Res Bull 2024; 209:110906. [PMID: 38395109 DOI: 10.1016/j.brainresbull.2024.110906] [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: 06/13/2023] [Revised: 01/31/2024] [Accepted: 02/20/2024] [Indexed: 02/25/2024]
Abstract
OBJECTIVE To investigate the role of the striatal extracellular signal-regulated kinase (Erk1/2) and its phosphorylation (p-Erk1/2) in aerobic training to alleviate the development of the L-DOPA induced dyskinesia (LID) in PD mice. METHODS Forty-eight male C57BL/6 N mice were randomly divided into the 6-OHDA surgery group (6-OHDA, n=42) and the sham surgery group (Sham, n=6). A two-point injection of 6-OHDA into the right striatum was used to establish a lateralized injury PD model. PD mice were randomly divided into a PD control group (PD, n=13) and a PD exercise group (PDE, n=16), this is followed by 4 weeks of L-DOPA treatment, and PDE mice received concurrent running table training (18 m/min, 40 min/day, 5 times/week). AIM scores were performed weekly, and mice were assessed for motor function after 4 weeks using the rotarod, open field, and gait tests. Immunohistochemistry was used to test nigrostriatal TH expression, Western blot was used to determine Erk1/2 and p-Erk1/2 protein expression, and immunofluorescence double-labeling technique was used to detect Erk1/2 and p-Erk1/2 co-expression with prodynorphin (PDYN). RESULTS (1) All AIM scores of PD and PDE mice increased significantly (P < 0.05) with the prolongation of L-DOPA treatment. Compared with PD, all AIM scores were significantly lower in PDE mice (P < 0.05). (2) After 4 weeks, the motor function of PD mice was significantly reduced compared with Sham (P < 0.05 or P < 0.01); compared with PD, the motor function of PDE mice was significantly increased (P < 0.05). (3) Compared with Sham, the expression of Erk1/2 protein, the number of positive cells of Erk1/2 and p-Erk1/2 and the number of positive cells co-expressed with PDYN were significantly increased in PD mice (P < 0.05); compared with PD, Erk1/2 protein expression was significantly decreased in PDE mice (P < 0.05), and the number of Erk1/2 and p-Erk1/2 positive cells was significantly reduced (P < 0.05). CONCLUSION 4 weeks of aerobic exercise can effectively alleviate the development of L-DOPA-induced dyskinesia and improve motor function in PD mice. The related mechanism may be related to the inhibition of striatal Erk/MAPK signaling pathway overactivation by aerobic exercise, but this change did not occur selectively in D1-MSNs.
Collapse
Affiliation(s)
- Fangyuan Zhou
- School of Physical Education, Hebei Normal University, Shijiazhuang, Hebei 050024, China
| | - Longwei Wei
- School of Physical Education, Hebei Normal University, Shijiazhuang, Hebei 050024, China
| | - Yinhao Wang
- School of Physical Education, Hebei Normal University, Shijiazhuang, Hebei 050024, China
| | - Wei Chen
- School of Physical Education, Hebei Normal University, Shijiazhuang, Hebei 050024, China; Key Laboratory of Measurement and Evaluation in Exercise Bioinformation of Hebei Province, School of Physical Education, Hebei Normal University, Shijiazhuang, Hebei 050024, China.
| |
Collapse
|
5
|
Zhu J, Hu Z, Luo Y, Liu Y, Luo W, Du X, Luo Z, Hu J, Peng S. Diabetic peripheral neuropathy: pathogenetic mechanisms and treatment. Front Endocrinol (Lausanne) 2024; 14:1265372. [PMID: 38264279 PMCID: PMC10803883 DOI: 10.3389/fendo.2023.1265372] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2023] [Accepted: 12/14/2023] [Indexed: 01/25/2024] Open
Abstract
Diabetic peripheral neuropathy (DPN) refers to the development of peripheral nerve dysfunction in patients with diabetes when other causes are excluded. Diabetic distal symmetric polyneuropathy (DSPN) is the most representative form of DPN. As one of the most common complications of diabetes, its prevalence increases with the duration of diabetes. 10-15% of newly diagnosed T2DM patients have DSPN, and the prevalence can exceed 50% in patients with diabetes for more than 10 years. Bilateral limb pain, numbness, and paresthesia are the most common clinical manifestations in patients with DPN, and in severe cases, foot ulcers can occur, even leading to amputation. The etiology and pathogenesis of diabetic neuropathy are not yet completely clarified, but hyperglycemia, disorders of lipid metabolism, and abnormalities in insulin signaling pathways are currently considered to be the initiating factors for a range of pathophysiological changes in DPN. In the presence of abnormal metabolic factors, the normal structure and function of the entire peripheral nervous system are disrupted, including myelinated and unmyelinated nerve axons, perikaryon, neurovascular, and glial cells. In addition, abnormalities in the insulin signaling pathway will inhibit neural axon repair and promote apoptosis of damaged cells. Here, we will discuss recent advances in the study of DPN mechanisms, including oxidative stress pathways, mechanisms of microvascular damage, mechanisms of damage to insulin receptor signaling pathways, and other potential mechanisms associated with neuroinflammation, mitochondrial dysfunction, and cellular oxidative damage. Identifying the contributions from each pathway to neuropathy and the associations between them may help us to further explore more targeted screening and treatment interventions.
Collapse
Affiliation(s)
- Jinxi Zhu
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
- The Second Clinical Medical College of Nanchang University, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Ziyan Hu
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
- The Second Clinical Medical College of Nanchang University, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Yifan Luo
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Yinuo Liu
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Wei Luo
- Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Xiaohong Du
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Zhenzhong Luo
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Jialing Hu
- Department of Emergency Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Shengliang Peng
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| |
Collapse
|
6
|
Gómez-Pineda VG, Nieto-Mendoza E, Torres-Cruz FM, Hernández-Echeagaray E. Neurotrophin-3 Rescues Striatal Synaptic Plasticity in Model of Neurodegeneration by PLC Signaling Activation. CNS & NEUROLOGICAL DISORDERS DRUG TARGETS 2024; 23:1488-1498. [PMID: 38859788 DOI: 10.2174/0118715273298919240531110022] [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] [Received: 01/18/2024] [Revised: 04/30/2024] [Accepted: 05/14/2024] [Indexed: 06/12/2024]
Abstract
BACKGROUND Neurotrophins are essential factors for neural growth and function; they play a crucial role in neurodegenerative diseases where their expression levels are altered. Our previous research has demonstrated changes in synaptic plasticity and neurotrophin expression levels in a pharmacological model of Huntington's disease (HD) induced by 3-nitropropionic acid (3-NP). In the 3-NP-induced HD model, corticostriatal Long Term Depression (LTD) was impaired, but neurotrophin- 3 (NT-3) restored striatal LTD. This study delves into the NT-3-induced signaling pathways involved in modulating and restoring striatal synaptic plasticity in cerebral slices from 3-NPinduced striatal degeneration in mice in vivo. METHODS Phospholipase C (PLC), phosphatidylinositol-3-kinase (PI3K), and mitogen-activated protein kinase (MEK)/extracellular signal-regulated kinase (ERK) pathways activated by NT-3 were analyzed by means of field electrophysiological recordings in brain slices from control and 3-NP treated in the presence of specific inhibitors of the signaling pathways. RESULTS Using specific inhibitors, PLC, PI3K, and MEK/ERK signaling pathways contribute to NT-3-mediated plasticity modulation in striatal tissue slices recorded from control animals. However, in the neurodegeneration model induced by 3-NP, the recovery of striatal LTD induced by NT-3 was prevented only by the PLC inhibitor. Moreover, the PLC signaling pathway appeared to trigger downstream activation of the endocannabinoid system, evidenced by AM 251, an inhibitor of the CB1 receptor, also hindered NT-3 plasticity recovery. CONCLUSION Our finding highlights the specific involvement of the PLC pathway in the neuroprotective effects of NT-3 in mitigating synaptic dysfunction under neurodegenerative conditions.
Collapse
Affiliation(s)
- Victor G Gómez-Pineda
- Laboratorio de Neurofisiología del Desarrollo y la Neurodegeneración, Facultad de Estudios Superiores Iztacala, Unidad de Investigación en Biomedicina, Universidad Nacional Autónoma de México, Av. De los Barrios No. 1, Los Reyes Iztacala, Tlalnepantla, Estado de México, C.P. 54090, México
| | - Elizabeth Nieto-Mendoza
- Laboratorio de Neurofisiología del Desarrollo y la Neurodegeneración, Facultad de Estudios Superiores Iztacala, Unidad de Investigación en Biomedicina, Universidad Nacional Autónoma de México, Av. De los Barrios No. 1, Los Reyes Iztacala, Tlalnepantla, Estado de México, C.P. 54090, México
| | - Francisco M Torres-Cruz
- Laboratorio de Neurofisiología del Desarrollo y la Neurodegeneración, Facultad de Estudios Superiores Iztacala, Unidad de Investigación en Biomedicina, Universidad Nacional Autónoma de México, Av. De los Barrios No. 1, Los Reyes Iztacala, Tlalnepantla, Estado de México, C.P. 54090, México
| | - Elizabeth Hernández-Echeagaray
- Laboratorio de Neurofisiología del Desarrollo y la Neurodegeneración, Facultad de Estudios Superiores Iztacala, Unidad de Investigación en Biomedicina, Universidad Nacional Autónoma de México, Av. De los Barrios No. 1, Los Reyes Iztacala, Tlalnepantla, Estado de México, C.P. 54090, México
| |
Collapse
|
7
|
Jastrzębski MK, Wójcik P, Stępnicki P, Kaczor AA. Effects of small molecules on neurogenesis: Neuronal proliferation and differentiation. Acta Pharm Sin B 2024; 14:20-37. [PMID: 38239239 PMCID: PMC10793103 DOI: 10.1016/j.apsb.2023.10.007] [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: 06/07/2023] [Revised: 09/06/2023] [Accepted: 09/13/2023] [Indexed: 01/22/2024] Open
Abstract
Neurons are believed to be non-proliferating cells. However, neuronal stem cells are still present in certain areas of the adult brain, although their proliferation diminishes with age. Just as with other cells, their proliferation and differentiation are modulated by various mechanisms. These mechanisms are foundational to the strategies developed to induce neuronal proliferation and differentiation, with potential therapeutic applications for neurodegenerative diseases. The most common among these diseases are Parkinson's disease and Alzheimer's disease, associated with the formation of β -amyloid (Aβ ) aggregates which cause a reduction in the number of neurons. Compounds such as LiCl, 4-aminothiazoles, Pregnenolone, ACEA, harmine, D2AAK1, methyl 3,4-dihydroxybenzoate, and shikonin may induce neuronal proliferation/differentiation through the activation of pathways: MAPK ERK, PI3K/AKT, NFκ B, Wnt, BDNF, and NPAS3. Moreover, combinations of these compounds can potentially transform somatic cells into neurons. This transformation process involves the activation of neuron-specific transcription factors such as NEUROD1, NGN2, ASCL1, and SOX2, which subsequently leads to the transcription of downstream genes, culminating in the transformation of somatic cells into neurons. Neurodegenerative diseases are not the only conditions where inducing neuronal proliferation could be beneficial. Consequently, the impact of pro-proliferative compounds on neurons has also been researched in mouse models of Alzheimer's disease.
Collapse
Affiliation(s)
- Michał K. Jastrzębski
- Department of Synthesis and Chemical Technology of Pharmaceutical Substances with Computer Modeling Laboratory, Medical University of Lublin, Faculty of Pharmacy, Lublin PL-20093, Poland
| | - Piotr Wójcik
- Department of Synthesis and Chemical Technology of Pharmaceutical Substances with Computer Modeling Laboratory, Medical University of Lublin, Faculty of Pharmacy, Lublin PL-20093, Poland
| | - Piotr Stępnicki
- Department of Synthesis and Chemical Technology of Pharmaceutical Substances with Computer Modeling Laboratory, Medical University of Lublin, Faculty of Pharmacy, Lublin PL-20093, Poland
| | - Agnieszka A. Kaczor
- Department of Synthesis and Chemical Technology of Pharmaceutical Substances with Computer Modeling Laboratory, Medical University of Lublin, Faculty of Pharmacy, Lublin PL-20093, Poland
- School of Pharmacy, University of Eastern Finland, Kuopio FI-70211, Finland
| |
Collapse
|
8
|
Ye Q, Srivastava P, Al-Kuwari N, Chen X. Oncogenic BRAFV600E induces microglial proliferation through extracellular signal-regulated kinase and neuronal death through c-Jun N-terminal kinase. Neural Regen Res 2023; 18:1613-1622. [PMID: 36571370 PMCID: PMC10075110 DOI: 10.4103/1673-5374.361516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 06/13/2022] [Accepted: 10/18/2022] [Indexed: 11/19/2022] Open
Abstract
Activating V600E in v-Raf murine sarcoma viral oncogene homolog B (BRAF) is a common driver mutation in cancers of multiple tissue origins, including melanoma and glioma. BRAFV600E has also been implicated in neurodegeneration. The present study aims to characterize BRAFV600E during cell death and proliferation of three major cell types of the central nervous system: neurons, astrocytes, and microglia. Multiple primary cultures (primary cortical mixed culture) and cell lines of glial cells (BV2) and neurons (SH-SY5Y) were employed. BRAFV600E and BRAFWT expression was mediated by lentivirus or retrovirus. Blockage of downstream effectors (extracellular signal-regulated kinase 1/2 and JNK1/2) were achieved by siRNA. In astrocytes and microglia, BRAFV600E induces cell proliferation, and the proliferative effect in microglia is mediated by activated extracellular signal-regulated kinase, but not c-Jun N-terminal kinase. Conditioned medium from BRAFV600E-expressing microglia induced neuronal death. In neuronal cells, BRAFV600E directly induces neuronal death, through c-Jun N-terminal kinase but not extracellular signal-regulated kinase. We further show that BRAF-related genes are enriched in pathways in patients with Parkinson's disease. Our study identifies distinct consequences mediated by distinct downstream effectors in dividing glial cells and in neurons following the same BRAF mutational activation and a causal link between BRAF-activated microglia and neuronal cell death that does not require physical proximity. It provides insight into a possibly important role of BRAF in neurodegeneration as a result of either dysregulated BRAF in neurons or its impact on glial cells.
