1
|
Zhang X, Vlkolinsky R, Wu C, Dolatabadi N, Scott H, Prikhodko O, Zhang A, Blanco M, Lang N, Piña-Crespo J, Nakamura T, Roberto M, Lipton SA. S-Nitrosylation of CRTC1 in Alzheimer's disease impairs CREB-dependent gene expression induced by neuronal activity. Proc Natl Acad Sci U S A 2025; 122:e2418179122. [PMID: 40014571 PMCID: PMC11892585 DOI: 10.1073/pnas.2418179122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Accepted: 01/10/2025] [Indexed: 03/01/2025] Open
Abstract
cAMP response element-binding protein (CREB)-regulated transcription coactivator 1 (CRTC1) plays an important role in synaptic plasticity, learning, and long-term memory formation through the regulation of neuronal activity-dependent gene expression, and CRTC1 dysregulation is implicated in Alzheimer's disease (AD). Here, we show that increased S-nitrosylation of CRTC1 (forming SNO-CRTC1), as seen in cell-based, animal-based, and human-induced pluripotent stem cell (hiPSC)-derived cerebrocortical neuron-based AD models, disrupts its binding with CREB and diminishes the activity-dependent gene expression mediated by the CRTC1/CREB pathway. We identified Cys216 of CRTC1 as the primary target of S-nitrosylation by nitric oxide (NO)-related species. Using CRISPR/Cas9 techniques, we mutated Cys216 to Ala in hiPSC-derived cerebrocortical neurons bearing one allele of the APPSwe mutation (AD-hiPSC neurons). Introduction of this nonnitrosylatable CRTC1 mutant rescued defects in AD-hiPSC neurons, including decreased neurite length and increased neuronal cell death. Additionally, expression of nonnitrosylatable CRTC1 in vivo in the hippocampus rescued synaptic plasticity in the form of long-term potentiation in 5XFAD mice. Taken together, these results demonstrate that formation of SNO-CRTC1 contributes to the pathogenesis of AD by attenuating the neuronal activity-dependent CREB transcriptional pathway, and suggests a therapeutic target for AD.
Collapse
Affiliation(s)
- Xu Zhang
- Neurodegeneration New Medicines Center and Department of Molecular & Cellular Biology, The Scripps Research Institute, La Jolla, CA92037
| | - Roman Vlkolinsky
- Department of Translational Medicine, The Scripps Research Institute, La Jolla, CA92037
| | - Chongyang Wu
- Neurodegeneration New Medicines Center and Department of Molecular & Cellular Biology, The Scripps Research Institute, La Jolla, CA92037
| | - Nima Dolatabadi
- Neurodegeneration New Medicines Center and Department of Molecular & Cellular Biology, The Scripps Research Institute, La Jolla, CA92037
| | - Henry Scott
- Neurodegeneration New Medicines Center and Department of Molecular & Cellular Biology, The Scripps Research Institute, La Jolla, CA92037
| | - Olga Prikhodko
- Department of Neurosciences, University of California San Diego, School of Medicine, La Jolla, CA92093
| | - Andrew Zhang
- Neurodegeneration New Medicines Center and Department of Molecular & Cellular Biology, The Scripps Research Institute, La Jolla, CA92037
| | - Mayra Blanco
- Neurodegeneration New Medicines Center and Department of Molecular & Cellular Biology, The Scripps Research Institute, La Jolla, CA92037
| | - Nhi Lang
- Neurodegeneration New Medicines Center and Department of Molecular & Cellular Biology, The Scripps Research Institute, La Jolla, CA92037
| | - Juan Piña-Crespo
- Neurodegeneration New Medicines Center and Department of Molecular & Cellular Biology, The Scripps Research Institute, La Jolla, CA92037
| | - Tomohiro Nakamura
- Neurodegeneration New Medicines Center and Department of Molecular & Cellular Biology, The Scripps Research Institute, La Jolla, CA92037
| | - Marisa Roberto
- Department of Translational Medicine, The Scripps Research Institute, La Jolla, CA92037
| | - Stuart A. Lipton
- Neurodegeneration New Medicines Center and Department of Molecular & Cellular Biology, The Scripps Research Institute, La Jolla, CA92037
- Department of Neurosciences, University of California San Diego, School of Medicine, La Jolla, CA92093
| |
Collapse
|
2
|
Kaiser J, Risteska A, Muller AG, Sun H, Lei B, Nay K, Means AR, Cousin MA, Drewry DH, Oakhill JS, Kemp BE, Hannan AJ, Berk M, Febbraio MA, Gundlach AL, Hill-Yardin EL, Scott JW. Convergence on CaMK4: A Key Modulator of Autism-Associated Signaling Pathways in Neurons. Biol Psychiatry 2025; 97:439-449. [PMID: 39442785 DOI: 10.1016/j.biopsych.2024.10.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 09/03/2024] [Accepted: 10/15/2024] [Indexed: 10/25/2024]
Abstract
Although the precise underlying cause(s) of autism spectrum disorder remain unclear, more than 1000 rare genetic variations are associated with the condition. For many people living with profound autism, this genetic heterogeneity has impeded the identification of common biological targets for therapy development for core and comorbid traits that include significant impairments in social communication and repetitive and restricted behaviors. A substantial number of genes associated with autism encode proteins involved in signal transduction and synaptic transmission that are critical for brain development and function. CAMK4 is an emerging risk gene for autism spectrum disorder that encodes the CaMK4 (calcium/calmodulin-dependent protein kinase 4) enzyme. CaMK4 is a key component of a Ca2+-activated signaling pathway that regulates neurodevelopment and synaptic plasticity. In this review, we discuss 3 genetic variants of CAMK4 found in individuals with hyperkinetic movement disorder and comorbid neurological symptoms including autism spectrum disorder that are likely pathogenic with monogenic effect. We also comment on 4 other genetic variations in CAMK4 that show associations with autism spectrum disorder, as well as 12 examples of autism-associated variations in other genes that impact CaMK4 signaling pathways. Finally, we highlight 3 environmental risk factors that impact CaMK4 signaling based on studies of preclinical models of autism and/or clinical cohorts. Overall, we review molecular, genetic, physiological, and environmental evidence that suggest that defects in the CaMK4 signaling pathway may play an important role in a common autism pathogenesis network across numerous patient groups, and we propose CaMK4 as a potential therapeutic target.
Collapse
Affiliation(s)
- Jacqueline Kaiser
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Melbourne, Victoria, Australia; St. Vincent's Institute of Medical Research, Melbourne, Victoria, Australia; Mary McKillop Institute for Health Research, Australian Catholic University, Melbourne, Victoria, Australia
| | - Alana Risteska
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Melbourne, Victoria, Australia
| | - Abbey G Muller
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Melbourne, Victoria, Australia; Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, Melbourne, Victoria, Australia
| | - Haoxiong Sun
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Melbourne, Victoria, Australia
| | - Bethany Lei
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Melbourne, Victoria, Australia
| | - Kevin Nay
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Melbourne, Victoria, Australia
| | - Anthony R Means
- Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas
| | - Margot A Cousin
- Center for Individualized Medicine, College of Medicine, Mayo Clinic, Rochester, Minnesota
| | - David H Drewry
- Structural Genomics Consortium, UNC Eshelman School of Pharmacy, the University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Jonathan S Oakhill
- St. Vincent's Institute of Medical Research, Melbourne, Victoria, Australia; Mary McKillop Institute for Health Research, Australian Catholic University, Melbourne, Victoria, Australia
| | - Bruce E Kemp
- St. Vincent's Institute of Medical Research, Melbourne, Victoria, Australia
| | - Anthony J Hannan
- Florey Institute of Neuroscience and Mental Health, Melbourne, Victoria, Australia; Department of Anatomy and Physiology, the University of Melbourne, Melbourne, Victoria, Australia
| | - Michael Berk
- Florey Institute of Neuroscience and Mental Health, Melbourne, Victoria, Australia; The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Geelong, Victoria, Australia; Orygen, The National Centre of Excellence in Youth Mental Health, Parkville, Melbourne, Australia
| | - Mark A Febbraio
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Melbourne, Victoria, Australia
| | - Andrew L Gundlach
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Melbourne, Victoria, Australia; St. Vincent's Institute of Medical Research, Melbourne, Victoria, Australia; Florey Institute of Neuroscience and Mental Health, Melbourne, Victoria, Australia; Department of Anatomy and Physiology, the University of Melbourne, Melbourne, Victoria, Australia
| | - Elisa L Hill-Yardin
- Department of Anatomy and Physiology, the University of Melbourne, Melbourne, Victoria, Australia; School of Health and Biomedical Sciences, STEM College, RMIT University, Melbourne, Victoria, Australia.
| | - John W Scott
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Melbourne, Victoria, Australia; St. Vincent's Institute of Medical Research, Melbourne, Victoria, Australia; Florey Institute of Neuroscience and Mental Health, Melbourne, Victoria, Australia.
| |
Collapse
|
3
|
Mizuno K, Sugahara M, Kutomi O, Kato R, Itoh T, Fujita S, Yamada M. Direct observation of importin α family member KPNA1 in axonal transport with or without a schizophrenia-related mutation. J Biol Chem 2025; 301:108343. [PMID: 40010609 DOI: 10.1016/j.jbc.2025.108343] [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: 08/12/2024] [Revised: 02/03/2025] [Accepted: 02/18/2025] [Indexed: 02/28/2025] Open
Abstract
Karyopherin α1 (KPNA1)/(human importin α5; mouse importin α1) facilitates cargo transport into the nucleus by forming a complex with a nuclear localization sequence containing cargo and importin β1 (IPOB1). The elevated KPNA1 expression in neurons and the correlation between mutations and psychiatric disorders suggest its broader significance beyond nucleocytoplasmic transport. Although KPNA1 is localized in the neurites of neurons, its role in axonal transport mechanisms remains unclear, and data on the connection between psychiatric disorders and signaling at the periphery of neurons remain limited. To address this knowledge gap, we investigated the dynamics of KPNA1 and related factors within axons. Our results showed that many of the axonal KPNA1 did not form a complex with IPOB1 in noninjured steady-state neurons. Axonal KPNA1 exhibited relatively stationary mobility and some showed bidirectional motility with fluctuating motion. KPNA1 partly comigrated with endosome/lysosome-associated factors, suggesting the presence of novel mechanisms underlie axonal transport and nucleocytoplasmic shuttling involving KPNA1 and IPOB1. Mutated KPNA1, which has been shown to be associated with psychiatric disorders (KPNA1E448X), was predominantly localized to the nucleus and lost from the axon. Incorporating a nuclear export signal (KPNA1E448X-NES) enhanced its subcellular localization and dynamics in the axon. Our findings demonstrate that KPNA1 functions not only as a shuttle between the cytoplasm and nucleus but also as a transporter in neuronal axons, relying on the endosomes for movement away from the nucleus with relatively slow net motions. Furthermore, a mutation in the Kpna1 gene can affect the dynamics of axonal transport. The insights from these mutations provide valuable knowledge for expanding our understanding of psychiatric disorders and facilitate the development of novel treatment strategies.
Collapse
Affiliation(s)
- Katsutoshi Mizuno
- Department of Cell Biology and Biochemistry, Division of Medicine, Faculty of Medical Sciences, University of Fukui, Yoshida-gun, Fukui Prefecture, Japan; Life Science Innovation Center, University of Fukui, Fukui City, Fukui Prefecture, Japan
| | - Masaki Sugahara
- Department of Cell Biology and Biochemistry, Division of Medicine, Faculty of Medical Sciences, University of Fukui, Yoshida-gun, Fukui Prefecture, Japan; Department of Frontier Fiber Technology and Science, Graduate School of Engineering, University of Fukui, Fukui City, Fukui Prefecture, Japan
| | - Osamu Kutomi
- Department of Cell Biology and Biochemistry, Division of Medicine, Faculty of Medical Sciences, University of Fukui, Yoshida-gun, Fukui Prefecture, Japan; Life Science Innovation Center, University of Fukui, Fukui City, Fukui Prefecture, Japan
| | - Ryota Kato
- Department of Cell Biology and Biochemistry, Division of Medicine, Faculty of Medical Sciences, University of Fukui, Yoshida-gun, Fukui Prefecture, Japan; Department of Frontier Fiber Technology and Science, Graduate School of Engineering, University of Fukui, Fukui City, Fukui Prefecture, Japan
| | - Takafumi Itoh
- Department of Bioscience and Biotechnology, Fukui Prefectural University, Yoshida-gun, Fukui Prefecture, Japan
| | - Satoshi Fujita
- Life Science Innovation Center, University of Fukui, Fukui City, Fukui Prefecture, Japan; Department of Frontier Fiber Technology and Science, Graduate School of Engineering, University of Fukui, Fukui City, Fukui Prefecture, Japan
| | - Masami Yamada
- Department of Cell Biology and Biochemistry, Division of Medicine, Faculty of Medical Sciences, University of Fukui, Yoshida-gun, Fukui Prefecture, Japan; Life Science Innovation Center, University of Fukui, Fukui City, Fukui Prefecture, Japan.
| |
Collapse
|
4
|
Lim CR, Ogawa S, Kumari Y. Exploring β-caryophyllene: a non-psychotropic cannabinoid's potential in mitigating cognitive impairment induced by sleep deprivation. Arch Pharm Res 2025; 48:1-42. [PMID: 39653971 DOI: 10.1007/s12272-024-01523-z] [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: 08/02/2024] [Accepted: 11/25/2024] [Indexed: 01/04/2025]
Abstract
Sleep deprivation or sleep loss, a prevalent issue in modern society, is linked to cognitive impairment, leading to heightened risks of errors and accidents. Chronic sleep deprivation affects various cognitive functions, including memory, attention, and decision-making, and is associated with an increased risk of neurodegenerative diseases, cardiovascular issues, and metabolic disorders. This review examines the potential of β-caryophyllene, a dietary non-psychotropic cannabinoid, and FDA-approved flavoring agent, as a therapeutic solution for sleep loss-induced cognitive impairment. It highlights β-caryophyllene's ability to mitigate key contributors to sleep loss-induced cognitive impairment, such as inflammation, oxidative stress, neuronal death, and reduced neuroplasticity, by modulating various signaling pathways, including TLR4/NF-κB/NLRP3, MAPK, Nrf2/HO-1, PI3K/Akt, and cAMP/PKA/CREB. As a naturally occurring, non-psychotropic compound with low toxicity, β-caryophyllene emerges as a promising candidate for further investigation. The review underscores the therapeutic potential of β-caryophyllene for sleep loss-induced cognitive impairment and provides mechanistic insights into its action on crucial pathways, suggesting that β-caryophyllene could be a valuable addition to strategies aimed at combating cognitive impairment and other health issues due to sleep loss.
