1
|
Park IK, Choi YS, Jo SY. Development of quantitative detection methods for four Alzheimer's disease specific biomarker panels using electrochemical immunosensors based on enzyme immunoassay. ANAL SCI 2024; 40:1809-1821. [PMID: 38884905 DOI: 10.1007/s44211-024-00614-7] [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: 03/04/2024] [Accepted: 06/02/2024] [Indexed: 06/18/2024]
Abstract
Accurate and timely diagnosis of Alzheimer's disease (AD) is necessary to maximize the effectiveness of treatment and using biomarkers for diagnosis is attracting attention as a minimally invasive method with few side effects. Electrochemical immunosensor (EI) is a method that is in the spotlight in the medical and bioanalytical fields due to its portability and field usability. Here, we quantified four AD specific biomarkers using EIs based on enzyme immunoassay. We selected and developed quantitative methods for the biomarkers using screen-printed gold electrodes. For three biomarkers, quantification was performed using competition immunoassays in which antigen-antibody premix mixtures were applied to antigen-immobilized electrodes and the limit of detection (LOD) values were secured, 1.20 ng/ml, 1.30 ng/ml, and 1.74 ng/ml, respectively. For the other, a sandwich immunoassay using antibody pair was selected for quantification and LOD was also achieved as 0.077 ng/ml. All four biomarkers in buffer samples were successfully quantified and reliable R2 values were obtained, and reliable calibration curves were secured for three biomarkers in spiked human serum samples. The immunosensors developed and will be optimized are expected to be used in various fields, including detection of biomarkers for not only AD but also related diseases.
Collapse
Affiliation(s)
- Il Kyu Park
- JHK Medical Science Inc., Yuseong-gu, Daejeon, 34013, Republic of Korea
| | - Young Sun Choi
- JHK Medical Science Inc., Yuseong-gu, Daejeon, 34013, Republic of Korea
| | - Seo Yun Jo
- JHK Medical Science Inc., Yuseong-gu, Daejeon, 34013, Republic of Korea.
| |
Collapse
|
2
|
Nheu D, Petratos S. How does Nogo-A signalling influence mitochondrial function during multiple sclerosis pathogenesis? Neurosci Biobehav Rev 2024; 163:105767. [PMID: 38885889 DOI: 10.1016/j.neubiorev.2024.105767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2024] [Revised: 05/30/2024] [Accepted: 06/08/2024] [Indexed: 06/20/2024]
Abstract
Multiple sclerosis (MS) is a severe neurological disorder that involves inflammation in the brain, spinal cord and optic nerve with key disabling neuropathological outcomes being axonal damage and demyelination. When degeneration of the axo-glial union occurs, a consequence of inflammatory damage to central nervous system (CNS) myelin, dystrophy and death can lead to large membranous structures from dead oligodendrocytes and degenerative myelin deposited in the extracellular milieu. For the first time, this review covers mitochondrial mechanisms that may be operative during MS-related neurodegenerative changes directly activated during accumulating extracellular deposits of myelin associated inhibitory factors (MAIFs), that include the potent inhibitor of neurite outgrowth, Nogo-A. Axonal damage may occur when Nogo-A binds to and signals through its cognate receptor, NgR1, a multimeric complex, to initially stall axonal transport and limit the delivery of important growth-dependent cargo and subcellular organelles such as mitochondria for metabolic efficiency at sites of axo-glial disintegration as a consequence of inflammation. Metabolic efficiency in axons fails during active demyelination and progressive neurodegeneration, preceded by stalled transport of functional mitochondria to fuel axo-glial integrity.
Collapse
Affiliation(s)
- Danica Nheu
- Department of Neuroscience, School of Translational Medicine, Monash University, Prahran, VIC 3004, Australia
| | - Steven Petratos
- Department of Neuroscience, School of Translational Medicine, Monash University, Prahran, VIC 3004, Australia.
| |
Collapse
|
3
|
Perez-Miller S, Gomez K, Khanna R. Peptide and Peptidomimetic Inhibitors Targeting the Interaction of Collapsin Response Mediator Protein 2 with the N-Type Calcium Channel for Pain Relief. ACS Pharmacol Transl Sci 2024; 7:1916-1936. [PMID: 39022365 PMCID: PMC11249630 DOI: 10.1021/acsptsci.4c00181] [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: 03/28/2024] [Revised: 05/13/2024] [Accepted: 05/23/2024] [Indexed: 07/20/2024]
Abstract
Ion channels serve pleiotropic functions. Often found in complexes, their activities and functions are sculpted by auxiliary proteins. We discovered that collapsin response mediator protein 2 (CRMP2) is a binding partner and regulator of the N-type voltage-gated calcium channel (CaV2.2), a genetically validated contributor to chronic pain. Herein, we trace the discovery of a new peptidomimetic modulator of this interaction, starting from the identification and development of CBD3, a CRMP2-derived CaV binding domain peptide. CBD3 uncouples CRMP2-CaV2.2 binding to decrease CaV2.2 surface localization and calcium currents. These changes occur at presynaptic sites of nociceptive neurons and indeed, CBD3 ameliorates chronic pain in preclinical models. In pursuit of a CBD3 peptidomimetic, we exploited a unique approach to identify a dipeptide with low conformational flexibility and high solvent accessibility that anchors binding to CaV2.2. From a pharmacophore screen, we obtained CBD3063, a small-molecule that recapitulated CBD3's activity, reversing nociceptive behaviors in rodents of both sexes without sensory, affective, or cognitive effects. By disrupting the CRMP2-CaV2.2 interaction, CBD3063 exerts these effects indirectly through modulating CaV2.2 trafficking, supporting CRMP2 as an auxiliary subunit of CaV2.2. The parent peptide CBD3 was also found by us and others to have neuroprotective properties at postsynaptic sites, through N-methyl-d-aspartate receptor and plasmalemmal Na+/Ca2+ exchanger 3, potentially acting as an auxiliary subunit for these pathways as well. Our new compound is poised to address several open questions regarding CRMP2's role in regulating the CaV2.2 pathways to treat pain with the potential added benefit of neuroprotection.
Collapse
Affiliation(s)
- Samantha Perez-Miller
- Department
of Pharmacology & Therapeutics, College of Medicine, University of Florida, 1200 Newell Drive, ARB R5-234, Gainesville, Florida 32610-0267, United States
| | - Kimberly Gomez
- Department
of Pharmacology & Therapeutics, College of Medicine, University of Florida, 1200 Newell Drive, ARB R5-234, Gainesville, Florida 32610-0267, United States
| | - Rajesh Khanna
- Department
of Pharmacology & Therapeutics, College of Medicine, University of Florida, 1200 Newell Drive, ARB R5-234, Gainesville, Florida 32610-0267, United States
- Pain
and Addiction Therapeutics (PATH) Collaboratory, College of Medicine, University of Florida, Gainesville, Florida 32610, United States
| |
Collapse
|
4
|
Jiang L, Li Y, Gu Y, Zheng J, Wei L, Wei M, Zou J, Wei C, Mo B, Pan L, Zhao L, Wang D. Identification of the Beta Subunit Fas1p of Fatty Acid Synthetase as an Interacting Partner of Yeast Calcium/Calmodulin-Dependent Protein Kinase Cmk2p Through Mass Spectrometry Analysis. Appl Biochem Biotechnol 2024:10.1007/s12010-024-04891-w. [PMID: 38411936 DOI: 10.1007/s12010-024-04891-w] [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] [Accepted: 02/12/2024] [Indexed: 02/28/2024]
Abstract
The calcium/calmodulin-dependent protein kinase II (CaMKII) is a mediator of calcium signals and regulates fatty acid metabolism in mammalian cells. Cmk2p is a yeast homolog of CaMKII and functions as a negative regulator of calcium signaling. However, its substrates remain to be identified. Combination of immunoprecipitation (IP) and mass spectrometry has been proven to be very useful for identification of interacting partner proteins and interactome. In this study, through these approaches, we have identified 65 and 110 potential Cmk2p-interacting proteins in yeast cells in the absence or presence of calcium stress, respectively. In yeast cells expressing both CMK2-HA and FAS1-GFP fusion proteins, in the absence or presence of calcium stress, less amounts of FAS1-GFP proteins are present in cell lysates after IP with anti-HA antibody than cell lysates before IP, while FAS1-GFP proteins are detected on both types of IP beads. However, as an internal control, similar amounts of Pgk1p proteins were detected in both after-IP and before-IP cell lysates but not on the IP beads. Therefore, our biochemical analysis demonstrates that the β subunit Fas1p of fatty acid synthetase interacts with Cmk2p in yeast cells independent of calcium stress. It is also interesting to note that, in addition to the expected 52-kDa CMK2-HA band, a faster-moving 48-kDa CMK2-HA band is present in the calcium-stressed cell lysate but not in the cell lysate without calcium stress. Our data would provide important clues for understanding the functions of CaMKII in the regulation of fatty acid metabolism as well as related diseases such as cancers, diabetes, and obesity.
Collapse
Affiliation(s)
- Linghuo Jiang
- Laboratory of Yeast Biology and Fermentation Technology, National Engineering Research Center for Non-Food Biorefinery, National Key Laboratory of Non-Food Biomass Energy Technology, Guangxi Biomass Engineering Technology Research Center, Institute of Biological Sciences and Technology, Guangxi Academy of Sciences, Nanning, 530007, Guangxi, China.
| | - Yiwu Li
- College of Chemistry and Materials, Guangxi Key Laboratory of Natural Polymer Chemistry and Physics, Nanning Normal University, Nanning, 530001, China
| | - Yiying Gu
- Laboratory of Yeast Biology and Fermentation Technology, National Engineering Research Center for Non-Food Biorefinery, National Key Laboratory of Non-Food Biomass Energy Technology, Guangxi Biomass Engineering Technology Research Center, Institute of Biological Sciences and Technology, Guangxi Academy of Sciences, Nanning, 530007, Guangxi, China
| | - Jiashi Zheng
- College of Chemistry and Materials, Guangxi Key Laboratory of Natural Polymer Chemistry and Physics, Nanning Normal University, Nanning, 530001, China
| | - Liudan Wei
- Laboratory of Yeast Biology and Fermentation Technology, National Engineering Research Center for Non-Food Biorefinery, National Key Laboratory of Non-Food Biomass Energy Technology, Guangxi Biomass Engineering Technology Research Center, Institute of Biological Sciences and Technology, Guangxi Academy of Sciences, Nanning, 530007, Guangxi, China
| | - Min Wei
- College of Chemistry and Materials, Guangxi Key Laboratory of Natural Polymer Chemistry and Physics, Nanning Normal University, Nanning, 530001, China
| | - Jie Zou
- College of Chemistry and Materials, Guangxi Key Laboratory of Natural Polymer Chemistry and Physics, Nanning Normal University, Nanning, 530001, China
| | - Chunyu Wei
- College of Chemistry and Materials, Guangxi Key Laboratory of Natural Polymer Chemistry and Physics, Nanning Normal University, Nanning, 530001, China
| | - Bei Mo
- College of Chemistry and Materials, Guangxi Key Laboratory of Natural Polymer Chemistry and Physics, Nanning Normal University, Nanning, 530001, China
| | - Lingxin Pan
- College of Chemistry and Materials, Guangxi Key Laboratory of Natural Polymer Chemistry and Physics, Nanning Normal University, Nanning, 530001, China
| | - Lijiao Zhao
- College of Chemistry and Materials, Guangxi Key Laboratory of Natural Polymer Chemistry and Physics, Nanning Normal University, Nanning, 530001, China
| | - Dan Wang
- College of Chemistry and Materials, Guangxi Key Laboratory of Natural Polymer Chemistry and Physics, Nanning Normal University, Nanning, 530001, China
| |
Collapse
|
5
|
Lakha R, Hachicho C, Mehlenbacher MR, Wilcox DE, Austin RN, Vizcarra CL. Metallothionein-3 attenuates the effect of Cu 2+ ions on actin filaments. J Inorg Biochem 2023; 242:112157. [PMID: 36801620 DOI: 10.1016/j.jinorgbio.2023.112157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 02/01/2023] [Accepted: 02/02/2023] [Indexed: 02/10/2023]
Abstract
Metallothionein 3 (MT-3) is a cysteine-rich metal-binding protein that is expressed in the mammalian central nervous system and kidney. Various reports have posited a role for MT-3 in regulating the actin cytoskeleton by promoting the assembly of actin filaments. We generated purified, recombinant mouse MT-3 of known metal compositions, either with zinc (Zn), lead (Pb), or copper/zinc (Cu/Zn) bound. None of these forms of MT-3 accelerated actin filament polymerization in vitro, either with or without the actin binding protein profilin. Furthermore, using a co-sedimentation assay, we did not observe Zn-bound MT-3 in complex with actin filaments. Cu2+ ions on their own induced rapid actin polymerization, an effect that we attribute to filament fragmentation. This effect of Cu2+ is reversed by adding either EGTA or Zn-bound MT-3, indicating that either molecule can chelate Cu2+ from actin. Altogether, our data indicate that purified recombinant MT-3 does not directly bind actin but it does attenuate the Cu-induced fragmentation of actin filaments.
