1
|
Tavalin SJ. Familial Alzheimer's disease mutations in amyloid precursor protein impair calcineurin signaling to NMDA receptors. J Biol Chem 2025; 301:108147. [PMID: 39732167 PMCID: PMC11910330 DOI: 10.1016/j.jbc.2024.108147] [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: 03/24/2024] [Revised: 12/19/2024] [Accepted: 12/21/2024] [Indexed: 12/30/2024] Open
Abstract
Familial Alzheimer's disease (FAD) is frequently associated with mutations in the amyloid precursor protein (APP), which are thought to lead to cognitive deficits by impairing NMDA receptor (NMDAR)-dependent forms of synaptic plasticity. Given the reliance of synaptic plasticity on NMDAR-mediated Ca2+ entry, shaping of NMDAR activity by APP and/or its disease-causing variants could provide a basis for understanding synaptic plasticity impairments associated with FAD. A region of APP (residues 639-644 within APP695) processed by the γ-secretase complex, which generates amyloid-β peptides, is a hotspot for FAD mutations. This region bears similarity to a binding motif for calcineurin (CaN), a Ca2+/calmodulin-dependent phosphatase. Interaction assays confirm that APP associates with CaN in native tissue as well as in a heterologous expression system. This capacity to bind CaN extends to APP family members amyloid precursor-like protein 1 and amyloid precursor-like protein 2 (APLP1 and APLP2, respectively). Electrophysiological analysis demonstrates that APP and its family members limit NMDAR activity, in a manner consistent with CaN-dependent regulation of NMDAR desensitization. FAD mutations, in this region of APP, impair this regulation and consequently enhance NMDAR activity. Thus, by altering the landscape for CaN regulation of NMDA receptors, FAD mutations in APP may contribute to faulty information processing by modifying the dynamic range and temporal window of a critical signal for synaptic plasticity.
Collapse
Affiliation(s)
- Steven J Tavalin
- Department of Pharmacology, Addiction Science, and Toxicology, College of Medicine, The University of Tennessee Health Science Center, Memphis, USA.
| |
Collapse
|
2
|
Walker SL, Glasper ER. Unraveling sex differences in maternal and paternal care impacts on social behaviors and neurobiological responses to early-life adversity. Front Neuroendocrinol 2025; 76:101162. [PMID: 39561882 PMCID: PMC11811932 DOI: 10.1016/j.yfrne.2024.101162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 11/11/2024] [Accepted: 11/11/2024] [Indexed: 11/21/2024]
Abstract
Early-life stress (ELS) affects the development of prosocial behaviors and social-cognitive function, often leading to structural brain changes and increased psychosocial disorders. Recent studies suggest that mother- and father-child relationships independently influence social development in a sex-specific manner, but the effects of impaired father-child relationships are often overlooked. This review examines preclinical rodent studies to explore how parental neglect impacts neuroplasticity and social behaviors in offspring. We highlight that disruptions in maternal interactions may affect male pups more in uniparental rodents, while impaired paternal interactions in biparental rodents tend to impact female pups more. Due to limited research, the separate effects of maternal and paternal neglect on brain development and social behaviors in biparental species remain unclear. Addressing these gaps could clarify the sex-specific mechanisms underlying social and neurobiological deficits following parental neglect.
Collapse
Affiliation(s)
- Shakeera L Walker
- Department of Neuroscience, The Ohio State University, Columbus, OH, 43210, United States; Neuroscience Graduate Program, The Ohio State University, Columbus, OH, 43210, United States
| | - Erica R Glasper
- Department of Neuroscience, The Ohio State University, Columbus, OH, 43210, United States; Neuroscience Graduate Program, The Ohio State University, Columbus, OH, 43210, United States; Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH, 43210, United States.
| |
Collapse
|
3
|
Chimura T, Manabe T. Ca2+-PP2B-PSD-95 axis: A novel regulatory mechanism of the phosphorylation state of Serine 295 of PSD-95. PLoS One 2024; 19:e0313441. [PMID: 39509447 PMCID: PMC11542788 DOI: 10.1371/journal.pone.0313441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 10/23/2024] [Indexed: 11/15/2024] Open
Abstract
The phosphorylation state of PSD-95 at Serine 295 (Ser295) is important for the regulation of synaptic plasticity. Although the activation of NMDA receptors (NMDARs), which initiates an intracellular calcium signaling cascade, decreases phosphorylated Ser295 (pS295) of PSD-95, the molecular mechanisms are not fully understood. We found that the calcium-activated protein phosphatase PP2B dephosphorylated pS295 not only in basal conditions but also in NMDAR-activated conditions in cultured neurons. The biochemical assay also revealed the dephosphorylation of pS295 by PP2B, consistently supporting the results obtained using neurons. The newly identified calcium signaling cascade "Ca2+-PP2B-PSD-95 axis" would play an important role in the molecular mechanism for NMDA receptor-dependent plasticity.
Collapse
Affiliation(s)
- Takahiko Chimura
- Department of Basic Medical Sciences, Institute of Medical Science, Division of Neuronal Network, University of Tokyo, Tokyo, Japan
| | - Toshiya Manabe
- Department of Basic Medical Sciences, Institute of Medical Science, Division of Neuronal Network, University of Tokyo, Tokyo, Japan
| |
Collapse
|
4
|
Mathew J, Tumin D, Beng-Ongey H. Predictors of academic delay post-pediatric kidney transplant in the USA. Pediatr Nephrol 2024; 39:3309-3316. [PMID: 38963554 DOI: 10.1007/s00467-024-06445-y] [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: 03/19/2024] [Revised: 06/02/2024] [Accepted: 06/17/2024] [Indexed: 07/05/2024]
Abstract
BACKGROUND Pediatric patients with kidney failure often experience cognitive delays. However, academic delay (being more than one grade level below age-appropriate grade, or in special education) after pediatric kidney transplantation (KTx) has not been explored. We sought to identify patient characteristics associated with a higher risk of academic delay 1 year post-KTx. METHODS We used the United Network for Organ Sharing (UNOS) database to identify children aged 6-17 years who received a primary KTx between 2014 and 2021 and had a functioning graft 1 year after KTx. The primary outcome was the patient's academic progress at 1 year post-transplant. The secondary outcome was change in academic progress between transplant and 1-year follow-up: onset of new delay, resolution of pre-existing delay, persistence of delay, or no delay at either timepoint. Binomial and multinomial mixed effects logistic regression models were used to predict each outcome based on patient characteristics. RESULTS The study included 2197 patients, of whom 14% demonstrated academic delay at 1 year post-KTx, 4% demonstrated a new onset of academic delay, 5% demonstrated a resolution of academic delay, and 10% demonstrated persistent academic delay. Patients undergoing transplantation at a younger age, receiving a deceased donor kidney, experiencing longer waitlist times, and undergoing KTx for vascular or other disease indications for KTx were more likely to experience academic delays, including new-onset academic delays. CONCLUSIONS Academic delays are frequently reported among pediatric KTx recipients. Additional academic support may help resolving or preventing academic delay for at-risk subgroups of children undergoing KTx.
Collapse
Affiliation(s)
- Joanna Mathew
- East Carolina University, E 5thSt, Greenville, NC, 27858, USA.
| | - Dmitry Tumin
- Department of Pediatrics, Brody School of Medicine at East, Carolina University, Greenville, NC, USA
- Department of Academic Affairs, Brody School of Medicine at East, Carolina University, Greenville, NC, USA
| | - Hostensia Beng-Ongey
- Department of Pediatrics, Brody School of Medicine at East, Carolina University, Greenville, NC, USA
| |
Collapse
|
5
|
Scaduto P, Marcatti M, Bhatt N, Kayed R, Taglialatela G. Calcineurin inhibition prevents synaptic plasticity deficit induced by brain-derived tau oligomers. Brain Commun 2024; 6:fcae277. [PMID: 39239152 PMCID: PMC11375858 DOI: 10.1093/braincomms/fcae277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 06/08/2024] [Accepted: 08/13/2024] [Indexed: 09/07/2024] Open
Abstract
Compelling evidence suggests that cognitive decline in Alzheimer's disease is associated with the accumulation and aggregation of tau protein, with the most toxic aggregates being in the form of oligomers. This underscores the necessity for direct isolation and analysis of brain-derived tau oligomers from patients with Alzheimer's disease, potentially offering novel perspectives into tau toxicity. Alzheimer's brain-derived tau oligomers are potent inhibitors of synaptic plasticity; however, the involved mechanism is still not fully understood. We previously reported a significantly reduced incidence of Alzheimer's disease in ageing humans chronically treated with a Food and Drug Administration-approved calcineurin inhibitor, FK506 (tacrolimus), used as an immunosuppressant after solid organ transplant. Using a combination of electrophysiological and RNA-sequencing techniques, we provide here evidence that FK506 has the potential to block the acute toxic effect of brain-derived tau oligomers on synaptic plasticity, as well as to restore the levels of some key synaptic mRNAs. These results further support FK506 as a promising novel therapeutic strategy for the treatment of Alzheimer's disease.
Collapse
Affiliation(s)
- Pietro Scaduto
- Department of Neurology, Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch (UTMB), 301 University Blvd, Galveston, TX 77555, USA
| | - Michela Marcatti
- Department of Neurology, Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch (UTMB), 301 University Blvd, Galveston, TX 77555, USA
| | - Nemil Bhatt
- Department of Neurology, Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch (UTMB), 301 University Blvd, Galveston, TX 77555, USA
| | - Rakez Kayed
- Department of Neurology, Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch (UTMB), 301 University Blvd, Galveston, TX 77555, USA
| | - Giulio Taglialatela
- Department of Neurology, Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch (UTMB), 301 University Blvd, Galveston, TX 77555, USA
| |
Collapse
|
6
|
Takasaki T, Bamba A, Kukita Y, Nishida A, Kanbayashi D, Hagihara K, Satoh R, Ishihara K, Sugiura R. Rcn1, the fission yeast homolog of human DSCR1, regulates arsenite tolerance independently from calcineurin. Genes Cells 2024; 29:589-598. [PMID: 38715219 DOI: 10.1111/gtc.13122] [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: 03/11/2024] [Revised: 04/29/2024] [Accepted: 04/30/2024] [Indexed: 07/06/2024]
Abstract
Calcineurin (CN) is a conserved Ca2+/calmodulin-dependent phosphoprotein phosphatase that plays a key role in Ca2+ signaling. Regulator of calcineurin 1 (RCAN1), also known as Down syndrome critical region gene 1 (DSCR1), interacts with calcineurin and inhibits calcineurin-dependent signaling in various organisms. Ppb1, the fission yeast calcineurin regulates Cl--homeostasis, and Ppb1 deletion induces MgCl2 hypersensitivity. Here, we characterize the conserved and novel roles of the fission yeast RCAN1 homolog rcn1+. Consistent with its role as an endogenous calcineurin inhibitor, Rcn1 overproduction reproduced the calcineurin-null phenotypes, including MgCl2 hypersensitivity and inhibition of calcineurin signaling upon extracellular Ca2+ stimuli as evaluated by the nuclear translocation and transcriptional activation of the calcineurin substrate Prz1. Notably, overexpression of rcn1+ causes hypersensitivity to arsenite, whereas calcineurin deletion induces arsenite tolerance, showing a phenotypic discrepancy between Rcn1 overexpression and calcineurin deletion. Importantly, although Rcn1 deletion induces modest sensitivities to arsenite and MgCl2 in wild-type cells, the arsenite tolerance, but not MgCl2 sensitivity, associated with Ppb1 deletion was markedly suppressed by Rcn1 deletion. Collectively, our findings reveal a previously unrecognized functional collaboration between Rcn1 and calcineurin, wherein Rcn1 not only negatively regulates calcineurin in the Cl- homeostasis, but also Rcn1 mediates calcineurin signaling to modulate arsenite cytotoxicity.
Collapse
Affiliation(s)
- Teruaki Takasaki
- Laboratory of Molecular Pharmacogenomics, Department of Pharmaceutical Sciences, Kindai University, Osaka, Japan
| | - Asuka Bamba
- Laboratory of Molecular Pharmacogenomics, Department of Pharmaceutical Sciences, Kindai University, Osaka, Japan
| | - Yuka Kukita
- Laboratory of Molecular Pharmacogenomics, Department of Pharmaceutical Sciences, Kindai University, Osaka, Japan
| | - Aiko Nishida
- Laboratory of Molecular Pharmacogenomics, Department of Pharmaceutical Sciences, Kindai University, Osaka, Japan
| | - Daiki Kanbayashi
- Laboratory of Molecular Pharmacogenomics, Department of Pharmaceutical Sciences, Kindai University, Osaka, Japan
| | - Kanako Hagihara
- Laboratory of Molecular Pharmacogenomics, Department of Pharmaceutical Sciences, Kindai University, Osaka, Japan
- Laboratory of Hygienic Science, Department of Pharmacy, School of Pharmacy, Hyogo Medical University, Kobe, Japan
| | - Ryosuke Satoh
- Laboratory of Molecular Pharmacogenomics, Department of Pharmaceutical Sciences, Kindai University, Osaka, Japan
| | - Keiichi Ishihara
- Laboratory of Pathological Biochemistry, Kyoto Pharmaceutical University, Kyoto, Japan
| | - Reiko Sugiura
- Laboratory of Molecular Pharmacogenomics, Department of Pharmaceutical Sciences, Kindai University, Osaka, Japan
| |
Collapse
|
7
|
Weiss BE, Kraner SD, Artiushin IA, Norris CM. Elevated calcineurin activity in primary astrocytes leads to the dephosphorylation of connexin 43 in conjunction with increased membrane permeability. Neuroreport 2024; 35:673-678. [PMID: 38813906 PMCID: PMC11279532 DOI: 10.1097/wnr.0000000000002051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/31/2024]
Abstract
Hyperactivation of the Ca2+/calmodulin-dependent phosphatase calcineurin (CN) is observed in reactive astrocytes associated with neuroinflammation and progressive degenerative diseases, like Alzheimer's disease. Apart from key transcription factors (e.g. nuclear factor of activated t cells and nuclear factor-κB) very few other CN-dependent pathways have been studied in astrocytes. The hemichannel protein, connexin 43 (Cx43) is found at high levels in astrocytes and contains a CN-sensitive Ser residue near its carboxy terminus. CN-dependent dephosphorylation of Cx43 has been reported in primary astrocytes treated with injurious stimuli, but much remains unknown about CN/Cx43 interactions in the context of neuroinflammation and disease. Western blots were used to assess total Cx43 and dephosphorylated Cx43 subtypes in rat embryonic primary astrocytes treated with a hyperactive CN fragment (ΔCN, via adenovirus), or with a proinflammatory cytokine cocktail. Under similar treatment conditions, an ethidium bromide (EtBr) uptake assay was used to assess membrane permeability. Effects of ΔCN and cytokines were tested in the presence or absence of the CN inhibitor, cyclosporin A. A connexin inhibitor, carbenoxolone was also used in EtBr assays to assess the involvement of connexins in membrane permeability. Treatment with ΔCN or cytokines increased dephosphorylated Cx43 levels in conjunction with increased membrane permeability (elevated EtBr uptake). Effects of ΔCN or cytokine treatment were blocked by cyclosporine A. Treatment-induced changes in EtBr uptake were also inhibited by carbenoxolone. The results suggest that Cx43 hemichannels could be an important mechanism through which astrocytic CN disrupts neurologic function associated with neurodegenerative disease.
