1
|
Townsend J, Gross N, Peng Z, Peñagaricano F, Yang Z, Ahsan N, Khatib H. The embryonic DPPA3 gene stimulates the expression of pregnancy-related genes in bovine endometrial cells. J Dairy Sci 2025:S0022-0302(25)00230-9. [PMID: 40222672 DOI: 10.3168/jds.2024-25872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Accepted: 03/12/2025] [Indexed: 04/15/2025]
Abstract
Extracellular vesicles (EVs) released by cells contain mRNAs, miRNAs, lncRNAs, lipids, and proteins, playing crucial roles in cell-cell communication. While full-length mRNA transcripts have been documented in EVs secreted by cancer cells, there are no reports on full transcripts secreted by embryos. Our study aimed to identify EV mRNAs in the culture media of bovine embryos and investigate their roles in embryo-maternal communication. Following the isolation of EVs from in vitro fertilization media samples and RNA sequencing, we identified a full mRNA transcript of DPPA3, known to play an essential role in embryo development. To examine the role of DPPA3 in embryo-maternal communication, an in vitro transcribed mRNA of DPPA3 was transfected into bovine endometrial epithelial cells. Transfected and control cells were subsequently analyzed with RNA sequencing and proteomics to assess the effects of DPPA3 on gene expression. A total of 24 genes were found to be upregulated, and one gene was downregulated (FDR <0.01) following DPPA3 transfection, many with known functions in pregnancy recognition. Proteomic analysis revealed 28 differentially expressed proteins, with 19 upregulated and 15 downregulated. Two proteins, ISG15 and MX1, overlapped with the differentially expressed mRNAs. To mimic the natural transfer of EVs from embryos to endometrial cells, we performed coculture with day-8 blastocysts or supplemented the cells with embryo-conditioned culture media. DPPA3 presence was detected in endometrial cells exposed to embryo-conditioned media after just 30 min. Overall, our study highlights the significant role of EVs in cell-cell communication through mRNA signaling from the embryo to the mother.
Collapse
Affiliation(s)
- Jessica Townsend
- The Department of Animal and Dairy Sciences, The University of Wisconsin, Madison, Wisconsin, USA
| | - Nicole Gross
- The Department of Animal and Dairy Sciences, The University of Wisconsin, Madison, Wisconsin, USA
| | - Zongkai Peng
- The Department of Chemistry and Biochemistry, The University of Oklahoma, Norman, Oklahoma, USA
| | - Francisco Peñagaricano
- The Department of Animal and Dairy Sciences, The University of Wisconsin, Madison, Wisconsin, USA
| | - Zhibo Yang
- The Department of Chemistry and Biochemistry, The University of Oklahoma, Norman, Oklahoma, USA
| | - Nagib Ahsan
- The Department of Chemistry and Biochemistry, The University of Oklahoma, Norman, Oklahoma, USA; Mass Spectrometry, Proteomics and Metabolomics Core Facility, Stephenson Life Sciences Research Center, The University of Oklahoma, Norman, Oklahoma, USA
| | - Hasan Khatib
- The Department of Animal and Dairy Sciences, The University of Wisconsin, Madison, Wisconsin, USA.
| |
Collapse
|
2
|
Yamaguchi M. Extracellular Regucalcin: A Potent Suppressor in the Cancer Cell Microenvironment. Cancers (Basel) 2025; 17:240. [PMID: 39858022 PMCID: PMC11763602 DOI: 10.3390/cancers17020240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 01/03/2025] [Accepted: 01/07/2025] [Indexed: 01/27/2025] Open
Abstract
The regucalcin gene is located on the X chromosome, comprising seven exons and six introns. This gene and protein are expressed in various tissues and cells and is predominantly expressed in human liver, kidney, and adrenal tissues. Regucalcin gene expression is enhanced via a mechanism mediated by several signaling molecules and transcription factors. Regucalcin plays a multifunctional role in cellular regulation in maintaining cell homeostasis. In addition, regucalcin has been implicated in several metabolic disorders and diseases. In particular, regucalcin plays a role as a novel suppressor in several types of cancer patients. Increased expression of regucalcin suppresses the growth of human cancer cells, suggesting its pivotal role in suppressing tumor development. The survival time of cancer patients is prolonged with increased expression of regucalcin in the tumor tissues. The adhesion, migration, invasion, and bone metastatic activity of cancer cells are blocked by the overexpression of regucalcin, promoting dormancy in cancer patients. Interestingly, regucalcin is also found in human serum, suggesting its character as a novel biomarker in various diseases. This extracellular regucalcin has been shown to suppress human cancer cells' growth and bone metastatic activity. Thus, extracellular regucalcin may play a vital role as a suppressor of human cancer activity. Alteration of the serum regucalcin levels in physiological and pathophysiological conditions may influence the activity of cancer cells in the microenvironment. This review will discuss the potential role of extracellular regucalcin in cancer cell activity as a critical suppressor in the cancer microenvironment.
Collapse
Affiliation(s)
- Masayoshi Yamaguchi
- Cancer Biology Program, University of Hawaii Cancer Center, University of Hawaii at Manoa, 701 Ilalo Street, Honolulu, HI 96813, USA
| |
Collapse
|
3
|
Guo X, Luo X, Huang X, Zhang Y, Ji J, Wang X, Wang K, Wang J, Pan X, Chen B, Tan Y, Luo X. The Role of 3' Regulatory Region Flanking Kinectin 1 Gene in Schizophrenia. ALPHA PSYCHIATRY 2024; 25:413-420. [PMID: 39148597 PMCID: PMC11322729 DOI: 10.5152/alphapsychiatry.2024.241616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Accepted: 04/18/2024] [Indexed: 08/17/2024]
Abstract
Objective Schizophrenia is often associated with volumetric reductions in cortices and expansions in basal ganglia, particularly the putamen. Recent genome-wide association studies have highlighted the significance of variants in the 3' regulatory region adjacent to the kinectin 1 gene (KTN1) in regulating gray matter volume (GMV) of the putamen. This study aimed to comprehensively investigate the involvement of this region in schizophrenia. Methods We analyzed 1136 single-nucleotide polymorphisms (SNPs) covering the entire 3' regulatory region in 4 independent dbGaP samples (4604 schizophrenia patients vs. 4884 healthy subjects) and 3 independent Psychiatric Genomics Consortium samples (107 240 cases vs. 210 203 controls) to identify consistent associations. Additionally, we examined the regulatory effects of schizophrenia-associated alleles on KTN1 mRNA expression in 16 brain areas among 348 subjects, as well as GMVs of 7 subcortical nuclei in 38 258 subjects, and surface areas (SA) and thickness (TH) of the entire cortex and 34 cortical areas in 36 936 subjects. Results The major alleles (f > 0.5) of 25 variants increased (β > 0) the risk of schizophrenia across 2 to 5 independent samples (8.4 × 10-4 ≤ P ≤ .049). These schizophrenia-associated alleles significantly elevated (β > 0) GMVs of basal ganglia, including the putamen (6.0 × 10-11 ≤ P ≤ 1.1 × 10-4), caudate (8.7 × 10-4 ≤ P ≤ 9.4 × 10-3), pallidum (P = 6.0 × 10-4), and nucleus accumbens (P = 2.7 × 10-5). Moreover, they potentially augmented (β > 0) the SA of posterior cingulate and insular cortices, as well as the TH of frontal (pars triangularis and medial orbitofrontal), parietal (superior, precuneus, and inferior), and temporal (transverse) cortices, but potentially reduced (β < 0) the SA of the whole, frontal (medial orbitofrontal), and temporal (pole, superior, middle, and entorhinal) cortices, as well as the TH of rostral middle frontal and superior frontal cortices (8.9 × 10-4 ≤ P ≤ .050). Conclusion Our findings identify significant and functionally relevant risk alleles in the 3' regulatory region adjacent to KTN1, implicating their crucial roles in the development of schizophrenia.
Collapse
Affiliation(s)
- Xiaoyun Guo
- Shanghai Mental Health Center, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Xinqun Luo
- Department of Neurosurgery, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Xiaoyi Huang
- Shanghai Mental Health Center, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Yong Zhang
- Institute of Mental Health, Tianjin Anding Hospital, Mental Health Center of Tianjin Medical University, Tianjin, China
| | - Jiawu Ji
- Department of Psychiatry, Fujian Medical University Affiliated Fuzhou Neuropsychiatric Hospital, Fuzhou, Fujian, China
| | - Xiaoping Wang
- Department of Neurology, Jiading Branch of Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Kesheng Wang
- Department of Family and Community Health, School of Nursing, Health Sciences Center, West Virginia University, Morgantown, WV, USA
| | - Jijun Wang
- Shanghai Mental Health Center, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Xinghua Pan
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southern Medical University, and Guangdong Provincial Key Laboratory of Single Cell Technology and Application, Guangzhou, China
| | - Bin Chen
- Department of Cardiovascular Medicine, Shengli Clinical Medical College of Fujian Medical University, Fujian Medical University, Fujian Provincial Hospital, Fuzhou, China
| | - Yunlong Tan
- Beijing Huilongguan Hospital, Peking University Huilongguan School of Clinical Medicine, Beijing, China
| | - Xingguang Luo
- Beijing Huilongguan Hospital, Peking University Huilongguan School of Clinical Medicine, Beijing, China
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| |
Collapse
|
4
|
Leung HH, Mansour C, Rousseau M, Nakhla A, Kiselyov K, Venkatachalam K, Wong CO. Drosophila tweety facilitates autophagy to regulate mitochondrial homeostasis and bioenergetics in Glia. Glia 2024; 72:433-451. [PMID: 37870193 PMCID: PMC10842981 DOI: 10.1002/glia.24484] [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: 06/02/2023] [Revised: 09/12/2023] [Accepted: 10/08/2023] [Indexed: 10/24/2023]
Abstract
Mitochondria support the energetic demands of the cells. Autophagic turnover of mitochondria serves as a critical pathway for mitochondrial homeostasis. It is unclear how bioenergetics and autophagy are functionally connected. Here, we identify an endolysosomal membrane protein that facilitates autophagy to regulate ATP production in glia. We determined that Drosophila tweety (tty) is highly expressed in glia and localized to endolysosomes. Diminished fusion between autophagosomes and endolysosomes in tty-deficient glia was rescued by expressing the human Tweety Homolog 1 (TTYH1). Loss of tty in glia attenuated mitochondrial turnover, elevated mitochondrial oxidative stress, and impaired locomotor functions. The cellular and organismal defects were partially reversed by antioxidant treatment. We performed live-cell imaging of genetically encoded metabolite sensors to determine the impact of tty and autophagy deficiencies on glial bioenergetics. We found that tty-deficient glia exhibited reduced mitochondrial pyruvate consumption accompanied by a shift toward glycolysis for ATP production. Likewise, genetic inhibition of autophagy in glia resulted in a similar glycolytic shift in bioenergetics. Furthermore, the survival of mutant flies became more sensitive to starvation, underlining the significance of tty in the crosstalk between autophagy and bioenergetics. Together, our findings uncover the role for tty in mitochondrial homeostasis via facilitating autophagy, which determines bioenergetic balance in glia.