Collapse
Affiliation(s)
- Rabina Lakha
- Department of Chemistry, Barnard College, New York, NY 10027, USA
| | - Carla Hachicho
- Department of Chemistry, Barnard College, New York, NY 10027, USA
| | | | - Dean E Wilcox
- Department of Chemistry, Dartmouth College, Hanover, NH 03755, USA
| | - Rachel N Austin
- Department of Chemistry, Barnard College, New York, NY 10027, USA
| | | |
Collapse
|
6
|
Vantaggiato L, Shaba E, Carleo A, Bezzini D, Pannuzzo G, Luddi A, Piomboni P, Bini L, Bianchi L. Neurodegenerative Disorder Risk in Krabbe Disease Carriers. Int J Mol Sci 2022; 23:13537. [PMID: 36362324 PMCID: PMC9654610 DOI: 10.3390/ijms232113537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 10/26/2022] [Accepted: 10/30/2022] [Indexed: 11/09/2022] Open
Abstract
Krabbe disease (KD) is a rare autosomal recessive disorder caused by mutations in the galactocerebrosidase gene (GALC). Defective GALC causes aberrant metabolism of galactolipids present almost exclusively in myelin, with consequent demyelinization and neurodegeneration of the central and peripheral nervous system (NS). KD shares some similar features with other neuropathies and heterozygous carriers of GALC mutations are emerging with an increased risk in developing NS disorders. In this work, we set out to identify possible variations in the proteomic profile of KD-carrier brain to identify altered pathways that may imbalance its homeostasis and that may be associated with neurological disorders. The differential analysis performed on whole brains from 33-day-old twitcher (galc -/-), heterozygous (galc +/-), and wild-type mice highlighted the dysregulation of several multifunctional factors in both heterozygous and twitcher mice. Notably, the KD-carrier mouse, despite its normal phenotype, presents the deregulation of vimentin, receptor of activated protein C kinase 1 (RACK1), myelin basic protein (MBP), 2',3'-cyclic-nucleotide 3'-phosphodiesterase (CNP), transitional endoplasmic reticulum ATPase (VCP), and N-myc downstream regulated gene 1 protein (NDRG1) as well as changes in the ubiquitinated-protein pattern. Our findings suggest the carrier may be affected by dysfunctions classically associated with neurodegeneration: (i) alteration of (mechano) signaling and intracellular trafficking, (ii) a generalized affection of proteostasis and lipid metabolism, with possible defects in myelin composition and turnover, and (iii) mitochondrion and energy supply dysfunctions.
Collapse
Affiliation(s)
- Lorenza Vantaggiato
- Functional Proteomics Laboratory, Department of Life Sciences, University of Siena, 53100 Siena, Italy
| | - Enxhi Shaba
- Functional Proteomics Laboratory, Department of Life Sciences, University of Siena, 53100 Siena, Italy
| | - Alfonso Carleo
- Department of Pulmonology, Hannover Medical School, Carl-Neuberg-Straße 1, 30625 Hannover, Germany
| | - Daiana Bezzini
- Department of Life Sciences, University of Siena, 53100 Siena, Italy
| | - Giovanna Pannuzzo
- Department of Biochemical and Biotechnological Sciences, Section of Physiology, University of Catania, 95121 Catania, Italy
| | - Alice Luddi
- Department of Molecular and Developmental Medicine, University of Siena, 53100 Siena, Italy
| | - Paola Piomboni
- Department of Molecular and Developmental Medicine, University of Siena, 53100 Siena, Italy
| | - Luca Bini
- Functional Proteomics Laboratory, Department of Life Sciences, University of Siena, 53100 Siena, Italy
| | - Laura Bianchi
- Functional Proteomics Laboratory, Department of Life Sciences, University of Siena, 53100 Siena, Italy
| |
Collapse
|
7
|
Vibert R, Mignot C, Keren B, Chantot-Bastaraud S, Portnoï MF, Nouguès MC, Moutard ML, Faudet A, Whalen S, Haye D, Garel C, Chatron N, Rossi M, Vincent-Delorme C, Boute O, Delobel B, Andrieux J, Devillard F, Coutton C, Puechberty J, Pebrel-Richard C, Colson C, Gerard M, Missirian C, Sigaudy S, Busa T, Doco-Fenzy M, Malan V, Rio M, Doray B, Sanlaville D, Siffroi JP, Héron D, Heide S. Neurodevelopmental phenotype in 36 new patients with 8p inverted duplication-deletion: Genotype-phenotype correlation for anomalies of the corpus callosum. Clin Genet 2021; 101:307-316. [PMID: 34866188 DOI: 10.1111/cge.14096] [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: 10/06/2021] [Revised: 11/26/2021] [Accepted: 12/02/2021] [Indexed: 11/26/2022]
Abstract
Inverted duplication deletion 8p [invdupdel(8p)] is a complex and rare chromosomal rearrangement that combines a distal deletion and an inverted interstitial duplication of the short arm of chromosome 8. Carrier patients usually have developmental delay and intellectual disability (ID), associated with various cerebral and extra-cerebral malformations. Invdupdel(8p) is the most common recurrent chromosomal rearrangement in ID patients with anomalies of the corpus callosum (AnCC). Only a minority of invdupdel(8p) cases reported in the literature to date had both brain cerebral imaging and chromosomal microarray (CMA) with precise breakpoints of the rearrangements, making genotype-phenotype correlation studies for AnCC difficult. In this study, we report the clinical, radiological, and molecular data from 36 new invdupdel(8p) cases including three fetuses and five individuals from the same family, with breakpoints characterized by CMA. Among those, 97% (n = 32/33) of patients presented with mild to severe developmental delay/ID and 34% had seizures with mean age of onset of 3.9 years (2 months-9 years). Moreover, out of the 24 patients with brain MRI and 3 fetuses with neuropathology analysis, 63% (n = 17/27) had AnCC. We review additional data from 99 previously published patients with invdupdel(8p) and compare data of 17 patients from the literature with both CMA analysis and brain imaging to refine genotype-phenotype correlations for AnCC. This led us to refine a region of 5.1 Mb common to duplications of patients with AnCC and discuss potential candidate genes within this region.
Collapse
Affiliation(s)
- Roseline Vibert
- Département de Génétique, Hôpital Armand-Trousseau and Groupe Hospitalier Pitié-Salpêtrière, Centre de Référence Déficiences Intellectuelles de Causes Rares, APHP-Sorbonne Université, Paris, France
| | - Cyril Mignot
- Département de Génétique, Hôpital Armand-Trousseau and Groupe Hospitalier Pitié-Salpêtrière, Centre de Référence Déficiences Intellectuelles de Causes Rares, APHP-Sorbonne Université, Paris, France
| | - Boris Keren
- UF de Génomique du Développement, Département de Génétique, Groupe Hospitalier Pitié-Salpêtrière, APHP-Sorbonne Université, Paris, France
| | | | - Marie-France Portnoï
- Department of Cytogenetics, Armand Trousseau Hospital, APHP-Sorbonne Université, Paris, France
| | - Marie-Christine Nouguès
- Service of Pediatric Neurology, Armand Trousseau Hospital, APHP-Sorbonne Université, Paris, France
| | - Marie-Laure Moutard
- Service of Pediatric Neurology, Armand Trousseau Hospital, APHP-Sorbonne Université, Paris, France
| | - Anne Faudet
- Département de Génétique, Hôpital Armand-Trousseau and Groupe Hospitalier Pitié-Salpêtrière, Centre de Référence Déficiences Intellectuelles de Causes Rares, APHP-Sorbonne Université, Paris, France
| | - Sandra Whalen
- UF de Génétique Clinique et Centre de Référence Maladies Rares des Anomalies du Développement et Syndromes Malformatifs, Hôpital Armand Trousseau, ERN ITHACA, APHP-Sorbonne Université, Paris, France
| | - Damien Haye
- Département de Génétique, Hôpital Armand-Trousseau and Groupe Hospitalier Pitié-Salpêtrière, Centre de Référence Déficiences Intellectuelles de Causes Rares, APHP-Sorbonne Université, Paris, France
| | - Catherine Garel
- Department of Radiology, Armand Trousseau Hospital, APHP-Sorbonne Université, Paris, France
| | - Nicolas Chatron
- Departments of Genetics, Lyon University Hospitals, Lyon, France
| | - Massimiliano Rossi
- Genetics Department, Referral Centre for Developmental Abnormalities, Lyon University Hospital, and INSERM U1028, CNRS UMR5292, Lyon Neuroscience Research Centre, GENDEV Team, Claude Bernard Lyon 1 University, Bron, France
| | | | - Odile Boute
- Service of Clinical Genetic, Jeanne de Flandre Hospital, Lille, France
| | - Bruno Delobel
- Service of Cytogenetics, Institut Catholique de Lille, Lille, France
| | - Joris Andrieux
- Institute of Medical Genetics, Jeanne de Flandre Hospital, Lille, France
| | - Françoise Devillard
- Service de Génétique, Génomique, et Procréation, Centre Hospitalier Universitaire Grenoble Alpes, 38700 La Tronche, France; INSERM 1209, CNRS UMR 5309, Institute for Advanced Biosciences, Université Grenoble Alpes, Grenoble, France
| | - Charles Coutton
- Service de Génétique, Génomique, et Procréation, Centre Hospitalier Universitaire Grenoble Alpes, 38700 La Tronche, France; INSERM 1209, CNRS UMR 5309, Institute for Advanced Biosciences, Université Grenoble Alpes, Grenoble, France
| | - Jacques Puechberty
- Department of Medical Genetics, Arnaud de Villeneuve Hospital, Montpellier, France
| | - Céline Pebrel-Richard
- Service of Cytogenetic, Clermont-Ferrand's University Hospital, Clermont-Ferrand, France
| | - Cindy Colson
- Service of Clinical Genetic, Caen's University Hospital, Caen, France
| | - Marion Gerard
- Service of Clinical Genetic, Caen's University Hospital, Caen, France
| | - Chantal Missirian
- APHM, Laboratory of Genetic, Timone Enfants' Hospital, Marseille, France
| | - Sabine Sigaudy
- Department of Medical Genetics, Timone Enfants' Hospital, Marseille, France
| | - Tiffany Busa
- Department of Medical Genetics, Timone Enfants' Hospital, Marseille, France
| | | | - Valérie Malan
- APHP, Service de Médecine Génomique, Hôpital Necker-Enfants Malades, Paris, Université de Paris, Paris, France
| | - Marlène Rio
- Department of Genetics, Hôpital Necker-Enfants Malades, AP-HP, Paris, France
| | - Bérénice Doray
- Service of Genetic, Felix Guyon Hospital, La Réunion, France
| | | | - Jean-Pierre Siffroi
- Department of Cytogenetics, Armand Trousseau Hospital, APHP-Sorbonne Université, Paris, France
| | - Delphine Héron
- Département de Génétique, Hôpital Armand-Trousseau and Groupe Hospitalier Pitié-Salpêtrière, Centre de Référence Déficiences Intellectuelles de Causes Rares, APHP-Sorbonne Université, Paris, France
| | - Solveig Heide
- Département de Génétique, Hôpital Armand-Trousseau and Groupe Hospitalier Pitié-Salpêtrière, Centre de Référence Déficiences Intellectuelles de Causes Rares, APHP-Sorbonne Université, Paris, France
| |
Collapse
|
8
|
Morales X, Peláez R, Garasa S, Ortiz de Solórzano C, Rouzaut A. CRMP2 as a Candidate Target to Interfere with Lung Cancer Cell Migration. Biomolecules 2021; 11:biom11101533. [PMID: 34680167 PMCID: PMC8533992 DOI: 10.3390/biom11101533] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 10/12/2021] [Accepted: 10/14/2021] [Indexed: 12/15/2022] Open
Abstract
Collapsin response mediator protein 2 (CRMP2) is an adaptor protein that adds tubulin dimers to the growing tip of a microtubule. First described in neurons, it is now considered a ubiquitous protein that intervenes in processes such as cytoskeletal remodeling, synaptic connection and trafficking of voltage channels. Mounting evidence supports that CRMP2 plays an essential role in neuropathology and, more recently, in cancer. We have previously described a positive correlation between nuclear phosphorylation of CRMP2 and poor prognosis in lung adenocarcinoma patients. In this work, we studied whether this cytoskeleton molding protein is involved in cancer cell migration. To this aim, we evaluated CRMP2 phosphorylation and localization in the extending lamella of lung adenocarcinoma migrating cells using in vitro assays and in vivo confocal microscopy. We demonstrated that constitutive phosphorylation of CRMP2 impaired lamella formation, cell adhesion and oriented migration. In search of a mechanistic explanation of this phenomenon, we discovered that CRMP2 Ser522 phospho-mimetic mutants display unstable tubulin polymers, unable to bind EB1 plus-Tip protein and the cortical actin adaptor IQGAP1. In addition, integrin recycling is defective and invasive structures are less evident in these mutants. Significantly, mouse xenograft tumors of NSCLC expressing CRMP2 phosphorylation mimetic mutants grew significantly less than wild-type tumors. Given the recent development of small molecule inhibitors of CRMP2 phosphorylation to treat neurodegenerative diseases, our results open the door for their use in cancer treatment.