Collapse
Affiliation(s)
- Blaine E. Weiss
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY 40536
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY 40536
| | - Susan D. Kraner
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY 40536
| | - Irina A. Artiushin
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY 40536
| | - Christopher M. Norris
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY 40536
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY 40536
| |
Collapse
|
8
|
Tapella L, Dematteis G, La Vitola P, Leva S, Tonelli E, Raddi M, Delconti M, Dacomo L, La Macchia A, Murari E, Talmon M, Malecka J, Chrostek G, Grilli M, Colombo L, Salmona M, Forloni G, Genazzani AA, Balducci C, Lim D. Genetic deletion of astrocytic calcineurin B1 prevents cognitive impairment and neuropathology development in acute and chronic mouse models of Alzheimer's disease. Glia 2024; 72:899-915. [PMID: 38288580 DOI: 10.1002/glia.24509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 01/15/2024] [Accepted: 01/18/2024] [Indexed: 03/20/2024]
Abstract
Alzheimer's disease (AD) represents an urgent yet unmet challenge for modern society, calling for exploration of innovative targets and therapeutic approaches. Astrocytes, main homeostatic cells in the CNS, represent promising cell-target. Our aim was to investigate if deletion of the regulatory CaNB1 subunit of calcineurin in astrocytes could mitigate AD-related memory deficits, neuropathology, and neuroinflammation. We have generated two, acute and chronic, AD mouse models with astrocytic CaNB1 ablation (ACN-KO). In the former, we evaluated the ability of β-amyloid oligomers (AβOs) to impair memory and activate glial cells once injected in the cerebral ventricle of conditional ACN-KO mice. Next, we generated a tamoxifen-inducible astrocyte-specific CaNB1 knock-out in 3xTg-AD mice (indACNKO-AD). CaNB1 was deleted, by tamoxifen injection, in 11.7-month-old 3xTg-AD mice for 4.4 months. Spatial memory was evaluated using the Barnes maze; β-amyloid plaques burden, neurofibrillary tangle deposition, reactive gliosis, and neuroinflammation were also assessed. The acute model showed that ICV injected AβOs in 2-month-old wild type mice impaired recognition memory and fostered a pro-inflammatory microglia phenotype, whereas in ACN-KO mice, AβOs were inactive. In indACNKO-AD mice, 4.4 months after CaNB1 depletion, we found preservation of spatial memory and cognitive flexibility, abolishment of amyloidosis, and reduction of neurofibrillary tangles, gliosis, and neuroinflammation. Our results suggest that ACN is crucial for the development of cognitive impairment, AD neuropathology, and neuroinflammation. Astrocyte-specific CaNB1 deletion is beneficial for both the abolishment of AβO-mediated detrimental effects and treatment of ongoing AD-related pathology, hence representing an intriguing target for AD therapy.
Collapse
Affiliation(s)
- Laura Tapella
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale, Novara, Italy
| | - Giulia Dematteis
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale, Novara, Italy
| | - Pietro La Vitola
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milano, Italy
| | - Susanna Leva
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milano, Italy
| | - Elisa Tonelli
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale, Novara, Italy
| | - Marco Raddi
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale, Novara, Italy
| | - Marta Delconti
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale, Novara, Italy
| | - Letizia Dacomo
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milano, Italy
| | - Alberto La Macchia
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milano, Italy
| | - Elisa Murari
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milano, Italy
| | - Maria Talmon
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale, Novara, Italy
| | - Justyna Malecka
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale, Novara, Italy
| | - Gabriela Chrostek
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale, Novara, Italy
| | - Mariagrazia Grilli
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale, Novara, Italy
| | - Laura Colombo
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milano, Italy
| | - Mario Salmona
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milano, Italy
| | - Gianluigi Forloni
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milano, Italy
| | - Armando A Genazzani
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale, Novara, Italy
| | - Claudia Balducci
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milano, Italy
| | - Dmitry Lim
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale, Novara, Italy
| |
Collapse
|
9
|
Kraner SD, Sompol P, Prateeptrang S, Promkan M, Hongthong S, Thongsopha N, Nelson PT, Norris CM. Development of a monoclonal antibody specific for a calpain-generated ∆48 kDa calcineurin fragment, a marker of distressed astrocytes. J Neurosci Methods 2024; 402:110012. [PMID: 37984591 PMCID: PMC10841921 DOI: 10.1016/j.jneumeth.2023.110012] [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: 09/14/2023] [Revised: 10/23/2023] [Accepted: 11/09/2023] [Indexed: 11/22/2023]
Abstract
BACKGROUND Calcineurin (CN) is a Ca2+/calmodulin-dependent protein phosphatase. In healthy tissue, CN exists mainly as a full-length (∼60 kDa) highly-regulated protein phosphatase involved in essential cellular functions. However, in diseased or injured tissue, CN is proteolytically converted to a constitutively active fragment that has been causatively-linked to numerous pathophysiologic processes. These calpain-cleaved CN fragments (∆CN) appear at high levels in human brain at early stages of cognitive decline associated with Alzheimer's disease (AD). NEW METHOD We developed a monoclonal antibody to ∆CN, using an immunizing peptide corresponding to the C-terminal end of the ∆CN fragment. RESULTS We obtained a mouse monoclonal antibody, designated 26A6, that selectively detects ∆CN in Western analysis of calpain-cleaved recombinant human CN. Using this antibody, we screened both pathological and normal human brain sections provided by the University of Kentucky's Alzheimer's Disease Research Center. 26A6 showed low reactivity towards normal brain tissue, but detected astrocytes both surrounding AD amyloid plaques and throughout AD brain tissue. In brain tissue with infarcts, there was considerable concentration of 26A6-positive astrocytes within/around infarcts, suggesting a link with anoxic/ischemia pathways. COMPARISON WITH EXISTING METHOD The results obtained with the new monoclonal are similar to those obtained with a polyclonal we had previously developed. However, the monoclonal is an abundant tool available to the dementia research community. CONCLUSIONS The new monoclonal 26A6 antibody is highly selective for the ∆CN proteolytic fragment and labels a subset of astrocytes, and could be a useful tool for marking insidious brain pathology and identifying novel astrocyte phenotypes.
Collapse
Affiliation(s)
| | - Pradoldej Sompol
- Sanders Brown Center on Aging, USA; Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY 40536, USA
| | - Siriyagon Prateeptrang
- Sanders Brown Center on Aging, USA; School of Allied Health Science, Walailak University, Nakhon Si Thammarat 80160, Thailand
| | - Moltira Promkan
- Sanders Brown Center on Aging, USA; Department of Clinical Microscopy, Faculty of Medical Technology, Mahidol University, Nakhon Pathom 73170, Thailand
| | - Suthida Hongthong
- Sanders Brown Center on Aging, USA; School of Allied Health Science, Walailak University, Nakhon Si Thammarat 80160, Thailand
| | - Napasorn Thongsopha
- Sanders Brown Center on Aging, USA; School of Allied Health Science, Walailak University, Nakhon Si Thammarat 80160, Thailand
| | - Peter T Nelson
- Sanders Brown Center on Aging, USA; Department of Pathology, University of Kentucky, Lexington, KY 40536, USA
| | - Christopher M Norris
- Sanders Brown Center on Aging, USA; Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY 40536, USA.
| |
Collapse
|
10
|
Lullmann O, van der Plas E, Harshman LA. Understanding the impact of pediatric kidney transplantation on cognition: A review of the literature. Pediatr Transplant 2023; 27:e14597. [PMID: 37664967 PMCID: PMC11034761 DOI: 10.1111/petr.14597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 08/15/2023] [Accepted: 08/17/2023] [Indexed: 09/05/2023]
Abstract
BACKGROUND Chronic kidney disease (CKD) is a relatively rare childhood disease that is associated with a wide array of medical comorbidities. Roughly half of all pediatric patients acquire CKD due to congenital anomalies of the kidneys and urinary tract, and of those with congenital disease, 50% will progress to end-stage kidney disease (ESKD) necessitating a kidney transplantation. The medical sequelae of advanced CKD/ESKD improve dramatically following successful kidney transplantation; however, the impact of kidney transplantation on neurocognition in children is less clear. It is generally thought that cognition improves following kidney transplantation; however, our knowledge on this topic is limited by the sparsity of high-quality data in the context of the relative rarity of pediatric CKD/ESKD. METHOD We conducted a narrative review to gauge the scope of the literature, using the PubMed database and the following keywords: cognition, kidney, brain, pediatric, neurocognition, intelligence, executive function, transplant, immunosuppression, and neuroimaging. RESULTS There are few published longitudinal studies, and existing work often includes wide heterogeneity in age at transplant, variable dialysis exposure/duration prior to transplant, and unaccounted cofounders which persist following transplantation, including socio-economic status. Furthermore, the impact of long-term maintenance immunosuppression on the brain and cognitive function of pediatric kidney transplant (KT) recipients remains unknown. CONCLUSION In this educational review, we highlight what is known on the topic of neurocognition and neuroimaging in the pediatric KT population.
Collapse
Affiliation(s)
- Olivia Lullmann
- Stead Family Department of Pediatrics, University of Iowa Carver College of Medicine; Iowa City, IA
| | - Ellen van der Plas
- Stead Family Department of Pediatrics, University of Iowa Carver College of Medicine; Iowa City, IA
- Department of Pediatrics, University of Arkansas for Medical Sciences College of Medicine; Little Rock, AR
| | - Lyndsay A. Harshman
- Stead Family Department of Pediatrics, University of Iowa Carver College of Medicine; Iowa City, IA
| |
Collapse
|
11
|
Rodríguez-Vega A, Dutra-Tavares AC, Souza TP, Semeão KA, Filgueiras CC, Ribeiro-Carvalho A, Manhães AC, Abreu-Villaça Y. Nicotine Exposure in a Phencyclidine-Induced Mice Model of Schizophrenia: Sex-Selective Medial Prefrontal Cortex Protein Markers of the Combined Insults in Adolescent Mice. Int J Mol Sci 2023; 24:14634. [PMID: 37834084 PMCID: PMC10572990 DOI: 10.3390/ijms241914634] [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: 07/19/2023] [Revised: 09/20/2023] [Accepted: 09/22/2023] [Indexed: 10/15/2023] Open
Abstract
Tobacco misuse as a comorbidity of schizophrenia is frequently established during adolescence. However, comorbidity markers are still missing. Here, the method of label-free proteomics was used to identify deregulated proteins in the medial prefrontal cortex (prelimbic and infralimbic) of male and female mice modelled to schizophrenia with a history of nicotine exposure during adolescence. Phencyclidine (PCP), used to model schizophrenia (SCHZ), was combined with an established model of nicotine minipump infusions (NIC). The combined insults led to worse outcomes than each insult separately when considering the absolute number of deregulated proteins and that of exclusively deregulated ones. Partially shared Reactome pathways between sexes and between PCP, NIC and PCPNIC groups indicate functional overlaps. Distinctively, proteins differentially expressed exclusively in PCPNIC mice reveal unique effects associated with the comorbidity model. Interactome maps of these proteins identified sex-selective subnetworks, within which some proteins stood out: for females, peptidyl-prolyl cis-trans isomerase (Fkbp1a) and heat shock 70 kDa protein 1B (Hspa1b), both components of the oxidative stress subnetwork, and gamma-enolase (Eno2), a component of the energy metabolism subnetwork; and for males, amphiphysin (Amph), a component of the synaptic transmission subnetwork. These are proposed to be further investigated and validated as markers of the combined insult during adolescence.
Collapse
Affiliation(s)
- Andrés Rodríguez-Vega
- Laboratório de Neurofisiologia, Departamento de Ciências Fisiológicas, Instituto de Biologia Roberto Alcantara Gomes, Universidade do Estado do Rio de Janeiro (UERJ), Rio de Janeiro 20550-170, RJ, Brazil; (A.R.-V.); (A.C.D.-T.); (T.P.S.); (K.A.S.); (C.C.F.); (A.C.M.)
| | - Ana Carolina Dutra-Tavares
- Laboratório de Neurofisiologia, Departamento de Ciências Fisiológicas, Instituto de Biologia Roberto Alcantara Gomes, Universidade do Estado do Rio de Janeiro (UERJ), Rio de Janeiro 20550-170, RJ, Brazil; (A.R.-V.); (A.C.D.-T.); (T.P.S.); (K.A.S.); (C.C.F.); (A.C.M.)
| | - Thainá P. Souza
- Laboratório de Neurofisiologia, Departamento de Ciências Fisiológicas, Instituto de Biologia Roberto Alcantara Gomes, Universidade do Estado do Rio de Janeiro (UERJ), Rio de Janeiro 20550-170, RJ, Brazil; (A.R.-V.); (A.C.D.-T.); (T.P.S.); (K.A.S.); (C.C.F.); (A.C.M.)
| | - Keila A. Semeão
- Laboratório de Neurofisiologia, Departamento de Ciências Fisiológicas, Instituto de Biologia Roberto Alcantara Gomes, Universidade do Estado do Rio de Janeiro (UERJ), Rio de Janeiro 20550-170, RJ, Brazil; (A.R.-V.); (A.C.D.-T.); (T.P.S.); (K.A.S.); (C.C.F.); (A.C.M.)
| | - Claudio C. Filgueiras
- Laboratório de Neurofisiologia, Departamento de Ciências Fisiológicas, Instituto de Biologia Roberto Alcantara Gomes, Universidade do Estado do Rio de Janeiro (UERJ), Rio de Janeiro 20550-170, RJ, Brazil; (A.R.-V.); (A.C.D.-T.); (T.P.S.); (K.A.S.); (C.C.F.); (A.C.M.)
| | - Anderson Ribeiro-Carvalho
- Departamento de Ciências, Faculdade de Formação de Professores da Universidade do Estado do Rio de Janeiro, São Gonçalo 24435-005, RJ, Brazil;
| | - Alex C. Manhães
- Laboratório de Neurofisiologia, Departamento de Ciências Fisiológicas, Instituto de Biologia Roberto Alcantara Gomes, Universidade do Estado do Rio de Janeiro (UERJ), Rio de Janeiro 20550-170, RJ, Brazil; (A.R.-V.); (A.C.D.-T.); (T.P.S.); (K.A.S.); (C.C.F.); (A.C.M.)
| | - Yael Abreu-Villaça
- Laboratório de Neurofisiologia, Departamento de Ciências Fisiológicas, Instituto de Biologia Roberto Alcantara Gomes, Universidade do Estado do Rio de Janeiro (UERJ), Rio de Janeiro 20550-170, RJ, Brazil; (A.R.-V.); (A.C.D.-T.); (T.P.S.); (K.A.S.); (C.C.F.); (A.C.M.)
| |
Collapse
|
12
|
Griffioen G. Calcium Dyshomeostasis Drives Pathophysiology and Neuronal Demise in Age-Related Neurodegenerative Diseases. Int J Mol Sci 2023; 24:13243. [PMID: 37686048 PMCID: PMC10487569 DOI: 10.3390/ijms241713243] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 08/21/2023] [Accepted: 08/23/2023] [Indexed: 09/10/2023] Open
Abstract
This review postulates that age-related neurodegeneration entails inappropriate activation of intrinsic pathways to enable brain plasticity through deregulated calcium (Ca2+) signalling. Ca2+ in the cytosol comprises a versatile signal controlling neuronal cell physiology to accommodate adaptive structural and functional changes of neuronal networks (neuronal plasticity) and, as such, is essential for brain function. Although disease risk factors selectively affect different neuronal cell types across age-related neurodegenerative diseases (NDDs), these appear to have in common the ability to impair the specificity of the Ca2+ signal. As a result, non-specific Ca2+ signalling facilitates the development of intraneuronal pathophysiology shared by age-related NDDs, including mitochondrial dysfunction, elevated reactive oxygen species (ROS) levels, impaired proteostasis, and decreased axonal transport, leading to even more Ca2+ dyshomeostasis. These core pathophysiological processes and elevated cytosolic Ca2+ levels comprise a self-enforcing feedforward cycle inevitably spiralling toward high levels of cytosolic Ca2+. The resultant elevated cytosolic Ca2+ levels ultimately gear otherwise physiological effector pathways underlying plasticity toward neuronal demise. Ageing impacts mitochondrial function indiscriminately of the neuronal cell type and, therefore, contributes to the feedforward cycle of pathophysiology development seen in all age-related NDDs. From this perspective, therapeutic interventions to safely restore Ca2+ homeostasis would mitigate the excessive activation of neuronal destruction pathways and, therefore, are expected to have promising neuroprotective potential.