Collapse
Affiliation(s)
- Qing Ye
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
- Department of Neurology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Pranay Srivastava
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Nasser Al-Kuwari
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Xiqun Chen
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| |
Collapse
|
9
|
Guo Y, Fu Y, Sun W. 50 Hz Magnetic Field Exposure Inhibited Spontaneous Movement of Zebrafish Larvae through ROS-Mediated syn2a Expression. Int J Mol Sci 2023; 24:ijms24087576. [PMID: 37108734 PMCID: PMC10144198 DOI: 10.3390/ijms24087576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 03/21/2023] [Accepted: 04/15/2023] [Indexed: 04/29/2023] Open
Abstract
Extremely low frequency electromagnetic field (ELF-EMF) exists widely in public and occupational environments. However, its potential adverse effects and the underlying mechanism on nervous system, especially behavior are still poorly understood. In this study, zebrafish embryos (including a transfected synapsin IIa (syn2a) overexpression plasmid) at 3 h post-fertilization (hpf) were exposed to a 50-Hz magnetic field (MF) with a series of intensities (100, 200, 400 and 800 μT, respectively) for 1 h or 24 h every day for 5 days. Results showed that, although MF exposure did not affect the basic development parameters including hatching rate, mortality and malformation rate, yet MF at 200 μT could significantly induce spontaneous movement (SM) hypoactivity in zebrafish larvae. Histological examination presented morphological abnormalities of the brain such as condensed cell nucleus and cytoplasm, increased intercellular space. Moreover, exposure to MF at 200 μT inhibited syn2a transcription and expression, and increased reactive oxygen species (ROS) level as well. Overexpression of syn2a could effectively rescue MF-induced SM hypoactivity in zebrafish. Pretreatment with N-acetyl-L-cysteine (NAC) could not only recover syn2a protein expression which was weakened by MF exposure, but also abolish MF-induced SM hypoactivity. However, syn2a overexpression did not affect MF-increased ROS. Taken together, the findings suggested that exposure to a 50-Hz MF inhibited spontaneous movement of zebrafish larvae via ROS-mediated syn2a expression in a nonlinear manner.
Collapse
Affiliation(s)
- Yixin Guo
- Bioelectromagnetics Key Laboratory, Zhejiang University School of Medicine, Hangzhou 310058, China
- Institute of Environmental Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Yiti Fu
- Institute of Environmental Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Wenjun Sun
- Bioelectromagnetics Key Laboratory, Zhejiang University School of Medicine, Hangzhou 310058, China
- Institute of Environmental Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China
| |
Collapse
|
10
|
Montanez-Miranda C, Bramlett SN, Hepler JR. RGS14 expression in CA2 hippocampus, amygdala, and basal ganglia: Implications for human brain physiology and disease. Hippocampus 2023; 33:166-181. [PMID: 36541898 PMCID: PMC9974931 DOI: 10.1002/hipo.23492] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 11/09/2022] [Accepted: 12/07/2022] [Indexed: 12/24/2022]
Abstract
RGS14 is a multifunctional scaffolding protein that is highly expressed within postsynaptic spines of pyramidal neurons in hippocampal area CA2. Known roles of RGS14 in CA2 include regulating G protein, H-Ras/ERK, and calcium signaling pathways to serve as a natural suppressor of synaptic plasticity and postsynaptic signaling. RGS14 also shows marked postsynaptic expression in major structures of the limbic system and basal ganglia, including the amygdala and both the ventral and dorsal subdivisions of the striatum. In this review, we discuss the signaling functions of RGS14 and its role in postsynaptic strength (long-term potentiation) and spine structural plasticity in CA2 hippocampal neurons, and how RGS14 suppression of plasticity impacts linked behaviors such as spatial learning, object memory, and fear conditioning. We also review RGS14 expression in the limbic system and basal ganglia and speculate on its possible roles in regulating plasticity in these regions, with a focus on behaviors related to emotion and motivation. Finally, we explore the functional implications of RGS14 in various brain circuits and speculate on its possible roles in certain disease states such as hippocampal seizures, addiction, and anxiety disorders.
Collapse
Affiliation(s)
| | | | - John R. Hepler
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA, 30322-3090
| |
Collapse
|
11
|
Pérez-Arancibia R, Cisternas-Olmedo M, Sepúlveda D, Troncoso-Escudero P, Vidal RL. Small molecules to perform big roles: The search for Parkinson's and Huntington's disease therapeutics. Front Neurosci 2023; 16:1084493. [PMID: 36699535 PMCID: PMC9868863 DOI: 10.3389/fnins.2022.1084493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Accepted: 12/20/2022] [Indexed: 01/12/2023] Open
Abstract
Neurological motor disorders (NMDs) such as Parkinson's disease and Huntington's disease are characterized by the accumulation and aggregation of misfolded proteins that trigger cell death of specific neuronal populations in the central nervous system. Differential neuronal loss initiates the impaired motor control and cognitive function in the affected patients. Although major advances have been carried out to understand the molecular basis of these diseases, to date there are no treatments that can prevent, cure, or significantly delay the progression of the disease. In this context, strategies such as gene editing, cellular therapy, among others, have gained attention as they effectively reduce the load of toxic protein aggregates in different models of neurodegeneration. Nevertheless, these strategies are expensive and difficult to deliver into the patients' nervous system. Thus, small molecules and natural products that reduce protein aggregation levels are highly sought after. Numerous drug discovery efforts have analyzed large libraries of synthetic compounds for the treatment of different NMDs, with a few candidates reaching clinical trials. Moreover, the recognition of new druggable targets for NMDs has allowed the discovery of new small molecules that have demonstrated their efficacy in pre-clinical studies. It is also important to recognize the contribution of natural products to the discovery of new candidates that can prevent or cure NMDs. Additionally, the repurposing of drugs for the treatment of NMDs has gained huge attention as they have already been through clinical trials confirming their safety in humans, which can accelerate the development of new treatment. In this review, we will focus on the new advances in the discovery of small molecules for the treatment of Parkinson's and Huntington's disease. We will begin by discussing the available pharmacological treatments to modulate the progression of neurodegeneration and to alleviate the motor symptoms in these diseases. Then, we will analyze those small molecules that have reached or are currently under clinical trials, including natural products and repurposed drugs.
Collapse
Affiliation(s)
- Rodrigo Pérez-Arancibia
- Center for Integrative Biology, Faculty of Sciences, Universidad Mayor, Santiago, Chile
- Departamento de Ciencias Básicas, Faculty of Medicine and Science, Universidad San Sebastián, Santiago, Chile
| | - Marisol Cisternas-Olmedo
- Center for Integrative Biology, Faculty of Sciences, Universidad Mayor, Santiago, Chile
- Biomedical Neuroscience Institute, Faculty of Medicine, University of Chile, Santiago, Chile
- Center for Geroscience, Brain Health and Metabolism, Santiago, Chile
| | - Denisse Sepúlveda
- Center for Integrative Biology, Faculty of Sciences, Universidad Mayor, Santiago, Chile
- Biomedical Neuroscience Institute, Faculty of Medicine, University of Chile, Santiago, Chile
- Center for Geroscience, Brain Health and Metabolism, Santiago, Chile
| | - Paulina Troncoso-Escudero
- Molecular Diagnostic and Biomarkers Laboratory, Department of Pathology, Faculty of Medicine Clínica Alemana, Universidad del Desarrollo, Santiago, Chile
| | - Rene L. Vidal
- Center for Integrative Biology, Faculty of Sciences, Universidad Mayor, Santiago, Chile
- Biomedical Neuroscience Institute, Faculty of Medicine, University of Chile, Santiago, Chile
- Center for Geroscience, Brain Health and Metabolism, Santiago, Chile
| |
Collapse
|
12
|
Wei Z, Cheng Q, Xu N, Zhao C, Xu J, Kang L, Lou X, Yu L, Feng W. Investigation of CRS-associated cytokines in CAR-T therapy with meta-GNN and pathway crosstalk. BMC Bioinformatics 2022; 23:373. [PMID: 36100873 PMCID: PMC9469618 DOI: 10.1186/s12859-022-04917-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Accepted: 09/06/2022] [Indexed: 11/24/2022] Open
Abstract
Background Chimeric antigen receptor T-cell (CAR-T) therapy is a new and efficient cellular immunotherapy. The therapy shows significant efficacy, but also has serious side effects, collectively known as cytokine release syndrome (CRS). At present, some CRS-related cytokines and their roles in CAR-T therapy have been confirmed by experimental studies. However, the mechanism of CRS remains to be fully understood. Methods Based on big data for human protein interactions and meta-learning graph neural network, we employed known CRS-related cytokines to comprehensively investigate the CRS associated cytokines in CAR-T therapy through protein interactions. Subsequently, the clinical data for 119 patients who received CAR-T therapy were examined to validate our prediction results. Finally, we systematically explored the roles of the predicted cytokines in CRS occurrence by protein interaction network analysis, functional enrichment analysis, and pathway crosstalk analysis. Results We identified some novel cytokines that would play important roles in biological process of CRS, and investigated the biological mechanism of CRS from the perspective of functional analysis. Conclusions 128 cytokines and related molecules had been found to be closely related to CRS in CAR-T therapy, where several important ones such as IL6, IFN-γ, TNF-α, ICAM-1, VCAM-1 and VEGFA were highlighted, which can be the key factors to predict CRS. Supplementary Information The online version contains supplementary material available at 10.1186/s12859-022-04917-2.
Collapse
Affiliation(s)
- Zhenyu Wei
- College of Intelligent Systems Science and Engineering, Institute of Intelligent System and Bioinformatics, Harbin Engineering University, Harbin, 150001, China
| | - Qi Cheng
- College of Intelligent Systems Science and Engineering, Institute of Intelligent System and Bioinformatics, Harbin Engineering University, Harbin, 150001, China
| | - Nan Xu
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, Institute of Biomedical Engineering and Technology, East China Normal University, No. 3663 North Zhongshan Road, Shanghai, 200065, China.,Shanghai Unicar-Therapy Bio-Medicine Technology Co., Ltd, Shanghai, China
| | - Chengkui Zhao
- College of Intelligent Systems Science and Engineering, Institute of Intelligent System and Bioinformatics, Harbin Engineering University, Harbin, 150001, China
| | - Jiayu Xu
- College of Intelligent Systems Science and Engineering, Institute of Intelligent System and Bioinformatics, Harbin Engineering University, Harbin, 150001, China
| | - Liqing Kang
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, Institute of Biomedical Engineering and Technology, East China Normal University, No. 3663 North Zhongshan Road, Shanghai, 200065, China.,Shanghai Unicar-Therapy Bio-Medicine Technology Co., Ltd, Shanghai, China
| | - Xiaoyan Lou
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, Institute of Biomedical Engineering and Technology, East China Normal University, No. 3663 North Zhongshan Road, Shanghai, 200065, China.,Shanghai Unicar-Therapy Bio-Medicine Technology Co., Ltd, Shanghai, China
| | - Lei Yu
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, Institute of Biomedical Engineering and Technology, East China Normal University, No. 3663 North Zhongshan Road, Shanghai, 200065, China. .,Shanghai Unicar-Therapy Bio-Medicine Technology Co., Ltd, Shanghai, China.
| | - Weixing Feng
- College of Intelligent Systems Science and Engineering, Institute of Intelligent System and Bioinformatics, Harbin Engineering University, Harbin, 150001, China.
| |
Collapse
|
13
|
Zhu L, Wu F, Yan Z, He L, Wang S, Hu H, Goh ELK, Zhu Y, Guan F, Chen T. A novel microRNA, novel-m009C, regulates methamphetamine rewarding effects. Mol Psychiatry 2022; 27:3885-3897. [PMID: 35715487 PMCID: PMC9708597 DOI: 10.1038/s41380-022-01651-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 05/18/2022] [Accepted: 05/31/2022] [Indexed: 02/08/2023]
Abstract
Methamphetamine (METH) is a widely abused psychostimulant, whose hyper-rewarding property is believed to underlie its addictive effect, but the molecular mechanism regulating this effect remains unclear. We previously reported that decreased expression of a novel microRNA (miRNA), novel-m009C, is implicated in the regulation of METH hyperlocomotion. Here, we found that novel-m009C may be homologous to hsa-miR-604. Its expression is consistently downregulated in the nucleus accumbens (NAc) of mice when exposed to METH and cocaine, whereas significant alterations in novel-m009C expression were not observed in the NAc of mice subjected to other rewarding and psychiatric stimuli, such as sucrose, morphine and MK-801. We further found the substantial reduction in novel-m009C expression may be regulated by both dopamine receptor D1 (D1R) and D2 (D2R). Increasing novel-m009C levels in the NAc attenuated METH-induced conditioned place preference (CPP) and hyperlocomotion, whereas inhibiting novel-m009C expression in the NAc enhanced these effects but did not change the preference of mice for a natural reward, i.e., sucrose. These effects may involve targeting of genes important for the synaptic transmission, such as Grin1 (NMDAR subunit 1). Our findings demonstrate an important role for NAc novel-m009C in regulating METH reward, reveal a novel molecular regulator of the actions of METH on brain reward circuitries and provide a new strategy for treating METH addiction based on the modulation of small non-coding RNAs.