Collapse
Affiliation(s)
- Ho Hang Leung
- Department of Biological Sciences, Rutgers University, Newark, NJ 07102, USA
- Present address: South Australian Health and Medical Research Institute (SAHMRI), Adelaide, SA 5000, Australia
| | - Christina Mansour
- Department of Biological Sciences, Rutgers University, Newark, NJ 07102, USA
| | - Morgan Rousseau
- Department of Integrative Biology and Pharmacology, McGovern Medical School at the University of Texas Health Sciences Center (UTHealth), Houston, TX 77030, USA
| | - Anwar Nakhla
- Department of Biological Sciences, Rutgers University, Newark, NJ 07102, USA
| | - Kirill Kiselyov
- Department of Biological Sciences, University of Pittsburgh, PA 15260, USA
| | - Kartik Venkatachalam
- Department of Integrative Biology and Pharmacology, McGovern Medical School at the University of Texas Health Sciences Center (UTHealth), Houston, TX 77030, USA
| | - Ching-On Wong
- Department of Biological Sciences, Rutgers University, Newark, NJ 07102, USA
| |
Collapse
|
5
|
Kojima R, Paslawski W, Lyu G, Arenas E, Zhang X, Svenningsson P. Secretome Analyses Identify FKBP4 as a GBA1-Associated Protein in CSF and iPS Cells from Parkinson's Disease Patients with GBA1 Mutations. Int J Mol Sci 2024; 25:683. [PMID: 38203854 PMCID: PMC10779269 DOI: 10.3390/ijms25010683] [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: 12/10/2023] [Revised: 12/31/2023] [Accepted: 01/02/2024] [Indexed: 01/12/2024] Open
Abstract
Mutations in the GBA1 gene increase the risk of developing Parkinson's disease (PD). However, most carriers of GBA1 mutations do not develop PD throughout their lives. The mechanisms of how GBA1 mutations contribute to PD pathogenesis remain unclear. Cerebrospinal fluid (CSF) is used for detecting pathological conditions of diseases, providing insights into the molecular mechanisms underlying neurodegenerative disorders. In this study, we utilized the proximity extension assay to examine the levels of metabolism-linked protein in the CSF from 17 PD patients carrying GBA1 mutations (GBA1-PD) and 17 idiopathic PD (iPD). The analysis of CSF secretome in GBA1-PD identified 11 significantly altered proteins, namely FKBP4, THOP1, GLRX, TXNDC5, GAL, SEMA3F, CRKL, APLP1, LRP11, CD164, and NPTXR. To investigate GBA1-associated CSF changes attributed to specific neuronal subtypes responsible for PD, we analyzed the cell culture supernatant from GBA1-PD-induced pluripotent stem cell (iPSC)-derived midbrain dopaminergic (mDA) neurons. The secretome analysis of GBA1-PD iPSC-derived mDA neurons revealed that five differently regulated proteins overlapped with those identified in the CSF analysis: FKBP4, THOP1, GLRX, GAL, and CRKL. Reduced intracellular level of the top hit, FKPB4, was confirmed via Western Blot. In conclusion, our findings identify significantly altered CSF GBA1-PD-associated proteins with FKPB4 being firmly attributed to mDA neurons.
Collapse
Affiliation(s)
- Rika Kojima
- Department of Clinical Neuroscience, Karolinska Institutet, 171 76 Stockholm, Sweden; (R.K.)
| | - Wojciech Paslawski
- Department of Clinical Neuroscience, Karolinska Institutet, 171 76 Stockholm, Sweden; (R.K.)
| | - Guochang Lyu
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Ernest Arenas
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Xiaoqun Zhang
- Department of Clinical Neuroscience, Karolinska Institutet, 171 76 Stockholm, Sweden; (R.K.)
| | - Per Svenningsson
- Department of Clinical Neuroscience, Karolinska Institutet, 171 76 Stockholm, Sweden; (R.K.)
| |
Collapse
|
6
|
da Silva Beraldo IJ, Prates Rodrigues M, Polanczyk RS, Verano-Braga T, Lopes-Aguiar C. Proteomic-Based Studies on Memory Formation in Normal and Neurodegenerative Disease-Affected Brains. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1443:129-158. [PMID: 38409419 DOI: 10.1007/978-3-031-50624-6_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/28/2024]
Abstract
A critical aspect of cognition is the ability to acquire, consolidate, and evoke memories, which is considerably impaired by neurodegenerative disorders such as Alzheimer's and Parkinson's diseases. These mnemonic processes are dependent on signaling cascades, which involve protein expression and degradation. Recent mass spectrometry (MS)-based proteomics has opened a range of possibilities for the study of memory formation and impairment, making it possible to research protein systems not studied before. However, in the context of synaptic proteome related to learning processes and memory formation, a deeper understanding of the synaptic proteome temporal dynamics after induction of synaptic plasticity and the molecular changes underlying the cognitive deficits seen in neurodegenerative diseases is needed. This review analyzes the applications of proteomics for understanding memory processes in both normal and neurodegenerative conditions. Moreover, the most critical experimental studies have been summarized using the PANTHER overrepresentation test. Finally, limitations associated with investigations of memory studies in physiological and neurodegenerative disorders have also been discussed.
Collapse
Affiliation(s)
- Ikaro Jesus da Silva Beraldo
- Universidade Federal de Minas Gerais, Departamento de Fisiologia e Biofísica, Laboratório de Neurociências Comportamental e Molecular (LANEC), Belo Horizonte, Brazil
| | - Mateus Prates Rodrigues
- Universidade Federal de Minas Gerais, Departamento de Fisiologia e Biofísica, Laboratório de Neurociências Comportamental e Molecular (LANEC), Belo Horizonte, Brazil
| | - Rafaela Schuttenberg Polanczyk
- Universidade Federal de Minas Gerais, Departamento de Fisiologia e Biofísica, Laboratório de Neurociências Comportamental e Molecular (LANEC), Belo Horizonte, Brazil
| | - Thiago Verano-Braga
- Universidade Federal de Minas Gerais, Departamento de Fisiologia e Biofísica, Núcleo de Proteômica Funcional (NPF), Belo Horizonte, Brazil
- Instituto Nacional de Ciência e Tecnologia em Nano-Biofarmacêutica (INCT-Nanobiofar), Belo Horizonte, Brazil
| | - Cleiton Lopes-Aguiar
- Universidade Federal de Minas Gerais, Departamento de Fisiologia e Biofísica, Laboratório de Neurociências Comportamental e Molecular (LANEC), Belo Horizonte, Brazil.
| |
Collapse
|
7
|
Downs M, Zaia J, Sethi MK. Mass spectrometry methods for analysis of extracellular matrix components in neurological diseases. MASS SPECTROMETRY REVIEWS 2023; 42:1848-1875. [PMID: 35719114 PMCID: PMC9763553 DOI: 10.1002/mas.21792] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 04/12/2022] [Accepted: 05/24/2022] [Indexed: 06/15/2023]
Abstract
The brain extracellular matrix (ECM) is a highly glycosylated environment and plays important roles in many processes including cell communication, growth factor binding, and scaffolding. The formation of structures such as perineuronal nets (PNNs) is critical in neuroprotection and neural plasticity, and the formation of molecular networks is dependent in part on glycans. The ECM is also implicated in the neuropathophysiology of disorders such as Alzheimer's disease (AD), Parkinson's disease (PD), and Schizophrenia (SZ). As such, it is of interest to understand both the proteomic and glycomic makeup of healthy and diseased brain ECM. Further, there is a growing need for site-specific glycoproteomic information. Over the past decade, sample preparation, mass spectrometry, and bioinformatic methods have been developed and refined to provide comprehensive information about the glycoproteome. Core ECM molecules including versican, hyaluronan and proteoglycan link proteins, and tenascin are dysregulated in AD, PD, and SZ. Glycomic changes such as differential sialylation, sulfation, and branching are also associated with neurodegeneration. A more thorough understanding of the ECM and its proteomic, glycomic, and glycoproteomic changes in brain diseases may provide pathways to new therapeutic options.