Collapse
Affiliation(s)
- Cher Ryn Lim
- Neurological Disorder and Aging Research Group (NDA), Neuroscience Research Strength (NRS), Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, 47500, Selangor, Malaysia
| | - Satoshi Ogawa
- Neuroscience Research Strength (NRS), Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, 47500, Selangor, Malaysia
| | - Yatinesh Kumari
- Neurological Disorder and Aging Research Group (NDA), Neuroscience Research Strength (NRS), Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, 47500, Selangor, Malaysia.
| |
Collapse
|
5
|
El-Khatib SM, Vagadia AR, Le ACD, Baulch JE, Ng DQ, Du M, Johnston KG, Tan Z, Xu X, Chan A, Acharya MM. BDNF augmentation reverses cranial radiation therapy-induced cognitive decline and neurodegenerative consequences. Acta Neuropathol Commun 2024; 12:190. [PMID: 39696694 DOI: 10.1186/s40478-024-01906-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Accepted: 11/29/2024] [Indexed: 12/20/2024] Open
Abstract
Cranial radiation therapy (RT) for brain cancers is often associated with the development of radiation-induced cognitive dysfunction (RICD). RICD significantly impacts the quality of life for cancer survivors, highlighting an unmet medical need. Previous human studies revealed a marked reduction in plasma brain-derived neurotrophic factor (BDNF) post-chronic chemotherapy, linking this decline to a substantial cognitive dysfunction among cancer survivors. Moreover, riluzole (RZ)-mediated increased BDNF in vivo in the chemotherapy-exposed mice reversed cognitive decline. RZ is an FDA-approved medication for ALS known to increase BDNF in vivo. In an effort to mitigate the detrimental effects of RT-induced BDNF decline in RICD, we tested the efficacy of RZ in a cranially irradiated (9 Gy) adult mouse model. Notably, RT-exposed mice exhibited significantly reduced hippocampal BDNF, accompanied by increased neuroinflammation, loss of neuronal plasticity-related immediate early gene product, cFos, and synaptic density. Spatial transcriptomic profiling comparing the RT + Vehicle with the RT + RZ group showed gene expression signatures of neuroprotection of hippocampal excitatory neurons post-RZ. RT-exposed mice performed poorly on learning and memory, and memory consolidation tasks. However, irradiated mice receiving RZ (13 mg/kg, drinking water) for 6-7 weeks showed a significant improvement in cognitive function compared to RT-exposed mice receiving vehicle. Dual-immunofluorescence staining, spatial transcriptomics, and biochemical assessment of RZ-treated irradiated brains demonstrated preservation of synaptic integrity and mature neuronal plasticity but not neurogenesis and reduced neuroinflammation concurrent with elevated BDNF levels and transcripts compared to vehicle-treated irradiated brains. In summary, oral administration of RZ represents a viable and translationally feasible neuroprotective approach against RICD.
Collapse
Affiliation(s)
- Sanad M El-Khatib
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, USA
| | - Arya R Vagadia
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, USA
| | - Anh C D Le
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, USA
| | - Janet E Baulch
- Department of Radiation Oncology, School of Medicine, University of California, Irvine, USA
| | - Ding Quan Ng
- Department of Clinical Pharmacy Practice, School of Pharmacy and Pharmaceutical Sciences, University of California, Irvine, USA
| | - Mingyu Du
- Center for Neural Circuit Mapping, School of Medicine, University of California, Irvine, USA
| | - Kevin G Johnston
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, USA
- Center for Neural Circuit Mapping, School of Medicine, University of California, Irvine, USA
| | - Zhiqun Tan
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, USA
- Center for Neural Circuit Mapping, School of Medicine, University of California, Irvine, USA
| | - Xiangmin Xu
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, USA
- Center for Neural Circuit Mapping, School of Medicine, University of California, Irvine, USA
| | - Alexandre Chan
- Department of Clinical Pharmacy Practice, School of Pharmacy and Pharmaceutical Sciences, University of California, Irvine, USA.
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University of California, Irvine, USA.
| | - Munjal M Acharya
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, USA.
- Department of Radiation Oncology, School of Medicine, University of California, Irvine, USA.
| |
Collapse
|
6
|
El-Khatib SM, Vagadia AR, Le ACD, Ng DQ, Baulch JE, Du M, Tan Z, Xu X, Chan A, Acharya MM. BDNF augmentation reverses cranial radiation therapy-induced cognitive decline and neurodegenerative consequences. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.23.614590. [PMID: 39386496 PMCID: PMC11463616 DOI: 10.1101/2024.09.23.614590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
Cranial radiation therapy (RT) for brain cancers is often associated with the development of radiation-induced cognitive dysfunction (RICD). RICD significantly impacts the quality of life for cancer survivors, highlighting an unmet medical need. Previous human studies revealed a marked reduction in plasma brain-derived neurotrophic factor (BDNF) post-chronic chemotherapy, linking this decline to a substantial cognitive dysfunction among cancer survivors. Moreover, riluzole (RZ)-mediated increased BDNF in vivo in the chemotherapy-exposed mice reversed cognitive decline. RZ is an FDA-approved medication for ALS known to increase BDNF in vivo . In an effort to mitigate the detrimental effects of RT-induced BDNF decline in RICD, we tested the efficacy of RZ in a cranially irradiated (9 Gy) adult mouse model. Notably, RT-exposed mice exhibited significantly reduced hippocampal BDNF, accompanied by increased neuroinflammation, loss of neuronal plasticity-related immediate early gene product, cFos, and synaptic density. Spatial transcriptomic profiling comparing the RT+Veh with the RT+RZ group showed gene expression signatures of neuroprotection of hippocampal excitatory neurons post-RZ. RT-exposed mice performed poorly on learning and memory, and memory consolidation tasks. However, irradiated mice receiving RZ (13 mg/kg, drinking water) for 6-7 weeks showed a significant improvement in cognitive function compared to RT-exposed mice receiving vehicle. Dual-immunofluorescence staining, spatial transcriptomics, and biochemical assessment of RZ-treated irradiated brains demonstrated preservation of synaptic integrity and neuronal plasticity but not neurogenesis and reduced neuroinflammation concurrent with elevated BDNF levels and transcripts compared to vehicle-treated irradiated brains. In summary, oral administration of RZ represents a viable and translationally feasible neuroprotective approach against RICD.
Collapse
|
7
|
Yasmin T, Sadia A, Nadeem L, Basra MAR, Rice ML, Raza MH. Whole Genome Analysis in Consanguineous Families Reveals New Loci for Speech Sound Disorder (SSD). Genes (Basel) 2024; 15:1069. [PMID: 39202429 PMCID: PMC11354014 DOI: 10.3390/genes15081069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 08/09/2024] [Accepted: 08/09/2024] [Indexed: 09/03/2024] Open
Abstract
Speech is the most common means of communication in humans. Any defect in accurate speech production ability results in the development of speech sound disorder (SSD), a condition that can significantly impair an individual's academic performance, social interactions, and relationships with peers and adults. This study investigated the genetic basis of SSD in three Pakistani families. We performed family-based genome-wide parametric linkage analysis and homozygosity mapping in three consanguineous families with SSD from the Punjab province of Pakistan. The Test for Assessment of Articulation and Phonology in Urdu (TAAPU) was used to analyze the speech articulation data and determine the Percentage Correct Consonants (PCC) score. The PCC score defined the affected and unaffected individuals in each family. Parametric linkage analysis revealed a linkage to chromosome 5 (5q21.3-5q23.1) with a significant logarithm of the odds (LOD) score of 3.13 in a Pakistani family with specific language impairment-97 (PKSLI-97) under an autosomal recessive mode of inheritance. The other two families showed a suggestive linkage at 6p22.1, 14q12, and 16q12.1 under the recessive mode of inheritance. Interestingly, homozygosity mapping showed a loss of heterozygosity in the linkage region at 5q15-5q23.1, shared among seven affected (mostly in the younger generation) and one unaffected individual of PKSLI-97. Our analysis identified the 6p22 locus previously implicated in dyslexia, childhood apraxia of speech (CAS), and language impairment, confirming the role of KIAA0319 and DCDC2 in this locus. These findings provide statistical evidence for the genomic regions associated with articulation disorder and offer future opportunities to further the role of genes in speech production.
Collapse
Affiliation(s)
- Tahira Yasmin
- Centre for Clinical and Nutritional Chemistry, School of Chemistry, University of the Punjab, Lahore 54590, Pakistan; (T.Y.); (A.S.); (L.N.); (M.A.R.B.)
| | - Aatika Sadia
- Centre for Clinical and Nutritional Chemistry, School of Chemistry, University of the Punjab, Lahore 54590, Pakistan; (T.Y.); (A.S.); (L.N.); (M.A.R.B.)
- Laboratory of Organic Electronics, Department of Science and Technology, Linköping University, Norrköping Campus, 60221 Norrköping, Sweden
| | - Laraib Nadeem
- Centre for Clinical and Nutritional Chemistry, School of Chemistry, University of the Punjab, Lahore 54590, Pakistan; (T.Y.); (A.S.); (L.N.); (M.A.R.B.)
| | - Muhammad Asim Raza Basra
- Centre for Clinical and Nutritional Chemistry, School of Chemistry, University of the Punjab, Lahore 54590, Pakistan; (T.Y.); (A.S.); (L.N.); (M.A.R.B.)
| | - Mabel L. Rice
- Speech-Language-Hearing Sciences & Disorders, University of Kansas, Lawrence, KS 66045, USA;
| | - Muhammad Hashim Raza
- Speech-Language-Hearing Sciences & Disorders, University of Kansas, Lawrence, KS 66045, USA;
| |
Collapse
|
8
|
Zhu Z, McClintock TS, Bieberich E. Transcriptomics analysis reveals potential regulatory role of nSMase2 (Smpd3) in nervous system development and function of middle-aged mouse brains. GENES, BRAIN, AND BEHAVIOR 2024; 23:e12911. [PMID: 39171374 PMCID: PMC11339599 DOI: 10.1111/gbb.12911] [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: 03/29/2024] [Revised: 07/22/2024] [Accepted: 07/29/2024] [Indexed: 08/23/2024]
Abstract
Neutral sphingomyelinase-2 (nSMase2), gene name sphingomyelin phosphodiesterase-3 (Smpd3), is a key regulatory enzyme responsible for generating the sphingolipid ceramide. The function of nSMase2 in the brain is still controversial. To better understand the functional roles of nSMase2 in the aging mouse brain, we applied RNA-seq analysis, which identified a total of 1462 differentially abundant mRNAs between +/fro and fro/fro, of which 891 were increased and 571 were decreased in nSMase2-deficient mouse brains. The most strongly enriched GO and KEGG annotation terms among transcripts increased in fro/fro mice included synaptogenesis, synapse development, synaptic signaling, axon development, and axonogenesis. Among decreased transcripts, enriched annotations included ribosome assembly and mitochondrial protein complex functions. KEGG analysis of decreased transcripts also revealed overrepresentation of annotations for Alzheimer's disease (AD), Parkinson's disease (PD), and Huntington disease (HD). Ingenuity Pathway Analysis (IPA) tools predicted lower susceptibility to these neurodegenerative disorders, as well as predictions agreeing with stronger synaptic function, learning, and memory in fro/fro mice. The IPA tools identified signaling proteins, epigenetic regulators, and microRNAs as likely upstream regulators of the broader set of genes encoding the affected transcripts. It also revealed 16 gene networks, each linked to biological processes identified as overrepresented annotations among the affected transcripts by multiple analysis methods. Therefore, the analysis of these RNA-seq data indicates that nSMase2 impacts synaptic function and neural development, and may contribute to the onset and development of neurodegenerative diseases in middle-aged mice.
Collapse
Affiliation(s)
- Zhihui Zhu
- Department of PhysiologyUniversity of Kentucky College of MedicineLexingtonKentuckyUSA
| | - Timothy S. McClintock
- Department of PhysiologyUniversity of Kentucky College of MedicineLexingtonKentuckyUSA
| | - Erhard Bieberich
- Department of PhysiologyUniversity of Kentucky College of MedicineLexingtonKentuckyUSA
- Veterans Affairs Medical CenterLexingtonKentuckyUSA
| |
Collapse
|
9
|
Ifejeokwu OV, Do A, El Khatib SM, Ho NH, Zavala A, Othy S, Acharya MM. Immune Checkpoint Inhibition-related Neuroinflammation Disrupts Cognitive Function. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.01.601087. [PMID: 39005282 PMCID: PMC11244914 DOI: 10.1101/2024.07.01.601087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/16/2024]
Abstract
Combinatorial blockade of Cytotoxic T-lymphocyte associated protein 4 (CTLA-4) and Programmed Cell Death Protein 1 (PD-1) significantly improve the progression-free survival of individuals with metastatic cancers, including melanoma. In addition to unleashing anti-tumor immunity, combination immune checkpoint inhibition (ICI) disrupts immune-regulatory networks critical for maintaining homeostasis in various tissues, including the central nervous system (CNS). Although ICI- and cancer-related cognitive impairments (CRCI) in survivors are increasingly becoming evident, our understanding of ICI-induced immune-related adverse effects (IREA) in the CNS remains incomplete. Here, our murine melanoma model reveals that combination ICI impairs hippocampal-dependent learning and memory, as well as memory consolidation processes. Mechanistically, combination ICI disrupted synaptic integrity, and neuronal plasticity, reduced myelin, and further predisposed CNS for exaggerated experimental autoimmune encephalomyelitis. Combination ICI substantially altered both lymphoid and myeloid cells in the CNS. Neurogenesis was unaffected, however, microglial activation persisted for two-months post- ICI, concurrently with cognitive deficits, which parallels clinical observations in survivors. Overall, our results demonstrate that blockade of CTLA-4 and PD-1 alters neuro-immune homeostasis and activates microglia, promoting long-term neurodegeneration and driving cognitive impairments. Therefore, limiting microglial activation is a potential avenue to mitigate CNS IRAE while maintaining the therapeutic benefits of rapidly evolving ICIs and their combinations. SIGNIFICANCE Despite the superior therapeutic efficacy of immune checkpoint inhibition (ICI) for cancers, its undesired effects on brain function are not fully understood. Here, we demonstrate that combination ICI elevates neuroinflammation, activates microglia, leading to detrimental neurodegenerative and neurocognitive sequelae.
Collapse
|
10
|
Scherlinger M, Li H, Pan W, Li W, Karino K, Vichos T, Boulougoura A, Yoshida N, Tsokos MG, Tsokos GC. CaMK4 controls follicular helper T cell expansion and function during normal and autoimmune T-dependent B cell responses. Nat Commun 2024; 15:840. [PMID: 38287012 PMCID: PMC10825135 DOI: 10.1038/s41467-024-45080-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Accepted: 01/11/2024] [Indexed: 01/31/2024] Open
Abstract
Systemic lupus erythematosus (SLE) is an autoimmune disease characterized by dysregulated B cell compartment responsible for the production of autoantibodies. Here, we show that T cell-specific expression of calcium/calmodulin-dependent protein kinase IV (CaMK4) leads to T follicular helper (Tfh) cells expansion in models of T-dependent immunization and autoimmunity. Mechanistically, CaMK4 controls the Tfh-specific transcription factor B cell lymphoma 6 (Bcl6) at the transcriptional level through the cAMP responsive element modulator α (CREMα). In the absence of CaMK4 in T cells, germinal center formation and humoral immunity is impaired in immunized mice, resulting in reduced anti-dsDNA titres, as well as IgG and complement kidney deposition in the lupus-prone B6.lpr mouse. In human Tfh cells, CaMK4 inhibition reduced BCL6 expression and IL-21 secretion ex vivo, resulting in impaired plasmablast formation and IgG production. In patients with SLE, CAMK4 mRNA levels in Tfh cells correlated with those of BCL6. In conclusion, we identify CaMK4/CREMα as a driver of T cell-dependent B cell dysregulation in autoimmunity.
Collapse
Affiliation(s)
- Marc Scherlinger
- Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA.
- Rheumatology department, Strasbourg University Hospital of Hautepierre, Strasbourg, France.