Collapse
Affiliation(s)
- Xabier Morales
- Solid Tumors and Biomarkers Program, Center for Applied Medical Research, University of Navarra, Avda Pío XII, 55, 31008 Pamplona, Spain; (X.M.); (C.O.d.S.)
- Instituto de Investigación Sanitaria de Navarra (IdISNA), 31008 Pamplona, Spain;
| | - Rafael Peláez
- Center for Biomedical Research of La Rioja (CIBIR), Neurodegeneration Area, Biomarkers and Molecular Signaling Group, Piqueras 98, 26006 Logroño, Spain;
| | - Saray Garasa
- Instituto de Investigación Sanitaria de Navarra (IdISNA), 31008 Pamplona, Spain;
- Division of Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), University of Navarra, 31008 Pamplona, Spain
| | - Carlos Ortiz de Solórzano
- Solid Tumors and Biomarkers Program, Center for Applied Medical Research, University of Navarra, Avda Pío XII, 55, 31008 Pamplona, Spain; (X.M.); (C.O.d.S.)
- Instituto de Investigación Sanitaria de Navarra (IdISNA), 31008 Pamplona, Spain;
| | - Ana Rouzaut
- Instituto de Investigación Sanitaria de Navarra (IdISNA), 31008 Pamplona, Spain;
- Department Biochemistry and Genetics, University of Navarra, Irunlarrea 1, 31080 Pamplona, Spain
- Correspondence:
| |
Collapse
|
9
|
Clinical evidence that a dysregulated master neural network modulator may aid in diagnosing schizophrenia. Proc Natl Acad Sci U S A 2021; 118:2100032118. [PMID: 34330827 PMCID: PMC8346854 DOI: 10.1073/pnas.2100032118] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
There are no biomarkers for schizophrenia (SCZ), a disorder of dysfunctional neural networks. We demonstrate that a master regulator of cytoskeleton (“CRMP2”) and, hence, neural circuitry, may form the basis for such a biomarker because its activity is uniquely imbalanced in SCZ patients. We show that SCZ patients are characterized by an excess of active CRMP2 not only in their brains (where it is correlated with dendritic abnormalities) but also in their peripheral blood lymphocytes. The abundance of active CRMP2 and insufficiency of opposing inactive p-CRMP2 likely disrupts neuronal function. Because peripheral blood CRMP2 appears to reflect intracerebral processes, it could form the basis of a rapid, minimally invasive, sensitive, and specific clinical diagnostic aid for SCZ in young patients. There are no validated biomarkers for schizophrenia (SCZ), a disorder linked to neural network dysfunction. We demonstrate that collapsin response mediator protein-2 (CRMP2), a master regulator of cytoskeleton and, hence, neural circuitry, may form the basis for a biomarker because its activity is uniquely imbalanced in SCZ patients. CRMP2’s activity depends upon its phosphorylation state. While an equilibrium between inactive (phosphorylated) and active (nonphosphorylated) CRMP2 is present in unaffected individuals, we show that SCZ patients are characterized by excess active CRMP2. We examined CRMP2 levels first in postmortem brains (correlated with neuronal morphometrics) and then, because CRMP2 is expressed in lymphocytes as well, in the peripheral blood of SCZ patients versus age-matched unaffected controls. In the brains and, more starkly, in the lymphocytes of SCZ patients <40 y old, we observed that nonphosphorylated CRMP2 was higher than in controls, while phosphorylated CRMP2 remained unchanged from control. In the brain, these changes were associated with dendritic structural abnormalities. The abundance of active CRMP2 with insufficient opposing inactive p-CRMP2 yielded a unique lowering of the p-CRMP2:CRMP2 ratio in SCZ patients, implying a disruption in the normal equilibrium between active and inactive CRMP2. These clinical data suggest that measuring CRMP2 and p-CRMP2 in peripheral blood might reflect intracerebral processes and suggest a rapid, minimally invasive, sensitive, and specific adjunctive diagnostic aid for early SCZ: increased CRMP2 or a decreased p-CRMP2:CRMP2 ratio may help cinch the diagnosis in a newly presenting young patient suspected of SCZ (versus such mimics as mania in bipolar disorder, where the ratio is high).
Collapse
|
10
|
Mast N, Petrov AM, Prendergast E, Bederman I, Pikuleva IA. Brain Acetyl-CoA Production and Phosphorylation of Cytoskeletal Proteins Are Targets of CYP46A1 Activity Modulation and Altered Sterol Flux. Neurotherapeutics 2021; 18:2040-2060. [PMID: 34235635 PMCID: PMC8609074 DOI: 10.1007/s13311-021-01079-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/23/2021] [Indexed: 02/04/2023] Open
Abstract
Cholesterol and 24-hydroxycholesterol are the most abundant brain sterols and represent the substrate and product, respectively, of cytochrome P450 46A1 (CYP46A1), a CNS-specific enzyme. CYP46A1 controls cholesterol elimination and turnover in the brain, the two processes that determine the rate of brain sterol flux through the plasma membranes and thereby the properties of these membranes. Brain sterol flux is decreased in Cyp46a1-/- mice compared to wild-type mice and increased in 5XFAD mice (a model of Alzheimer's disease) when they are treated with a small dose of efavirenz, a CYP46A1 activator. Herein, we first assessed the brain proteome (synaptosomal fractions) and phospho-proteome (synaptosomal fractions and brain homogenates) of efavirenz-treated and control 5XFAD mice. Then, based on the pattern of protein abundance change, we conducted acetyl-CoA measurements (brain homogenates and mitochondria) and metabolic profiling (brain homogenates). The phospho-proteomics datasets were used for comparative analyses with the datasets obtained by us previously on mice with the same changes (efavirenz-treated and control 5XFAD mice from a different treatment paradigm) or with changes in the opposite direction (Cyp46a1-/- vs wild-type mice) in brain sterol flux. We found that CYP46A1 activity or the rate of brain sterol flux affects acetyl-CoA-related metabolic pathways as well as phosphorylation of cytoskeletal and other proteins. Knowledge of the key roles of acetyl-CoA and cytoskeletal phosphorylation in cell biology expands our understanding of the significance of CYP46A1-mediated cholesterol 24-hydroxylation in the brain and provides an additional explanation for why CYP46A1 activity modulations are beneficial in mouse models of different brain diseases.
Collapse
Affiliation(s)
- Natalia Mast
- Department of Ophthalmology and Visual Sciences, Case Western Reserve University, Cleveland, OH, USA
| | - Alexey M Petrov
- Department of Ophthalmology and Visual Sciences, Case Western Reserve University, Cleveland, OH, USA
- Laboratory of Biophysics of Synaptic Processes, Kazan Institute of Biochemistry and Biophysics, Federal Research Center, Kazan Scientific Center of RAS", 2/31 Lobachevsky Street, Box 30, 420111, Kazan, Russia
- Institute of Neuroscience, Kazan State Medial University, 49 Butlerova Street, 420012, Kazan, Russia
| | - Erin Prendergast
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH, USA
| | - Ilya Bederman
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH, USA
| | - Irina A Pikuleva
- Department of Ophthalmology and Visual Sciences, Case Western Reserve University, Cleveland, OH, USA.
| |
Collapse
|
11
|
Liu X, Zhang Y. Bioinformatics Analysis of Dysregulated MicroRNA-Messenger RNA Networks in Small Cell Lung Cancer. J BIOMATER TISS ENG 2021. [DOI: 10.1166/jbt.2021.2563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
The present study aimed to identify a key module of differentially expressed miRNAs (DE-miRNAs) together with the corresponding differentially expressed mRNAs (DE-mRNAs) within small cell lung cancer (SCLC). Linear models were applied to ascertain the DE-miRNAs and DE-mRNAs in SCLC
versus matched non-carcinoma samples obtained from the RNA expression datasets of GSE19945, GSE74190 and GSE6044. The common DE-miRNAs were identified using the Venn plot. Then, 3 databases were used to retrieve the DE-miRNAs target genes, and the intersection was taken for validating the
shared target genes. Besides, Cytoscape was utilized for constructing the miRNAmRNA network for SCLC. Finally, a key module of five DE-miRNAs and four hub genes was determined based on the degree. In addition, the Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses
were conducted for exploring those hub genes in terms of their functions along with the involved signal transduction pathways. Altogether 106 shared DE-miRNAs were identified, which were used to predict 63 common target genes. In addition, a key module of five DE-miRNAs (hsa-miR-17-5p, hsa-miR-20a-5p,
hsa-miR-20b-5p, hsa-miR-93-5p and hsa-miR- 106b-5p) and four hub genes (SOX4, DPYSL2, TGFBR2 and F3) were extracted from the miRNAmRNA network according to their degree. Finally, the hub genes were subjected to GO as well as KEGG analysis, which revealed that cell cycle G1/S phase transition,
the extracellular matrix, and cellular senescence signaling pathways exerted vial parts during SCLC progression. A key module of five DE-miRNAs and four hub genes may be potentially used as clinical biomarkers to predict SCLC.
