1
|
Tseng HC, Wang YS, Pan CY. Glutamate gradually elevates [Zn 2+] i via the CaM-CaMKII-NOS cascade in primary cultured rat embryonic cortical neurons. Sci Rep 2025; 15:15205. [PMID: 40307298 PMCID: PMC12043812 DOI: 10.1038/s41598-025-99142-1] [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: 06/10/2024] [Accepted: 04/17/2025] [Indexed: 05/02/2025] Open
Abstract
Zn2+ is essential for neuronal signaling, but imbalance cause cell death and neurodegenerative disorders. While the buffering system maintains low cytosolic Zn2+ concentration ([Zn2+]i), the details on physiological stimuli elevating [Zn2+]i for neuronal processes remain limited. Our previous reports have demonstrated that dopamine elevates [Zn2+]i through the cAMP-NO pathway, activating autophagy and inflammation in neurons. In this study, we adopted the Zn2+ imaging technique to verify how glutamate elevated [Zn2+]i in cultured cortical neurons and examined the inflammatory response. Our results showed that glutamate elevates the [Zn2+]i, by activating ionotropic glutamate receptors. Inhibitors of calmodulin (CaM), CaM-dependent protein kinase II (CaMKII), and NO synthase (NOS) blocked the glutamate-induced Zn2+ response. High-K+ buffer induced-membrane depolarization significantly elevated the intracellular Ca2+ concentration ([Ca2+]i) but only slightly increased [Zn2+]i and NO production. Glutamate also transiently increased NOS phosphorylation at Ser1417 within 15 min. The Zn2+ chelator, TPEN suppressed glutamate-induced inflammasome formation. These results indicate that glutamate-induced local increment in [Ca2+]i via the ionotropic glutamate receptors activates the CaM-CaMKII-NOS complex to produce NO and elevate [Zn2+]i. which trigger inflammation in cultured neurons. Henceforth, this novel glutamate-Zn2+ signaling pathway after glutamate depolarization elevates [Ca2+]i indicates the involvement of Zn2+ in modulating long-term neuronal activities.
Collapse
Affiliation(s)
- Hui-Chiun Tseng
- Department of Life Science, National Taiwan University, 1 Roosevelt Rd. Sec 4, Taipei, 106, Taiwan
| | - Yong-Sheng Wang
- Department of Life Science, National Taiwan University, 1 Roosevelt Rd. Sec 4, Taipei, 106, Taiwan
| | - Chien-Yuan Pan
- Department of Life Science, National Taiwan University, 1 Roosevelt Rd. Sec 4, Taipei, 106, Taiwan.
- Graduate Institute of Brain and Mind Science, National Taiwan University, Taipei, Taiwan.
| |
Collapse
|
2
|
Massey S, Ang CS, Davidson NM, Quigley A, Rollo B, Harris AR, Kapsa RMI, Christodoulou J, Van Bergen NJ. Novel CDKL5 targets identified in human iPSC-derived neurons. Cell Mol Life Sci 2024; 81:347. [PMID: 39136782 PMCID: PMC11335273 DOI: 10.1007/s00018-024-05389-8] [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: 03/26/2024] [Revised: 07/01/2024] [Accepted: 07/31/2024] [Indexed: 08/22/2024]
Abstract
CDKL5 Deficiency Disorder (CDD) is a debilitating epileptic encephalopathy disorder affecting young children with no effective treatments. CDD is caused by pathogenic variants in Cyclin-Dependent Kinase-Like 5 (CDKL5), a protein kinase that regulates key phosphorylation events in neurons. For therapeutic intervention, it is essential to understand molecular pathways and phosphorylation targets of CDKL5. Using an unbiased phosphoproteomic approach we identified novel targets of CDKL5, including GTF2I, PPP1R35, GATAD2A and ZNF219 in human iPSC-derived neuronal cells. The phosphoserine residue in the target proteins lies in the CDKL5 consensus motif. We validated direct phosphorylation of GTF2I and PPP1R35 by CDKL5 using complementary approaches. GTF2I controls axon guidance, cell cycle and neurodevelopment by regulating expression of neuronal genes. PPP1R35 is critical for centriole elongation and cilia morphology, processes that are impaired in CDD. PPP1R35 interacts with CEP131, a known CDKL5 phospho-target. GATAD2A and ZNF219 belong to the Nucleosome Remodelling Deacetylase (NuRD) complex, which regulates neuronal activity-dependent genes and synaptic connectivity. In-depth knowledge of molecular pathways regulated by CDKL5 will allow a better understanding of druggable disease pathways to fast-track therapeutic development.