Collapse
Affiliation(s)
- Margaret Downs
- Department of Biochemistry, Center for Biomedical Mass Spectrometry, Boston University, Boston, Massachusetts, USA
| | - Joseph Zaia
- Department of Biochemistry, Center for Biomedical Mass Spectrometry, Boston University, Boston, Massachusetts, USA
- Bioinformatics Program, Boston University, Boston, Massachusetts, USA
| | - Manveen K Sethi
- Department of Biochemistry, Center for Biomedical Mass Spectrometry, Boston University, Boston, Massachusetts, USA
| |
Collapse
|
8
|
Fissolo N, Calvo-Barreiro L, Eixarch H, Boschert U, Villar LM, Costa-Frossard L, Ferrer M, Sanchez A, Borràs E, Sabidó E, Espejo C, Montalban X, Comabella M. Molecular signature associated with cladribine treatment in patients with multiple sclerosis. Front Immunol 2023; 14:1233546. [PMID: 37559720 PMCID: PMC10408299 DOI: 10.3389/fimmu.2023.1233546] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Accepted: 07/07/2023] [Indexed: 08/11/2023] Open
Abstract
Introduction Little is known about the molecular profiling associated with the effect of cladribine in patients with multiple sclerosis (MS). Here, we aimed first to characterize the transcriptomic and proteomic profiles induced by cladribine in blood cells, and second to identify potential treatment response biomarkers to cladribine in patients with MS. Methods Gene, protein and microRNA (miRNA) expression profiles were determined by microarrays (genes, miRNAs) and mass spectrometry (proteins) in peripheral blood mononuclear cells (PBMCs) from MS patients after in vitro treatment with cladribine in its active and inactive forms. Two bioinformatics approaches to integrate the three obtained datasets were applied: (i) a multiomics discriminant analysis (DIABLO - Data Integration Analysis for Biomarker discovery using Latent variable approaches for Omics studies); and (ii) a multi-stage integration of features selected in differential expression analysis on each dataset and then merged. Selected molecules from the in vitro study were quantified by qPCR ex vivo in PBMCs from MS patients receiving cladribine. Results PBMCs treated in vitro with cladribine were characterized by a major downregulation of gene, protein, and miRNA expression compared with the untreated cells. An intermediate pattern between the cladribine-treated and untreated conditions was observed in PBMCs treated with cladribine in its inactive form. The differential expression analysis of each dataset led to the identification of four genes and their encoded proteins, and twenty-two miRNAs regulating their expression, that were associated with cladribine treatment. Two of these genes (PPIF and NHLRC2), and three miRNAs (miR-21-5p, miR-30b-5p, and miR-30e-5p) were validated ex vivo in MS patients treated with cladribine. Discussion By using a combination of omics data and bioinformatics approaches we were able to identify a multiomics molecular profile induced by cladribine in vitro in PBMCs. We also identified a number of biomarkers that were validated ex vivo in PBMCs from patients with MS treated with cladribine that have the potential to become treatment response biomarkers to this drug.
Collapse
Affiliation(s)
- Nicolas Fissolo
- Servei de Neurologia, Centre d’Esclerosi Múltiple de Catalunya (Cemcat) Institut de Recerca Vall d’Hebron (VHIR), Hospital Universitari Vall d’Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Laura Calvo-Barreiro
- Servei de Neurologia, Centre d’Esclerosi Múltiple de Catalunya (Cemcat) Institut de Recerca Vall d’Hebron (VHIR), Hospital Universitari Vall d’Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Herena Eixarch
- Servei de Neurologia, Centre d’Esclerosi Múltiple de Catalunya (Cemcat) Institut de Recerca Vall d’Hebron (VHIR), Hospital Universitari Vall d’Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Ursula Boschert
- Ares Trading SA, Eysins, Switzerland, an affiliate of Merck KGaA, Darmstadt, Germany
| | - Luisa M. Villar
- Department of Immunology, Multiple Sclerosis Unit, Hospital Ramon y Cajal, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| | - Lucienne Costa-Frossard
- Department of Neurology, Multiple Sclerosis Unit, Hospital Ramon y Cajal, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| | - Mireia Ferrer
- Statistics and Bioinformatics Unit, Vall d’Hebron Institut de Recerca (VHIR), Barcelona, Spain
| | - Alex Sanchez
- Statistics and Bioinformatics Unit, Vall d’Hebron Institut de Recerca (VHIR), Barcelona, Spain
- Genetics, Microbiology and Statistics Department, Universitat de Barcelona, Barcelona, Spain
| | - Eva Borràs
- Proteomics Unit, Centre de Regulació Genòmica (CRG), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
- Proteomics Unit, Universitat Pompeu Fabra, Barcelona, Spain
| | - Eduard Sabidó
- Proteomics Unit, Centre de Regulació Genòmica (CRG), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
- Proteomics Unit, Universitat Pompeu Fabra, Barcelona, Spain
| | - Carmen Espejo
- Servei de Neurologia, Centre d’Esclerosi Múltiple de Catalunya (Cemcat) Institut de Recerca Vall d’Hebron (VHIR), Hospital Universitari Vall d’Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Xavier Montalban
- Servei de Neurologia, Centre d’Esclerosi Múltiple de Catalunya (Cemcat) Institut de Recerca Vall d’Hebron (VHIR), Hospital Universitari Vall d’Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Manuel Comabella
- Servei de Neurologia, Centre d’Esclerosi Múltiple de Catalunya (Cemcat) Institut de Recerca Vall d’Hebron (VHIR), Hospital Universitari Vall d’Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| |
Collapse
|
9
|
Tallgren A, Kager L, O’Grady G, Tuominen H, Körkkö J, Kuismin O, Feucht M, Wilson C, Behunova J, England E, Kurki MI, Palotie A, Hallman M, Kaarteenaho R, Laccone F, Boztug K, Hinttala R, Uusimaa J. Novel patients with NHLRC2 variants expand the phenotypic spectrum of FINCA disease. Front Neurosci 2023; 17:1123327. [PMID: 37179546 PMCID: PMC10173879 DOI: 10.3389/fnins.2023.1123327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Accepted: 02/08/2023] [Indexed: 05/15/2023] Open
Abstract
Purpose FINCA disease (Fibrosis, Neurodegeneration and Cerebral Angiomatosis, OMIM 618278) is an infantile-onset neurodevelopmental and multiorgan disease. Since our initial report in 2018, additional patients have been described. FINCA is the first human disease caused by recessive variants in the highly conserved NHLRC2 gene. Our previous studies have shown that Nhlrc2-null mouse embryos die during gastrulation, indicating the essential role of the protein in embryonic development. Defect in NHLRC2 leads to cerebral neurodegeneration and severe pulmonary, hepatic and cardiac fibrosis. Despite having a structure suggestive of an enzymatic role and the clinical importance of NHLRC2 in multiple organs, the specific physiological role of the protein is unknown. Methods The clinical histories of five novel FINCA patients diagnosed with whole exome sequencing were reviewed. Segregation analysis of the biallelic, potentially pathogenic NHLRC2 variants was performed using Sanger sequencing. Studies on neuropathology and NHLRC2 expression in different brain regions were performed on autopsy samples of three previously described deceased FINCA patients. Results One patient was homozygous for the pathogenic variant c.442G > T, while the other four were compound heterozygous for this variant and two other pathogenic NHLRC2 gene variants. All five patients presented with multiorgan dysfunction with neurodevelopmental delay, recurrent infections and macrocytic anemia as key features. Interstitial lung disease was pronounced in infancy but often stabilized. Autopsy samples revealed widespread, albeit at a lower intensity than the control, NHLRC2 expression in the brain. Conclusion This report expands on the characteristic clinical features of FINCA disease. Presentation is typically in infancy, and although patients can live to late adulthood, the key clinical and histopathological features are fibrosis, infection susceptibility/immunodeficiency/intellectual disability, neurodevelopmental disorder/neurodegeneration and chronic anemia/cerebral angiomatosis (hence the acronym FINCA) that enable an early diagnosis confirmed by genetic investigations.
Collapse
Affiliation(s)
- Antti Tallgren
- Research Unit of Clinical Medicine and Medical Research Center, Oulu University Hospital, University of Oulu, Oulu, Finland
| | - Leo Kager
- St. Anna Children’s Hospital, Vienna, Austria
- Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, Vienna, Austria
- St. Anna Children’s Cancer Research Institute (CCRI), Vienna, Austria
| | - Gina O’Grady
- Paediatric Neuroservices, Starship Children’s Health, Te Whatu Ora Health New Zealand, Auckland, New Zealand
| | - Hannu Tuominen
- Department of Pathology, Oulu University Hospital, University of Oulu, Oulu, Finland
| | - Jarmo Körkkö
- Center for Intellectual Disability Care, Oulu University Hospital, Oulu, Finland
| | - Outi Kuismin
- Research Unit of Clinical Medicine and Medical Research Center, Oulu University Hospital, University of Oulu, Oulu, Finland
- Department of Clinical Genetics, Oulu University Hospital, Oulu, Finland
| | - Martha Feucht
- Department of Paediatrics, Center for Rare and Complex Epilepsies, Medical University of Vienna, Vienna, Austria
| | - Callum Wilson
- National Metabolic Service, Auckland City Hospital, Auckland, New Zealand
| | - Jana Behunova
- Department of Medical Genetics, Medical University of Vienna, Vienna, Austria
| | - Eleina England
- Mendelian Genomics, Programme in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, United States
| | - Mitja I. Kurki
- Programme in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, United States
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, United States
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, United States
| | - Aarno Palotie
- Programme in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, United States
- Psychiatric and Neurodevelopmental Genetics Unit, Department of Psychiatry, Massachusetts General Hospital, Boston, MA, United States
- Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital, Boston, MA, United States
- Department of Neurology, Massachusetts General Hospital, Boston, MA, United States
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
| | - Mikko Hallman
- Research Unit of Clinical Medicine and Medical Research Center, Oulu University Hospital, University of Oulu, Oulu, Finland
- Clinic for Children and Adolescents, Oulu University Hospital, Oulu, Finland
| | - Riitta Kaarteenaho
- Research Unit of Internal Medicine, University of Oulu, Oulu, Finland
- Center of Internal Medicine and Respiratory Medicine and Medical Research Center Oulu, Oulu University Hospital, Oulu, Finland
| | - Franco Laccone
- Department of Medical Genetics, Medical University of Vienna, Vienna, Austria
| | - Kaan Boztug
- St. Anna Children’s Hospital, Vienna, Austria
- Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, Vienna, Austria
- St. Anna Children’s Cancer Research Institute (CCRI), Vienna, Austria
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Reetta Hinttala
- Research Unit of Clinical Medicine and Medical Research Center, Oulu University Hospital, University of Oulu, Oulu, Finland
- Biocenter Oulu, University of Oulu, Oulu, Finland
| | - Johanna Uusimaa
- Research Unit of Clinical Medicine and Medical Research Center, Oulu University Hospital, University of Oulu, Oulu, Finland
- Clinic for Children and Adolescents, Oulu University Hospital, Oulu, Finland
| |
Collapse
|
10
|
Rike WA, Stern S. Proteins and Transcriptional Dysregulation of the Brain Extracellular Matrix in Parkinson's Disease: A Systematic Review. Int J Mol Sci 2023; 24:ijms24087435. [PMID: 37108598 PMCID: PMC10138539 DOI: 10.3390/ijms24087435] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 04/06/2023] [Accepted: 04/12/2023] [Indexed: 04/29/2023] Open
Abstract
The extracellular matrix (ECM) of the brain is a dynamic structure made up of a vast network of bioactive macromolecules that modulate cellular events. Structural, organizational, and functional changes in these macromolecules due to genetic variation or environmental stressors are thought to affect cellular functions and may result in disease. However, most mechanistic studies to date usually focus on the cellular aspects of diseases and pay less attention to the relevance of the processes governing the dynamic nature of the extracellular matrix in disease pathogenesis. Thus, due to the ECM's diversified biological roles, increasing interest in its involvement in disease, and the lack of sufficient compiled evidence regarding its relationship with Parkinson's disease (PD) pathology, we aimed to compile the existing evidence to boost the current knowledge on the area and provide refined guidance for the future research. Here, in this review, we gathered postmortem brain tissue and induced pluripotent stem cell (iPSC)-related studies from PubMed and Google Scholar to identify, summarize and describe common macromolecular alterations in the expression of brain ECM components in Parkinson's disease (PD). A literature search was conducted up until 10 February 2023. The overall hits from the database and manual search for proteomic and transcriptome studies were 1243 and 1041 articles, respectively. Following a full-text review, 10 articles from proteomic and 24 from transcriptomic studies were found to be eligible for inclusion. According to proteomic studies, proteins such as collagens, fibronectin, annexins, and tenascins were recognized to be differentially expressed in Parkinson's disease. Transcriptomic studies displayed dysregulated pathways including ECM-receptor interaction, focal adhesion, and cell adhesion molecules in Parkinson's disease. A limited number of relevant studies were accessed from our search, indicating that much work remains to be carried out to better understand the roles of the ECM in neurodegeneration and Parkinson's disease. However, we believe that our review will elicit focused primary studies and thus support the ongoing efforts of the discovery and development of diagnostic biomarkers as well as therapeutic agents for Parkinson's disease.