Collapse
Affiliation(s)
- Xingsheng Liu
- Department of Thoracic Surgery, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Yi Zhang
- Department of Thoracic Surgery, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| |
Collapse
|
12
|
Sugeno A, Piao W, Yamazaki M, Takahashi K, Arikawa K, Matsunaga H, Hosokawa M, Tominaga D, Goshima Y, Takeyama H, Ohshima T. Cortical transcriptome analysis after spinal cord injury reveals the regenerative mechanism of central nervous system in CRMP2 knock-in mice. Neural Regen Res 2021; 16:1258-1265. [PMID: 33318403 PMCID: PMC8284262 DOI: 10.4103/1673-5374.301035] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
Recent studies have shown that mutation at Ser522 causes inhibition of collapsin response mediator protein 2 (CRMP2) phosphorylation and induces axon elongation and partial recovery of the lost sensorimotor function after spinal cord injury (SCI). We aimed to reveal the intracellular mechanism in axotomized neurons in the CRMP2 knock-in (CRMP2KI) mouse model by performing transcriptome analysis in mouse sensorimotor cortex using micro-dissection punching system. Prior to that, we analyzed the structural pathophysiology in axotomized or neighboring neurons after SCI and found that somatic atrophy and dendritic spine reduction in sensorimotor cortex were suppressed in CRMP2KI mice. Further analysis of the transcriptome has aided in the identification of four hemoglobin genes Hba-a1, Hba-a2, Hbb-bs, and Hbb-bt that are significantly upregulated in wild-type mice with concomitant upregulation of genes involved in the oxidative phosphorylation and ribosomal pathways after SCI. However, we observed substantial upregulation in channel activity genes and downregulation of genes regulating vesicles, synaptic function, glial cell differentiation in CRMP2KI mice. Moreover, the transcriptome profile of CRMP2KI mice has been discussed wherein energy metabolism and neuronal pathways were found to be differentially regulated. Our results showed that CRMP2KI mice displayed improved SCI pathophysiology not only via microtubule stabilization in neurons, but also possibly via the whole metabolic system in the central nervous system, response changes in glial cells, and synapses. Taken together, we reveal new insights on SCI pathophysiology and the regenerative mechanism of central nervous system by the inhibition of CRMP2 phosphorylation at Ser522. All these experiments were performed in accordance with the guidelines of the Institutional Animal Care and Use Committee at Waseda University, Japan (2017-A027 approved on March 21, 2017; 2018-A003 approved on March 25, 2018; 2019-A026 approved on March 25, 2019).
Collapse
Affiliation(s)
- Ayaka Sugeno
- Laboratory for Molecular Brain Science, Department of Life Science and Medical Bioscience, Graduate School of Advanced Science and Engineering, Waseda University; Computational Bio Big-Data Open Innovation Laboratory (CBBD-OIL), National Institute of Advanced Industrial Science and Technology (AIST), Tokyo, Japan
| | - Wenhui Piao
- Laboratory for Molecular Brain Science, Department of Life Science and Medical Bioscience, Graduate School of Advanced Science and Engineering, Waseda University, Tokyo, Japan
| | - Miki Yamazaki
- Biomolecular Engineering Laboratory, Department of Life Science and Medical Bioscience, Graduate School of Advanced Science and Engineering, Waseda University; Computational Bio Big-Data Open Innovation Laboratory (CBBD-OIL), National Institute of Advanced Industrial Science and Technology (AIST), Tokyo, Japan
| | - Kiyofumi Takahashi
- Research Organization for Nano and Life Innovation, Waseda University, Tokyo, Japan
| | - Koji Arikawa
- Research Organization for Nano and Life Innovation, Waseda University, Tokyo, Japan
| | - Hiroko Matsunaga
- Research Organization for Nano and Life Innovation, Waseda University, Tokyo, Japan
| | - Masahito Hosokawa
- Biomolecular Engineering Laboratory, Department of Life Science and Medical Bioscience, Graduate School of Advanced Science and Engineering; Research Organization for Nano and Life Innovation; Institute for Advanced Research of Biosystem Dynamics, Waseda Research Institute for Science and Engineering, Graduate School of Advanced Science and Engineering, Waseda University, Tokyo, Japan
| | - Daisuke Tominaga
- Computational Bio Big-Data Open Innovation Laboratory (CBBD-OIL), National Institute of Advanced Industrial Science and Technology (AIST), Tokyo; Cellular and Molecular Biotechnology Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba, Japan
| | - Yoshio Goshima
- Department of Molecular Pharmacology and Neurobiology, Graduate School of Medicine, Yokohama City University, Yokohama, Japan
| | - Haruko Takeyama
- Biomolecular Engineering Laboratory, Department of Life Science and Medical Bioscience, Graduate School of Advanced Science and Engineering, Waseda University; Computational Bio Big-Data Open Innovation Laboratory (CBBD-OIL), National Institute of Advanced Industrial Science and Technology (AIST); Research Organization for Nano and Life Innovation; Institute for Advanced Research of Biosystem Dynamics, Waseda Research Institute for Science and Engineering, Graduate School of Advanced Science and Engineering, Waseda University, Tokyo, Japan
| | - Toshio Ohshima
- Laboratory for Molecular Brain Science, Department of Life Science and Medical Bioscience; Institute for Advanced Research of Biosystem Dynamics, Waseda Research Institute for Science and Engineering, Graduate School of Advanced Science and Engineering, Waseda University, Tokyo, Japan
| |
Collapse
|
13
|
Khanna R, Moutal A, Perez-Miller S, Chefdeville A, Boinon L, Patek M. Druggability of CRMP2 for Neurodegenerative Diseases. ACS Chem Neurosci 2020; 11:2492-2505. [PMID: 32693579 DOI: 10.1021/acschemneuro.0c00307] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Collapsin response mediator proteins (CRMPs) are ubiquitously expressed phosphoproteins that coordinate cytoskeletal formation and regulate cellular division, migration, polarity, and synaptic connection. CRMP2, the most studied of the five family members, is best known for its affinity for tubulin heterodimers and function in regulating the microtubule network. Accumulating evidence has also demonstrated a key role for CRMP2 in trafficking of voltage- and ligand-gated ion channels. These functions are tightly regulated by post-translational modifications including phosphorylation and SUMOylation (addition of a small ubiquitin like modifier). Over the past decade, it has become increasingly clear that dysregulated post-translational modifications of CRMP2 contribute to the pathomechanisms of diverse diseases, including cancer, neurodegenerative diseases, chronic pain, and bipolar disorder. Here, we review the discovery, functions, and current putative preclinical and clinical therapeutics targeting CRMP2. These potential therapeutics include CRMP2-based peptides that inhibit protein-protein interactions and small-molecule compounds. Capitalizing on the availability of structural information, we identify druggable pockets on CRMP2 and predict binding modes for five known CRMP2-targeting compounds, setting the stage for optimization and de novo drug discovery targeting this multifunctional protein.
Collapse
Affiliation(s)
- Rajesh Khanna
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, Arizona 85724, United States
- Graduate Interdisciplinary Program in Neuroscience, College of Medicine, University of Arizona, Tucson, Arizona 85724, United States
- The Center for Innovation in Brain Sciences, The University of Arizona Health Sciences, Tucson, Arizona 85724, United States
- Regulonix LLC, Tucson, Arizona 85718, United States
| | - Aubin Moutal
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, Arizona 85724, United States
| | - Samantha Perez-Miller
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, Arizona 85724, United States
| | - Aude Chefdeville
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, Arizona 85724, United States
| | - Lisa Boinon
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, Arizona 85724, United States
| | - Marcel Patek
- BrightRock Path, LLC, Tucson, Arizona 85704, United States
| |
Collapse
|
14
|
Stratton H, Boinon L, Moutal A, Khanna R. Coordinating Synaptic Signaling with CRMP2. Int J Biochem Cell Biol 2020; 124:105759. [PMID: 32437854 DOI: 10.1016/j.biocel.2020.105759] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 03/27/2020] [Accepted: 04/21/2020] [Indexed: 10/24/2022]
Abstract
Synaptic transmission is a complex process, dysregulation of which underlies several neurological conditions. Collapsin response mediator protein 2 (CRMP2) is a microtubule associated protein expressed ubiquitously in the central nervous system. Identified initially in the context of Semaphorin 3A (Collapsin) induced growth cone collapse, more recent findings revealed the involvement of CRMP2 in ion channel trafficking, kinesin-dependent axonal transport and maintenance of intracellular calcium homeostasis. CRMP2 is a synaptic protein, expressed at pre- and post-synaptic sites. Interactions with proteins such as N-methyl-D-aspartate receptors, syntaxin1A as well as voltage-gated calcium and sodium channels, suggest that CRMP2 may control both the electrical and chemical components of synaptic transmission. This short review will outline the known synaptic interactions of CRMP2 and illustrate its role in synaptic transmission, thereby introducing CRMP2 as a prospective target for the pathophysiological modulation of aberrant synaptic activity.
Collapse
Affiliation(s)
- Harrison Stratton
- Department of Pharmacology, College of Medicine, University of Arizona, United States
| | - Lisa Boinon
- Department of Pharmacology, College of Medicine, University of Arizona, United States
| | - Aubin Moutal
- Department of Pharmacology, College of Medicine, University of Arizona, United States
| | - Rajesh Khanna
- Department of Pharmacology, College of Medicine, University of Arizona, United States; BIO5 Institute, University of Arizona, United States; The Center for Innovation in Brain Sciences, The University of Arizona Health Sciences, Tucson, Arizona, United States.
| |
Collapse
|
15
|
Cai S, Shan Z, Zhang Z, Moutal A, Khanna R. Activity of T-type calcium channels is independent of CRMP2 in sensory neurons. Channels (Austin) 2020; 13:147-152. [PMID: 31025580 PMCID: PMC6527066 DOI: 10.1080/19336950.2019.1608129] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Amongst the regulators of voltage-gated ion channels is the collapsin response mediator protein 2 (CRMP2). CRMP2 regulation of the activity and trafficking of NaV1.7 voltage-gated sodium channels as well as the N-type (CaV2.2) voltage-gated calcium channel (VGCC) has been reported. On the other hand, CRMP2 does not appear to regulate L- (CaV1.x), P/Q- (CaV2.1), and R- (CaV2.3) type high VGCCs. Whether CRMP2 regulates low VGCCs remains an open question. Here, we asked if CRMP2 could regulate the low voltage-gated (T-type/CaV3.x) channels in sensory neurons. Reducing CRMP2 protein levels with short interfering RNAs yielded no change in macroscopic currents carried by T-type channels. No change in biophysical properties of the T-type currents was noted. Future studies pursuing CRMP2 druggability in neuropathic pain will benefit from the findings that CRMP2 regulates only the N-type (CaV2.2) calcium channels.
Collapse
Affiliation(s)
- Song Cai
- a Department of Pharmacology, College of Medicine , The University of Arizona Health Sciences , Tucson , AZ , USA
| | - Zhiming Shan
- a Department of Pharmacology, College of Medicine , The University of Arizona Health Sciences , Tucson , AZ , USA.,b Department of Anesthesiology , Shenzhen People's Hospital & Second Clinical Medical College of Jinan University , Shenzhen , P.R. China
| | - Zhongjun Zhang
- b Department of Anesthesiology , Shenzhen People's Hospital & Second Clinical Medical College of Jinan University , Shenzhen , P.R. China
| | - Aubin Moutal
- a Department of Pharmacology, College of Medicine , The University of Arizona Health Sciences , Tucson , AZ , USA
| | - Rajesh Khanna
- a Department of Pharmacology, College of Medicine , The University of Arizona Health Sciences , Tucson , AZ , USA.,c The Center for Innovation in Brain Sciences , The University of Arizona Health Sciences , Tucson , AZ , USA
| |
Collapse
|
16
|
Chang YH, Tsai JN, Chang SW, Hsu WT, Yang CP, Hsiao CW, Shiau MY. Regulation of Adipogenesis and Lipid Deposits by Collapsin Response Mediator Protein 2. Int J Mol Sci 2020; 21:ijms21062172. [PMID: 32245267 PMCID: PMC7139951 DOI: 10.3390/ijms21062172] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 03/17/2020] [Accepted: 03/19/2020] [Indexed: 12/17/2022] Open
Abstract
As emerging evidence suggesting neurodegenerative diseases and metabolic diseases have common pathogenesis, we hypothesized that the neurite outgrowth-controlling collapsin response mediator protein 2 (CRMP2) was involved in energy homeostasis. Therefore, putative roles of CRMP2 in adipocyte differentiation (adipogenesis) and lipid metabolism were explored and addressed in this study. CRMP2 expression profiles were in vitro and in vivo characterized during adipogenic process of 3T3-L1 pre-adipocytes and diet-induced obese (DIO) mice, respectively. Effects of CRMP2 on lipid metabolism and deposits were also analyzed. Our data revealed that CRMP2 expression pattern was coupled with adipogenic stages. CRMP2 overexpression inhibited cell proliferation at MCE phase, and significantly reduced lipid contents by down-regulating adipogenesis-driving transcription factors and lipid-synthesizing enzymes. Interestingly, GLUT4 translocation and the lipid droplets fusion were disturbed in CRMP2-silencing cells by affecting actin polymerization. Moreover, adipose CRMP2 was significantly increased in DIO mice, indicating CRMP2 is associated with obesity. Accordingly, CRMP2 exerts multiple functions in adipogenesis and lipid deposits through mediating cell proliferation, glucose/lipid metabolism and cytoskeleton dynamics. The present study identifies novel roles of CRMP2 in mediating adipogenesis and possible implication in metabolic disorders, as well as provides molecular evidence supporting the link of pathogenesis between neurodegenerative diseases and metabolic abnormalities.
