1
|
Priya P, Srivastava A, Yadav N, Mittal R, Anand S, Banerjee J, Tripathi M, Chandra PS, Doddamani R, Sharma MC, Lalwani S, Siraj F, Dixit AB. Subunit specific altered expression and activity of casein kinase 2 in the brain tissues resected from mesial temporal lobe epilepsy with hippocampal sclerosis patients & rodent temporal lobe epilepsy model. Neuroscience 2025; 572:108-121. [PMID: 40064363 DOI: 10.1016/j.neuroscience.2025.03.001] [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: 02/28/2025] [Accepted: 03/01/2025] [Indexed: 03/14/2025]
Abstract
INTRODUCTION Mesial temporal lobe epilepsy (MTLE), is associated with dysregulated excitatory-inhibitory balance in the brain. Numerous enzymes, protein kinases, that are modulated through phosphorylation, have been linked with key processes involved in the pathogenesis of epilepsy. Therefore, in this study, we determined the subunit specific expression and activity of multi-subunit casein Kinase 2 (CK2) which influences NMDARs through phosphorylation events, in MTS patients as well as pilocarpine model of TLE. METHODS mRNA expression of CK2 (α, α', β) & NR2B was measured by real time PCR andprotein expression of CK2 (α, α', β), NR2B, and NR2B Ser1480 were evaluated using western blotting and immunohistochemistry in experimental models of TLE and MTS patients. CK2 α and α' activity was measured by kinase assay. RESULTS Significant increase in CK2α', CK2β, and NR2B mRNA expression were noted in chronic TLE rat model. Similarly, MTS patients displayed upregulated CK2α' and CK2β expressions, but NR2B mRNA remained unchanged. CK2α', CK2β, and NR2B Ser1480 protein expressions were higher in chronic TLE and MTS patients in relation to controls (p < 0.05), as was kinase activity (p < 0.05). In acute TLE rats, only NR2B protein expression was upregulated (p < 0.05). CONCLUSION Our research demonstrated for the first time the upregulation of CK2α' subunit and its increased kinase activityin resected brain samples from MTS patients as well as pilocarpine model of TLE. Altered expression and higher activity of CK2 α' highlights subunit specific contribution, suggesting the modulation of NMDA receptors by Casein Kinase 2 may contribute to hyperexcitability in MTLE.
Collapse
Affiliation(s)
- Priya Priya
- Dr. B R Ambedkar Centre for Biomedical Research, University of Delhi, Delhi, India
| | | | - Nitin Yadav
- Dr. B R Ambedkar Centre for Biomedical Research, University of Delhi, Delhi, India; Department of Biomedical Science, Bhaskaracharya College of Applied Sciences, University of Delhi, Delhi, India
| | - Radhika Mittal
- Dr. B R Ambedkar Centre for Biomedical Research, University of Delhi, Delhi, India
| | - Sneha Anand
- Dr. B R Ambedkar Centre for Biomedical Research, University of Delhi, Delhi, India
| | | | | | | | | | | | - Sanjeev Lalwani
- Department of Forensic Medicine &Toxicology, AIIMS, New Delhi, India
| | - Fouzia Siraj
- National Institute of Pathology, New Delhi, India
| | | |
Collapse
|
2
|
Dóra F, Hajdu T, Renner É, Paál K, Alpár A, Palkovits M, Chinopoulos C, Dobolyi A. Reverse phase protein array-based investigation of mitochondrial genes reveals alteration of glutaminolysis in the parahippocampal cortex of people who died by suicide. Transl Psychiatry 2024; 14:479. [PMID: 39604371 PMCID: PMC11603240 DOI: 10.1038/s41398-024-03137-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 09/20/2024] [Accepted: 09/26/2024] [Indexed: 11/29/2024] Open
Abstract
A moderating hub between resting state networks (RSNs) and the medial temporal lobe (MTL) is the parahippocampal cortex (PHC). Abnormal activity has been reported in depressed patients and suicide attempters in this region. Alterations in neuronal mitochondrial function may contribute to depression and suicidal behavior. However, little is known about the underlying molecular level changes in relevant structures. Specifically, expressional changes related to suicide have not been reported in the PHC. In this study, we compared the protein expression levels of genes encoding tricarboxylic acid (TCA) cycle enzymes in the PHC of adult individuals who died by suicide by reverse phase protein array (RPPA), which was corroborated by qRT-PCR at the mRNA level. Postmortem human brain samples were collected from 12 control and 10 suicidal individuals. The entorhinal cortex, which is topographically anterior to the PHC in the parahippocampal gyrus, and some other cortical brain regions were utilized for comparison. The results of the RPPA analysis revealed that the protein levels of DLD, OGDH, SDHB, SUCLA2, and SUCLG2 subunits were significantly elevated in the PHC but not in other cortical brain regions. In accordance with these findings, the mRNA levels of the respective subunits were also increased in the PHC. The subunits with altered levels are implicated in enzyme complexes involved in the oxidative decarboxylation branch of glutamine catabolism. These data suggest a potential role of glutaminolysis in the pathophysiology of suicidal behavior in the PHC.
Collapse
Affiliation(s)
- Fanni Dóra
- Human Brain Tissue Bank, Semmelweis University, Budapest, 1094, Hungary
- Laboratory of Neuromorphology, Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, 1094, Hungary
| | - Tamara Hajdu
- Laboratory of Neuromorphology, Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, 1094, Hungary
- Department of Physiology and Neurobiology, Eötvös Loránd University, Budapest, 1117, Hungary
| | - Éva Renner
- Human Brain Tissue Bank, Semmelweis University, Budapest, 1094, Hungary
| | - Krisztina Paál
- Department of Biochemistry and Molecular Biology, Semmelweis University, Budapest, 1094, Hungary
| | - Alán Alpár
- Human Brain Tissue Bank, Semmelweis University, Budapest, 1094, Hungary
- Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, 1094, Hungary
| | - Miklós Palkovits
- Human Brain Tissue Bank, Semmelweis University, Budapest, 1094, Hungary
| | - Christos Chinopoulos
- Department of Biochemistry and Molecular Biology, Semmelweis University, Budapest, 1094, Hungary.
| | - Arpád Dobolyi
- Laboratory of Neuromorphology, Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, 1094, Hungary.
- Department of Physiology and Neurobiology, Eötvös Loránd University, Budapest, 1117, Hungary.
| |
Collapse
|
3
|
Zwirner J, Devananthan P, Docherty PD, Ondruschka B, Kabaliuk N. The influence of cooling on biomechanical time since death estimations using ovine brain tissue. Int J Legal Med 2024; 138:2541-2549. [PMID: 38902542 PMCID: PMC11490522 DOI: 10.1007/s00414-024-03267-3] [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: 02/22/2024] [Accepted: 06/03/2024] [Indexed: 06/22/2024]
Abstract
The significance of biomechanical analyses for forensic time since death estimations has recently been demonstrated. Previous biomechanical analyses successfully discriminated post-mortem brain tissue from tissue with a post-mortem interval of at least one day when held at 20 °C. However, the practical utility of such analyses beyond day one at 20 °C was limited. This study investigates the storage, loss, and complex shear modulus of various brain regions in sheep stored at 4 °C in 24-hour intervals over four days post-mortem using rheometry tests. The aim is to identify the critical biomechanical tissue property values to predict post-mortem time and assess the temperature sensitivity of the rheometry method by comparing results to recent findings at 20 °C. Thirty sheep brains were examined, including the frontal lobe, parietal lobe, anterior and posterior deep brain, superior colliculi, pons, medulla, and cerebellum. Rheometry tests were conducted, and receiver operator characteristic analyses were employed to establish cut-off values. At 4 °C storage, all investigated biomechanical properties of the examined brain regions remained stable for at least one day post-mortem. Using cerebellar samples stored at 4 °C, a post-mortem interval of at least two days could be determined with excellent diagnostic ability. Complex shear modulus values below 1435 Pa or storage modulus values below 1313 Pa allowed prediction of two or more days post-mortem. Comparisons between 4 °C and 20 °C revealed brain region-specific results. For instance, the complex shear moduli of the anterior deep brain at 4 °C were significantly higher on all individual testing days when compared to 20 °C. In contrast, the combined medulla and pons samples were similar on each day. Rheometry testing of brain tissue consistently stored at 4 °C since death proved valuable for forensic time since death estimations starting from two days after death.
Collapse
Affiliation(s)
- Johann Zwirner
- Institute of Legal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
- Department of Oral Sciences, University of Otago, Dunedin, New Zealand.
| | - Pavithran Devananthan
- Department of Mechanical Engineering, University of Canterbury, Christchurch, New Zealand
- Biomolecular Interaction Centre, University of Canterbury, Christchurch, New Zealand
| | - Paul D Docherty
- Department of Mechanical Engineering, University of Canterbury, Christchurch, New Zealand
| | - Benjamin Ondruschka
- Institute of Legal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Natalia Kabaliuk
- Department of Mechanical Engineering, University of Canterbury, Christchurch, New Zealand
- Biomolecular Interaction Centre, University of Canterbury, Christchurch, New Zealand
| |
Collapse
|
4
|
Bonelli M, Di Giuseppe F, Tupone N, Di Virgilio V, Catena AM, Locatelli M, Ascani G, Giammaria G, Ciccarelli R, D’Ovidio C, Angelucci S. Proteomic Characterization of Changes in Mouse Brain Cortex Protein Expression at Different Post-Mortem Intervals: A Preliminary Study for Forensic Biomarker Identification. Int J Mol Sci 2024; 25:8736. [PMID: 39201424 PMCID: PMC11354345 DOI: 10.3390/ijms25168736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 08/01/2024] [Accepted: 08/07/2024] [Indexed: 09/02/2024] Open
Abstract
Accuracy in the evaluation of death-induced tissue degradation for thanato-chronological purposes is strictly dependent on the condition of the biological source as well as on the precision of post-mortem interval (PMI) estimation. Thus, the optimization of tissue handling and identification of sensitive post-mortem biomarkers could help establish a timeline for post-mortem events. To this aim, we investigated the proteome changes in cortex samples of 6-week-old female SAMR1 mice over a post-mortem time course. After death, brain tissue was removed immediately (T0), and after 4, 8, 12, 24, and 32 h, four mice were used for each time period, and animals were maintained at 4 °C until brain removal. Dissected tissues were frozen at -80 °C until processed. Proteomic analysis, performed on samples related to early and late PMIs (<24 h and >24 h post-mortem, respectively) showed protein level changes as compared to T0 samples, with a remarkable increase in Calpain11 in the early PMI, as well as in Caspases 7 and 8 together with Gasdermin 3 in late PMI. These findings were confirmed by LIFT mass spectrometry technology and western blot analysis and, although requiring further investigation in other biological samples, suggest that these proteins could be considered as putative biomarkers of different PMIs.
Collapse
Affiliation(s)
- Martina Bonelli
- Department of Medicine and Aging Sciences, University “G. d’Annunzio” of Chieti-Pescara, Via dei Vestini 31, 66100 Chieti, Italy; (M.B.); (C.D.)
- Center for Advanced Studies and Technologies (CAST), University “G. d’Annunzio” of Chieti-Pescara, Via Luigi Polacchi 13, 66100 Chieti, Italy; (F.D.G.); (N.T.); (V.D.V.); (S.A.)
| | - Fabrizio Di Giuseppe
- Center for Advanced Studies and Technologies (CAST), University “G. d’Annunzio” of Chieti-Pescara, Via Luigi Polacchi 13, 66100 Chieti, Italy; (F.D.G.); (N.T.); (V.D.V.); (S.A.)
- Department of Innovative Technologies in Medicine and Dentistry, University “G. d’Annunzio” of Chieti-Pescara, Via dei Vestini 31, 66100 Chieti, Italy
| | - Nicola Tupone
- Center for Advanced Studies and Technologies (CAST), University “G. d’Annunzio” of Chieti-Pescara, Via Luigi Polacchi 13, 66100 Chieti, Italy; (F.D.G.); (N.T.); (V.D.V.); (S.A.)
- Department of Innovative Technologies in Medicine and Dentistry, University “G. d’Annunzio” of Chieti-Pescara, Via dei Vestini 31, 66100 Chieti, Italy
| | - Vimal Di Virgilio
- Center for Advanced Studies and Technologies (CAST), University “G. d’Annunzio” of Chieti-Pescara, Via Luigi Polacchi 13, 66100 Chieti, Italy; (F.D.G.); (N.T.); (V.D.V.); (S.A.)
- Department of Innovative Technologies in Medicine and Dentistry, University “G. d’Annunzio” of Chieti-Pescara, Via dei Vestini 31, 66100 Chieti, Italy
| | - Antonio Maria Catena
- Forensic Medicine, Facoltà di Medicina e Chirurgia Via Montpellier, Tor Vergata University, 100133 Roma, Italy;
| | - Marcello Locatelli
- Department of Pharmacy, University “G. d’Annunzio” of Chieti-Pescara, Via dei Vestini 31, 66100 Chieti, Italy;
| | - Giuliano Ascani
- UOSD Maxillofacial Surgery, Azienda Sanitaria Locale di Pescara, Via Renato Paolini 47, 65124 Pescara, Italy;
| | | | - Renata Ciccarelli
- Center for Advanced Studies and Technologies (CAST), University “G. d’Annunzio” of Chieti-Pescara, Via Luigi Polacchi 13, 66100 Chieti, Italy; (F.D.G.); (N.T.); (V.D.V.); (S.A.)
| | - Cristian D’Ovidio
- Department of Medicine and Aging Sciences, University “G. d’Annunzio” of Chieti-Pescara, Via dei Vestini 31, 66100 Chieti, Italy; (M.B.); (C.D.)
- Center for Advanced Studies and Technologies (CAST), University “G. d’Annunzio” of Chieti-Pescara, Via Luigi Polacchi 13, 66100 Chieti, Italy; (F.D.G.); (N.T.); (V.D.V.); (S.A.)
| | - Stefania Angelucci
- Center for Advanced Studies and Technologies (CAST), University “G. d’Annunzio” of Chieti-Pescara, Via Luigi Polacchi 13, 66100 Chieti, Italy; (F.D.G.); (N.T.); (V.D.V.); (S.A.)
- Department of Innovative Technologies in Medicine and Dentistry, University “G. d’Annunzio” of Chieti-Pescara, Via dei Vestini 31, 66100 Chieti, Italy
| |
Collapse
|
5
|
Dean B, Seymour N, Gibbons A. Lower levels of TRAF1 in Brodmann's area 24, but not 46, in bipolar disorders are not detectable in major depressive disorders. J Affect Disord 2024; 356:316-322. [PMID: 38583598 DOI: 10.1016/j.jad.2024.04.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 04/01/2024] [Accepted: 04/03/2024] [Indexed: 04/09/2024]
Abstract
INTRODUCTION Multiple lines of research implicate inflammation-related pathways in the molecular pathology of mood disorders, with our data suggesting a critical role for aberrant cortical tumour necrosis factor α (TNF)-signaling in the molecular pathology of bipolar disorders (BPD) and major depressive disorders (MDD). METHODS To extend our understanding of changes in TNF-signaling pathways in mood disorders we used Western blotting to measure levels of tumour necrosis factor receptor associated factor 1 (TRAF1) and transmembrane TNF receptor superfamily member 1B (tmTNFRSF1B) in Brodmann's areas (BA) 24 and 46 from people with BPD and MDD. These proteins are key rate-limiting components within TNF-signaling pathways. RESULTS Compared to controls, there were higher levels of TRAF1 of large effect size (η = 0.19, Cohen's d = 0.97) in BA 24, but not BA 46, from people with BPD. Levels of TRAF1 were not altered in MDD and levels of tmTNFRSF1B were not altered in either disorder. LIMITATIONS The cases studied had been treated with psychotropic drugs prior to death which is an unresolvable study confound. Cohort sizes are relatively small but not untypical of postmortem CNS studies. CONCLUSIONS To facilitate post-synaptic signaling, TRAF1 is known to associate with tmTNFRSF1B after that receptor takes its activated conformation which occurs predominantly after it binds to transmembrane TNF (tmTNF). Simultaneously, when tmTNFRSF1B binds to tmTNF reverse signaling through tmTNF is activated. Hence our findings in BA 24 argues that bidirectional TNF-signaling may be an important component of the molecular pathology of BPD.