Collapse
|
13
|
Rodrigues YE, Tigaret CM, Marie H, O'Donnell C, Veltz R. A stochastic model of hippocampal synaptic plasticity with geometrical readout of enzyme dynamics. eLife 2023; 12:e80152. [PMID: 37589251 PMCID: PMC10435238 DOI: 10.7554/elife.80152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 03/22/2023] [Indexed: 08/18/2023] Open
Abstract
Discovering the rules of synaptic plasticity is an important step for understanding brain learning. Existing plasticity models are either (1) top-down and interpretable, but not flexible enough to account for experimental data, or (2) bottom-up and biologically realistic, but too intricate to interpret and hard to fit to data. To avoid the shortcomings of these approaches, we present a new plasticity rule based on a geometrical readout mechanism that flexibly maps synaptic enzyme dynamics to predict plasticity outcomes. We apply this readout to a multi-timescale model of hippocampal synaptic plasticity induction that includes electrical dynamics, calcium, CaMKII and calcineurin, and accurate representation of intrinsic noise sources. Using a single set of model parameters, we demonstrate the robustness of this plasticity rule by reproducing nine published ex vivo experiments covering various spike-timing and frequency-dependent plasticity induction protocols, animal ages, and experimental conditions. Our model also predicts that in vivo-like spike timing irregularity strongly shapes plasticity outcome. This geometrical readout modelling approach can be readily applied to other excitatory or inhibitory synapses to discover their synaptic plasticity rules.
Collapse
Affiliation(s)
- Yuri Elias Rodrigues
- Université Côte d’AzurNiceFrance
- Institut de Pharmacologie Moléculaire et Cellulaire (IPMC), CNRSValbonneFrance
- Inria Center of University Côte d’Azur (Inria)Sophia AntipolisFrance
| | - Cezar M Tigaret
- Neuroscience and Mental Health Research Innovation Institute, Division of Psychological Medicine and Clinical Neurosciences,School of Medicine, Cardiff UniversityCardiffUnited Kingdom
| | - Hélène Marie
- Université Côte d’AzurNiceFrance
- Institut de Pharmacologie Moléculaire et Cellulaire (IPMC), CNRSValbonneFrance
| | - Cian O'Donnell
- School of Computing, Engineering, and Intelligent Systems, Magee Campus, Ulster UniversityLondonderryUnited Kingdom
- School of Computer Science, Electrical and Electronic Engineering, and Engineering Mathematics, University of BristolBristolUnited Kingdom
| | - Romain Veltz
- Inria Center of University Côte d’Azur (Inria)Sophia AntipolisFrance
| |
Collapse
|
14
|
Teramoto H, Hirashima N, Tanaka M. Calcineurin B1 Deficiency Reduces Proliferation, Increases Apoptosis, and Alters Secretion in Enteric Glial Cells of Mouse Small Intestine in Culture. Cells 2023; 12:1867. [PMID: 37508531 PMCID: PMC10378349 DOI: 10.3390/cells12141867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2023] [Revised: 06/30/2023] [Accepted: 07/15/2023] [Indexed: 07/30/2023] Open
Abstract
To investigate the roles of calcineurin (CN) in glial cells, we previously generated conditional knockout (CKO) mice lacking CNB1 in glial cells. Because these CKO mice showed dysfunction and inflammation of the small intestine in addition to growth impairment and postweaning death, we have focused on enteric glial cells (EGCs) in the small intestine. In this study, we examined the effects of CNB1 deficiency on the proliferation and survival of EGCs and the expression and secretion of EGC-derived substances in culture to reveal the mechanisms of how CNB1 deficiency leads to dysfunction and inflammation of the small intestine. In primary myenteric cultures of the small intestine, EGCs from the CKO mice showed reduced proliferation and increased apoptosis compared with EGCs from control mice. In purified EGC cultures from the CKO mice, Western blot analysis showed increased expression of S100B, iNOS, GFAP, and GDNF, and increased phosphorylation of NF-κB p65. In the supernatants of purified EGC cultures from the CKO mice, ELISA showed reduced secretion of TGF-β1. In contrast, GDNF secretion was not altered in purified EGC cultures from the CKO mice. Furthermore, treatment with an S100B inhibitor partially rescued the CKO mice from growth impairment and postweaning death in vivo. In conclusion, CNB1 deficiency leads to reduced proliferation and increased apoptosis of EGCs and abnormal expression and secretion of EGC-derived substances, which may contribute to dysfunction and inflammation of the small intestine.
Collapse
Affiliation(s)
- Hikaru Teramoto
- Department of Cellular Biophysics, Graduate School of Pharmaceutical Sciences, Nagoya City University, Tanabe-dori, Mizuho-ku, Nagoya 467-8603, Japan
| | - Naohide Hirashima
- Department of Cellular Biophysics, Graduate School of Pharmaceutical Sciences, Nagoya City University, Tanabe-dori, Mizuho-ku, Nagoya 467-8603, Japan
| | - Masahiko Tanaka
- Department of Cellular Biophysics, Graduate School of Pharmaceutical Sciences, Nagoya City University, Tanabe-dori, Mizuho-ku, Nagoya 467-8603, Japan
| |
Collapse
|
15
|
Lim D, Tapella L, Dematteis G, Talmon M, Genazzani AA. Calcineurin Signalling in Astrocytes: From Pathology to Physiology and Control of Neuronal Functions. Neurochem Res 2023; 48:1077-1090. [PMID: 36083398 PMCID: PMC10030417 DOI: 10.1007/s11064-022-03744-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Revised: 07/31/2022] [Accepted: 08/29/2022] [Indexed: 10/14/2022]
Abstract
Calcineurin (CaN), a Ca2+/calmodulin-activated serine/threonine phosphatase, acts as a Ca2+-sensitive switch regulating cellular functions through protein dephosphorylation and activation of gene transcription. In astrocytes, the principal homeostatic cells in the CNS, over-activation of CaN is known to drive pathological transcriptional remodelling, associated with neuroinflammation in diseases such as Alzheimer's disease, epilepsy and brain trauma. Recent reports suggest that, in physiological conditions, the activity of CaN in astrocytes is transcription-independent and is required for maintenance of basal protein synthesis rate and activation of astrocytic Na+/K+ pump thereby contributing to neuronal functions such as neuronal excitability and memory formation. In this contribution we overview the role of Ca2+ and CaN signalling in astroglial pathophysiology focusing on the emerging physiological role of CaN in astrocytes. We propose a model for the context-dependent switch of CaN activity from the post-transcriptional regulation of cell proteostasis in healthy astrocytes to the CaN-dependent transcriptional activation in neuroinflammation-associated diseases.
Collapse
Affiliation(s)
- Dmitry Lim
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale "Amedeo Avogadro", Via Bovio 6, 28100, Novara, Italy.
| | - Laura Tapella
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale "Amedeo Avogadro", Via Bovio 6, 28100, Novara, Italy
| | - Giulia Dematteis
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale "Amedeo Avogadro", Via Bovio 6, 28100, Novara, Italy
| | - Maria Talmon
- Department of Health Sciences, School of Medicine, Università del Piemonte Orientale "Amedeo Avogadro", Via Solaroli 17, 28100, Novara, Italy
| | - Armando A Genazzani
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale "Amedeo Avogadro", Via Bovio 6, 28100, Novara, Italy.
| |
Collapse
|
16
|
Chi X, Wang L, Liu H, Zhang Y, Shen W. Post-stroke cognitive impairment and synaptic plasticity: A review about the mechanisms and Chinese herbal drugs strategies. Front Neurosci 2023; 17:1123817. [PMID: 36937659 PMCID: PMC10014821 DOI: 10.3389/fnins.2023.1123817] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 02/13/2023] [Indexed: 03/06/2023] Open
Abstract
Post-stroke cognitive impairment, is a major complication of stroke, characterized by cognitive dysfunction, which directly affects the quality of life. Post-stroke cognitive impairment highlights the causal relationship between stroke and cognitive impairment. The pathological damage of stroke, including the increased release of excitatory amino acids, oxidative stress, inflammatory responses, apoptosis, changed neurotrophic factor levels and gene expression, influence synaptic plasticity. Synaptic plasticity refers to the activity-dependent changes in the strength of synaptic connections and efficiency of synaptic transmission at pre-existing synapses and can be divided into structural synaptic plasticity and functional synaptic plasticity. Changes in synaptic plasticity have been proven to play important roles in the occurrence and treatment of post-stroke cognitive impairment. Evidence has indicated that Chinese herbal drugs have effect of treating post-stroke cognitive impairment. In this review, we overview the influence of pathological damage of stroke on synaptic plasticity, analyze the changes of synaptic plasticity in post-stroke cognitive impairment, and summarize the commonly used Chinese herbal drugs whose active ingredient or extracts can regulate synaptic plasticity. This review will summarize the relationship between post-stroke cognitive impairment and synaptic plasticity, provide new ideas for future exploration of the mechanism of post-stroke cognitive impairment, compile evidence of applying Chinese herbal drugs to treat post-stroke cognitive impairment and lay a foundation for the development of novel formulas for treating post-stroke cognitive impairment.
Collapse
Affiliation(s)
- Xiansu Chi
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Liuding Wang
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Hongxi Liu
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Yunling Zhang
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Wei Shen
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
17
|
Gore SV, Kakodkar R, Del Rosario Hernández T, Edmister ST, Creton R. Zebrafish Larvae Position Tracker (Z-LaP Tracker): a high-throughput deep-learning behavioral approach for the identification of calcineurin pathway-modulating drugs using zebrafish larvae. Sci Rep 2023; 13:3174. [PMID: 36823315 PMCID: PMC9950053 DOI: 10.1038/s41598-023-30303-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Accepted: 02/21/2023] [Indexed: 02/25/2023] Open
Abstract
Brain function studies greatly depend on quantification and analysis of behavior. While behavior can be imaged efficiently, the quantification of specific aspects of behavior is labor-intensive and may introduce individual biases. Recent advances in deep learning and artificial intelligence-based tools have made it possible to precisely track individual features of freely moving animals in diverse environments without any markers. In the current study, we developed Zebrafish Larvae Position Tracker (Z-LaP Tracker), a modification of the markerless position estimation software DeepLabCut, to quantify zebrafish larval behavior in a high-throughput 384-well setting. We utilized the high-contrast features of our model animal, zebrafish larvae, including the eyes and the yolk for our behavioral analysis. Using this experimental setup, we quantified relevant behaviors with similar accuracy to the analysis performed by humans. The changes in behavior were organized in behavioral profiles, which were examined by K-means and hierarchical cluster analysis. Calcineurin inhibitors exhibited a distinct behavioral profile characterized by increased activity, acoustic hyperexcitability, reduced visually guided behaviors, and reduced habituation to acoustic stimuli. The developed methodologies were used to identify 'CsA-type' drugs that might be promising candidates for the prevention and treatment of neurological disorders.
Collapse
Affiliation(s)
- Sayali V. Gore
- grid.40263.330000 0004 1936 9094Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, 185 Meeting Street, Providence, RI 02912 USA
| | - Rohit Kakodkar
- grid.40263.330000 0004 1936 9094Center for Computation and Visualization, Brown University, Providence, RI USA
| | - Thaís Del Rosario Hernández
- grid.40263.330000 0004 1936 9094Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, 185 Meeting Street, Providence, RI 02912 USA
| | - Sara Tucker Edmister
- grid.40263.330000 0004 1936 9094Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, 185 Meeting Street, Providence, RI 02912 USA
| | - Robbert Creton
- grid.40263.330000 0004 1936 9094Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, 185 Meeting Street, Providence, RI 02912 USA
| |
Collapse
|
18
|
Zarei F, Moazedi AA, Salimi Z, Pourmotabbed A, Yousofvand N, Farshad M, Akrami MR. Activation of androgen receptors alters hippocampal synaptic plasticity and memory retention through modulation of L-type calcium channels. Life Sci 2023; 314:121155. [PMID: 36379312 DOI: 10.1016/j.lfs.2022.121155] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Revised: 10/27/2022] [Accepted: 11/02/2022] [Indexed: 11/15/2022]
Abstract
AIMS It has been revealed that membrane androgen receptor activation modulates avoidance memory and synaptic plasticity. In a previous study, we showed that Calcineurin, a calcium dependent phosphatase, could be a potential mediator of these AR effects. Also, it is reported that AR activation leads to L-type calcium channel activation. The aim of the current study is to test whether L-type calcium channels are downstream of AR and whether this signal pathway mediates the impairment effect of androgenic steroids on passive avoidance memory and synaptic plasticity. MATERIALS AND METHODS We measured the effect of Nandrolone Decanoate (AR agonist), AR antagonist (Nilutamide) plus ND or L-type calcium channel inhibitor (Nifedipine) plus ND on passive avoidance performance of adolescent male rats. For extracellular field potential recordings hippocampal slices were perfused with ND, Nilutamide-ND or Nifedipine-ND. KEY FINDINGS Our results clarified that AR activation by ND could impair avoidance behavior as step through latency decreased in ND-treated group while application of both Nilutamide and Nifedipine reestablished normal avoidance behavior. Also, LTP induction in the CA1 area of hippocampus was diminished by ND perfusion and both AR antagonist and L-type calcium channel inhibitor application lead to normal LTP. These findings support our hypothesis that activation of L-type calcium channels are involved in ARs mechanism effects on both avoidance behavior and hippocampal synaptic plasticity. SIGNIFICANCE Understanding the biological effects of AR agonists on cognitive processes and its cellular mechanism may be a new/supplementary way to treating fear-related disorders.