Collapse
Affiliation(s)
- Yih-Hsin Chang
- Department of Biotechnology and Laboratory Science in Medicine, National Yang-Ming University, Taipei 112, Taiwan; (Y.-H.C.); (S.-W.C.); (W.-T.H.); (C.-P.Y.); (C.-W.H.)
| | - Jen-Ning Tsai
- Department of Medical Laboratory and Biotechnology, Chung Shan Medical University, Taichung 402, Taiwan;
- Clinical Laboratory, Chung Shan Medical University Hospital, Taichung 402, Taiwan
| | - Shu-Wen Chang
- Department of Biotechnology and Laboratory Science in Medicine, National Yang-Ming University, Taipei 112, Taiwan; (Y.-H.C.); (S.-W.C.); (W.-T.H.); (C.-P.Y.); (C.-W.H.)
| | - Wei-Ting Hsu
- Department of Biotechnology and Laboratory Science in Medicine, National Yang-Ming University, Taipei 112, Taiwan; (Y.-H.C.); (S.-W.C.); (W.-T.H.); (C.-P.Y.); (C.-W.H.)
| | - Ching-Ping Yang
- Department of Biotechnology and Laboratory Science in Medicine, National Yang-Ming University, Taipei 112, Taiwan; (Y.-H.C.); (S.-W.C.); (W.-T.H.); (C.-P.Y.); (C.-W.H.)
| | - Chiao-Wan Hsiao
- Department of Biotechnology and Laboratory Science in Medicine, National Yang-Ming University, Taipei 112, Taiwan; (Y.-H.C.); (S.-W.C.); (W.-T.H.); (C.-P.Y.); (C.-W.H.)
- Program in Molecular Medicine, National Yang-Ming University and Academia Sinica, Taipei 112, Taiwan
| | - Ming-Yuh Shiau
- Department of Nursing, College of Nursing, Hungkuang University, Taichung 433, Taiwan
- Correspondence: or ; Tel.: +886-4-26318652 (ext. 7090); Fax: +886-4-26331198
| |
Collapse
|
17
|
Xiang D, Xiao J, Sun S, Fu L, Yao L, Wang G, Liu Z. Differential Regulation of DNA Methylation at the CRMP2 Promoter Region Between the Hippocampus and Prefrontal Cortex in a CUMS Depression Model. Front Psychiatry 2020; 11:141. [PMID: 32256396 PMCID: PMC7093734 DOI: 10.3389/fpsyt.2020.00141] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Accepted: 02/14/2020] [Indexed: 12/21/2022] Open
Abstract
Current evidence supports the idea that neural plasticity is a potential cause of depression. Abundant studies indicate that CRMP2 has important roles in neural plasticity. Moreover, CRMP2 may contribute to the etiology of depression. However, the regulatory mechanisms underlying the role of CRMP2 remain unclear. DNA methylation alteration is generally acknowledged to be involved in the development of depression. The aim of this study was to explore the relationship between the expression and DNA methylation of CRMP2 in the hippocampus and prefrontal cortex of a rat depression model. Chronic unpredictable mild stress (CUMS) was used to establish a rat depression model, and body weight and behavioral tests were used to evaluate the effects of stress. Real-time PCR and Western blotting were used to test CRMP2 mRNA and protein expression, respectively, in the hippocampus and prefrontal cortex of rats. DNA methylation levels of the CRMP2 promoter were analyzed by bisulfite sequencing PCR (BSP). CUMS caused depressive-like behavior in rats, as evidenced by: decreased body weight and sucrose preference rate; decreases in the total distance traveled, rearing frequency, velocity, and duration in the center in the open field test (OFT); and prolonged immobility in the forced swimming test (FST). CRMP2 mRNA and protein expression in the hippocampus and prefrontal cortex were significantly decreased in the CUMS group compared with the control group. The levels of CRMP2 promoter DNA methylation in the hippocampus of the CUMS group were significantly higher than those of the control group, while these changes were not observed in the prefrontal cortex of CUMS rats. Our data provide evidence that altered expression of CRMP2 in the hippocampus and prefrontal cortex is associated with the pathogenesis of depression. Moreover, the results also suggest regional differences in the regulation of DNA methylation in the CRMP2 promoter between the hippocampus and prefrontal cortex during the development of depression.
Collapse
Affiliation(s)
- Dan Xiang
- Department of Psychiatry, Renmin Hospital of Wuhan University, Wuhan, China
| | - Jiawei Xiao
- Department of Psychiatry, Renmin Hospital of Wuhan University, Wuhan, China
| | - Siqi Sun
- Department of Psychiatry, Renmin Hospital of Wuhan University, Wuhan, China
| | - Linyan Fu
- Department of Psychiatry, Renmin Hospital of Wuhan University, Wuhan, China
| | - Lihua Yao
- Department of Psychiatry, Renmin Hospital of Wuhan University, Wuhan, China
| | - Gaohua Wang
- Department of Psychiatry, Renmin Hospital of Wuhan University, Wuhan, China.,Institute of Neuropsychiatry, Renmin Hospital, Wuhan University, Wuhan, China
| | - Zhongchun Liu
- Department of Psychiatry, Renmin Hospital of Wuhan University, Wuhan, China.,Institute of Neuropsychiatry, Renmin Hospital, Wuhan University, Wuhan, China
| |
Collapse
|
18
|
Landi C, Luddi A, Bianchi L, Pannuzzo G, Pavone V, Piomboni P, Bini L. Proteostasis network alteration in lysosomal storage disorders: Insights from the mouse model of Krabbe disease. J Neurosci Res 2019; 98:718-733. [PMID: 31797419 DOI: 10.1002/jnr.24558] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 10/09/2019] [Accepted: 10/29/2019] [Indexed: 12/19/2022]
Abstract
In Krabbe disease, a mutation in GALC gene causes widespread demyelination determining cell death by apoptosis, mainly in oligodendrocytes and Schwann cells. Less is known on the molecular mechanisms induced by this deficiency. Here, we report an impairment in protein synthesis and degradation and in proteasomal clearance with a potential accumulation of the misfolded proteins and induction of the endoplasmic reticulum stress in the brain of 6-day-old twitcher mice (TM) (model of Krabbe disease). In particular, an imbalance of the immunoproteasome function was highlighted, useful for shaping adaptive immune response by neurological cells. Moreover, our data show an involvement of cytoskeleton remodeling in Krabbe pathogenesis, with a lamin meshwork disaggregation in twitcher oligodendrocytes in 6-day-old TM. This study provides interesting protein targets and mechanistic insight on the early onset of Krabbe disease that may be promising options to be tested in combination with currently available therapies to rescue Krabbe phenotype.
Collapse
Affiliation(s)
- Claudia Landi
- Department of Life Sciences, University of Siena, Siena, Italy
| | - Alice Luddi
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | - Laura Bianchi
- Department of Life Sciences, University of Siena, Siena, Italy
| | - Giovanna Pannuzzo
- Department of Biomedical and Biotechnological Sciences, Section of Physiology, University of Catania, Catania, Italy
| | - Valentina Pavone
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | - Paola Piomboni
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | - Luca Bini
- Department of Life Sciences, University of Siena, Siena, Italy
| |
Collapse
|
19
|
Guo K, Elzinga S, Eid S, Figueroa-Romero C, Hinder LM, Pacut C, Feldman EL, Hur J. Genome-wide DNA methylation profiling of human diabetic peripheral neuropathy in subjects with type 2 diabetes mellitus. Epigenetics 2019; 14:766-779. [PMID: 31132961 PMCID: PMC6615525 DOI: 10.1080/15592294.2019.1615352] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
DNA methylation is an epigenetic mechanism important for the regulation of gene expression, which plays a vital role in the interaction between genetic and environmental factors. Aberrant epigenetic changes are implicated in the pathogenesis of diabetes and diabetic complications, but the role of DNA methylation in diabetic peripheral neuropathy (DPN) is not well understood. Therefore, our aim in this study was to explore the role of DNA methylation in the progression of DPN in type 2 diabetes. We compared genome-wide DNA methylation profiles of human sural nerve biopsies from subjects with stable or improving nerve fibre counts to biopsies from subjects with progressive loss of nerve fibres. Nerve fibre counts were determined by comparing myelinated nerve fibre densities between an initial and repeat biopsy separated by 52 weeks. Subjects with significant nerve regeneration (regenerators) and subjects with significant nerve degeneration (degenerators) represent the two extreme DPN phenotypes. Using reduced representation bisulfite sequencing, we identified 3,460 differentially methylated CpG dinucleotides between the two groups. The genes associated with differentially methylated CpGs were highly enriched in biological processes that have previously been implicated in DPN such as nervous system development, neuron development, and axon guidance, as well as glycerophospholipid metabolism and mitogen-activated protein kinase (MAPK) signalling. These findings are the first to provide a comprehensive analysis of DNA methylation profiling in human sural nerves of subjects with DPN and suggest that epigenetic regulation has an important role in the progression of this prevalent diabetic complication.
Collapse
Affiliation(s)
- Kai Guo
- a Department of Biomedical Sciences, School of Medicine and Health Sciences , University of North Dakota , Grand Forks , ND , USA
| | - Sarah Elzinga
- b Department of Neurology, School of Medicine , University of Michigan , Ann Arbor , MI , USA
| | - Stephanie Eid
- b Department of Neurology, School of Medicine , University of Michigan , Ann Arbor , MI , USA
| | - Claudia Figueroa-Romero
- b Department of Neurology, School of Medicine , University of Michigan , Ann Arbor , MI , USA
| | - Lucy M Hinder
- b Department of Neurology, School of Medicine , University of Michigan , Ann Arbor , MI , USA
| | - Crystal Pacut
- b Department of Neurology, School of Medicine , University of Michigan , Ann Arbor , MI , USA
| | - Eva L Feldman
- b Department of Neurology, School of Medicine , University of Michigan , Ann Arbor , MI , USA
| | - Junguk Hur
- a Department of Biomedical Sciences, School of Medicine and Health Sciences , University of North Dakota , Grand Forks , ND , USA
| |
Collapse
|
20
|
Ohtani-Kaneko R. Crmp4-KO Mice as an Animal Model for Investigating Certain Phenotypes of Autism Spectrum Disorders. Int J Mol Sci 2019; 20:E2485. [PMID: 31137494 PMCID: PMC6566569 DOI: 10.3390/ijms20102485] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Revised: 05/17/2019] [Accepted: 05/18/2019] [Indexed: 12/21/2022] Open
Abstract
Previous research has demonstrated that the collapsin response mediator protein (CRMP) family is involved in the formation of neural networks. A recent whole-exome sequencing study identified a de novo variant (S541Y) of collapsin response mediator protein 4 (CRMP4) in a male patient with autism spectrum disorder (ASD). In addition, Crmp4-knockout (KO) mice show some phenotypes similar to those observed in human patients with ASD. For example, compared with wild-type mice, Crmp4-KO mice exhibit impaired social interaction, abnormal sensory sensitivities, broader distribution of activated (c-Fos expressing) neurons, altered dendritic formation, and aberrant patterns of neural gene expressions, most of which have sex differences. This review summarizes current knowledge regarding the role of CRMP4 during brain development and discusses the possible contribution of CRMP4 deficiencies or abnormalities to the pathogenesis of ASD. Crmp4-KO mice represent an appropriate animal model for investigating the mechanisms underlying some ASD phenotypes, such as impaired social behavior, abnormal sensory sensitivities, and sex-based differences, and other neurodevelopmental disorders associated with sensory processing disorders.