Collapse
Affiliation(s)
- Brian Dean
- The Molecular Psychiatry Laboratory, The Florey Institute of Neuroscience and Mental Health, Parkville, Victoria, Australia; The University of Melbourne Florey Department of Neuroscience and Mental Health, Parkville, Victoria, Australia.
| | - Natasha Seymour
- The Molecular Psychiatry Laboratory, The Florey Institute of Neuroscience and Mental Health, Parkville, Victoria, Australia; The University of Melbourne Florey Department of Neuroscience and Mental Health, Parkville, Victoria, Australia
| | - Andrew Gibbons
- Department of Psychiatry, School of Clinical Sciences at Monash Health, Monash University, Clayton, Victoria, Australia
| |
Collapse
|
6
|
Seiffer S, Brendler J, Schulz A, Ricken A. Reliable detection of RNA in hippocampus sections of mice by FISH up to a post-mortem delay of 24 h. Histochem Cell Biol 2024; 161:539-547. [PMID: 38582805 PMCID: PMC11162364 DOI: 10.1007/s00418-024-02277-x] [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] [Accepted: 03/18/2024] [Indexed: 04/08/2024]
Abstract
Proteins can be successfully localized in post-mortem (PM) brain tissue sections if the time until PM tissue sampling is not too long. In this study, we show that this also applies to the localization of RNA and in particular to the RNA of microglia-specific receptor proteins using the probes and the RNAscope™ Multiplex Fluorescent Detection Kit v2 from Advanced Cell Diagnostics. Brains were removed from killed mice after different PM delays and processed into paraffin sections. In sections of brains from animals whose cadavers had been kept at room temperature (21 °C) before tissue removal, ubiquitously expressed RNAs of genes with low to high expression levels (Polr2a, PPIB, and UBC) were reliably detected in the brain sections even if tissue removal was delayed by up to 48 h. In addition, microglia-specific G protein-coupled receptor RNA (Gpr34, P2ry12) could be reliably assigned to microglia by simultaneous labeling of the microglia with microglia-specific antibodies (Iba1 or P2ry12). Only after a delay of 48 h until tissue removal were the receptor RNA signals significantly lower. The reduction in receptor RNA signals could be delayed if the animal cadavers were stored at 4 °C until the brains were removed. Tissue sections of PM brain samples allow the spatial and cellular localization of specific RNA, at least if the sampling takes place within the first 24 h of PM.
Collapse
Affiliation(s)
- Sophie Seiffer
- Institute of Anatomy, Medical Faculty, Leipzig University, Leipzig, Germany
| | - Jana Brendler
- Institute of Anatomy, Medical Faculty, Leipzig University, Leipzig, Germany
| | - Angela Schulz
- Medical Faculty, Rudolf Schönheimer Institute of Biochemistry, Leipzig University, Leipzig, Germany
| | - Albert Ricken
- Institute of Anatomy, Medical Faculty, Leipzig University, Leipzig, Germany.
| |
Collapse
|
7
|
Dean B, Duce J, Li QX, Masters CL, Scarr E. Lower levels of soluble β-amyloid precursor protein, but not β-amyloid, in the frontal cortex in schizophrenia. Psychiatry Res 2024; 331:115656. [PMID: 38071879 DOI: 10.1016/j.psychres.2023.115656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 11/28/2023] [Accepted: 12/01/2023] [Indexed: 01/02/2024]
Abstract
We identified a sub-group (25%) of people with schizophrenia (muscarinic receptor deficit schizophrenia (MRDS)) that are characterised because of markedly lower levels of cortical muscarinic M1 receptors (CHRM1) compared to most people with the disorder (non-MRDS). Notably, bioinformatic analyses of our cortical gene expression data shows a disturbance in the homeostasis of a biochemical pathway that regulates levels of CHRM1. A step in this pathway is the processing of β-amyloid precursor protein (APP) and therefore we postulated there would be altered levels of APP in the frontal cortex from people with MRDS. Here we measure levels of CHRM1 using [3H]pirenzepine binding, soluble APP (sAPP) using Western blotting and amyloid beta peptides (Aβ1-40 and Aβ1-42) using ELISA in the frontal cortex (Brodmann's area 6: BA 6; MRDS = 14, non-MRDS = 14, controls = 14). We confirmed the MRDS cohort in this study had the expected low levels of [3H]pirenzepine binding. In addition, we showed that people with schizophrenia, independent of their sub-group status, had lower levels of sAPP compared to controls but did not have altered levels of Aβ1-40 or Aβ1-42. In conclusion, whilst changes in sAPP are not restricted to MRDS our data could indicate a role of APP, which is important in axonal and synaptic pruning, in the molecular pathology of the syndrome of schizophrenia.
Collapse
Affiliation(s)
- Brian Dean
- The Florey, Parkville, Victoria, Australia; The University of Melbourne of Melbourne Florey Department of Neuroscience and Mental Health, Parkville, Victoria, Australia.
| | - James Duce
- MSD Discovery Centre, 120 Moorgate, London, UK
| | - Qiao-Xin Li
- The Florey, Parkville, Victoria, Australia; The University of Melbourne of Melbourne Florey Department of Neuroscience and Mental Health, Parkville, Victoria, Australia
| | - Colin L Masters
- The Florey, Parkville, Victoria, Australia; The University of Melbourne of Melbourne Florey Department of Neuroscience and Mental Health, Parkville, Victoria, Australia
| | - Elizabeth Scarr
- The Department of Psychiatry, The University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
8
|
Obis E, Sol J, Andres-Benito P, Martín-Gari M, Mota-Martorell N, Galo-Licona JD, Piñol-Ripoll G, Portero-Otin M, Ferrer I, Jové M, Pamplona R. Lipidomic Alterations in the Cerebral Cortex and White Matter in Sporadic Alzheimer's Disease. Aging Dis 2023; 14:1887-1916. [PMID: 37196109 PMCID: PMC10529741 DOI: 10.14336/ad.2023.0217] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Accepted: 02/17/2023] [Indexed: 05/19/2023] Open
Abstract
Non-targeted LC-MS/MS-based lipidomic analysis was conducted in post-mortem human grey matter frontal cortex area 8 (GM) and white matter of the frontal lobe centrum semi-ovale (WM) to identify lipidome fingerprints in middle-aged individuals with no neurofibrillary tangles and senile plaques, and cases at progressive stages of sporadic Alzheimer's disease (sAD). Complementary data were obtained using RT-qPCR and immunohistochemistry. The results showed that WM presents an adaptive lipid phenotype resistant to lipid peroxidation, characterized by a lower fatty acid unsaturation, peroxidizability index, and higher ether lipid content than the GM. Changes in the lipidomic profile are more marked in the WM than in GM in AD with disease progression. Four functional categories are associated with the different lipid classes affected in sAD: membrane structural composition, bioenergetics, antioxidant protection, and bioactive lipids, with deleterious consequences affecting both neurons and glial cells favoring disease progression.
Collapse
Affiliation(s)
- Elia Obis
- Department of Experimental Medicine, Lleida University (UdL), Lleida Biomedical Research Institute (IRBLleida), Lleida, Spain.
| | - Joaquim Sol
- Department of Experimental Medicine, Lleida University (UdL), Lleida Biomedical Research Institute (IRBLleida), Lleida, Spain.
- Catalan Institute of Health (ICS), Lleida, Spain, Research Support Unit (USR), Fundació Institut Universitari per a la Recerca en Atenció Primària de Salut Jordi Gol i Gurina (IDIAP JGol), Lleida, Spain.
| | - Pol Andres-Benito
- CIBERNED (Network Centre of Biomedical Research of Neurodegenerative Diseases), Institute of Health Carlos III, Ministry of Economy and Competitiveness, Madrid, Spain.
- Bellvitge University Hospital-Bellvitge Biomedical Research Institute (IDIBELL), E-08907 Hospitalet de Llobregat, Barcelona, Spain.
| | - Meritxell Martín-Gari
- Department of Experimental Medicine, Lleida University (UdL), Lleida Biomedical Research Institute (IRBLleida), Lleida, Spain.
| | - Natàlia Mota-Martorell
- Department of Experimental Medicine, Lleida University (UdL), Lleida Biomedical Research Institute (IRBLleida), Lleida, Spain.
| | - José Daniel Galo-Licona
- Department of Experimental Medicine, Lleida University (UdL), Lleida Biomedical Research Institute (IRBLleida), Lleida, Spain.
| | - Gerard Piñol-Ripoll
- Unitat Trastorns Cognitius, Clinical Neuroscience Research, Santa Maria University Hospital, IRBLleida, Lleida, Spain.
| | - Manuel Portero-Otin
- Department of Experimental Medicine, Lleida University (UdL), Lleida Biomedical Research Institute (IRBLleida), Lleida, Spain.
| | - Isidro Ferrer
- CIBERNED (Network Centre of Biomedical Research of Neurodegenerative Diseases), Institute of Health Carlos III, Ministry of Economy and Competitiveness, Madrid, Spain.
- Bellvitge University Hospital-Bellvitge Biomedical Research Institute (IDIBELL), E-08907 Hospitalet de Llobregat, Barcelona, Spain.
- Department of Pathology and Experimental Therapeutics, University of Barcelona, L’Hospitalet de Llobregat, Barcelona, Spain.
| | - Mariona Jové
- Department of Experimental Medicine, Lleida University (UdL), Lleida Biomedical Research Institute (IRBLleida), Lleida, Spain.
| | - Reinald Pamplona
- Department of Experimental Medicine, Lleida University (UdL), Lleida Biomedical Research Institute (IRBLleida), Lleida, Spain.
| |
Collapse
|
9
|
Exton J, Higgins JMG, Chen J. Acute brain slice elastic modulus decreases over time. Sci Rep 2023; 13:12826. [PMID: 37550376 PMCID: PMC10406937 DOI: 10.1038/s41598-023-40074-z] [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: 02/10/2023] [Accepted: 08/04/2023] [Indexed: 08/09/2023] Open
Abstract
A common benchmark in the brain tissue mechanics literature is that the properties of acute brain slices should be measured within 8 h of the experimental animal being sacrificed. The core assumption is that-since there is no substantial protein degradation during this time-there will be no change to elastic modulus. This assumption overlooks the possibility of other effects (such as osmotic swelling) that may influence the mechanical properties of the tissue. To achieve consistent and accurate analysis of brain mechanics, it is important to account for or mitigate these effects. Using atomic force microscopy (AFM), tissue hydration and volume measurements, we find that acute brain slices in oxygenated artificial cerebrospinal fluid (aCSF) with a standard osmolarity of 300 mOsm/l experience rapid swelling, softening, and increases in hydration within the first 2 hours after slicing. Reductions in elastic modulus can be partly mitigated by addition of chondroitinase ABC enzyme (CHABC). Increasing aCSF osmolarity to 400 mOsm/l does not prevent softening but may hasten equilibration of samples to a point where measurements of relative elastic modulus are consistent across experiments.
Collapse
Affiliation(s)
- John Exton
- School of Engineering, Newcastle University, Newcastle Upon Tyne, NE1 7RU, UK
| | - Jonathan M G Higgins
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Framlington Place, Newcastle Upon Tyne, NE2 4HH, UK
| | - Jinju Chen
- School of Engineering, Newcastle University, Newcastle Upon Tyne, NE1 7RU, UK.
| |
Collapse
|
10
|
Moors TE, Mona D, Luehe S, Duran-Pacheco G, Spycher L, Mundigl O, Kaluza K, Huber S, Hug MN, Kremer T, Ritter M, Dziadek S, Dernick G, van de Berg WDJ, Britschgi M. Multi-platform quantitation of alpha-synuclein human brain proteoforms suggests disease-specific biochemical profiles of synucleinopathies. Acta Neuropathol Commun 2022; 10:82. [PMID: 35659116 PMCID: PMC9164351 DOI: 10.1186/s40478-022-01382-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 05/16/2022] [Indexed: 12/02/2022] Open
Abstract
Based on immunostainings and biochemical analyses, certain post-translationally modified alpha-synuclein (aSyn) variants, including C-terminally truncated (CTT) and Serine-129 phosphorylated (pSer129) aSyn, are proposed to be involved in the pathogenesis of synucleinopathies such as Parkinson’s disease with (PDD) and without dementia (PD), dementia with Lewy bodies (DLB), and multiple system atrophy (MSA). However, quantitative information about aSyn proteoforms in the human brain in physiological and different pathological conditions is still limited. To address this, we generated sequential biochemical extracts of the substantia nigra, putamen and hippocampus from 28 donors diagnosed and neuropathologically-confirmed with different synucleinopathies (PD/PDD/DLB/MSA), as well as Alzheimer’s disease, progressive supranuclear palsy, and aged normal subjects. The tissue extracts were used to build a reverse phase array including 65 aSyn antibodies for detection. In this multiplex approach, we observed increased immunoreactivity in donors with synucleinopathies compared to controls in detergent-insoluble fractions, mainly for antibodies against CT aSyn and pSer129 aSyn. In addition, despite of the restricted sample size, clustering analysis suggested disease-specific immunoreactivity signatures in patient groups with different synucleinopathies. We aimed to validate and quantify these findings using newly developed immunoassays towards total, 119 and 122 CTT, and pSer129 aSyn. In line with previous studies, we found that synucleinopathies shared an enrichment of post-translationally modified aSyn in detergent-insoluble fractions compared to the other analyzed groups. Our measurements allowed for a quantitative separation of PDD/DLB patients from other synucleinopathies based on higher detergent-insoluble pSer129 aSyn concentrations in the hippocampus. In addition, we found that MSA stood out due to enrichment of CTT and pSer129 aSyn also in the detergent-soluble fraction of the SN and putamen. Together, our results achieved by multiplexed and quantitative immunoassay-based approaches in human brain extracts of a limited sample set point to disease-specific biochemical aSyn proteoform profiles in distinct neurodegenerative disorders.
Collapse
|
11
|
Neuroplastic alterations in cannabinoid receptors type 1 (CB1) in animal models of epileptic seizures. Neurosci Biobehav Rev 2022; 137:104675. [PMID: 35460705 DOI: 10.1016/j.neubiorev.2022.104675] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Revised: 03/16/2022] [Accepted: 04/17/2022] [Indexed: 01/01/2023]
Abstract
Currently, there is an urgent need to better comprehend neuroplastic alterations in cannabinoid receptors type 1 (CB1) and to understand the biological meaning of these alterations in epileptic disorders. The present study reviewed neuroplastic changes in CB1 distribution, expression, and functionality in animal models of epileptic seizures. Neuroplastic alterations in CB1 were consistently observed in chemical, genetic, electrical, and febrile seizure models. Most studies assessed changes in hippocampal and cortical CB1, while thalamic, hypothalamic, and brainstem nuclei were rarely investigated. Additionally, the relationship between CB1 alteration and the control of brain excitability through modulation of specific neuronal networks, such as striatonigral, nigrotectal and thalamocortical pathways, and inhibitory projections to hippocampal pyramidal neurons, were all presented and discussed in the present review. Neuroplastic alterations in CB1 detected in animal models of epilepsy may reflect two different scenarios: (1) endogenous adaptations aimed to control neuronal hyperexcitability in epilepsy or (2) pathological alterations that facilitate neuronal hyperexcitability. Additionally, a better comprehension of neuroplastic and functional alterations in CB1 can improve pharmacological therapies for epilepsies and their comorbidities.
Collapse
|
12
|
Wu X, Deng C, Su Y, Zhang C, Chen M, Tian K, Wu H, Xu S. The effect of prolonged formalin fixation on the expression of proteins in human brain tissues. Acta Histochem 2022; 124:151879. [PMID: 35358895 DOI: 10.1016/j.acthis.2022.151879] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 03/04/2022] [Accepted: 03/14/2022] [Indexed: 11/24/2022]
Abstract
Formalin-fixed, paraffin-embedded (FFPE) tissues have been widely used in researches. Proteins and nucleic acids in prolonged FFPE tissues display different degrees of degradation. We investigated the effect of prolonged formalin fixation on protein expression in human brain tissues. Twenty-eight middle prefrontal front cortex tissue blocks from human brains prefixed in formalin were obtained from a brain bank. The tissue blocks were divided into two groups, the control group and the prolonged fixation group. Quantitative immunocytochemistry was used to analyse the biological markers of Fox-3, Rbfox3 (NeuN), glial fibrillary acidic protein (GFAP), ionized calcium binding adapter molecule-1 (IBA-1) and glyceraldehyde-3-phosphate dehydrogenase (GAPDH). Nissl staining showed that positive signaling of Nissl body was significantly decreased by 16.6% in the prolonged fixation group. In addition, the staining intensity of Nissl body was negatively correlated with fixation time. The level of NeuN immunoreactivity (ir) was significantly reduced by 19.31% in the prolonged fixation group. Moreover, there was a significant negative correlation between NeuN-ir and fixation time. There were no significant changes in GFAP-ir, IBA-1-ir and GAPDH-ir between control group and the prolonged fixation group. Prolonged formalin-fixed tissues showed time- and molecule-dependent protein changes, which may be potential confounders in the clinic and researches. Our study suggested short formalin fixation time is recommended when using PPFE brain tissues.