- Laboratoire d'ImmunoRhumatologie Moléculaire, Institut National de la Santé et de la Recherche Médicale (INSERM) UMR_S 1109, Strasbourg, France.
| | - Hao Li
- Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Wenliang Pan
- Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Wei Li
- Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Kohei Karino
- Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Theodoros Vichos
- Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | | | - Nobuya Yoshida
- Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Maria G Tsokos
- Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - George C Tsokos
- Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA.
| |
Collapse
|
11
|
Liu X, Zheng Y, Wang Q, Zhao L, Zhang Z, Wang H, Yang Y, Song N, Xiang J, Shen Y, Fan S. Artificially reprogrammed stem cells deliver transcytosable nanocomplexes for improved spinal cord repair. J Control Release 2023; 364:601-617. [PMID: 37926244 DOI: 10.1016/j.jconrel.2023.10.051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 10/12/2023] [Accepted: 10/31/2023] [Indexed: 11/07/2023]
Abstract
Stem cell transplantation holds great promise for restoring function after spinal cord injury (SCI), but its therapeutic efficacy heavily depends on the innate capabilities of the cells and the microenvironment at the lesion site. Herein, a potent cell therapeutic (NCs@SCs) is engineered by artificially reprogramming bone marrow mesenchymal stem cells (BMSCs) with oxidation-responsive transcytosable gene-delivery nanocomplexes (NCs), which endows cells with robust oxidative stress resistance and improved cytokine secretion. NCs@SCs can accumulate in the injured spinal cord after intravenous administration via chemotaxis and boost successive transcytosis to deliver NCs to neurons, augmenting ciliary neurotrophic factor (CNTF) production in both BMSCs and neurons in response to elevated ROS levels. Furthermore, NCs@SCs can actively sense and eliminate ROS and re-educate recruited M1-like macrophages into the anti-inflammatory M2 phenotype via a paracrine pathway, ultimately reshaping the inflammatory microenvironment. Synergistically, NCs@SCs exhibit durable survival and provide neuroprotection against secondary damage, enabling significant locomotor function recovery in SCI rats. Transcriptome analysis reveals that regulation of the ROS/MAPK signaling pathway is involved in SCI therapy by NCs@SCs. This study presents a nanomaterial-mediated cell-reprogramming approach for developing live cell therapeutics, showing significant potential in the treatment of SCI and other neuro-injury disorders.
Collapse
Affiliation(s)
- Xin Liu
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310016, China; Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, Zhejiang 310016, China
| | - Yufei Zheng
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310016, China; Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, Zhejiang 310016, China
| | - Qingqing Wang
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310016, China; Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, Zhejiang 310016, China
| | - Lan Zhao
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310016, China; Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, Zhejiang 310016, China
| | - Zhaowei Zhang
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310016, China; Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, Zhejiang 310016, China
| | - Haoli Wang
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310016, China; Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, Zhejiang 310016, China
| | - Yang Yang
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310016, China; Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, Zhejiang 310016, China
| | - Nan Song
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310016, China; Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, Zhejiang 310016, China
| | - Jiajia Xiang
- Zhejiang Key Laboratory of Smart Biomaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, Zhejiang 310058, China; ZJU-Hangzhou Global Scientific and Technological Innovation Center, Zhejiang University, Hangzhou, Zhejiang 311215, China.
| | - Youqing Shen
- Zhejiang Key Laboratory of Smart Biomaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, Zhejiang 310058, China.
| | - Shunwu Fan
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310016, China; Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, Zhejiang 310016, China.
| |
Collapse
|
12
|
Liu B, Zhu X, Zhou Q, Su Y, Qian Y, Ma Z, Gu X, Xia T. Activating ryanodine receptor improves isoflurane-induced cognitive dysfunction. Brain Res Bull 2023; 204:110790. [PMID: 37852420 DOI: 10.1016/j.brainresbull.2023.110790] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Revised: 09/30/2023] [Accepted: 10/16/2023] [Indexed: 10/20/2023]
Abstract
BACKGROUND Postoperative cognitive dysfunction (POCD) is characterized by impaired learning and memory. 6 h duration isoflurane anesthesia is an important factor to induce POCD, and the dysfunction of ryanodine receptor (RyR) in the hippocampus may be involved in this process. We investigated the expression of RyR3 in the hippocampus of mice after 6-h duration isoflurane anesthesia, as well as the improvement of RyR receptor agonist caffeine on POCD mice, while attempting to identify the underlying molecular mechanism. MATERIALS We constructed a POCD model using 8-week-old male C57BL/6J mice that were exposed to 6-h duration isoflurane. Prior to the three-day cognitive behavioral experiment, RyR agonist caffeine were injected. Fear conditioning and location memory tests were used in behavioral studies. We also exposed the mouse neuroblastoma cell line Neuro-2a (N2A) to 6-h duration isoflurane exposure to simulate the conditions of in vivo cognitive dysfunction. We administered ryanodine receptor agonist (caffeine) and inhibitor (ryanodine) to N2a cells. Following that, we performed a series of bioinformatics analysis to discover proteins that are involved in the development of cognitive dysfunction. Rt-PCR and Western blot were used to assess mRNA level and protein expression. RESULTS 6-h duration isoflurane anesthesia induced cognitive dysfunction and increased RyR3 mRNA levels in hippocampus. The mRNA levels of RyR3 in cultured N2a cells after anesthesia were comparable to those in vivo, and the RyR agonist caffeine corrected the expression of some cognitive-related phenotypic proteins that were disturbed after anesthesia. Intraperitoneal injection of RyR agonist caffeine can improve cognitive function after isoflurane anesthesia in mice, and bioinformatics analyses suggest that CaMKⅣ may be involved in the molecular mechanism. CONCLUSION Ryanodine receptor agonist caffeine may improve cognitive dysfunction in mice after isoflurane anesthesia.
Collapse
Affiliation(s)
- Binwen Liu
- Department of Anesthesiology, Affiliated Drum Tower Hospital of Medical School of Nanjing University, Nanjing 210008, China; Medical School, Nanjing University, Nanjing 210093, China; Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing, China; State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, China.
| | - Xurui Zhu
- Department of Anesthesiology, Affiliated Drum Tower Hospital of Medical School of Nanjing University, Nanjing 210008, China.
| | - Qingyun Zhou
- Department of Anesthesiology, Affiliated Drum Tower Hospital of Medical School of Nanjing University, Nanjing 210008, China; Medical School, Nanjing University, Nanjing 210093, China; Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing, China; State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, China.
| | - Yan Su
- Department of Anesthesiology, Affiliated Drum Tower Hospital of Medical School of Nanjing University, Nanjing 210008, China; Medical School, Nanjing University, Nanjing 210093, China; Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing, China; State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, China.
| | - Yue Qian
- Department of Anesthesiology, Affiliated Drum Tower Hospital of Medical School of Nanjing University, Nanjing 210008, China.
| | - Zhengliang Ma
- Department of Anesthesiology, Affiliated Drum Tower Hospital of Medical School of Nanjing University, Nanjing 210008, China.
| | - Xiaoping Gu
- Department of Anesthesiology, Affiliated Drum Tower Hospital of Medical School of Nanjing University, Nanjing 210008, China.
| | - Tianjiao Xia
- Medical School, Nanjing University, Nanjing 210093, China; Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing, China; State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, China.
| |
Collapse
|
13
|
Chong YS, Wong LW, Gaunt J, Lee YJ, Goh CS, Morris RGM, Ch'ng TH, Sajikumar S. Distinct contributions of ventral CA1/amygdala co-activation to the induction and maintenance of synaptic plasticity. Cereb Cortex 2023; 33:676-690. [PMID: 35253866 DOI: 10.1093/cercor/bhac093] [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: 12/24/2021] [Revised: 02/11/2022] [Accepted: 02/12/2022] [Indexed: 02/03/2023] Open
Abstract
The amygdala is known to modulate hippocampal synaptic plasticity. One role could be an immediate effect of basolateral amygdala (BLA) in priming synaptic plasticity in the hippocampus. Another role could be through associative synaptic co-operation and competition that triggers events involved in the maintenance of synaptic potentiation. We present evidence that the timing and activity level of BLA stimulation are important factors for the induction and maintenance of long-term potentiation (LTP) in ventral hippocampal area CA1. A 100 Hz BLA co-stimulation facilitated the induction of LTP, whereas 200 Hz co-stimulation attenuated induction. A 100 Hz BLA co-stimulation also caused enhanced persistence, sufficient to prevent synaptic competition. This maintenance effect is likely through translational mechanisms, as mRNA expression of primary response genes was unaffected, whereas protein level of plasticity-related products was increased. Further understanding of the neural mechanisms of amygdala modulation on hippocampus could provide insights into the mechanisms of emotional disorders.
Collapse
Affiliation(s)
- Yee Song Chong
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, SIngapore 117597, Singapore.,Life Sciences Institute Neurobiology Programme, National University of Singapore, Singapore 117456, Singapore
| | - Lik-Wei Wong
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, SIngapore 117597, Singapore.,Life Sciences Institute Neurobiology Programme, National University of Singapore, Singapore 117456, Singapore.,Healthy Longevity Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117456, Singapore
| | - Jessica Gaunt
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232, Singapore
| | - Yan Jun Lee
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232, Singapore.,Interdisciplinary Graduate School, Nanyang Technological University, Singapore 637335, Singapore
| | - Cai Shan Goh
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, SIngapore 117597, Singapore.,Life Sciences Institute Neurobiology Programme, National University of Singapore, Singapore 117456, Singapore
| | - Richard G M Morris
- Laboratory for Cognitive Neuroscience, Centre for Cognitive and Neural Systems, University of Edinburgh, Edinburgh EH8 9JZ, Scotland
| | - Toh Hean Ch'ng
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232, Singapore.,School of Biological Sciences, Nanyang Technological University, Singapore 637551, Singapore
| | - Sreedharan Sajikumar
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, SIngapore 117597, Singapore.,Life Sciences Institute Neurobiology Programme, National University of Singapore, Singapore 117456, Singapore.,Healthy Longevity Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117456, Singapore
| |
Collapse
|
14
|
Sanderson JL, Freund RK, Castano AM, Benke TA, Dell'Acqua ML. The Ca V1.2 G406R mutation decreases synaptic inhibition and alters L-type Ca 2+ channel-dependent LTP at hippocampal synapses in a mouse model of Timothy Syndrome. Neuropharmacology 2022; 220:109271. [PMID: 36162529 PMCID: PMC9644825 DOI: 10.1016/j.neuropharm.2022.109271] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 09/09/2022] [Accepted: 09/20/2022] [Indexed: 11/17/2022]
Abstract
Genetic alterations in autism spectrum disorders (ASD) frequently disrupt balance between synaptic excitation and inhibition and alter plasticity in the hippocampal CA1 region. Individuals with Timothy Syndrome (TS), a genetic disorder caused by CaV1.2 L-type Ca2+ channel (LTCC) gain-of function mutations, such as G406R, exhibit social deficits, repetitive behaviors, and cognitive impairments characteristic of ASD that are phenocopied in TS2-neo mice expressing G406R. Here, we characterized hippocampal CA1 synaptic function in male TS2-neo mice and found basal excitatory transmission was slightly increased and inhibitory transmission strongly decreased. We also found distinct impacts on two LTCC-dependent forms of long-term potentiation (LTP) synaptic plasticity that were not readily consistent with LTCC gain-of-function. LTP induced by high-frequency stimulation (HFS) was strongly impaired in TS2-neo mice, suggesting decreased LTCC function. Yet, CaV1.2 expression, basal phosphorylation, and current density were similar for WT and TS2-neo. However, this HFS-LTP also required GABAA receptor activity, and thus may be impaired in TS2-neo due to decreased inhibitory transmission. In contrast, LTP induced in WT mice by prolonged theta-train (PTT) stimulation in the presence of a β-adrenergic receptor agonist to increase CaV1.2 phosphorylation was partially induced in TS2-neo mice by PTT stimulation alone, consistent with increased LTCC function. Overall, our findings provide insights regarding how altered CaV1.2 channel function disrupts basal transmission and plasticity that could be relevant for neurobehavioral alterations in ASD.
Collapse
Affiliation(s)
- Jennifer L Sanderson
- Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, 12800 E. 19th Ave, Mail Stop 8303, Aurora, CO, 80045, USA
| | - Ronald K Freund
- Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, 12800 E. 19th Ave, Mail Stop 8303, Aurora, CO, 80045, USA
| | - Anna M Castano
- Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, 12800 E. 19th Ave, Mail Stop 8303, Aurora, CO, 80045, USA
| | - Timothy A Benke
- Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, 12800 E. 19th Ave, Mail Stop 8303, Aurora, CO, 80045, USA; Departments of Pediatrics, Neurology, and Otolaryngology, University of Colorado School of Medicine, Anschutz Medical Campus, 12800 E. 19th Ave, Mail Stop 8303, Aurora, CO, 80045, USA
| | - Mark L Dell'Acqua
- Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, 12800 E. 19th Ave, Mail Stop 8303, Aurora, CO, 80045, USA.
| |
Collapse
|
15
|
Glucocorticoid-Regulated Kinase CAMKIγ in the Central Amygdala Controls Anxiety-like Behavior in Mice. Int J Mol Sci 2022; 23:ijms232012328. [PMID: 36293185 PMCID: PMC9604347 DOI: 10.3390/ijms232012328] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 09/30/2022] [Accepted: 10/04/2022] [Indexed: 11/07/2022] Open
Abstract
The expression of the Calcium/Calmodulin-Dependent Protein Kinase I gamma (encoded by the Camk1g gene) depends on the activation of glucocorticoid receptors (GR) and is strongly regulated by stress. Since Camk1g is primarily expressed in neuronal cells of the limbic system in the brain, we hypothesized that it could be involved in signaling mechanisms that underlie the adaptive or maladaptive responses to stress. Here, we find that restraint-induced stress and the GR agonist dexamethasone robustly increase the expression of Camk1g in neurons of the amygdalar nuclei in the mouse brain. To assess the functional role of Camk1g expression, we performed a virally induced knock-down of the transcript. Mice with bilateral amygdala-specific Camk1g knock-down showed increased anxiety-like behaviors in the light-dark box, and an increase in freezing behavior after fear-conditioning, but normal spatial working memory during exploration of a Y-maze. Thus, we confirm that Camk1g is a neuron-specific GR-regulated transcript, and show that it is specifically involved in behaviors related to anxiety, as well as responses conditioned by aversive stimuli.
Collapse
|
16
|
Espasandín C, Rivero S, Bengoa L, Cal K, Romanelli G, Benech JC, Damián JP. CaMKIV/CREB/BDNF signaling pathway expression in prefrontal cortex, amygdala, hippocampus and hypothalamus in streptozotocin-induced diabetic mice with anxious-like behavior. Exp Brain Res 2022; 240:2687-2699. [PMID: 35984483 DOI: 10.1007/s00221-022-06446-z] [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: 03/04/2022] [Accepted: 08/14/2022] [Indexed: 11/04/2022]
Abstract
Individuals with diabetes mellitus (DM) tend to manifest anxiety and depression, which could be related to changes in the expression of calcium/calmodulin-dependent protein kinase IV (CaMKIV), transcription factor cyclic AMP-responsive element binding protein (CREB), phosphorylated CREB (pCREB) and brain-derived neurotrophic factor (BDNF) in different brain regions. The objective of this study was to determine whether mice with type 1 diabetes (T1DM) induced with streptozotocin show a profile of anxious-type behaviors and alterations in the expression/activity of CaMKIV, CREB, pCREB and BDNF in different regions of the brain (prefrontal cortex, amygdala, hippocampus and hypothalamus) in comparison to non-diabetic mice (NDB). Mice with 3 months of chronic DM showed an anxious-like behavioral profile in two anxiety tests (Open Field and Elevated Plus Maze), when compared to NDB. There were significant differences in the expression of cell signaling proteins: diabetic mice had a lower expression of CaMKIV in the hippocampus, a greater expression of CREB in the amygdala and hypothalamus, as well as a lower pCREB/CREB in hypothalamus than NDB mice (P < 0.05). This is the first study evaluating the expression of CaMKIV in the brain of animals with DM, who presented lower expression of this protein in the hippocampus. In addition, it is the first time that CREB was evaluated in amygdala and hypothalamus of animals with DM, who presented a higher expression. Further research is necessary to determine the possible link between expression of CaMKIV and CREB, and the behavioral profile of anxiety in diabetic animals.