Collapse
Affiliation(s)
- Sean Massey
- Brain and Mitochondrial Research Group, Murdoch Children's Research Institute, Royal Children's Hospital, Melbourne, VIC, 3052, Australia
| | - Ching-Seng Ang
- The Bio21 Institute of Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, VIC, Australia
| | - Nadia M Davidson
- Murdoch Children's Research Institute, Royal Children's Hospital, Melbourne, 3052, Australia
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, 3052, Australia
| | - Anita Quigley
- Electrical and Biomedical Engineering, School of Engineering, RMIT University, Melbourne, VIC, Australia
- Aikenhead Centre for Medical Discovery, St Vincent's Hospital Melbourne, Fitzroy, Melbourne, VIC, 3065, Australia
- Centre for Clinical Neurosciences and Neurological Research, St. Vincent's Hospital Melbourne, Fitzroy, Melbourne, VIC, 3065, Australia
- Department of Medicine, St Vincent's Hospital Melbourne, The University of Melbourne, Fitzroy, Melbourne, VIC, 3065, Australia
| | - Ben Rollo
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Australia
| | - Alexander R Harris
- Department of Biomedical Engineering, University of Melbourne, Melbourne, 3010, Australia
| | - Robert M I Kapsa
- Electrical and Biomedical Engineering, School of Engineering, RMIT University, Melbourne, VIC, Australia
- Aikenhead Centre for Medical Discovery, St Vincent's Hospital Melbourne, Fitzroy, Melbourne, VIC, 3065, Australia
- Centre for Clinical Neurosciences and Neurological Research, St. Vincent's Hospital Melbourne, Fitzroy, Melbourne, VIC, 3065, Australia
- Department of Medicine, St Vincent's Hospital Melbourne, The University of Melbourne, Fitzroy, Melbourne, VIC, 3065, Australia
| | - John Christodoulou
- Brain and Mitochondrial Research Group, Murdoch Children's Research Institute, Royal Children's Hospital, Melbourne, VIC, 3052, Australia
- Murdoch Children's Research Institute, Royal Children's Hospital, Melbourne, 3052, Australia
- Victorian Clinical Genetics Services, Royal Children's Hospital, Melbourne, VIC, 3052, Australia
- Discipline of Child and Adolescent Health, Sydney Medical School, University of Sydney, Sydney, NSW, Australia
| | - Nicole J Van Bergen
- Brain and Mitochondrial Research Group, Murdoch Children's Research Institute, Royal Children's Hospital, Melbourne, VIC, 3052, Australia.
- Murdoch Children's Research Institute, Royal Children's Hospital, Melbourne, 3052, Australia.
- Department of Paediatrics, University of Melbourne, c/o MCRI, 50 Flemington Road, Parkville, VIC, 3052, Australia.
| |
Collapse
|
3
|
Neumann S, Kuteykin-Teplyakov K, Heumann R. Neuronal Protection by Ha-RAS-GTPase Signaling through Selective Downregulation of Plasmalemmal Voltage-Dependent Anion Channel-1. Int J Mol Sci 2024; 25:3030. [PMID: 38474278 DOI: 10.3390/ijms25053030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 02/29/2024] [Accepted: 03/01/2024] [Indexed: 03/14/2024] Open
Abstract
The small GTPase RAS acts as a plasma membrane-anchored intracellular neurotrophin counteracting neuronal degeneration in the brain, but the underlying molecular mechanisms are largely unknown. In transgenic mice expressing constitutively activated V12-Ha-RAS selectively in neurons, proteome analysis uncovered a 70% decrease in voltage-dependent anion channel-1 (VDAC-1) in the cortex and hippocampus. We observed a corresponding reduction in the levels of mRNA splicing variant coding for plasma membrane-targeted VDAC-1 (pl-VDAC-1) while mRNA levels for mitochondrial membrane VDAC-1 (mt-VDAC-1) remained constant. In primary cortical neurons derived from V12-Ha-RAS animals, a decrease in pl-VDAC-1 mRNA levels was observed, accompanied by a concomitant reduction in the ferricyanide reductase activity associated with VDAC-1 protein. Application of MEK inhibitor U0126 to transgenic cortical neurons reconstituted pl-VDAC-1 mRNA to reach wild-type levels. Excitotoxic glutamate-induced cell death was strongly attenuated in transgenic V12-Ha-RAS overexpressing cortical cultures. Consistently, a neuroprotective effect could also be achieved in wild-type cortical cultures by the extracellular application of channel-blocking antibody targeting the N-terminus of VDAC-1. These results may encourage novel therapeutic approaches toward blocking pl-VDAC-1 by monoclonal antibody targeting for complementary treatments in transplantation and neurodegenerative disease.
Collapse
Affiliation(s)
- Sebastian Neumann
- Department of Biochemistry II-Molecular Neurobiochemistry, Faculty of Chemistry and Biochemistry, Ruhr-Universität Bochum, 44801 Bochum, Germany
| | - Konstantin Kuteykin-Teplyakov
- Department of Biochemistry II-Molecular Neurobiochemistry, Faculty of Chemistry and Biochemistry, Ruhr-Universität Bochum, 44801 Bochum, Germany
| | - Rolf Heumann
- Department of Biochemistry II-Molecular Neurobiochemistry, Faculty of Chemistry and Biochemistry, Ruhr-Universität Bochum, 44801 Bochum, Germany
| |
Collapse
|
4
|
Werner B, Yadav S. Phosphoregulation of the septin cytoskeleton in neuronal development and disease. Cytoskeleton (Hoboken) 2023; 80:275-289. [PMID: 36127729 PMCID: PMC10025170 DOI: 10.1002/cm.21728] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 08/13/2022] [Accepted: 09/12/2022] [Indexed: 11/06/2022]
Abstract
Septins are highly conserved GTP-binding proteins that oligomerize and form higher order structures. The septin cytoskeleton plays an important role in cellular organization, intracellular transport, and cytokinesis. Kinase-mediated phosphorylation of septins regulates various aspects of their function, localization, and dynamics. Septins are enriched in the mammalian nervous system where they contribute to neurodevelopment and neuronal function. Emerging research has implicated aberrant changes in septin cytoskeleton in several human diseases. The mechanisms through which aberrant phosphorylation by kinases contributes to septin dysfunction in neurological disorders are poorly understood and represent an important question for future research with therapeutic implications. This review summarizes the current state of knowledge of the diversity of kinases that interact with and phosphorylate mammalian septins, delineates how phosphoregulation impacts septin dynamics, and describes how aberrant septin phosphorylation contributes to neurological disorders.