Collapse
|
13
|
Faber J, Hinrichsen J, Greiner A, Reiter N, Budday S. Tissue-Scale Biomechanical Testing of Brain Tissue for the Calibration of Nonlinear Material Models. Curr Protoc 2022; 2:e381. [PMID: 35384412 DOI: 10.1002/cpz1.381] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 01/14/2022] [Indexed: 06/14/2023]
Abstract
Brain tissue is one of the most complex and softest tissues in the human body. Due to its ultrasoft and biphasic nature, it is difficult to control the deformation state during biomechanical testing and to quantify the highly nonlinear, time-dependent tissue response. In numerous experimental studies that have investigated the mechanical properties of brain tissue over the last decades, stiffness values have varied significantly. One reason for the observed discrepancies is the lack of standardized testing protocols and corresponding data analyses. The tissue properties have been tested on different length and time scales depending on the testing technique, and the corresponding data have been analyzed based on simplifying assumptions. In this review, we highlight the advantage of using nonlinear continuum mechanics based modeling and finite element simulations to carefully design experimental setups and protocols as well as to comprehensively analyze the corresponding experimental data. We review testing techniques and protocols that have been used to calibrate material model parameters and discuss artifacts that might falsify the measured properties. The aim of this work is to provide standardized procedures to reliably quantify the mechanical properties of brain tissue and to more accurately calibrate appropriate constitutive models for computational simulations of brain development, injury and disease. Computational models can not only be used to predictively understand brain tissue behavior, but can also serve as valuable tools to assist diagnosis and treatment of diseases or to plan neurosurgical procedures. © 2022 The Authors. Current Protocols published by Wiley Periodicals LLC.
Collapse
Affiliation(s)
- Jessica Faber
- Friedrich-Alexander-Universität Erlangen-Nürnberg, Institute of Applied Mechanics, Egerlandstraße 5, 91058 Erlangen, Germany
| | - Jan Hinrichsen
- Friedrich-Alexander-Universität Erlangen-Nürnberg, Institute of Applied Mechanics, Egerlandstraße 5, 91058 Erlangen, Germany
| | - Alexander Greiner
- Friedrich-Alexander-Universität Erlangen-Nürnberg, Institute of Applied Mechanics, Egerlandstraße 5, 91058 Erlangen, Germany
| | - Nina Reiter
- Friedrich-Alexander-Universität Erlangen-Nürnberg, Institute of Applied Mechanics, Egerlandstraße 5, 91058 Erlangen, Germany
| | - Silvia Budday
- Friedrich-Alexander-Universität Erlangen-Nürnberg, Institute of Applied Mechanics, Egerlandstraße 5, 91058 Erlangen, Germany
| |
Collapse
|
14
|
Qian L, Wang S, Zhou S, Sun Y, Zhao H. Influence of pia-arachnoid complex on the indentation response of porcine brain at different length scales. J Mech Behav Biomed Mater 2022; 127:104925. [DOI: 10.1016/j.jmbbm.2021.104925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 08/16/2021] [Accepted: 10/25/2021] [Indexed: 11/26/2022]
|
15
|
Biovalue in Human Brain Banking: Applications and Challenges for Research in Neurodegenerative Diseases. Methods Mol Biol 2021. [PMID: 34558013 DOI: 10.1007/978-1-0716-1783-0_17] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/12/2024]
Abstract
Brain banking occupies a central role for the advancement of the study of human neurodegenerative and neuropsychiatric diseases. The smooth functioning and effectiveness of a brain bank is largely a multidisciplinary effort and requires the cooperation and participation of several players including neurologists, neuropathologists, and research coordinators to guarantee that donated tissue is properly processed and archived. If properly run, brain banks can ultimately lay the foundation for new brain research and pioneer the discovery of new therapies for a variety of neurological diseases.
Collapse
|
16
|
Snelleksz M, Dean B. Lower levels of tubulin alpha 1b in the frontal pole in schizophrenia supports a role for changed cytoskeletal dynamics in the aetiology of the disorder. Psychiatry Res 2021; 303:114096. [PMID: 34274903 DOI: 10.1016/j.psychres.2021.114096] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 06/30/2021] [Accepted: 07/03/2021] [Indexed: 11/26/2022]
Abstract
Our transcriptomic study suggested there were markedly lower levels of tubulin alpha 1b (TUBA1B) expression in BA 10, but not BA 9, from patients with schizophrenia. We now use Western blotting to compare levels of TUBA1B protein in BA 9 and 10 from patients with schizophrenia and BA 10 from patients with mood disorders to controls as well as in the frontal cortex from rats after treatment with haloperidol, chlorpromazine or vehicle for 28 days. Levels of TUBA1B were significantly lower (- 18.6%) in BA 10, but not BA 9, from patients with schizophrenia. Levels of TUBA1B did not differ significantly from controls in BA 10 from patients with mood disorders or in the cortex of rats after antipsychotic drug treatments. Levels of TUBA1B were significantly lower (- 30%) in BA 10 from patients with schizophrenia who were not being treated with antipsychotic drugs close to death compared to those who were treated close to death. These data suggest that lower levels of TUBA1B, a cytoskeletal protein, in BA 10 from patients with schizophrenia are not a simple drug effect and therefore add to the hypothesis that a breakdown in cytoskeletal homoeostasis may be contributing to the genesis of the symptoms of the disorder.
Collapse
Affiliation(s)
- Megan Snelleksz
- The Molecular Psychiatry Laboratory, The Florey Institute for Neuroscience and Mental Health, Parkville, Victoria, Australia; The Florey Department of Neuroscience and Mental Health, Parkville, Victoria, Australia
| | - Brian Dean
- The Molecular Psychiatry Laboratory, The Florey Institute for Neuroscience and Mental Health, Parkville, Victoria, Australia; The Florey Department of Neuroscience and Mental Health, Parkville, Victoria, Australia.
| |
Collapse
|
17
|
Zissler A, Stoiber W, Geissenberger J, Steinbacher P, Monticelli FC, Pittner S. Influencing Factors on Postmortem Protein Degradation for PMI Estimation: A Systematic Review. Diagnostics (Basel) 2021; 11:1146. [PMID: 34201836 PMCID: PMC8304065 DOI: 10.3390/diagnostics11071146] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 06/09/2021] [Accepted: 06/16/2021] [Indexed: 11/17/2022] Open
Abstract
The present review provides an overview of the current research status on the effects of influencing factors on postmortem protein degradation used to estimate the PMI (postmortem interval). Focus was set on characteristics of internal and external influencing factors and the respective susceptibility and/or robustness of protein degradation. A systematic literature search up to December 2020 was conducted on the effect of influencing factors investigated in the context of postmortem protein degradation in the tissues of animals and humans using the scientific databases PubMed and Google Scholar, as well as the reference lists of eligible articles. We identified ten studies investigating a total of seven different influencing factors in degrading tissues/organs (n = 7) of humans and animals using six different methodological approaches. Although comparison of study outcomes was impeded by the high variety of investigated factors, and by high risk of bias appraisals, it was evident that the majority of the influencing factors concerned affected protein degradation, thus being able to modulate the precision of protein degradation-based PMI estimation. The results clearly highlight the need for a thorough screening for corresponding factors to enable the introduction of appropriate correction factors and exclusion criteria. This seems especially relevant for the protein degradation-based study of human PMI to increase the reliability and precision of the method and to facilitate a broader applicability in routine forensic casework.
Collapse
Affiliation(s)
- Angela Zissler
- Department of Biosciences, University of Salzburg, 5020 Salzburg, Austria; (A.Z.); (W.S.); (J.G.); (P.S.)
| | - Walter Stoiber
- Department of Biosciences, University of Salzburg, 5020 Salzburg, Austria; (A.Z.); (W.S.); (J.G.); (P.S.)
| | - Janine Geissenberger
- Department of Biosciences, University of Salzburg, 5020 Salzburg, Austria; (A.Z.); (W.S.); (J.G.); (P.S.)
| | - Peter Steinbacher
- Department of Biosciences, University of Salzburg, 5020 Salzburg, Austria; (A.Z.); (W.S.); (J.G.); (P.S.)
| | - Fabio C. Monticelli
- Department of Forensic Medicine, University of Salzburg, 5020 Salzburg, Austria;
| | - Stefan Pittner
- Department of Forensic Medicine, University of Salzburg, 5020 Salzburg, Austria;
| |
Collapse
|
18
|
Andrés‐Benito P, Gelpi E, Jové M, Mota‐Martorell N, Obis È, Portero‐Otin M, Povedano M, Pujol A, Pamplona R, Ferrer I. Lipid alterations in human frontal cortex in ALS-FTLD-TDP43 proteinopathy spectrum are partly related to peroxisome impairment. Neuropathol Appl Neurobiol 2021; 47:544-563. [PMID: 33332650 PMCID: PMC8248144 DOI: 10.1111/nan.12681] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 10/26/2020] [Accepted: 12/13/2020] [Indexed: 01/08/2023]
Abstract
AIM Peroxisomes play a key role in lipid metabolism, and peroxisome defects have been associated with neurodegenerative diseases such as X-adrenoleukodystrophy and Alzheimer's disease. This study aims to elucidate the contribution of peroxisomes in lipid alterations of area 8 of the frontal cortex in the spectrum of TDP43-proteinopathies. Cases of frontotemporal lobar degeneration-TDP43 (FTLD-TDP), manifested as sporadic (sFTLD-TDP) or linked to mutations in various genes including expansions of the non-coding region of C9ORF72 (c9FTLD), and of sporadic amyotrophic lateral sclerosis (sALS) as the most common TDP43 proteinopathies, were analysed. METHODS We used transcriptomics and lipidomics methods to define the steady-state levels of gene expression and lipid profiles. RESULTS Our results show alterations in gene expression of some components of peroxisomes and related lipid pathways in frontal cortex area 8 in sALS, sFTLD-TDP and c9FTLD. Additionally, we identify a lipidomic pattern associated with the ALS-FTLD-TDP43 proteinopathy spectrum, notably characterised by down-regulation of ether lipids and acylcarnitine among other lipid species, as well as alterations in the lipidome of each phenotype of TDP43 proteinopathy, which reveals commonalities and disease-dependent differences in lipid composition. CONCLUSION Globally, lipid alterations in the human frontal cortex of the ALS-FTLD-TDP43 proteinopathy spectrum, which involve cell membrane composition and signalling, vulnerability against cellular stress and possible glucose metabolism, are partly related to peroxisome impairment.
Collapse
Affiliation(s)
- Pol Andrés‐Benito
- NeuropathologyBellvitge University Hospital‐Bellvitge Biomedical Research Institute (IDIBELLHospitalet de Llobregat, BarcelonaSpain
- Department of Pathology and Experimental TherapeuticsUniversity of BarcelonaBarcelonaSpain
- CIBERNED (Network Centre of Biomedical Research of Neurodegenerative DiseasesInstitute of Health Carlos IIIMinistry of Economy and CompetitivenessMadridSpain
- International Initiative for Treatment and Research Initiative to Cure ALS (TRICALSUtrechtThe Netherlands
| | - Ellen Gelpi
- Neurological Tissue Bank of the Biobanc‐Hospital Clínic‐Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPSBarcelonaSpain
- Institute of NeurologyMedical University of ViennaViennaAustria
| | - Mariona Jové
- Department of Experimental MedicineUniversity of Lleida ‐ Lleida Biomedical Research Institute (UdL‐IRBLleidaLleidaSpain
| | - Natalia Mota‐Martorell
- Department of Experimental MedicineUniversity of Lleida ‐ Lleida Biomedical Research Institute (UdL‐IRBLleidaLleidaSpain
| | - Èlia Obis
- Department of Experimental MedicineUniversity of Lleida ‐ Lleida Biomedical Research Institute (UdL‐IRBLleidaLleidaSpain
| | - Manuel Portero‐Otin
- Department of Experimental MedicineUniversity of Lleida ‐ Lleida Biomedical Research Institute (UdL‐IRBLleidaLleidaSpain
| | - Mònica Povedano
- International Initiative for Treatment and Research Initiative to Cure ALS (TRICALSUtrechtThe Netherlands
- Functional Unit of Amyotrophic Lateral Sclerosis (UFELAService of NeurologyBellvitge University HospitalHospitalet de LlobregatSpain
| | - Aurora Pujol
- Catalan Institution for Research and Advanced Studies (ICREABarcelonaSpain
- Neurometabolic Diseases LaboratoryBellvitge Biomedical Research InstituteHospital Duran i ReynalsHospitalet de Llobregat, BarcelonaSpain
- Center for Biomedical Research on Rare Diseases (CIBERERInstitute of Health Carlos IIIMadridSpain
| | - Reinald Pamplona
- Department of Experimental MedicineUniversity of Lleida ‐ Lleida Biomedical Research Institute (UdL‐IRBLleidaLleidaSpain
| | - Isidro Ferrer
- NeuropathologyBellvitge University Hospital‐Bellvitge Biomedical Research Institute (IDIBELLHospitalet de Llobregat, BarcelonaSpain
- Department of Pathology and Experimental TherapeuticsUniversity of BarcelonaBarcelonaSpain
- CIBERNED (Network Centre of Biomedical Research of Neurodegenerative DiseasesInstitute of Health Carlos IIIMinistry of Economy and CompetitivenessMadridSpain
- International Initiative for Treatment and Research Initiative to Cure ALS (TRICALSUtrechtThe Netherlands
- Institute of NeurosciencesUniversity of BarcelonaBarcelonaSpain
| |
Collapse
|
19
|
Synaptic Reshaping and Neuronal Outcomes in the Temporal Lobe Epilepsy. Int J Mol Sci 2021; 22:ijms22083860. [PMID: 33917911 PMCID: PMC8068229 DOI: 10.3390/ijms22083860] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 04/02/2021] [Accepted: 04/04/2021] [Indexed: 12/11/2022] Open
Abstract
Temporal lobe epilepsy (TLE) is one of the most common types of focal epilepsy, characterized by recurrent spontaneous seizures originating in the temporal lobe(s), with mesial TLE (mTLE) as the worst form of TLE, often associated with hippocampal sclerosis. Abnormal epileptiform discharges are the result, among others, of altered cell-to-cell communication in both chemical and electrical transmissions. Current knowledge about the neurobiology of TLE in human patients emerges from pathological studies of biopsy specimens isolated from the epileptogenic zone or, in a few more recent investigations, from living subjects using positron emission tomography (PET). To overcome limitations related to the use of human tissue, animal models are of great help as they allow the selection of homogeneous samples still presenting a more various scenario of the epileptic syndrome, the presence of a comparable control group, and the availability of a greater amount of tissue for in vitro/ex vivo investigations. This review provides an overview of the structural and functional alterations of synaptic connections in the brain of TLE/mTLE patients and animal models.
Collapse
|
20
|
Dachet F, Brown JB, Valyi-Nagy T, Narayan KD, Serafini A, Boley N, Gingeras TR, Celniker SE, Mohapatra G, Loeb JA. Selective time-dependent changes in activity and cell-specific gene expression in human postmortem brain. Sci Rep 2021; 11:6078. [PMID: 33758256 PMCID: PMC7988150 DOI: 10.1038/s41598-021-85801-6] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Accepted: 02/24/2021] [Indexed: 12/15/2022] Open
Abstract
As a means to understand human neuropsychiatric disorders from human brain samples, we compared the transcription patterns and histological features of postmortem brain to fresh human neocortex isolated immediately following surgical removal. Compared to a number of neuropsychiatric disease-associated postmortem transcriptomes, the fresh human brain transcriptome had an entirely unique transcriptional pattern. To understand this difference, we measured genome-wide transcription as a function of time after fresh tissue removal to mimic the postmortem interval. Within a few hours, a selective reduction in the number of neuronal activity-dependent transcripts occurred with relative preservation of housekeeping genes commonly used as a reference for RNA normalization. Gene clustering indicated a rapid reduction in neuronal gene expression with a reciprocal time-dependent increase in astroglial and microglial gene expression that continued to increase for at least 24 h after tissue resection. Predicted transcriptional changes were confirmed histologically on the same tissue demonstrating that while neurons were degenerating, glial cells underwent an outgrowth of their processes. The rapid loss of neuronal genes and reciprocal expression of glial genes highlights highly dynamic transcriptional and cellular changes that occur during the postmortem interval. Understanding these time-dependent changes in gene expression in post mortem brain samples is critical for the interpretation of research studies on human brain disorders.