Collapse
Affiliation(s)
- Wote Amelo Rike
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa 3498838, Israel
| | - Shani Stern
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa 3498838, Israel
| |
Collapse
|
11
|
Chithra Y, Dey G, Ghose V, Chandramohan V, Gowthami N, Vasudev V, Srinivas Bharath MM. Mitochondrial Complex I Inhibition in Dopaminergic Neurons Causes Altered Protein Profile and Protein Oxidation: Implications for Parkinson's disease. Neurochem Res 2023:10.1007/s11064-023-03907-x. [PMID: 36964824 DOI: 10.1007/s11064-023-03907-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 02/25/2023] [Accepted: 03/03/2023] [Indexed: 03/26/2023]
Abstract
Mitochondrial dysfunction and oxidative stress are critical to neurodegeneration in Parkinson's disease (PD). Mitochondrial dysfunction in PD entails inhibition of the mitochondrial complex I (CI) in the dopaminergic neurons of substantia nigra. The events contributing to CI inhibition and downstream pathways are not completely elucidated. We conducted proteomic analysis in a dopaminergic neuronal cell line exposed individually to neurotoxic CI inhibitors: rotenone (Rot), paraquat (Pq) and 1-methyl-4-phenylpyridinium (MPP+). Mass spectrometry (MS) revealed the involvement of biological processes including cell death pathways, structural changes and metabolic processes among others, most of which were common across all models. The proteomic changes induced by Pq were significantly higher than those induced by Rot and MPP+. Altered metabolic processes included downregulated mitochondrial proteins such as CI subunits. MS of CI isolated from the models revealed oxidative post-translational modifications with Tryptophan (Trp) oxidation as the predominant modification. Further, 62 peptides in 22 subunits of CI revealed Trp oxidation with 16 subunits common across toxins. NDUFV1 subunit had the greatest number of oxidized Trp and Rot model displayed the highest number of Trp oxidation events compared to the other models. Molecular dynamics simulation (MDS) of NDUFV1 revealed that oxidized Trp 433 altered the local conformation thereby changing the distance between the Fe-S clusters, Fe-S 301(N1a) to Fe-S 502 (N3) and Fe-S 802 (N4) to Fe-S 801 (N5), potentially affecting the efficiency of electron transfer. The events triggered by the neurotoxins represent CI damage, mitochondrial dysfunction and neurodegeneration in PD.
Collapse
Affiliation(s)
- Yogeshachar Chithra
- Department of Bioscience, P.G. Center, Hemagangotri, University of Mysore, Hassan, Karnataka, 573220, India
| | - Gourav Dey
- Institute of Bioinformatics, International Tech Park, Bangalore, 560066, India
| | - Vivek Ghose
- Manipal Academy of Higher Education, Udupi, Karnataka, 576104, India
| | - Vivek Chandramohan
- Department of Biotechnology, Siddaganga Institute of Technology, Tumkur, Karnataka, 572103, India
| | - Niya Gowthami
- Department of Clinical Psychopharmacology and Neurotoxicology, National Institute of Mental Health and Neurosciences (NIMHANS), No. 2900, Hosur Road, Lakkasandra, Bangalore, 560029, India
| | - V Vasudev
- Department of Bioscience, P.G. Center, Hemagangotri, University of Mysore, Hassan, Karnataka, 573220, India
| | - M M Srinivas Bharath
- Department of Clinical Psychopharmacology and Neurotoxicology, National Institute of Mental Health and Neurosciences (NIMHANS), No. 2900, Hosur Road, Lakkasandra, Bangalore, 560029, India.
| |
Collapse
|
12
|
Mao Q, Lin X, Yin Q, Liu P, Zhang Y, Qu S, Xu J, Cheng W, Luo X, Kang L, Taximaimaiti R, Zheng C, Zhang H, Wang X, Ren H, Cao Y, Lin J, Luo X. A significant, functional and replicable risk KTN1 variant block for schizophrenia. Sci Rep 2023; 13:3890. [PMID: 36890161 PMCID: PMC9995530 DOI: 10.1038/s41598-023-27448-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: 10/12/2022] [Accepted: 01/02/2023] [Indexed: 03/10/2023] Open
Abstract
Cortical and subcortical structural alteration has been extensively reported in schizophrenia, including the unusual expansion of gray matter volumes (GMVs) of basal ganglia (BG), especially putamen. Previous genome-wide association studies pinpointed kinectin 1 gene (KTN1) as the most significant gene regulating the GMV of putamen. In this study, the role of KTN1 variants in risk and pathogenesis of schizophrenia was explored. A dense set of SNPs (n = 849) covering entire KTN1 was analyzed in three independent European- or African-American samples (n = 6704) and one mixed European and Asian Psychiatric Genomics Consortium sample (n = 56,418 cases vs. 78,818 controls), to identify replicable SNP-schizophrenia associations. The regulatory effects of schizophrenia-associated variants on the KTN1 mRNA expression in 16 cortical or subcortical regions in two European cohorts (n = 138 and 210, respectively), the total intracranial volume (ICV) in 46 European cohorts (n = 18,713), the GMVs of seven subcortical structures in 50 European cohorts (n = 38,258), and the surface areas (SA) and thickness (TH) of whole cortex and 34 cortical regions in 50 European cohorts (n = 33,992) and eight non-European cohorts (n = 2944) were carefully explored. We found that across entire KTN1, only 26 SNPs within the same block (r2 > 0.85) were associated with schizophrenia across ≥ 2 independent samples (7.5 × 10-5 ≤ p ≤ 0.048). The schizophrenia-risk alleles, which increased significantly risk for schizophrenia in Europeans (q < 0.05), were all minor alleles (f < 0.5), consistently increased (1) the KTN1 mRNA expression in 12 brain regions significantly (5.9 × 10-12 ≤ p ≤ 0.050; q < 0.05), (2) the ICV significantly (6.1 × 10-4 ≤ p ≤ 0.008; q < 0.05), (3) the SA of whole (9.6 × 10-3 ≤ p ≤ 0.047) and two regional cortices potentially (2.5 × 10-3 ≤ p ≤ 0.042; q > 0.05), and (4) the TH of eight regional cortices potentially (0.006 ≤ p ≤ 0.050; q > 0.05), and consistently decreased (1) the BG GMVs significantly (1.8 × 10-19 ≤ p ≤ 0.050; q < 0.05), especially putamen GMV (1.8 × 10-19 ≤ p ≤ 1.0 × 10-4; q < 0.05, (2) the SA of four regional cortices potentially (0.010 ≤ p ≤ 0.048), and (3) the TH of four regional cortices potentially (0.015 ≤ p ≤ 0.049) in Europeans. We concluded that we identified a significant, functional, and robust risk variant block covering entire KTN1 that might play a critical role in the risk and pathogenesis of schizophrenia.
Collapse
Affiliation(s)
- Qiao Mao
- Department of Psychosomatic Medicine, People's Hospital of Deyang City, Deyang, 618000, Sichuan, China
| | - Xiandong Lin
- Laboratory of Radiation Oncology and Radiobiology, Fujian Provincial Cancer Hospital, the Teaching Hospital of Fujian Medical University, Fuzhou, 350014, Fujian, China
| | - Qin Yin
- Department of Respiratory and Critical Care Medicine, Hubei Provincial Hospital of Integrated Chinese and Western Medicine, Wuhan, 430000, Hubei, China
| | - Ping Liu
- Department of Psychosomatic Medicine, People's Hospital of Deyang City, Deyang, 618000, Sichuan, China
| | - Yong Zhang
- Tianjin Mental Health Center, Tianjin, 300222, China
| | - Shihao Qu
- Zhuhai Center for Maternal and Child Health Care, Zhuhai, Guangdong, 519001, China
| | - Jianying Xu
- Zhuhai Center for Maternal and Child Health Care, Zhuhai, Guangdong, 519001, China
| | - Wenhong Cheng
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China
| | - Xinqun Luo
- Department of Neurosurgery, The First Hospital, Fujian Medical University, Fuzhou, 350004, Fujian, China
| | - Longli Kang
- Key Laboratory for Molecular Genetic Mechanisms and Intervention Research On High Altitude Diseases of Tibet Autonomous Region, Xizang Minzu University School of Medicine, Xiangyang, 712082, Shaanxi, China
| | - Reyisha Taximaimaiti
- Department of Neurology, Shanghai Tongren Hospital, Shanghai Jiao Tong University, Shanghai, 200080, China
| | - Chengchou Zheng
- Minqing Psychiatric Hospital, Minqing, 350800, Fujian, China
| | - Huihao Zhang
- The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350001, China
| | - Xiaoping Wang
- Department of Neurology, The 1st People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 201620, USA
| | - Honggang Ren
- Department of Internal Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuping Cao
- Department of Psychiatry, Second Xiangya Hospital, Central South University, China National Clinical Research Center On Mental Disorders, China National Technology Institute On Mental Disorders, Changsha, 410011, Hunan, China.
| | - Jie Lin
- Fujian Center for Disease Control and Prevention, Fuzhou, 350012, Fujian, China.