Collapse
Affiliation(s)
- Bailey Werner
- Department of Pharmacology, University of Washington, Seattle, WA, United States
| | - Smita Yadav
- Department of Pharmacology, University of Washington, Seattle, WA, United States
| |
Collapse
|
5
|
Ameen SS, Griem-Krey N, Dufour A, Hossain MI, Hoque A, Sturgeon S, Nandurkar H, Draxler DF, Medcalf RL, Kamaruddin MA, Lucet IS, Leeming MG, Liu D, Dhillon A, Lim JP, Basheer F, Zhu HJ, Bokhari L, Roulston CL, Paradkar PN, Kleifeld O, Clarkson AN, Wellendorph P, Ciccotosto GD, Williamson NA, Ang CS, Cheng HC. N-Terminomic Changes in Neurons During Excitotoxicity Reveal Proteolytic Events Associated With Synaptic Dysfunctions and Potential Targets for Neuroprotection. Mol Cell Proteomics 2023; 22:100543. [PMID: 37030595 PMCID: PMC10199228 DOI: 10.1016/j.mcpro.2023.100543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 02/23/2023] [Accepted: 04/04/2023] [Indexed: 04/10/2023] Open
Abstract
Excitotoxicity, a neuronal death process in neurological disorders such as stroke, is initiated by the overstimulation of ionotropic glutamate receptors. Although dysregulation of proteolytic signaling networks is critical for excitotoxicity, the identity of affected proteins and mechanisms by which they induce neuronal cell death remain unclear. To address this, we used quantitative N-terminomics to identify proteins modified by proteolysis in neurons undergoing excitotoxic cell death. We found that most proteolytically processed proteins in excitotoxic neurons are likely substrates of calpains, including key synaptic regulatory proteins such as CRMP2, doublecortin-like kinase I, Src tyrosine kinase and calmodulin-dependent protein kinase IIβ (CaMKIIβ). Critically, calpain-catalyzed proteolytic processing of these proteins generates stable truncated fragments with altered activities that potentially contribute to neuronal death by perturbing synaptic organization and function. Blocking calpain-mediated proteolysis of one of these proteins, Src, protected against neuronal loss in a rat model of neurotoxicity. Extrapolation of our N-terminomic results led to the discovery that CaMKIIα, an isoform of CaMKIIβ, undergoes differential processing in mouse brains under physiological conditions and during ischemic stroke. In summary, by identifying the neuronal proteins undergoing proteolysis during excitotoxicity, our findings offer new insights into excitotoxic neuronal death mechanisms and reveal potential neuroprotective targets for neurological disorders.
Collapse
Affiliation(s)
- S Sadia Ameen
- Department of Biochemistry and Pharmacology, University of Melbourne, Parkville, Victoria, Australia; Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, Victoria, Australia
| | - Nane Griem-Krey
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Antoine Dufour
- Department of Physiology and Pharmacology, University of Calgary, Calgary, Alberta, Canada
| | - M Iqbal Hossain
- Department of Biochemistry and Pharmacology, University of Melbourne, Parkville, Victoria, Australia; Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, Victoria, Australia; Department of Pharmacology and Toxicology, University of Alabama, Birmingham, Alabama, USA
| | - Ashfaqul Hoque
- St Vincent's Institute of Medical Research, Fitzroy, Victoria, Australia
| | - Sharelle Sturgeon
- Australian Centre for Blood Diseases, Monash University, Melbourne, Victoria, Australia
| | - Harshal Nandurkar
- Australian Centre for Blood Diseases, Monash University, Melbourne, Victoria, Australia
| | - Dominik F Draxler
- Australian Centre for Blood Diseases, Monash University, Melbourne, Victoria, Australia
| | - Robert L Medcalf
- Australian Centre for Blood Diseases, Monash University, Melbourne, Victoria, Australia
| | - Mohd Aizuddin Kamaruddin
- Department of Biochemistry and Pharmacology, University of Melbourne, Parkville, Victoria, Australia; Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, Victoria, Australia
| | - Isabelle S Lucet
- Chemical Biology Division, The Walter and Eliza Hall Institute for Medical Research, Parkville, Victoria, Australia; Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
| | - Michael G Leeming
- Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, Victoria, Australia
| | - Dazhi Liu
- Department of Neurology, School of Medicine, University of California, Davis, California, USA
| | - Amardeep Dhillon
- Faculty of Health, Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Waurn Ponds, Victoria, Australia
| | - Jet Phey Lim
- Faculty of Health, Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Waurn Ponds, Victoria, Australia
| | - Faiza Basheer
- Faculty of Health, Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Waurn Ponds, Victoria, Australia
| | - Hong-Jian Zhu
- Department of Surgery (Royal Melbourne Hospital), University of Melbourne, Parkville, Victoria, Australia
| | - Laita Bokhari
- Department of Biochemistry and Pharmacology, University of Melbourne, Parkville, Victoria, Australia; Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, Victoria, Australia
| | - Carli L Roulston
- Florey Institute of Neuroscience and Mental Health, Parkville, Victoria, Australia
| | - Prasad N Paradkar
- CSIRO Health & Biosecurity, Australian Centre for Disease Preparedness, East Geelong, Victoria, Australia
| | - Oded Kleifeld
- Faculty of Biology, Technion-Israel Institute of Technology, Technion City, Haifa, Israel
| | - Andrew N Clarkson
- Department of Anatomy, Brain Health Research Centre and Brain Research New Zealand, University of Otago, Dunedin, New Zealand
| | - Petrine Wellendorph
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Giuseppe D Ciccotosto
- Department of Biochemistry and Pharmacology, University of Melbourne, Parkville, Victoria, Australia; Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, Victoria, Australia.