Collapse
Affiliation(s)
- Fabien Dachet
- University of Illinois at Chicago, Chicago, IL, 60612, USA.
| | - James B Brown
- Lawrence Berkeley National Laboratory, Berkeley, CA, 94720, USA
| | | | | | - Anna Serafini
- University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Nathan Boley
- University of California, Berkeley, CA, 94720, USA
| | | | | | | | - Jeffrey A Loeb
- University of Illinois at Chicago, Chicago, IL, 60612, USA.
| |
Collapse
|
21
|
Fernandez-Cerado C, Legarda GP, Velasco-Andrada MS, Aguil A, Ganza-Bautista NG, Lagarde JBB, Soria J, Jamora RDG, Acuña PJ, Vanderburg C, Sapp E, DiFiglia M, Murcar MG, Campion L, Ozelius LJ, Alessi AK, Singh-Bains MK, Waldvogel HJ, Faull RLM, Macalintal-Canlas R, Muñoz EL, Penney EB, Ang MA, Diesta CCE, Bragg DC, Acuña-Sunshine G. Promise and challenges of dystonia brain banking: establishing a human tissue repository for studies of X-Linked Dystonia-Parkinsonism. J Neural Transm (Vienna) 2021; 128:575-587. [PMID: 33439365 PMCID: PMC8099813 DOI: 10.1007/s00702-020-02286-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Accepted: 12/01/2020] [Indexed: 01/20/2023]
Abstract
X-Linked Dystonia-Parkinsonism (XDP) is a neurodegenerative disease affecting individuals with ancestry to the island of Panay in the Philippines. In recent years there has been considerable progress at elucidating the genetic basis of XDP and candidate disease mechanisms in patient-derived cellular models, but the neural substrates that give rise to XDP in vivo are still poorly understood. Previous studies of limited XDP postmortem brain samples have reported a selective dropout of medium spiny neurons within the striatum, although neuroimaging of XDP patients has detected additional abnormalities in multiple brain regions beyond the basal ganglia. Given the need to fully define the CNS structures that are affected in this disease, we created a brain bank in Panay to serve as a tissue resource for detailed studies of XDP-related neuropathology. Here we describe this platform, from donor recruitment and consent to tissue collection, processing, and storage, that was assembled within a predominantly rural region of the Philippines with limited access to medical and laboratory facilities. Thirty-six brains from XDP individuals have been collected over an initial 4 years period. Tissue quality was assessed based on histologic staining of cortex, RNA integrity scores, detection of neuronal transcripts in situ by fluorescent hybridization chain reaction, and western blotting of neuronal and glial proteins. The results indicate that this pipeline preserves tissue integrity to an extent compatible with a range of morphologic, molecular, and biochemical analyses. Thus the algorithms that we developed for working in rural communities may serve as a guide for establishing similar brain banks for other rare diseases in indigenous populations.
Collapse
Affiliation(s)
| | - G Paul Legarda
- Sunshine Care Foundation, 5800, Roxas City, Capiz, Philippines
| | | | - Abegail Aguil
- Sunshine Care Foundation, 5800, Roxas City, Capiz, Philippines
| | | | | | - Jasmin Soria
- Sunshine Care Foundation, 5800, Roxas City, Capiz, Philippines
| | - Roland Dominic G Jamora
- Department of Neurosciences, College of Medicine-Philippine General Hospital, University of the Philippines Manila, Manila, Philippines
| | - Patrick J Acuña
- Sunshine Care Foundation, 5800, Roxas City, Capiz, Philippines.,Department of Neurology, The Collaborative Center for X-linked Dystonia-Parkinsonism, Massachusetts General Hospital, Boston, MA, 02129, USA
| | - Charles Vanderburg
- Stanley Center for Psychiatric Research, Broad Institute, Cambridge, MA, 02142, USA
| | - Ellen Sapp
- Department of Neurology, The Collaborative Center for X-linked Dystonia-Parkinsonism, Massachusetts General Hospital, Boston, MA, 02129, USA
| | - Marian DiFiglia
- Department of Neurology, The Collaborative Center for X-linked Dystonia-Parkinsonism, Massachusetts General Hospital, Boston, MA, 02129, USA
| | - Micaela G Murcar
- Department of Neurology, The Collaborative Center for X-linked Dystonia-Parkinsonism, Massachusetts General Hospital, Boston, MA, 02129, USA
| | - Lindsey Campion
- Department of Neurology, The Collaborative Center for X-linked Dystonia-Parkinsonism, Massachusetts General Hospital, Boston, MA, 02129, USA
| | - Laurie J Ozelius
- Department of Neurology, The Collaborative Center for X-linked Dystonia-Parkinsonism, Massachusetts General Hospital, Boston, MA, 02129, USA
| | - Amy K Alessi
- Department of Neurology, The Collaborative Center for X-linked Dystonia-Parkinsonism, Massachusetts General Hospital, Boston, MA, 02129, USA
| | - Malvindar K Singh-Bains
- Department of Anatomy with Medical Imaging, Centre for Brain Research, University of Auckland, Auckland, New Zealand
| | - Henry J Waldvogel
- Department of Anatomy with Medical Imaging, Centre for Brain Research, University of Auckland, Auckland, New Zealand
| | - Richard L M Faull
- Department of Anatomy with Medical Imaging, Centre for Brain Research, University of Auckland, Auckland, New Zealand
| | | | - Edwin L Muñoz
- Department of Pathology, College of Medicine, University of the Philippines, Manila, Philippines
| | - Ellen B Penney
- Department of Neurology, The Collaborative Center for X-linked Dystonia-Parkinsonism, Massachusetts General Hospital, Boston, MA, 02129, USA
| | - Mark A Ang
- Department of Pathology, College of Medicine, University of the Philippines, Manila, Philippines
| | | | - D Cristopher Bragg
- Department of Neurology, The Collaborative Center for X-linked Dystonia-Parkinsonism, Massachusetts General Hospital, Boston, MA, 02129, USA.
| | - Geraldine Acuña-Sunshine
- Sunshine Care Foundation, 5800, Roxas City, Capiz, Philippines. .,Department of Neurology, The Collaborative Center for X-linked Dystonia-Parkinsonism, Massachusetts General Hospital, Boston, MA, 02129, USA.
| |
Collapse
|
22
|
Gibbons AS, Hoyer D, Dean B. SMAD4 protein is decreased in the dorsolateral prefrontal and anterior cingulate cortices in schizophrenia. World J Biol Psychiatry 2021; 22:70-77. [PMID: 32081064 DOI: 10.1080/15622975.2020.1733081] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
OBJECTIVES Signal transduction through the mothers against decapentaplegic (SMAD) is a family of signal transduction factors that mediate signalling of the transforming growth factor B (TGFB)-superfamily of cell regulatory proteins. A recent transcriptomic analysis of post-mortem, cortical tissue from subjects with schizophrenia found decreased mRNA expression of SMAD2 and SMAD4 in the dorsolateral prefrontal cortex (DLPFC) associated with the disorder. To expand this initial finding, we sought to determine whether SMAD2 and SMAD4 protein were also altered in the cortex from subjects with schizophrenia. METHODS Western blotting was used to measure SMAD2 and SMAD4 protein levels in DLPFC and anterior cingulate cortex (ACC) taken post-mortem from subjects with schizophrenia (n = 20) and matched control (n = 20) subjects. RESULTS Compared to controls, levels of SMAD4 were 25% lower in the DLPFC and 38% lower in the ACC from subjects with schizophrenia. By contrast, SMAD2 levels were not altered in either DLPFC or ACC. CONCLUSIONS Our finding of lower SMAD4 protein in the cortex suggests there are likely to be abnormalities in cortical TGFB-superfamily signalling in schizophrenia.
Collapse
Affiliation(s)
- Andrew S Gibbons
- The Florey Institute for Neuroscience and Mental Health, Parkville, Victoria, Australia.,The Department of Psychiatry, Monash University, Clayton, Victoria, Australia
| | - Daniel Hoyer
- The Florey Institute for Neuroscience and Mental Health, Parkville, Victoria, Australia.,Department of Pharmacology and Therapeutics, School of Biomedical Sciences, University of Melbourne, Parkville, Victoria, Australia.,Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA
| | - Brian Dean
- The Florey Institute for Neuroscience and Mental Health, Parkville, Victoria, Australia.,Brain and Psychological Sciences Research Centre, Swinburne University of Technology, Hawthorn, Victoria, Australia
| |
Collapse
|
23
|
Zissler A, Stoiber W, Steinbacher P, Geissenberger J, Monticelli FC, Pittner S. Postmortem Protein Degradation as a Tool to Estimate the PMI: A Systematic Review. Diagnostics (Basel) 2020; 10:E1014. [PMID: 33256203 PMCID: PMC7760775 DOI: 10.3390/diagnostics10121014] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 11/22/2020] [Accepted: 11/23/2020] [Indexed: 12/12/2022] Open
Abstract
Objectives: We provide a systematic review of the literature to evaluate the current research status of protein degradation-based postmortem interval (PMI) estimation. Special attention is paid to the applicability of the proposed approaches/methods in forensic routine practice. Method: A systematic review of the literature on protein degradation in tissues and organs of animals and humans was conducted. Therefore, we searched the scientific databases Pubmed and Ovid for publications until December 2019. Additional searches were performed in Google Scholar and the reference lists of eligible articles. Results: A total of 36 studies were included. This enabled us to consider the degradation pattern of over 130 proteins from 11 different tissues, studied with different methods including well-established and modern approaches. Although comparison between studies is complicated by the heterogeneity of study designs, tissue types, methods, proteins and outcome measurement, there is clear evidence for a high explanatory power of protein degradation analysis in forensic PMI analysis. Conclusions: Although only few approaches have yet exceeded a basic research level, the current research status provides strong evidence in favor of the applicability of a protein degradation-based PMI estimation method in routine forensic practice. Further targeted research effort towards specific aims (also addressing influencing factors and exclusion criteria), especially in human tissue will be required to obtain a robust, reliable laboratory protocol, and collect sufficient data to develop accurate multifactorial mathematical decomposition models.
Collapse
Affiliation(s)
- Angela Zissler
- Department of Biosciences, University of Salzburg, 5020 Salzburg, Austria; (A.Z.); (W.S.); (P.S.); (J.G.)
| | - Walter Stoiber
- Department of Biosciences, University of Salzburg, 5020 Salzburg, Austria; (A.Z.); (W.S.); (P.S.); (J.G.)
| | - Peter Steinbacher
- Department of Biosciences, University of Salzburg, 5020 Salzburg, Austria; (A.Z.); (W.S.); (P.S.); (J.G.)
| | - Janine Geissenberger
- Department of Biosciences, University of Salzburg, 5020 Salzburg, Austria; (A.Z.); (W.S.); (P.S.); (J.G.)
| | - Fabio C. Monticelli
- Department of Forensic Medicine, University of Salzburg, 5020 Salzburg, Austria;
| | - Stefan Pittner
- Department of Forensic Medicine, University of Salzburg, 5020 Salzburg, Austria;
| |
Collapse
|
24
|
Towards animal surrogates for characterising large strain dynamic mechanical properties of human brain tissue. BRAIN MULTIPHYSICS 2020. [DOI: 10.1016/j.brain.2020.100018] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
|
25
|
Sandstrom TS, Burke Schinkel SC, Angel JB. Medical Assistance in Death as a Unique Opportunity to Advance Human Immunodeficiency Virus Cure Research. Clin Infect Dis 2020; 69:1063-1067. [PMID: 30715211 DOI: 10.1093/cid/ciz068] [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: 10/03/2018] [Revised: 11/12/2018] [Accepted: 01/18/2019] [Indexed: 12/31/2022] Open
Abstract
The inability to sample deep-tissue reservoirs in individuals living with human immunodeficiency virus (HIV) has greatly hindered accurate estimates of viral reservoir size and distribution. Animal models and collection of tissues during autopsies of HIV-positive individuals are 2 proposed solutions to this problem. Each, however, has its limitations. In this Viewpoint, we argue that tissue donation following medical assistance in death (MAiD) will form an invaluable resource for the characterization of the viral reservoir in the context of current HIV cure research. In support, we discuss a recent instance in which an individual living with HIV chose to donate their body/tissues to HIV research prior to undergoing MAiD at our institution. Going forward, we hope this will help provide support to individuals in their decisions around tissue donation following MAiD, while highlighting how healthcare providers, by complying with such wishes, can affect patient satisfaction in the last days of life.
Collapse
Affiliation(s)
- Teslin S Sandstrom
- Ottawa Hospital Research Institute.,Department of Biochemistry, Microbiology & Immunology, University of Ottawa
| | | | - Jonathan B Angel
- Ottawa Hospital Research Institute.,Department of Biochemistry, Microbiology & Immunology, University of Ottawa.,Division of Infectious Diseases, Ottawa Hospital-General Campus, Canada
| |
Collapse
|
26
|
Tang K, Wan M, Zhang H, Zhang Q, Yang Q, Chen K, Wang N, Zhang D, Qiu W, Ma C. The top 100 most-cited articles citing human brain banking from 1970 to 2020: a bibliometric analysis. Cell Tissue Bank 2020; 21:685-697. [PMID: 32761559 DOI: 10.1007/s10561-020-09849-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2020] [Accepted: 07/09/2020] [Indexed: 02/08/2023]
Abstract
Many articles involving human brain banks have been published. Bibliometric analysis can determine the history of the development of research and future research trends in a specific field. Three independent researchers retrieved and reviewed articles from the Web of Science database using the following strategy: "TS = (((brain OR cerebral) AND (bank* OR biobank*)) OR brainbank*)." The top 100 most-cited articles were identified and listed in descending order by total citations. Web of Science was used to identify ten recent articles describing bank construction. GeenMedical ( https://www.geenmedical.com/ ) was used to identify ten recent articles from journals with an impact factor (IF) > 20. The top 100 most-cited articles citing human brain banks were published between 1991 and 2017. Fifty-two percent of the articles focused on a specific type of neurodegenerative disease, and 16% discussed the construction and development of human brain banks. Articles using brain tissue had more total and annual citations than those on bank construction. Ten articles with high IFs were published from 2017 to 2019, and they were primarily studies using novel research techniques such RNA sequencing and genome-wide association studies. Most studies were published in journals specializing in neurology or neuroscience such as Movement Disorders (10%), and had been conducted in the United States (52%) by neurologists (62%). The top 100 most-cited articles and recent publications citing human brain banks and their bibliometric characteristics were identified and analyzed, which may serve as a useful reference and pave the way for further research.
Collapse
Affiliation(s)
- Keyun Tang
- Department of Human Anatomy, Histology and Embryology, Institute of Basic Medical Sciences, Neuroscience Center, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China.,Eight-Year MD Program, Peking Union Medical College, Beijing, China
| | - Mengyao Wan
- Department of Human Anatomy, Histology and Embryology, Institute of Basic Medical Sciences, Neuroscience Center, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China.,Eight-Year MD Program, Peking Union Medical College, Beijing, China
| | - Hanlin Zhang
- Department of Human Anatomy, Histology and Embryology, Institute of Basic Medical Sciences, Neuroscience Center, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China.,Eight-Year MD Program, Peking Union Medical College, Beijing, China
| | - Qing Zhang
- Department of Human Anatomy, Histology and Embryology, Institute of Basic Medical Sciences, Neuroscience Center, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Qian Yang
- Department of Human Anatomy, Histology and Embryology, Institute of Basic Medical Sciences, Neuroscience Center, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Kang Chen
- Department of Human Anatomy, Histology and Embryology, Institute of Basic Medical Sciences, Neuroscience Center, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China.,Eight-Year MD Program, Peking Union Medical College, Beijing, China
| | - Naili Wang
- Department of Human Anatomy, Histology and Embryology, Institute of Basic Medical Sciences, Neuroscience Center, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China.,National Experimental Demonstration Center of Basic Medicine, Peking Union Medical College, Beijing, China
| | - Di Zhang
- Department of Human Anatomy, Histology and Embryology, Institute of Basic Medical Sciences, Neuroscience Center, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China.,National Experimental Demonstration Center of Basic Medicine, Peking Union Medical College, Beijing, China
| | - Wenying Qiu
- Department of Human Anatomy, Histology and Embryology, Institute of Basic Medical Sciences, Neuroscience Center, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China.
| | - Chao Ma
- Department of Human Anatomy, Histology and Embryology, Institute of Basic Medical Sciences, Neuroscience Center, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China. .,Joint Laboratory of Anesthesia and Pain, Peking Union Medical College, Beijing, China.
| |
Collapse
|
27
|
Esteves AR, Cardoso SM. Differential protein expression in diverse brain areas of Parkinson's and Alzheimer's disease patients. Sci Rep 2020; 10:13149. [PMID: 32753661 PMCID: PMC7403590 DOI: 10.1038/s41598-020-70174-z] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Accepted: 07/10/2020] [Indexed: 11/09/2022] Open
Abstract
Many hypotheses have been postulated to define the etiology of sporadic Parkinson's and Alzheimer's disorders (PD and AD) but there is no consensus on what causes these devastating age-related diseases. Braak staging of both pathologies helped researchers to better understand the progression and to identify their prodromal and symptomatic phases. Indeed, it is well accepted that Lewy body pathology and neurofibrillary tangles appearance correlates with disease progression and severity of symptoms in PD and AD, respectively. Additionally, several studies in PD and AD models try to disclose which cellular mechanisms are defaulted and trigger the neurodegenerative process that culminates with neuronal death causing PD and AD classical symptomatology. Herein, we determined expression levels of proteins involved in microtubule assembly, autophagic-lysosomal pathway and unfolded protein response in the cortex, hippocampus and SNpc of PD and AD patients, vascular dementia patients and aged-match controls. The differential expression allowed us to determine which pathways are determinant to synaptic dysfunction and to establish a time line for disease progression. Our results allow us to challenge the hypothesis that both PD and AD pathologies are caused by α-synuclein or Aβ pathology propagation throughout the brain in a prion-like manner.