Collapse
Affiliation(s)
- Li Zhu
- College of Forensic Medicine, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, Shaanxi, PR China
- The Key Laboratory of Health Ministry for Forensic Science, Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, PR China
| | - Feifei Wu
- College of Forensic Medicine, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, Shaanxi, PR China
- The Key Laboratory of Health Ministry for Forensic Science, Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, PR China
| | - Zhilan Yan
- College of Forensic Medicine, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, Shaanxi, PR China
- The Key Laboratory of Health Ministry for Forensic Science, Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, PR China
| | - Lijun He
- College of Forensic Medicine, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, Shaanxi, PR China
- The Key Laboratory of Health Ministry for Forensic Science, Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, PR China
| | - Shufei Wang
- College of Forensic Medicine, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, Shaanxi, PR China
- The Key Laboratory of Health Ministry for Forensic Science, Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, PR China
| | - Haohao Hu
- College of Forensic Medicine, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, Shaanxi, PR China
- The Key Laboratory of Health Ministry for Forensic Science, Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, PR China
| | - Eyleen L K Goh
- Neuroscience and Mental Health Faculty, Lee Kong Chian School of Medicine, Nanyang Technological University, 308232, Singapore, Singapore
| | - Yingjie Zhu
- Shenzhen Key Laboratory of Drug Addiction, CAS Key Laboratory of Brain Connectome and Manipulation, the Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, PR China
- Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, 518055, PR China
| | - Fanglin Guan
- College of Forensic Medicine, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, Shaanxi, PR China.
- The Key Laboratory of Health Ministry for Forensic Science, Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, PR China.
| | - Teng Chen
- College of Forensic Medicine, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, Shaanxi, PR China.
- The Key Laboratory of Health Ministry for Forensic Science, Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, PR China.
| |
Collapse
|
14
|
Xu W, Zheng J, Wang X, Zhou B, Chen H, Li G, Yan F. tRF-Val-CAC-016 modulates the transduction of CACNA1d-mediated MAPK signaling pathways to suppress the proliferation of gastric carcinoma. Cell Commun Signal 2022; 20:68. [PMID: 35590368 PMCID: PMC9118711 DOI: 10.1186/s12964-022-00857-9] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2021] [Accepted: 03/04/2022] [Indexed: 11/25/2022] Open
Abstract
Background As a new kind of non-coding RNAs (ncRNAs), tRNA derivatives play an important role in gastric carcinoma (GC). Nevertheless, the underlying mechanism tRNA derivatives were involved in was rarely illustrated. Methods We screened out the tRNA derivative, tRF-Val-CAC-016, based on the tsRNA sequencing and demonstrated the effect tRF-Val-CAC-016 exerted on GC proliferation in vitro and in vivo. We applied Dual-luciferase reporter assay, RIP assay, and bioinformatic analysis to discover the downstream target of tRF-Val-CAC-016. Then CACNA1d was selected, and the oncogenic characteristics were verified. Subsequently, we detected the possible regulation of the canonical MAPK signaling pathway to further explore the downstream mechanism of tRF-Val-CAC-016. Results As a result, we found that tRF-Val-CAC-016 was low-expressed in GC, and upregulation of tRF-Val-CAC-016 could significantly suppress the proliferation of GC cell lines. Meanwhile, tRF-Val-CAC-016 regulated the canonical MAPK signaling pathway by targeting CACNA1d. Conclusions tRF-Val-CAC-016 modulates the transduction of CACNA1d-mediated MAPK signaling pathways to suppress the proliferation of gastric carcinoma. This study discussed the function and mechanism of tRF-Val-CAC-016 in GC for the first time. The pioneering work has contributed to our present understanding of tRNA derivative, which might provide an alternative mean for the targeted therapy of GC. Video abstract
Supplementary Information The online version contains supplementary material available at 10.1186/s12964-022-00857-9.
Collapse
Affiliation(s)
- Weiguo Xu
- Department of General Surgery, Jiangsu Cancer Hospital & The Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Institute of Cancer Research, Nanjing, China
| | - Junyu Zheng
- Department of Clinical Laboratory, Jiangsu Cancer Hospital & The Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Institute of Cancer Research, Baiziting No. 42, Nanjing, 210009, Jiangsu, China
| | - Xiao Wang
- Department of Radiology, Jiangsu Cancer Hospital & The Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Institute of Cancer Research, Nanjing, China
| | - Bin Zhou
- Department of Gastric Surgery, Jiangsu Cancer Hospital & The Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Institute of Cancer Research, Baiziting No. 42, Nanjing, 210009, Jiangsu, China
| | - Huanqiu Chen
- Department of Gastric Surgery, Jiangsu Cancer Hospital & The Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Institute of Cancer Research, Baiziting No. 42, Nanjing, 210009, Jiangsu, China.
| | - Gang Li
- Department of Gastric Surgery, Jiangsu Cancer Hospital & The Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Institute of Cancer Research, Baiziting No. 42, Nanjing, 210009, Jiangsu, China.
| | - Feng Yan
- Department of Clinical Laboratory, Jiangsu Cancer Hospital & The Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Institute of Cancer Research, Baiziting No. 42, Nanjing, 210009, Jiangsu, China.
| |
Collapse
|
15
|
Wang X, Wang Y, Chen J, Li J, Liu Y, Chen W. Aerobic exercise improves motor function and striatal MSNs-Erk/MAPK signaling in mice with 6-OHDA-induced Parkinson's disease. Exp Brain Res 2022; 240:1713-1725. [PMID: 35384454 PMCID: PMC8985567 DOI: 10.1007/s00221-022-06360-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2021] [Accepted: 03/25/2022] [Indexed: 01/30/2023]
Abstract
In Parkinson’s disease (PD) state, with progressive loss of dopaminergic neurons in the substantia nigra, the striatal dopamine (DA) and glutamate (Glu) levels change, resulting in dysfunction of basal ganglia motor regulation. The PD patient presents motor dysfunction such as resting tremor, bradykinesia, and muscular rigidity. To investigate the mechanism of aerobic exercise to improve PD-related motor dysfunction, in the current study, 6-hydroxydopamine (6-OHDA) was used to induce the PD mice model, and the motor function of PD mice was comprehensively evaluated by open-field test, rotarod test, and gait test. The co-expression of prodynorphin (PDYN) and proenkephalin (PENK) with extracellular signal-regulated kinase (Erk1/2) and phosphorylation Erk1/2 (p-Erk1/2) were detected by double-labeling immunofluorescence. The results showed that a 4-week aerobic exercise intervention could effectively improve the motor dysfunction of PD mice. Moreover, it was found that the expressions of Erk1/2 and p-Erk1/2 in the dorsal striatum (Str) of PD mice were significantly increased, and the number of positive cells co-expressed by Erk1/2, p-Erk1/2, and PENK was significantly higher than PDYN. The above phenomenon was reversed by a 4-week aerobic exercise intervention. Therefore, this study suggests that the mechanism by which aerobic exercise improves PD-related motor dysfunction may be related to that the aerobic exercise intervention alleviates the activity of extracellular signal-regulated kinase/mitogen-activated protein kinases (Erk/MAPK) signaling pathway in striatal medium spiny neurons expressing D2-like receptors (D2-MSNs) of PD mice by regulating the striatal DA and Glu signaling.
Collapse
Affiliation(s)
- Xiaodong Wang
- Key Laboratory of Measurement and Evaluation in Exercise Bioinformation of Hebei Province, School of Physical Education, Hebei Normal University, Shijiazhuang, Hebei, China
| | - Yinhao Wang
- Key Laboratory of Measurement and Evaluation in Exercise Bioinformation of Hebei Province, School of Physical Education, Hebei Normal University, Shijiazhuang, Hebei, China
| | - Jian Chen
- Department of General Surgery, Bethune International Peace Hospital of The People's Liberation Army, Shijiazhuang, Hebei, China
| | - Juan Li
- Key Laboratory of Measurement and Evaluation in Exercise Bioinformation of Hebei Province, School of Physical Education, Hebei Normal University, Shijiazhuang, Hebei, China
| | - Yang Liu
- Key Laboratory of Measurement and Evaluation in Exercise Bioinformation of Hebei Province, School of Physical Education, Hebei Normal University, Shijiazhuang, Hebei, China
| | - Wei Chen
- Key Laboratory of Measurement and Evaluation in Exercise Bioinformation of Hebei Province, School of Physical Education, Hebei Normal University, Shijiazhuang, Hebei, China.
| |
Collapse
|
16
|
Selective Manipulation of G-Protein γ 7 Subunit in Mice Provides New Insights into Striatal Control of Motor Behavior. J Neurosci 2021; 41:9065-9081. [PMID: 34544837 DOI: 10.1523/jneurosci.1211-21.2021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 08/26/2021] [Accepted: 09/11/2021] [Indexed: 01/15/2023] Open
Abstract
Stimulatory coupling of dopamine D1 (D1R) and adenosine A2A receptors (A2AR) to adenylyl cyclase within the striatum is mediated through a specific Gαolfβ2γ7 heterotrimer to ultimately modulate motor behaviors. To dissect the individual roles of the Gαolfβ2γ7 heterotrimer in different populations of medium spiny neurons (MSNs), we produced and characterized conditional mouse models, in which the Gng7 gene was deleted in either the D1R- or A2AR/D2R-expressing MSNs. We show that conditional loss of γ7 disrupts the cell type-specific assembly of the Gαolfβ2γ7 heterotrimer, thereby identifying its circumscribed roles acting downstream of either the D1Rs or A2ARs in coordinating motor behaviors, including in vivo responses to psychostimulants. We reveal that Gαolfβ2γ7/cAMP signal in D1R-MSNs does not impact spontaneous and amphetamine-induced locomotor behaviors in male and female mice, while its loss in A2AR/D2R-MSNs results in a hyperlocomotor phenotype and enhanced locomotor response to amphetamine. Additionally, Gαolfβ2γ7/cAMP signal in either D1R- or A2AR/D2R-expressing MSNs is not required for the activation of PKA signaling by amphetamine. Finally, we show that Gαolfβ2γ7 signaling acting downstream of D1Rs is selectively implicated in the acute locomotor-enhancing effects of morphine. Collectively, these results support the general notion that receptors use specific Gαβγ proteins to direct the fidelity of downstream signaling pathways and to elicit a diverse repertoire of cellular functions. Specifically, these findings highlight the critical role for the γ7 protein in determining the cellular level, and hence, the function of the Gαolfβ2γ7 heterotrimer in several disease states associated with dysfunctional striatal signaling.SIGNIFICANCE STATEMENT Dysfunction or imbalance of cAMP signaling in the striatum has been linked to several neurologic and neuropsychiatric disorders, including Parkinson's disease, dystonia, schizophrenia, and drug addiction. By genetically targeting the γ7 subunit in distinct striatal neuronal subpopulations in mice, we demonstrate that the formation and function of the Gαolfβ2γ7 heterotrimer, which represents the rate-limiting step for cAMP production in the striatum, is selectively disrupted. Furthermore, we reveal cell type-specific roles for Gαolfβ2γ7-mediated cAMP production in the control of spontaneous locomotion as well as behavioral and molecular responses to psychostimulants. Our findings identify the γ7 protein as a novel therapeutic target for disease states associated with dysfunctional striatal cAMP signaling.
Collapse
|
17
|
Isaac AR, de Velasco PC, Fraga KYD, Tavares-do-Carmo MDG, Campos RMP, Iannotti FA, Verde R, Martins DBG, Santos TA, Ferreira BK, de Mello FG, Di Marzo V, Andrade-da-Costa BLDS, de Melo Reis RA. Maternal omega-3 intake differentially affects the endocannabinoid system in the progeny`s neocortex and hippocampus: Impact on synaptic markers. J Nutr Biochem 2021; 96:108782. [PMID: 34038760 DOI: 10.1016/j.jnutbio.2021.108782] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 03/16/2021] [Accepted: 04/29/2021] [Indexed: 12/16/2022]
Abstract
Omega-3 (n-3) polyunsaturated fatty acids (PUFA) and the endocannabinoid system (ECS) modulate several functions through neurodevelopment including synaptic plasticity mechanisms. The interplay between n-3PUFA and the ECS during the early stages of development, however, is not fully understood. This study investigated the effects of maternal n-3PUFA supplementation (n-3Sup) or deficiency (n-3Def) on ECS and synaptic markers in postnatal offspring. Female rats were fed with a control, n-3Def, or n-3Sup diet from 15 days before mating and during pregnancy. The cerebral cortex and hippocampus of mothers and postnatal 1-2 days offspring were analyzed. In the mothers, a n-3 deficiency reduced CB1 receptor (CB1R) protein levels in the cortex and increased CB2 receptor (CB2R) in both cortex and hippocampus. In neonates, a maternal n-3 deficiency reduced the hippocampal CB1R amount while it increased CB2R. Additionally, total GFAP isoform expression was increased in both cortex and hippocampus in neonates of the n-3Def group. Otherwise, maternal n-3 supplementation increased the levels of n-3-derived endocannabinoids, DHEA and EPEA, in the cortex and hippocampus and reduced 2-arachidonoyl-glycerol (2-AG) concentrations in the cortex of the offspring. Furthermore, maternal n-3 supplementation also increased PKA phosphorylation in the cortex and ERK phosphorylation in the hippocampus. Synaptophysin immunocontent in both regions was also increased. In vitro assays showed that the increase of synaptophysin in the n-3Sup group was independent of CB1R activation. The findings show that variations in maternal dietary omega-3 PUFA levels may impact differently on the ECS and molecular markers in the cerebral cortex and hippocampus of the progeny.