| | - Nicholas A Williamson
- Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, Victoria, Australia.
| | - Ching-Seng Ang
- Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, Victoria, Australia.
| | - Heung-Chin Cheng
- Department of Biochemistry and Pharmacology, University of Melbourne, Parkville, Victoria, Australia; Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, Victoria, Australia.
| |
Collapse
|
6
|
Regulatory Effects of Curcumin on Platelets: An Update and Future Directions. Biomedicines 2022; 10:biomedicines10123180. [PMID: 36551934 PMCID: PMC9775400 DOI: 10.3390/biomedicines10123180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 11/29/2022] [Accepted: 12/01/2022] [Indexed: 12/13/2022] Open
Abstract
The rhizomatous plant turmeric, which is frequently used as a spice and coloring ingredient, yields curcumin, a bioactive compound. Curcumin inhibits platelet activation and aggregation and improves platelet count. Platelets dysfunction results in several disorders, including inflammation, atherothrombosis, and thromboembolism. Several studies have proved the beneficial role of curcumin on platelets and hence proved it is an important candidate for the treatment of the aforementioned diseases. Moreover, curcumin is also frequently employed as an anti-inflammatory agent in conventional medicine. In arthritic patients, it has been shown to reduce the generation of pro-inflammatory eicosanoids and to reduce edema, morning stiffness, and other symptoms. Curcumin taken orally also reduced rats' acute inflammation brought on by carrageenan. Curcumin has also been proven to prevent atherosclerosis and platelet aggregation, as well as to reduce angiogenesis in adipose tissue. In the cerebral microcirculation, curcumin significantly lowered platelet and leukocyte adhesion. It largely modulated the endothelium to reduce platelet adhesion. Additionally, P-selectin expression and mice survival after cecal ligation and puncture were improved by curcumin, which also altered platelet and leukocyte adhesion and blood-brain barrier dysfunction. Through regulating many processes involved in platelet aggregation, curcuminoids collectively demonstrated detectable antiplatelet activity. Curcuminoids may therefore be able to prevent disorders linked to platelet activation as possible therapeutic agents. This review article proposes to highlight and discuss the regulatory effects of curcumin on platelets.
Collapse
|
7
|
Van Berkel AA, Koopmans F, Gonzalez-Lozano MA, Lammertse HCA, Feringa F, Bryois J, Sullivan PF, Smit AB, Toonen RF, Verhage M. Dysregulation of synaptic and developmental transcriptomic/proteomic profiles upon depletion of MUNC18-1. eNeuro 2022; 9:ENEURO.0186-22.2022. [PMID: 36257704 PMCID: PMC9668351 DOI: 10.1523/eneuro.0186-22.2022] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 06/27/2022] [Accepted: 07/14/2022] [Indexed: 11/23/2022] Open
Abstract
Absence of presynaptic protein MUNC18-1 (gene: Stxbp1) leads to neuronal cell death at an immature stage before synapse formation. Here, we performed transcriptomic and proteomic profiling of immature Stxbp1 knockout (KO) cells to discover which cellular processes depend on MUNC18-1. Hippocampi of Stxbp1 KO mice showed cell-type specific dysregulation of 2123 transcripts primarily related to synaptic transmission and immune response. To further investigate direct, neuron-specific effects of MUNC18-1 depletion, a proteomic screen was performed on murine neuronal cultures at two developmental timepoints prior to onset of neuron degeneration. 399 proteins were differentially expressed, which were primarily involved in synaptic function (especially synaptic vesicle exocytosis) and neuron development. We further show that many of the downregulated proteins upon loss of MUNC18-1 are normally upregulated during this developmental stage. Thus, absence of MUNC18-1 extensively dysregulates the transcriptome and proteome, primarily affecting synaptic and developmental profiles. Lack of synaptic activity is unlikely to underlie these effects, as the changes were observed in immature neurons without functional synapses, and minimal overlap was found to activity-dependent proteins. We hypothesize that presence of MUNC18-1 is essential to advance neuron development, serving as a 'checkpoint' for neurons to initiate cell death in its absence.Significance StatementPresynaptic protein MUNC18-1 is essential for neuronal functioning. Pathogenic variants in its gene, STXBP1, are among the most common found in patients with developmental delay and epilepsy. To discern the pathogenesis in these patients, a thorough understanding of MUNC18-1's function in neurons is required. Here, we show that loss of MUNC18-1 results in extensive dysregulation of synaptic and developmental proteins in immature neurons before synapse formation. Many of the downregulated proteins are normally upregulated during this developmental stage. This indicates that MUNC18-1 is a critical regulator of neuronal development, which could play an important role in the pathogenesis of STXBP1 variant carriers.