Collapse
Affiliation(s)
- A R Esteves
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Largo Marquês de Pombal, 3004-517, Coimbra, Portugal.,CIBB-Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
| | - S M Cardoso
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Largo Marquês de Pombal, 3004-517, Coimbra, Portugal. .,CIBB-Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal. .,Institute of Cellular and Molecular Biology, Faculty of Medicine, University of Coimbra, Coimbra, Portugal.
| |
Collapse
|
28
|
Hildebrand S, Schueth A, Wangenheim KV, Mattheyer C, Pampaloni F, Bratzke H, Roebroeck AF, Galuske RAW. hFRUIT: An optimized agent for optical clearing of DiI-stained adult human brain tissue. Sci Rep 2020; 10:9950. [PMID: 32561795 PMCID: PMC7305111 DOI: 10.1038/s41598-020-66999-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Accepted: 05/28/2020] [Indexed: 11/09/2022] Open
Abstract
Here, we describe a new immersion-based clearing method suitable for optical clearing of thick adult human brain samples while preserving its lipids and lipophilic labels such as 1,1'-dioctadecyl-3,3,3',3'-tetramethylindocarbocyanine perchlorate (DiI). This clearing procedure is simple, easy to implement, and allowed for clearing of 5 mm thick human brain tissue samples within 12 days. Furthermore, we show for the first time the advantageous effect of the Periodate-Lysine-Paraformaldehyde (PLP) fixation as compared to the more commonly used 4% paraformaldehyde (PFA) on clearing performance.
Collapse
Affiliation(s)
- Sven Hildebrand
- Department of Cognitive Neuroscience, Faculty of Psychology & Neuroscience, Maastricht University, 6229 EV, Maastricht, the Netherlands.
| | - Anna Schueth
- Department of Cognitive Neuroscience, Faculty of Psychology & Neuroscience, Maastricht University, 6229 EV, Maastricht, the Netherlands
| | - Klaus von Wangenheim
- Department of Systemic Neurophysiology, Technische Universität Darmstadt, Schnittspahnstraße 3, Darmstadt, 64287, Germany
| | - Christian Mattheyer
- Johann Wolfgang Goethe-Universität Frankfurt am Main, Faculty 15 Biological Sciences, Institute for Cell Biology and Neuroscience, Buchmann Institute for Molecular Life Sciences, Max-von-Laue-Str. 15, Frankfurt am Main, D-60438, Germany
| | - Francesco Pampaloni
- Johann Wolfgang Goethe-Universität Frankfurt am Main, Faculty 15 Biological Sciences, Institute for Cell Biology and Neuroscience, Buchmann Institute for Molecular Life Sciences, Max-von-Laue-Str. 15, Frankfurt am Main, D-60438, Germany
| | - Hansjürgen Bratzke
- Johann Wolfgang Goethe-Universität Frankfurt am Main, Institute for Forensic Medicine, Kennedy Allee 104, 60596, Frankfurt am Main, Germany
| | - Alard F Roebroeck
- Department of Cognitive Neuroscience, Faculty of Psychology & Neuroscience, Maastricht University, 6229 EV, Maastricht, the Netherlands
| | - Ralf A W Galuske
- Department of Systemic Neurophysiology, Technische Universität Darmstadt, Schnittspahnstraße 3, Darmstadt, 64287, Germany.
| |
Collapse
|
29
|
Synaptic Loss, ER Stress and Neuro-Inflammation Emerge Late in the Lateral Temporal Cortex and Associate with Progressive Tau Pathology in Alzheimer's Disease. Mol Neurobiol 2020; 57:3258-3272. [PMID: 32514860 PMCID: PMC7340653 DOI: 10.1007/s12035-020-01950-1] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Accepted: 05/22/2020] [Indexed: 11/17/2022]
Abstract
The complex multifactorial nature of AD pathogenesis has been highlighted by evidence implicating additional neurodegenerative mechanisms, beyond that of amyloid-β (Aβ) and tau. To provide insight into cause and effect, we here investigated the temporal profile and associations of pathological changes in synaptic, endoplasmic reticulum (ER) stress and neuro-inflammatory markers. Quantifications were established via immunoblot and immunohistochemistry protocols in post-mortem lateral temporal cortex (n = 46). All measures were assessed according to diagnosis (non-AD vs. AD), neuropathological severity (low (Braak ≤ 2) vs. moderate (3–4) vs. severe (≥ 5)) and individual Braak stage, and were correlated with Aβ and tau pathology and cognitive scores. Postsynaptic PSD-95, but not presynaptic synaptophysin, was decreased in AD cases and demonstrated a progressive decline across disease severity and Braak stage, yet not with cognitive scores. Of all investigated ER stress markers, only phospho-protein kinase RNA-like ER kinase (p-PERK) correlated with Braak stage and was increased in diagnosed AD cases. A similar relationship was observed for the astrocytic glial fibrillary acidic protein (GFAP); however, the associated aquaporin 4 and microglial Iba1 remained unchanged. Pathological alterations in these markers preferentially correlated with measures of tau over those related to Aβ. Notably, GFAP also correlated strongly with Aβ markers and with all assessments of cognition. Lateral temporal cortex-associated synaptic, ER stress and neuro-inflammatory pathologies are here determined as late occurrences in AD progression, largely associated with tau pathology. Moreover, GFAP emerged as the most robust indicator of disease progression, tau/Aβ pathology, and cognitive impairment.
Collapse
|
30
|
Parkin GM, Gibbons A, Udawela M, Dean B. Excitatory amino acid transporter (EAAT)1 and EAAT2 mRNA levels are altered in the prefrontal cortex of subjects with schizophrenia. J Psychiatr Res 2020; 123:151-158. [PMID: 32065951 DOI: 10.1016/j.jpsychires.2020.02.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 02/06/2020] [Accepted: 02/07/2020] [Indexed: 12/21/2022]
Abstract
Excitatory amino acid transporter (EAAT)1 and EAAT2 mediate glutamatergic neurotransmission and prevent excitotoxicity through binding and transportation of glutamate into glia. These EAATs may be regulated by metabotropic glutamate receptor 5 (mGluR5), which is also expressed by glia. Whilst we have data from an Affymetrix™ Human Exon 1.0 ST Array showing higher levels of EAAT1 mRNA (+36%) in Brodmann's are (BA)9 of subjects with schizophrenia, there is evidence that EAAT1 and EAAT2, as well as mGluR5 levels, are altered in the cortex of subjects with the disorder. Hence, we measured mRNA levels of these genes in other cortical regions in subjects with that disorder. EAAT1, EAAT2 and mGluR5 mRNA were measured, in triplicate, using Quantitative PCR in BA10 and BA46 from subjects with schizophrenia (n = 20) and age and sex matched controls (n = 18). Levels of mRNA were normalised to the geometric mean of two reference genes, transcription factor B1, mitochondrial (TFB1M) and S-phase kinase-associated protein 1A (SKP1A), for which mRNA did not vary between diagnostic groups in either region. Normalised levels of EAAT1 and EAAT2 mRNA were significantly higher in BA10 (EAAT1: U = 58, p = 0.0002; EAAT2 U = 70, p = 0.0009), but not BA46 (EAAT1: U = 122, p = 0.09; EAAT2: U = 136, p = 0.21), from subjects with schizophrenia compared to controls. mGluR5 levels in BA10 (U = 173, p=0.85) and BA46 (U = 178, p = 0.96) did not vary by cohort. Our data suggests that region-specific increases in cortical EAAT1 and EAAT2 mRNA are involved in schizophrenia pathophysiology and that disrupted glutamate uptake in schizophrenia may be of particular significance in BA10.
Collapse
Affiliation(s)
- Georgia M Parkin
- The Molecular Psychiatry Laboratory, The Florey Institute for Neuroscience and Mental Health, Parkville, Victoria, Australia; The Cooperative Research Centre for Mental Health, Parkville, Victoria, Australia.
| | - Andrew Gibbons
- The Molecular Psychiatry Laboratory, The Florey Institute for Neuroscience and Mental Health, Parkville, Victoria, Australia
| | - Madhara Udawela
- The Molecular Psychiatry Laboratory, The Florey Institute for Neuroscience and Mental Health, Parkville, Victoria, Australia; The Cooperative Research Centre for Mental Health, Parkville, Victoria, Australia
| | - Brian Dean
- The Molecular Psychiatry Laboratory, The Florey Institute for Neuroscience and Mental Health, Parkville, Victoria, Australia; The Cooperative Research Centre for Mental Health, Parkville, Victoria, Australia; The Centre for Mental Health, The Faculty of Health, Arts and Design, Swinburne University, Hawthorne, Victoria, Australia
| |
Collapse
|
31
|
Dean B, Tsatsanis A, Lam LQ, Scarr E, Duce JA. Changes in cortical protein markers of iron transport with gender, major depressive disorder and suicide. World J Biol Psychiatry 2020; 21:119-126. [PMID: 30513246 DOI: 10.1080/15622975.2018.1555377] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Objectives: The objective of this study was to determine whether a breakdown in proteins regulating cortical iron homeostasis could be involved in the pathophysiology of mood disorders.Methods: Levels of select proteins responsible for cortical iron transport were quantitated by Western blotting of Brodmann's (BA) areas 6 and 10 from patients with major depressive disorder (n = 13), bipolar disorder (n = 12) and age/sex matched controls (n = 13).Results: We found the inactive form of ceruloplasmin was lower in BA 6 from males compared to females. Levels of copper containing ceruloplasmin was lower in BA 6 from suicide completers whilst levels of amyloid precursor protein, TAU and transferrin were higher in BA 10 from those individuals. The level of prion protein was lower in BA 6 from subjects with major depressive disorder.Conclusions: Our data suggests that perturbation in cortical iron transport proteins is not prevalent in mood disorders. By contrast, our data suggests changes in iron transport proteins in BA 6 and BA 10 are present after suicide completion. If these changes were present before death, they could have had a role in the genesis of the contemplation and completion of suicide.
Collapse
Affiliation(s)
- Brian Dean
- The Molecular Psychiatry Laboratory, The Florey Institute of Neuroscience and Mental Health, Parkville, Australia.,The Centre for Mental Health, the Faculty of Health, Arts and Design, Swinburne University, Hawthorne, Australia
| | - Andrew Tsatsanis
- Melbourne Dementia Research Centre, The Florey Institute of Neuroscience and Mental Health, Parkville, Australia.,School of Biomedical Sciences, University of Leeds, Leeds, UK
| | - Linh Q Lam
- Melbourne Dementia Research Centre, The Florey Institute of Neuroscience and Mental Health, Parkville, Australia.,Neuropharmacology Laboratory, Department of Pharmacology and Therapeutics, The University of Melbourne, Parkville, Australia
| | - Elizabeth Scarr
- The Molecular Psychiatry Laboratory, The Florey Institute of Neuroscience and Mental Health, Parkville, Australia.,Melbourne Veterinary School, Faculty of Veterinary and Agricultural Sciences, The University of Melbourne, Parkville, Australia
| | - James A Duce
- Melbourne Dementia Research Centre, The Florey Institute of Neuroscience and Mental Health, Parkville, Australia.,School of Biomedical Sciences, University of Leeds, Leeds, UK.,The ALBORADO Drug Discovery Institute, University of Cambridge, Cambridge, UK
| |
Collapse
|
32
|
Dean B, Parkin GM, Gibbons AS. Associations between catechol-O-methyltransferase (COMT) genotypes at rs4818 and rs4680 and gene expression in human dorsolateral prefrontal cortex. Exp Brain Res 2020; 238:477-486. [DOI: 10.1007/s00221-020-05730-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Accepted: 01/07/2020] [Indexed: 12/28/2022]
|
33
|
Esteva-Socias M, Artiga MJ, Bahamonde O, Belar O, Bermudo R, Castro E, Escámez T, Fraga M, Jauregui-Mosquera L, Novoa I, Peiró-Chova L, Rejón JD, Ruiz-Miró M, Vieiro-Balo P, Villar-Campo V, Zazo S, Rábano A, Villena C. In search of an evidence-based strategy for quality assessment of human tissue samples: report of the tissue Biospecimen Research Working Group of the Spanish Biobank Network. J Transl Med 2019; 17:370. [PMID: 31718661 PMCID: PMC6852937 DOI: 10.1186/s12967-019-2124-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Accepted: 11/01/2019] [Indexed: 01/10/2023] Open
Abstract
The purpose of the present work is to underline the importance of obtaining a standardized procedure to ensure and evaluate both clinical and research usability of human tissue samples. The study, which was carried out by the Biospecimen Science Working Group of the Spanish Biobank Network, is based on a general overview of the current situation about quality assurance in human tissue biospecimens. It was conducted an exhaustive review of the analytical techniques used to evaluate the quality of human tissue samples over the past 30 years, as well as their reference values if they were published, and classified them according to the biomolecules evaluated: (i) DNA, (ii) RNA, and (iii) soluble or/and fixed proteins for immunochemistry. More than 130 publications released between 1989 and 2019 were analysed, most of them reporting results focused on the analysis of tumour and biopsy samples. A quality assessment proposal with an algorithm has been developed for both frozen tissue samples and formalin-fixed paraffin-embedded (FFPE) samples, according to the expected quality of sample based on the available pre-analytical information and the experience of the participants in the Working Group. The high heterogeneity of human tissue samples and the wide number of pre-analytic factors associated to quality of samples makes it very difficult to harmonize the quality criteria. However, the proposed method to assess human tissue sample integrity and antigenicity will not only help to evaluate whether stored human tissue samples fit for the purpose of biomarker development, but will also allow to perform further studies, such as assessing the impact of different pre-analytical factors on very well characterized samples or evaluating the readjustment of tissue sample collection, processing and storing procedures. By ensuring the quality of the samples used on research, the reproducibility of scientific results will be guaranteed.
Collapse
Affiliation(s)
- Margalida Esteva-Socias
- Centro de Investigación Biomédica en Red Respiratory Diseases (CIBERES), Plataforma Biobanco Pulmonar CIBERES, Hospital Universitari Son Espases, Palma, Spain.,Grupo de Inflamación, reparación y cáncer en enfermedades respiratorias, Institut d'Investigació Sanitària de les Illes Balears (IdISBa), Hospital Universitari Son Espases, Palma, Spain
| | | | | | - Oihana Belar
- Basque Foundation for Health Innovation and Research, Basque Biobank, Barakaldo, Spain
| | - Raquel Bermudo
- Hospital Clínic-IDIBAPS Biobank, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Erika Castro
- Basque Foundation for Health Innovation and Research, Basque Biobank, Barakaldo, Spain
| | - Teresa Escámez
- IMIB Biobank, Instituto Murciano de Investigación Biosanitaria, Murcia, Spain
| | - Máximo Fraga
- Depto. de Ciencias Forenses, Anatomía Patolóxica, Xinecología e Obstetricia, e Pediatría, Facultade de Medicina, Universidade de Santiago de Compostela (USC), Santiago, Spain.,Biobanco Complejo Hospitalario Universitario de Santiago de Compostela (CHUS), SERGAS, Santiago, Spain
| | | | - Isabel Novoa
- Vall d'Hebron University Hospital Biobank, Vall d'Hebron Hospital Research Institute, Barcelona, Spain
| | | | - Juan-David Rejón
- Biobanco del Sistema Sanitario Público de Andalucía, Granada, Spain
| | - María Ruiz-Miró
- IRBLleida Biobank, Instituto de Investigaciones Biomédica de Lleida-Fundación Dr. Pifarre, Lérida, Spain
| | - Paula Vieiro-Balo
- Biobanco Complejo Hospitalario Universitario de Santiago de Compostela (CHUS), SERGAS, Santiago, Spain
| | | | - Sandra Zazo
- Department of Pathology, IIS-Fundación Jiménez Díaz, Madrid, Spain
| | - Alberto Rábano
- Banco de Tejidos, Fundación CIEN, Instituto de Salud Carlos III, Madrid, Spain
| | - Cristina Villena
- Centro de Investigación Biomédica en Red Respiratory Diseases (CIBERES), Plataforma Biobanco Pulmonar CIBERES, Hospital Universitari Son Espases, Palma, Spain. .,Grupo de Inflamación, reparación y cáncer en enfermedades respiratorias, Institut d'Investigació Sanitària de les Illes Balears (IdISBa), Hospital Universitari Son Espases, Palma, Spain.
| |
Collapse
|
34
|
Ramos DM, d’Ydewalle C, Gabbeta V, Dakka A, Klein SK, Norris DA, Matson J, Taylor SJ, Zaworski PG, Prior TW, Snyder PJ, Valdivia D, Hatem CL, Waters I, Gupte N, Swoboda KJ, Rigo F, Bennett CF, Naryshkin N, Paushkin S, Crawford TO, Sumner CJ. Age-dependent SMN expression in disease-relevant tissue and implications for SMA treatment. J Clin Invest 2019; 129:4817-4831. [PMID: 31589162 PMCID: PMC6819103 DOI: 10.1172/jci124120] [Citation(s) in RCA: 123] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Accepted: 08/07/2019] [Indexed: 12/11/2022] Open
Abstract
BACKGROUNDSpinal muscular atrophy (SMA) is caused by deficient expression of survival motor neuron (SMN) protein. New SMN-enhancing therapeutics are associated with variable clinical benefits. Limited knowledge of baseline and drug-induced SMN levels in disease-relevant tissues hinders efforts to optimize these treatments.METHODSSMN mRNA and protein levels were quantified in human tissues isolated during expedited autopsies.RESULTSSMN protein expression varied broadly among prenatal control spinal cord samples, but was restricted at relatively low levels in controls and SMA patients after 3 months of life. A 2.3-fold perinatal decrease in median SMN protein levels was not paralleled by comparable changes in SMN mRNA. In tissues isolated from nusinersen-treated SMA patients, antisense oligonucleotide (ASO) concentration and full-length (exon 7 including) SMN2 (SMN2-FL) mRNA level increases were highest in lumbar and thoracic spinal cord. An increased number of cells showed SMN immunolabeling in spinal cord of treated patients, but was not associated with an increase in whole-tissue SMN protein levels.CONCLUSIONSA normally occurring perinatal decrease in whole-tissue SMN protein levels supports efforts to initiate SMN-inducing therapies as soon after birth as possible. Limited ASO distribution to rostral spinal and brain regions in some patients likely limits clinical response of motor units in these regions for those patients. These results have important implications for optimizing treatment of SMA patients and warrant further investigations to enhance bioavailability of intrathecally administered ASOs.FUNDINGSMA Foundation, SMART, NIH (R01-NS096770, R01-NS062869), Ionis Pharmaceuticals, and PTC Therapeutics. Biogen provided support for absolute real-time RT-PCR.