Collapse
Affiliation(s)
- Alinny Rosendo Isaac
- Instituto de Biofísica Carlos Chagas Filho (IBCCF), Centro de Ciências da Saúde, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.
| | | | - Karla Yasmin Dias Fraga
- Instituto de Nutrição Josué de Castro (INJC), Centro de Ciências da Saúde, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Maria das Graças Tavares-do-Carmo
- Instituto de Nutrição Josué de Castro (INJC), Centro de Ciências da Saúde, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Raquel Maria Pereira Campos
- Instituto de Biofísica Carlos Chagas Filho (IBCCF), Centro de Ciências da Saúde, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Fabio Arturo Iannotti
- Endocannabinoid Research Group, Istituto di Chimica Biomolecolare (ICB), Consiglio Nazionale delle Ricerche (CNR), Pozzuoli (NA), Italy
| | - Roberta Verde
- Endocannabinoid Research Group, Istituto di Chimica Biomolecolare (ICB), Consiglio Nazionale delle Ricerche (CNR), Pozzuoli (NA), Italy
| | - Danyelly Bruneska Gondim Martins
- Grupo de Bioinformática e prospecção molecular, Laboratório de Imunopatologia Keizo Asami, Universidade Federal de Pernambuco, Recife, Pernambuco, Brazil
| | - Thaysa Aragão Santos
- Grupo de Bioinformática e prospecção molecular, Laboratório de Imunopatologia Keizo Asami, Universidade Federal de Pernambuco, Recife, Pernambuco, Brazil
| | - Bruna Klippel Ferreira
- Departamento de Bioquímica, Centro de Ciências da Saúde, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Fernando Garcia de Mello
- Instituto de Biofísica Carlos Chagas Filho (IBCCF), Centro de Ciências da Saúde, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Vincenzo Di Marzo
- Endocannabinoid Research Group, Istituto di Chimica Biomolecolare (ICB), Consiglio Nazionale delle Ricerche (CNR), Pozzuoli (NA), Italy; Canada Exellence Research Chair on the Microbiome-Endocannabinoidome Axis in Metabolic Health, CRIUCPQ and NUTRISS-INAF Universitè Laval, Quebec City, Canada
| | | | - Ricardo Augusto de Melo Reis
- Instituto de Biofísica Carlos Chagas Filho (IBCCF), Centro de Ciências da Saúde, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
18
|
Qin D, Zhou Y, Zhang P, Liu B, Zheng Q, Zhang Z. Azadirachtin downregulates the expression of the CREB gene and protein in the brain and directly or indirectly affects the cognitive behavior of the Spodoptera litura fourth-instar larvae. PEST MANAGEMENT SCIENCE 2021; 77:1873-1885. [PMID: 33284470 DOI: 10.1002/ps.6212] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 11/21/2020] [Accepted: 12/07/2020] [Indexed: 06/12/2023]
Abstract
BACKGROUND Azadirachtin has the potential to be used for pest control. Nevertheless, few studies have investigated the effects of azadirachtin on the cognitive behavior of pests. In this study, expression of the cAMP response element-binding protein (CREB) and its gene were studied via a series of experiments in Spodoptera litura larvae treated with azadirachtin. RESULTS RNA-Seq analysis of S. litura larvae treated with azadirachtin was undertaken. According to Kyoto Encyclopedia of Genes and Genomes analysis, the top 20 enriched pathways included neuroactive ligand-receptor interaction pathways, with seven significantly differentially expressed genes including CREB. Quantitative real time polymerase chain reaction (qRT-PCR) results indicated that the CREB gene was expressed during all developmental stages of S. litura, but relative expression of the CREB gene was significantly downregulated after treatment with azadirachtin. Grayscale statistical analysis also showed that expression levels of protein kinase A (PKA), extracellular signal-regulated kinase (ERK) and CREB proteins were significantly downregulated after treatment with azadirachtin. Moreover, RNA interference results showed that the effect of azadirachtin on the cognitive behavior of S. litura was consistent with that seen after interfering with CREB. In addition, larval selectivity to addictive odor sources was reduced, and the initial reaction time was increased. CONCLUSIONS This study clarified that azadirachtin can affect the cognitive behavior of S. litura and treatment with azadirachtin resulted in a downregulation of gene and protein expression of CREB and its pathway proteins. © 2020 Society of Chemical Industry.
Collapse
Affiliation(s)
- Deqiang Qin
- Key Laboratory of Natural Pesticide and Chemical Biology, Ministry of Education, South China Agricultural University, Guangzhou, China
| | - You Zhou
- Key Laboratory of Natural Pesticide and Chemical Biology, Ministry of Education, South China Agricultural University, Guangzhou, China
| | - Peiwen Zhang
- Key Laboratory of Natural Pesticide and Chemical Biology, Ministry of Education, South China Agricultural University, Guangzhou, China
| | - Benju Liu
- Key Laboratory of Natural Pesticide and Chemical Biology, Ministry of Education, South China Agricultural University, Guangzhou, China
| | - Qun Zheng
- Key Laboratory of Natural Pesticide and Chemical Biology, Ministry of Education, South China Agricultural University, Guangzhou, China
| | - Zhixiang Zhang
- Key Laboratory of Natural Pesticide and Chemical Biology, Ministry of Education, South China Agricultural University, Guangzhou, China
| |
Collapse
|
19
|
Talley MJ, Nardini D, Qin S, Prada CE, Ehrman LA, Waclaw RR. A role for sustained MAPK activity in the mouse ventral telencephalon. Dev Biol 2021; 476:137-147. [PMID: 33775695 DOI: 10.1016/j.ydbio.2021.03.019] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 03/14/2021] [Accepted: 03/21/2021] [Indexed: 11/28/2022]
Abstract
The MAPK pathway is a major growth signal that has been implicated during the development of progenitors, neurons, and glia in the embryonic brain. Here, we show that the MAPK pathway plays an important role in the generation of distinct cell types from progenitors in the ventral telencephalon. Our data reveal that phospho-p44/42 (called p-ERK1/2) and the ETS transcription factor Etv5, both downstream effectors in the MAPK pathway, show a regional bias in expression during ventral telencephalic development, with enriched expression in the dorsal region of the LGE and ventral region of the MGE at E13.5 and E15.5. Interestingly, expression of both factors becomes more uniform in ventricular zone (VZ) progenitors by E18.5. To gain insight into the role of MAPK activity during progenitor cell development, we used a cre inducible constitutively active MEK1 allele (RosaMEK1DD/+) in combination with a ventral telencephalon enriched cre (Gsx2e-cre) or a dorsal telencephalon enriched cre (Emx1cre/+). Sustained MEK/MAPK activity in the ventral telencephalon (Gsx2e-cre; RosaMEK1DD/+) expanded dorsal lateral ganglionic eminence (dLGE) enriched genes (Gsx2 and Sp8) and oligodendrocyte progenitor cell (OPC) markers (Olig2, Pdgfrα, and Sox10), and also reduced markers in the ventral (v) LGE domain (Isl1 and Foxp1). Activation of MEK/MAPK activity in the dorsal telencephalon (Emx1cre/+; RosaMEK1DD/+) did not initially activate the expression of dLGE or OPC genes at E15.5 but ectopic expression of Gsx2 and OPC markers were observed at E18.5. These results support the idea that MAPK activity as readout by p-ERK1/2 and Etv5 expression is enriched in distinct subdomains of ventral telencephalic progenitors during development. In addition, sustained activation of the MEK/MAPK pathway in the ventral or dorsal telencephalon influences dLGE and OPC identity from progenitors.
Collapse
Affiliation(s)
- Mary Jo Talley
- Graduate Program in Molecular and Developmental Biology, Cincinnati Children's Hospital Research Foundation, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| | - Diana Nardini
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229, USA
| | - Shenyue Qin
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229, USA
| | - Carlos E Prada
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229, USA
| | - Lisa A Ehrman
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229, USA
| | - Ronald R Waclaw
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229, USA; Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229, USA; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA.
| |
Collapse
|
20
|
Chen MJ, Ramesha S, Weinstock LD, Gao T, Ping L, Xiao H, Dammer EB, Duong DD, Levey AI, Lah JJ, Seyfried NT, Wood LB, Rangaraju S. Extracellular signal-regulated kinase regulates microglial immune responses in Alzheimer's disease. J Neurosci Res 2021; 99:1704-1721. [PMID: 33729626 DOI: 10.1002/jnr.24829] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 02/20/2021] [Accepted: 03/02/2021] [Indexed: 12/12/2022]
Abstract
The importance of mitogen-activated protein kinase (MAPK) pathway signaling in regulating microglia-mediated neuroinflammation in Alzheimer's disease (AD) remains unclear. We examined the role of MAPK signaling in microglia using a preclinical model of AD pathology and quantitative proteomics studies of postmortem human brains. In multiplex immunoassay analyses of MAPK phosphoproteins in acutely isolated microglia and brain tissue from 5xFAD mice, we found phosphorylated extracellular signal-regulated kinase (ERK) was the most strongly upregulated phosphoprotein within the MAPK pathway in acutely isolated microglia, but not whole-brain tissue from 5xFAD mice. The importance of ERK signaling in primary microglia cultures was next investigated using transcriptomic profiling and functional assays of amyloid-β and neuronal phagocytosis, which confirmed that ERK is a critical regulator of IFNγ-mediated pro-inflammatory activation of microglia, although it was also partly important for constitutive microglial functions. Phospho-ERK was an upstream regulator of disease-associated microglial gene expression (Trem2, Tyrobp), as well as several human AD risk genes (Bin1, Cd33, Trem2, Cnn2), indicative of the importance of microglial ERK signaling in AD pathology. Quantitative proteomic analyses of postmortem human brain showed that ERK1 and ERK2 were the only MAPK proteins with increased protein expression and positive associations with neuropathological grade. In a human brain phosphoproteomic study, we found evidence for increased flux through the ERK signaling pathway in AD. Overall, our analyses strongly suggest that ERK phosphorylation, particularly in microglia in mouse models, is a regulator of pro-inflammatory immune responses in AD pathogenesis.
Collapse
Affiliation(s)
- Michael J Chen
- Department of Neurology, Emory University, Atlanta, GA, USA
| | | | - Laura D Weinstock
- Parker H. Petit Institute for Bioengineering & Bioscience, Georgia Institute of Technology, Atlanta, GA, USA.,The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Tianwen Gao
- Department of Neurology, Emory University, Atlanta, GA, USA
| | - Lingyan Ping
- Department of Biochemistry, Emory University, Atlanta, GA, USA
| | - Hailian Xiao
- Department of Neurology, Emory University, Atlanta, GA, USA
| | - Eric B Dammer
- Department of Biochemistry, Emory University, Atlanta, GA, USA
| | - Duc D Duong
- Department of Biochemistry, Emory University, Atlanta, GA, USA
| | - Allan I Levey
- Department of Neurology, Emory University, Atlanta, GA, USA
| | - James J Lah
- Department of Neurology, Emory University, Atlanta, GA, USA
| | | | - Levi B Wood
- Parker H. Petit Institute for Bioengineering & Bioscience, Georgia Institute of Technology, Atlanta, GA, USA.,The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA.,George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | | |
Collapse
|
21
|
Holter MC, Hewitt LT, Nishimura KJ, Knowles SJ, Bjorklund GR, Shah S, Fry NR, Rees KP, Gupta TA, Daniels CW, Li G, Marsh S, Treiman DM, Olive MF, Anderson TR, Sanabria F, Snider WD, Newbern JM. Hyperactive MEK1 Signaling in Cortical GABAergic Neurons Promotes Embryonic Parvalbumin Neuron Loss and Defects in Behavioral Inhibition. Cereb Cortex 2021; 31:3064-3081. [PMID: 33570093 DOI: 10.1093/cercor/bhaa413] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 12/22/2020] [Accepted: 12/23/2020] [Indexed: 12/20/2022] Open
Abstract
Many developmental syndromes have been linked to genetic mutations that cause abnormal ERK/MAPK activity; however, the neuropathological effects of hyperactive signaling are not fully understood. Here, we examined whether hyperactivation of MEK1 modifies the development of GABAergic cortical interneurons (CINs), a heterogeneous population of inhibitory neurons necessary for cortical function. We show that GABAergic-neuron specific MEK1 hyperactivation in vivo leads to increased cleaved caspase-3 labeling in a subpopulation of immature neurons in the embryonic subpallial mantle zone. Adult mutants displayed a significant loss of parvalbumin (PV), but not somatostatin, expressing CINs and a reduction in perisomatic inhibitory synapses on excitatory neurons. Surviving mutant PV-CINs maintained a typical fast-spiking phenotype but showed signs of decreased intrinsic excitability that coincided with an increased risk of seizure-like phenotypes. In contrast to other mouse models of PV-CIN loss, we discovered a robust increase in the accumulation of perineuronal nets, an extracellular structure thought to restrict plasticity. Indeed, we found that mutants exhibited a significant impairment in the acquisition of behavioral response inhibition capacity. Overall, our data suggest PV-CIN development is particularly sensitive to hyperactive MEK1 signaling, which may underlie certain neurological deficits frequently observed in ERK/MAPK-linked syndromes.