Collapse
Affiliation(s)
- Ritsuko Ohtani-Kaneko
- Graduate School of Life Sciences, Toyo University, 1-1-1 Itakura, Oura 374-0193, Japan.
| |
Collapse
|
21
|
Limiting Neuronal Nogo Receptor 1 Signaling during Experimental Autoimmune Encephalomyelitis Preserves Axonal Transport and Abrogates Inflammatory Demyelination. J Neurosci 2019; 39:5562-5580. [PMID: 31061088 DOI: 10.1523/jneurosci.1760-18.2019] [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: 07/12/2018] [Revised: 04/05/2019] [Accepted: 04/29/2019] [Indexed: 11/21/2022] Open
Abstract
We previously identified that ngr1 allele deletion limits the severity of experimental autoimmune encephalomyelitis (EAE) by preserving axonal integrity. However, whether this favorable outcome observed in EAE is a consequence of an abrogated neuronal-specific pathophysiological mechanism, is yet to be defined. Here we show that, Cre-loxP-mediated neuron-specific deletion of ngr1 preserved axonal integrity, whereas its re-expression in ngr1-/- female mice potentiated EAE-axonopathy. As a corollary, myelin integrity was preserved under Cre deletion in ngr1flx/flx , retinal ganglion cell axons whereas, significant demyelination occurred in the ngr1-/- optic nerves following the re-introduction of NgR1. Moreover, Cre-loxP-mediated axon-specific deletion of ngr1 in ngr1flx/flx mice also demonstrated efficient anterograde transport of fluorescently-labeled ChTxβ in the optic nerves of EAE-induced mice. However, the anterograde transport of ChTxβ displayed accumulation in optic nerve degenerative axons of EAE-induced ngr1-/- mice, when NgR1 was reintroduced but was shown to be transported efficiently in the contralateral non- recombinant adeno-associated virus serotype 2-transduced optic nerves of these mutant mice. We further identified that the interaction between the axonal motor protein, Kinesin-1 and collapsin response mediator protein 2 (CRMP2) was unchanged upon Cre deletion of ngr1 Whereas, this Kinesin-1/CRMP2 association was reduced when NgR1 was re-expressed in the ngr1-/- optic nerves. Our data suggest that NgR1 governs axonal degeneration in the context of inflammatory-mediated demyelination through the phosphorylation of CRMP2 by stalling axonal vesicular transport. Moreover, axon-specific deletion of ngr1 preserves axonal transport mechanisms, blunting the induction of inflammatory demyelination and limiting the severity of EAE.SIGNIFICANCE STATEMENT Multiple sclerosis (MS) is commonly induced by aberrant immune-mediated destruction of the protective sheath of nerve fibers (known as myelin). However, it has been shown that MS lesions do not only consist of this disease pattern, exhibiting heterogeneity with continual destruction of axons. Here we investigate how neuronal NgR1 can drive inflammatory-mediated axonal degeneration and demyelination within the optic nerve by analyzing its downstream signaling events that govern axonal vesicular transport. We identify that abrogating the NgR1/pCRMP2 signaling cascade can maintain Kinesin-1-dependent anterograde axonal transport to limit inflammatory-mediated axonopathy and demyelination. The ability to differentiate between primary and secondary mechanisms of axonal degeneration may uncover therapeutic strategies to limit axonal damage and progressive MS.
Collapse
|
22
|
Faigle W, Cruciani C, Wolski W, Roschitzki B, Puthenparampil M, Tomas-Ojer P, Sellés-Moreno C, Zeis T, Jelcic I, Schaeren-Wiemers N, Sospedra M, Martin R. Brain Citrullination Patterns and T Cell Reactivity of Cerebrospinal Fluid-Derived CD4 + T Cells in Multiple Sclerosis. Front Immunol 2019; 10:540. [PMID: 31024521 PMCID: PMC6467957 DOI: 10.3389/fimmu.2019.00540] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Accepted: 02/27/2019] [Indexed: 01/11/2023] Open
Abstract
Immune responses to citrullinated peptides have been described in autoimmune diseases like rheumatoid arthritis (RA) and multiple sclerosis (MS). We investigated the post-translational modification (PTM), arginine to citrulline, in brain tissue of MS patients and controls (C) by proteomics and subsequently the cellular immune response of cerebrospinal fluid (CSF)-infiltrating T cells to citrullinated and unmodified peptides of myelin basic protein (MBP). Using specifically adapted tissue extraction- and combined data interpretation protocols we could establish a map of citrullinated proteins by identifying more than 80 proteins with two or more citrullinated peptides in human brain tissue. We report many of them for the first time. For the already described citrullinated proteins MBP, GFAP, and vimentin, we could identify additional citrullinated sites. The number of modified proteins in MS white matter was higher than control tissue. Citrullinated peptides are considered neoepitopes that may trigger autoreactivity. We used newly identified epitopes and previously reported immunodominant myelin peptides in their citrullinated and non-citrullinated form to address the recognition of CSF-infiltrating CD4+ T cells from 22 MS patients by measuring proliferation and IFN-γ secretion. We did not detect marked responses to citrullinated peptides, but slightly more strongly to the non-modified version. Based on these data, we conclude that citrullination does not appear to be an important activating factor of a T cell response, but could be the consequence of an immune- or inflammatory response. Our approach allowed us to perform a deep proteome analysis and opens new technical possibilities to analyze complex PTM patterns on minute quantities of rare tissue samples.
Collapse
Affiliation(s)
- Wolfgang Faigle
- Neuroimmunology and MS Research Section, Neurology Clinic, University Zurich, University Hospital Zurich, Zurich, Switzerland
| | - Carolina Cruciani
- Neuroimmunology and MS Research Section, Neurology Clinic, University Zurich, University Hospital Zurich, Zurich, Switzerland
| | - Witold Wolski
- Functional Genomics Center Zurich, ETH Zurich & University of Zurich, Zurich, Switzerland
| | - Bernd Roschitzki
- Functional Genomics Center Zurich, ETH Zurich & University of Zurich, Zurich, Switzerland
| | - Marco Puthenparampil
- Neuroimmunology and MS Research Section, Neurology Clinic, University Zurich, University Hospital Zurich, Zurich, Switzerland
| | - Paula Tomas-Ojer
- Neuroimmunology and MS Research Section, Neurology Clinic, University Zurich, University Hospital Zurich, Zurich, Switzerland
| | - Carla Sellés-Moreno
- Neuroimmunology and MS Research Section, Neurology Clinic, University Zurich, University Hospital Zurich, Zurich, Switzerland
| | - Thomas Zeis
- Neurobiology, Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Ivan Jelcic
- Neuroimmunology and MS Research Section, Neurology Clinic, University Zurich, University Hospital Zurich, Zurich, Switzerland
| | - Nicole Schaeren-Wiemers
- Neurobiology, Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Mireia Sospedra
- Neuroimmunology and MS Research Section, Neurology Clinic, University Zurich, University Hospital Zurich, Zurich, Switzerland
| | - Roland Martin
- Neuroimmunology and MS Research Section, Neurology Clinic, University Zurich, University Hospital Zurich, Zurich, Switzerland
| |
Collapse
|
23
|
Ramos-Miguel A, Barakauskas V, Alamri J, Miyauchi M, Barr AM, Beasley CL, Rosoklija G, Mann JJ, Dwork AJ, Moradian A, Morin GB, Honer WG. The SNAP25 Interactome in Ventromedial Caudate in Schizophrenia Includes the Mitochondrial Protein ARF1. Neuroscience 2019; 420:97-111. [PMID: 30610939 DOI: 10.1016/j.neuroscience.2018.12.045] [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: 08/28/2018] [Revised: 12/21/2018] [Accepted: 12/24/2018] [Indexed: 12/11/2022]
Abstract
Abnormalities of SNAP25 (synaptosome-associated protein 25) amount and protein-protein interactions occur in schizophrenia, and may contribute to abnormalities of neurotransmitter release in patients. However, presynaptic terminal function depends on multiple subcellular mechanisms, including energy provided by mitochondria. To explore the SNAP25 interactome in schizophrenia, we immunoprecipitated SNAP25 along with interacting proteins from the ventromedial caudate of 15 cases of schizophrenia and 13 controls. Proteins were identified with mass spectrometry-based proteomics. As well as 15 SNARE- (soluble N-ethylmaleimide-sensitive factor attachment protein receptor) associated proteins, we identified 17 mitochondria-associated and four other proteins. The mitochondrial small GTPase ARF1 (ADP-ribosylation factor 1) was identified in eight schizophrenia SNAP25 immunoprecipitates and none from controls (P = 0.004). Although the ARF1-SNAP25 interaction may be increased, immunoblotting demonstrated 21% lower ARF1-21 (21 kiloDaltons) in schizophrenia samples (P = 0.04). In contrast, the mitochondrial protein UQCRC1 (ubiquinol-cytochrome c reductase core protein 1) did not differ. Lower ARF1-21 levels were associated with the previously reported increased SNAP25-syntaxin interaction in schizophrenia (r = -0.39, P = 0.04). Additional immunoprecipitation studies confirmed the ARF1-21-SNAP25 interaction, independent of UQCRC1. Both ARF1 and SNAP25 were localized to synaptosomes. Confocal microscopy demonstrated co-localization of ARF1 and SNAP25, and further suggested fivefold enrichment of ARF1 in synaptosomes containing an excitatory marker (vesicular glutamate transporter) compared with synaptosomes containing an inhibitory marker (vesicular GABA transporter). The present findings suggest an association between abnormalities of SNARE proteins involved with vesicular neurotransmission and the mitochondrial protein ARF1 that may contribute to the pathophysiology of schizophrenia.
Collapse
Affiliation(s)
- Alfredo Ramos-Miguel
- BC Mental Health and Addictions Research Institute, 938 West 28th Ave, Vancouver, BC V5Z 4H4, Canada; Department of Psychiatry, University of British Columbia, 2255 Wesbrook Mall, Vancouver, BC V6T 2A1, Canada; Department of Pharmacology, University of the Basque Country, Centro de Investigación Biomédica en Red de Salud Mental, CIBERSAM, Barrio Sarriena, s/n, 48940 Leioa, Biscay, Spain
| | - Vilte Barakauskas
- BC Mental Health and Addictions Research Institute, 938 West 28th Ave, Vancouver, BC V5Z 4H4, Canada; Department of Pathology and Laboratory Medicine, University of British Columbia, 2J9-4500 Oak St., Vancouver, BC V6H 3B1, Canada
| | - Jehan Alamri
- BC Mental Health and Addictions Research Institute, 938 West 28th Ave, Vancouver, BC V5Z 4H4, Canada
| | - Masatoshi Miyauchi
- BC Mental Health and Addictions Research Institute, 938 West 28th Ave, Vancouver, BC V5Z 4H4, Canada; Department of Psychiatry, University of British Columbia, 2255 Wesbrook Mall, Vancouver, BC V6T 2A1, Canada
| | - Alasdair M Barr
- BC Mental Health and Addictions Research Institute, 938 West 28th Ave, Vancouver, BC V5Z 4H4, Canada; Department of Anesthesiology, Pharmacology, & Therapeutics, University of British Columbia, 2176 Health Sciences Mall Vancouver, BC V6T 1Z3, Canada
| | - Clare L Beasley
- BC Mental Health and Addictions Research Institute, 938 West 28th Ave, Vancouver, BC V5Z 4H4, Canada; Department of Psychiatry, University of British Columbia, 2255 Wesbrook Mall, Vancouver, BC V6T 2A1, Canada
| | - Gorazd Rosoklija
- Department of Psychiatry, Columbia University, 1051 Riverside Drive, New York, NY 10032, USA
| | - J John Mann
- Department of Psychiatry, Columbia University, 1051 Riverside Drive, New York, NY 10032, USA
| | - Andrew J Dwork
- Department of Psychiatry, Columbia University, 1051 Riverside Drive, New York, NY 10032, USA
| | - Annie Moradian
- Department of Medical Genetics, University of British Columbia, C234-4500 Oak St., Vancouver, BC V6H 3B1, Canada
| | - Gregg B Morin
- Department of Medical Genetics, University of British Columbia, C234-4500 Oak St., Vancouver, BC V6H 3B1, Canada
| | - William G Honer
- BC Mental Health and Addictions Research Institute, 938 West 28th Ave, Vancouver, BC V5Z 4H4, Canada; Department of Psychiatry, University of British Columbia, 2255 Wesbrook Mall, Vancouver, BC V6T 2A1, Canada.
| |
Collapse
|
24
|
Park DI, Turck CW. Interactome Studies of Psychiatric Disorders. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1118:163-173. [PMID: 30747422 DOI: 10.1007/978-3-030-05542-4_8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
High comorbidity and complexity have precluded reliable diagnostic assessment and treatment of psychiatric disorders. Impaired molecular interactions may be relevant for underlying mechanisms of psychiatric disorders but by and large remain unknown. With the help of a number of publicly available databases and various technological tools, recent research has filled the paucity of information by generating a novel dataset of psychiatric interactomes. Different technological platforms including yeast two-hybrid screen, co-immunoprecipitation-coupled with mass spectrometry-based proteomics, and transcriptomics have been widely used in combination with cellular and molecular techniques to interrogate the psychiatric interactome. Novel molecular interactions have been identified in association with different psychiatric disorders including autism spectrum disorders, schizophrenia, bipolar disorder, and major depressive disorder. However, more extensive and sophisticated interactome research needs to be conducted to overcome the current limitations such as incomplete interactome databases and a lack of functional information among components. Ultimately, integrated psychiatric interactome databases will contribute to the implementation of biomarkers and therapeutic intervention.