Collapse
Affiliation(s)
- Fatemeh Zarei
- Department of Physiology, Kermanshah University of Medical Sciences, Kermanshah, Iran; Fertility and Infertility Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran; Department of Biology, Shahid Chamran University of Ahvaz, Ahvaz, Iran; Department of Biology, Faculty of Sciences, Razi University, Kermanshah, Iran.
| | - Ahmad Ali Moazedi
- Department of Biology, Shahid Chamran University of Ahvaz, Ahvaz, Iran; Stem Cell and Transgenic Technology Research Center, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| | - Zahra Salimi
- Department of Physiology, Kermanshah University of Medical Sciences, Kermanshah, Iran; Fertility and Infertility Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran; Department of Biology, Centre for Biomedical Research, University of Victoria, Victoria, BC V8P 5C2, Canada; Department of Biology, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| | - Ali Pourmotabbed
- Department of Physiology, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Namdar Yousofvand
- Department of Biology, Centre for Biomedical Research, University of Victoria, Victoria, BC V8P 5C2, Canada
| | - Moradpour Farshad
- Department of Physiology, Kermanshah University of Medical Sciences, Kermanshah, Iran; Fertility and Infertility Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran; Department of Biology, Centre for Biomedical Research, University of Victoria, Victoria, BC V8P 5C2, Canada
| | - Mohammad Reza Akrami
- Department of Neurosurgery, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| |
Collapse
|
19
|
Armstrong NS, Frank CA. The calcineurin regulator Sarah enables distinct forms of homeostatic plasticity at the Drosophila neuromuscular junction. Front Synaptic Neurosci 2023; 14:1033743. [PMID: 36685082 PMCID: PMC9846150 DOI: 10.3389/fnsyn.2022.1033743] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 12/05/2022] [Indexed: 01/05/2023] Open
Abstract
Introduction: The ability of synapses to maintain physiological levels of evoked neurotransmission is essential for neuronal stability. A variety of perturbations can disrupt neurotransmission, but synapses often compensate for disruptions and work to stabilize activity levels, using forms of homeostatic synaptic plasticity. Presynaptic homeostatic potentiation (PHP) is one such mechanism. PHP is expressed at the Drosophila melanogaster larval neuromuscular junction (NMJ) synapse, as well as other NMJs. In PHP, presynaptic neurotransmitter release increases to offset the effects of impairing muscle transmitter receptors. Prior Drosophila work has studied PHP using different ways to perturb muscle receptor function-either acutely (using pharmacology) or chronically (using genetics). Some of our prior data suggested that cytoplasmic calcium signaling was important for expression of PHP after genetic impairment of glutamate receptors. Here we followed up on that observation. Methods: We used a combination of transgenic Drosophila RNA interference and overexpression lines, along with NMJ electrophysiology, synapse imaging, and pharmacology to test if regulators of the calcium/calmodulin-dependent protein phosphatase calcineurin are necessary for the normal expression of PHP. Results: We found that either pre- or postsynaptic dysregulation of a Drosophila gene regulating calcineurin, sarah (sra), blocks PHP. Tissue-specific manipulations showed that either increases or decreases in sra expression are detrimental to PHP. Additionally, pharmacologically and genetically induced forms of expression of PHP are functionally separable depending entirely upon which sra genetic manipulation is used. Surprisingly, dual-tissue pre- and postsynaptic sra knockdown or overexpression can ameliorate PHP blocks revealed in single-tissue experiments. Pharmacological and genetic inhibition of calcineurin corroborated this latter finding. Discussion: Our results suggest tight calcineurin regulation is needed across multiple tissue types to stabilize peripheral synaptic outputs.
Collapse
Affiliation(s)
- Noah S. Armstrong
- Department of Anatomy and Cell Biology, University of Iowa Carver College of Medicine, Iowa City, IA, United States,Interdisciplinary Graduate Program in Neuroscience, University of Iowa, Iowa City, IA, United States
| | - C. Andrew Frank
- Department of Anatomy and Cell Biology, University of Iowa Carver College of Medicine, Iowa City, IA, United States,Interdisciplinary Graduate Program in Neuroscience, University of Iowa, Iowa City, IA, United States,Iowa Neuroscience Institute, University of Iowa, Iowa City, IA, United States,*Correspondence: C. Andrew Frank
| |
Collapse
|
20
|
Norris CM. Is It Time to Repurpose Calcineurin Inhibitors for the Treatment of Cognitive Impairment and Dementia? J Alzheimers Dis 2023; 95:599-602. [PMID: 37661889 PMCID: PMC10947791 DOI: 10.3233/jad-230780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/05/2023]
Abstract
Numerous preclinical and human tissue studies implicate the protein phosphatase calcineurin (CN) as a pathophysiologic mechanism in Alzheimer's disease (AD) and other neurodegenerative conditions. Using public electronic records of tens of thousands of individuals across the United States, Silva et al. (2023) show that use of the FDA-approved CN inhibitor, tacrolimus (for purposes of immunosuppression) is also associated with reduced prevalence of dementia-related symptoms. Notably, the study controls for age, sex, and race as well as multiple risk factors for AD. The results suggest that tacrolimus, and possibly other immunosuppressants could be repurposed for the treatment of AD-related dementia.
Collapse
Affiliation(s)
- Christopher M. Norris
- Sanders-Brown Center on Aging, University of Kentucky College of Medicine, Lexington, KY, USA
| |
Collapse
|
21
|
Long-term cyclosporine A treatment promotes anxiety-like behavior: Possible relation with glutamate signaling in rat hippocampus. JOURNAL OF AFFECTIVE DISORDERS REPORTS 2022. [DOI: 10.1016/j.jadr.2022.100394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
|
22
|
Hamoudi W, Tripathi MK, Ojha SK, Amal H. A cross-talk between nitric oxide and the glutamatergic system in a Shank3 mouse model of autism. Free Radic Biol Med 2022; 188:83-91. [PMID: 35716826 DOI: 10.1016/j.freeradbiomed.2022.06.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Revised: 06/06/2022] [Accepted: 06/07/2022] [Indexed: 11/21/2022]
Abstract
Nitric oxide (NO) is a multifunctional signaling molecule that plays a crucial role in synaptic transmission and neuronal function. Pioneering studies show that nitric oxide (NO) and S-nitrosylation (SNO, the NO-mediated posttranslational modification) can engender nitrosative stress in the brain, contributing to neurodegenerative diseases. Little is known, however, about the aberrant NO signaling in neurodevelopmental disorders including autism spectrum disorder (ASD). We have recently shown that the Shank3 mutation in mice representing a model of ASD causes excessive NO levels and aberrant protein SNO. The glutamatergic system is involved in ASD, specifically in SHANK3 pathology. We used SNOTRAP technology to identify the SNO-proteome in the brain of the Shank3 mutant mice to understand the role of SNO in the glutamatergic system during the development of these mice. We conducted a systems biology analysis of the SNO-proteome to investigate the biological processes and signaling pathways in the brain of juvenile and adult Shank3 mutant and wild-type mice. The Shank3 mutation caused significant SNO-enrichment of a glutamate signaling pathway in the juvenile and adult mutant mice, although different protein subsets were S-nitrosylated in both ages. Cellular compartments analysis showed that "glutamatergic Synapse" is SNO-enriched significantly in the mutant mice of both ages. We also found eight S-nitrosylated proteins involved in glutamate transmission in both ages. 38 SNO-proteins found in the mutant mice are among the high-risk SFARI gene list. Biochemical examination shows a reduction in the levels of NMDA Receptor (NR1) in the cortex and striatum of the mutant mice of both ages. Neuronal NOS knockdown in SHSY-5Y rescued NR1 levels. In conclusion, this study reveals novel SNO of key glutamatergic proteins in Shank3 mutant mice and a cross-talk between nitric oxide and the glutamatergic system.
Collapse
Affiliation(s)
- Wajeha Hamoudi
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, 91120, Israel
| | - Manish Kumar Tripathi
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, 91120, Israel
| | - Shashank Kumar Ojha
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, 91120, Israel
| | - Haitham Amal
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, 91120, Israel.
| |
Collapse
|
23
|
The optimized core peptide derived from CABIN1 efficiently inhibits calcineurin-mediated T-cell activation. Exp Mol Med 2022; 54:613-625. [PMID: 35550603 PMCID: PMC9166766 DOI: 10.1038/s12276-022-00772-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 01/11/2022] [Accepted: 02/15/2022] [Indexed: 11/08/2022] Open
Abstract
The C-terminal fragment of CABIN1 interacts with calcineurin and represses the transcriptional activity of the nuclear factor of activated T cells (NFAT). However, the specific sequences and mechanisms through which it binds to calcineurin are unclear. This study determined that decameric peptide (CABIN1 residues 2146–2155) is minimally required for binding to calcineurin. This peptide contains a unique “PPTP” C-terminal sequence and a “PxIxIT” N-terminal motif. Furthermore, p38MAPK phosphorylated the threonine residue of the “PPTP” sequence under physiological conditions, dramatically enhancing the peptide’s binding affinity to calcineurin. Therefore, the CABIN1 peptide inhibited the calcineurin-NFAT pathway and the activation of T cells more efficiently than the VIVIT peptide without affecting calcineurin’s phosphatase activity. The CABIN1 peptide could thus be a more potent calcineurin inhibitor and provide therapeutic opportunities for various diseases caused by the calcineurin-NFAT pathway. A peptide with therapeutic potential binds strongly to the cellular enzyme calcineurin and may prove valuable in anti-cancer and autoimmune disease treatments. Many cancers and autoimmune diseases are linked with overactivation of a key calcineurin-related pathway which is heavily involved in T cell activation. This pathway has long been a therapeutic target, but existing drugs show problems with stability and delivery, and can cause serious side effects. One known inhibitor of calcineurin is the protein CABIN1, but precisely how well it binds and how useful it may be is unclear. Now, Hong-Duk Youn at Seoul National University College of Medicine, South Korea, and co-workers have identified how one specific peptide from CABIN1 binds strongly to calcineurin. The CABIN1 peptide was stable and displayed greater efficiency at inhibiting calcineurin than another recently identified peptide candidate.
Collapse
|
24
|
Stacho M, Manahan-Vaughan D. The Intriguing Contribution of Hippocampal Long-Term Depression to Spatial Learning and Long-Term Memory. Front Behav Neurosci 2022; 16:806356. [PMID: 35548697 PMCID: PMC9084281 DOI: 10.3389/fnbeh.2022.806356] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Accepted: 03/10/2022] [Indexed: 01/03/2023] Open
Abstract
Long-term potentiation (LTP) and long-term depression (LTD) comprise the principal cellular mechanisms that fulfill established criteria for the physiological correlates of learning and memory. Traditionally LTP, that increases synaptic weights, has been ascribed a prominent role in learning and memory whereas LTD, that decreases them, has often been relegated to the category of "counterpart to LTP" that serves to prevent saturation of synapses. In contradiction of these assumptions, studies over the last several years have provided functional evidence for distinct roles of LTD in specific aspects of hippocampus-dependent associative learning and information encoding. Furthermore, evidence of the experience-dependent "pruning" of excitatory synapses, the majority of which are located on dendritic spines, by means of LTD has been provided. In addition, reports exist of the temporal and physical restriction of LTP in dendritic compartments by means of LTD. Here, we discuss the role of LTD and LTP in experience-dependent information encoding based on empirical evidence derived from conjoint behavioral and electrophysiological studies conducted in behaving rodents. We pinpoint the close interrelation between structural modifications of dendritic spines and the occurrence of LTP and LTD. We report on findings that support that whereas LTP serves to acquire the general scheme of a spatial representation, LTD enables retention of content details. We argue that LTD contributes to learning by engaging in a functional interplay with LTP, rather than serving as its simple counterpart, or negator. We propose that similar spatial experiences that share elements of neuronal representations can be modified by means of LTD to enable pattern separation. Therewith, LTD plays a crucial role in the disambiguation of similar spatial representations and the prevention of generalization.
Collapse
|
25
|
Dematteis G, Restelli E, Vanella VV, Manfredi M, Marengo E, Corazzari M, Genazzani AA, Chiesa R, Lim D, Tapella L. Calcineurin Controls Cellular Prion Protein Expression in Mouse Astrocytes. Cells 2022; 11:cells11040609. [PMID: 35203261 PMCID: PMC8870693 DOI: 10.3390/cells11040609] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Revised: 02/04/2022] [Accepted: 02/08/2022] [Indexed: 02/05/2023] Open
Abstract
Prion diseases arise from the conformational conversion of the cellular prion protein (PrPC) into a self-replicating prion isoform (PrPSc). Although this process has been studied mostly in neurons, a growing body of evidence suggests that astrocytes express PrPC and are able to replicate and accumulate PrPSc. Currently, prion diseases remain incurable, while downregulation of PrPC represents the most promising therapy due to the reduction of the substrate for prion conversion. Here we show that the astrocyte-specific genetic ablation or pharmacological inhibition of the calcium-activated phosphatase calcineurin (CaN) reduces PrPC expression in astrocytes. Immunocytochemical analysis of cultured CaN-KO astrocytes and isolation of synaptosomal compartments from the hippocampi of astrocyte-specific CaN-KO (ACN-KO) mice suggest that PrPC is downregulated both in vitro and in vivo. The downregulation occurs without affecting the glycosylation of PrPC and without alteration of its proteasomal or lysosomal degradation. Direct assessment of the protein synthesis rate and shotgun mass spectrometry proteomics analysis suggest that the reduction of PrPC is related to the impairment of global protein synthesis in CaN-KO astrocytes. When WT-PrP and PrP-D177N, a mouse homologue of a human mutation associated with the inherited prion disease fatal familial insomnia, were expressed in astrocytes, CaN-KO astrocytes showed an aberrant localization of both WT-PrP and PrP-D177N variants with predominant localization to the Golgi apparatus, suggesting that ablation of CaN affects both WT and mutant PrP proteins. These results provide new mechanistic details in relation to the regulation of PrP expression in astrocytes, suggesting the therapeutic potential of astroglial cells.
Collapse
Affiliation(s)
- Giulia Dematteis
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale “Amedeo Avogadro”, 28100 Novara, Italy; (G.D.); (A.A.G.)
| | - Elena Restelli
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, 20156 Milan, Italy; (E.R.); (R.C.)
| | - Virginia Vita Vanella
- Department of Translational Medicine, Center for Translational Research on Autoimmune and Allergic Disease (CAAD), Università del Piemonte Orientale “Amedeo Avogadro”, 28100 Novara, Italy; (V.V.V.); (M.M.)
| | - Marcello Manfredi
- Department of Translational Medicine, Center for Translational Research on Autoimmune and Allergic Disease (CAAD), Università del Piemonte Orientale “Amedeo Avogadro”, 28100 Novara, Italy; (V.V.V.); (M.M.)
| | - Emilio Marengo
- Department of Sciences and Technological Innovation, Università del Piemonte Orientale “Amedeo Avogadro”, 28100 Novara, Italy;
| | - Marco Corazzari
- Department of Health Science (DSS), Center for Translational Research on Autoimmune and Allergic Disease (CAAD) & Interdisciplinary Research Center of Autoimmune Diseases (IRCAD), Università del Piemonte Orientale “Amedeo Avogadro”, 28100 Novara, Italy;
| | - Armando A. Genazzani
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale “Amedeo Avogadro”, 28100 Novara, Italy; (G.D.); (A.A.G.)
| | - Roberto Chiesa
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, 20156 Milan, Italy; (E.R.); (R.C.)
| | - Dmitry Lim
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale “Amedeo Avogadro”, 28100 Novara, Italy; (G.D.); (A.A.G.)
- Correspondence: (D.L.); (L.T.); Tel.: +39-0321-375822 (L.T.)
| | - Laura Tapella
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale “Amedeo Avogadro”, 28100 Novara, Italy; (G.D.); (A.A.G.)