Collapse
Affiliation(s)
- Michael C Holter
- School of Life Sciences, Arizona State University, Tempe, AZ 85287, USA
| | - Lauren T Hewitt
- School of Life Sciences, Arizona State University, Tempe, AZ 85287, USA.,Interdepartmental Neuroscience Graduate Program, University of Texas, Austin, TX 78712, USA
| | - Kenji J Nishimura
- School of Life Sciences, Arizona State University, Tempe, AZ 85287, USA.,Interdepartmental Neuroscience Graduate Program, University of Texas, Austin, TX 78712, USA
| | - Sara J Knowles
- School of Life Sciences, Arizona State University, Tempe, AZ 85287, USA
| | | | - Shiv Shah
- School of Life Sciences, Arizona State University, Tempe, AZ 85287, USA
| | - Noah R Fry
- School of Life Sciences, Arizona State University, Tempe, AZ 85287, USA
| | - Katherina P Rees
- School of Life Sciences, Arizona State University, Tempe, AZ 85287, USA
| | - Tanya A Gupta
- Department of Psychology, Arizona State University, Tempe, AZ 85287, USA
| | - Carter W Daniels
- Department of Psychology, Arizona State University, Tempe, AZ 85287, USA.,Department of Psychiatry, Columbia University, New York, NY 10032, USA
| | - Guohui Li
- College of Medicine, University of Arizona, Phoenix, AZ 85004, USA
| | - Steven Marsh
- Barrow Neurological Institute, Phoenix, AZ 85013, USA
| | | | | | - Trent R Anderson
- College of Medicine, University of Arizona, Phoenix, AZ 85004, USA
| | - Federico Sanabria
- Department of Psychology, Arizona State University, Tempe, AZ 85287, USA
| | - William D Snider
- University of North Carolina Neuroscience Center, The University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| | - Jason M Newbern
- School of Life Sciences, Arizona State University, Tempe, AZ 85287, USA
| |
Collapse
|
22
|
Melis C, Beauvais G, Muntean BS, Cirnaru MD, Otrimski G, Creus-Muncunill J, Martemyanov KA, Gonzalez-Alegre P, Ehrlich ME. Striatal Dopamine Induced ERK Phosphorylation Is Altered in Mouse Models of Monogenic Dystonia. Mov Disord 2021; 36:1147-1157. [PMID: 33458877 DOI: 10.1002/mds.28476] [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: 10/16/2020] [Revised: 12/02/2020] [Accepted: 12/04/2020] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Similar to some monogenic forms of dystonia, levodopa-induced dyskinesia is a hyperkinetic movement disorder with abnormal nigrostriatal dopaminergic neurotransmission. Molecularly, it is characterized by hyper-induction of phosphorylation of extracellular signal-related kinase in response to dopamine in medium spiny neurons of the direct pathway. OBJECTIVES The objective of this study was to determine if mouse models of monogenic dystonia exhibit molecular features of levodopa-induced dyskinesia. METHODS Western blotting and quantitative immunofluorescence was used to assay baseline and/or dopamine-induced levels of the phosphorylated kinase in the striatum in mouse models of DYT1, DYT6, and DYT25 expressing a reporter in dopamine D1 receptor-expressing projection neurons. Cyclic adenosine monophosphate (cAMP) immunoassay and adenylyl cyclase activity assays were also performed. RESULTS In DYT1 and DYT6 models, blocking dopamine reuptake with cocaine leads to enhanced extracellular signal-related kinase phosphorylation in dorsomedial striatal medium spiny neurons in the direct pathway, which is abolished by pretreatment with the N-methyl-d-aspartate antagonist MK-801. Phosphorylation is decreased in a model of DYT25. Levels of basal and stimulated cAMP and adenylyl cyclase activity were normal in the DYT1 and DYT6 mice and decreased in the DYT25 mice. Oxotremorine induced increased abnormal movements in the DYT1 knock-in mice. CONCLUSIONS The increased dopamine induction of extracellular signal-related kinase phosphorylation in 2 genetic types of dystonia, similar to what occurs in levodopa-induced dyskinesia, and its decrease in a third, suggests that abnormal signal transduction in response to dopamine in the postsynaptic nigrostriatal pathway might be a point of convergence for dystonia and other hyperkinetic movement disorders, potentially offering common therapeutic targets. © 2021 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Chiara Melis
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Genevieve Beauvais
- Raymond G. Perelman Center for Cellular and Molecular Therapy, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Brian S Muntean
- Department of Neuroscience, The Scripps Research Institute, Jupiter, Florida, USA
| | - Maria-Daniela Cirnaru
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Garrett Otrimski
- Raymond G. Perelman Center for Cellular and Molecular Therapy, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Jordi Creus-Muncunill
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Kirill A Martemyanov
- Department of Neuroscience, The Scripps Research Institute, Jupiter, Florida, USA
| | - Pedro Gonzalez-Alegre
- Raymond G. Perelman Center for Cellular and Molecular Therapy, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA.,Department of Neurology, The University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Michelle E Ehrlich
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, New York, USA.,Departments of Pediatrics and Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| |
Collapse
|
23
|
Cocaine-Dependent Acquisition of Locomotor Sensitization and Conditioned Place Preference Requires D1 Dopaminergic Signaling through a Cyclic AMP, NCS-Rapgef2, ERK, and Egr-1/Zif268 Pathway. J Neurosci 2020; 41:711-725. [PMID: 33268547 DOI: 10.1523/jneurosci.1497-20.2020] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 11/08/2020] [Accepted: 11/15/2020] [Indexed: 11/21/2022] Open
Abstract
Elucidation of the mechanism of dopamine signaling to ERK that underlies plasticity in dopamine D1 receptor-expressing neurons leading to acquired cocaine preference is incomplete. NCS-Rapgef2 is a novel cAMP effector, expressed in neuronal and endocrine cells in adult mammals, that is required for D1 dopamine receptor-dependent ERK phosphorylation in mouse brain. In this report, we studied the effects of abrogating NCS-Rapgef2 expression on cAMP-dependent ERK→Egr-1/Zif268 signaling in cultured neuroendocrine cells; in D1 medium spiny neurons of NAc slices; and in either male or female mouse brain in a region-specific manner. NCS-Rapgef2 gene deletion in the NAc in adult mice, using adeno-associated virus-mediated expression of cre recombinase, eliminated cocaine-induced ERK phosphorylation and Egr-1/Zif268 upregulation in D1-medium spiny neurons and cocaine-induced behaviors, including locomotor sensitization and conditioned place preference. Abrogation of NCS-Rapgef2 gene expression in mPFC and BLA, by crossing mice bearing a floxed Rapgef2 allele with a cre mouse line driven by calcium/calmodulin-dependent kinase IIα promoter also eliminated cocaine-induced phospho-ERK activation and Egr-1/Zif268 induction, but without effect on the cocaine-induced behaviors. Our results indicate that NCS-Rapgef2 signaling to ERK in dopamine D1 receptor-expressing neurons in the NAc, but not in corticolimbic areas, contributes to cocaine-induced locomotor sensitization and conditioned place preference. Ablation of cocaine-dependent ERK activation by elimination of NCS-Rapgef2 occurred with no effect on phosphorylation of CREB in D1 dopaminoceptive neurons of NAc. This study reveals a new cAMP-dependent signaling pathway for cocaine-induced behavioral adaptations, mediated through NCS-Rapgef2/phospho-ERK activation, independently of PKA/CREB signaling.SIGNIFICANCE STATEMENT ERK phosphorylation in dopamine D1 receptor-expressing neurons exerts a pivotal role in psychostimulant-induced neuronal gene regulation and behavioral adaptation, including locomotor sensitization and drug preference in rodents. In this study, we examined the role of dopamine signaling through the D1 receptor via a novel pathway initiated through the cAMP-activated guanine nucleotide exchange factor NCS-Rapgef2 in mice. NCS-Rapgef2 in the NAc is required for activation of ERK and Egr-1/Zif268 in D1 dopaminoceptive neurons after acute cocaine administration, and subsequent enhanced locomotor response and drug seeking behavior after repeated cocaine administration. This novel component in dopamine signaling provides a potential new target for intervention in psychostimulant-shaped behaviors, and new understanding of how D1-medium spiny neurons encode the experience of psychomotor stimulant exposure.
Collapse
|
24
|
Song N, Du J, Gao Y, Yang S. Epitranscriptome of the ventral tegmental area in a deep brain-stimulated chronic unpredictable mild stress mouse model. Transl Neurosci 2020; 11:402-418. [PMID: 33343932 PMCID: PMC7724003 DOI: 10.1515/tnsci-2020-0146] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 09/08/2020] [Accepted: 10/02/2020] [Indexed: 12/20/2022] Open
Abstract
Deep brain stimulation (DBS) applied to the nucleus accumbens (NAc) alleviates the depressive symptoms of major depressive disorders. We investigated the mechanism of this effect by assessing gene expression and RNA methylation changes in the ventral tegmental area (VTA) following NAc-DBS in a chronic unpredictable mild stress (CUMS) mouse model of depression. Gene expression and N 6-methyladenosine (m6A) levels in the VTA were measured in mice subjected to CUMS and then DBS, and transcriptome-wide m6A changes were profiled using immunoprecipitated methylated RNAs with microarrays, prior to gene ontology analysis. The expression levels of genes linked to neurotransmitter receptors, transporters, transcription factors, neuronal activities, synaptic functions, and mitogen-activated protein kinase and dopamine signaling were upregulated in the VTA upon NAc-DBS. Furthermore, m6A modifications included both hypermethylation and hypomethylation, and changes were positively correlated with the upregulation of some genes. Moreover, the effects of CUMS on gene expression and m6A-mRNA modification were reversed by DBS for some genes. Interestingly, while the expression of certain genes was not changed by DBS, long-term stimulation did alter their m6A modifications. NAc-DBS-induced modifications are correlated largely with upregulation but sometimes downregulation of genes in CUMS mice. Our findings improve the current understanding of the molecular mechanisms underlying DBS effects on depression.
Collapse
Affiliation(s)
- Nan Song
- Center of Military Brain Science, Institute of Military Cognition and Brain Sciences, Academy of Military Medical Sciences (AMMS), The Academy of Military Sciences, No. 27 Taiping Road, Haidian District, Beijing, China, 100850
| | - Jun Du
- Center of Military Brain Science, Institute of Military Cognition and Brain Sciences, Academy of Military Medical Sciences (AMMS), The Academy of Military Sciences, No. 27 Taiping Road, Haidian District, Beijing, China, 100850
| | - Yan Gao
- Center of Military Brain Science, Institute of Military Cognition and Brain Sciences, Academy of Military Medical Sciences (AMMS), The Academy of Military Sciences, No. 27 Taiping Road, Haidian District, Beijing, China, 100850
| | - Shenglian Yang
- Center of Military Brain Science, Institute of Military Cognition and Brain Sciences, Academy of Military Medical Sciences (AMMS), The Academy of Military Sciences, No. 27 Taiping Road, Haidian District, Beijing, China, 100850
| |
Collapse
|
25
|
Zhang J, Zhang T, Zhang W, Zou C, Zhang Q, Ma X, Zhu Y. Circular RNA-DENND4C in H9c2 cells relieves OGD/R-induced injury by down regulation of microRNA-320. Cell Cycle 2020; 19:3074-3085. [PMID: 33090893 DOI: 10.1080/15384101.2020.1831253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
Abstract
Ischemic heart disease (IHD) is one of the most deadly diseases worldwide. To detect the regulatory mechanism, the circular RNA (circRNA)-differentially expressed in normal cells and neoplasia domain containing 4 C (DENND4C) was explored in the H9c2 cells. The circRNA-DENND4C overexpressing plasmid, si-circRNA-DENND4C and miR-320 mimic were transfected into the H9c2 cells and treated with OGD/R stimulation. We took CCK-8 method, Annexin V-FITC/PI-flow cytometer to search for viability and apoptotic ability. With the help of qRT-PCR and western blot, the expression of circRNA-DENND4C and miR-320, as well as the Bax, Cleaved PARP/caspase 3 and signal proteins were separately determined. Regulation of circRNA-DENND4C and miR-320 was confirmed by dual-luciferase reporter assay. OGD/R induced suppression of cell viability, but enhancement of apoptosis and block of ERK and mTOR pathways. Moreover, circRNA-DENND4C was up-regulated after OGD/R stimulation and augmented OGD/R-stimulated damage while circRNA-DENND4C silencing displayed opposite influences. miR-320 was negatively controlled and targeted by the circRNA-DENND4C.The overexpressed miR-320 impeded the effects of circRNA-DENND4C. Besides, circRNA-DENND4C relieved the suppression of ERK and mTOR pathways caused by OGD/R stimulation, and all promoting impacts of circRNA-DENND4C were reversed by the miR-320 mimic. Overexpressed circRNA-DENND4C in H9c2 cells attenuated OGD/R-induced injuries by the down-regulation of miR-320 through the ERK and mTOR activation.