- Fujian Institute of Preventive Medicine, Fuzhou, 350012, Fujian, China.
| | - Xingguang Luo
- Beijing Huilongguan Hospital, Peking University Huilongguan School of Clinical Medicine, Beijing, 100096, China.
| |
Collapse
|
13
|
Functional pleiotropy of calcium binding protein Regucalcin in signaling and diseases. Cell Signal 2023; 102:110533. [PMID: 36442591 DOI: 10.1016/j.cellsig.2022.110533] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 11/17/2022] [Accepted: 11/21/2022] [Indexed: 11/27/2022]
Abstract
Regucalcin (Mr ∼ 33.38 kDa) is a calcium binding protein, discovered in rat liver. In humans, gene for regucalcin is located on chromosome-11 (p11.3-q11.2) consisting of seven exons and six introns. The protein differs from other calcium binding protein in the way that it lacks EF-hand motif of calcium binding domain. It is also called as Senescence Marker Protein-30 (SMP-30) as previously its weight assumes to be 30 kDa and expression of this protein decreases with aging in androgen independent manner. Among vertebrates, it is a highly conserved protein showing gene homology in Drosophila, Xenopus, fireflies and others too. It is primarily expressed in liver and kidney in addition to brain, lungs, and skeletal muscles. Regucalcin acts as a Ca2+ regulatory protein and controls various cellular functions in liver and other organs. It suppresses protein phosphatase, protein kinase, DNA and RNA synthesis. Published evidences suggest regucalcin to be a reliable biomarker in various disorders of liver, kidney, brain and ocular. In over expressed state, it subdues apoptosis in cloned rat hepatoma cells and also induces hyperlipidemia and osteoblastogenesis by regulating various factors. Owing to the multi-functionality of regucalcin this review is presented to elaborate its importance in order to understand its involvement in cellular signaling during various pathologies.
Collapse
|
14
|
Jang Y, Pletnikova O, Troncoso JC, Pantelyat AY, Dawson TM, Rosenthal LS, Na CH. Mass Spectrometry-Based Proteomics Analysis of Human Substantia Nigra From Parkinson's Disease Patients Identifies Multiple Pathways Potentially Involved in the Disease. Mol Cell Proteomics 2023; 22:100452. [PMID: 36423813 PMCID: PMC9792365 DOI: 10.1016/j.mcpro.2022.100452] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Revised: 10/26/2022] [Accepted: 11/18/2022] [Indexed: 11/23/2022] Open
Abstract
Parkinson's disease (PD) is the second most prevalent neurodegenerative disorder characterized by the loss of dopaminergic neurons in the substantia nigra (SN) of the brain. Despite decades of studies, the precise pathogenic mechanism of PD is still elusive. An unbiased proteomic analysis of PD patient's brain allows the identification of critical proteins and molecular pathways that lead to dopamine cell death and α-synuclein deposition and the resulting devastating clinical symptoms. In this study, we conducted an in-depth proteome analysis of human SN tissues from 15 PD patients and 15 healthy control individuals combining Orbitrap mass spectrometry with the isobaric tandem mass tag-based multiplexing technology. We identified 10,040 proteins with 1140 differentially expressed proteins in the SN of PD patients. Pathway analysis showed that the ribosome pathway was the most enriched one, followed by gamma-aminobutyric acidergic synapse, retrograde endocannabinoid signaling, cell adhesion molecules, morphine addiction, Prion disease, and PD pathways. Strikingly, the majority of the proteins enriched in the ribosome pathway were mitochondrial ribosomal proteins (mitoribosomes). The subsequent protein-protein interaction analysis and the weighted gene coexpression network analysis confirmed that the mitoribosome is the most enriched protein cluster. Furthermore, the mitoribosome was also identified in our analysis of a replication set of ten PD and nine healthy control SN tissues. This study provides potential disease pathways involved in PD and paves the way to study further the pathogenic mechanism of PD.
Collapse
Affiliation(s)
- Yura Jang
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA; Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Olga Pletnikova
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Juan C Troncoso
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA; Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Alexander Y Pantelyat
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Ted M Dawson
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA; Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA; Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA; Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA; Adrienne Helis Malvin Medical Research Foundation, New Orleans, Louisiana, USA; Diana Helis Henry Medical Research Foundation, New Orleans, Louisiana, USA.
| | - Liana S Rosenthal
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
| | - Chan Hyun Na
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA; Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
| |
Collapse
|
15
|
Caligiore D, Giocondo F, Silvetti M. The Neurodegenerative Elderly Syndrome (NES) hypothesis: Alzheimer and Parkinson are two faces of the same disease. IBRO Neurosci Rep 2022; 13:330-343. [PMID: 36247524 PMCID: PMC9554826 DOI: 10.1016/j.ibneur.2022.09.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 09/07/2022] [Accepted: 09/21/2022] [Indexed: 11/25/2022] Open
Abstract
Increasing evidence suggests that Alzheimer's disease (AD) and Parkinson's disease (PD) share monoamine and alpha-synuclein (αSyn) dysfunctions, often beginning years before clinical manifestations onset. The triggers for these impairments and the causes leading these early neurodegenerative processes to become AD or PD remain unclear. We address these issues by proposing a radically new perspective to frame AD and PD: they are different manifestations of one only disease we call "Neurodegenerative Elderly Syndrome (NES)". NES goes through three phases. The seeding stage, which starts years before clinical signs, and where the part of the brain-body affected by the initial αSyn and monoamine dysfunctions, influences the future possible progression of NES towards PD or AD. The compensatory stage, where the clinical symptoms are still silent thanks to compensatory mechanisms keeping monoamine concentrations homeostasis. The bifurcation stage, where NES becomes AD or PD. We present recent literature supporting NES and discuss how this hypothesis could radically change the comprehension of AD and PD comorbidities and the design of novel system-level diagnostic and therapeutic actions.
Collapse
Affiliation(s)
- Daniele Caligiore
- Computational and Translational Neuroscience Laboratory, Institute of Cognitive Sciences and Technologies, National Research Council (CTNLab-ISTC-CNR), Via San Martino della Battaglia 44, Rome 00185, Italy
- AI2Life s.r.l., Innovative Start-Up, ISTC-CNR Spin-Off, Via Sebino 32, Rome 00199, Italy
| | - Flora Giocondo
- Laboratory of Embodied Natural and Artificial Intelligence, Institute of Cognitive Sciences and Technologies, National Research Council (LENAI-ISTC-CNR), Via San Martino della Battaglia 44, Rome 00185, Italy
| | - Massimo Silvetti
- Computational and Translational Neuroscience Laboratory, Institute of Cognitive Sciences and Technologies, National Research Council (CTNLab-ISTC-CNR), Via San Martino della Battaglia 44, Rome 00185, Italy
| |
Collapse
|
16
|
Li Y, Zhang Y, Jiang G, Wang Y, He C, Zhao X, Liu L, Li L. Case report: novel mutations of NDUFS6 and NHLRC2 genes potentially cause the quick postnatal death of a Chinese Hani minority neonate with mitochondrial complex I deficiency and FINCA syndrome. Medicine (Baltimore) 2022; 101:e29239. [PMID: 35801790 PMCID: PMC9259100 DOI: 10.1097/md.0000000000029239] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
INTRODUCTION Mitochondrial complex I deficiency (MCID) and abbFINCA syndrome are lethal congenital diseases and cases in the neonatal period are rarely reported. Here, we identified a Chinese Hani minority neonate with rare MCID and FINCA syndrome. This study was to analyze the clinical manifestations and pathogenic gene variations, and to investigate causes of quick postnatal death of patient and possible molecular pathogenic mechanisms. PATIENT CONCERNS A 17-day-old patient had reduced muscle tension, diminished primitive reflexes, significantly abnormal blood gas analysis, and progressively increased blood lactate and blood glucose. Imaging studies revealed pneumonia, pulmonary hypertension, and brain abnormalities. DIAGNOSIS Whole-exome sequencing revealed that the NDUFS6 gene of the patient carried c. 344G > T (p.C115F) novel homozygous variation, and the NHLRC2 gene carried c. 1749C > G (p.F583L) and c. 2129C > T (p.T710M) novel compound heterozygous variation. INTERVENTIONS AND OUTCOMES The patient was given endotracheal intubation, respiratory support, high-frequency ventilation, antishock therapy, as well as iNO and Alprostadil to reduce pulmonary hypertension and maintain homeostatic equilibrium. However, the patient was critically ill and died in 27 days. CONCLUSION The patient has MCID due to a novel mutation in NDUFS6 and FINCA syndrome due to novel mutations in NHLRC2, which is the main reason for the rapid onset and quick death of the patient.