- Correspondence: (D.L.); (L.T.); Tel.: +39-0321-375822 (L.T.)
| |
Collapse
|
26
|
Presynaptic NMDA Receptors Influence Ca2+ Dynamics by Interacting with Voltage-Dependent Calcium Channels during the Induction of Long-Term Depression. Neural Plast 2022; 2022:2900875. [PMID: 35178084 PMCID: PMC8844386 DOI: 10.1155/2022/2900875] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2021] [Revised: 12/28/2021] [Accepted: 01/18/2022] [Indexed: 12/29/2022] Open
Abstract
Spike-timing-dependent long-term depression (t-LTD) of glutamatergic layer (L)4-L2/3 synapses in developing neocortex requires activation of astrocytes by endocannabinoids (eCBs), which release glutamate onto presynaptic NMDA receptors (preNMDARs). The exact function of preNMDARs in this context is still elusive and strongly debated. To elucidate their function, we show that bath application of the eCB 2-arachidonylglycerol (2-AG) induces a preNMDAR-dependent form of chemically induced LTD (eCB-LTD) in L2/3 pyramidal neurons in the juvenile somatosensory cortex of rats. Presynaptic Ca2+ imaging from L4 spiny stellate axons revealed that action potential (AP) evoked Ca2+ transients show a preNMDAR-dependent broadening during eCB-LTD induction. However, blockade of voltage-dependent Ca2+ channels (VDCCs) did not uncover direct preNMDAR-mediated Ca2+ transients in the axon. This suggests that astrocyte-mediated glutamate release onto preNMDARs does not result in a direct Ca2+ influx, but that it instead leads to an indirect interaction with presynaptic VDCCs, boosting axonal Ca2+ influx. These results reveal one of the main remaining missing pieces in the signaling cascade of t-LTD at developing cortical synapses.
Collapse
|
27
|
Sun J, Liu Y, Hao X, Lin W, Su W, Chiang E, Baudry M, Bi X. LAMTOR1 inhibition of TRPML1‐dependent lysosomal calcium release regulates dendritic lysosome trafficking and hippocampal neuronal function. EMBO J 2022; 41:e108119. [PMID: 35099830 PMCID: PMC8886530 DOI: 10.15252/embj.2021108119] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 12/12/2021] [Accepted: 12/21/2021] [Indexed: 02/05/2023] Open
Abstract
Lysosomes function not only as degradatory compartments but also as dynamic intracellular calcium ion stores. The transient receptor potential mucolipin 1 (TRPML1) channel mediates lysosomal Ca2+ release, thereby participating in multiple cellular functions. The pentameric Ragulator complex, which plays a critical role in the activation of mTORC1, is also involved in lysosomal trafficking and is anchored to lysosomes through its LAMTOR1 subunit. Here, we report that the Ragulator restricts lysosomal trafficking in dendrites of hippocampal neurons via LAMTOR1‐mediated tonic inhibition of TRPML1 activity, independently of mTORC1. LAMTOR1 directly interacts with TRPML1 through its N‐terminal domain. Eliminating this inhibition in hippocampal neurons by LAMTOR1 deletion or by disrupting LAMTOR1‐TRPML1 binding increases TRPML1‐mediated Ca2+ release and facilitates dendritic lysosomal trafficking powered by dynein. LAMTOR1 deletion in the hippocampal CA1 region of adult mice results in alterations in synaptic plasticity, and in impaired object‐recognition memory and contextual fear conditioning, due to TRPML1 activation. Mechanistically, changes in synaptic plasticity are associated with increased GluA1 dephosphorylation by calcineurin and lysosomal degradation. Thus, LAMTOR1‐mediated inhibition of TRPML1 is critical for regulating dendritic lysosomal motility, synaptic plasticity, and learning.
Collapse
Affiliation(s)
- Jiandong Sun
- College of Osteopathic Medicine of the Pacific Western University of Health Sciences Pomona CA USA
| | - Yan Liu
- Graduate College of Biomedical Sciences Western University of Health Sciences Pomona CA USA
| | - Xiaoning Hao
- College of Osteopathic Medicine of the Pacific Western University of Health Sciences Pomona CA USA
| | - Weiju Lin
- Graduate College of Biomedical Sciences Western University of Health Sciences Pomona CA USA
| | - Wenyue Su
- Graduate College of Biomedical Sciences Western University of Health Sciences Pomona CA USA
| | - Emerald Chiang
- Graduate College of Biomedical Sciences Western University of Health Sciences Pomona CA USA
| | - Michel Baudry
- Graduate College of Biomedical Sciences Western University of Health Sciences Pomona CA USA
| | - Xiaoning Bi
- College of Osteopathic Medicine of the Pacific Western University of Health Sciences Pomona CA USA
| |
Collapse
|
28
|
A recurrent SHANK1 mutation implicated in autism spectrum disorder causes autistic-like core behaviors in mice via downregulation of mGluR1-IP3R1-calcium signaling. Mol Psychiatry 2022; 27:2985-2998. [PMID: 35388181 PMCID: PMC9205781 DOI: 10.1038/s41380-022-01539-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 03/09/2022] [Accepted: 03/21/2022] [Indexed: 12/27/2022]
Abstract
The genetic etiology and underlying mechanism of autism spectrum disorder (ASD) remain elusive. SHANK family genes (SHANK1/2/3) are well known ASD-related genes. However, little is known about how SHANK missense mutations contribute to ASD. Here, we aimed to clarify the molecular mechanism of and the multilevel neuropathological features induced by Shank1 mutations in knock-in (KI) mice. In this study, by sequencing the SHANK1 gene in a cohort of 615 ASD patients and 503 controls, we identified an ASD-specific recurrent missense mutation, c.2621 G > A (p.R874H). This mutation demonstrated strong pathogenic potential in in vitro experiments, and we generated the corresponding Shank1 R882H-KI mice. Shank1 R882H-KI mice displayed core symptoms of ASD, namely, social disability and repetitive behaviors, without confounding comorbidities of abnormal motor function and heightened anxiety. Brain structural changes in the frontal cortex, hippocampus and cerebellar cortex were observed in Shank1 R882H-KI mice via structural magnetic resonance imaging. These key brain regions also showed severe and consistent downregulation of mGluR1-IP3R1-calcium signaling, which subsequently affected the release of intracellular calcium. Corresponding cellular structural and functional changes were present in Shank1 R882H-KI mice, including decreased spine size, reduced spine density, abnormal morphology of postsynaptic densities, and impaired hippocampal long-term potentiation and basal excitatory transmission. These findings demonstrate the causative role of SHANK1 in ASD and elucidate the underlying biological mechanism of core symptoms of ASD. We also provide a reliable model of ASD with core symptoms for future studies, such as biomarker identification and therapeutic intervention studies.
Collapse
|
29
|
Trask S, Ferrara NC, Jasnow AM, Kwapis JL. Contributions of the rodent cingulate-retrosplenial cortical axis to associative learning and memory: A proposed circuit for persistent memory maintenance. Neurosci Biobehav Rev 2021; 130:178-184. [PMID: 34450181 PMCID: PMC8511298 DOI: 10.1016/j.neubiorev.2021.08.023] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 08/18/2021] [Accepted: 08/22/2021] [Indexed: 10/20/2022]
Abstract
While the anterior cingulate (ACC) and retrosplenial (RSC) cortices have been extensively studied for their role in spatial navigation, less is known about how they contribute to associative learning and later memory recall. The limited work that has been conducted on this topic suggests that each of these cortical regions makes distinct, but similar contributions to associative learning and memory. Here, we review evidence from the rodent literature demonstrating that while ACC activity seems to be necessary at remote time points associated with imprecise or generalized memories, the role of the RSC seems to be uniform over time. Together, the lines of evidence reviewed here suggest that the ACC and RSC likely function together to support memory formation and maintenance following associative learning.
Collapse
Affiliation(s)
- Sydney Trask
- Department of Psychological Sciences, Purdue University, West Lafayette, IN, 47907, United States
| | - Nicole C Ferrara
- Department of Pharmacology, Rosalind Franklin University of Medicine and Science, North Chicago, IL, 60064, United States
| | - Aaron M Jasnow
- Department of Pharmacology, Physiology & Neuroscience, University of South Carolina School of Medicine, Columbia, SC, 29209, United States
| | - Janine L Kwapis
- Department of Biology, Pennsylvania State University, University Park, PA, 16802, United States.
| |
Collapse
|
30
|
Tapella L, Dematteis G, Ruffinatti FA, Ponzoni L, Fiordaliso F, Corbelli A, Albanese E, Pistolato B, Pagano J, Barberis E, Marengo E, Balducci C, Forloni G, Verpelli C, Sala C, Distasi C, Sala M, Manfredi M, Genazzani AA, Lim D. Deletion of calcineurin from astrocytes reproduces proteome signature of Alzheimer's disease and epilepsy and predisposes to seizures. Cell Calcium 2021; 100:102480. [PMID: 34607180 DOI: 10.1016/j.ceca.2021.102480] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2021] [Revised: 09/18/2021] [Accepted: 09/22/2021] [Indexed: 02/07/2023]
Abstract
Calcineurin (CaN), acting downstream of intracellular calcium signals, orchestrates cellular remodeling in many cellular types. In astrocytes, major homeostatic players in the central nervous system (CNS), CaN is involved in neuroinflammation and gliosis, while its role in healthy CNS or in early neuro-pathogenesis is poorly understood. Here we report that in mice with conditional deletion of CaN in GFAP-expressing astrocytes (astroglial calcineurin KO, ACN-KO), at 1 month of age, transcription was largely unchanged, while the proteome was deranged in the hippocampus and cerebellum. Gene ontology analysis revealed overrepresentation of annotations related to myelin sheath, mitochondria, ribosome and cytoskeleton. Over-represented pathways were related to protein synthesis, oxidative phosphorylation, mTOR and neurological disorders, including Alzheimer's disease (AD) and seizure disorder. Comparison with published proteomic datasets showed significant overlap with the proteome of a familial AD mouse model and of human subjects with drug-resistant seizures. ACN-KO mice showed no alterations of motor activity, equilibrium, anxiety or depressive state. However, in Barnes maze ACN-KO mice learned the task but adopted serial search strategy. Strikingly, beginning from about 5 months of age ACN-KO mice developed spontaneous tonic-clonic seizures with an inflammatory signature of epileptic brains. Altogether, our data suggest that the deletion of astroglial CaN produces features of neurological disorders and predisposes mice to seizures. We suggest that calcineurin in astrocytes may serve as a novel Ca2+-sensitive switch which regulates protein expression and homeostasis in the central nervous system.
Collapse
Affiliation(s)
- Laura Tapella
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale "Amedeo Avogadro", Via Bovio 6, 28100, Novara, Italy
| | - Giulia Dematteis
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale "Amedeo Avogadro", Via Bovio 6, 28100, Novara, Italy
| | - Federico Alessandro Ruffinatti
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale "Amedeo Avogadro", Via Bovio 6, 28100, Novara, Italy
| | - Luisa Ponzoni
- BIOMETRA, University of Milan and Fondazione Zardi-Gori, Milan, Italy
| | - Fabio Fiordaliso
- Department of Molecular Biochemistry and Pharmacology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Alessandro Corbelli
- Department of Molecular Biochemistry and Pharmacology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Enrico Albanese
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale "Amedeo Avogadro", Via Bovio 6, 28100, Novara, Italy
| | - Beatrice Pistolato
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale "Amedeo Avogadro", Via Bovio 6, 28100, Novara, Italy
| | | | - Elettra Barberis
- Department of Sciences and Technological Innovation, Università del Piemonte Orientale, Alessandria, Italy; Department of Translational Medicine, University of Piemonte Orientale, Novara, Italy
| | - Emilio Marengo
- Department of Sciences and Technological Innovation, Università del Piemonte Orientale, Alessandria, Italy
| | - Claudia Balducci
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Gianluigi Forloni
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | | | - Carlo Sala
- CNR Neuroscience Institute, Milan, Italy
| | - Carla Distasi
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale "Amedeo Avogadro", Via Bovio 6, 28100, Novara, Italy
| | | | - Marcello Manfredi
- Department of Translational Medicine, University of Piemonte Orientale, Novara, Italy; Center for Translational Research on Autoimmune and Allergic Diseases, University of Piemonte Orientale, Novara, Italy
| | - Armando A Genazzani
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale "Amedeo Avogadro", Via Bovio 6, 28100, Novara, Italy.
| | - Dmitry Lim
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale "Amedeo Avogadro", Via Bovio 6, 28100, Novara, Italy.
| |
Collapse
|
31
|
Wang Z, Zhao M, Gao S. Epileptic Seizures After Allogeneic Hematopoietic Stem Cell Transplantation. Front Neurol 2021; 12:675756. [PMID: 34335446 PMCID: PMC8322618 DOI: 10.3389/fneur.2021.675756] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Accepted: 06/17/2021] [Indexed: 12/02/2022] Open
Abstract
Technique in allogeneic hematopoietic stem cell transplantation has greatly advanced over the past decades, which has led to an increase in the number of patients receiving transplantation, but the complex procedure places these transplant recipients at high risk of a large spectrum of complications including neurologic involvement. As a common manifestation of neurological disorders, epileptic seizures after transplantation have been of great concern to clinicians because it seriously affects the survival rate and living quality of those recipients. The aim of this review is to elucidate the incidence of seizures after allogeneic hematopoietic stem cell transplantation, and to further summarize in detail its etiologies, possible mechanisms, clinical manifestations, therapeutic schedule, and prognosis, hoping to improve doctors' understandings of concurrent seizures following transplantation, so they can prevent, process, and eventually improve the survival and outlook for patients in a timely manner and correctly.
Collapse
Affiliation(s)
- Zhuo Wang
- Department of Hematology, The First Hospital of Jilin University, Changchun, China
| | - Munan Zhao
- Department of Oncology, The First Hospital of Jilin University, Changchun, China
| | - Sujun Gao
- Department of Hematology, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
32
|
Stewart S, Le Bleu HK, Yette GA, Henner AL, Robbins AE, Braunstein JA, Stankunas K. longfin causes cis-ectopic expression of the kcnh2a ether-a-go-go K+ channel to autonomously prolong fin outgrowth. Development 2021; 148:dev199384. [PMID: 34061172 PMCID: PMC8217709 DOI: 10.1242/dev.199384] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Accepted: 04/19/2021] [Indexed: 12/11/2022]
Abstract
Organs stop growing to achieve a characteristic size and shape in scale with the body of an animal. Likewise, regenerating organs sense injury extents to instruct appropriate replacement growth. Fish fins exemplify both phenomena through their tremendous diversity of form and remarkably robust regeneration. The classic zebrafish mutant longfint2 develops and regenerates dramatically elongated fins and underlying ray skeleton. We show longfint2 chromosome 2 overexpresses the ether-a-go-go-related voltage-gated potassium channel kcnh2a. Genetic disruption of kcnh2a in cis rescues longfint2, indicating longfint2 is a regulatory kcnh2a allele. We find longfint2 fin overgrowth originates from prolonged outgrowth periods by showing Kcnh2a chemical inhibition during late stage regeneration fully suppresses overgrowth. Cell transplantations demonstrate longfint2-ectopic kcnh2a acts tissue autonomously within the fin intra-ray mesenchymal lineage. Temporal inhibition of the Ca2+-dependent phosphatase calcineurin indicates it likewise entirely acts late in regeneration to attenuate fin outgrowth. Epistasis experiments suggest longfint2-expressed Kcnh2a inhibits calcineurin output to supersede growth cessation signals. We conclude ion signaling within the growth-determining mesenchyme lineage controls fin size by tuning outgrowth periods rather than altering positional information or cell-level growth potency.