Collapse
Affiliation(s)
- Jun Zhang
- Department of Cardiac Surgery, Shandong Provincial Hospital Affiliated to Shandong University , Jinan, China
| | - Tao Zhang
- Department of Cardiac Surgery, Shandong Provincial Hospital Affiliated to Shandong University , Jinan, China
| | - Wenlong Zhang
- Department of Cardiac Surgery, Shandong Provincial Hospital Affiliated to Shandong University , Jinan, China
| | - Chengwei Zou
- Department of Cardiac Surgery, Shandong Provincial Hospital Affiliated to Shandong University , Jinan, China
| | - Qian Zhang
- Department of Cardiac Surgery, Shandong Provincial Hospital Affiliated to Shandong University , Jinan, China
| | - Xiaochun Ma
- Department of Cardiac Surgery, Shandong Provincial Hospital Affiliated to Shandong University , Jinan, China
| | - Yanhui Zhu
- Department of Cardiac Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University , Jinan, China
| |
Collapse
|
26
|
Brakatselos C, Delis F, Asprogerakas MZ, Lekkas P, Tseti I, Tzimas PS, Petrakis EA, Halabalaki M, Skaltsounis LA, Antoniou K. Cannabidiol Modulates the Motor Profile and NMDA Receptor-related Alterations Induced by Ketamine. Neuroscience 2020; 454:105-115. [PMID: 32950556 DOI: 10.1016/j.neuroscience.2020.09.029] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 09/07/2020] [Accepted: 09/08/2020] [Indexed: 02/01/2023]
Abstract
Cannabidiol (CBD) is a non-addictive ingredient of cannabis with antipsychotic potential, while ketamine (KET), an uncompetitive NMDA receptor inhibitor, has been extensively used as a psychotomimetic. Only few studies have focused on the role of CBD on the KET-induced motor profile, while no study has investigated the impact of CBD on KET-induced alterations in NMDA receptor subunit expression and ERK phosphorylation state, in brain regions related to the neurobiology and treatment of schizophrenia. Therefore, the aim of the present study is to evaluate the role of CBD on KET-induced motor response and relevant glutamatergic signaling in the prefrontal cortex, the nucleus accumbens, the dorsal and ventral hippocampus. The present study demonstrated that CBD pre-administration did not reverse KET-induced short-lasting hyperactivity, but it prolonged it over time. CBD alone decreased motor activity at the highest dose tested (30 mg/kg) while KET increased motor activity at the higher doses (30, 60 mg/kg). Moreover, KET induced regionally-dependent alterations in NR1 and NR2B expression and ERK phosphorylation that were reversed by CBD pre-administration. Interestingly, in the nucleus accumbens KET per se reduced NR2B and p-ERK levels, while the CBD/KET combination increased NR2B and p-ERK levels, as compared to control. This study is the first to show that CBD prolongs KET-induced motor stimulation and restores KET-induced effects on glutamatergic signaling and neuroplasticity-related markers. These findings contribute to the understanding of CBD effects on the behavioral and neurobiological profiles of psychotogenic KET.
Collapse
Affiliation(s)
- Charalampos Brakatselos
- Department of Pharmacology, Faculty of Medicine, School of Health Sciences, University of Ioannina, 45110 Ioannina, Greece
| | - Foteini Delis
- Department of Pharmacology, Faculty of Medicine, School of Health Sciences, University of Ioannina, 45110 Ioannina, Greece
| | - Michail-Zois Asprogerakas
- Department of Pharmacology, Faculty of Medicine, School of Health Sciences, University of Ioannina, 45110 Ioannina, Greece
| | - Panagiotis Lekkas
- Department of Pharmacology, Faculty of Medicine, School of Health Sciences, University of Ioannina, 45110 Ioannina, Greece
| | - Ioulia Tseti
- INTERMED: Pharmaceutical Laboratories Ioulia and Eirini Tseti, Kaliftaki 27, 14564 Athens, Greece
| | - Petros S Tzimas
- Department of Pharmacognosy and Natural Product Chemistry, Faculty of Pharmacy, National and Kapodistrian University of Athens, 15771 Athens, Greece
| | - Eleftherios A Petrakis
- Department of Pharmacognosy and Natural Product Chemistry, Faculty of Pharmacy, National and Kapodistrian University of Athens, 15771 Athens, Greece
| | - Maria Halabalaki
- Department of Pharmacognosy and Natural Product Chemistry, Faculty of Pharmacy, National and Kapodistrian University of Athens, 15771 Athens, Greece
| | - Leandros A Skaltsounis
- Department of Pharmacognosy and Natural Product Chemistry, Faculty of Pharmacy, National and Kapodistrian University of Athens, 15771 Athens, Greece
| | - Katerina Antoniou
- Department of Pharmacology, Faculty of Medicine, School of Health Sciences, University of Ioannina, 45110 Ioannina, Greece.
| |
Collapse
|
27
|
Bilge SS, Günaydin C, Önger ME, Bozkurt A, Avci B. Neuroprotective action of agmatine in rotenone-induced model of Parkinson's disease: Role of BDNF/cREB and ERK pathway. Behav Brain Res 2020; 392:112692. [PMID: 32479847 DOI: 10.1016/j.bbr.2020.112692] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Revised: 04/14/2020] [Accepted: 05/03/2020] [Indexed: 01/10/2023]
Abstract
Numerous studies have investigated the role of agmatine in the central nervous system and indicated neuroprotective properties. In addition to its potent antioxidant effects, agmatine is an endogenous neuromodulator and has wide spectrum molecular actions on different receptor subtypes (NMDA, Imidazoline 1-2, alpha-2 adrenoreceptor, 5-HT2a, 5-HT3) and cellular signaling pathways (MAPK, PKA, NO, BDNF). Although the neuroprotective effects of agmatine demonstrated in experimental Parkinson's disease model, the effects of agmatine with the aspect of neuroplasticity and possible signaling mechanisms behind agmatine actions have not been investigated. Herein, in this study, we investigated the role of the of agmatine on rotenone-induced Parkinson's disease model. Agmatine at the dose of 100 mg/kg i.p., was mitigated oxidative damage and alleviated motor impairments which were the results of the rotenone insult. Additionally, agmatine decreased neuronal loss, tyrosine hydroxylase immunoreactivity and increased cREB, BDNF and ERK1/2 expression in the striatum, which are crucial neuroplasticity elements of striatal integrity. Taken together, the present study expands the knowledge of molecular mechanisms behind neuroprotective actions of agmatine in Parkinson's disease, and as far as we have known, this is the first study to delineate agmatine treated activation of cellular pathways which are important elements in neuronal cell survival.
Collapse
Affiliation(s)
- S Sırrı Bilge
- Ondokuz Mayıs University, School of Medicine, Department of Pharmacology, Samsun, Turkey.
| | - Caner Günaydin
- Ondokuz Mayıs University, School of Medicine, Department of Pharmacology, Samsun, Turkey.
| | - M Emin Önger
- Ondokuz Mayıs University, School of Medicine, Department of Histology and Embryology, Samsun, Turkey.
| | - Ayhan Bozkurt
- Ondokuz Mayıs University, School of Medicine, Department of Physiology, Samsun, Turkey.
| | - Bahattin Avci
- Ondokuz Mayıs University, School of Medicine, Department of Biochemistry, Samsun, Turkey.
| |
Collapse
|
28
|
Liu Z, Wu C, Zou X, Shen W, Yang J, Zhang X, Hu X, Wang H, Liao Y, Jing T. Exosomes derived from mesenchymal stem cells inhibit neointimal hyperplasia by activating the Erk1/2 signalling pathway in rats. Stem Cell Res Ther 2020; 11:220. [PMID: 32513275 PMCID: PMC7278178 DOI: 10.1186/s13287-020-01676-w] [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: 12/06/2019] [Revised: 03/17/2020] [Accepted: 04/13/2020] [Indexed: 01/29/2023] Open
Abstract
Background Restenosis is a serious problem in patients who have undergone percutaneous transluminal angioplasty. Endothelial injury resulting from surgery can lead to endothelial dysfunction and neointimal formation by inducing aberrant proliferation and migration of vascular smooth muscle cells. Exosomes secreted by mesenchymal stem cells have been a hot topic in cardioprotective research. However, to date, exosomes derived from mesenchymal stem cells (MSC-Exo) have rarely been reported in association with restenosis after artery injury. The aim of this study was to investigate whether MSC-Exo inhibit neointimal hyperplasia in a rat model of carotid artery balloon-induced injury and, if so, to explore the underlying mechanisms. Methods Characterization of MSC-Exo immunophenotypes was performed by electron microscopy, nanoparticle tracking analysis and western blot assays. To investigate whether MSC-Exo inhibited neointimal hyperplasia, rats were intravenously injected with normal saline or MSC-Exo after carotid artery balloon-induced injury. Haematoxylin-eosin staining was performed to examine the intimal and media areas. Evans blue dye staining was performed to examine re-endothelialization. Moreover, immunohistochemistry and immunofluorescence were performed to examine the expression of CD31, vWF and α-SMA. To further investigate the involvement of MSC-Exo-induced re-endothelialization, the underlying mechanisms were studied by cell counting kit-8, cell scratch, immunofluorescence and western blot assays. Results Our data showed that MSC-Exo were ingested by endothelial cells and that systemic injection of MSC-Exo suppressed neointimal hyperplasia after artery injury. The Evans blue staining results showed that MSC-Exo could accelerate re-endothelialization compared to the saline group. The immunofluorescence and immunohistochemistry results showed that MSC-Exo upregulated the expression of CD31 and vWF but downregulated the expression of α-SMA. Furthermore, MSC-Exo mechanistically facilitated proliferation and migration by activating the Erk1/2 signalling pathway. The western blot results showed that MSC-Exo upregulated the expression of PCNA, Cyclin D1, Vimentin, MMP2 and MMP9 compared to that in the control group. Interestingly, an Erk1/2 inhibitor reversed the expression of the above proteins. Conclusion Our data suggest that MSC-Exo can inhibit neointimal hyperplasia after carotid artery injury by accelerating re-endothelialization, which is accompanied by activation of the Erk1/2 signalling pathway. Importantly, our study provides a novel cell-free approach for the treatment of restenosis diseases after intervention.
Collapse
Affiliation(s)
- Zhihui Liu
- Department of Cardiology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, 400038, China.,State Key Laboratory of Silkworm Genome Biology, The Institute of Sericulture and Systems Biology, Southwest University, Chongqing, China
| | - Chao Wu
- State Key Laboratory of Silkworm Genome Biology, The Institute of Sericulture and Systems Biology, Southwest University, Chongqing, China.,Department of Thoracic Surgery, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Xinliang Zou
- Department of Cardiology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, 400038, China
| | - Weiming Shen
- Laboratory of Integrative Medicine, School of Basic Medical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jiacai Yang
- The Institute of Burn Research, South-West Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Xiaorong Zhang
- The Institute of Burn Research, South-West Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Xiaohong Hu
- The Institute of Burn Research, South-West Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Haidong Wang
- Department of Thoracic Surgery, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Yi Liao
- Department of Thoracic Surgery, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Tao Jing
- Department of Cardiology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, 400038, China.
| |
Collapse
|
29
|
Malvaez M. Neural substrates of habit. J Neurosci Res 2020; 98:986-997. [PMID: 31693205 PMCID: PMC7183880 DOI: 10.1002/jnr.24552] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2019] [Revised: 09/27/2019] [Accepted: 10/22/2019] [Indexed: 12/11/2022]
Abstract
Active reward pursuit is supported by the balance between the cognitive and habitual control of behavior. The cognitive, goal-directed strategy relies on the prospective evaluation of anticipated consequences, which allows behavior to readily adapt when circumstances change. Repetition of successful actions promotes less cognitively taxing habits, in which behavior is automatically executed without prospective consideration. Disruption in either of these behavioral regulatory systems contributes to the symptoms that underlie many psychiatric disorders. Here, I review recently identified neural substrates, at multiple neural levels, that contribute to habits and outline gaps in knowledge that must be addressed to fully understand the neural mechanisms of behavioral control.
Collapse
|
30
|
Shi P, Fan F, Chen H, Xu Z, Cheng S, Lu W, Du M. A bovine lactoferrin–derived peptide induced osteogenesis via regulation of osteoblast proliferation and differentiation. J Dairy Sci 2020; 103:3950-3960. [DOI: 10.3168/jds.2019-17425] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Accepted: 01/13/2020] [Indexed: 01/24/2023]
|
31
|
Gait Deficits and Loss of Striatal Tyrosine Hydroxlase/Trk-B are Restored Following 7,8-Dihydroxyflavone Treatment in a Progressive MPTP Mouse Model of Parkinson’s Disease. Neuroscience 2020; 433:53-71. [DOI: 10.1016/j.neuroscience.2020.02.046] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Revised: 02/26/2020] [Accepted: 02/27/2020] [Indexed: 12/13/2022]
|
32
|
Wu J, Meng X, Jia Y, Chai J, Wang J, Xue X, Dang T. Long non-coding RNA HNF1A-AS1 upregulates OTX1 to enhance angiogenesis in colon cancer via the binding of transcription factor PBX3. Exp Cell Res 2020; 393:112025. [PMID: 32325080 DOI: 10.1016/j.yexcr.2020.112025] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Revised: 03/25/2020] [Accepted: 04/17/2020] [Indexed: 12/21/2022]
Abstract
Colon cancer shows characteristics of metastasis, which is associated with angiogenesis. Increasing evidence highlights long non-coding RNAs (lncRNAs) as important participants in angiogenesis of cancers, including colon cancer. Hence, this study investigated the role of HNF1A-AS1 in angiogenesis of colon cancer. RT-qPCR and Western blot analysis were applied to detect HNF1A-AS1 and OTX1 expression in colon cancer tissues and cell lines. Then the interactions among HNF1A-AS1, PBX3, OTX1 and ERK/MAPK pathway were evaluated with RNA pull-down, RIP, ChIP and dual-luciferase reporter gene assays. Next, HCT116 and SW620 cells were treated with si-HNF1A-AS1 and/or oe-OTX1 plasmids to assess the effects of HNF1A-AS1 and OTX1 on angiogenesis, which was further evaluated in nude mice injected with SW620 cells transfected with sh-HNF1A-AS1 or sh-OTX1 lentivirus. HNF1A-AS1 and OTX1 were highly expressed in colon cancer. Silencing of HNF1A-AS1 inhibited angiogenesis of colon cancer in vivo and in vitro. HNF1A-AS1 increased the OTX1 expression by binding to transcription factor PBX3 to promote angiogenesis in colon cancer. Further, HNF1A-AS1 upregulated OTX1 to activate the ERK/MAPK pathway. Altogether, our findings identified HNF1A-AS1 as a tumor-promoting RNA in colon cancer, which could serve as a potential therapeutic target for colon cancer treatment.