Collapse
Affiliation(s)
- A A Van Berkel
- Dept. Functional Genomics, CNCR, VU University Amsterdam, 1081 HV Amsterdam, The Netherlands
- Functional Genomics, Department of Human Genetics, CNCR, Amsterdam UMC, 1081 HV Amsterdam, The Netherlands
| | - F Koopmans
- Dept. Functional Genomics, CNCR, VU University Amsterdam, 1081 HV Amsterdam, The Netherlands
- Dept. Molecular & Cellular Neurobiology, CNCR, VU University Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - M A Gonzalez-Lozano
- Dept. Molecular & Cellular Neurobiology, CNCR, VU University Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - H C A Lammertse
- Dept. Functional Genomics, CNCR, VU University Amsterdam, 1081 HV Amsterdam, The Netherlands
- Functional Genomics, Department of Human Genetics, CNCR, Amsterdam UMC, 1081 HV Amsterdam, The Netherlands
| | - F Feringa
- Functional Genomics, Department of Human Genetics, CNCR, Amsterdam UMC, 1081 HV Amsterdam, The Netherlands
| | - J Bryois
- Karolinska Institutet, Department of Medical Epidemiology and Biostatistics, Nobels vag 12A, 171 77 Stockholm, Sweden
| | - P F Sullivan
- UNC Center for Psychiatric Genomics, University of North Carolina at Chapel Hill, 101 Manning Drive, Chapel Hill, NC 27599-7160, USA
- Karolinska Institutet, Department of Medical Epidemiology and Biostatistics, Nobels vag 12A, 171 77 Stockholm, Sweden
| | - A B Smit
- Dept. Molecular & Cellular Neurobiology, CNCR, VU University Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - R F Toonen
- Dept. Functional Genomics, CNCR, VU University Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - M Verhage
- Dept. Functional Genomics, CNCR, VU University Amsterdam, 1081 HV Amsterdam, The Netherlands
- Functional Genomics, Department of Human Genetics, CNCR, Amsterdam UMC, 1081 HV Amsterdam, The Netherlands
| |
Collapse
|
8
|
Calpain Inhibitors as Potential Therapeutic Modulators in Neurodegenerative Diseases. Neurochem Res 2022; 47:1125-1149. [PMID: 34982393 DOI: 10.1007/s11064-021-03521-9] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 12/27/2021] [Accepted: 12/28/2021] [Indexed: 02/07/2023]
Abstract
It is considered a significant challenge to understand the neuronal cell death mechanisms with a suitable cure for neurodegenerative disorders in the coming years. Calpains are one of the best-considered "cysteine proteases activated" in brain disorders. Calpain is an important marker and mediator in the pathophysiology of neurodegeneration. Calpain activation being the essential neurodegenerative factor causing apoptotic machinery activation, it is crucial to develop reliable and effective approaches to prevent calpain-mediated apoptosis in degenerating neurons. It has been recently seen that the "inhibition of calpain activation" has appeared as a possible therapeutic target for managing neurodegenerative diseases. A systematic literature review of PubMed, Medline, Bentham, Scopus, and EMBASE (Elsevier) databases was conducted. The present article reviews the basic pathobiology and role of selective calpain inhibitors used in various neurodegenerative diseases as a therapeutic target.
Collapse
|
9
|
CK2 Regulation: Perspectives in 2021. Biomedicines 2021; 9:biomedicines9101361. [PMID: 34680478 PMCID: PMC8533506 DOI: 10.3390/biomedicines9101361] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 09/23/2021] [Accepted: 09/26/2021] [Indexed: 12/20/2022] Open
Abstract
The protein kinase CK2 (CK2) family encompasses a small number of acidophilic serine/threonine kinases that phosphorylate substrates involved in numerous biological processes including apoptosis, cell proliferation, and the DNA damage response. CK2 has also been implicated in many human malignancies and other disorders including Alzheimer′s and Parkinson’s diseases, and COVID-19. Interestingly, no single mechanism describes how CK2 is regulated, including activation by external proteins or domains, phosphorylation, or dimerization. Furthermore, the kinase has an elongated activation loop that locks the kinase into an active conformation, leading CK2 to be labelled a constitutively active kinase. This presents an interesting paradox that remains unanswered: how can a constitutively active kinase regulate biological processes that require careful control? Here, we highlight a selection of studies where CK2 activity is regulated at the substrate level, and discuss them based on the regulatory mechanism. Overall, this review describes numerous biological processes where CK2 activity is regulated, highlighting how a constitutively active kinase can still control numerous cellular activities. It is also evident that more research is required to fully elucidate the mechanisms that regulate CK2 and what causes aberrant CK2 signaling in disease.