Collapse
Affiliation(s)
- Scott Stewart
- Institute of Molecular Biology, University of Oregon, 273 Onyx Bridge, 1318 Franklin Blvd, Eugene, OR 97403-1229, USA
| | - Heather K. Le Bleu
- Institute of Molecular Biology, University of Oregon, 273 Onyx Bridge, 1318 Franklin Blvd, Eugene, OR 97403-1229, USA
- Department of Biology, University of Oregon, 77 Klamath Hall, Eugene, OR 97403-1210, USA
| | - Gabriel A. Yette
- Institute of Molecular Biology, University of Oregon, 273 Onyx Bridge, 1318 Franklin Blvd, Eugene, OR 97403-1229, USA
- Department of Biology, University of Oregon, 77 Klamath Hall, Eugene, OR 97403-1210, USA
| | - Astra L. Henner
- Institute of Molecular Biology, University of Oregon, 273 Onyx Bridge, 1318 Franklin Blvd, Eugene, OR 97403-1229, USA
| | - Amy E. Robbins
- Institute of Molecular Biology, University of Oregon, 273 Onyx Bridge, 1318 Franklin Blvd, Eugene, OR 97403-1229, USA
- Department of Biology, University of Oregon, 77 Klamath Hall, Eugene, OR 97403-1210, USA
| | - Joshua A. Braunstein
- Institute of Molecular Biology, University of Oregon, 273 Onyx Bridge, 1318 Franklin Blvd, Eugene, OR 97403-1229, USA
| | - Kryn Stankunas
- Institute of Molecular Biology, University of Oregon, 273 Onyx Bridge, 1318 Franklin Blvd, Eugene, OR 97403-1229, USA
- Department of Biology, University of Oregon, 77 Klamath Hall, Eugene, OR 97403-1210, USA
| |
Collapse
|
33
|
The Relevance of Amyloid β-Calmodulin Complexation in Neurons and Brain Degeneration in Alzheimer's Disease. Int J Mol Sci 2021; 22:ijms22094976. [PMID: 34067061 PMCID: PMC8125740 DOI: 10.3390/ijms22094976] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 05/02/2021] [Accepted: 05/05/2021] [Indexed: 12/13/2022] Open
Abstract
Intraneuronal amyloid β (Aβ) oligomer accumulation precedes the appearance of amyloid plaques or neurofibrillary tangles and is neurotoxic. In Alzheimer’s disease (AD)-affected brains, intraneuronal Aβ oligomers can derive from Aβ peptide production within the neuron and, also, from vicinal neurons or reactive glial cells. Calcium homeostasis dysregulation and neuronal excitability alterations are widely accepted to play a key role in Aβ neurotoxicity in AD. However, the identification of primary Aβ-target proteins, in which functional impairment initiating cytosolic calcium homeostasis dysregulation and the critical point of no return are still pending issues. The micromolar concentration of calmodulin (CaM) in neurons and its high affinity for neurotoxic Aβ peptides (dissociation constant ≈ 1 nM) highlight a novel function of CaM, i.e., the buffering of free Aβ concentrations in the low nanomolar range. In turn, the concentration of Aβ-CaM complexes within neurons will increase as a function of time after the induction of Aβ production, and free Aβ will rise sharply when accumulated Aβ exceeds all available CaM. Thus, Aβ-CaM complexation could also play a major role in neuronal calcium signaling mediated by calmodulin-binding proteins by Aβ; a point that has been overlooked until now. In this review, we address the implications of Aβ-CaM complexation in the formation of neurotoxic Aβ oligomers, in the alteration of intracellular calcium homeostasis induced by Aβ, and of dysregulation of the calcium-dependent neuronal activity and excitability induced by Aβ.
Collapse
|
34
|
Serine/Threonine Phosphatases in LTP: Two B or Not to Be the Protein Synthesis Blocker-Induced Impairment of Early Phase. Int J Mol Sci 2021; 22:ijms22094857. [PMID: 34064311 PMCID: PMC8125358 DOI: 10.3390/ijms22094857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 04/27/2021] [Accepted: 04/30/2021] [Indexed: 11/25/2022] Open
Abstract
Dephosphorylation of target proteins at serine/threonine residues is one of the most crucial mechanisms regulating their activity and, consequently, the cellular functions. The role of phosphatases in synaptic plasticity, especially in long-term depression or depotentiation, has been reported. We studied serine/threonine phosphatase activity during the protein synthesis blocker (PSB)-induced impairment of long-term potentiation (LTP). Established protein phosphatase 2B (PP2B, calcineurin) inhibitor cyclosporin A prevented the LTP early phase (E-LTP) decline produced by pretreatment of hippocampal slices with cycloheximide or anisomycin. For the first time, we directly measured serine/threonine phosphatase activity during E-LTP, and its significant increase in PSB-treated slices was demonstrated. Nitric oxide (NO) donor SNAP also heightened phosphatase activity in the same manner as PSB, and simultaneous application of anisomycin + SNAP had no synergistic effect. Direct measurement of the NO production in hippocampal slices by the NO-specific fluorescent probe DAF-FM revealed that PSBs strongly stimulate the NO concentration in all studied brain areas: CA1, CA3, and dentate gyrus (DG). Cyclosporin A fully abolished the PSB-induced NO production in the hippocampus, suggesting a close relationship between nNOS and PP2B activity. Surprisingly, cyclosporin A alone impaired short-term plasticity in CA1 by decreasing paired-pulse facilitation, which suggests bi-directionality of the influences of PP2B in the hippocampus. In conclusion, we proposed a minimal model of signaling events that occur during LTP induction in normal conditions and the PSB-treated slices.
Collapse
|
35
|
Lim D, Semyanov A, Genazzani A, Verkhratsky A. Calcium signaling in neuroglia. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2021; 362:1-53. [PMID: 34253292 DOI: 10.1016/bs.ircmb.2021.01.003] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Glial cells exploit calcium (Ca2+) signals to perceive the information about the activity of the nervous tissue and the tissue environment to translate this information into an array of homeostatic, signaling and defensive reactions. Astrocytes, the best studied glial cells, use several Ca2+ signaling generation pathways that include Ca2+ entry through plasma membrane, release from endoplasmic reticulum (ER) and from mitochondria. Activation of metabotropic receptors on the plasma membrane of glial cells is coupled to an enzymatic cascade in which a second messenger, InsP3 is generated thus activating intracellular Ca2+ release channels in the ER endomembrane. Astrocytes also possess store-operated Ca2+ entry and express several ligand-gated Ca2+ channels. In vivo astrocytes generate heterogeneous Ca2+ signals, which are short and frequent in distal processes, but large and relatively rare in soma. In response to neuronal activity intracellular and inter-cellular astrocytic Ca2+ waves can be produced. Astrocytic Ca2+ signals are involved in secretion, they regulate ion transport across cell membranes, and are contributing to cell morphological plasticity. Therefore, astrocytic Ca2+ signals are linked to fundamental functions of the central nervous system ranging from synaptic transmission to behavior. In oligodendrocytes, Ca2+ signals are generated by plasmalemmal Ca2+ influx, or by release from intracellular stores, or by combination of both. Microglial cells exploit Ca2+ permeable ionotropic purinergic receptors and transient receptor potential channels as well as ER Ca2+ release. In this contribution, basic morphology of glial cells, glial Ca2+ signaling toolkit, intracellular Ca2+ signals and Ca2+-regulated functions are discussed with focus on astrocytes.
Collapse
Affiliation(s)
- Dmitry Lim
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale, Novara, Italy.
| | - Alexey Semyanov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia; Faculty of Biology, Moscow State University, Moscow, Russia; Sechenov First Moscow State Medical University, Moscow, Russia
| | - Armando Genazzani
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale, Novara, Italy
| | - Alexei Verkhratsky
- Sechenov First Moscow State Medical University, Moscow, Russia; Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom; Achucarro Centre for Neuroscience, IKERBASQUE, Basque Foundation for Science, Bilbao, Spain.
| |
Collapse
|
36
|
Nikitin VP, Kozyrev SA, Solntseva SV, Nikitin PV. Protein synthesis inhibitor administration before a reminder caused recovery from amnesia induced by memory reconsolidation impairment with NMDA glutamate receptor antagonist. Brain Res Bull 2021; 171:44-55. [PMID: 33722648 DOI: 10.1016/j.brainresbull.2021.03.008] [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: 10/23/2020] [Revised: 01/19/2021] [Accepted: 03/09/2021] [Indexed: 11/15/2022]
Abstract
Memory recovery in amnestic animals is one of the most poorly studied processes. In this paper, we examine the role of protein synthesis and a reminder in the mechanisms of amnesia and memory recovery in grape snails trained to conditioned food aversion. Amnesia was induced by the impairment of memory reconsolidation using NMDA (N-methyl d-aspartate) glutamate receptor antagonists. In an early stage of amnesia (day 3), injections of protein synthesis inhibitors into animals combined with a reminder by a conditioned stimulus (CS) led to the recovery of aversive reactions to its presentation. Two types of changes in reactions to CS were revealed. In most animals, a persistent recovery of memory retrieval was found that lasted for at least 10 days. In other snails, aversive responses to CS persisted for 24 h. Isolated injections of inhibitors, injections of inhibitors and a reminder by the learning environment (without presenting a CS), usage of a differentiating stimulus instead of a CS, or inhibitor injections after the reminder did not affect the development of amnesia. The administration of protein synthesis inhibitors and a reminder in the late period after amnesia induction (10 days) did not affect its development or caused a short-term memory recovery. We suggest that amnesia is an active process that develops over time. The reminder induces the reactivation of the amnesia process dependent on protein synthesis, while the administration of protein synthesis inhibitors leads to the impairment of amnesia reactivation and recovery of the state formed before amnesia induction (i.e., recovery of conditioned food aversion memory).
Collapse
Affiliation(s)
- V P Nikitin
- P.K. Anokhin Institute of Normal Physiology, 125315, Baltiyskaya Street, 8, Moscow, Russia.
| | - S A Kozyrev
- P.K. Anokhin Institute of Normal Physiology, 125315, Baltiyskaya Street, 8, Moscow, Russia.
| | - S V Solntseva
- P.K. Anokhin Institute of Normal Physiology, 125315, Baltiyskaya Street, 8, Moscow, Russia.
| | - P V Nikitin
- P.K. Anokhin Institute of Normal Physiology, 125315, Baltiyskaya Street, 8, Moscow, Russia.
| |
Collapse
|
37
|
Role of calcineurin biosignaling in cell secretion and the possible regulatory mechanisms. Saudi J Biol Sci 2021; 28:116-124. [PMID: 33424288 PMCID: PMC7783665 DOI: 10.1016/j.sjbs.2020.08.042] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Revised: 08/02/2020] [Accepted: 08/30/2020] [Indexed: 11/22/2022] Open
Abstract
Cyclic adenosine monophosphate (cAMP) and calcium ions (Ca2+) are two chemical molecules that play a central role in the stimulus-dependent secretion processes within cells. Ca2+ acts as the basal signaling molecule responsible to initiate cell secretion. cAMP primarily acts as an intracellular second messenger in a myriad of cellular processes by activating cAMP-dependent protein kinases through association with such kinases in order to mediate post-translational phosphorylation of those protein targets. Put succinctly, both Ca2+ and cAMP act by associating or activating other proteins to ensure successful secretion. Calcineurin is one such protein regulated by Ca2+; its action depends on the intracellular levels of Ca2+. Being a phosphatase, calcineurin dephosphorylate and other proteins, as is the case with most other phosphatases, such as protein phosphatase 2A (PP2A), PP2C, and protein phosphatase-1 (PP1), will likely be activated by phosphorylation. Via this process, calcineurin is able to affect different intracellular signaling with clinical importance, some of which has been the basis for development of different calcineurin inhibitors. In this review, the cAMP-dependent calcineurin bio-signaling, protein-protein interactions and their physiological implications as well as regulatory signaling within the context of cellular secretion are explored.
Collapse
|
38
|
Mahmoodkhani M, Ghasemi M, Derafshpour L, Amini M, Mehranfard N. Long-Term Decreases in the Expression of Calcineurin and GABAA Receptors Induced by Early Maternal Separation Are Associated with Increased Anxiety-Like Behavior in Adult Male Rats. Dev Neurosci 2020; 42:135-144. [PMID: 33341802 DOI: 10.1159/000512221] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Accepted: 10/09/2020] [Indexed: 12/17/2022] Open
Abstract
INTRODUCTION Early life stress is a well-described risk factor of anxiety disorders in adulthood. Dysfunction in GABA/glutamate receptors and their functional regulator, calcineurin, is linked to anxiety disorders. Here, we investigated the effect of early life stress, such as repeated maternal separation (MS; 3 h per day from postnatal day [P] 2 to 11), on changes in the expression of calcineurin as well as the ionotropic glutamatergic and GABAergic receptors including α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA), N-methyl-D-aspartate (NMDA) and GABAA receptors in the hippocampus and prefrontal cortex (PFC) of adolescent (P35) and adult (P62) male Wistar rats and their correlations with anxiety-like behavior in adulthood. METHODS The protein levels were assessed by Western blot analysis. Anxiety-like behavior was measured in the elevated plus maze (EPM) and open field (OF) tests. RESULTS MS induced a regional transient decrease of glutamate receptors expression at P35, with decreased NMDA and AMPA receptor levels, respectively, in the hippocampus and PFC, suggesting a possible decrease in excitatory synaptic strength. In contrast to glutamate receptors, MS had long-lasting influence on GABAA receptor and calcineurin levels, with reduced expression of GABAA receptor and calcineurin in both brain regions at P35 that continued into adulthood. These results were accompanied by increased anxiety behavior in adulthood, shown by lower percentage of number of total entries and time spent in the open arms of the EPM, and by lower time spent and number of entries in the OF central area. CONCLUSIONS Together, our study suggests that GABAA receptors via calcineurin-dependent signaling pathways may play an important role in the expression of stress-induced anxiety-like behavior.
Collapse
Affiliation(s)
- Maryam Mahmoodkhani
- Neurophysiology Research Center, Cellular and Molecular Medicine Institute, Urmia University of Medical Sciences, Urmia, Iran
| | - Maedeh Ghasemi
- Department of Physiology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Leila Derafshpour
- Neurophysiology Research Center, Cellular and Molecular Medicine Institute, Urmia University of Medical Sciences, Urmia, Iran
| | - Mohammad Amini
- Neurophysiology Research Center, Cellular and Molecular Medicine Institute, Urmia University of Medical Sciences, Urmia, Iran
| | - Nasrin Mehranfard
- Neurophysiology Research Center, Cellular and Molecular Medicine Institute, Urmia University of Medical Sciences, Urmia, Iran,
| |
Collapse
|
39
|
Vasilopoulou F, Griñán-Ferré C, Rodríguez-Arévalo S, Bagán A, Abás S, Escolano C, Pallàs M. I 2 imidazoline receptor modulation protects aged SAMP8 mice against cognitive decline by suppressing the calcineurin pathway. GeroScience 2020; 43:965-983. [PMID: 33128688 PMCID: PMC8110656 DOI: 10.1007/s11357-020-00281-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Accepted: 09/28/2020] [Indexed: 12/26/2022] Open
Abstract
Brain aging and dementia are current problems that must be solved. The levels of imidazoline 2 receptors (I2-IRs) are increased in the brain in Alzheimer's disease (AD) and other neurodegenerative diseases. We tested the action of the specific and selective I2-IR ligand B06 in a mouse model of accelerated aging and AD, the senescence-accelerated mouse prone 8 (SAMP8) model. Oral administration of B06 for 4 weeks improved SAMP8 mouse behavior and cognition and reduced AD hallmarks, oxidative stress, and apoptotic and neuroinflammation markers. Likewise, B06 regulated glial excitatory amino acid transporter 2 and N-methyl-D aspartate 2A and 2B receptor subunit protein levels. Calcineurin (CaN) is a phosphatase that controls the phosphorylation levels of cAMP response element-binding (CREB), apoptotic mediator BCL-2-associated agonist of cell death (BAD) and GSK3β, among other molecules. Interestingly, B06 was able to reduce the levels of the CaN active form (CaN A). Likewise, CREB phosphorylation, BAD gene expression, and other factors were modified after B06 treatment. Moreover, phosphorylation of a target of CaN, nuclear factor of activated T-cells, cytoplasmic 1 (NFATC1), was increased in B06-treated mice, impeding the transcription of genes related to neuroinflammation and neural plasticity. In summary, this I2 imidazoline ligand can exert its beneficial effects on age-related conditions by modulating CaN pathway action and affecting several molecular pathways, playing a neuroprotective role in SAMP8 mice.