Collapse
Affiliation(s)
- Jinbao Wu
- Inner Mongolia Institute of Digestive Diseases, The Second Affiliated Hospital of Baotou Medical College, Inner Mongolia University of Science and Technology, Baotou, 014030, PR China.
| | - Xianmei Meng
- Inner Mongolia Institute of Digestive Diseases, The Second Affiliated Hospital of Baotou Medical College, Inner Mongolia University of Science and Technology, Baotou, 014030, PR China.
| | - Yanbin Jia
- Inner Mongolia Institute of Digestive Diseases, The Second Affiliated Hospital of Baotou Medical College, Inner Mongolia University of Science and Technology, Baotou, 014030, PR China; Nursing College of Baotou Medical College, Inner Mongolia University of Science and Technology, Baotou, 014030, PR China.
| | - Jianyuan Chai
- Inner Mongolia Institute of Digestive Diseases, The Second Affiliated Hospital of Baotou Medical College, Inner Mongolia University of Science and Technology, Baotou, 014030, PR China.
| | - Jing Wang
- Inner Mongolia Institute of Digestive Diseases, The Second Affiliated Hospital of Baotou Medical College, Inner Mongolia University of Science and Technology, Baotou, 014030, PR China.
| | - Xiaohui Xue
- Inner Mongolia Institute of Digestive Diseases, The Second Affiliated Hospital of Baotou Medical College, Inner Mongolia University of Science and Technology, Baotou, 014030, PR China.
| | - Tong Dang
- Inner Mongolia Institute of Digestive Diseases, The Second Affiliated Hospital of Baotou Medical College, Inner Mongolia University of Science and Technology, Baotou, 014030, PR China.
| |
Collapse
|
33
|
Wu QL, Cheng YQ, Liu AJ, Zhang WD. Formononetin recovered injured nerve functions by enhancing synaptic plasticity in ischemic stroke rats. Biochem Biophys Res Commun 2020; 525:S0006-291X(20)30281-3. [PMID: 32081422 DOI: 10.1016/j.bbrc.2020.02.015] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Accepted: 02/05/2020] [Indexed: 11/16/2022]
Abstract
BACKGROUND AND OBJECTIVES Formononetin has protective effect against ischemic stroke. It's unclear whether it can restore the nerve functions after stroke. METHODS SD rats were subjected with middle cerebral artery occlusion (MCAO), and divided into sham, model and formononetin (30 mg/kg) groups. Neurobehavioral tests (modified Neurological Severity Score [mNSS] and rotarod) were performed before and at 1, 3, 7 and 14 days after MCAO. Then, the rats were sacrificed and the brain sections were processed for neuronal differentiation and synaptic plasticity. RESULTS Compared with the sham group, the scores of mNSS were significantly increased, and the residence time on the rotating drum was significantly decreased in the MCAO rats. Compared with the model group, the scores of mNSS were significantly decreased, and the residence time on the rotating drum was increased in the formononetin (30 mg/kg) group. Formononetin significantly increased the number of neuronal dendritic spines and the expression of β III-tubulin, GAP-43, NGF, BDNF, p-Trk A, p-Trk B, p-AKT and p-ERK 1/2. CONCLUSIONS Formononetin recovered injured nerve functions after ischemic stroke. PI3K/AKT/ERK pathway might involve in the beneficial effect of formononetin on the neuronal differentiation and synaptic plasticity.
Collapse
Affiliation(s)
- Qiu-Ling Wu
- School of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, China; Department of Pharmacology, School of Pharmacy, Second Military Medical University, Shanghai, 200433, China
| | - Yan-Qiong Cheng
- Department of Pharmacology, School of Pharmacy, Second Military Medical University, Shanghai, 200433, China; Department of Pharmacy Research, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200437, China
| | - Ai-Jun Liu
- Department of Pharmacology, School of Pharmacy, Second Military Medical University, Shanghai, 200433, China; Department of Pharmacy Research, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200437, China.
| | - Wei-Dong Zhang
- School of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, China; Department of Phytochemistry, School of Pharmacy, Second Military Medical University, Shanghai, 200433, China.
| |
Collapse
|
34
|
Sutton LP, Muntean BS, Ostrovskaya O, Zucca S, Dao M, Orlandi C, Song C, Xie K, Martemyanov KA. NF1-cAMP signaling dissociates cell type-specific contributions of striatal medium spiny neurons to reward valuation and motor control. PLoS Biol 2019; 17:e3000477. [PMID: 31600280 PMCID: PMC6805008 DOI: 10.1371/journal.pbio.3000477] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Revised: 10/22/2019] [Accepted: 09/17/2019] [Indexed: 12/11/2022] Open
Abstract
The striatum plays a fundamental role in motor learning and reward-related behaviors that are synergistically shaped by populations of D1 dopamine receptor (D1R)- and D2 dopamine receptor (D2R)-expressing medium spiny neurons (MSNs). How various neurotransmitter inputs converging on common intracellular pathways are parsed out to regulate distinct behavioral outcomes in a neuron-specific manner is poorly understood. Here, we reveal that distinct contributions of D1R-MSNs and D2R-MSNs towards reward and motor behaviors are delineated by the multifaceted signaling protein neurofibromin 1 (NF1). Using genetic mouse models, we show that NF1 in D1R-MSN modulates opioid reward, whereas loss of NF1 in D2R-MSNs delays motor learning by impeding the formation and consolidation of repetitive motor sequences. We found that motor learning deficits upon NF1 loss were associated with the disruption in dopamine signaling to cAMP in D2R-MSN. Restoration of cAMP levels pharmacologically or chemogenetically rescued the motor learning deficits seen upon NF1 loss in D2R-MSN. Our findings illustrate that multiplex signaling capabilities of MSNs are deployed at the level of intracellular pathways to achieve cell-specific control over behavioral outcomes. A mouse genetic study reveals that the multifaceted signaling protein neurofibromin (known for its role in the human genetic disease neurofibromatosis type 1) plays a key role in differential routing of motor and reward signals in populations of striatal medium spiny neurons.
Collapse
Affiliation(s)
- Laurie P. Sutton
- Department of Neuroscience, The Scripps Research Institute, Jupiter, Florida, United States of America
| | - Brian S. Muntean
- Department of Neuroscience, The Scripps Research Institute, Jupiter, Florida, United States of America
| | - Olga Ostrovskaya
- Department of Neuroscience, The Scripps Research Institute, Jupiter, Florida, United States of America
| | - Stefano Zucca
- Department of Neuroscience, The Scripps Research Institute, Jupiter, Florida, United States of America
| | - Maria Dao
- Department of Neuroscience, The Scripps Research Institute, Jupiter, Florida, United States of America
| | - Cesare Orlandi
- Department of Neuroscience, The Scripps Research Institute, Jupiter, Florida, United States of America
| | - Chenghui Song
- Department of Neuroscience, The Scripps Research Institute, Jupiter, Florida, United States of America
| | - Keqiang Xie
- Department of Neuroscience, The Scripps Research Institute, Jupiter, Florida, United States of America
| | - Kirill A. Martemyanov
- Department of Neuroscience, The Scripps Research Institute, Jupiter, Florida, United States of America
- * E-mail:
| |
Collapse
|
35
|
Gangarossa G, Castell L, Castro L, Tarot P, Veyrunes F, Vincent P, Bertaso F, Valjent E. Contrasting patterns of ERK activation in the tail of the striatum in response to aversive and rewarding signals. J Neurochem 2019; 151:204-226. [PMID: 31245856 DOI: 10.1111/jnc.14804] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Revised: 05/13/2019] [Accepted: 06/19/2019] [Indexed: 01/08/2023]
Abstract
The caudal part of the striatum, also named the tail of the striatum (TS), defines a fourth striatal domain. Determining whether rewarding, aversive and salient stimuli regulate the activity of striatal spiny projections neurons (SPNs) of the TS is therefore of paramount importance to understand its functions, which remain largely elusive. Taking advantage of genetically encoded biosensors (A-kinase activity reporter 3) to record protein kinase A signals and by analyzing the distribution of dopamine D1R- and D2R-SPNs in the TS, we characterized three subterritories: a D2R/A2aR-lacking, a D1R/D2R-intermingled and a D1R/D2R-SPNs-enriched area (corresponding to the amygdalostriatal transition). In addition, we provide evidence that the distribution of D1R- and D2R-SPNs in the TS is evolutionarily conserved (mouse, rat, gerbil). The in vivo analysis of extracellular signal-regulated kinase (ERK) phosphorylation in these TS subterritories in response to distinct appetitive, aversive and pharmacological stimuli revealed that SPNs of the TS are not recruited by stimuli triggering innate aversive responses, fasting, satiety, or palatable signals whereas a reduction in ERK phosphorylation occurred following learned avoidance. In contrast, D1R-SPNs of the intermingled and D2R/A2aR-lacking areas were strongly activated by both D1R agonists and psychostimulant drugs (d-amphetamine, cocaine, 3,4-methyl enedioxy methamphetamine, or methylphenidate), but not by hallucinogens. Finally, a similar pattern of ERK activation was observed by blocking selectively dopamine reuptake. Together, our results reveal that the caudal TS might participate in the processing of specific reward signals and discrete aversive stimuli. Cover Image for this issue: doi: 10.1111/jnc.14526. Open Science: This manuscript was awarded with the Open Materials Badge For more information see: https://cos.io/our-services/open-science-badges/.
Collapse
Affiliation(s)
- Giuseppe Gangarossa
- IGF, CNRS, INSERM, University of Montpellier, Montpellier, France.,Université de Paris, BFA, UMR 8251, CNRS, Paris, France
| | - Laia Castell
- IGF, CNRS, INSERM, University of Montpellier, Montpellier, France
| | - Liliana Castro
- Sorbonne Université, CNRS, Biological Adaptation and Ageing, Paris, France
| | - Pauline Tarot
- IGF, CNRS, INSERM, University of Montpellier, Montpellier, France
| | - Frederic Veyrunes
- Institut des Sciences de l'Evolution de Montpellier, ISEM, Université de Montpellier, CNRS, EPHE, IRD, Montpellier, France
| | - Pierre Vincent
- Sorbonne Université, CNRS, Biological Adaptation and Ageing, Paris, France
| | - Federica Bertaso
- IGF, CNRS, INSERM, University of Montpellier, Montpellier, France
| | - Emmanuel Valjent
- IGF, CNRS, INSERM, University of Montpellier, Montpellier, France
| |
Collapse
|
36
|
Mariani LL, Longueville S, Girault JA, Hervé D, Gervasi N. Differential enhancement of ERK, PKA and Ca 2+ signaling in direct and indirect striatal neurons of Parkinsonian mice. Neurobiol Dis 2019; 130:104506. [PMID: 31220556 DOI: 10.1016/j.nbd.2019.104506] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 06/06/2019] [Accepted: 06/13/2019] [Indexed: 12/29/2022] Open
Abstract
Parkinson's disease (PD) is characterized by severe locomotor deficits due to the disappearance of dopamine (DA) from the dorsal striatum. The development of PD symptoms and treatment-related complications such as dyskinesia have been proposed to result from complex alterations in intracellular signaling in both direct and indirect pathway striatal projection neurons (dSPNs and iSPNs, respectively) following loss of DA afferents. To identify cell-specific and dynamical modifications of signaling pathways associated with PD, we used a hemiparkinsonian mouse model with 6-hydroxydopamine (6-OHDA) lesion combined with two-photon fluorescence biosensors imaging in adult corticostriatal slices. After DA lesion, extracellular signal-regulated kinase (ERK) activation was increased in response to DA D1 receptor (D1R) or α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) stimulation. The cAMP-dependent protein kinase (PKA) pathway contributing to ERK activation displayed supersensitive responses to D1R stimulation after 6-OHDA lesion. This cAMP/PKA supersensitivity was specific of D1R-responding SPNs and resulted from Gαolf upregulation and deficient phosphodiesterase activity. In lesioned striatum, the number of D1R-SPNs with spontaneous Ca2+ transients augmented while Ca2+ response to AMPA receptor stimulation specifically increased in iSPNs. Our work reveals distinct cell type-specific signaling alterations in the striatum after DA denervation. It suggests that over-activation of ERK pathway, observed in PD striatum, known to contribute to dyskinesia, may be linked to the combined dysregulation of DA and glutamate signaling pathways in the two populations of SPNs. These findings bring new insights into the implication of these respective neuronal populations in PD motor symptoms and the occurrence of PD treatment complications.