Collapse
|
10
|
Abstract
Intrauterine growth restriction is a condition that prevents normal fetal development, and previous studies have reported that intrauterine growth restriction is caused by adverse intrauterine factors. This condition affects both short- and long-term neurodevelopmental disorders. Studies have revealed that neurodevelopmental disorders can contribute to gray and white matter damage and decrease the brain volume of affected individuals. Further, these disorders are associated with increased risks of mental retardation, cognitive impairment, and cerebral palsy, which seriously affect the quality of life. Although the mechanisms underlying the neurologic injury associated with intrauterine growth restriction are not completely clear, studies have revealed that neuronal apoptosis, neuroinflammation, oxidative stress, excitatory toxicity, disruption of blood-brain barrier, and epigenetics may be involved in this process. This article reviews the manifestations and possible mechanisms underlying neurologic injury in intrauterine growth restriction and provides a theoretical basis for the effective prevention and treatment of this condition.
Collapse
Affiliation(s)
- Lijia Wan
- Department of Pediatrics, 70566The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Laboratory of Neonatal Disease, Institute of Pediatrics, Central South University, Changsha, Hunan, China
| | - Kaiju Luo
- Department of Pediatrics, 70566The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Laboratory of Neonatal Disease, Institute of Pediatrics, Central South University, Changsha, Hunan, China
| | - Pingyang Chen
- Department of Pediatrics, 70566The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Laboratory of Neonatal Disease, Institute of Pediatrics, Central South University, Changsha, Hunan, China
| |
Collapse
|
11
|
Leeming MG, O'Callaghan S, Licata L, Iannuccelli M, Lo Surdo P, Micarelli E, Ang CS, Nie S, Varshney S, Ameen S, Cheng HC, Williamson NA. Phosphomatics: interactive interrogation of substrate-kinase networks in global phosphoproteomics datasets. Bioinformatics 2021; 37:1635-1636. [PMID: 33119075 DOI: 10.1093/bioinformatics/btaa916] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 09/14/2020] [Accepted: 10/15/2020] [Indexed: 11/14/2022] Open
Abstract
MOTIVATION Mass spectrometry-based phosphoproteomics can routinely identify and quantify thousands of phosphorylated peptides from a single experiment. However interrogating possible upstream kinases and identifying key literature for phosphorylation sites is laborious and time-consuming. RESULTS Here, we present Phosphomatics-a publicly available web resource for interrogating phosphoproteomics data. Phosphomatics allows researchers to upload phosphoproteomics data and interrogate possible relationships from a substrate-, kinase- or pathway-centric viewpoint. AVAILABILITY AND IMPLEMENTATION Phosphomatics is freely available via the internet at: https://phosphomatics.com. SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Michael G Leeming
- Melbourne Mass Spectrometry and Proteomics Facility, Bio21 Molecular Science & Biotechnology Institute, The University of Melbourne, Melbourne, VIC 3010, Australia
| | | | - Luana Licata
- Department of Biology, University of Rome Tor Vergata, Rome 00133, Italy
| | - Marta Iannuccelli
- Department of Biochemistry and Molecular Biology, The University of Melbourne, Melbourne, VIC 3010, Australia
| | - Prisca Lo Surdo
- Department of Biology, University of Rome Tor Vergata, Rome 00133, Italy
| | - Elisa Micarelli
- Department of Biology, University of Rome Tor Vergata, Rome 00133, Italy
| | - Ching-Seng Ang
- Melbourne Mass Spectrometry and Proteomics Facility, Bio21 Molecular Science & Biotechnology Institute, The University of Melbourne, Melbourne, VIC 3010, Australia
| | - Shuai Nie
- Melbourne Mass Spectrometry and Proteomics Facility, Bio21 Molecular Science & Biotechnology Institute, The University of Melbourne, Melbourne, VIC 3010, Australia
| | - Swati Varshney
- Melbourne Mass Spectrometry and Proteomics Facility, Bio21 Molecular Science & Biotechnology Institute, The University of Melbourne, Melbourne, VIC 3010, Australia
| | - Sadia Ameen
- Department of Biochemistry and Molecular Biology, The University of Melbourne, Melbourne, VIC 3010, Australia
| | - Heung-Chin Cheng
- Department of Biochemistry and Molecular Biology, The University of Melbourne, Melbourne, VIC 3010, Australia
| | - Nicholas A Williamson
- Melbourne Mass Spectrometry and Proteomics Facility, Bio21 Molecular Science & Biotechnology Institute, The University of Melbourne, Melbourne, VIC 3010, Australia
| |
Collapse
|
12
|
Borgo C, D'Amore C, Cesaro L, Sarno S, Pinna LA, Ruzzene M, Salvi M. How can a traffic light properly work if it is always green? The paradox of CK2 signaling. Crit Rev Biochem Mol Biol 2021; 56:321-359. [PMID: 33843388 DOI: 10.1080/10409238.2021.1908951] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
CK2 is a constitutively active protein kinase that assuring a constant level of phosphorylation to its numerous substrates supports many of the most important biological functions. Nevertheless, its activity has to be controlled and adjusted in order to cope with the varying needs of a cell, and several examples of a fine-tune regulation of its activity have been described. More importantly, aberrant regulation of this enzyme may have pathological consequences, e.g. in cancer, chronic inflammation, neurodegeneration, and viral infection. Our review aims at summarizing our current knowledge about CK2 regulation. In the first part, we have considered the most important stimuli shown to affect protein kinase CK2 activity/expression. In the second part, we focus on the molecular mechanisms by which CK2 can be regulated, discussing controversial aspects and future perspectives.