Collapse
Affiliation(s)
- Foteini Vasilopoulou
- Pharmacology Section, Department of Pharmacology, Toxicology and Medicinal Chemistry, Faculty of Pharmacy and Food Sciences, and Institute of Neurociencies, University of Barcelona, Av. Joan XXIII, 27-31, E-08028, Barcelona, Spain
| | - Christian Griñán-Ferré
- Pharmacology Section, Department of Pharmacology, Toxicology and Medicinal Chemistry, Faculty of Pharmacy and Food Sciences, and Institute of Neurociencies, University of Barcelona, Av. Joan XXIII, 27-31, E-08028, Barcelona, Spain
| | - Sergio Rodríguez-Arévalo
- Laboratory of Medicinal Chemistry (Associated Unit to CSIC), Department of Pharmacology, Toxicology and Medicinal Chemistry, Faculty of Pharmacy and Food Sciences, and Institute of Biomedicine (IBUB), University of Barcelona, Av. Joan XXIII, 27-31, E-08028, Barcelona, Spain
| | - Andrea Bagán
- Laboratory of Medicinal Chemistry (Associated Unit to CSIC), Department of Pharmacology, Toxicology and Medicinal Chemistry, Faculty of Pharmacy and Food Sciences, and Institute of Biomedicine (IBUB), University of Barcelona, Av. Joan XXIII, 27-31, E-08028, Barcelona, Spain
| | - Sònia Abás
- Laboratory of Medicinal Chemistry (Associated Unit to CSIC), Department of Pharmacology, Toxicology and Medicinal Chemistry, Faculty of Pharmacy and Food Sciences, and Institute of Biomedicine (IBUB), University of Barcelona, Av. Joan XXIII, 27-31, E-08028, Barcelona, Spain
| | - Carmen Escolano
- Laboratory of Medicinal Chemistry (Associated Unit to CSIC), Department of Pharmacology, Toxicology and Medicinal Chemistry, Faculty of Pharmacy and Food Sciences, and Institute of Biomedicine (IBUB), University of Barcelona, Av. Joan XXIII, 27-31, E-08028, Barcelona, Spain
| | - Mercè Pallàs
- Pharmacology Section, Department of Pharmacology, Toxicology and Medicinal Chemistry, Faculty of Pharmacy and Food Sciences, and Institute of Neurociencies, University of Barcelona, Av. Joan XXIII, 27-31, E-08028, Barcelona, Spain.
| |
Collapse
|
40
|
Amal H, Barak B, Bhat V, Gong G, Joughin BA, Wang X, Wishnok JS, Feng G, Tannenbaum SR. Shank3 mutation in a mouse model of autism leads to changes in the S-nitroso-proteome and affects key proteins involved in vesicle release and synaptic function. Mol Psychiatry 2020; 25:1835-1848. [PMID: 29988084 PMCID: PMC6614015 DOI: 10.1038/s41380-018-0113-6] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Revised: 05/14/2018] [Accepted: 06/05/2018] [Indexed: 12/25/2022]
Abstract
Mutation in the SHANK3 human gene leads to different neuropsychiatric diseases including Autism Spectrum Disorder (ASD), intellectual disabilities and Phelan-McDermid syndrome. Shank3 disruption in mice leads to dysfunction of synaptic transmission, behavior, and development. Protein S-nitrosylation, the nitric oxide (NO•)-mediated posttranslational modification (PTM) of cysteine thiols (SNO), modulates the activity of proteins that regulate key signaling pathways. We tested the hypothesis that Shank3 mutation would generate downstream effects on PTM of critical proteins that lead to modification of synaptic functions. SNO-proteins in two ASD-related brain regions, cortex and striatum of young and adult InsG3680(+/+) mice (a human mutation-based Shank3 mouse model), were identified by an innovative mass spectrometric method, SNOTRAP. We found changes of the SNO-proteome in the mutant compared to WT in both ages. Pathway analysis showed enrichment of processes affected in ASD. SNO-Calcineurin in mutant led to a significant increase of phosphorylated Synapsin1 and CREB, which affect synaptic vesicle mobilization and gene transcription, respectively. A significant increase of 3-nitrotyrosine was found in the cortical regions of the adult mutant, signaling both oxidative and nitrosative stress. Neuronal NO• Synthase (nNOS) was examined for levels and localization in neurons and no significant difference was found in WT vs. mutant. S-nitrosoglutathione concentrations were higher in mutant mice compared to WT. This is the first study on NO•-related molecular changes and SNO-signaling in the brain of an ASD mouse model that allows the characterization and identification of key proteins, cellular pathways, and neurobiological mechanisms that might be affected in ASD.
Collapse
Affiliation(s)
- Haitham Amal
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA.
| | - Boaz Barak
- McGovern Institute for Brain Research, Massachusetts
Institute of Technology, Cambridge, MA 02139, USA
| | | | - Guanyu Gong
- Department of Biological Engineering, Massachusetts
Institute of Technology, Cambridge, MA 02139, USA
| | - Brian A. Joughin
- Department of Biological Engineering, Massachusetts
Institute of Technology, Cambridge, MA 02139, USA,Koch Institute for Integrative Cancer Research,
Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Xin Wang
- Department of Biological Engineering, Massachusetts
Institute of Technology, Cambridge, MA 02139, USA
| | - John S. Wishnok
- Department of Biological Engineering, Massachusetts
Institute of Technology, Cambridge, MA 02139, USA
| | - Guoping Feng
- McGovern Institute for Brain Research, Massachusetts
Institute of Technology, Cambridge, MA 02139, USA
| | - Steven R. Tannenbaum
- Department of Biological Engineering, Massachusetts
Institute of Technology, Cambridge, MA 02139, USA,Department of Chemistry, Massachusetts Institute of
Technology, Cambridge, MA 02139, USA
| |
Collapse
|
41
|
Wigington CP, Roy J, Damle NP, Yadav VK, Blikstad C, Resch E, Wong CJ, Mackay DR, Wang JT, Krystkowiak I, Bradburn DA, Tsekitsidou E, Hong SH, Kaderali MA, Xu SL, Stearns T, Gingras AC, Ullman KS, Ivarsson Y, Davey NE, Cyert MS. Systematic Discovery of Short Linear Motifs Decodes Calcineurin Phosphatase Signaling. Mol Cell 2020; 79:342-358.e12. [PMID: 32645368 DOI: 10.1016/j.molcel.2020.06.029] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 03/24/2020] [Accepted: 05/26/2020] [Indexed: 12/17/2022]
Abstract
Short linear motifs (SLiMs) drive dynamic protein-protein interactions essential for signaling, but sequence degeneracy and low binding affinities make them difficult to identify. We harnessed unbiased systematic approaches for SLiM discovery to elucidate the regulatory network of calcineurin (CN)/PP2B, the Ca2+-activated phosphatase that recognizes LxVP and PxIxIT motifs. In vitro proteome-wide detection of CN-binding peptides, in vivo SLiM-dependent proximity labeling, and in silico modeling of motif determinants uncovered unanticipated CN interactors, including NOTCH1, which we establish as a CN substrate. Unexpectedly, CN shows SLiM-dependent proximity to centrosomal and nuclear pore complex (NPC) proteins-structures where Ca2+ signaling is largely uncharacterized. CN dephosphorylates human and yeast NPC proteins and promotes accumulation of a nuclear transport reporter, suggesting conserved NPC regulation by CN. The CN network assembled here provides a resource to investigate Ca2+ and CN signaling and demonstrates synergy between experimental and computational methods, establishing a blueprint for examining SLiM-based networks.
Collapse
Affiliation(s)
| | - Jagoree Roy
- Department of Biology, Stanford University, Stanford, CA, USA
| | - Nikhil P Damle
- Department of Biology, Stanford University, Stanford, CA, USA
| | - Vikash K Yadav
- Department of Chemistry - BMC, Uppsala University, Uppsala, Sweden
| | - Cecilia Blikstad
- Department of Chemistry - BMC, Uppsala University, Uppsala, Sweden
| | - Eduard Resch
- Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Branch for Translational Medicine and Pharmacology TMP, Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany
| | - Cassandra J Wong
- Lunenfeld-Tanenbaum Research Institute at Mount Sinai Hospital, University of Toronto, Toronto, ON, Canada
| | - Douglas R Mackay
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Jennifer T Wang
- Department of Biology, Stanford University, Stanford, CA, USA
| | - Izabella Krystkowiak
- Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin 4, Ireland
| | | | | | - Su Hyun Hong
- Department of Plant Biology, Carnegie Institution for Science, Stanford, CA, USA
| | - Malika Amyn Kaderali
- Department of Plant Biology, Carnegie Institution for Science, Stanford, CA, USA
| | - Shou-Ling Xu
- Department of Plant Biology, Carnegie Institution for Science, Stanford, CA, USA
| | - Tim Stearns
- Department of Biology, Stanford University, Stanford, CA, USA
| | - Anne-Claude Gingras
- Lunenfeld-Tanenbaum Research Institute at Mount Sinai Hospital, University of Toronto, Toronto, ON, Canada; Department of Molecular Genetics, University of Toronto, Toronto, M5S 3H7 ON, Canada
| | - Katharine S Ullman
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Ylva Ivarsson
- Department of Chemistry - BMC, Uppsala University, Uppsala, Sweden
| | - Norman E Davey
- Division of Cancer Biology, The Institute of Cancer Research, 237 Fullham Road, London SW3 6JB, UK
| | - Martha S Cyert
- Department of Biology, Stanford University, Stanford, CA, USA.
| |
Collapse
|
42
|
Tapella L, Soda T, Mapelli L, Bortolotto V, Bondi H, Ruffinatti FA, Dematteis G, Stevano A, Dionisi M, Ummarino S, Di Ruscio A, Distasi C, Grilli M, Genazzani AA, D'Angelo E, Moccia F, Lim D. Deletion of calcineurin from GFAP-expressing astrocytes impairs excitability of cerebellar and hippocampal neurons through astroglial Na + /K + ATPase. Glia 2020; 68:543-560. [PMID: 31626368 DOI: 10.1002/glia.23737] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Revised: 10/02/2019] [Accepted: 10/04/2019] [Indexed: 01/29/2023]
Abstract
Astrocytes perform important housekeeping functions in the nervous system including maintenance of adequate neuronal excitability, although the regulatory mechanisms are currently poorly understood. The astrocytic Ca2+ /calmodulin-activated phosphatase calcineurin (CaN) is implicated in the development of reactive gliosis and neuroinflammation, but its roles, including the control of neuronal excitability, in healthy brain is unknown. We have generated a mouse line with conditional knockout (KO) of CaN B1 (CaNB1) in glial fibrillary acidic protein-expressing astrocytes (astroglial calcineurin KO [ACN-KO]). Here, we report that postnatal and astrocyte-specific ablation of CaNB1 did not alter normal growth and development as well as adult neurogenesis. Yet, we found that specific deletion of astrocytic CaN selectively impairs intrinsic neuronal excitability in hippocampal CA1 pyramidal neurons and cerebellar granule cells (CGCs). This impairment was associated with a decrease in after hyperpolarization in CGC, while passive properties were unchanged, suggesting impairment of K+ homeostasis. Indeed, blockade of Na+ /K+ -ATPase (NKA) with ouabain phenocopied the electrophysiological alterations observed in ACN-KO CGCs. In addition, NKA activity was significantly lower in cerebellar and hippocampal lysates and in pure astrocytic cultures from ACN-KO mice. While no changes were found in protein levels, NKA activity was inhibited by the specific CaN inhibitor FK506 in both cerebellar lysates and primary astroglia from control mice, suggesting that CaN directly modulates NKA activity and in this manner controls neuronal excitability. In summary, our data provide formal evidence for the notion that astroglia is fundamental for controlling basic neuronal functions and place CaN center-stage as an astrocytic Ca2+ -sensitive switch.
Collapse
Affiliation(s)
- Laura Tapella
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale "Amedeo Avogadro", Novara, Italy
| | - Teresa Soda
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
| | - Lisa Mapelli
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
| | - Valeria Bortolotto
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale "Amedeo Avogadro", Novara, Italy
| | - Heather Bondi
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale "Amedeo Avogadro", Novara, Italy
| | - Federico A Ruffinatti
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale "Amedeo Avogadro", Novara, Italy
| | - Giulia Dematteis
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale "Amedeo Avogadro", Novara, Italy
| | - Alessio Stevano
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale "Amedeo Avogadro", Novara, Italy
| | - Marianna Dionisi
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale "Amedeo Avogadro", Novara, Italy
| | - Simone Ummarino
- Center of Life Science, Medical School Initiative for RNA Medicine, Harvard Medical School, Boston, Massachusetts
- Department of Translational Medicine, Università del Piemonte Orientale, Novara, Italy
| | - Annalisa Di Ruscio
- Center of Life Science, Medical School Initiative for RNA Medicine, Harvard Medical School, Boston, Massachusetts
- Department of Translational Medicine, Università del Piemonte Orientale, Novara, Italy
| | - Carla Distasi
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale "Amedeo Avogadro", Novara, Italy
| | - Mariagrazia Grilli
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale "Amedeo Avogadro", Novara, Italy
| | - Armando A Genazzani
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale "Amedeo Avogadro", Novara, Italy
| | - Egidio D'Angelo
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
- IRCCS Mondino Foundation, Pavia, Italy
| | - Francesco Moccia
- Department of Biology and Biotechnology "Lazzaro Spallanzani", University of Pavia, Pavia, Italy
| | - Dmitry Lim
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale "Amedeo Avogadro", Novara, Italy
| |
Collapse
|
43
|
Rich MT, Huang YH, Torregrossa MM. Calcineurin Promotes Neuroplastic Changes in the Amygdala Associated with Weakened Cocaine-Cue Memories. J Neurosci 2020; 40:1344-1354. [PMID: 31862855 PMCID: PMC7002139 DOI: 10.1523/jneurosci.0453-19.2019] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Revised: 11/23/2019] [Accepted: 12/10/2019] [Indexed: 01/03/2023] Open
Abstract
Interfering with memory reconsolidation or inducing memory extinction are two approaches for weakening maladaptive memories in disorders such as addiction and post-traumatic stress disorder. Both extinction and reconsolidation are regulated by intracellular protein kinases and phosphatases, and interfering with these signaling molecules can alter memory strength. The calcium-dependent protein phosphatase, calcineurin (CaN), has been implicated in both the consolidation and extinction of fear memories. However, the role of CaN in regulating drug-cue associative memories has not been investigated. Prior studies have demonstrated that plasticity at thalamo-lateral amygdala (T-LA) synapses is critically involved in the regulation of cocaine-cue memories. Therefore, in the present study, we tested the effects of LA administration of an activator of CaN, chlorogenic acid (CGA), on behavioral and electrophysiological indices of cocaine cue memory reconsolidation and extinction. Male, Sprague-Dawley rats were trained to self-administer cocaine paired with an audiovisual cue. The cue memory was then either briefly reactivated, extinguished, or not manipulated, followed immediately by LA infusion of CGA. Rats were tested 24 h later for cue-induced reinstatement, or LA slices were prepared for electrophysiological recordings. We found that intra-LA infusions of CGA following cue extinction or reconsolidation reduced cue-induced reinstatement, which was blocked by co-infusion of the CaN inhibitor, FK-506. Similarly, CGA infusions following cue re-exposure significantly attenuated EPSC amplitude at T-LA synapses, suggesting that CaN affects cocaine-cue memory reconsolidation and extinction by altering T-LA synaptic strength. Therefore, CaN signaling in the LA may represent a novel target for disrupting cocaine-associated memories to reduce relapse.SIGNIFICANCE STATEMENT Repetitive drug use induces synaptic plasticity that underlies the formation of long-lasting associative memories for environmental cues paired with the drug. We previously identified thalamo-amygdala synapses (T-LA) that project via the interal capsule, as an important locus for the regulation of cocaine-cue memories. These synapses are strengthened by repeated cocaine-cue pairings, but this is reversed by extinction training or by optogenetic induction of in vivo long-term depression (LTD). Here, we demonstrate that activating calcineurin, a calcium-dependent phosphatase, following the reactivation or extinction of a cocaine-cue memory, induces LTD-like changes at T-LA synapses, and a corresponding decrease in cue-induced reinstatement, suggesting that calcineurin may be a potential therapeutic target for relapse prevention.