Collapse
Affiliation(s)
- Yangfang Li
- Department of Neonatology, Kunming Children’s Hospital, Kunming 650228, Yunnan, China
| | - Yu Zhang
- Kunming Key Laboratory of Children Infection and Immunity, Yunnan Key Laboratory of Children’s Major Disease Research, Yunnan Institute of Pediatrics, Kunming Children’s Hospital, Kunming 650228, Yunnan, China
| | - Gengpan Jiang
- Department of Neonatology, Kunming Children’s Hospital, Kunming 650228, Yunnan, China
| | - Yan Wang
- Kunming Key Laboratory of Children Infection and Immunity, Yunnan Key Laboratory of Children’s Major Disease Research, Yunnan Institute of Pediatrics, Kunming Children’s Hospital, Kunming 650228, Yunnan, China
| | - Canlin He
- Department of Neonatology, Kunming Children’s Hospital, Kunming 650228, Yunnan, China
| | - Xiaofen Zhao
- Department of Neonatology, Kunming Children’s Hospital, Kunming 650228, Yunnan, China
| | - Ling Liu
- Department of Neonatology, Kunming Children’s Hospital, Kunming 650228, Yunnan, China
| | - Li Li
- Kunming Key Laboratory of Children Infection and Immunity, Yunnan Key Laboratory of Children’s Major Disease Research, Yunnan Institute of Pediatrics, Kunming Children’s Hospital, Kunming 650228, Yunnan, China
- *Correspondence: Li Li, Kunming Key Laboratory of Children Infection and Immunity, Yunnan Key Laboratory of Children’s Major Disease Research, Yunnan Institute of Pediatrics, Kunming Children’s Hospital, Kunming 650228, Yunnan, China (e-mail: )
| |
Collapse
|
17
|
Ruffini N, Klingenberg S, Heese R, Schweiger S, Gerber S. The Big Picture of Neurodegeneration: A Meta Study to Extract the Essential Evidence on Neurodegenerative Diseases in a Network-Based Approach. Front Aging Neurosci 2022; 14:866886. [PMID: 35832065 PMCID: PMC9271745 DOI: 10.3389/fnagi.2022.866886] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 05/13/2022] [Indexed: 12/12/2022] Open
Abstract
The common features of all neurodegenerative diseases, including Alzheimer's disease, Parkinson's disease, Amyotrophic Lateral Sclerosis (ALS), and Huntington's disease, are the accumulation of aggregated and misfolded proteins and the progressive loss of neurons, leading to cognitive decline and locomotive dysfunction. Still, they differ in their ultimate manifestation, the affected brain region, and the kind of proteinopathy. In the last decades, a vast number of processes have been described as associated with neurodegenerative diseases, making it increasingly harder to keep an overview of the big picture forming from all those data. In this meta-study, we analyzed genomic, transcriptomic, proteomic, and epigenomic data of the aforementioned diseases using the data of 234 studies in a network-based approach to study significant general coherences but also specific processes in individual diseases or omics levels. In the analysis part, we focus on only some of the emerging findings, but trust that the meta-study provided here will be a valuable resource for various other researchers focusing on specific processes or genes contributing to the development of neurodegeneration.
Collapse
Affiliation(s)
- Nicolas Ruffini
- Institute of Human Genetics, University Medical Center, Johannes Gutenberg University, Mainz, Germany
- Leibniz Institute for Resilience Research, Leibniz Association, Mainz, Germany
| | - Susanne Klingenberg
- Institute of Human Genetics, University Medical Center, Johannes Gutenberg University, Mainz, Germany
| | - Raoul Heese
- Fraunhofer Institute for Industrial Mathematics (ITWM), Kaiserslautern, Germany
| | - Susann Schweiger
- Institute of Human Genetics, University Medical Center, Johannes Gutenberg University, Mainz, Germany
| | - Susanne Gerber
- Institute of Human Genetics, University Medical Center, Johannes Gutenberg University, Mainz, Germany
| |
Collapse
|
18
|
Holland N, Robbins TW, Rowe JB. The role of noradrenaline in cognition and cognitive disorders. Brain 2021; 144:2243-2256. [PMID: 33725122 PMCID: PMC8418349 DOI: 10.1093/brain/awab111] [Citation(s) in RCA: 94] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 01/08/2021] [Accepted: 01/23/2021] [Indexed: 01/09/2023] Open
Abstract
Many aspects of cognition and behaviour are regulated by noradrenergic projections to the forebrain originating from the locus coeruleus, acting through alpha and beta adrenoreceptors. Loss of these projections is common in neurodegenerative diseases and contributes to their cognitive and behavioural deficits. We review the evidence for a noradrenergic modulation of cognition in its contribution to Alzheimer's disease, Parkinson's disease and other cognitive disorders. We discuss the advances in human imaging and computational methods that quantify the locus coeruleus and its function in humans, and highlight the potential for new noradrenergic treatment strategies.
Collapse
Affiliation(s)
- Negin Holland
- Department of Clinical Neurosciences, University of Cambridge, Cambridge CB2 0SZ, UK
| | - Trevor W Robbins
- Department of Psychology, University of Cambridge, Cambridge CB2 3EB, UK
- Behavioural and Clinical Neuroscience Institute, University of Cambridge, Cambridge CB2 3EB, UK
| | - James B Rowe
- Department of Clinical Neurosciences, University of Cambridge, Cambridge CB2 0SZ, UK
- Behavioural and Clinical Neuroscience Institute, University of Cambridge, Cambridge CB2 3EB, UK
- MRC Cognition and Brain Sciences Unit, University of Cambridge, Cambridge CB2 7EF, UK
| |
Collapse
|
19
|
Nalamalapu RR, Yue M, Stone AR, Murphy S, Saha MS. The tweety Gene Family: From Embryo to Disease. Front Mol Neurosci 2021; 14:672511. [PMID: 34262434 PMCID: PMC8273234 DOI: 10.3389/fnmol.2021.672511] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 05/18/2021] [Indexed: 12/31/2022] Open
Abstract
The tweety genes encode gated chloride channels that are found in animals, plants, and even simple eukaryotes, signifying their deep evolutionary origin. In vertebrates, the tweety gene family is highly conserved and consists of three members—ttyh1, ttyh2, and ttyh3—that are important for the regulation of cell volume. While research has elucidated potential physiological functions of ttyh1 in neural stem cell maintenance, proliferation, and filopodia formation during neural development, the roles of ttyh2 and ttyh3 are less characterized, though their expression patterns during embryonic and fetal development suggest potential roles in the development of a wide range of tissues including a role in the immune system in response to pathogen-associated molecules. Additionally, members of the tweety gene family have been implicated in various pathologies including cancers, particularly pediatric brain tumors, and neurodegenerative diseases such as Alzheimer’s and Parkinson’s disease. Here, we review the current state of research using information from published articles and open-source databases on the tweety gene family with regard to its structure, evolution, expression during development and adulthood, biochemical and cellular functions, and role in human disease. We also identify promising areas for further research to advance our understanding of this important, yet still understudied, family of genes.
Collapse
Affiliation(s)
- Rithvik R Nalamalapu
- Department of Biology, College of William and Mary, Williamsburg, VA, United States
| | - Michelle Yue
- Department of Biology, College of William and Mary, Williamsburg, VA, United States
| | - Aaron R Stone
- Department of Biology, College of William and Mary, Williamsburg, VA, United States
| | - Samantha Murphy
- Department of Biology, College of William and Mary, Williamsburg, VA, United States
| | - Margaret S Saha
- Department of Biology, College of William and Mary, Williamsburg, VA, United States
| |
Collapse
|
20
|
Lashuel HA, Novello S. Lewy body-associated proteins: victims, instigators, or innocent bystanders? The case of AIMP2 and alpha-synuclein. Neurobiol Dis 2021; 156:105417. [PMID: 34102275 DOI: 10.1016/j.nbd.2021.105417] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 06/01/2021] [Accepted: 06/02/2021] [Indexed: 01/21/2023] Open
Abstract
Lewy bodies (LBs), one of the neuropathological defining hallmarks of Parkinson's disease (PD), are composed of a complex mixture of alpha-synuclein (aSyn) filaments and hundreds of proteins, lipids, and membranous organelles. However, these proteins' role in aSyn aggregation and the biogenesis of LBs remains poorly understood. Previous studies have focused on investigating the role of these proteins as modifiers of aSyn aggregation, inclusion formation, and toxicity; very often, one protein at a time. In a recent study, Ham et al. suggest that one of these proteins, aminoacyl tRNA synthase complex-interacting multifunctional protein 2 (AIMP2), plays a primary role in the initiation of aSyn aggregation and is essential for aSyn inclusion formation and toxicity in cells and several models of synucleinopathies (Ham et al., 2020). Based on in vitro aggregation studies, they proposed a model in which AIMP2 self-associates to form amyloid-like aggregates that interact with monomeric aSyn and catalyze/seed the formation of aSyn fibrils and, eventually, LB-like inclusions. Herein, we present a critical analysis of their results and conclusions, review previous studies on AIMP2 aggregation, and reexamine the role of AIMP2 in regulating aSyn inclusion formation and clearance and aSyn-induced neurodegeneration in Parkinson's disease. We conclude by presenting lesson learned and recommendations on experimental factors and approaches that should be considered in future studies aimed at investigating the potential of targeting LBs-associated proteins, including AIMP2, for developing therapies to treat PD and other synucleinopathies.
Collapse
Affiliation(s)
- Hilal A Lashuel
- Laboratory of Molecular and Chemical Biology of Neurodegeneration, School of Life Sciences, Brain Mind Institute, Ecole Polytechnique Fédérale de Lausanne (EPFL), CH-1015 Lausanne, Switzerland.
| | - Salvatore Novello
- Laboratory of Molecular and Chemical Biology of Neurodegeneration, School of Life Sciences, Brain Mind Institute, Ecole Polytechnique Fédérale de Lausanne (EPFL), CH-1015 Lausanne, Switzerland
| |
Collapse
|
21
|
Fanning S, Selkoe D, Dettmer U. Vesicle trafficking and lipid metabolism in synucleinopathy. Acta Neuropathol 2021; 141:491-510. [PMID: 32607605 DOI: 10.1007/s00401-020-02177-z] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Revised: 06/05/2020] [Accepted: 06/06/2020] [Indexed: 12/12/2022]
Abstract
The neuronal protein α-synuclein (αS) is central to the pathogenesis of Parkinson's disease and other progressive brain diseases such as Lewy body dementia and multiple system atrophy. These diseases, collectively referred to as 'synucleinopathies', have long been considered purely proteinopathies: diseases characterized by the misfolding of a protein into small and large aggregates mainly consisting of that protein (in this case: α-synuclein). However, recent morphological insights into Lewy bodies, the hallmark neuropathology of human synucleinopathies, suggests these lesions are also rich in vesicles and other membranous organelles. Moreover, αS physiology and pathology are both strongly associated with various aspects of intracellular vesicle trafficking and lipid biology. αS physiologically binds to synaptic and other small vesicles, and several functions of αS in regulating vesicle biology have been proposed. Familial PD-linked αS excess and missense mutations have been shown to impair vesicle trafficking and alter lipid homeostasis. On the other hand, vesicle trafficking and lipid-related genes have emerged as Parkinson's risk factors, suggesting a bidirectional relationship. The answer to the question "Does abnormal αS accumulation cause impaired vesicle trafficking and lipid dyshomeostasis or is αS aggregation the consequence of such impairments?" may be "Both". Here, we review current knowledge of the αS-lipid and αS-vesicle trafficking interplay, with a special focus on Parkinson's disease and Lewy body dementia.