Collapse
Affiliation(s)
- Christian Borgo
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Claudio D'Amore
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Luca Cesaro
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Stefania Sarno
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Lorenzo A Pinna
- Department of Biomedical Sciences, University of Padova, Padova, Italy.,CNR Institute of Neurosciences, Padova, Italy
| | - Maria Ruzzene
- Department of Biomedical Sciences, University of Padova, Padova, Italy.,CNR Institute of Neurosciences, Padova, Italy
| | - Mauro Salvi
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| |
Collapse
|
13
|
Rehfeldt SCH, Laufer S, Goettert MI. A Highly Selective In Vitro JNK3 Inhibitor, FMU200, Restores Mitochondrial Membrane Potential and Reduces Oxidative Stress and Apoptosis in SH-SY5Y Cells. Int J Mol Sci 2021; 22:ijms22073701. [PMID: 33918172 PMCID: PMC8037381 DOI: 10.3390/ijms22073701] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 03/29/2021] [Accepted: 03/30/2021] [Indexed: 12/31/2022] Open
Abstract
Current treatments for neurodegenerative diseases (ND) are symptomatic and do not affect disease progression. Slowing this progression remains a crucial unmet need for patients and their families. c-Jun N-terminal kinase 3 (JNK3) are related to several ND hallmarks including apoptosis, oxidative stress, excitotoxicity, mitochondrial dysfunction, and neuroinflammation. JNK inhibitors can play an important role in addressing neuroprotection. This research aims to evaluate the neuroprotective, anti-inflammatory, and antioxidant effects of a synthetic compound (FMU200) with known JNK3 inhibitory activity in SH-SY5Y and RAW264.7 cell lines. SH-SY5Y cells were pretreated with FMU200 and cell damage was induced by 6-hydroxydopamine (6-OHDA) or hydrogen peroxide (H2O2). Cell viability and neuroprotective effect were assessed with an MTT assay. Flow cytometric analysis was performed to evaluate cell apoptosis. The H2O2-induced reactive oxygen species (ROS) generation and mitochondrial membrane potential (ΔΨm) were evaluated by DCFDA and JC-1 assays, respectively. The anti-inflammatory effect was determined in LPS-induced RAW264.7 cells by ELISA assay. In undifferentiated SH-SY5Y cells, FMU200 decreased neurotoxicity induced by 6-OHDA in approximately 20%. In RA-differentiated cells, FMU200 diminished cell death in approximately 40% and 90% after 24 and 48 h treatment, respectively. FMU200 reduced both early and late apoptotic cells, decreased ROS levels, restored mitochondrial membrane potential, and downregulated JNK phosphorylation after H2O2 exposure. In LPS-stimulated RAW264.7 cells, FMU200 reduced TNF-α levels after a 3 h treatment. FMU200 protects neuroblastoma SH-SY5Y cells against 6-OHDA- and H2O2-induced apoptosis, which may result from suppressing the JNK pathways. Our findings show that FMU200 can be a useful candidate for the treatment of neurodegenerative disorders.
Collapse
Affiliation(s)
| | - Stefan Laufer
- Department of Pharmaceutical and Medicinal Chemistry, Institute of Pharmacy, Eberhard Karls Universität Tübingen, D-72076 Tübingen, Germany
- Tübingen Center for Academic Drug Discovery (TüCAD2), D-72076 Tübingen, Germany
- Correspondence: (S.L.); (M.I.G.); Tel.: +55-(51)3714-7000 (ext. 5445) (M.I.G.)
| | - Márcia Inês Goettert
- Graduate Program in Biotechnology, University of Vale do Taquari (Univates), Lajeado, RS 95914-014, Brazil;
- Correspondence: (S.L.); (M.I.G.); Tel.: +55-(51)3714-7000 (ext. 5445) (M.I.G.)
| |
Collapse
|
14
|
eNOS-dependent S-nitrosylation of the NF-κB subunit p65 has neuroprotective effects. Cell Death Dis 2021; 12:4. [PMID: 33414434 PMCID: PMC7790835 DOI: 10.1038/s41419-020-03338-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 12/02/2020] [Accepted: 12/09/2020] [Indexed: 01/29/2023]
Abstract
Cell death by glutamate excitotoxicity, mediated by N-methyl-D-aspartate (NMDA) receptors, negatively impacts brain function, including but not limited to hippocampal neurons. The NF-κB transcription factor (composed mainly of p65/p50 subunits) contributes to neuronal death in excitotoxicity, while its inhibition should improve cell survival. Using the biotin switch method, subcellular fractionation, immunofluorescence, and luciferase reporter assays, we found that NMDA-stimulated NF-κB activity selectively in hippocampal neurons, while endothelial nitric oxide synthase (eNOS), an enzyme expressed in neurons, is involved in the S-nitrosylation of p65 and consequent NF-κB inhibition in cerebrocortical, i.e., resistant neurons. The S-nitro proteomes of cortical and hippocampal neurons revealed that different biological processes are regulated by S-nitrosylation in susceptible and resistant neurons, bringing to light that protein S-nitrosylation is a ubiquitous post-translational modification, able to influence a variety of biological processes including the homeostatic inhibition of the NF-κB transcriptional activity in cortical neurons exposed to NMDA receptor overstimulation.