Collapse
Affiliation(s)
- Camila Espasandín
- Departamento de Biociencias Veterinarias, Facultad de Veterinaria, Universidad de la República, Lasplaces 1550, 11600, Montevideo, CP, Uruguay
- Laboratorio de Señalización Celular y Nanobiología, Instituto de Investigaciones Biológicas Clemente Estable, Avenida Italia 3318, 11600, Montevideo, CP, Uruguay
| | - Sofía Rivero
- Departamento de Biociencias Veterinarias, Facultad de Veterinaria, Universidad de la República, Lasplaces 1550, 11600, Montevideo, CP, Uruguay
| | - Laura Bengoa
- Departamento de Biociencias Veterinarias, Facultad de Veterinaria, Universidad de la República, Lasplaces 1550, 11600, Montevideo, CP, Uruguay
| | - Karina Cal
- Departamento de Biociencias Veterinarias, Facultad de Veterinaria, Universidad de la República, Lasplaces 1550, 11600, Montevideo, CP, Uruguay
- Laboratorio de Patologías del Metabolismo y el Envejecimiento, Institut Pasteur Montevideo, Mataojo 2020, 11400, Montevideo, CP, Uruguay
| | - Gerardo Romanelli
- Laboratorio de Señalización Celular y Nanobiología, Instituto de Investigaciones Biológicas Clemente Estable, Avenida Italia 3318, 11600, Montevideo, CP, Uruguay
| | - Juan Claudio Benech
- Laboratorio de Señalización Celular y Nanobiología, Instituto de Investigaciones Biológicas Clemente Estable, Avenida Italia 3318, 11600, Montevideo, CP, Uruguay
| | - Juan Pablo Damián
- Departamento de Biociencias Veterinarias, Facultad de Veterinaria, Universidad de la República, Lasplaces 1550, 11600, Montevideo, CP, Uruguay.
| |
Collapse
|
17
|
Propolis Promotes Memantine-Dependent Rescue of Cognitive Deficits in APP-KI Mice. Mol Neurobiol 2022; 59:4630-4646. [PMID: 35587310 DOI: 10.1007/s12035-022-02876-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 05/12/2022] [Indexed: 10/18/2022]
Abstract
Propolis is a complex resinous substance that is relevant as a therapeutic target for Alzheimer's disease (AD) and other neurodegenerative diseases. In this study, we confirmed that propolis (Brazilian green propolis) further enhances the rescue of cognitive deficits by the novel AD drug memantine in APP-KI mice. In memory-related behavior tasks, administration of a single dose of propolis at 1-100 mg/kg p.o. significantly enhanced the rescue of cognitive deficits by memantine at 1 mg/kg p.o. in APP-KI mice. In in vitro studies, propolis significantly increased intracellular Ca2+ concentration and calcium/calmodulin-dependent protein kinase II (CaMKII) autophosphorylation in Kir6.2-overexpressed N2A cells treated with memantine. Propolis also significantly increased adenosine 5'-triphosphate (ATP) contents and CaMKII autophosphorylation, which was impaired in Aβ-treated Kir6.2-overexpressed N2A cells. Similarly, repeated administration of propolis at 100 mg/kg p.o. for 8 weeks further enhanced the rescue of cognitive deficits by memantine in APP-KI mice. Consistent with the rescued cognitive deficits in APP-KI mice, repeated administration of propolis markedly ameliorated memantine-dependent rescue of injured long-term potentiation (LTP) in APP-KI mice, concomitant with increased CaMKII autophosphorylation and calcium/calmodulin-dependent protein kinase IV (CaMKIV) phosphorylation in the hippocampal CA1 region. Furthermore, repeated administration of both memantine and propolis significantly restored the decreased ATP contents in the CA1 region of APP-KI mice. Finally, we confirmed that repeated administration of memantine at 1 mg/kg p.o. and propolis at 100 mg/kg p.o. for 8 weeks failed to restore the cognitive deficits in Kir6.2-/- mice. Our study demonstrates that propolis increases ATP contents and promotes the amelioration of cognitive deficits rescued by memantine via Kir6.2 channel inhibition in the CA1 region.
Collapse
|
18
|
Jyoti Dutta B, Singh S, Seksaria S, Das Gupta G, Bodakhe SH, Singh A. Potential role of IP3/Ca 2+ signaling and phosphodiesterases: Relevance to neurodegeneration in Alzheimer's disease and possible therapeutic strategies. Biochem Pharmacol 2022; 201:115071. [PMID: 35525328 DOI: 10.1016/j.bcp.2022.115071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 04/27/2022] [Accepted: 04/28/2022] [Indexed: 11/02/2022]
Abstract
Despite large investments by industry and governments, no disease-modifying medications for the treatment of patients with Alzheimer's disease (AD) have been found. The failures of various clinical trials indicate the need for a more in-depth understanding of the pathophysiology of AD and for innovative therapeutic strategies for its treatment. Here, we review the rational for targeting IP3 signaling, cytosolic calcium dysregulation, phosphodiesterases (PDEs), and secondary messengers like cGMP and cAMP, as well as their correlations with the pathophysiology of AD. Various drugs targeting these signaling cascades are still in pre-clinical and clinical trials which support the ideas presented in this article. Further, we describe different molecular mechanisms and medications currently being used in various pre-clinical and clinical trials involving IP3/Ca+2 signaling. We also highlight various isoforms, as well as the functions and pharmacology of the PDEs broadly expressed in different parts of the brain and attempt to unravel the potential benefits of PDE inhibitors for use as novel medications to alleviate the pathogenesis of AD.
Collapse
Affiliation(s)
- Bhaskar Jyoti Dutta
- Department of Pharmacology, ISF College of Pharmacy, GT Road, Ghal Kalan, Moga-142001, Punjab, India
| | - Shamsher Singh
- Department of Pharmacology, ISF College of Pharmacy, GT Road, Ghal Kalan, Moga-142001, Punjab, India
| | - Sanket Seksaria
- Department of Pharmacology, ISF College of Pharmacy, GT Road, Ghal Kalan, Moga-142001, Punjab, India
| | - Ghanshyam Das Gupta
- Department of Pharmacology, ISF College of Pharmacy, GT Road, Ghal Kalan, Moga-142001, Punjab, India
| | - Surendra H Bodakhe
- Department of Pharmacy, Guru Ghasidas Vishwavidyalaya (A Central University), Bilaspur - 495009, Chhattisgarh, India
| | - Amrita Singh
- Department of Pharmacology, ISF College of Pharmacy, GT Road, Ghal Kalan, Moga-142001, Punjab, India.
| |
Collapse
|
19
|
Vickovic S, Lötstedt B, Klughammer J, Mages S, Segerstolpe Å, Rozenblatt-Rosen O, Regev A. SM-Omics is an automated platform for high-throughput spatial multi-omics. Nat Commun 2022; 13:795. [PMID: 35145087 PMCID: PMC8831571 DOI: 10.1038/s41467-022-28445-y] [Citation(s) in RCA: 81] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Accepted: 01/24/2022] [Indexed: 12/12/2022] Open
Abstract
The spatial organization of cells and molecules plays a key role in tissue function in homeostasis and disease. Spatial transcriptomics has recently emerged as a key technique to capture and positionally barcode RNAs directly in tissues. Here, we advance the application of spatial transcriptomics at scale, by presenting Spatial Multi-Omics (SM-Omics) as a fully automated, high-throughput all-sequencing based platform for combined and spatially resolved transcriptomics and antibody-based protein measurements. SM-Omics uses DNA-barcoded antibodies, immunofluorescence or a combination thereof, to scale and combine spatial transcriptomics and spatial antibody-based multiplex protein detection. SM-Omics allows processing of up to 64 in situ spatial reactions or up to 96 sequencing-ready libraries, of high complexity, in a ~2 days process. We demonstrate SM-Omics in the mouse brain, spleen and colorectal cancer model, showing its broad utility as a high-throughput platform for spatial multi-omics.
Collapse
Affiliation(s)
- S Vickovic
- Klarman Cell Observatory Broad Institute of MIT and Harvard, Cambridge, MA, USA. .,Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA. .,New York Genome Center, New York, NY, USA. .,Science for Life Laboratory, Department of Biochemistry and Biophysics, Stockholm University, Solna, Sweden.
| | - B Lötstedt
- Klarman Cell Observatory Broad Institute of MIT and Harvard, Cambridge, MA, USA.,Science for Life Laboratory, Department of Gene Technology, KTH Royal Institute of Technology, Stockholm, Sweden.,Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - J Klughammer
- Klarman Cell Observatory Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - S Mages
- Klarman Cell Observatory Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Å Segerstolpe
- Klarman Cell Observatory Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - O Rozenblatt-Rosen
- Klarman Cell Observatory Broad Institute of MIT and Harvard, Cambridge, MA, USA.,Genentech, 1 DNA Way, South San Francisco, CA, USA
| | - A Regev
- Klarman Cell Observatory Broad Institute of MIT and Harvard, Cambridge, MA, USA. .,Howard Hughes Medical Institute and Koch Institute for Integrative Cancer Research, Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA. .,Genentech, 1 DNA Way, South San Francisco, CA, USA.
| |
Collapse
|
20
|
Zhang R, Gao Y, Li Y, Geng D, Liang Y, He Q, Wang L, Cui H. Nrf2 improves hippocampal synaptic plasticity, learning and memory through the circ-Vps41/miR-26a-5p/CaMKIV regulatory network. Exp Neurol 2022; 351:113998. [PMID: 35143833 DOI: 10.1016/j.expneurol.2022.113998] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 01/21/2022] [Accepted: 02/01/2022] [Indexed: 12/24/2022]
Abstract
Antioxidant response transcription factor nuclear factor erythroid-2-related factor 2 (Nrf2/Nfe2l2) is a neuroprotective agent in learning and memory impairment. This study provides a new perspective to explore the regulatory mechanisms of Nrf2. Here, we found that Nrf2 regulated circular RNA circ-Vps41 to increase hippocampal synaptic plasticity; Nrf2 bound the Vps41 promoter to activate transcription of the Vps41 gene and promote expression of circ-Vps41; circ-Vps41 positively correlated with Nrf2, synaptic plasticity, and learning and memory but negatively correlated with reactive oxygen species; and Nrf2 promoted CaMKIV expression by increasing levels of circ-Vps41, which can absorb miR-26a-5p that targets CaMKIV. Our findings revealed a new circRNA-based regulatory network regulated by Nrf2 and provided novel insights into the potential mechanism involved in the improvement of learning and memory impairment.
Collapse
Affiliation(s)
- Runjiao Zhang
- Department of Human Anatomy, Hebei Medical University, Shijiazhuang, Hebei 050017, PR China; Institute of Medicine and Health, Hebei Medical University, Shijiazhuang, Hebei 050017, PR China
| | - Yanjing Gao
- Department of Human Anatomy, Hebei Medical University, Shijiazhuang, Hebei 050017, PR China; Institute of Medicine and Health, Hebei Medical University, Shijiazhuang, Hebei 050017, PR China
| | - Yibo Li
- Department of Human Anatomy, Hebei Medical University, Shijiazhuang, Hebei 050017, PR China; Institute of Medicine and Health, Hebei Medical University, Shijiazhuang, Hebei 050017, PR China
| | - Dandan Geng
- Department of Human Anatomy, Hebei Medical University, Shijiazhuang, Hebei 050017, PR China; Institute of Medicine and Health, Hebei Medical University, Shijiazhuang, Hebei 050017, PR China; The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, Hebei 050017, PR China
| | - Yuxiang Liang
- School of Basic Medicine, Hebei Medical University, Shijiazhuang, Hebei 050017, PR China
| | - Qingwen He
- School of Basic Medicine, Hebei Medical University, Shijiazhuang, Hebei 050017, PR China
| | - Lei Wang
- Department of Human Anatomy, Hebei Medical University, Shijiazhuang, Hebei 050017, PR China; Institute of Medicine and Health, Hebei Medical University, Shijiazhuang, Hebei 050017, PR China; The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, Hebei 050017, PR China.
| | - Huixian Cui
- Department of Human Anatomy, Hebei Medical University, Shijiazhuang, Hebei 050017, PR China; Institute of Medicine and Health, Hebei Medical University, Shijiazhuang, Hebei 050017, PR China; National University of Ireland Galway Stem Cell Research Center, Hebei Medical University, Shijiazhuang, Hebei 050017, PR China.
| |
Collapse
|
21
|
Stojanovic T, Velarde Gamez D, Schuld GJ, Bormann D, Cabatic M, Uhrin P, Lubec G, Monje FJ. Age-Dependent and Pathway-Specific Bimodal Action of Nicotine on Synaptic Plasticity in the Hippocampus of Mice Lacking the miR-132/212 Genes. Cells 2022; 11:261. [PMID: 35053378 PMCID: PMC8774101 DOI: 10.3390/cells11020261] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2021] [Revised: 01/06/2022] [Accepted: 01/11/2022] [Indexed: 12/19/2022] Open
Abstract
Nicotine addiction develops predominantly during human adolescence through smoking. Self-administration experiments in rodents verify this biological preponderance to adolescence, suggesting evolutionary-conserved and age-defined mechanisms which influence the susceptibility to nicotine addiction. The hippocampus, a brain region linked to drug-related memory storage, undergoes major morpho-functional restructuring during adolescence and is strongly affected by nicotine stimulation. However, the signaling mechanisms shaping the effects of nicotine in young vs. adult brains remain unclear. MicroRNAs (miRNAs) emerged recently as modulators of brain neuroplasticity, learning and memory, and addiction. Nevertheless, the age-dependent interplay between miRNAs regulation and hippocampal nicotinergic signaling remains poorly explored. We here combined biophysical and pharmacological methods to examine the impact of miRNA-132/212 gene-deletion (miRNA-132/212-/-) and nicotine stimulation on synaptic functions in adolescent and mature adult mice at two hippocampal synaptic circuits: the medial perforant pathway (MPP) to dentate yrus (DG) synapses (MPP-DG) and CA3 Schaffer collaterals to CA1 synapses (CA3-CA1). Basal synaptic transmission and short-term (paired-pulse-induced) synaptic plasticity was unaltered in adolescent and adult miRNA-132/212-/- mice hippocampi, compared with wild-type controls. However, nicotine stimulation promoted CA3-CA1 synaptic potentiation in mature adult (not adolescent) wild-type and suppressed MPP-DG synaptic potentiation in miRNA-132/212-/- mice. Altered levels of CREB, Phospho-CREB, and acetylcholinesterase (AChE) expression were further detected in adult miRNA-132/212-/- mice hippocampi. These observations propose miRNAs as age-sensitive bimodal regulators of hippocampal nicotinergic signaling and, given the relevance of the hippocampus for drug-related memory storage, encourage further research on the influence of miRNAs 132 and 212 in nicotine addiction in the young and the adult brain.
Collapse
Affiliation(s)
- Tamara Stojanovic
- Center for Physiology and Pharmacology, Department of Neurophysiology and Neuropharmacology, Medical University of Vienna, 1090 Vienna, Austria; (D.V.G.); (G.J.S.); (D.B.); (M.C.)
| | - David Velarde Gamez
- Center for Physiology and Pharmacology, Department of Neurophysiology and Neuropharmacology, Medical University of Vienna, 1090 Vienna, Austria; (D.V.G.); (G.J.S.); (D.B.); (M.C.)
| | - Gabor Jorrid Schuld
- Center for Physiology and Pharmacology, Department of Neurophysiology and Neuropharmacology, Medical University of Vienna, 1090 Vienna, Austria; (D.V.G.); (G.J.S.); (D.B.); (M.C.)
| | - Daniel Bormann
- Center for Physiology and Pharmacology, Department of Neurophysiology and Neuropharmacology, Medical University of Vienna, 1090 Vienna, Austria; (D.V.G.); (G.J.S.); (D.B.); (M.C.)