Collapse
|
22
|
Luo X, Guo X, Luo X, Tan Y, Zhang P, Yang K, Xie T, Shi J, Zhang Y, Xu J, Zuo L, Li CSR. Significant, replicable, and functional associations between KTN1 variants and alcohol and drug codependence. Addict Biol 2021; 26:e12888. [PMID: 32115811 PMCID: PMC7641293 DOI: 10.1111/adb.12888] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Revised: 02/04/2020] [Accepted: 02/13/2020] [Indexed: 01/01/2023]
Abstract
The gray matter volume (GMV) of the putamen has been reported to be regulated by kinectin 1 gene (KTN1). As a hub of the dopaminergic circuit, the putamen is widely implicated in the etiological processes of substance use disorders (SUD). Here, we aimed to identify robust and reliable associations between KTN1 SNPs and SUD across multiple samples. We examined the associations between SUD and KTN1 SNPs in four independent population-based or family-based samples (n = 10,209). The potential regulatory effects of the risk alleles on the putamen GMVs, the effects of alcohol, nicotine, marijuana and cocaine on KTN1 mRNA expression, and the relationship between KTN1 mRNA expression and SUD were explored. We found that a total of 23 SNPs were associated with SUD across at least two independent samples (1.4 × 10-4 ≤ p ≤ 0.049), including one SNP (rs12895072) across three samples (8.8 × 10-3 ≤ p ≤ 0.049). Four other SNPs were significantly or suggestively associated with SUD only in European-Australians (4.8 × 10-4 ≤ p ≤ 0.058). All of the SUD-risk alleles of these 27 SNPs increased (β > 0) the putamen GMVs and represented major alleles (f > 0.5) in Europeans. Twenty-two SNPs were potentially biologically functional. Alcohol, nicotine and cocaine significantly affected the KTN1 mRNA expression, and the KTN1 mRNA was differentially expressed between nicotine or cocaine dependent and control subjects. We concluded that there was a replicable and robust relationship among the KTN1 variants, KTN1 mRNA expression, putamen GMVs, molecular effects of substances, and SUD, suggesting that some risk KTN1 alleles might increase kinectin 1 expression in the putamen, altering putamen structures and functions, and leading to SUD.
Collapse
Affiliation(s)
- Xingguang Luo
- Biological Psychiatry Research Center, Beijing Huilongguan Hospital, Beijing 100096, China
| | - Xiaoyun Guo
- Department of Psychiatry, Shanghai Mental Health Center, Shanghai 200030, China
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Xingqun Luo
- Department of Clinical Medicine, College of Integrated Traditional Chinese and Western Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350004, China
| | - Yunlong Tan
- Biological Psychiatry Research Center, Beijing Huilongguan Hospital, Beijing 100096, China
| | - Ping Zhang
- Biological Psychiatry Research Center, Beijing Huilongguan Hospital, Beijing 100096, China
| | - Kebing Yang
- Biological Psychiatry Research Center, Beijing Huilongguan Hospital, Beijing 100096, China
| | - Ting Xie
- Biological Psychiatry Research Center, Beijing Huilongguan Hospital, Beijing 100096, China
| | - Jing Shi
- Biological Psychiatry Research Center, Beijing Huilongguan Hospital, Beijing 100096, China
| | - Yong Zhang
- Department of Psychiatry, Tianjin Mental Health Center, Tianjin 300222, China
| | - Jianying Xu
- Department of Obstetrics and Gynecology, Zhuhai Municipal Maternal and Children’s Health Hospital, Zhuhai, Guangdong 519000, China
| | - Lingjun Zuo
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Chiang-Shan R. Li
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT 06510, USA
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT 06510, USA
| |
Collapse
|
23
|
Reverse engineering Lewy bodies: how far have we come and how far can we go? Nat Rev Neurosci 2021; 22:111-131. [PMID: 33432241 DOI: 10.1038/s41583-020-00416-6] [Citation(s) in RCA: 104] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/23/2020] [Indexed: 12/19/2022]
Abstract
Lewy bodies (LBs) are α-synuclein (α-syn)-rich intracellular inclusions that are an important pathological hallmark of Parkinson disease and several other neurodegenerative diseases. Increasing evidence suggests that the aggregation of α-syn has a central role in LB formation and is one of the key processes that drive neurodegeneration and pathology progression in Parkinson disease. However, little is known about the mechanisms underlying the formation of LBs, their biochemical composition and ultrastructural properties, how they evolve and spread with disease progression, and their role in neurodegeneration. In this Review, we discuss current knowledge of α-syn pathology, including the biochemical, structural and morphological features of LBs observed in different brain regions. We also review the most used cellular and animal models of α-syn aggregation and pathology spreading in relation to the extent to which they reproduce key features of authentic LBs. Finally, we provide important insights into molecular and cellular determinants of LB formation and spreading, and highlight the critical need for more detailed and systematic characterization of α-syn pathology, at both the biochemical and structural levels. This would advance our understanding of Parkinson disease and other neurodegenerative diseases and allow the development of more-reliable disease models and novel effective therapeutic strategies.
Collapse
|
24
|
Hiltunen AE, Kangas SM, Ohlmeier S, Pietilä I, Hiltunen J, Tanila H, McKerlie C, Govindan S, Tuominen H, Kaarteenaho R, Hallman M, Uusimaa J, Hinttala R. Variant in NHLRC2 leads to increased hnRNP C2 in developing neurons and the hippocampus of a mouse model of FINCA disease. Mol Med 2020; 26:123. [PMID: 33297935 PMCID: PMC7724728 DOI: 10.1186/s10020-020-00245-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 11/18/2020] [Indexed: 12/13/2022] Open
Abstract
Background FINCA disease is a pediatric cerebropulmonary disease caused by variants in the NHL repeat-containing 2 (NHLRC2) gene. Neurological symptoms are among the first manifestations of FINCA disease, but the consequences of NHLRC2 deficiency in the central nervous system are currently unexplored. Methods The orthologous mouse gene is essential for development, and its complete loss leads to early embryonic lethality. In the current study, we used CRISPR/Cas9 to generate an Nhlrc2 knockin (KI) mouse line, harboring the FINCA patient missense mutation (c.442G > T, p.Asp148Tyr). A FINCA mouse model, resembling the compound heterozygote genotype of FINCA patients, was obtained by crossing the KI and Nhlrc2 knockout mouse lines. To reveal NHLRC2-interacting proteins in developing neurons, we compared cortical neuronal precursor cells of E13.5 FINCA and wild-type mouse embryos by two-dimensional difference gel electrophoresis. Results Despite the significant decrease in NHLRC2, the mice did not develop severe early onset multiorgan disease in either sex. We discovered 19 altered proteins in FINCA neuronal precursor cells; several of which are involved in vesicular transport pathways and actin dynamics which have been previously reported in other cell types including human to have an association with dysfunctional NHLRC2. Interestingly, isoform C2 of hnRNP C1/C2 was significantly increased in both developing neurons and the hippocampus of adult female FINCA mice, connecting NHLRC2 dysfunction with accumulation of RNA binding protein. Conclusions We describe here the first NHLRC2-deficient mouse model to overcome embryonic lethality, enabling further studies on predisposing and causative mechanisms behind FINCA disease. Our novel findings suggest that disrupted RNA metabolism may contribute to the neurodegeneration observed in FINCA patients.
Collapse
Affiliation(s)
- Anniina E Hiltunen
- Medical Research Center Oulu and PEDEGO Research Unit, University of Oulu and Oulu University Hospital, PO Box 5000, 90014, Oulu, Finland. .,Biocenter Oulu, University of Oulu, Oulu, Finland.
| | - Salla M Kangas
- Medical Research Center Oulu and PEDEGO Research Unit, University of Oulu and Oulu University Hospital, PO Box 5000, 90014, Oulu, Finland.,Biocenter Oulu, University of Oulu, Oulu, Finland
| | - Steffen Ohlmeier
- Proteomics Core Facility, Biocenter Oulu, Faculty of Biochemistry and Molecular Medicine, University of Oulu, PO Box 5400, Oulu, 90014, Finland
| | - Ilkka Pietilä
- Medical Research Center Oulu and PEDEGO Research Unit, University of Oulu and Oulu University Hospital, PO Box 5000, 90014, Oulu, Finland.,Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, Rudbeck Laboratory, Uppsala, Sweden
| | - Jori Hiltunen
- Medical Research Center Oulu and PEDEGO Research Unit, University of Oulu and Oulu University Hospital, PO Box 5000, 90014, Oulu, Finland
| | - Heikki Tanila
- A.I. Virtanen Institute, University of Eastern Finland, Kuopio, Finland
| | - Colin McKerlie
- The Hospital for Sick Children, Toronto, Canada.,Faculty of Medicine, University of Toronto, Toronto, Canada
| | - Subashika Govindan
- Tissue Engineering Laboratory, Hepia/HES-SO, University of Applied Sciences Western Switzerland, Geneva, Switzerland
| | - Hannu Tuominen
- Department of Pathology, Cancer and Translational Medicine Research Unit, University of Oulu, Oulu, Finland.,Department of Pathology, Oulu University Hospital, Oulu, Finland
| | - Riitta Kaarteenaho
- Research Unit of Internal Medicine, Respiratory Research, University of Oulu, Oulu, Finland.,Medical Research Center Oulu and Unit of Internal Medicine and Respiratory Medicine, Oulu University Hospital, Oulu, Finland
| | - Mikko Hallman
- Medical Research Center Oulu and PEDEGO Research Unit, University of Oulu and Oulu University Hospital, PO Box 5000, 90014, Oulu, Finland
| | - Johanna Uusimaa
- Medical Research Center Oulu and PEDEGO Research Unit, University of Oulu and Oulu University Hospital, PO Box 5000, 90014, Oulu, Finland.,Clinic for Children and Adolescents, Paediatric Neurology Unit, Oulu University Hospital, Oulu, Finland
| | - Reetta Hinttala
- Medical Research Center Oulu and PEDEGO Research Unit, University of Oulu and Oulu University Hospital, PO Box 5000, 90014, Oulu, Finland.,Biocenter Oulu, University of Oulu, Oulu, Finland
| |
Collapse
|
25
|
Ruffini N, Klingenberg S, Schweiger S, Gerber S. Common Factors in Neurodegeneration: A Meta-Study Revealing Shared Patterns on a Multi-Omics Scale. Cells 2020; 9:E2642. [PMID: 33302607 PMCID: PMC7764447 DOI: 10.3390/cells9122642] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 11/24/2020] [Accepted: 12/04/2020] [Indexed: 02/06/2023] Open
Abstract
Neurodegenerative diseases such as Alzheimer's disease (AD), Parkinson's disease (PD), Huntington's disease (HD), and amyotrophic lateral sclerosis (ALS) are heterogeneous, progressive diseases with frequently overlapping symptoms characterized by a loss of neurons. Studies have suggested relations between neurodegenerative diseases for many years (e.g., regarding the aggregation of toxic proteins or triggering endogenous cell death pathways). We gathered publicly available genomic, transcriptomic, and proteomic data from 177 studies and more than one million patients to detect shared genetic patterns between the neurodegenerative diseases on three analyzed omics-layers. The results show a remarkably high number of shared differentially expressed genes between the transcriptomic and proteomic levels for all conditions, while showing a significant relation between genomic and proteomic data between AD and PD and AD and ALS. We identified a set of 139 genes being differentially expressed in several transcriptomic experiments of all four diseases. These 139 genes showed overrepresented gene ontology (GO) Terms involved in the development of neurodegeneration, such as response to heat and hypoxia, positive regulation of cytokines and angiogenesis, and RNA catabolic process. Furthermore, the four analyzed neurodegenerative diseases (NDDs) were clustered by their mean direction of regulation throughout all transcriptomic studies for this set of 139 genes, with the closest relation regarding this common gene set seen between AD and HD. GO-Term and pathway analysis of the proteomic overlap led to biological processes (BPs), related to protein folding and humoral immune response. Taken together, we could confirm the existence of many relations between Alzheimer's disease, Parkinson's disease, Huntington's disease, and amyotrophic lateral sclerosis on transcriptomic and proteomic levels by analyzing the pathways and GO-Terms arising in these intersections. The significance of the connection and the striking relation of the results to processes leading to neurodegeneration between the transcriptomic and proteomic data for all four analyzed neurodegenerative diseases showed that exploring many studies simultaneously, including multiple omics-layers of different neurodegenerative diseases simultaneously, holds new relevant insights that do not emerge from analyzing these data separately. Furthermore, the results shed light on processes like the humoral immune response that have previously been described only for certain diseases. Our data therefore suggest human patients with neurodegenerative diseases should be addressed as complex biological systems by integrating multiple underlying data sources.