Collapse
Affiliation(s)
- Dong Ik Park
- Danish Research Institute of Translational Neuroscience (DANDRITE), Department of Biomedicine, Aarhus University, Aarhus, Denmark.
| | - Christoph W Turck
- Proteomics and Biomarkers, Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, Munich, Germany
| |
Collapse
|
25
|
Sarakul M, Elzo MA, Koonawootrittriron S, Suwanasopee T, Jattawa D, Laodim T. Characterization of biological pathways associated with semen traits in the Thai multibreed dairy population. Anim Reprod Sci 2018; 197:324-334. [PMID: 30213568 DOI: 10.1016/j.anireprosci.2018.09.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Revised: 08/28/2018] [Accepted: 09/04/2018] [Indexed: 10/28/2022]
Abstract
The objective of this research was to characterize biological pathways associated with semen volume (VOL), number of sperm (NS), and sperm motility (MOT) of dairy bulls in the Thai multibreed dairy population. Phenotypes for VOL (n = 13,535), NS (n = 12,773), and MOT (n = 12,660) came from 131 bulls of the Dairy Farming Promotion Organization of Thailand. Genotypic data consisted of 76,519 imputed and actual single nucleotide polymorphisms (SNP) from 72 animals. The SNP variances for VOL, NS, and MOT were estimated using a three-trait genomic-polygenic repeatability model. Fixed effects were contemporary group, ejaculate order, age of bull, ambient temperature, and heterosis. Random effects were animal additive genetic, permanent environmental, and residual. Individual SNP explaining at least 0.001% of the total genetic variance for each trait were selected to identify associated genes in the NCBI database (UMD Bos taurus 3.1 assembly) using the R package Map2NCBI. A set of 1,999 NCBI genes associated with all three semen traits was utilized for the pathway analysis conducted with the ClueGO plugin of Cytoscape using information from the Kyoto Encyclopedia of Genes and Genomes database. The pathway analysis revealed seven significant biological pathways involving 127 genes that explained 1.04% of the genetic variance for VOL, NS, and MOT. These genes were known to affect cell structure, motility, migration, proliferation, differentiation, survival, apoptosis, signal transduction, oxytocin release, calcium channel, neural development, and immune system functions related to sperm morphology and physiology during spermatogenesis.
Collapse
Affiliation(s)
- Mattaneeya Sarakul
- Department of Animal Science, Kasetsart University, Bangkok, 10900, Thailand
| | - Mauricio A Elzo
- Department of Animal Sciences, University of Florida, Gainesville, FL, 32611-0910, USA
| | | | | | - Danai Jattawa
- Department of Animal Science, Kasetsart University, Bangkok, 10900, Thailand
| | - Thawee Laodim
- Department of Animal Science, Kasetsart University, Bangkok, 10900, Thailand
| |
Collapse
|
26
|
Ryskalin L, Limanaqi F, Frati A, Busceti CL, Fornai F. mTOR-Related Brain Dysfunctions in Neuropsychiatric Disorders. Int J Mol Sci 2018; 19:ijms19082226. [PMID: 30061532 PMCID: PMC6121884 DOI: 10.3390/ijms19082226] [Citation(s) in RCA: 74] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Revised: 07/26/2018] [Accepted: 07/27/2018] [Indexed: 12/12/2022] Open
Abstract
The mammalian target of rapamycin (mTOR) is an ubiquitously expressed serine-threonine kinase, which senses and integrates several intracellular and environmental cues to orchestrate major processes such as cell growth and metabolism. Altered mTOR signalling is associated with brain malformation and neurological disorders. Emerging evidence indicates that even subtle defects in the mTOR pathway may produce severe effects, which are evident as neurological and psychiatric disorders. On the other hand, administration of mTOR inhibitors may be beneficial for a variety of neuropsychiatric alterations encompassing neurodegeneration, brain tumors, brain ischemia, epilepsy, autism, mood disorders, drugs of abuse, and schizophrenia. mTOR has been widely implicated in synaptic plasticity and autophagy activation. This review addresses the role of mTOR-dependent autophagy dysfunction in a variety of neuropsychiatric disorders, to focus mainly on psychiatric syndromes including schizophrenia and drug addiction. For instance, amphetamines-induced addiction fairly overlaps with some neuropsychiatric disorders including neurodegeneration and schizophrenia. For this reason, in the present review, a special emphasis is placed on the role of mTOR on methamphetamine-induced brain alterations.
Collapse
Affiliation(s)
- Larisa Ryskalin
- Human Anatomy, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via Roma 55, 56126 Pisa, Italy.
| | - Fiona Limanaqi
- Human Anatomy, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via Roma 55, 56126 Pisa, Italy.
| | | | | | - Francesco Fornai
- Human Anatomy, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via Roma 55, 56126 Pisa, Italy.
- I.R.C.C.S. Neuromed, Via Atinense 18, 86077 Isernia, Italy.
| |
Collapse
|
27
|
Interaction of Recombinant Gallus gallus SEPT5 and Brain Proteins of H5N1-Avian Influenza Virus-Infected Chickens. Proteomes 2017; 5:proteomes5030023. [PMID: 28895884 PMCID: PMC5620540 DOI: 10.3390/proteomes5030023] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Revised: 09/05/2017] [Accepted: 09/07/2017] [Indexed: 11/16/2022] Open
Abstract
Septin forms a conserved family of cytoskeletal guanosine triphosphate (GTP) binding proteins that have diverse roles in protein scaffolding, vesicle trafficking, and cytokinesis. The involvement of septins in infectious viral disease pathogenesis has been demonstrated by the upregulation of SEPT5 protein and its mRNA in brain tissues of H5N1-infected chickens, thus, providing evidence for the potential importance of this protein in the pathogenesis of neurovirulence caused by the avian influenza virus. In this study, cloning, expression, and purification of Gallus gallus SEPT5 protein was performed in Escherichia coli. The SEPT5 gene was inserted into the pRSETB expression vector, transformed in the E. coli BL21 (DE3) strain and the expression of SEPT5 protein was induced by IPTG. The SEPT5 protein was shown to be authentic as it was able to be pulled down by a commercial anti-SEPT5 antibody in a co-immunoprecipitation assay. In vivo aggregation of the recombinant protein was limited by cultivation at a reduced temperature of 16 °C. Using co-immunoprecipitation techniques, the purified recombinant SEPT5 protein was used to pull down host’s interacting or binding proteins, i.e., proteins of brains of chickens infected with the H5N1 influenza virus. Interacting proteins, such as CRMP2, tubulin proteins, heat-shock proteins and other classes of septins were identified using LCMS/MS. Results from this study suggest that the codon-optimized SEPT5 gene can be efficiently expressed in the E. coli bacterial system producing authentic SEPT5 protein, thus, enabling multiple host’s proteins to interact with the SEPT5 protein.
Collapse
|
28
|
Yu-Kemp HC, Kemp JP, Brieher WM. CRMP-1 enhances EVL-mediated actin elongation to build lamellipodia and the actin cortex. J Cell Biol 2017. [PMID: 28630144 PMCID: PMC5551698 DOI: 10.1083/jcb.201606084] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
CRMP proteins regulate the cytoskeleton, but the underlying mechanisms are poorly understood. Yu-Kemp et al. show that CRMP-1 helps Ena/VASP proteins elongate actin filaments to assemble actin networks that are necessary for the integrity of epithelial sheets. Cells can control actin polymerization by nucleating new filaments or elongating existing ones. We recently identified CRMP-1 as a factor that stimulates the formation of Listeria monocytogenes actin comet tails, thereby implicating it in actin assembly. We now show that CRMP-1 is a major contributor to actin assembly in epithelial cells, where it works with the Ena/VASP family member EVL to assemble the actin cytoskeleton in the apical cortex and in protruding lamellipodia. CRMP-1 and EVL bind to one another and together accelerate actin filament barbed-end elongation. CRMP-1 also stimulates actin assembly in the presence of VASP and Mena in vitro, but CRMP-1–dependent actin assembly in MDCK cells is EVL specific. Our results identify CRMP-1 as a novel regulator of actin filament elongation and reveal a surprisingly important role for CRMP-1, EVL, and actin polymerization in maintaining the structural integrity of epithelial sheets.
Collapse
Affiliation(s)
- Hui-Chia Yu-Kemp
- Department of Cell and Developmental Biology, University of Illinois, Urbana-Champaign, IL
| | - James P Kemp
- Department of Cell and Developmental Biology, University of Illinois, Urbana-Champaign, IL
| | - William M Brieher
- Department of Cell and Developmental Biology, University of Illinois, Urbana-Champaign, IL
| |
Collapse
|
29
|
Abstract
ASCIZ/ATMIN is not required for ATM activation by replication stress in MEFs. ATM activation is normal in human ASCIZ/ATMIN KO cells. ASCIZ/ATMIN is dispensable for aphidicolin-induced 53BP1 focus formation.
The ATM kinase plays critical roles in the response to DNA double-strand breaks, and can also be activated by prolonged DNA replication blocks. It has recently been proposed that replication stress-dependent ATM activation is mediated by ASCIZ (also known as ATMIN, ZNF822), an essential developmental transcription factor. In contrast, we show here that ATM activation, and phosphorylation of its substrates KAP1, p53 and H2AX in response to the replication blocking agent aphidicolin was unaffected in both immortalized and primary ASCIZ/ATMIN-deficient murine embryonic fibroblasts compared to control cells. Similar results were also obtained in human ASCIZ/ATMIN-deleted lymphoma cells. The results demonstrate that ASCIZ/ATMIN is dispensable for ATM activation, and contradict the previously reported dependence of ATM on ASCIZ/ATMIN.
Collapse
|
30
|
Smith BJ, Cassoli JS, Guest PC, Martins-de-Souza D. Co-immunoprecipitation for Deciphering Protein Interactomes. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 974:229-236. [DOI: 10.1007/978-3-319-52479-5_19] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
31
|
Kuter K, Kratochwil M, Marx SH, Hartwig S, Lehr S, Sugawa MD, Dencher NA. Native DIGE proteomic analysis of mitochondria from substantia nigra and striatum during neuronal degeneration and its compensation in an animal model of early Parkinson's disease. Arch Physiol Biochem 2016; 122:238-256. [PMID: 27467289 DOI: 10.1080/13813455.2016.1197948] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Cause of Parkinson's disease (PD) is still not understood. Motor symptoms are not observed at early stages of disease due to compensatory processes. Dysfunction of mitochondria was indicated already at preclinical PD. Selective toxin 6-OHDA was applied to kill dopaminergic neurons in substantia nigra and disturb neuronal transmission in striatum. Early phase of active degeneration and later stage, when surviving cells adapted to function normally, were analysed. 2D BN/SDS difference gel electrophoresis (DIGE) of mitochondrial proteome enabled to point out crucial processes involved at both time-points in dopaminergic structures. Marker proteins such as DPYSL2, HSP60, ATP1A3, EAAT2 indicated structural remodelling, cytoskeleton rearrangement, organelle trafficking, axon outgrowth and regeneration. Adaptations in dopaminergic and glutamatergic neurotransmission, recycling of synaptic vesicles, along with enlargement of mitochondria mass were proposed as causative for compensation. Changed expression of carbohydrates metabolism and oxidative phosphorylation proteins were described, including their protein-protein interactions and supercomplex assembly.