- Laboratory for Cardiac and Thoracic Diagnosis, Department of Surgery, Regeneration and Applied Immunology, Medical University of Vienna, Research Laboratories Vienna General Hospital, Waehringer Guertel 18-20, 1090 Vienna, Austria
- Division of Thoracic Surgery, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| | - Maureen Cabatic
- Center for Physiology and Pharmacology, Department of Neurophysiology and Neuropharmacology, Medical University of Vienna, 1090 Vienna, Austria; (D.V.G.); (G.J.S.); (D.B.); (M.C.)
| | - Pavel Uhrin
- Center for Physiology and Pharmacology, Department of Vascular Biology and Thrombosis Research, Medical University of Vienna, 1090 Vienna, Austria;
| | - Gert Lubec
- Department of Neuroproteomics, Paracelsus Medical University, 5020 Salzburg, Austria;
| | - Francisco J. Monje
- Center for Physiology and Pharmacology, Department of Neurophysiology and Neuropharmacology, Medical University of Vienna, 1090 Vienna, Austria; (D.V.G.); (G.J.S.); (D.B.); (M.C.)
| |
Collapse
|
22
|
Zheng Z, Wang X, Wang Y, King JAC, Xie P, Wu S. CaMK4 is a downstream effector of the α 1G T-type calcium channel to determine the angiogenic potential of pulmonary microvascular endothelial cells. Am J Physiol Cell Physiol 2021; 321:C964-C977. [PMID: 34586897 DOI: 10.1152/ajpcell.00216.2021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Accepted: 09/27/2021] [Indexed: 01/25/2023]
Abstract
Pulmonary microvascular endothelial cells (PMVECs) uniquely express an α1G-subtype of voltage-gated T-type Ca2+ channel. We have previously revealed that the α1G channel functions as a background Ca2+ entry pathway that is critical for the cell proliferation, migration, and angiogenic potential of PMVECs, a novel function attributed to the coupling between α1G-mediated Ca2+ entry and constitutive Akt phosphorylation and activation. Despite this significance, mechanism(s) that link the α1G-mediated Ca2+ entry to Akt phosphorylation remain incompletely understood. In this study, we demonstrate that Ca2+/calmodulin-dependent protein kinase (CaMK) 4 serves as a downstream effector of the α1G-mediated Ca2+ entry to promote the angiogenic potential of PMVECs. Notably, CaMK2 and CaMK4 are both expressed in PMVECs. Pharmacological blockade or genetic knockdown of the α1G channel led to a significant reduction in the phosphorylation level of CaMK4 but not the phosphorylation level of CaMK2. Pharmacological inhibition as well as genetic knockdown of CaMK4 significantly decreased cell proliferation, migration, and network formation capacity in PMVECs. However, CaMK4 inhibition or knockdown did not alter Akt phosphorylation status in PMVECs, indicating that α1G/Ca2+/CaMK4 is independent of the α1G/Ca2+/Akt pathway in sustaining the cells' angiogenic potential. Altogether, these findings suggest a novel α1G-CaMK4 signaling complex that regulates the Ca2+-dominated angiogenic potential in PMVECs.
Collapse
Affiliation(s)
- Zhen Zheng
- Department of Anesthesiology and Perioperative Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Xuelin Wang
- Department of Respiratory and Critical Care Medicine, Henan Provincial People's Hospital, Zhengzhou, China
| | - Yuxia Wang
- Department of Anesthesiology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Judy A C King
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center - Shreveport, Shreveport, Louisiana
| | - Peilin Xie
- Department of Anesthesiology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Songwei Wu
- Department of Anesthesiology and Perioperative Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| |
Collapse
|
23
|
Cascella R, Cecchi C. Calcium Dyshomeostasis in Alzheimer's Disease Pathogenesis. Int J Mol Sci 2021; 22:ijms22094914. [PMID: 34066371 PMCID: PMC8124842 DOI: 10.3390/ijms22094914] [Citation(s) in RCA: 79] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 04/26/2021] [Accepted: 04/30/2021] [Indexed: 01/12/2023] Open
Abstract
Alzheimer’s disease (AD) is the most common age-related neurodegenerative disorder that is characterized by amyloid β-protein deposition in senile plaques, neurofibrillary tangles consisting of abnormally phosphorylated tau protein, and neuronal loss leading to cognitive decline and dementia. Despite extensive research, the exact mechanisms underlying AD remain unknown and effective treatment is not available. Many hypotheses have been proposed to explain AD pathophysiology; however, there is general consensus that the abnormal aggregation of the amyloid β peptide (Aβ) is the initial event triggering a pathogenic cascade of degenerating events in cholinergic neurons. The dysregulation of calcium homeostasis has been studied considerably to clarify the mechanisms of neurodegeneration induced by Aβ. Intracellular calcium acts as a second messenger and plays a key role in the regulation of neuronal functions, such as neural growth and differentiation, action potential, and synaptic plasticity. The calcium hypothesis of AD posits that activation of the amyloidogenic pathway affects neuronal Ca2+ homeostasis and the mechanisms responsible for learning and memory. Aβ can disrupt Ca2+ signaling through several mechanisms, by increasing the influx of Ca2+ from the extracellular space and by activating its release from intracellular stores. Here, we review the different molecular mechanisms and receptors involved in calcium dysregulation in AD and possible therapeutic strategies for improving the treatment.
Collapse
|
24
|
The Calcium/Calmodulin-Dependent Kinases II and IV as Therapeutic Targets in Neurodegenerative and Neuropsychiatric Disorders. Int J Mol Sci 2021; 22:ijms22094307. [PMID: 33919163 PMCID: PMC8122486 DOI: 10.3390/ijms22094307] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 04/13/2021] [Accepted: 04/17/2021] [Indexed: 12/14/2022] Open
Abstract
CaMKII and CaMKIV are calcium/calmodulin-dependent kinases playing a rudimentary role in many regulatory processes in the organism. These kinases attract increasing interest due to their involvement primarily in memory and plasticity and various cellular functions. Although CaMKII and CaMKIV are mostly recognized as the important cogs in a memory machine, little is known about their effect on mood and role in neuropsychiatric diseases etiology. Here, we aimed to review the structure and functions of CaMKII and CaMKIV, as well as how these kinases modulate the animals’ behavior to promote antidepressant-like, anxiolytic-like, and procognitive effects. The review will help in the understanding of the roles of the above kinases in the selected neurodegenerative and neuropsychiatric disorders, and this knowledge can be used in future drug design.
Collapse
|
25
|
Xing B, Mack NR, Guo KM, Zhang YX, Ramirez B, Yang SS, Lin L, Wang DV, Li YC, Gao WJ. A Subpopulation of Prefrontal Cortical Neurons Is Required for Social Memory. Biol Psychiatry 2021; 89:521-531. [PMID: 33190846 PMCID: PMC7867585 DOI: 10.1016/j.biopsych.2020.08.023] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2020] [Revised: 08/12/2020] [Accepted: 08/24/2020] [Indexed: 02/02/2023]
Abstract
BACKGROUND The medial prefrontal cortex (mPFC) is essential for social behaviors, yet whether and how it encodes social memory remains unclear. METHODS We combined whole-cell patch recording, morphological analysis, optogenetic/chemogenetic manipulation, and the TRAP (targeted recombination in active populations) transgenic mouse tool to study the social-associated neural populations in the mPFC. RESULTS Fos-TRAPed prefrontal social-associated neurons are excitatory pyramidal neurons with relatively small soma sizes and thin-tufted apical dendrite. These cells exhibit intrinsic firing features of dopamine D1 receptor-like neurons, show persisting firing pattern after social investigation, and project dense axons to nucleus accumbens. In behaving TRAP mice, selective inhibition of prefrontal social-associated neurons does not affect social investigation but does impair subsequent social recognition, whereas optogenetic reactivation of their projections to the nucleus accumbens enables recall of a previously encountered but "forgotten" mouse. Moreover, chemogenetic activation of mPFC-to-nucleus accumbens projections ameliorates MK-801-induced social memory impairments. CONCLUSIONS Our results characterize the electrophysiological and morphological features of social-associated neurons in the mPFC and indicate that these Fos-labeled, social-activated prefrontal neurons are necessary and sufficient for social memory.
Collapse
Affiliation(s)
- Bo Xing
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, Pennsylvania
| | - Nancy R Mack
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, Pennsylvania
| | - Kai-Ming Guo
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China; Department of Pharmacology, University of Virginia School of Medicine, Charlottesville, Virginia
| | - Yu-Xiang Zhang
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, Pennsylvania
| | - Billy Ramirez
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, Pennsylvania
| | - Sha-Sha Yang
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, Pennsylvania
| | - Li Lin
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China.
| | - Dong V Wang
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, Pennsylvania
| | - Yan-Chun Li
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, Pennsylvania
| | - Wen-Jun Gao
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, Pennsylvania.
| |
Collapse
|
26
|
Yamamoto Y, Owada Y. Possible involvement of fatty acid binding proteins in psychiatric disorders. Anat Sci Int 2021; 96:333-342. [PMID: 33604770 DOI: 10.1007/s12565-020-00598-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 12/26/2020] [Indexed: 12/19/2022]
Abstract
Polyunsaturated fatty acids (PUFAs) are essential for brain development and function. Increasing evidence has shown that an imbalance of PUFAs is associated with various human psychiatric disorders, including autism and schizophrenia. However, the mechanisms underlying the effects of PUFAs on brain functions at cellular and molecular levels remain unclear. Since PUFAs are insoluble in water, specific transporters are required to deliver PUFAs to appropriate intracellular compartments. Fatty acid-binding proteins (FABPs), the cellular chaperones of PUFAs, are involved in PUFA intracellular trafficking, signal transduction, and gene transcription. Therefore, we focused on the relationship between FABP-regulated PUFA homeostasis in the brain and neuronal plasticity. The authors previously reported that FABP3, which preferentially binds to n-6 PUFAs, is strongly expressed in the gamma-aminobutyric acid (GABAergic) inhibitory interneurons of the adult mouse anterior cingulate cortex (ACC), which is a component of the limbic cortex and is important for the coordination of cognitive and emotional behaviors. Interestingly, Fabp3 KO mice show increased GABA synthesis and abnormal excitatory/inhibitory balance in the ACC. In addition, studies have indicated that FABP7, which preferentially binds to n-3 PUFAs, controls lipid raft function in astrocytes, and astrocytic Fabp7 deficiency results in an altered response of astrocytes to external stimuli. Furthermore, Fabp7 KO mice exhibit aberrant dendritic morphology, and decreased spine density and excitatory synaptic transmission in pyramidal neurons. This review summarizes relationship between PUFAs or FABPs and human psychiatric disorders and discusses recent progress in elucidating the function of FABPs, especially FABP3 and 7, in the brain.
Collapse
Affiliation(s)
- Yui Yamamoto
- Department of Organ Anatomy, Tohoku University, Seiryo-machi Aoba-ku, Sendai, 980-8575, Japan. .,Department of Anatomy, Tohoku Medical and Pharmaceutical University, Fukumuro Miyagino-ku, Sendai, 980-8578, Japan.
| | - Yuji Owada
- Department of Organ Anatomy, Tohoku University, Seiryo-machi Aoba-ku, Sendai, 980-8575, Japan
| |
Collapse
|
27
|
Ye T, Gao HW, Xuan WT, Ye S, Zhou P, Li XQ, Wang Y, Song H, Liu YY, Cai B. The Regulating Mechanism of Chrysophanol on Protein Level of CaM-CaMKIV to Protect PC12 Cells Against Aβ 25-35-Induced Damage. DRUG DESIGN DEVELOPMENT AND THERAPY 2020; 14:2715-2723. [PMID: 32764873 PMCID: PMC7381772 DOI: 10.2147/dddt.s245128] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Accepted: 06/30/2020] [Indexed: 12/14/2022]
Abstract
Objective To investigate the neuroprotective effect of chrysophanol (CHR) on PC12 treated with Aβ25-35, and the involved mechanism. Methods After the establishment of an AD cell model induced by Aβ25-35, the cell survival rate was detected by MTT, cell apoptosis was assayed by Hoechst 33342 staining, mRNA expressions of calmodulin (CaM), calcium/calmodulin-dependent protein kinase kinase (CaMKK), calcium/calmodulin-dependent protein kinase IV (CaMKIV) and tau (MAPT; commonly known as tau) were determined by qRT-PCR, and protein levels of CaM, CaMKK, CaMKIV, phospho-CaMKIV (p-CaMKIV), tau and phospho-tau (p-tau) were detected by Western blot analysis. Results When pretreated with CHR before exposure to Aβ25-35, PC12 cells showed that increased cell viability and reduced apoptosis. The qRT-PCR results indicated that the deposition of Aβ25-35 triggers a decrease in levels of CaM, CaMKK, CaMKIV, and tau in PC12 cells. In addition, Western blot results also suggested that Aβ25-35 decreases the protein expression of CaM, CaMKK, CaMKIV, p-CaMKIV, and the ratio of p-tau to tau in PC12 cells. However, the above effects were significantly alleviated after the treatment of CHR. Conclusion CHR plays a neuroprotective role in AD though decreasing the protein level of CaM-CaMKK-CaMKIV and the expression of p-tau downstream.
Collapse
Affiliation(s)
- Ting Ye
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei 230012, People's Republic of China
| | - Hua-Wu Gao
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei 230012, People's Republic of China
| | - Wei-Ting Xuan
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei 230012, People's Republic of China
| | - Shu Ye
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei 230012, People's Republic of China.,Institute of Integrated Chinese and Western Medicine, Anhui Academy of Chinese Medicine, Hefei 230012, People's Republic of China.,Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei 230012, People's Republic of China
| | - Peng Zhou
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei 230012, People's Republic of China.,Institute of Integrated Chinese and Western Medicine, Anhui Academy of Chinese Medicine, Hefei 230012, People's Republic of China.,Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei 230012, People's Republic of China
| | - Xin-Quan Li
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei 230012, People's Republic of China
| | - Yan Wang
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei 230012, People's Republic of China.,Institute of Integrated Chinese and Western Medicine, Anhui Academy of Chinese Medicine, Hefei 230012, People's Republic of China.,Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei 230012, People's Republic of China
| | - Hang Song
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei 230012, People's Republic of China.,Institute of Integrated Chinese and Western Medicine, Anhui Academy of Chinese Medicine, Hefei 230012, People's Republic of China
| | - Yan-Yan Liu
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei 230012, People's Republic of China.,Institute of Integrated Chinese and Western Medicine, Anhui Academy of Chinese Medicine, Hefei 230012, People's Republic of China
| | - Biao Cai
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei 230012, People's Republic of China.,Institute of Integrated Chinese and Western Medicine, Anhui Academy of Chinese Medicine, Hefei 230012, People's Republic of China.,Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei 230012, People's Republic of China
| |
Collapse
|
28
|
Memantine Improves Depressive-like Behaviors via Kir6.1 Channel Inhibition in Olfactory Bulbectomized Mice. Neuroscience 2020; 442:264-273. [PMID: 32531473 DOI: 10.1016/j.neuroscience.2020.06.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 05/29/2020] [Accepted: 06/02/2020] [Indexed: 12/27/2022]
Abstract
Aberrant depressive-like behaviors in olfactory bulbectomized (OBX) mice have been documented by previous studies. Here, we show that memantine enhances adult neurogenesis in the subgranular zone of the hippocampal dentate gyrus (DG) and improves depressive-like behaviors via inhibition of the ATP-sensitive potassium (KATP) channel in OBX mice. Treatment with memantine (1-3 mg/kg; per os (p.o.)) for 14 days significantly improved depressive-like behaviors in OBX mice, as assessed using the tail-suspension and forced-swim tests. Treatment with memantine also increased the number of BrdU-positive neurons in the DG of OBX mice. In the immunoblot analysis, memantine significantly increased phosphorylation of CaMKIV (Thr-196) and Akt (Ser-473), but not ERK (Thr-202/Tyr-204), in the DG of OBX mice. Furthermore, phosphorylation of GSK3β (Ser-9) and CREB (Ser-133), and BDNF protein expression levels increased in the DG of OBX mice, possibly accounting for the increased adult neurogenesis owing to Akt activation. In contrast, both the improvement of depressive-like behaviors and increase in BrdU-positive neurons in the DG following treatment with memantine were unapparent in OBX-treated Kir6.1 heterozygous (+/-) mice but not OBX-treated Kir6.2 heterozygous (+/-) mice. Furthermore, the increase in CaMKIV (Thr-196) and Akt (Ser-473) phosphorylation and BDNF protein expression levels was not observed in OBX-treated Kir6.1 +/- mice. Overall, our study shows that memantine improves OBX-induced depressive-like behaviors by increasing adult neurogenesis in the DG via Kir6.1 channel inhibition.