Collapse
Affiliation(s)
| | - Constantin d’Ydewalle
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | | | - Amal Dakka
- PTC Therapeutics, South Plainfield, New Jersey, USA
| | | | | | - John Matson
- Ionis Pharmaceuticals, Carlsbad, California, USA
| | | | | | - Thomas W. Prior
- Center for Human Genetics, Case Western Reserve University, Cleveland, Ohio, USA
| | - Pamela J. Snyder
- Department of Pathology, Ohio State University, Columbus, Ohio, USA
| | - David Valdivia
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Christine L. Hatem
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Ian Waters
- The Sidney Kimmel Comprehensive Cancer Center, Department of Oncology, and
| | - Nikhil Gupte
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Kathryn J. Swoboda
- Center for Genomic Medicine, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Frank Rigo
- Ionis Pharmaceuticals, Carlsbad, California, USA
| | | | | | | | - Thomas O. Crawford
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Charlotte J. Sumner
- Department of Neuroscience and
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
35
|
Nucifora LG, MacDonald ML, Lee BJ, Peters ME, Norris AL, Orsburn BC, Yang K, Gleason K, Margolis RL, Pevsner J, Tamminga CA, Sweet RA, Ross CA, Sawa A, Nucifora FC. Increased Protein Insolubility in Brains From a Subset of Patients With Schizophrenia. Am J Psychiatry 2019; 176:730-743. [PMID: 31055969 DOI: 10.1176/appi.ajp.2019.18070864] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
OBJECTIVE The mechanisms leading to schizophrenia are likely to be diverse. However, there may be common pathophysiological pathways for subtypes of the disease. The authors tested the hypothesis that increased protein insolubility and ubiquitination underlie the pathophysiology for a subtype of schizophrenia. METHODS Prefrontal cortex and superior temporal gyrus from postmortem brains of individuals with and without schizophrenia were subjected to cold sarkosyl fractionation, separating proteins into soluble and insoluble fractions. Protein insolubility and ubiquitin levels were quantified for each insoluble fraction, with normalization to total homogenate protein. Mass spectrometry analysis was then performed to identify the protein contents of the insoluble fractions. The potential biological relevance of the detected proteins was assessed using Gene Ontology enrichment analysis and Ingenuity Pathway Analysis. RESULTS A subset of the schizophrenia brains showed an increase in protein insolubility and ubiquitination in the insoluble fraction. Mass spectrometry of the insoluble fraction revealed that brains with increased insolubility and ubiquitination exhibited a similar peptide expression by principal component analysis. The proteins that were significantly altered in the insoluble fraction were enriched for pathways relating to axon target recognition as well as nervous system development and function. CONCLUSIONS This study suggests a pathological process related to protein insolubility for a subset of patients with schizophrenia. Determining the molecular mechanism of this subtype of schizophrenia could lead to a better understanding of the pathways underlying the clinical phenotype in some patients with major mental illness as well as to improved nosology and identification of novel therapeutic targets.
Collapse
Affiliation(s)
- Leslie G Nucifora
- The Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore (L.G. Nucifora, Lee, Peters, Yang, Margolis, Pevsner, Ross, Sawa, F.C. Nucifora); the Departments of Psychiatry and Neurology, University of Pittsburgh, and the VISN 4 Mental Illness Research, Education, and Clinical Center, VA Pittsburgh Healthcare System, Pittsburgh (MacDonald, Sweet); the Department of Neurology, Kennedy Krieger Institute, Baltimore (Norris, Pevsner); the Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore (Norris, Pevsner, Ross, Sawa); Protein Characterization Laboratory, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Frederick, Md. (Orsburn); the Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas (Gleason, Tamminga); the Department of Neurology, Johns Hopkins University School of Medicine, Baltimore (Margolis, Ross, Sawa, F.C. Nucifora); Program in Cellular and Molecular Medicine, Johns Hopkins University School of Medicine, Baltimore (Lee, Sawa); the Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore (Ross)
| | - Matthew L MacDonald
- The Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore (L.G. Nucifora, Lee, Peters, Yang, Margolis, Pevsner, Ross, Sawa, F.C. Nucifora); the Departments of Psychiatry and Neurology, University of Pittsburgh, and the VISN 4 Mental Illness Research, Education, and Clinical Center, VA Pittsburgh Healthcare System, Pittsburgh (MacDonald, Sweet); the Department of Neurology, Kennedy Krieger Institute, Baltimore (Norris, Pevsner); the Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore (Norris, Pevsner, Ross, Sawa); Protein Characterization Laboratory, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Frederick, Md. (Orsburn); the Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas (Gleason, Tamminga); the Department of Neurology, Johns Hopkins University School of Medicine, Baltimore (Margolis, Ross, Sawa, F.C. Nucifora); Program in Cellular and Molecular Medicine, Johns Hopkins University School of Medicine, Baltimore (Lee, Sawa); the Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore (Ross)
| | - Brian J Lee
- The Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore (L.G. Nucifora, Lee, Peters, Yang, Margolis, Pevsner, Ross, Sawa, F.C. Nucifora); the Departments of Psychiatry and Neurology, University of Pittsburgh, and the VISN 4 Mental Illness Research, Education, and Clinical Center, VA Pittsburgh Healthcare System, Pittsburgh (MacDonald, Sweet); the Department of Neurology, Kennedy Krieger Institute, Baltimore (Norris, Pevsner); the Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore (Norris, Pevsner, Ross, Sawa); Protein Characterization Laboratory, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Frederick, Md. (Orsburn); the Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas (Gleason, Tamminga); the Department of Neurology, Johns Hopkins University School of Medicine, Baltimore (Margolis, Ross, Sawa, F.C. Nucifora); Program in Cellular and Molecular Medicine, Johns Hopkins University School of Medicine, Baltimore (Lee, Sawa); the Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore (Ross)
| | - Matthew E Peters
- The Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore (L.G. Nucifora, Lee, Peters, Yang, Margolis, Pevsner, Ross, Sawa, F.C. Nucifora); the Departments of Psychiatry and Neurology, University of Pittsburgh, and the VISN 4 Mental Illness Research, Education, and Clinical Center, VA Pittsburgh Healthcare System, Pittsburgh (MacDonald, Sweet); the Department of Neurology, Kennedy Krieger Institute, Baltimore (Norris, Pevsner); the Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore (Norris, Pevsner, Ross, Sawa); Protein Characterization Laboratory, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Frederick, Md. (Orsburn); the Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas (Gleason, Tamminga); the Department of Neurology, Johns Hopkins University School of Medicine, Baltimore (Margolis, Ross, Sawa, F.C. Nucifora); Program in Cellular and Molecular Medicine, Johns Hopkins University School of Medicine, Baltimore (Lee, Sawa); the Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore (Ross)
| | - Alexis L Norris
- The Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore (L.G. Nucifora, Lee, Peters, Yang, Margolis, Pevsner, Ross, Sawa, F.C. Nucifora); the Departments of Psychiatry and Neurology, University of Pittsburgh, and the VISN 4 Mental Illness Research, Education, and Clinical Center, VA Pittsburgh Healthcare System, Pittsburgh (MacDonald, Sweet); the Department of Neurology, Kennedy Krieger Institute, Baltimore (Norris, Pevsner); the Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore (Norris, Pevsner, Ross, Sawa); Protein Characterization Laboratory, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Frederick, Md. (Orsburn); the Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas (Gleason, Tamminga); the Department of Neurology, Johns Hopkins University School of Medicine, Baltimore (Margolis, Ross, Sawa, F.C. Nucifora); Program in Cellular and Molecular Medicine, Johns Hopkins University School of Medicine, Baltimore (Lee, Sawa); the Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore (Ross)
| | - Benjamin C Orsburn
- The Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore (L.G. Nucifora, Lee, Peters, Yang, Margolis, Pevsner, Ross, Sawa, F.C. Nucifora); the Departments of Psychiatry and Neurology, University of Pittsburgh, and the VISN 4 Mental Illness Research, Education, and Clinical Center, VA Pittsburgh Healthcare System, Pittsburgh (MacDonald, Sweet); the Department of Neurology, Kennedy Krieger Institute, Baltimore (Norris, Pevsner); the Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore (Norris, Pevsner, Ross, Sawa); Protein Characterization Laboratory, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Frederick, Md. (Orsburn); the Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas (Gleason, Tamminga); the Department of Neurology, Johns Hopkins University School of Medicine, Baltimore (Margolis, Ross, Sawa, F.C. Nucifora); Program in Cellular and Molecular Medicine, Johns Hopkins University School of Medicine, Baltimore (Lee, Sawa); the Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore (Ross)
| | - Kun Yang
- The Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore (L.G. Nucifora, Lee, Peters, Yang, Margolis, Pevsner, Ross, Sawa, F.C. Nucifora); the Departments of Psychiatry and Neurology, University of Pittsburgh, and the VISN 4 Mental Illness Research, Education, and Clinical Center, VA Pittsburgh Healthcare System, Pittsburgh (MacDonald, Sweet); the Department of Neurology, Kennedy Krieger Institute, Baltimore (Norris, Pevsner); the Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore (Norris, Pevsner, Ross, Sawa); Protein Characterization Laboratory, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Frederick, Md. (Orsburn); the Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas (Gleason, Tamminga); the Department of Neurology, Johns Hopkins University School of Medicine, Baltimore (Margolis, Ross, Sawa, F.C. Nucifora); Program in Cellular and Molecular Medicine, Johns Hopkins University School of Medicine, Baltimore (Lee, Sawa); the Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore (Ross)
| | - Kelly Gleason
- The Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore (L.G. Nucifora, Lee, Peters, Yang, Margolis, Pevsner, Ross, Sawa, F.C. Nucifora); the Departments of Psychiatry and Neurology, University of Pittsburgh, and the VISN 4 Mental Illness Research, Education, and Clinical Center, VA Pittsburgh Healthcare System, Pittsburgh (MacDonald, Sweet); the Department of Neurology, Kennedy Krieger Institute, Baltimore (Norris, Pevsner); the Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore (Norris, Pevsner, Ross, Sawa); Protein Characterization Laboratory, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Frederick, Md. (Orsburn); the Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas (Gleason, Tamminga); the Department of Neurology, Johns Hopkins University School of Medicine, Baltimore (Margolis, Ross, Sawa, F.C. Nucifora); Program in Cellular and Molecular Medicine, Johns Hopkins University School of Medicine, Baltimore (Lee, Sawa); the Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore (Ross)
| | - Russell L Margolis
- The Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore (L.G. Nucifora, Lee, Peters, Yang, Margolis, Pevsner, Ross, Sawa, F.C. Nucifora); the Departments of Psychiatry and Neurology, University of Pittsburgh, and the VISN 4 Mental Illness Research, Education, and Clinical Center, VA Pittsburgh Healthcare System, Pittsburgh (MacDonald, Sweet); the Department of Neurology, Kennedy Krieger Institute, Baltimore (Norris, Pevsner); the Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore (Norris, Pevsner, Ross, Sawa); Protein Characterization Laboratory, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Frederick, Md. (Orsburn); the Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas (Gleason, Tamminga); the Department of Neurology, Johns Hopkins University School of Medicine, Baltimore (Margolis, Ross, Sawa, F.C. Nucifora); Program in Cellular and Molecular Medicine, Johns Hopkins University School of Medicine, Baltimore (Lee, Sawa); the Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore (Ross)
| | - Jonathan Pevsner
- The Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore (L.G. Nucifora, Lee, Peters, Yang, Margolis, Pevsner, Ross, Sawa, F.C. Nucifora); the Departments of Psychiatry and Neurology, University of Pittsburgh, and the VISN 4 Mental Illness Research, Education, and Clinical Center, VA Pittsburgh Healthcare System, Pittsburgh (MacDonald, Sweet); the Department of Neurology, Kennedy Krieger Institute, Baltimore (Norris, Pevsner); the Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore (Norris, Pevsner, Ross, Sawa); Protein Characterization Laboratory, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Frederick, Md. (Orsburn); the Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas (Gleason, Tamminga); the Department of Neurology, Johns Hopkins University School of Medicine, Baltimore (Margolis, Ross, Sawa, F.C. Nucifora); Program in Cellular and Molecular Medicine, Johns Hopkins University School of Medicine, Baltimore (Lee, Sawa); the Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore (Ross)
| | - Carol A Tamminga
- The Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore (L.G. Nucifora, Lee, Peters, Yang, Margolis, Pevsner, Ross, Sawa, F.C. Nucifora); the Departments of Psychiatry and Neurology, University of Pittsburgh, and the VISN 4 Mental Illness Research, Education, and Clinical Center, VA Pittsburgh Healthcare System, Pittsburgh (MacDonald, Sweet); the Department of Neurology, Kennedy Krieger Institute, Baltimore (Norris, Pevsner); the Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore (Norris, Pevsner, Ross, Sawa); Protein Characterization Laboratory, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Frederick, Md. (Orsburn); the Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas (Gleason, Tamminga); the Department of Neurology, Johns Hopkins University School of Medicine, Baltimore (Margolis, Ross, Sawa, F.C. Nucifora); Program in Cellular and Molecular Medicine, Johns Hopkins University School of Medicine, Baltimore (Lee, Sawa); the Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore (Ross)
| | - Robert A Sweet
- The Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore (L.G. Nucifora, Lee, Peters, Yang, Margolis, Pevsner, Ross, Sawa, F.C. Nucifora); the Departments of Psychiatry and Neurology, University of Pittsburgh, and the VISN 4 Mental Illness Research, Education, and Clinical Center, VA Pittsburgh Healthcare System, Pittsburgh (MacDonald, Sweet); the Department of Neurology, Kennedy Krieger Institute, Baltimore (Norris, Pevsner); the Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore (Norris, Pevsner, Ross, Sawa); Protein Characterization Laboratory, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Frederick, Md. (Orsburn); the Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas (Gleason, Tamminga); the Department of Neurology, Johns Hopkins University School of Medicine, Baltimore (Margolis, Ross, Sawa, F.C. Nucifora); Program in Cellular and Molecular Medicine, Johns Hopkins University School of Medicine, Baltimore (Lee, Sawa); the Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore (Ross)
| | - Christopher A Ross
- The Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore (L.G. Nucifora, Lee, Peters, Yang, Margolis, Pevsner, Ross, Sawa, F.C. Nucifora); the Departments of Psychiatry and Neurology, University of Pittsburgh, and the VISN 4 Mental Illness Research, Education, and Clinical Center, VA Pittsburgh Healthcare System, Pittsburgh (MacDonald, Sweet); the Department of Neurology, Kennedy Krieger Institute, Baltimore (Norris, Pevsner); the Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore (Norris, Pevsner, Ross, Sawa); Protein Characterization Laboratory, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Frederick, Md. (Orsburn); the Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas (Gleason, Tamminga); the Department of Neurology, Johns Hopkins University School of Medicine, Baltimore (Margolis, Ross, Sawa, F.C. Nucifora); Program in Cellular and Molecular Medicine, Johns Hopkins University School of Medicine, Baltimore (Lee, Sawa); the Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore (Ross)
| | - Akira Sawa
- The Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore (L.G. Nucifora, Lee, Peters, Yang, Margolis, Pevsner, Ross, Sawa, F.C. Nucifora); the Departments of Psychiatry and Neurology, University of Pittsburgh, and the VISN 4 Mental Illness Research, Education, and Clinical Center, VA Pittsburgh Healthcare System, Pittsburgh (MacDonald, Sweet); the Department of Neurology, Kennedy Krieger Institute, Baltimore (Norris, Pevsner); the Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore (Norris, Pevsner, Ross, Sawa); Protein Characterization Laboratory, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Frederick, Md. (Orsburn); the Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas (Gleason, Tamminga); the Department of Neurology, Johns Hopkins University School of Medicine, Baltimore (Margolis, Ross, Sawa, F.C. Nucifora); Program in Cellular and Molecular Medicine, Johns Hopkins University School of Medicine, Baltimore (Lee, Sawa); the Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore (Ross)
| | - Frederick C Nucifora
- The Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore (L.G. Nucifora, Lee, Peters, Yang, Margolis, Pevsner, Ross, Sawa, F.C. Nucifora); the Departments of Psychiatry and Neurology, University of Pittsburgh, and the VISN 4 Mental Illness Research, Education, and Clinical Center, VA Pittsburgh Healthcare System, Pittsburgh (MacDonald, Sweet); the Department of Neurology, Kennedy Krieger Institute, Baltimore (Norris, Pevsner); the Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore (Norris, Pevsner, Ross, Sawa); Protein Characterization Laboratory, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Frederick, Md. (Orsburn); the Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas (Gleason, Tamminga); the Department of Neurology, Johns Hopkins University School of Medicine, Baltimore (Margolis, Ross, Sawa, F.C. Nucifora); Program in Cellular and Molecular Medicine, Johns Hopkins University School of Medicine, Baltimore (Lee, Sawa); the Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore (Ross)
| |
Collapse
|
36
|
The standardization of cerebrospinal fluid markers and neuropathological diagnoses brings to light the frequent complexity of concomitant pathology in Alzheimer's disease: The next challenge for biochemical markers? Clin Biochem 2019; 72:15-23. [PMID: 31194969 DOI: 10.1016/j.clinbiochem.2019.06.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 05/29/2019] [Accepted: 06/06/2019] [Indexed: 12/14/2022]
Abstract
During the last two decades, neuropathological examination of the brain has evolved both technically and scientifically. The increasing use of immunohistochemistry to detect protein aggregates paralleled a better understanding of neuroanatomical progression of protein deposition. As a consequence, an international effort was achieved to standardize hyperphosphorylated-Tau (phospho-TAU), ßAmyloid (Aß), alpha syncuclein (alpha-syn), phosphorylated transactive response DNA-binding protein 43 (phospho-TDP43) and vascular pathology detection. Meanwhile harmonized staging systems emerged in order to increase inter rater reproducibility. Therefore, a refined definition of Alzheimer's disease was recommended., a clearer picture of the neuropathological lesions diversity emerged secondarily to the systematic assessment of concomitant pathology highlighting finally a low rate of pure AD pathology. This brings new challenges to laboratory medicine in the field of cerebrospinal fluid (CSF) markers of Alzheimer's disease: how to further validate total Tau, phospho-TAU, Aß40 and Aß42 and new marker level cut-offs while autopsy rates are declining?