Collapse
Affiliation(s)
- Katarzyna Kuter
- a Department of Neuropsychopharmacology , Polish Academy of Sciences , Kraków , Poland
- b Physical Biochemistry, Department of Chemistry, Technische Universität Darmstadt , Darmstadt , Germany
| | - Manuela Kratochwil
- b Physical Biochemistry, Department of Chemistry, Technische Universität Darmstadt , Darmstadt , Germany
| | - Sven-Hendric Marx
- b Physical Biochemistry, Department of Chemistry, Technische Universität Darmstadt , Darmstadt , Germany
| | - Sonja Hartwig
- c Institute of Clinical Biochemistry and Pathobiochemistry, German Diabetes Center at the Heinrich-Heine-University Düsseldorf, Leibniz Center for Diabetes Research , Düsseldorf , Germany
- d German Center for Diabetes Research (DZD) , München, Neuherberg , Germany , and
| | - Stephan Lehr
- c Institute of Clinical Biochemistry and Pathobiochemistry, German Diabetes Center at the Heinrich-Heine-University Düsseldorf, Leibniz Center for Diabetes Research , Düsseldorf , Germany
- d German Center for Diabetes Research (DZD) , München, Neuherberg , Germany , and
| | - Michiru D Sugawa
- b Physical Biochemistry, Department of Chemistry, Technische Universität Darmstadt , Darmstadt , Germany
- e Clinical Neurobiology, Charité-Universitätsmedizin , Berlin , Germany
| | - Norbert A Dencher
- b Physical Biochemistry, Department of Chemistry, Technische Universität Darmstadt , Darmstadt , Germany
| |
Collapse
|
32
|
Pham X, Song G, Lao S, Goff L, Zhu H, Valle D, Avramopoulos D. The DPYSL2 gene connects mTOR and schizophrenia. Transl Psychiatry 2016; 6:e933. [PMID: 27801893 PMCID: PMC5314117 DOI: 10.1038/tp.2016.204] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Revised: 07/05/2016] [Accepted: 08/17/2016] [Indexed: 12/23/2022] Open
Abstract
We previously reported a schizophrenia-associated polymorphic CT di-nucleotide repeat (DNR) at the 5'-untranslated repeat (UTR) of DPYSL2, which responds to mammalian target of Rapamycin (mTOR) signaling with allelic differences in reporter assays. Now using microarray analysis, we show that the DNR alleles interact differentially with specific proteins, including the mTOR-related protein HuD/ELAVL4. We confirm the differential binding to HuD and other known mTOR effectors by electrophoretic mobility shift assays. We edit HEK293 cells by CRISPR/Cas9 to carry the schizophrenia risk variant (13DNR) and observe a significant reduction of the corresponding CRMP2 isoform. These edited cells confirm the response to mTOR inhibitors and show a twofold shortening of the cellular projections. Transcriptome analysis of these modified cells by RNA-seq shows changes in 12.7% of expressed transcripts at a false discovery rate of 0.05. These transcripts are enriched in immunity-related genes, overlap significantly with those modified by the schizophrenia-associated gene, ZNF804A, and have a reverse expression signature from that seen with antipsychotic drugs. Our results support the functional importance of the DPYSL2 DNR and a role for mTOR signaling in schizophrenia.
Collapse
Affiliation(s)
- X Pham
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA,Predoctoral Training Program in Human Genetics, McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - G Song
- Department of Pharmacology and Molecular Sciences, Baltimore, MD, USA
| | - S Lao
- University of Maryland, College Park, MD, USA
| | - L Goff
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - H Zhu
- Department of Pharmacology and Molecular Sciences, Baltimore, MD, USA
| | - D Valle
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA,Predoctoral Training Program in Human Genetics, McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - D Avramopoulos
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA,Predoctoral Training Program in Human Genetics, McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA,McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, MRB 512, 733N Wolfe Street Broadway, Baltimore, MD 21205, USA. E-mail:
| |
Collapse
|
33
|
Nascimento JM, Garcia S, Saia-Cereda VM, Santana AG, Brandao-Teles C, Zuccoli GS, Junqueira DG, Reis-de-Oliveira G, Baldasso PA, Cassoli JS, Martins-de-Souza D. Proteomics and molecular tools for unveiling missing links in the biochemical understanding of schizophrenia. Proteomics Clin Appl 2016; 10:1148-1158. [DOI: 10.1002/prca.201600021] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Revised: 06/21/2016] [Accepted: 07/14/2016] [Indexed: 12/20/2022]
Affiliation(s)
- Juliana M. Nascimento
- Department of Biochemistry and Tissue Biology; Laboratory of Neuroproteomics; Institute of Biology; University of Campinas (UNICAMP); Campinas São Paulo Brazil
| | - Sheila Garcia
- Department of Biochemistry and Tissue Biology; Laboratory of Neuroproteomics; Institute of Biology; University of Campinas (UNICAMP); Campinas São Paulo Brazil
| | - Verônica M. Saia-Cereda
- Department of Biochemistry and Tissue Biology; Laboratory of Neuroproteomics; Institute of Biology; University of Campinas (UNICAMP); Campinas São Paulo Brazil
| | - Aline G. Santana
- Department of Biochemistry and Tissue Biology; Laboratory of Neuroproteomics; Institute of Biology; University of Campinas (UNICAMP); Campinas São Paulo Brazil
| | - Caroline Brandao-Teles
- Department of Biochemistry and Tissue Biology; Laboratory of Neuroproteomics; Institute of Biology; University of Campinas (UNICAMP); Campinas São Paulo Brazil
| | - Giuliana S. Zuccoli
- Department of Biochemistry and Tissue Biology; Laboratory of Neuroproteomics; Institute of Biology; University of Campinas (UNICAMP); Campinas São Paulo Brazil
| | - Danielle G. Junqueira
- Department of Biochemistry and Tissue Biology; Laboratory of Neuroproteomics; Institute of Biology; University of Campinas (UNICAMP); Campinas São Paulo Brazil
| | - Guilherme Reis-de-Oliveira
- Department of Biochemistry and Tissue Biology; Laboratory of Neuroproteomics; Institute of Biology; University of Campinas (UNICAMP); Campinas São Paulo Brazil
| | - Paulo A. Baldasso
- Department of Biochemistry and Tissue Biology; Laboratory of Neuroproteomics; Institute of Biology; University of Campinas (UNICAMP); Campinas São Paulo Brazil
| | - Juliana S. Cassoli
- Department of Biochemistry and Tissue Biology; Laboratory of Neuroproteomics; Institute of Biology; University of Campinas (UNICAMP); Campinas São Paulo Brazil
| | - Daniel Martins-de-Souza
- Department of Biochemistry and Tissue Biology; Laboratory of Neuroproteomics; Institute of Biology; University of Campinas (UNICAMP); Campinas São Paulo Brazil
| |
Collapse
|
34
|
Domínguez M, de Oliveira E, Odena MA, Portero M, Pamplona R, Ferrer I. Redox proteomic profiling of neuroketal-adducted proteins in human brain: Regional vulnerability at middle age increases in the elderly. Free Radic Biol Med 2016; 95:1-15. [PMID: 26968793 DOI: 10.1016/j.freeradbiomed.2016.02.034] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Revised: 02/03/2016] [Accepted: 02/27/2016] [Indexed: 02/08/2023]
Abstract
Protein lipoxidation was assessed in the parietal cortex (PC), frontal cortex (FC), and cingulate gyrus (CG) in middle-aged and old-aged individuals with no clinical manifestations of cognitive impairment, in order to increase understanding of regional brain vulnerability to oxidative damage during aging. Twenty-five lipoxidized proteins were identified in all the three regions although with regional specificities, by using redox proteomics to detect target proteins of neuroketals (NKT) adduction. The number of cases with NKT-adducted proteins was higher in old-aged individuals but most oxidized proteins were already present in middle-aged individuals. Differences in vulnerability to oxidation were dependent on the sub-cellular localization, secondary structure, and external exposition of certain amino acids. Lipoxidized proteins included those involved in energy metabolism, cytoskeleton, proteostasis, neurotransmission and O2/CO2, and heme metabolism. Total NKT and soluble oligomer levels were estimated employing slot-blot, and these were compared between age groups. Oligomers increased with age in PC and FC; NKT significantly increased with age in FC, whereas total NKT and oligomer levels were not modified in CG, thus highlighting differences in brain regional vulnerability with age. Oligomers significantly correlated with NKT levels in the three cortical regions, suggesting that protein NKT adduction parallels soluble oligomer formation.
Collapse
Affiliation(s)
- Mayelín Domínguez
- Institute of Neuropathology, University Hospital of Bellvitge, IDIBELL (Biomedical Research Institute of Bellvitge), Carrer Feixa Llarga sn, 08907 Hospitalet de Llobregat, Spain.
| | | | | | - Manuel Portero
- Department of Experimental Medicine, University of Lleida-Biomedical Research Institute of Lleida, 25198 Lleida, Spain.
| | - Reinald Pamplona
- Department of Experimental Medicine, University of Lleida-Biomedical Research Institute of Lleida, 25198 Lleida, Spain.
| | - Isidro Ferrer
- Institute of Neuropathology, University Hospital of Bellvitge, IDIBELL (Biomedical Research Institute of Bellvitge), Carrer Feixa Llarga sn, 08907 Hospitalet de Llobregat, Spain; Department of Pathology and Experimental Therapeutics, University of Barcelona, Carrer Feixa Llarga sn, 08907 Hospitalet de Llobregat, Spain; CIBERNED (Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas), Spain.
| |
Collapse
|
35
|
Cassoli JS, Guest PC, Santana AG, Martins-de-Souza D. Employing proteomics to unravel the molecular effects of antipsychotics and their role in schizophrenia. Proteomics Clin Appl 2016; 10:442-55. [PMID: 26679983 DOI: 10.1002/prca.201500109] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2015] [Revised: 11/15/2015] [Accepted: 12/09/2015] [Indexed: 12/20/2022]
Abstract
Schizophrenia is an incurable neuropsychiatric disorder managed mostly by treatment of the patients with antipsychotics. However, the efficacy of these drugs has remained only low to moderate despite intensive research efforts since the early 1950s when chlorpromazine, the first antipsychotic, was synthesized. In addition, antipsychotic treatment can produce often undesired severe side effects in the patients and addressing these remains a large unmet clinical need. One of the reasons for the low effectiveness of these drugs is the limited knowledge about the molecular mechanisms of schizophrenia, which impairs the development of new and more effective treatments. Recently, proteomic studies of clinical and preclinical samples have identified changes in the levels of specific proteins in response to antipsychotic treatment, which have converged on molecular pathways such as cell communication and signaling, inflammation and cellular growth, and maintenance. The findings of these studies are summarized and discussed in this review and we suggest that this provides validation of proteomics as a useful tool for mining drug mechanisms of action and potentially for pinpointing novel molecular targets that may enable development of more effective medications.
Collapse
Affiliation(s)
- Juliana S Cassoli
- Laboratory of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
| | - Paul C Guest
- Laboratory of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
| | - Aline G Santana
- Laboratory of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
| | - Daniel Martins-de-Souza
- Laboratory of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, São Paulo, Brazil.,UNICAMP Neurobiology Center, Campinas, São Paulo, Brazil
| |
Collapse
|