Collapse
|
29
|
Jebelli A, Khalaj-Kondori M, Rahmati-Yamchi M. The Effect of Beta-Boswellic Acid on the Expression of Camk4 and Camk2α Genes in the PC12 Cell Line. Adv Pharm Bull 2020; 10:437-443. [PMID: 32665903 PMCID: PMC7335997 DOI: 10.34172/apb.2020.053] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 11/28/2019] [Accepted: 12/08/2019] [Indexed: 12/18/2022] Open
Abstract
Purpose: Beta-boswellic acid (βBA) may play central roles in neural plasticity. Neural plasticity has significant implications for learning and memory which are governed by strict memoryrelated molecular pathways. To gain insight into the molecular mechanism by which βBA affects these pathways this study analyzed the expression patterns of Camk2α and Camk4 genes in PC12 cells treated with βBA. Methods: The cytotoxic effects of different βBA concentrations on PC12 cells were examined by MTT assay. For gene expression analysis, cells were treated with concentrations of 1 and 10 µM of βBA for 12, 24, 48, and 72 hours. Total RNA was purified by RNX-Plus solution and reverse transcribed into cDNA using Thermo Scientific Reverse Transcription reagents. The expression patterns of Camk2α and Camk4 genes were quantified by quantitative reverse transcription polymerase chain reaction (qRT-PCR). Results: MTT assay indicated that βBA reduced PC12 cell viability in a time- and concentrationdependent manner. The 50% inhibitory concentrations for the 48 and 72 hours time points were 35 and 26 µM, respectively; while, the βBA concentrations up to 100 µM failed to kill 50% of the cells after 24 hours. According to the qRT-PCR data, the Camk2α variant is not expressed in either βBA-treated or untreated PC12 cells. However, a significant upregulation was observed inCamk4 after 12 hours of treatment with βBA, which followed by a significant downregulation after 24 hours and a persistent expression equal to the control until 72 hours. Conclusion: these findings indicate that PC12 cells not only does not express Camk2α but also its expression cannot be induced by βBA. However, βBA does modulate the expression of Camk4. This result provides further insight into the molecular mechanism by which βBA affects memory.
Collapse
Affiliation(s)
- Asiyeh Jebelli
- Department of Animal Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
| | - Mohammad Khalaj-Kondori
- Department of Animal Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
| | - Mohammad Rahmati-Yamchi
- Department of Clinical Biochemistry, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.,Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
30
|
Hagenston AM, Bading H, Bas-Orth C. Functional Consequences of Calcium-Dependent Synapse-to-Nucleus Communication: Focus on Transcription-Dependent Metabolic Plasticity. Cold Spring Harb Perspect Biol 2020; 12:cshperspect.a035287. [PMID: 31570333 DOI: 10.1101/cshperspect.a035287] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
In the nervous system, calcium signals play a major role in the conversion of synaptic stimuli into transcriptional responses. Signal-regulated gene transcription is fundamental for a range of long-lasting adaptive brain functions that include learning and memory, structural plasticity of neurites and synapses, acquired neuroprotection, chronic pain, and addiction. In this review, we summarize the diverse mechanisms governing calcium-dependent transcriptional regulation associated with central nervous system plasticity. We focus on recent advances in the field of synapse-to-nucleus communication that include studies of the signal-regulated transcriptome in human neurons, identification of novel regulatory mechanisms such as activity-induced DNA double-strand breaks, and the identification of novel forms of activity- and transcription-dependent adaptations, in particular, metabolic plasticity. We summarize the reciprocal interactions between different kinds of neuroadaptations and highlight the emerging role of activity-regulated epigenetic modifiers in gating the inducibility of signal-regulated genes.
Collapse
Affiliation(s)
- Anna M Hagenston
- Department of Neurobiology, Interdisciplinary Center for Neurosciences (IZN), Heidelberg University, 69120 Heidelberg, Germany
| | - Hilmar Bading
- Department of Neurobiology, Interdisciplinary Center for Neurosciences (IZN), Heidelberg University, 69120 Heidelberg, Germany
| | - Carlos Bas-Orth
- Department of Medical Cell Biology, Institute for Anatomy and Cell Biology, Heidelberg University, 69120 Heidelberg, Germany
| |
Collapse
|
31
|
Navabpour S, Kwapis JL, Jarome TJ. A neuroscientist's guide to transgenic mice and other genetic tools. Neurosci Biobehav Rev 2020; 108:732-748. [PMID: 31843544 PMCID: PMC8049509 DOI: 10.1016/j.neubiorev.2019.12.013] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 11/05/2019] [Accepted: 12/09/2019] [Indexed: 12/14/2022]
Abstract
The past decade has produced an explosion in the number and variety of genetic tools available to neuroscientists, resulting in an unprecedented ability to precisely manipulate the genome and epigenome in behaving animals. However, no single resource exists that describes all of the tools available to neuroscientists. Here, we review the genetic, transgenic, and viral techniques that are currently available to probe the complex relationship between genes and cognition. Topics covered include types of traditional transgenic mouse models (knockout, knock-in, reporter lines), inducible systems (Cre-loxP, Tet-On, Tet-Off) and cell- and circuit-specific systems (TetTag, TRAP, DIO-DREADD). Additionally, we provide details on virus-mediated and siRNA/shRNA approaches, as well as a comprehensive discussion of the myriad manipulations that can be made using the CRISPR-Cas9 system, including single base pair editing and spatially- and temporally-regulated gene-specific transcriptional control. Collectively, this review will serve as a guide to assist neuroscientists in identifying and choosing the appropriate genetic tools available to study the complex relationship between the brain and behavior.
Collapse
Affiliation(s)
- Shaghayegh Navabpour
- Fralin Biomedical Research Institute, Translational Biology, Medicine and Health, Roanoke, VA, USA
| | - Janine L Kwapis
- Department of Biology, Pennsylvania State University, College Park, PA, USA; Center for the Molecular Investigation of Neurological Disorders (CMIND), Pennsylvania State University, College Park, PA, USA.
| | - Timothy J Jarome
- Fralin Biomedical Research Institute, Translational Biology, Medicine and Health, Roanoke, VA, USA; Department of Animal and Poultry Sciences, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA; School of Neuroscience, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA.
| |
Collapse
|
32
|
Association of genes with phenotype in autism spectrum disorder. Aging (Albany NY) 2019; 11:10742-10770. [PMID: 31744938 PMCID: PMC6914398 DOI: 10.18632/aging.102473] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Accepted: 11/08/2019] [Indexed: 12/27/2022]
Abstract
Autism spectrum disorder (ASD) is a genetic heterogeneous neurodevelopmental disorder that is characterized by impairments in social interaction and speech development and is accompanied by stereotypical behaviors such as body rocking, hand flapping, spinning objects, sniffing and restricted behaviors. The considerable significance of the genetics associated with autism has led to the identification of many risk genes for ASD used for the probing of ASD specificity and shared cognitive features over the past few decades. Identification of ASD risk genes helps to unravel various genetic variants and signaling pathways which are involved in ASD. This review highlights the role of ASD risk genes in gene transcription and translation regulation processes, as well as neuronal activity modulation, synaptic plasticity, disrupted key biological signaling pathways, and the novel candidate genes that play a significant role in the pathophysiology of ASD. The current emphasis on autism spectrum disorders has generated new opportunities in the field of neuroscience, and further advancements in the identification of different biomarkers, risk genes, and genetic pathways can help in the early diagnosis and development of new clinical and pharmacological treatments for ASD.
Collapse
|
33
|
Sushma, Mondal AC. Role of GPCR signaling and calcium dysregulation in Alzheimer's disease. Mol Cell Neurosci 2019; 101:103414. [PMID: 31655116 DOI: 10.1016/j.mcn.2019.103414] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 09/17/2019] [Accepted: 09/24/2019] [Indexed: 12/12/2022] Open
Abstract
Alzheimer's disease (AD), a late onset neurodegenerative disorder is characterized by the loss of memory, disordered cognitive function, caused by accumulation of amyloid-β (Aβ) peptide and neurofibrillary tangles (NFTs) in the neocortex and hippocampal brain area. Extensive research has been done on the findings of the disease etiology or pathological causes of aggregation of Aβ and hyperphosphorylation of tau protein without much promising results. Recently, calcium dysregulation has been reported to play an important role in the pathophysiology of AD. Calcium ion acts as one of the major secondary messengers, regulates many signaling pathways involved in cell survival, proliferation, differentiation, transcription and apoptosis. Calcium signaling is one of the major signaling pathways involved in the formation of memory, generation of energy and other physiological functions. It also can modulate function of many proteins upon binding. Dysregulation in calcium homeostasis leads to many physiological changes leading to neurodegenerative diseases including AD. In AD, GPCRs generate secondary messengers which regulate calcium homeostasis inside the cell and is reported to be disturbed in the pathological condition. Calcium channels and receptors present on the plasma membrane and intracellular organelle maintain calcium homeostasis through different signaling mechanisms. In this review, we have summarized the different calcium channels and receptors involved in calcium dysregulation which in turn play a critical role in the pathogenesis of AD. Understanding the role of calcium channels and GPCRs to maintain calcium homeostasis is an attempt to develop effective AD treatments.
Collapse
Affiliation(s)
- Sushma
- Laboratory of Cellular and Molecular Neurobiology, School of Life Sciences, Jawaharlal Nehru University, New Delhi, Delhi, India
| | - Amal Chandra Mondal
- Laboratory of Cellular and Molecular Neurobiology, School of Life Sciences, Jawaharlal Nehru University, New Delhi, Delhi, India.
| |
Collapse
|
34
|
Duda P, Wójcicka O, Wiśniewski JR, Rakus D. Global quantitative TPA-based proteomics of mouse brain structures reveals significant alterations in expression of proteins involved in neuronal plasticity during aging. Aging (Albany NY) 2019; 10:1682-1697. [PMID: 30026405 PMCID: PMC6075443 DOI: 10.18632/aging.101501] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Accepted: 07/15/2018] [Indexed: 01/17/2023]
Abstract
Aging is believed to be the result of alterations of protein expression and accumulation of changes in biomolecules. Although there are numerous reports demonstrating changes in protein expression in brain during aging, only few of them describe global changes at the protein level. Here, we present the deepest quantitative proteomic analysis of three brain regions, hippocampus, cortex and cerebellum, in mice aged 1 or 12 months, using the total protein approach technique. In all the brain regions, both in young and middle-aged animals, we quantitatively measured over 5,200 proteins. We found that although the total protein expression in middle-aged brain structures is practically unaffected by aging, there are significant differences between young and middle-aged mice in the expression of some receptors and signaling cascade proteins proven to be significant for learning and memory formation. Our analysis demonstrates that the hippocampus is the most variable structure during natural aging and that the first symptoms of weakening of neuronal plasticity may be observed on protein level in middle-aged animals.
Collapse
Affiliation(s)
- Przemysław Duda
- Department of Molecular Physiology and Neurobiology, University of Wroclaw, Wroclaw 50-137, Poland
| | - Olga Wójcicka
- Department of Molecular Physiology and Neurobiology, University of Wroclaw, Wroclaw 50-137, Poland
| | - Jacek R Wiśniewski
- Department of Proteomics and Signal Transduction, Max-Planck-Institute of Biochemistry, Martinsried 82152, Germany
| | - Dariusz Rakus
- Department of Molecular Physiology and Neurobiology, University of Wroclaw, Wroclaw 50-137, Poland
| |
Collapse
|
35
|
Sabti M, Sasaki K, Gadhi C, Isoda H. Elucidation of the Molecular Mechanism Underlying Lippia citriodora(Lim.)-Induced Relaxation and Anti-Depression. Int J Mol Sci 2019; 20:E3556. [PMID: 31330819 PMCID: PMC6678442 DOI: 10.3390/ijms20143556] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Revised: 07/16/2019] [Accepted: 07/18/2019] [Indexed: 12/12/2022] Open
Abstract
Lippia citriodora ethanolic extract (VEE) and verbascoside (Vs), a phenypropanoid glycoside, have been demonstrated to exert relaxant and anxiolytic properties. However, the molecular mechanisms behind their effects are still unclear. In this work, we studied the effects and action mechanisms of VEE and Vs in vivo and in vitro, on human neurotypic SH-SY5Y cells.TST was conducted on mice treated orally with VEE (25, 50 and 100 mg/Kg), Vs (2.5 and 5 mg/Kg), Bupropion (20 mg/Kg) and Milli-Q water. Higher dose of VEE-treated mice showed an increase of immobility time compared to control groups, indicating an induction of relaxation. This effect was found to be induced by regulation of genes playing key roles in calcium homeostasis (calcium channels), cyclic AMP (cAMP) production and energy metabolism. On the other hand, low doses of VEE and Vs showed an antidepressant-like effect and was confirmed by serotonin, noradrenalin, dopamine and BDNF expressions. Finally, VEE and Vsenhancedcell viability, mitochondrial activity and calcium uptake in vitro confirming in vivo findings. Our results showed induction of relaxation and antidepressant-like effects depending on the administered dose of VEE and Vs, through modulation of cAMP and calcium.
Collapse
Affiliation(s)
- Mouad Sabti
- Alliance for Research on the Mediterranean and North Africa (ARENA), University of Tsukuba, 1-1-1 Tennodai, Tsukuba City 305-8572, Ibaraki, Japan
- Tsukuba Life Science Innovation Program (T-LSI), University of Tsukuba, Tennodai 1-1-1, Tsukuba City 305-8577, Ibaraki, Japan
| | - Kazunori Sasaki
- Alliance for Research on the Mediterranean and North Africa (ARENA), University of Tsukuba, 1-1-1 Tennodai, Tsukuba City 305-8572, Ibaraki, Japan
- Interdisciplinary Research Center for Catalytic Chemistry, National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba 305-8560, Japan
| | - Chemseddoha Gadhi
- Faculty of Sciences Semlalia, Cadi Ayyad University, Avenue Prince MoulayAbdellah, BP 2390, 40000 Marrakesh, Morocco
| | - Hiroko Isoda
- Alliance for Research on the Mediterranean and North Africa (ARENA), University of Tsukuba, 1-1-1 Tennodai, Tsukuba City 305-8572, Ibaraki, Japan.
- Tsukuba Life Science Innovation Program (T-LSI), University of Tsukuba, Tennodai 1-1-1, Tsukuba City 305-8577, Ibaraki, Japan.