Collapse
|
37
|
Bereczki E, Branca RM, Francis PT, Pereira JB, Baek JH, Hortobágyi T, Winblad B, Ballard C, Lehtiö J, Aarsland D. Synaptic markers of cognitive decline in neurodegenerative diseases: a proteomic approach. Brain 2019; 141:582-595. [PMID: 29324989 DOI: 10.1093/brain/awx352] [Citation(s) in RCA: 150] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Accepted: 10/30/2017] [Indexed: 01/12/2023] Open
Abstract
See Attems and Jellinger (doi:10.1093/brain/awx360) for a scientific commentary on this article.Cognitive changes occurring throughout the pathogenesis of neurodegenerative diseases are directly linked to synaptic loss. We used in-depth proteomics to compare 32 post-mortem human brains in the prefrontal cortex of prospectively followed patients with Alzheimer's disease, Parkinson's disease with dementia, dementia with Lewy bodies and older adults without dementia. In total, we identified 10 325 proteins, 851 of which were synaptic proteins. Levels of 25 synaptic proteins were significantly altered in the various dementia groups. Significant loss of SNAP47, GAP43, SYBU (syntabulin), LRFN2, SV2C, SYT2 (synaptotagmin 2), GRIA3 and GRIA4 were further validated on a larger cohort comprised of 92 brain samples using ELISA or western blot. Cognitive impairment before death and rate of cognitive decline significantly correlated with loss of SNAP47, SYBU, LRFN2, SV2C and GRIA3 proteins. Besides differentiating Parkinson's disease dementia, dementia with Lewy bodies, and Alzheimer's disease from controls with high sensitivity and specificity, synaptic proteins also reliably discriminated Parkinson's disease dementia from Alzheimer's disease patients. Our results suggest that these particular synaptic proteins have an important predictive and discriminative molecular fingerprint in neurodegenerative diseases and could be a potential target for early disease intervention.
Collapse
Affiliation(s)
- Erika Bereczki
- Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Division of Neurogeriatrics, Karolinska Institutet, Novum, Stockholm, Sweden
| | - Rui M Branca
- Department of Oncology-Pathology, Science for Life Laboratory, Karolinska Institutet, Stockholm, Sweden
| | - Paul T Francis
- King's College London, Wolfson Centre for Age-Related Diseases, London SE1 1UL, UK
| | - Joana B Pereira
- Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Division of Clinical Geriatrics, Karolinska Institutet, Novum, 14186 Stockholm, Sweden
| | - Jean-Ha Baek
- Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Division of Neurogeriatrics, Karolinska Institutet, Novum, Stockholm, Sweden
| | - Tibor Hortobágyi
- MTA-DE Cerebrovascular and Neurodegenerative Research Group, University of Debrecen, Debrecen, Hungary.,Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Bengt Winblad
- Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Division of Neurogeriatrics, Karolinska Institutet, Novum, Stockholm, Sweden
| | - Clive Ballard
- Department of Oncology-Pathology, Science for Life Laboratory, Karolinska Institutet, Stockholm, Sweden
| | - Janne Lehtiö
- Department of Oncology-Pathology, Science for Life Laboratory, Karolinska Institutet, Stockholm, Sweden
| | - Dag Aarsland
- Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Division of Neurogeriatrics, Karolinska Institutet, Novum, Stockholm, Sweden.,Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| |
Collapse
|
38
|
Sánchez RG, Parrish RR, Rich M, Webb WM, Lockhart RM, Nakao K, Ianov L, Buckingham SC, Broadwater DR, Jenkins A, de Lanerolle NC, Cunningham M, Eid T, Riley K, Lubin FD. Human and rodent temporal lobe epilepsy is characterized by changes in O-GlcNAc homeostasis that can be reversed to dampen epileptiform activity. Neurobiol Dis 2019; 124:531-543. [PMID: 30625365 PMCID: PMC6379093 DOI: 10.1016/j.nbd.2019.01.001] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Revised: 11/26/2018] [Accepted: 01/01/2019] [Indexed: 02/06/2023] Open
Abstract
Temporal Lobe Epilepsy (TLE) is frequently associated with changes in protein composition and post-translational modifications (PTM) that exacerbate the disorder. O-linked-β-N-acetyl glucosamine (O-GlcNAc) is a PTM occurring at serine/threonine residues that is derived from and closely associated with metabolic substrates. The enzymes O-GlcNActransferase (OGT) and O-GlcNAcase (OGA) mediate the addition and removal, respectively, of the O-GlcNAc modification. The goal of this study was to characterize OGT/OGA and protein O-GlcNAcylation in the epileptic hippocampus and to determine and whether direct manipulation of these proteins and PTM's alter epileptiform activity. We observed reduced global and protein specific O-GlcNAcylation and OGT expression in the kainate rat model of TLE and in human TLE hippocampal tissue. Inhibiting OGA with Thiamet-G elevated protein O-GlcNAcylation, and decreased both seizure duration and epileptic spike events, suggesting that OGA may be a therapeutic target for seizure control. These findings suggest that loss of O-GlcNAc homeostasis in the kainate model and in human TLE can be reversed via targeting of O-GlcNAc related pathways.
Collapse
Affiliation(s)
- Richard G Sánchez
- Department of Neurobiology, University of Alabama, Birmingham, AL, United States
| | - R Ryley Parrish
- Institute of Neuroscience, Newcastle University, Newcastle upon Tyne, UK
| | - Megan Rich
- Department of Neurobiology, University of Alabama, Birmingham, AL, United States
| | - William M Webb
- Department of Neurobiology, University of Alabama, Birmingham, AL, United States
| | - Roxanne M Lockhart
- Department of Neurobiology, University of Alabama, Birmingham, AL, United States
| | - Kazuhito Nakao
- Department of Neurobiology, University of Alabama, Birmingham, AL, United States
| | - Lara Ianov
- Civitan International Research Center, University of Alabama, Birmingham, AL, United States
| | - Susan C Buckingham
- Department of Neurobiology, University of Alabama, Birmingham, AL, United States
| | - Devin R Broadwater
- School of Medicine, University of Alabama, Birmingham, AL, United States
| | - Alistair Jenkins
- Department of Neurosurgery, Royal Victoria Infirmary, Newcastle upon Tyne NE1 4LP, UK
| | - Nihal C de Lanerolle
- Department of Laboratory Medicine and of Neurosurgery, Yale School of Medicine, New Haven, CT, United States
| | - Mark Cunningham
- Institute of Neuroscience, Newcastle University, Newcastle upon Tyne, UK
| | - Tore Eid
- Department of Laboratory Medicine and of Neurosurgery, Yale School of Medicine, New Haven, CT, United States
| | - Kristen Riley
- Department of Neurosurgery, University of Alabama, Birmingham, AL, United States
| | - Farah D Lubin
- Department of Neurobiology, University of Alabama, Birmingham, AL, United States.
| |
Collapse
|
39
|
Qian L, Sun Y, Tong Q, Tian J, Ren Z, Zhao H. Indentation response in porcine brain under electric fields. SOFT MATTER 2019; 15:623-632. [PMID: 30608501 DOI: 10.1039/c8sm01272e] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Electric fields in the environment can have profound effects on brain function and behavior. In clinical practice, some noninvasive/microinvasive therapies with electrical fields such as transcranial electrical stimulation (tES), deep brain stimulation (DBS), and electroconvulsive therapy (ECT) have emerged as powerful tools for the treatment of neuropsychiatric disorders and neuromodulation. Nonetheless, currently, most studies focus on the mechanisms and effects of therapies and do not to address the mechanical properties of brain tissue under electric fields. Thus, the mechanical behavior of brain tissue, which plays an important role in modulating both brain form and brain function, should be given attention. The present study addresses this paucity by presenting, for the first time, the mechanical properties of brain tissue under various intensities of direct current electric field (0, 2, 5, 10, 20, and 50 V) using a custom-designed indentation device. Prior to brain indentation, validation tests were performed in different hydrogels to ensure that there was no interference in the electric fields from the indentation device. Subsequently, the load trace data obtained from the indentation-relaxation tests was fitted to both linear elastic and viscoelastic models to characterize the sensitivity of the mechanical behavior of the brain tissue to the electric fields. The brain tissue was found to be softened at a higher electric field level and less viscous, and substantially responded more quickly with an increase in electric field. The explanations for the above behaviors were further discussed based on the analysis of the resistance and thermal responses during the testing process. Understanding the effect of electric fields on brain tissue at the mechanical level can provide a better understanding of the mechanisms of some therapies, which may be beneficial to guide therapy protocols.
Collapse
Affiliation(s)
- Long Qian
- School of Mechanical and Aerospace Engineering, Jilin University, Changchun 130025, China.
| | - Yifan Sun
- School of Mechanical and Aerospace Engineering, Jilin University, Changchun 130025, China.
| | - Qian Tong
- Department of Cardiology, The First Hospital of Jilin University, Changchun 130021, China.
| | - Jiyu Tian
- School of Mechanical and Aerospace Engineering, Jilin University, Changchun 130025, China.
| | - Zhuang Ren
- School of Mechanical and Aerospace Engineering, Jilin University, Changchun 130025, China.
| | - Hongwei Zhao
- School of Mechanical and Aerospace Engineering, Jilin University, Changchun 130025, China.
| |
Collapse
|
40
|
Dean B, Lam LQ, Scarr E, Duce JA. Cortical biometals: Changed levels in suicide and with mood disorders. J Affect Disord 2019; 243:539-544. [PMID: 30292148 DOI: 10.1016/j.jad.2018.09.026] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Revised: 08/13/2018] [Accepted: 09/11/2018] [Indexed: 01/04/2023]
Abstract
BACKGROUND Changes in levels of metals have been suggested to contribute to the pathophysiologies of several neurodegenerative disorders but to our knowledge this is the first metallomic study in CNS from patients with mood disorders. The focus of this study was on cortical regions affected by the pathophysiologies of bipolar disorders and major depressive disorders. METHODS Levels of metals were measured using inductively coupled plasma mass spectrometry in Brodmann's areas (BA) 6, 10 and 17 from patients with major depressive disorders (n = 13), bipolar disorders (n = 12) and age / sex matched controls (n = 13). RESULTS There were lower levels of cortical strontium (BA 6 & 10), ruthenium (BA 6 & 17) and cadmium (BA 10) from patients with major depressive disorder as well as lower levels of strontium in BA 10 from patients with bipolar disorders. Unexpectedly, there were changes in levels of 16 metals in the cortex, mainly BA 6, from suicide completers compared to those who died of other causes. LIMITATIONS Cohort sizes were relatively small but comparable with many studies using human postmortem CNS. Like all studies on non-treatment naïve patients, drug treatment was a potential confound in our experiments. CONCLUSIONS Our exploratory study suggests changes in levels of metals in bipolar disorders and major depressive disorders could be affecting cortical oxidative balance in patients with mood disorders. Our data raises the possibility that measuring levels of specific biometals in the blood could be used as a biomarker for increased risk of suicide.
Collapse
Affiliation(s)
- Brian Dean
- The Molecular Psychiatry Laboratory, The Florey Institute of Neuroscience and Mental Health, Parkville, Victoria, Australia; The Centre for Mental Health, the Faculty of Health, Arts and Design, Swinburne University, Hawthorne, Victoria, Australia
| | - Linh Q Lam
- The Oxidation Biology Unit, The Florey Institute of Neuroscience and Mental Health, Parkville, Victoria, Australia; Neuropharmacology Laboratory, Department of Pharmacology and Therapeutics, The University of Melbourne, Parkville, Victoria, Australia
| | - Elizabeth Scarr
- The Molecular Psychiatry Laboratory, The Florey Institute of Neuroscience and Mental Health, Parkville, Victoria, Australia; Melbourne Veterinary School, Faculty of Veterinary and Agricultural Sciences, The University of Melbourne, Victoria, Australia
| | - James A Duce
- The Oxidation Biology Unit, The Florey Institute of Neuroscience and Mental Health, Parkville, Victoria, Australia; The Alzheimer's Research UK Cambridge Drug Discovery Institute, University of Cambridge, Cambridge Biomedical Campus, Hills Road, Cambridge, UK
| |
Collapse
|
41
|
Parkin GM, Udawela M, Gibbons A, Dean B. Β-actin does not show the characteristics of a reference protein in human cortex. Electrophoresis 2018; 40:247-253. [DOI: 10.1002/elps.201800328] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Revised: 09/24/2018] [Accepted: 10/15/2018] [Indexed: 12/19/2022]
Affiliation(s)
- Georgia M. Parkin
- The Molecular Psychiatry Laboratory; the Florey Institute for Neuroscience and Mental Health; Parkville Victoria Australia
- The Cooperative Research Centre for Mental Health; Parkville Victoria Australia
| | - Madhara Udawela
- The Molecular Psychiatry Laboratory; the Florey Institute for Neuroscience and Mental Health; Parkville Victoria Australia
- The Cooperative Research Centre for Mental Health; Parkville Victoria Australia
| | - Andrew Gibbons
- The Molecular Psychiatry Laboratory; the Florey Institute for Neuroscience and Mental Health; Parkville Victoria Australia
| | - Brian Dean
- The Molecular Psychiatry Laboratory; the Florey Institute for Neuroscience and Mental Health; Parkville Victoria Australia
- The Cooperative Research Centre for Mental Health; Parkville Victoria Australia
- Centre for Mental Health; the Faculty of Health, Arts and Design; Swinburne University; Hawthorn Victoria Australia
| |
Collapse
|
42
|
Parkin GM, Udawela M, Gibbons A, Scarr E, Dean B. Catechol-O-methyltransferase (COMT) genotypes are associated with varying soluble, but not membrane-bound COMT protein in the human prefrontal cortex. J Hum Genet 2018; 63:1251-1258. [DOI: 10.1038/s10038-018-0511-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Revised: 08/22/2018] [Accepted: 08/23/2018] [Indexed: 11/09/2022]
|
43
|
Koss DJ, Dubini M, Buchanan H, Hull C, Platt B. Distinctive temporal profiles of detergent-soluble and -insoluble tau and Aβ species in human Alzheimer's disease. Brain Res 2018; 1699:121-134. [PMID: 30102892 DOI: 10.1016/j.brainres.2018.08.014] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Revised: 07/13/2018] [Accepted: 08/09/2018] [Indexed: 12/14/2022]
Abstract
Alzheimer's disease (AD) pathology relevant proteins tau and beta-amyloid (Aβ) exist as an array of post-translationally modified and conformationally altered species with varying abundance, solubility and toxicity. Insoluble neurofibrillary tau tangles and Aβ plaques are end-stage AD hallmarks, yet may carry less disease significance compared to soluble species. At present, it is unclear how soluble and insoluble tau and Aβ relate to each other as well as to disease progression. Here, detergent soluble and insoluble fractions generated from post-mortem human temporal lobe samples (Brodmann area 21) were probed for tau and Aβ markers in immuno-dot assays. Measures were quantified according to diagnosis (AD cf. Non-AD), neuropathological severity, and correlated with disease progression (Braak stages). All markers were elevated within AD cases cf. non-AD controls (p < 0.05) independent of solubility. However, when considered according to neuropathological severity, phospho-tau (detected via CP13 and AT8 antibodies) was elevated early within the soluble fraction (p < 0.05 intermediate cf. low severity) and emerged only later within the insoluble fraction (p < 0.05 high cf. low severity). In contrast, PHF1 phospho-tau, TOC1 reactive tau oligomers and amyloid markers rose within the two fractions simultaneously. Independent of solubility, cognitive correlations were observed for tau makers and for fibrillary amyloid (OC), however only soluble total Aβ was significantly correlated with intellectual impairment. Following the exclusion of end-stage cases, only soluble total Aβ remained correlated with cognition. The data indicate differential rates of protein aggregation during AD progression and confirm the disease relevance of early emerging soluble Aβ species.