Collapse
Affiliation(s)
- Louise-Laure Mariani
- Inserm UMR-S 1270, Paris, France; Sorbonne Université, Science and Engineering Faculty, Paris, France; Institut du Fer à Moulin, Paris, France
| | - Sophie Longueville
- Inserm UMR-S 1270, Paris, France; Sorbonne Université, Science and Engineering Faculty, Paris, France; Institut du Fer à Moulin, Paris, France
| | - Jean-Antoine Girault
- Inserm UMR-S 1270, Paris, France; Sorbonne Université, Science and Engineering Faculty, Paris, France; Institut du Fer à Moulin, Paris, France
| | - Denis Hervé
- Inserm UMR-S 1270, Paris, France; Sorbonne Université, Science and Engineering Faculty, Paris, France; Institut du Fer à Moulin, Paris, France.
| | - Nicolas Gervasi
- Inserm UMR-S 1270, Paris, France; Sorbonne Université, Science and Engineering Faculty, Paris, France; Institut du Fer à Moulin, Paris, France.
| |
Collapse
|
37
|
Nair PS, Kuusi T, Ahvenainen M, Philips AK, Järvelä I. Music-performance regulates microRNAs in professional musicians. PeerJ 2019; 7:e6660. [PMID: 30956902 PMCID: PMC6442922 DOI: 10.7717/peerj.6660] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Accepted: 02/19/2019] [Indexed: 12/12/2022] Open
Abstract
Musical training and performance require precise integration of multisensory and motor centres of the human brain and can be regarded as an epigenetic modifier of brain functions. Numerous studies have identified structural and functional differences between the brains of musicians and non-musicians and superior cognitive functions in musicians. Recently, music-listening and performance has also been shown to affect the regulation of several genes, many of which were identified in songbird singing. MicroRNAs affect gene regulation and studying their expression may give new insights into the epigenetic effect of music. Here, we studied the effect of 2 hours of classical music-performance on the peripheral blood microRNA expressions in professional musicians with respect to a control activity without music for the same duration. As detecting transcriptomic changes in the functional human brain remains a challenge for geneticists, we used peripheral blood to study music-performance induced microRNA changes and interpreted the results in terms of potential effects on brain function, based on the current knowledge about the microRNA function in blood and brain. We identified significant (FDR <0.05) up-regulation of five microRNAs; hsa-miR-3909, hsa-miR-30d-5p, hsa-miR-92a-3p, hsa-miR-222-3p and hsa-miR-30a-5p; and down-regulation of two microRNAs; hsa-miR-6803-3p and hsa-miR-1249-3p. hsa-miR-222-3p and hsa-miR-92a-3p putatively target FOXP2, which was found down-regulated by microRNA regulation in songbird singing. miR-30d and miR-222 corroborate microRNA response observed in zebra finch song-listening/learning. miR-222 is induced by ERK cascade, which is important for memory formation, motor neuron functions and neuronal plasticity. miR-222 is also activated by FOSL1, an immediate early gene from the FOS family of transcriptional regulators which are activated by auditory-motor stimuli. miR-222 and miR-92 promote neurite outgrowth by negatively regulating the neuronal growth inhibitor, PTEN, and by activating CREB expression and phosphorylation. The up-regulation of microRNAs previously found to be regulators of auditory and nervous system functions (miR-30d, miR-92a and miR-222) is indicative of the sensory perception processes associated with music-performance. Akt signalling pathway which has roles in cell survival, cell differentiation, activation of CREB signalling and dopamine transmission was one of the functions regulated by the up-regulated microRNAs; in accordance with functions identified from songbird learning. The up-regulated microRNAs were also found to be regulators of apoptosis, suggesting repression of apoptotic mechanisms in connection with music-performance. Furthermore, comparative analyses of the target genes of differentially expressed microRNAs with that of the song-responsive microRNAs in songbirds suggest convergent regulatory mechanisms underlying auditory perception.
Collapse
Affiliation(s)
| | - Tuire Kuusi
- DocMus Doctoral School, Sibelius Academy, University of the Arts, Helsinki, Finland
| | - Minna Ahvenainen
- Department of Medical Genetics, University of Helsinki, Helsinki, Finland
| | - Anju K Philips
- Department of Medical Genetics, University of Helsinki, Helsinki, Finland
| | - Irma Järvelä
- Department of Medical Genetics, University of Helsinki, Helsinki, Finland
| |
Collapse
|
38
|
Alvarez-Flores MP, Hébert A, Gouelle C, Geller S, Chudzinski-Tavassi AM, Pellerin L. Neuroprotective effect of rLosac on supplement-deprived mouse cultured cortical neurons involves maintenance of monocarboxylate transporter MCT2 protein levels. J Neurochem 2018; 148:80-96. [PMID: 30347438 DOI: 10.1111/jnc.14617] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2018] [Revised: 09/02/2018] [Accepted: 10/17/2018] [Indexed: 01/08/2023]
Abstract
The recombinant Lonomia obliqua Stuart-factor activator (rLosac) is a recombinant hemolin which belongs to the immunoglobulin superfamily of cell adhesion molecules. It is capable of inducing pro-survival activity in serum-deprived human umbilical vein endothelial cells (HUVECs) and fibroblasts by increasing mitochondrial metabolism. We hypothesize that it could promote neuronal survival by acting on neuroenergetics. Our study reveals that treatment of primary mouse cortical neurons cultured in neurobasal medium lacking B27 supplement with rLosac led to an enhancement of cell viability in a time- and concentration-dependent manner. In parallel, preserved or enhanced phosphorylation of Akt, p44, and p42 MAPK, as well as mTOR was observed following treatment with rLosac. During deprivation, as assessed by western blot and qRT-PCR, protein and mRNA expression of MCT2 (the predominant neuronal monocarboxylate transporter allowing lactate use as an alternative energy substrate) decreased significantly in B27 supplement-deprived cortical neurons and was hardly detected after 24 h of deprivation. Interestingly, rLosac maintained MCT2 protein expression after 24 h of deprivation including at the cell surface without preventing mRNA loss. MCT2 knockdown reduced rLosac-enhanced cell viability, confirming its involvement in rLosac effect. Enhanced uptake of lactate was detected following rLosac treatment and might contribute to rLosac-enhanced viability during deprivation. In the presence of both lactate and rLosac, cell viability was higher than in the presence of lactate alone. Our observations suggest that rLosac promotes cell viability in stressed (B27 supplement-deprived) neurons by facilitating the use of lactate as energy substrate via the preservation of MCT2 protein expression. OPEN SCIENCE BADGES: This article has received a badge for *Open Materials* because it provided all relevant information to reproduce the study in the manuscript. The complete Open Science Disclosure form for this article can be found at the end of the article. More information about the Open Practices badges can be found at https://cos.io/our-services/open-science-badges/.
Collapse
Affiliation(s)
- Miryam P Alvarez-Flores
- Department of Physiology, University of Lausanne, Lausanne, Switzerland.,Laboratory of Molecular Biology - Centre of Excellence in New Target Discover CENTD, Butantan Institute, São Paulo, Brazil
| | - Audrey Hébert
- Department of Physiology, University of Lausanne, Lausanne, Switzerland
| | - Cathy Gouelle
- Department of Physiology, University of Lausanne, Lausanne, Switzerland
| | - Sarah Geller
- Department of Physiology, University of Lausanne, Lausanne, Switzerland
| | - Ana M Chudzinski-Tavassi
- Laboratory of Molecular Biology - Centre of Excellence in New Target Discover CENTD, Butantan Institute, São Paulo, Brazil
| | - Luc Pellerin
- Department of Physiology, University of Lausanne, Lausanne, Switzerland.,Centre de Résonance Magnétique des Systèmes Biologiques, UMR5536 CNRS, LabEx TRAIL-IBIO, Université de Bordeaux, Bordeaux Cedex, France
| |
Collapse
|
39
|
Kandasamy M, Aigner L. Reactive Neuroblastosis in Huntington's Disease: A Putative Therapeutic Target for Striatal Regeneration in the Adult Brain. Front Cell Neurosci 2018; 12:37. [PMID: 29593498 PMCID: PMC5854998 DOI: 10.3389/fncel.2018.00037] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Accepted: 01/31/2018] [Indexed: 01/19/2023] Open
Abstract
The cellular and molecular mechanisms underlying the reciprocal relationship between adult neurogenesis, cognitive and motor functions have been an important focus of investigation in the establishment of effective neural replacement therapies for neurodegenerative disorders. While neuronal loss, reactive gliosis and defects in the self-repair capacity have extensively been characterized in neurodegenerative disorders, the transient excess production of neuroblasts detected in the adult striatum of animal models of Huntington's disease (HD) and in post-mortem brain of HD patients, has only marginally been addressed. This abnormal cellular response in the striatum appears to originate from the selective proliferation and ectopic migration of neuroblasts derived from the subventricular zone (SVZ). Based on and in line with the term "reactive astrogliosis", we propose to name the observed cellular event "reactive neuroblastosis". Although, the functional relevance of reactive neuroblastosis is unknown, we speculate that this process may provide support for the tissue regeneration in compensating the structural and physiological functions of the striatum in lieu of aging or of the neurodegenerative process. Thus, in this review article, we comprehend different possibilities for the regulation of striatal neurogenesis, neuroblastosis and their functional relevance in the context of HD.
Collapse
Affiliation(s)
- Mahesh Kandasamy
- Laboratory of Stem Cells and Neuroregeneration, Department of Animal Science, School of Life Sciences, Bharathidasan University, Tiruchirappalli, India
- Faculty Recharge Programme, University Grants Commission (UGC-FRP), New Delhi, India
| | - Ludwig Aigner
- Institute of Molecular Regenerative Medicine, Paracelsus Medical University, Salzburg, Austria
- Spinal Cord Injury and Tissue Regeneration Center, Paracelsus Medical University, Salzburg, Austria
| |
Collapse
|
40
|
Abstract
Habits are an essential and pervasive component of our daily lives that allow us to efficiently perform routine tasks. But their disruption contributes to the symptoms that underlie many psychiatric diseases. Emerging data are revealing the cellular and molecular mechanisms of habit formation in the dorsal striatum. New data suggest that in both the dorsolateral and dorsomedial striatum histone deacetylase (HDAC) activity acts as a critical negative regulator of the transcriptional processes underlying habit formation. In this review, we discuss this recent work and draw conclusions relevant to the treatment of diseases marked by maladaptive habits.
Collapse
Affiliation(s)
| | - Kate M Wassum
- Dept. of Psychology, UCLA, Los Angeles, CA 90095, USA.,Brain Research Institute, UCLA, Los Angeles, CA 90095, USA
| |
Collapse
|
41
|
Zhu X, Dong J, Han B, Huang R, Zhang A, Xia Z, Chang H, Chao J, Yao H. Neuronal Nitric Oxide Synthase Contributes to PTZ Kindling-Induced Cognitive Impairment and Depressive-Like Behavior. Front Behav Neurosci 2017; 11:203. [PMID: 29093670 PMCID: PMC5651248 DOI: 10.3389/fnbeh.2017.00203] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Accepted: 10/06/2017] [Indexed: 12/14/2022] Open
Abstract
Epilepsy is a chronic neurological disease which is usually associated with psychiatric comorbidities. Depsression and cognition impairment are considered to be the most common psychiatric comorbidities in epilepsy patients. However, the specific contribution of epilepsy made to these psychiatric comorbidities remains largely unknown. Here we use pentylenetetrazole (PTZ) kindling, a chronic epilepsy model, to identify neuronal nitric oxide synthase (nNOS) as a signaling molecule triggering PTZ kindling-induced cognitive impairment and depressive-like behavior. Furthermore, we identified that both hippocampal MAPK and PI3K/AKT signaling pathways were activated in response to PTZ kindling, and the increased MAPK and PI3K/AKT signaling activation was paralleled by increased level of reactive oxygen species (ROS) in the hippocampus. However, the PTZ kindling-induced MAPK, PI3K/AKT signaling activities and the ROS level were attenuated by nNOS gene deficiency, suggesting that nNOS may act through ROS-mediated MAPK and PI3K/AKT signaling pathways to trigger cognition deficit and depressive-like behavior in PTZ-kindled mice. Our findings thus define a specific mechanism for chronic epilepsy-induced cognitive impairment and depressive-like behavior, and identify a potential therapeutic target for psychiatric comorbidities in chronic epilepsy patients.
Collapse
Affiliation(s)
- Xinjian Zhu
- Department of Pharmacology, Medical School of Southeast University, Nanjing, China
| | - Jingde Dong
- Department of Geriatric Neurology, Nanjing Brain Hospital Affiliated to Nanjing Medical University, Nanjing, China
| | - Bing Han
- Department of Pharmacology, Medical School of Southeast University, Nanjing, China
| | - Rongrong Huang
- Department of Pharmacology, Medical School of Southeast University, Nanjing, China
| | - Aifeng Zhang
- Department of Pathology, Medical School of Southeast University, Nanjing, China
| | - Zhengrong Xia
- Analysis and Test Center of Nanjing Medical University, Nanjing, China
| | - Huanhuan Chang
- Nanjing Biomedical Research Institute of Nanjing University, Nanjing, China
| | - Jie Chao
- Department of Physiology, Medical School of Southeast University, Nanjing, China
| | - Honghong Yao
- Department of Pharmacology, Medical School of Southeast University, Nanjing, China
| |
Collapse
|