- Interdisciplinary Research Center for Catalytic Chemistry, National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba 305-8560, Japan.
| |
Collapse
|
36
|
Koga Y, Tsurumaki H, Aoki-Saito H, Sato M, Yatomi M, Takehara K, Hisada T. Roles of Cyclic AMP Response Element Binding Activation in the ERK1/2 and p38 MAPK Signalling Pathway in Central Nervous System, Cardiovascular System, Osteoclast Differentiation and Mucin and Cytokine Production. Int J Mol Sci 2019; 20:ijms20061346. [PMID: 30884895 PMCID: PMC6470985 DOI: 10.3390/ijms20061346] [Citation(s) in RCA: 96] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2019] [Revised: 03/08/2019] [Accepted: 03/13/2019] [Indexed: 11/26/2022] Open
Abstract
There are many downstream targets of mitogen-activated protein kinase (MAPK) signalling that are involved in neuronal development, cellular differentiation, cell migration, cancer, cardiovascular dysfunction and inflammation via their functions in promoting apoptosis and cell motility and regulating various cytokines. It has been reported that cyclic AMP response element-binding protein (CREB) is phosphorylated and activated by cyclic AMP signalling and calcium/calmodulin kinase. Recent evidence also points to CREB phosphorylation by the MAPK signalling pathway. However, the specific roles of CREB phosphorylation in MAPK signalling have not yet been reviewed in detail. Here, we describe the recent advances in the study of this MAPK-CREB signalling axis in human diseases. Overall, the crosstalk between extracellular signal-related kinase (ERK) 1/2 and p38 MAPK signalling has been shown to regulate various physiological functions, including central nervous system, cardiac fibrosis, alcoholic cardiac fibrosis, osteoclast differentiation, mucin production in the airway, vascular smooth muscle cell migration, steroidogenesis and asthmatic inflammation. In this review, we focus on ERK1/2 and/or p38 MAPK-dependent CREB activation associated with various diseases to provide insights for basic and clinical researchers.
Collapse
Affiliation(s)
- Yasuhiko Koga
- Department of Allergy and Respiratory Medicine, Gunma University Graduate School of Medicine, 3-39-15 sho-wa machi Maebashi, Gunma 371-8511, Japan.
| | - Hiroaki Tsurumaki
- Department of Allergy and Respiratory Medicine, Gunma University Graduate School of Medicine, 3-39-15 sho-wa machi Maebashi, Gunma 371-8511, Japan.
| | - Haruka Aoki-Saito
- Department of Allergy and Respiratory Medicine, Gunma University Graduate School of Medicine, 3-39-15 sho-wa machi Maebashi, Gunma 371-8511, Japan.
| | - Makiko Sato
- Department of Allergy and Respiratory Medicine, Gunma University Graduate School of Medicine, 3-39-15 sho-wa machi Maebashi, Gunma 371-8511, Japan.
| | - Masakiyo Yatomi
- Department of Allergy and Respiratory Medicine, Gunma University Graduate School of Medicine, 3-39-15 sho-wa machi Maebashi, Gunma 371-8511, Japan.
| | - Kazutaka Takehara
- Department of Allergy and Respiratory Medicine, Gunma University Graduate School of Medicine, 3-39-15 sho-wa machi Maebashi, Gunma 371-8511, Japan.
| | - Takeshi Hisada
- Gunma University Graduate School of Health Sciences, 3-39-22 sho-wa machi Maebashi, Gunma 371-8514, Japan.
| |
Collapse
|
37
|
Pagani MR, Merlo E. Kinase and Phosphatase Engagement Is Dissociated Between Memory Formation and Extinction. Front Mol Neurosci 2019; 12:38. [PMID: 30842725 PMCID: PMC6391346 DOI: 10.3389/fnmol.2019.00038] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Accepted: 01/31/2019] [Indexed: 01/18/2023] Open
Abstract
Associative long-term memories (LTMs) support long-lasting behavioral changes resulting from sensory experiences. Retrieval of a stable LTM by means of a large number of conditioned stimulus (CS) alone presentations produces inhibition of the original memory through extinction. Currently, there are two opposing hypotheses to account for the neural mechanisms supporting extinction. The unlearning hypothesis posits that extinction affects the original memory trace by reverting the synaptic changes supporting LTM. On the contrary, the new learning hypothesis proposes that extinction is simply the formation of a new associative memory that inhibits the expression of the original one. We propose that detailed analysis of extinction-associated molecular mechanisms could help distinguish between these hypotheses. Here we will review experimental evidence regarding the role of protein kinases and phosphatases (K&P) on LTM formation and extinction. Even though K&P regulate both memory processes, their participation appears to be dissociated. LTM formation recruits kinases, but is constrained by phosphatases. Memory extinction presents a more diverse molecular landscape, requiring phosphatases and some kinases, but also being constrained by kinase activity. Based on the available evidence, we propose a new theoretical model for memory extinction: a neuronal segregation of K&P supports a combination of time-dependent reversible inhibition of the original memory [CS-unconditioned stimulus (US)], with establishment of a new associative memory trace (CS-noUS).
Collapse
Affiliation(s)
- Mario Rafael Pagani
- Instituto de Fisiología y Biofísica Bernardo Houssay (IFIBIO)-Houssay, Facultad de Medicina, Universidad de Buenos Aires-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Emiliano Merlo
- Instituto de Fisiología y Biofísica Bernardo Houssay (IFIBIO)-Houssay, Facultad de Medicina, Universidad de Buenos Aires-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina.,Department of Psychology, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
38
|
Cattaneo A, Begni V, Malpighi C, Cattane N, Luoni A, Pariante C, Riva MA. Transcriptional Signatures of Cognitive Impairment in Rat Exposed to Prenatal Stress. Mol Neurobiol 2019; 56:6251-6260. [DOI: 10.1007/s12035-019-1523-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Accepted: 02/01/2019] [Indexed: 12/14/2022]
|
39
|
Zech M, Lam DD, Weber S, Berutti R, Poláková K, Havránková P, Fečíková A, Strom TM, Růžička E, Jech R, Winkelmann J. A unique de novo gain-of-function variant in CAMK4 associated with intellectual disability and hyperkinetic movement disorder. Cold Spring Harb Mol Case Stud 2018; 4:mcs.a003293. [PMID: 30262571 PMCID: PMC6318768 DOI: 10.1101/mcs.a003293] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Accepted: 09/04/2018] [Indexed: 01/12/2023] Open
Abstract
Calcium/calmodulin-dependent protein kinases (CaMKs) are key mediators of calcium signaling and underpin neuronal health. Although widely studied, the contribution of CaMKs to Mendelian disease is rather enigmatic. Here, we describe an unusual neurodevelopmental phenotype, characterized by milestone delay, intellectual disability, autism, ataxia, and mixed hyperkinetic movement disorder including severe generalized dystonia, in a proband who remained etiologically undiagnosed despite exhaustive testing. We performed trio whole-exome sequencing to identify a de novo essential splice-site variant (c.981+1G>A) in CAMK4, encoding CaMKIV. Through in silico evaluation and cDNA analyses, we demonstrated that c.981+1G>A alters CAMK4 pre-mRNA processing and results in a stable mRNA transcript containing a 77-nt out-of-frame deletion and a premature termination codon within the last exon. The expected protein, p.Lys303Serfs*28, exhibits selective loss of the carboxy-terminal regulatory domain of CaMKIV and bears striking structural resemblance to previously reported synthetic mutants that confer constitutive CaMKIV activity. Biochemical studies in proband-derived cells confirmed an activating effect of c.981+1G>A and indicated that variant-induced excessive CaMKIV signaling is sensitive to pharmacological manipulation. Additionally, we found that variants predicted to cause selective depletion of CaMKIV's regulatory domain are unobserved in diverse catalogs of human variation, thus revealing that c.981+1G>A is a unique molecular event. We propose that our proband's phenotype is explainable by a dominant CAMK4 splice-disrupting mutation that acts through a gain-of-function mechanism. Our findings highlight the importance of CAMK4 in human neurodevelopment, provide a foundation for future clinical research of CAMK4, and suggest the CaMKIV signaling pathway as a potential drug target in neurological disease.
Collapse
Affiliation(s)
- Michael Zech
- Institut für Neurogenomik, Helmholtz Zentrum München, Munich, 85764, Germany.,Klinik und Poliklinik für Neurologie, Klinikum rechts der Isar, Technische Universität München, Munich, 81675, Germany
| | - Daniel D Lam
- Institut für Neurogenomik, Helmholtz Zentrum München, Munich, 85764, Germany
| | - Sandrina Weber
- Institut für Neurogenomik, Helmholtz Zentrum München, Munich, 85764, Germany
| | - Riccardo Berutti
- Institut für Humangenetik, Helmholtz Zentrum München, Munich, 85764, Germany
| | - Kamila Poláková
- Department of Neurology and Center of Clinical Neuroscience, First Faculty of Medicine, Charles University and General Faculty Hospital, Prague, 120 00, Czech Republic
| | - Petra Havránková
- Department of Neurology and Center of Clinical Neuroscience, First Faculty of Medicine, Charles University and General Faculty Hospital, Prague, 120 00, Czech Republic
| | - Anna Fečíková
- Department of Neurology and Center of Clinical Neuroscience, First Faculty of Medicine, Charles University and General Faculty Hospital, Prague, 120 00, Czech Republic
| | - Tim M Strom
- Institut für Humangenetik, Helmholtz Zentrum München, Munich, 85764, Germany.,Institut für Humangenetik, Technische Universität München, Munich, 81675, Germany
| | - Evžen Růžička
- Department of Neurology and Center of Clinical Neuroscience, First Faculty of Medicine, Charles University and General Faculty Hospital, Prague, 120 00, Czech Republic
| | - Robert Jech
- Department of Neurology and Center of Clinical Neuroscience, First Faculty of Medicine, Charles University and General Faculty Hospital, Prague, 120 00, Czech Republic
| | - Juliane Winkelmann
- Institut für Neurogenomik, Helmholtz Zentrum München, Munich, 85764, Germany.,Institut für Humangenetik, Technische Universität München, Munich, 81675, Germany.,Lehrstuhl für Neurogenetik, Technische Universität München, Munich, 80333, Germany.,Munich Cluster for Systems Neurology, SyNergy, Munich, 81377, Germany
| |
Collapse
|
40
|
Effect of Exercise and Aβ Protein Infusion on Long-Term Memory-Related Signaling Molecules in Hippocampal Areas. Mol Neurobiol 2018; 56:4980-4987. [DOI: 10.1007/s12035-018-1425-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Accepted: 11/05/2018] [Indexed: 12/12/2022]
|
41
|
Vashisht A, Bach SV, Fetterhoff D, Morgan JW, McGee M, Hegde AN. Proteasome limits plasticity-related signaling to the nucleus in the hippocampus. Neurosci Lett 2018; 687:31-36. [PMID: 30219486 DOI: 10.1016/j.neulet.2018.09.017] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Revised: 08/21/2018] [Accepted: 09/10/2018] [Indexed: 01/22/2023]
Abstract
Proteolysis by the ubiquitin-proteasome pathway has pleiotropic effects on both induction and maintenance of long-term synaptic plasticity. In this study, we examined the effect of proteasome inhibition on signaling to the nucleus during late-phase long-term potentiation. When a subthreshold L-LTP induction protocol was used, proteasome inhibition led to a significant increase in phosphorylated CREB (pCREB) in the nucleus. Inhibitors of cAMP-dependent protein kinase/protein kinase A, extracellular signal-regulated kinase and cGMP-dependent protein kinase/protein kinase G all blocked the proteasome-inhibition-mediated increase in nuclear pCREB after subthreshold stimulation. These results lay the groundwork for understanding a novel role for the proteasome in limiting signaling to the nucleus in the absence of adequate synaptic stimulation.
Collapse
Affiliation(s)
- Anirudh Vashisht
- Department of Neurobiology and Anatomy & Wake Forest University Health Sciences, Medical Center Boulevard, Winston-Salem, NC, 27157, USA; Department of Biological and Environmental Sciences, Georgia College and State University, Milledgeville, GA, 31061, USA
| | - Svitlana V Bach
- Department of Neurobiology and Anatomy & Wake Forest University Health Sciences, Medical Center Boulevard, Winston-Salem, NC, 27157, USA
| | - Dustin Fetterhoff
- Department of Neurobiology and Anatomy & Wake Forest University Health Sciences, Medical Center Boulevard, Winston-Salem, NC, 27157, USA
| | - James W Morgan
- Department of Neurobiology and Anatomy & Wake Forest University Health Sciences, Medical Center Boulevard, Winston-Salem, NC, 27157, USA
| | - Maria McGee
- Plastic and Reconstructive Surgery Research, Wake Forest University Health Sciences, Medical Center Boulevard, Winston-Salem, NC, 27157, USA
| | - Ashok N Hegde
- Department of Neurobiology and Anatomy & Wake Forest University Health Sciences, Medical Center Boulevard, Winston-Salem, NC, 27157, USA; Department of Biological and Environmental Sciences, Georgia College and State University, Milledgeville, GA, 31061, USA.
| |
Collapse
|
42
|
Thompson JW, Sorum AW, Hsieh-Wilson LC. Deciphering the Functions of O-GlcNAc Glycosylation in the Brain: The Role of Site-Specific Quantitative O-GlcNAcomics. Biochemistry 2018; 57:4010-4018. [PMID: 29936833 PMCID: PMC6058732 DOI: 10.1021/acs.biochem.8b00516] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
The dynamic posttranslational modification O-linked β- N-acetylglucosamine glycosylation (O-GlcNAcylation) is present on thousands of intracellular proteins in the brain. Like phosphorylation, O-GlcNAcylation is inducible and plays important functional roles in both physiology and disease. Recent advances in mass spectrometry (MS) and bioconjugation methods are now enabling the mapping of O-GlcNAcylation events to individual sites in proteins. However, our understanding of which glycosylation events are necessary for regulating protein function and controlling specific processes, phenotypes, or diseases remains in its infancy. Given the sheer number of O-GlcNAc sites, methods for identifying promising sites and prioritizing them for time- and resource-intensive functional studies are greatly needed. Revealing sites that are dynamically altered by different stimuli or disease states will likely go a long way in this regard. Here, we describe advanced methods for identifying O-GlcNAc sites on individual proteins and across the proteome and for determining their stoichiometry in vivo. We also highlight emerging technologies for quantitative, site-specific MS-based O-GlcNAc proteomics (O-GlcNAcomics), which allow proteome-wide tracking of O-GlcNAcylation dynamics at individual sites. These cutting-edge technologies are beginning to bridge the gap between the high-throughput cataloguing of O-GlcNAcylated proteins and the relatively low-throughput study of individual proteins. By uncovering the O-GlcNAcylation events that change in specific physiological and disease contexts, these new approaches are providing key insights into the regulatory functions of O-GlcNAc in the brain, including their roles in neuroprotection, neuronal signaling, learning and memory, and neurodegenerative diseases.