Collapse
Affiliation(s)
- David J Koss
- School of Medicine, Medical Sciences & Nutrition, College of Life Sciences and Medicine, University of Aberdeen, Aberdeen AB25 2ZD, UK.
| | - Marina Dubini
- School of Medicine, Medical Sciences & Nutrition, College of Life Sciences and Medicine, University of Aberdeen, Aberdeen AB25 2ZD, UK
| | - Heather Buchanan
- School of Medicine, Medical Sciences & Nutrition, College of Life Sciences and Medicine, University of Aberdeen, Aberdeen AB25 2ZD, UK
| | - Claire Hull
- School of Medicine, Medical Sciences & Nutrition, College of Life Sciences and Medicine, University of Aberdeen, Aberdeen AB25 2ZD, UK
| | - Bettina Platt
- School of Medicine, Medical Sciences & Nutrition, College of Life Sciences and Medicine, University of Aberdeen, Aberdeen AB25 2ZD, UK.
| |
Collapse
|
44
|
|
45
|
Dean B, Gibbons A, Gogos A, Udawela M, Thomas E, Scarr E. Studies on Prostaglandin-Endoperoxide Synthase 1: Lower Levels in Schizophrenia and After Treatment with Antipsychotic Drugs in Conjunction with Aspirin. Int J Neuropsychopharmacol 2018; 21:216-225. [PMID: 30052978 PMCID: PMC5838806 DOI: 10.1093/ijnp/pyx092] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Revised: 09/18/2017] [Accepted: 10/04/2017] [Indexed: 12/12/2022] Open
Abstract
Background Antipsychotic drugs plus aspirin (acetylsalicylic acid), which targets prostaglandin-endoperoxide synthase 1 (PTGS1: COX1), improved therapeutic outcomes when treating schizophrenia. Our microarray data showed higher levels of PTGS1 mRNA in the dorsolateral prefrontal cortex from subjects with schizophrenia of long duration of illness, suggesting aspirin plus antipsychotic drugs could have therapeutic effects by lowering PTGS1 expression in the cortex of subjects with the disorder. Methods We used Western blotting to measure levels of PTSG1 protein in human postmortem CNS, rat and mouse cortex, and cells in culture. Results Compared with controls, PTGS1 levels were 41% lower in the dorsolateral prefrontal cortex (P<.01), but not the anterior cingulate or frontal pole, from subjects with schizophrenia. Levels of PTGS1 were not changed in the dorsolateral prefrontal cortex in mood disorders or in the cortex of rats treated with antipsychotic drugs. There was a strong trend (P=.05) to lower cortical PTGS1 10 months after mice were treated postnatally with polyinosinic-polycytidylic acid sodium salt (Poly I:C), consistent with cortical PTGS1 being lower in adult mice after exposure to an immune activator postnatally. In CCF-STTG1 cells, a human-derived astrocytic cell line, aspirin caused a dose-dependent decrease in PTGS1 that was decreased further with the addition of risperidone. Conclusions Our data suggest low levels of dorsolateral prefrontal cortex PTGS1 could be associated with the pathophysiology of schizophrenia, and improved therapeutic outcome from treating schizophrenia with antipsychotic drugs augmented with aspirin may be because such treatment lowers cortical PTGS1.
Collapse
MESH Headings
- Animals
- Anti-Inflammatory Agents, Non-Steroidal/pharmacology
- Anti-Inflammatory Agents, Non-Steroidal/therapeutic use
- Antipsychotic Agents/pharmacology
- Antipsychotic Agents/therapeutic use
- Aspirin/pharmacology
- Aspirin/therapeutic use
- Bipolar Disorder/drug therapy
- Bipolar Disorder/enzymology
- Brain/drug effects
- Brain/enzymology
- Cell Line
- Depressive Disorder, Major/drug therapy
- Depressive Disorder, Major/enzymology
- Disease Models, Animal
- Dose-Response Relationship, Drug
- Drug Therapy, Combination
- Female
- Humans
- Male
- Mice, Inbred BALB C
- Middle Aged
- Poly I-C
- Prostaglandin-Endoperoxide Synthases/metabolism
- RNA, Messenger/metabolism
- Rats, Sprague-Dawley
- Risperidone/pharmacology
- Risperidone/therapeutic use
- Schizophrenia/drug therapy
- Schizophrenia/enzymology
Collapse
Affiliation(s)
- Brian Dean
- The Florey Institute for Neuroscience and Mental Health, Parkville, Victoria, Australia
- Centre for Mental Health, Swinburne University of Technology, Hawthorn, Australia
| | | | - Andrea Gogos
- The Florey Institute for Neuroscience and Mental Health, Parkville, Victoria, Australia
| | - Madhara Udawela
- The Florey Institute for Neuroscience and Mental Health, Parkville, Victoria, Australia
| | | | - Elizabeth Scarr
- The Florey Institute for Neuroscience and Mental Health, Parkville, Victoria, Australia
- Melbourne Veterinary School, Faculty of Veterinary and Agricultural Sciences, The University of Melbourne, Victoria, Australia
| |
Collapse
|
46
|
Scarr E, Udawela M, Dean B. Changed frontal pole gene expression suggest altered interplay between neurotransmitter, developmental, and inflammatory pathways in schizophrenia. NPJ SCHIZOPHRENIA 2018; 4:4. [PMID: 29463818 PMCID: PMC5820249 DOI: 10.1038/s41537-018-0044-x] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Revised: 01/10/2018] [Accepted: 01/16/2018] [Indexed: 12/22/2022]
Abstract
Schizophrenia (Sz) probably occurs after genetically susceptible individuals encounter a deleterious environmental factor that triggers epigenetic mechanisms to change CNS gene expression. To determine if omnibus changes in CNS gene expression are present in Sz, we compared mRNA levels in the frontal pole (Brodmann’s area (BA) 10), the dorsolateral prefrontal cortex (BA 9) and cingulate cortex (BA 33) from 15 subjects with Sz and 15 controls using the Affymetrix™ Human Exon 1.0 ST Array. Differences in mRNA levels (±≥20%; p < 0.01) were identified (JMP Genomics 5.1) and used to predict pathways and gene x gene interactions that would be affected by the changes in gene expression using Ingenuity Pathway Analysis. There was significant variation in mRNA levels with diagnoses for 566 genes in BA 10, 65 genes in BA 9 and 40 genes in BA 33. In Sz, there was an over-representation of genes with changed expression involved in inflammation and development in BA 10, cell morphology in BA 9 and amino acid metabolism and small molecule biochemistry in BA 33. Using 94 genes with altered levels of expression in BA 10 from subjects with Sz, it was possible to construct an interactome of proven direct gene x gene interactions that was enriched for genes in inflammatory, developmental, oestrogen, serotonergic, cholinergic and NRG1 regulated pathways. Our data shows complex, regionally specific changes in cortical gene expression in Sz that are predicted to affect homeostasis between biochemical pathways already proposed to be important in the pathophysiology of the disorder. Anterior brain regions exhibit significant amounts of differentially-expressed genes which might cause dysfunction in schizophrenia. It’s thought that schizophrenia occurs when environmental factors trigger gene expression changes and downstream effects in the human brain, though this is not fully understood. An Australian research group led by Brian Dean, from the Florey Institute of Neuroscience and Mental Health, conducted a post-mortem human brain study in which they compared gene expression between 15 schizophrenia patients and 15 controls. They found 566 instances of altered gene expression in the most frontal part of the brain, Brodmann Area 10, and fewer changes in proximal regions. These are brain areas known to mediate schizophrenia-related traits and the changes in gene expression in these areas will affect a range of essential biological pathways. The group also found 97 differentially-expressed genes that have been shown to directly interact with each. This study paints a complex picture of the causes of schizophrenia but suggests modern technologies can help unravel these complexities.
Collapse
Affiliation(s)
- Elizabeth Scarr
- Molecular Psychiatry Laboratory, Florey Institute for Neuroscience and Mental Health, Parkville, VIC 3052, Australia.,CRC for Mental Health, Carlton, VIC, 3053, Australia.,Melbourne Veterinary School, Faculty of Veterinary and Agricultural Sciences, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Madhara Udawela
- Molecular Psychiatry Laboratory, Florey Institute for Neuroscience and Mental Health, Parkville, VIC 3052, Australia.,CRC for Mental Health, Carlton, VIC, 3053, Australia
| | - Brian Dean
- Molecular Psychiatry Laboratory, Florey Institute for Neuroscience and Mental Health, Parkville, VIC 3052, Australia. .,CRC for Mental Health, Carlton, VIC, 3053, Australia. .,Research Centre for Mental Health, the Faculty of Health, Arts and Design, Swinburne University, Hawthorne, VIC, 3122, Australia.
| |
Collapse
|
47
|
Scarr E, Udawela M, Thomas EA, Dean B. Changed gene expression in subjects with schizophrenia and low cortical muscarinic M1 receptors predicts disrupted upstream pathways interacting with that receptor. Mol Psychiatry 2018; 23:295-303. [PMID: 27801890 PMCID: PMC5794886 DOI: 10.1038/mp.2016.195] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Revised: 08/24/2016] [Accepted: 08/26/2016] [Indexed: 12/14/2022]
Abstract
We tested the hypothesis that, compared with subjects with no history of psychiatric illness (controls), changes in gene expression in the dorsolateral prefrontal cortex from two subgroups of subjects with schizophrenia, one with a marked deficit in muscarinic M1 receptors (muscarinic receptor-deficit schizophrenia (MRDS)), would identify different biochemical pathways that would be affected by their aetiologies. Hence, we measured levels of cortical (Brodmann area 9) mRNA in 15 MRDS subjects, 15 subjects with schizophrenia but without a deficit in muscarinic M1 receptors (non-MRDS) and 15 controls using Affymetrix Exon 1.0 ST arrays. Levels of mRNA for 65 genes were significantly different in the cortex of subjects with MRDS and predicted changes in pathways involved in cellular movement and cell-to-cell signalling. Levels of mRNA for 45 genes were significantly different in non-MRDS and predicted changes in pathways involved in cellular growth and proliferation as well as cellular function and maintenance. Changes in gene expression also predicted effects on pathways involved in amino acid metabolism, molecular transport and small-molecule biochemistry in both MRDS and non-MRDS. Overall, our data argue a prominent role for glial function in MRDS and neurodevelopment in non-MRDS. Finally, the interactions of gene with altered levels of mRNA in the cortex of subjects with MRDS suggest many of their affects will be upstream of the muscarinic M1 receptor. Our study gives new insight into the molecular pathways affected in the cortex of subjects with MRDS and supports the notion that studying subgroups within the syndrome of schizophrenia is worthwhile.
Collapse
Affiliation(s)
- E Scarr
- Molecular Psychiatry Laboratory, Florey Institute for Neuroscience and Mental Health, Parkville, VIC, Australia,CRC for Mental Health, Carlton, VIC, Australia,Department of Psychiatry, University of Melbourne, Parkville, VIC, Australia
| | - M Udawela
- Molecular Psychiatry Laboratory, Florey Institute for Neuroscience and Mental Health, Parkville, VIC, Australia,CRC for Mental Health, Carlton, VIC, Australia
| | - E A Thomas
- Department of Molecular and Cellular Neuroscience, The Scripps Research Institute, La Jolla, CA, Australia
| | - B Dean
- Molecular Psychiatry Laboratory, Florey Institute for Neuroscience and Mental Health, Parkville, VIC, Australia,CRC for Mental Health, Carlton, VIC, Australia,Molecular Psychiatry Laboratory, Florey Institute for Neuroscience and Mental Health, 30 Royal Parade, Parkville, VIC 3052, Australia. E-mail:
| |
Collapse
|
48
|
Ramirez EPC, Keller CE, Vonsattel JP. The New York Brain Bank of Columbia University: practical highlights of 35 years of experience. HANDBOOK OF CLINICAL NEUROLOGY 2018; 150:105-118. [PMID: 29496134 DOI: 10.1016/b978-0-444-63639-3.00008-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The New York Brain Bank processes brains and organs of clinically well-characterized patients with age-related neurodegenerative diseases, and for comparison, from individuals without neurologic or psychiatric impairments. The donors, either patients or individuals, were evaluated at healthcare facilities of the Columbia University of New York. Each source brain yields four categories of samples: fresh frozen blocks and crushed parenchyma, and formalin-fixed wet blocks and histology sections. A source brain is thoroughly evaluated to determine qualitatively and quantitatively any changes it might harbor using conventional neuropathologic techniques. The clinical and pathologic diagnoses are integrated to determine the distributive diagnosis assigned to the samples obtained from a source brain. The gradual standardization of the protocol was developed in 1981 in response to the evolving requirements of basic investigations on neurodegeneration. The methods assimilate long-standing experience from multiple centers. The resulting and current protocol includes a constant central core applied to all brains with conditional flexibility around it. The New York Brain Bank is an integral part of the department of pathology, where the expertise, teaching duties, and hardware are shared. Since details of the protocols are available online, this chapter focuses on practical issues in professionalizing brain banking.
Collapse
Affiliation(s)
- Etty Paola Cortes Ramirez
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Medical Center, New York, NY, United States; New York Brain Bank, Children's Hospital, New York, NY, United States
| | | | - Jean Paul Vonsattel
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Medical Center, New York, NY, United States; New York Brain Bank, Children's Hospital, New York, NY, United States; Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY, United States.
| |
Collapse
|
49
|
Abstract
Proteomics and lipidomics are powerful tools to the large-scale study of proteins and lipids, respectively. Several methods can be employed with particular benefits and limitations in the study of human brain. This is a review of the rationale use of current techniques with particular attention to limitations and pitfalls inherent to each one of the techniques, and more importantly, to their use in the study of post-mortem brain tissue. These aspects are cardinal to avoid false interpretations, errors and unreal expectancies. Other points are also stressed as exemplified in the analysis of human neurodegenerative diseases which are manifested by disease-, region-, and stage-specific modifications commonly in the context of aging. Information about certain altered protein clusters and proteins oxidatively damaged is summarized for Alzheimer and Parkinson diseases.
Collapse
Affiliation(s)
- Isidro Ferrer
- Pathologic Anatomy Service, Institute of Neuropathology, Bellvitge University Hospital; Department of Pathology and Experimental Therapeutics, Faculty of Medicine, University of Barcelona; and Network Center of Biomedical Research on Neurodegenerative Diseases, Institute Carlos III; Hospitalet de Llobregat, Llobregat, Spain.
| |
Collapse
|
50
|
Ferrer I. Sisyphus in Neverland. J Alzheimers Dis 2018; 62:1023-1047. [PMID: 29154280 PMCID: PMC5870014 DOI: 10.3233/jad-170609] [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] [Accepted: 08/21/2017] [Indexed: 11/24/2022]
Abstract
The study of life and living organisms and the way in which these interact and organize to form social communities have been central to my career. I have been fascinated by biology, neurology, and neuropathology, but also by history, sociology, and art. Certain current historical, political, and social events, some occurring proximally but others affecting people in apparently distant places, have had an impact on me. Epicurus, Seneca, and Camus shared their philosophical positions which I learned from. Many scientists from various disciplines have been exciting sources of knowledge as well. I have created a world of hypothesis and experiments but I have also got carried away by serendipity following unexpected observations. It has not been an easy path; errors and wanderings are not uncommon, and opponents close to home much more abundant than one might imagine. Ambition, imagination, resilience, and endurance have been useful in moving ahead in response to setbacks. In the end, I have enjoyed my dedication to science and I am grateful to have glimpsed beauty in it. These are brief memories of a Spanish neuropathologist born and raised in Barcelona, EU.
Collapse
Affiliation(s)
- Isidro Ferrer
- Department of Pathology and Experimental Therapeutics, University of Barcelona; Service of Pathological Anatomy, Bellvitge University Hospital; CIBERNED; Hospitalet de Llobregat, Barcelona, Spain
| |
Collapse
|