Collapse
Affiliation(s)
- Matthew T Rich
- Department of Psychiatry
- Center for Neuroscience, and
- Center for the Neural Basis of Cognition, University of Pittsburgh, Pittsburgh, Pennsylvania 15219
| | | | | |
Collapse
|
44
|
Bye-A-Jee H, Zaru R, Magrane M, Orchard S. Caenorhabditis elegans phosphatase complexes in UniProtKB and Complex Portal. FEBS J 2020; 287:2664-2684. [PMID: 31944606 DOI: 10.1111/febs.15213] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Revised: 11/25/2019] [Accepted: 01/13/2020] [Indexed: 02/06/2023]
Abstract
Phosphatases play an essential role in the regulation of protein phosphorylation. Less abundant than kinases, many phosphatases are components of one or more macromolecular complexes with different substrate specificities and specific functionalities. The expert scientific curation of phosphatase complexes for the UniProt and Complex Portal databases supports the whole scientific community by collating and organising small- and large-scale experimental data from the scientific literature into context-specific central resources, where the data can be freely accessed and used to further academic and translational research. In this review, we discuss how the diverse biological functions of phosphatase complexes are presented in UniProt and the Complex Portal, and how understanding the biological significance of phosphatase complexes in Caenorhabditis elegans offers insight into the mechanisms of substrate diversity in a variety of cellular and molecular processes.
Collapse
Affiliation(s)
- Hema Bye-A-Jee
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Cambridge, UK
| | - Rossana Zaru
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Cambridge, UK
| | - Michele Magrane
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Cambridge, UK
| | - Sandra Orchard
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Cambridge, UK
| | -
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Cambridge, UK.,SIB Swiss Institute of Bioinformatics, Centre Medical Universitaire, Geneva 4, Switzerland.,Protein Information Resource, Georgetown University Medical Center, Washington, DC, USA.,Protein Information Resource, University of Delaware, Newark, DE, USA
| |
Collapse
|
45
|
Li L, Lai M, Cole S, Le Novère N, Edelstein SJ. Neurogranin stimulates Ca2+/calmodulin-dependent kinase II by suppressing calcineurin activity at specific calcium spike frequencies. PLoS Comput Biol 2020; 16:e1006991. [PMID: 32049957 PMCID: PMC7041932 DOI: 10.1371/journal.pcbi.1006991] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 02/25/2020] [Accepted: 11/18/2019] [Indexed: 11/18/2022] Open
Abstract
Calmodulin sits at the center of molecular mechanisms underlying learning and memory. Its complex and sometimes opposite influences, mediated via the binding to various proteins, are yet to be fully understood. Calcium/calmodulin-dependent protein kinase II (CaMKII) and calcineurin (CaN) both bind open calmodulin, favoring Long-Term Potentiation (LTP) or Depression (LTD) respectively. Neurogranin binds to the closed conformation of calmodulin and its impact on synaptic plasticity is less clear. We set up a mechanistic computational model based on allosteric principles to simulate calmodulin state transitions and its interactions with calcium ions and the three binding partners mentioned above. We simulated calcium spikes at various frequencies and show that neurogranin regulates synaptic plasticity along three modalities. At low spike frequencies, neurogranin inhibits the onset of LTD by limiting CaN activation. At intermediate frequencies, neurogranin facilitates LTD, but limits LTP by precluding binding of CaMKII with calmodulin. Finally, at high spike frequencies, neurogranin promotes LTP by enhancing CaMKII autophosphorylation. While neurogranin might act as a calmodulin buffer, it does not significantly preclude the calmodulin opening by calcium. On the contrary, neurogranin synchronizes the opening of calmodulin's two lobes and promotes their activation at specific frequencies. Neurogranin suppresses basal CaN activity, thus increasing the chance of CaMKII trans-autophosphorylation at high-frequency calcium spikes. Taken together, our study reveals dynamic regulatory roles played by neurogranin on synaptic plasticity, which provide mechanistic explanations for opposing experimental findings.
Collapse
Affiliation(s)
- Lu Li
- Babraham Institute, Cambridge, United Kingdom
| | - Massimo Lai
- Quantitative Systems Pharmacology, CERTARA, Canterbury, United Kingdom
| | - Stephen Cole
- Cambridge Systems Biology Centre, University of Cambridge, Cambridge, United Kingdom
| | | | | |
Collapse
|
46
|
Speigel IA, Ma CM, Bichler EK, Gooch JL, García PS. Chronic Calcineurin Inhibition via Cyclosporine A Impairs Visuospatial Learning After Isoflurane Anesthesia. Anesth Analg 2020; 129:192-203. [PMID: 31082969 DOI: 10.1213/ane.0000000000004183] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
BACKGROUND Clinical studies implicate the perioperative period in cognitive complications, and increasing experimental evidence shows that the anesthetic agents can affect neuronal processes that underpin learning and memory. Calcineurin, a Ca-dependent phosphatase critically involved in synaptic plasticity, is activated after isoflurane exposure, but its role in the neurological response to anesthesia is unclear. METHODS We investigated the effect of chronic calcineurin inhibition on postanesthetic cognitive function. Mice were treated with 30 minutes of isoflurane anesthesia during a chronic cyclosporine A regimen. Behavioral end points during the perianesthesia period were quantified. Visuospatial learning was assessed with the water radial arm maze. Total and biotinylated surface protein expression of the α5β3γ2 γ-aminobutyric acid (GABA) type A receptors was measured. Expression of the GABA synthesis enzyme glutamate decarboxylase (GAD)-67 was also measured. RESULTS Mice treated with cyclosporine A before anesthesia showed significant deficits in visuospatial learning compared to sham and cyclosporine A-treated mice (n = 10 per group, P = .0152, Tukey post hoc test). Induction and emergence were unaltered by cyclosporine A. Analysis of hippocampal protein expression revealed an increased surface expression of the α5 GABA type A receptor subunit after isoflurane treatment (P = .019, Dunnett post hoc testing), as well as a decrease in GAD-67 expression. Cyclosporine A did not rescue either effect. CONCLUSIONS Our results confirm the work of others that isoflurane induces changes to inhibitory network function and exclude calcineurin inhibition via cyclosporine A as an intervention. Further, our studies suggest that calcineurin mediates a protective role in the neurological response to anesthesia, and patients receiving cyclosporine A may be an at-risk group for memory problems related to anesthesia.
Collapse
Affiliation(s)
- Iris A Speigel
- From the Neuroanesthesia Laboratory, Atlanta Veterans Affairs Medical Center/Emory University, Atlanta, Georgia.,Department of Anesthesiology, Emory University, Atlanta, Georgia
| | - Christopher M Ma
- Department of Anesthesiology, Emory University, Atlanta, Georgia.,Department of Nephrology, Emory University School of Medicine, Atlanta, Georgia
| | - Edyta K Bichler
- From the Neuroanesthesia Laboratory, Atlanta Veterans Affairs Medical Center/Emory University, Atlanta, Georgia.,Department of Anesthesiology, Emory University, Atlanta, Georgia
| | - Jennifer L Gooch
- Department of Nephrology, Emory University School of Medicine, Atlanta, Georgia
| | - Paul S García
- From the Neuroanesthesia Laboratory, Atlanta Veterans Affairs Medical Center/Emory University, Atlanta, Georgia.,Department of Anesthesiology, Emory University, Atlanta, Georgia
| |
Collapse
|
47
|
Fujii S, Yamazaki Y, Goto JI, Fujiwara H, Mikoshiba K. Depotentiation depends on IP 3 receptor activation sustained by synaptic inputs after LTP induction. ACTA ACUST UNITED AC 2020; 27:52-66. [PMID: 31949037 PMCID: PMC6970427 DOI: 10.1101/lm.050344.119] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Accepted: 10/29/2020] [Indexed: 12/14/2022]
Abstract
In CA1 neurons of guinea pig hippocampal slices, long-term potentiation (LTP) was induced in field excitatory postsynaptic potentials (EPSPs) or population spikes (PSs) by the delivery of high-frequency stimulation (HFS, 100 pulses at 100 Hz) to CA1 synapses, and was reversed by the delivery of a train of low-frequency stimulation (LFS, 1000 pulses at 2 Hz) at 30 min after HFS (depotentiation), and this effect was inhibited when test synaptic stimulation was halted for a 19-min period after HFS or for a 20-min period after LFS or applied over the same time period in the presence of an antagonist of N-methyl-D-aspartate receptors (NMDARs), group I metabotropic glutamate receptors (mGluRs), or inositol 1, 4, 5-trisphosphate receptors (IP3Rs). Depotentiation was also blocked by the application of a Ca2+/calmodulin-dependent protein kinase II (CaMKII) inhibitor or a calcineurin inhibitor applied in the presence of test synaptic input for a 10-min period after HFS or for a 20-min period after LFS. These results suggest that, in postsynaptic neurons, the coactivation of NMDARs and group I mGluRs due to sustained synaptic activity following LTP induction results in the activation of IP3Rs and CaMKII, which leads to the activation of calcineurin after LFS and depotentiation of CA1 synaptic responses.
Collapse
Affiliation(s)
- Satoshi Fujii
- Department of Physiology, Yamagata University School of Medicine, Yamagata 990-9585, Japan.,Laboratory for Developmental Neurobiology, Riken Brain Science Institute, Wako, Saitama 351-0198, Japan
| | - Yoshihiko Yamazaki
- Department of Physiology, Yamagata University School of Medicine, Yamagata 990-9585, Japan
| | - Jun-Ichi Goto
- Department of Physiology, Yamagata University School of Medicine, Yamagata 990-9585, Japan.,Laboratory for Developmental Neurobiology, Riken Brain Science Institute, Wako, Saitama 351-0198, Japan
| | - Hiroki Fujiwara
- Department of Physiology, Yamagata University School of Medicine, Yamagata 990-9585, Japan
| | - Katsuhiko Mikoshiba
- Laboratory for Developmental Neurobiology, Riken Brain Science Institute, Wako, Saitama 351-0198, Japan
| |
Collapse
|
48
|
Nikitin V, Solntseva S, Kozyrev S, Nikitin P. Long-term memory consolidation or reconsolidation impairment induces amnesia with key characteristics that are similar to key learning characteristics. Neurosci Biobehav Rev 2020; 108:542-558. [DOI: 10.1016/j.neubiorev.2019.12.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Revised: 09/16/2019] [Accepted: 12/03/2019] [Indexed: 02/07/2023]
|
49
|
Simplified calcium signaling cascade for synaptic plasticity. Neural Netw 2019; 123:38-51. [PMID: 31821949 DOI: 10.1016/j.neunet.2019.11.022] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2019] [Revised: 11/21/2019] [Accepted: 11/25/2019] [Indexed: 11/21/2022]
Abstract
We propose a model for synaptic plasticity based on a calcium signaling cascade. The model simplifies the full signaling pathways from a calcium influx to the phosphorylation (potentiation) and dephosphorylation (depression) of glutamate receptors that are gated by fictive C1 and C2 catalysts, respectively. This model is based on tangible chemical reactions, including fictive catalysts, for long-term plasticity rather than the conceptual theories commonplace in various models, such as preset thresholds of calcium concentration. Our simplified model successfully reproduced the experimental synaptic plasticity induced by different protocols such as (i) a synchronous pairing protocol and (ii) correlated presynaptic and postsynaptic action potentials (APs). Further, the ocular dominance plasticity (or the experimental verification of the celebrated Bienenstock-Cooper-Munro theory) was reproduced by two model synapses that compete by means of back-propagating APs (bAPs). The key to this competition is synapse-specific bAPs with reference to bAP-boosting on the physiological grounds.
Collapse
|
50
|
Merino P, Diaz A, Torre ER, Yepes M. Urokinase-type plasminogen activator (uPA) regulates the expression and function of growth-associated protein 43 (GAP-43) in the synapse. J Biol Chem 2019; 295:619-630. [PMID: 31819012 DOI: 10.1074/jbc.ra119.010644] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Revised: 11/19/2019] [Indexed: 11/06/2022] Open
Abstract
Growth-associated protein 43 (GAP-43) plays a central role in the formation of presynaptic terminals, synaptic plasticity, and axonal growth and regeneration. During development, GAP-43 is found in axonal extensions of most neurons. In contrast, in the mature brain, its expression is restricted to a few presynaptic terminals and scattered axonal growth cones. Urokinase-type plasminogen activator (uPA) is a serine proteinase that, upon binding to its receptor (uPAR), catalyzes the conversion of plasminogen into plasmin and activates signaling pathways that promote cell migration, proliferation, and survival. In the developing brain, uPA induces neuritogenesis and neuronal migration. In contrast, the expression and function of uPA in the mature brain are poorly understood. However, recent evidence reveals that different forms of injury induce release of uPA and expression of uPAR in neurons and that uPA/uPAR binding triggers axonal growth and synapse formation. Here we show that binding of uPA to uPAR induces not only the mobilization of GAP-43 from the axonal shaft to the presynaptic terminal but also its activation in the axonal bouton by PKC-induced calcium-dependent phosphorylation at Ser-41 (pGAP-43). We found that this effect requires open presynaptic N-methyl-d-aspartate receptors but not plasmin generation. Furthermore, our work reveals that, following its activation by uPA/uPAR binding, pGAP-43 colocalizes with presynaptic vesicles and triggers their mobilization to the synaptic release site. Together, these data reveal a novel role of uPA as an activator of the synaptic vesicle cycle in cerebral cortical neurons via its ability to induce presynaptic recruitment and activation of GAP-43.
Collapse
Affiliation(s)
- Paola Merino
- Division of Neuropharmacology and Neurologic Diseases, Yerkes National Primate Research Center, Atlanta, Georgia 30329-4208
| | - Ariel Diaz
- Division of Neuropharmacology and Neurologic Diseases, Yerkes National Primate Research Center, Atlanta, Georgia 30329-4208
| | - Enrique R Torre
- Division of Neuropharmacology and Neurologic Diseases, Yerkes National Primate Research Center, Atlanta, Georgia 30329-4208
| | - Manuel Yepes
- Division of Neuropharmacology and Neurologic Diseases, Yerkes National Primate Research Center, Atlanta, Georgia 30329-4208; Department of Neurology and Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, Georgia 30322-0001; Department of Neurology, Veterans Affairs Medical Center, Atlanta, Georgia 30033-4004.
| |
Collapse
|