Collapse
Affiliation(s)
- John W. Thompson
- Department of Chemistry and Chemical Engineering, California Institute
of Technology, Pasadena, California 91125, United States
| | - Alexander W. Sorum
- Department of Chemistry and Chemical Engineering, California Institute
of Technology, Pasadena, California 91125, United States
| | - Linda C. Hsieh-Wilson
- Department of Chemistry and Chemical Engineering, California Institute
of Technology, Pasadena, California 91125, United States
| |
Collapse
|
43
|
Beheshti S, Ghorbanpour Skakakomi A, Ghaedi K, Dehestani H. Frankincense upregulates the hippocampal calcium/calmodulin kinase II‐α during development of the rat brain and improves memory performance. Int J Dev Neurosci 2018; 69:44-48. [DOI: 10.1016/j.ijdevneu.2018.06.011] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Revised: 05/24/2018] [Accepted: 06/24/2018] [Indexed: 11/29/2022] Open
Affiliation(s)
- Siamak Beheshti
- Division of Animal Sciences, Department of Biology, Faculty of SciencesUniversity of IsfahanIsfahanIran
| | | | - Kamran Ghaedi
- Division of Cellular and Molecular Biology, Department of Biology, Faculty of SciencesUniversity of IsfahanIsfahanIran
| | - Hadi Dehestani
- Division of Animal Sciences, Department of Biology, Faculty of SciencesUniversity of IsfahanIsfahanIran
| |
Collapse
|
44
|
Calmodulin shuttling mediates cytonuclear signaling to trigger experience-dependent transcription and memory. Nat Commun 2018; 9:2451. [PMID: 29934532 PMCID: PMC6015085 DOI: 10.1038/s41467-018-04705-8] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Accepted: 05/14/2018] [Indexed: 12/18/2022] Open
Abstract
Learning and memory depend on neuronal plasticity originating at the synapse and requiring nuclear gene expression to persist. However, how synapse-to-nucleus communication supports long-term plasticity and behavior has remained elusive. Among cytonuclear signaling proteins, γCaMKII stands out in its ability to rapidly shuttle Ca2+/CaM to the nucleus and thus activate CREB-dependent transcription. Here we show that elimination of γCaMKII prevents activity-dependent expression of key genes (BDNF, c-Fos, Arc), inhibits persistent synaptic strengthening, and impairs spatial memory in vivo. Deletion of γCaMKII in adult excitatory neurons exerts similar effects. A point mutation in γCaMKII, previously uncovered in a case of intellectual disability, selectively disrupts CaM sequestration and CaM shuttling. Remarkably, this mutation is sufficient to disrupt gene expression and spatial learning in vivo. Thus, this specific form of cytonuclear signaling plays a key role in learning and memory and contributes to neuropsychiatric disease. Activity-dependent gene expression is thought to involve translocation of Ca2+/calmodulin (CaM) to the nucleus. Here, the authors examine a translocation-deficient mutant of γCaMKII, a Ca2+/CaM shuttle protein, to show that translocation of Ca2+/CaM is required for memory and synaptic plasticity.
Collapse
|
45
|
Charpentier M. Calcium Signals in the Plant Nucleus: Origin and Function. JOURNAL OF EXPERIMENTAL BOTANY 2018; 69:4986421. [PMID: 29718301 DOI: 10.1093/jxb/ery160] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Indexed: 06/08/2023]
Abstract
The universality of calcium as an intracellular messenger depends on the dynamics of its spatial and temporal release from calcium stores. Accumulating evidence over the past two decades supports an essential role for nuclear calcium signalling in the transduction of specific stimuli into cellular responses. This review focusses on mechanisms underpinning changes in nuclear calcium concentrations and discusses what is known so far, about the origin of the nuclear calcium signals identified, primarily in the context of microbial symbioses and abiotic stresses.
Collapse
Affiliation(s)
- Myriam Charpentier
- John Innes Centre, Department of Cell and developmental Biology, Colney Lane, Norwich, UK
| |
Collapse
|
46
|
Clocking In Time to Gate Memory Processes: The Circadian Clock Is Part of the Ins and Outs of Memory. Neural Plast 2018; 2018:6238989. [PMID: 29849561 PMCID: PMC5925033 DOI: 10.1155/2018/6238989] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Revised: 01/22/2018] [Accepted: 02/05/2018] [Indexed: 01/11/2023] Open
Abstract
Learning, memory consolidation, and retrieval are processes known to be modulated by the circadian (circa: about; dies: day) system. The circadian regulation of memory performance is evolutionarily conserved, independent of the type and complexity of the learning paradigm tested, and not specific to crepuscular, nocturnal, or diurnal organisms. In mammals, long-term memory (LTM) formation is tightly coupled to de novo gene expression of plasticity-related proteins and posttranslational modifications and relies on intact cAMP/protein kinase A (PKA)/protein kinase C (PKC)/mitogen-activated protein kinase (MAPK)/cyclic adenosine monophosphate response element-binding protein (CREB) signaling. These memory-essential signaling components cycle rhythmically in the hippocampus across the day and night and are clearly molded by an intricate interplay between the circadian system and memory. Important components of the circadian timing mechanism and its plasticity are members of the Period clock gene family (Per1, Per2). Interestingly, Per1 is rhythmically expressed in mouse hippocampus. Observations suggest important and largely unexplored roles of the clock gene protein PER1 in synaptic plasticity and in the daytime-dependent modulation of learning and memory. Here, we review the latest findings on the role of the clock gene Period 1 (Per1) as a candidate molecular and mechanistic blueprint for gating the daytime dependency of memory processing.
Collapse
|
47
|
Johnson JJ, Loeffert AC, Stokes J, Olympia RP, Bramley H, Hicks SD. Association of Salivary MicroRNA Changes With Prolonged Concussion Symptoms. JAMA Pediatr 2018; 172:65-73. [PMID: 29159407 PMCID: PMC5833519 DOI: 10.1001/jamapediatrics.2017.3884] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
IMPORTANCE Approximately one-third of children who experience a concussion develop prolonged concussion symptoms. To our knowledge, there are currently no objective or easily administered tests for predicting prolonged concussion symptoms. Several studies have identified alterations in epigenetic molecules known as microRNAs (miRNAs) following traumatic brain injury. No studies have examined whether miRNA expression can detect prolonged concussion symptoms. OBJECTIVE To evaluate the efficacy of salivary miRNAs for identifying children with concussion who are at risk for prolonged symptoms. DESIGN, SETTING, AND PARTICIPANTS This prospective cohort study at the Penn State Medical Center observed 52 patients aged 7 to 21 years presenting for evaluation of concussion within 14 days of initial head injury, with follow-up at 4 and 8 weeks. EXPOSURES All patients had a clinical diagnosis of concussion. MAIN OUTCOMES AND MEASURES Salivary miRNA expression was measured at the time of initial clinical presentation in all patients. Patients with a Sport Concussion Assessment Tool (SCAT3) symptom score of 5 or greater on self-report or parent report 4 weeks after injury were designated as having prolonged symptoms. RESULTS Of the 52 included participants, 22 (42%) were female, and the mean (SD) age was 14 (3) years. Participants were split into the prolonged symptom group (n = 30) and acute symptom group (n = 22). Concentrations of 15 salivary miRNAs spatially differentiated prolonged and acute symptom groups on partial least squares discriminant analysis and demonstrated functional relationships with neuronal regulatory pathways. Levels of 5 miRNAs (miR-320c-1, miR-133a-5p, miR-769-5p, let-7a-3p, and miR-1307-3p) accurately identified patients with prolonged symptoms on logistic regression (area under the curve, 0.856; 95% CI, 0.822-0.890). This accuracy exceeded accuracy of symptom burden on child (area under the curve, 0.649; 95% CI, 0.388-0.887) or parent (area under the curve, 0.562; 95% CI, 0.219-0.734) SCAT3 score. Levels of 3 miRNAs were associated with specific symptoms 4 weeks after injury; miR-320c-1 was associated with memory difficulty (R, 0.55; false detection rate, 0.02), miR-629 was associated with headaches (R, 0.47; false detection rate, 0.04), and let-7b-5p was associated with fatigue (R, 0.45; false detection rate, 0.04). CONCLUSIONS AND RELEVANCE Salivary miRNA levels may identify the duration and character of concussion symptoms. This could reduce parental anxiety and improve care by providing a tool for concussion management. Further validation of this approach is needed.
Collapse
Affiliation(s)
- Jeremiah J. Johnson
- Department of Pediatrics, Penn State College of Medicine, Pennsylvania State University, Hershey
| | - Andrea C. Loeffert
- Department of Pediatrics, Penn State College of Medicine, Pennsylvania State University, Hershey
| | - Jennifer Stokes
- Department of Pediatrics, Penn State College of Medicine, Pennsylvania State University, Hershey
| | - Robert P. Olympia
- Department of Pediatrics, Penn State College of Medicine, Pennsylvania State University, Hershey,Department of Emergency Medicine, Penn State College of Medicine, Pennsylvania State University, Hershey
| | - Harry Bramley
- Department of Pediatrics, Penn State College of Medicine, Pennsylvania State University, Hershey
| | - Steven D. Hicks
- Department of Pediatrics, Penn State College of Medicine, Pennsylvania State University, Hershey
| |
Collapse
|
48
|
Arambula SE, Jima D, Patisaul HB. Prenatal bisphenol A (BPA) exposure alters the transcriptome of the neonate rat amygdala in a sex-specific manner: a CLARITY-BPA consortium study. Neurotoxicology 2017; 65:207-220. [PMID: 29097150 DOI: 10.1016/j.neuro.2017.10.005] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Revised: 10/23/2017] [Accepted: 10/24/2017] [Indexed: 12/11/2022]
Abstract
Bisphenol A (BPA) is a widely recognized endocrine disruptor prevalent in many household items. Because experimental and epidemiological data suggest links between prenatal BPA exposure and altered affective behaviors in children, even at levels below the current US FDA No Observed Adverse Effect Level (NOAEL) of 5mg/kg body weight (bw)/day, there is concern that early life exposure may alter neurodevelopment. The current study was conducted as part of the CLARITY-BPA (Consortium Linking Academic and Regulatory Insights on BPA Toxicity) program and examined the full amygdalar transcriptome on postnatal day (PND) 1, with the hypothesis that prenatal BPA exposure would alter the expression of genes and pathways fundamental to sex-specific affective behaviors. NCTR Sprague-Dawley dams were gavaged from gestational day 6 until parturition with BPA (2.5, 25, 250, 2500, or 25000μg/kg bw/day), a reference estrogen (0.05 or 0.5μg ethinyl estradiol (EE2)/kg bw/day), or vehicle. PND 1 amygdalae were microdissected and gene expression was assessed with qRT-PCR (all exposure groups) and RNAseq (vehicle, 25 and 250μg BPA, and 0.5μg EE2 groups only). Our results demonstrate that that prenatal BPA exposure can disrupt the transcriptome of the neonate amygdala, at doses below the FDA NOAEL, in a sex-specific manner and indicate that the female amygdala may be more sensitive to BPA exposure during fetal development. We also provide additional evidence that developmental BPA exposure can interfere with estrogen, oxytocin, and vasopressin signaling pathways in the developing brain and alter signaling pathways critical for synaptic organization and transmission.
Collapse
Affiliation(s)
- Sheryl E Arambula
- Department of Biological Sciences, North Carolina State University, Raleigh, NC 27695, USA; WM Keck Center for Behavioral Biology, North Carolina State University, Raleigh, NC 27695, USA
| | - Dereje Jima
- Center for Human Health and the Environment, North Carolina State University, Raleigh, NC 27695, USA; Bioinformatics Research Center, North Carolina State University, Raleigh, NC 27695
| | - Heather B Patisaul
- Department of Biological Sciences, North Carolina State University, Raleigh, NC 27695, USA; WM Keck Center for Behavioral Biology, North Carolina State University, Raleigh, NC 27695, USA; Center for Human Health and the Environment, North Carolina State University, Raleigh, NC 27695, USA.
| |
Collapse
|
49
|
Abstract
Alzheimer's disease (AD) is a devastating neurodegenerative disorder and the most common cause of dementia among aged people whose population is rapidly increasing. AD not only seriously affects the patient's physical health and quality of life, but also adds a heavy burden to the patient's family and society. It is urgent to understand AD pathogenesis and develop the means of prevention and treatment. AD is a chronic devastating neurodegenerative disease without effective treatment. Current approaches for management focus on helping patients relieve or delay the symptoms of cognitive dysfunction. The calcium ion (Ca2+) is an important second messenger in the function and structure of nerve cell circuits in the brain such as neuronal growth, exocytosis, as well as in synaptic and cognitive function. Increasing numbers of studies suggested that disruption of intracellular Ca2+ homeostasis, especially the abnormal and excessive Ca2+ release from the endoplasmic reticulum (ER) via the ryanodine receptor (RYR), plays important roles in orchestrating the dynamic of the neuropathology of AD and associated memory loss, cognitive dysfunction. Dantrolene, a known antagonist of the RYR and a clinically available drug to treat malignant hyperthermia, can ameliorate the abnormal Ca2+ release from the RYR in AD and the subsequent pathogenesis, such as increased β-secretase and γ-secretase activities, production of Amyloid-β 42 (Aβ 42) and its oligomer, impaired autophagy, synapse dysfunction, and memory loss. However, more studies are needed to confirm the efficacy and safety repurposing dantrolene as a therapeutic drug in AD.
Collapse
Affiliation(s)
- Yong Wang
- Department of Anesthesiology and Critical Care, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.,Department of Anesthesiology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Yun Shi
- Department of Anesthesiology and Critical Care, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.,Department of Anesthesiology, Children's Hospital of Fudan University, Shanghai, 201102, China
| | - Huafeng Wei
- Department of Anesthesiology and Critical Care, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
50
|
Han YY, Wang XD, Liu L, Guo HM, Cong W, Yan WW, Huang JN, Xiao P, Li CH. L-type VDCCs participate in behavioral-LTP and memory retention. Neurobiol Learn Mem 2017; 145:75-83. [PMID: 28866469 DOI: 10.1016/j.nlm.2017.08.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2016] [Revised: 08/09/2017] [Accepted: 08/29/2017] [Indexed: 11/25/2022]
Abstract
Although L-type voltage-dependent calcium channels (VDCCs) have been reported to display different even contrary actions on cognitive functions and long-term potentiation (LTP) formation, there is little information regarding the role of L-type VDCCs in behavioral LTP, a learning-induced LTP model, in the intact brain of freely behaving animals. Here we investigated the effects of verapamil, a non-selective blocker of L-type VDCCs, on behavioral LTP and cognitive functions. Population spikes (PS) were recorded by using electrophysiological methods to examine the role of verapamil in behavioral LTP in the hippocampal dentate gyrus (DG) region. Y-maze assay was used to evaluate the effects of verapamil on learning and memory. Electron microscope was used to observe the changes on synaptic ultrastructural morphology in hippocampal DG area. We found that intrahippocampal verapamil treatments had no significant changes on the PS amplitude during a 90min recordings period. However, intrahippocampal applications of verapamil, including pre- or post-training, reduced behavioral LTP magnitude and memory retention but did not prevent the induction of behavioral LTP and the acquisition of learning. The saline group with behaving trainings showed obvious increases in the number of smile synapses, the length of active zones and the thickness of postsynaptic density as compared to the baseline group, but verapamil with pre-training treatment almost returned these changes to the baseline levels except for the synaptic interface curvature. In conclusion, our results suggest that L-type VDCCs may only contribute to the magnitude of behavioral LTP and the memory maintenance with an activity-independent relationship. L-type VDCCs may be critical to new information long-term storage rather than acquisition in hippocampus.
Collapse
Affiliation(s)
- Yuan-Yuan Han
- School of Life Science, South China Normal University, Guangzhou 510631, China
| | - Xiao-Dong Wang
- School of Life Science, South China Normal University, Guangzhou 510631, China
| | - Li Liu
- School of Life Science, South China Normal University, Guangzhou 510631, China
| | - Hong-Mei Guo
- School of Life Science, South China Normal University, Guangzhou 510631, China
| | - Wei Cong
- Henan Medical Equipment Inspection Institute, Zhengzhou 450003, China
| | - Wen-Wen Yan
- School of Life Science, South China Normal University, Guangzhou 510631, China
| | - Jun-Ni Huang
- School of Life Science, South China Normal University, Guangzhou 510631, China
| | - Peng Xiao
- School of Life Science, South China Normal University, Guangzhou 510631, China.
| | - Chu-Hua Li
- School of Life Science, South China Normal University, Guangzhou 510631, China; Brain Science Institute, South China Normal University, Guangzhou 510631, China.
| |
Collapse
|