Collapse
|
15
|
Bartley AF, Abiraman K, Stewart LT, Hossain MI, Gahan DM, Kamath AV, Burdette MK, Andrabe S, Foulger SH, McMahon LL, Dobrunz LE. LSO:Ce Inorganic Scintillators Are Biocompatible With Neuronal and Circuit Function. Front Synaptic Neurosci 2019; 11:24. [PMID: 31551750 PMCID: PMC6733890 DOI: 10.3389/fnsyn.2019.00024] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Accepted: 08/06/2019] [Indexed: 12/19/2022] Open
Abstract
Optogenetics is widely used in neuroscience to control neural circuits. However, non-invasive methods for light delivery in brain are needed to avoid physical damage caused by current methods. One potential strategy could employ x-ray activation of radioluminescent particles (RPLs), enabling localized light generation within the brain. RPLs composed of inorganic scintillators can emit light at various wavelengths depending upon composition. Cerium doped lutetium oxyorthosilicate (LSO:Ce), an inorganic scintillator that emits blue light in response to x-ray or ultraviolet (UV) stimulation, could potentially be used to control neural circuits through activation of channelrhodopsin-2 (ChR2), a light-gated cation channel. Whether inorganic scintillators themselves negatively impact neuronal processes and synaptic function is unknown, and was investigated here using cellular, molecular, and electrophysiological approaches. As proof of principle, we applied UV stimulation to 4 μm LSO:Ce particles during whole-cell recording of CA1 pyramidal cells in acute hippocampal slices from mice that expressed ChR2 in glutamatergic neurons. We observed an increase in frequency and amplitude of spontaneous excitatory postsynaptic currents (sEPSCs), indicating activation of ChR2 and excitation of neurons. Importantly, LSO:Ce particles did not affect survival of primary mouse cortical neurons, even after 24 h of exposure. In extracellular dendritic field potential recordings, no change in the strength of basal glutamatergic transmission was observed during exposure to LSO:Ce microparticles. However, the amplitude of the fiber volley was slightly reduced with high stimulation. Additionally, there was a slight decrease in the frequency of sEPSCs in whole-cell voltage-clamp recordings from CA1 pyramidal cells, with no change in current amplitudes. The amplitude and frequency of spontaneous inhibitory postsynaptic currents were unchanged. Finally, long term potentiation (LTP), a synaptic modification believed to underlie learning and memory and a robust measure of synaptic integrity, was successfully induced, although the magnitude was slightly reduced. Together, these results show LSO:Ce particles are biocompatible even though there are modest effects on baseline synaptic function and long-term synaptic plasticity. Importantly, we show that light emitted from LSO:Ce particles is able to activate ChR2 and modify synaptic function. Therefore, LSO:Ce inorganic scintillators are potentially viable for use as a new light delivery system for optogenetics.
Collapse
Affiliation(s)
- Aundrea F. Bartley
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL, United States
- Civitan International Research Center, University of Alabama at Birmingham, Birmingham, AL, United States
- Evelyn F. McKnight Brain Institute, University of Alabama at Birmingham, Birmingham, AL, United States
- Comprehensive Neuroscience Center, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Kavitha Abiraman
- Comprehensive Neuroscience Center, University of Alabama at Birmingham, Birmingham, AL, United States
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Luke T. Stewart
- Comprehensive Neuroscience Center, University of Alabama at Birmingham, Birmingham, AL, United States
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Mohammed Iqbal Hossain
- Department of Pharmacology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - David M. Gahan
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Abhishek V. Kamath
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Mary K. Burdette
- Department of Materials Science and Engineering, Clemson University, Anderson, SC, United States
| | - Shaida Andrabe
- Department of Pharmacology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Stephen H. Foulger
- Department of Materials Science and Engineering, Clemson University, Anderson, SC, United States
- Center for Optical Materials Science and Engineering Technologies, Clemson University, Anderson, SC, United States
- Department of Bioengineering, Clemson University, Clemson, SC, United States
| | - Lori L. McMahon
- Civitan International Research Center, University of Alabama at Birmingham, Birmingham, AL, United States
- Evelyn F. McKnight Brain Institute, University of Alabama at Birmingham, Birmingham, AL, United States
- Comprehensive Neuroscience Center, University of Alabama at Birmingham, Birmingham, AL, United States
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Lynn E. Dobrunz
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL, United States
- Civitan International Research Center, University of Alabama at Birmingham, Birmingham, AL, United States
- Evelyn F. McKnight Brain Institute, University of Alabama at Birmingham, Birmingham, AL, United States
- Comprehensive Neuroscience Center, University of Alabama at Birmingham, Birmingham, AL, United States
| |
Collapse
|