Collapse
Affiliation(s)
- Nicolas Ruffini
- Institute for Human Genetics, University Medical Center, Johannes Gutenberg University, 55131 Mainz, Germany; (N.R.); (S.K.); (S.S.)
- Leibniz Institute for Resilience Research, Leibniz Association, Wallstraße 7, 55122 Mainz, Germany
| | - Susanne Klingenberg
- Institute for Human Genetics, University Medical Center, Johannes Gutenberg University, 55131 Mainz, Germany; (N.R.); (S.K.); (S.S.)
| | - Susann Schweiger
- Institute for Human Genetics, University Medical Center, Johannes Gutenberg University, 55131 Mainz, Germany; (N.R.); (S.K.); (S.S.)
| | - Susanne Gerber
- Institute for Human Genetics, University Medical Center, Johannes Gutenberg University, 55131 Mainz, Germany; (N.R.); (S.K.); (S.S.)
| |
Collapse
|
26
|
Mao Q, Wang X, Chen B, Fan L, Wang S, Zhang Y, Lin X, Cao Y, Wu YC, Ji J, Xu J, Zheng J, Zhang H, Zheng C, Chen W, Cheng W, Luo X, Wang K, Zuo L, Kang L, Li CSR, Luo X. KTN1 Variants Underlying Putamen Gray Matter Volumes and Parkinson's Disease. Front Neurosci 2020; 14:651. [PMID: 32655362 PMCID: PMC7324786 DOI: 10.3389/fnins.2020.00651] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Accepted: 05/26/2020] [Indexed: 11/13/2022] Open
Abstract
Background Selective loss of dopaminergic neurons and diminished putamen gray matter volume (GMV) represents a central feature of Parkinson’s disease (PD). Recent studies have reported specific effects of kinectin 1 gene (KTN1) variants on the putamen GMV. Objective To examine the relationship of KTN1 variants, KTN1 mRNA expression in the putamen and substantia nigra pars compacta (SNc), putamen GMV, and PD. Methods We examined the associations between PD and a total of 1847 imputed KTN1 single nucleotide polymorphisms (SNPs) in one discovery sample [2,000 subjects with PD vs. 1,986 healthy controls (HC)], and confirmed the nominally significant associations (p < 0.05) in two replication samples (900 PD vs. 867 HC, and 940 PD vs. 801 HC, respectively). The regulatory effects of risk variants on the KTN1 mRNA expression in putamen and SNc and the putamen GMV were tested. We also quantified the expression levels of KTN1 mRNA in the putamen and/or SNc for comparison between PD and HC in five independent cohorts. Results Six replicable and two non-replicable KTN1-PD associations were identified (0.009 ≤ p ≤ 0.049). The major alleles of five SNPs, including rs12880292, rs8017172, rs17253792, rs945270, and rs4144657, significantly increased risk for PD (0.020 ≤ p ≤ 0.049) and putamen GMVs (19.08 ≤ β ≤ 60.38; 2.82 ≤ Z ≤ 15.03; 5.0 × 10–51 ≤ p ≤ 0.018). The risk alleles of five SNPs, including rs8017172, rs17253792, rs945270, rs4144657, and rs1188184 also significantly increased the KTN1 mRNA expression in the putamen or SNc (0.021 ≤ p ≤ 0.046). The KTN1 mRNA was abundant in the putamen and/or SNc across five independent cohorts and differentially expressed in the SNc between PD and HC in one cohort (p = 0.047). Conclusion There was a consistent, significant, replicable, and robust positive relationship among the KTN1 variants, PD risk, KTN1 mRNA expression in putamen, and putamen volumes, and a modest relation between PD risk and KTN1 mRNA expression in SNc, suggesting that KTN1 may play a functional role in the development of PD.
Collapse
Affiliation(s)
- Qiao Mao
- Department of Psychosomatic Medicine, People's Hospital of Deyang, Deyang, China
| | - Xiaoping Wang
- Department of Neurology, Shanghai Tongren Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Bin Chen
- Department of Cardiovascular Medicine, Fujian Provincial Hospital, Fuzhou, China
| | - Longhua Fan
- Qingpu Branch, Department of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Shuhong Wang
- Department of Neurology, Shanghai Tongren Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Yong Zhang
- Tianjin Mental Health Center, Tianjin, China
| | - Xiandong Lin
- Laboratory of Radiation Oncology and Radiobiology, Fujian Provincial Cancer Hospital, Teaching Hospital of Fujian Medical University, Fuzhou, China
| | - Yuping Cao
- Department of Psychiatry, Second Xiangya Hospital, Central South University, Changsha, China
| | - Yun-Cheng Wu
- Department of Neurology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jiawu Ji
- Department of Psychiatry, Fuzhou Neuropsychiatric Hospital, Fujian Medical University, Fuzhou, China
| | - Jianying Xu
- Zhuhai Municipal Maternal and Children's Health Hospital, Zhuhai, China
| | - Jianming Zheng
- Huashan Hospital, Fudan University School of Medicine, Shanghai, China
| | - Huihao Zhang
- The First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | | | - Wenzhong Chen
- Department of Psychiatry, Shanghai Mental Health Center, Shanghai, China
| | - Wenhong Cheng
- Department of Psychiatry, Shanghai Mental Health Center, Shanghai, China
| | - Xingqun Luo
- Department of Clinical Medicine, College of Integrated Traditional Chinese and Western Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Kesheng Wang
- Department of Family and Community Health, School of Nursing, Health Sciences Center, West Virginia University, Morgantown, WV, United States
| | - Lingjun Zuo
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, United States
| | - Longli Kang
- Key Laboratory for Molecular Genetic Mechanisms and Intervention Research on High Altitude Diseases of Tibet Autonomous Region, Xizang Minzu University School of Medicine, Xiangyang, China
| | - Chiang-Shan R Li
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, United States
| | - Xingguang Luo
- Biological Psychiatry Research Center, Beijing Huilongguan Hospital, Beijing, China
| |
Collapse
|
27
|
Parkinson's disease: proteinopathy or lipidopathy? NPJ PARKINSONS DISEASE 2020; 6:3. [PMID: 31909184 PMCID: PMC6941970 DOI: 10.1038/s41531-019-0103-7] [Citation(s) in RCA: 142] [Impact Index Per Article: 28.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Accepted: 12/02/2019] [Indexed: 12/16/2022]
Abstract
Lipids play a more significant role in Parkinson’s disease and its related brain disorders than is currently recognized, supporting a “lipid cascade”. The 14 kDa protein α-synuclein (αS) is strongly associated with Parkinson’s disease (PD), dementia with Lewy bodies (DLB), other synucleinopathies such as multiple system atrophy, and even certain forms of Alzheimer’s disease. Rigorously deciphering the biochemistry of αS in native systems is the key to developing treatments. αS is highly expressed in the brain, the second most lipid-rich organ, and has been proposed to be a lipid-binding protein that physiologically interacts with phospholipids and fatty acids (FAs). αS-rich cytoplasmic inclusions called Lewy bodies and Lewy neurites are the hallmark lesions of synucleinopathies. Excess αS–membrane interactions may trigger proteinaceous αS aggregation by stimulating its primary nucleation. However, αS may also exert its toxicity prior to or independent of its self-aggregation, e.g., via excessive membrane interactions, which may be promoted by certain lipids and FAs. A complex αS-lipid landscape exists, which comprises both physiological and pathological states of αS. As novel insights about the composition of Lewy lesions occur, new lipid-related PD drug candidates emerge, and genome-wide association studies (GWAS) increasingly validate new hits in lipid-associated pathways, it seems timely to review our current knowledge of lipids in PD and consider the roles for these pathways in synucleinopathies.αS ↔ lipid interplay: aspects of cellular αS homeostasis (blue oval), aspects of lipid homeostasis (green oval), and overlapping aspects. Pathological states are labeled in red. Simplified schematic of both select αS and select lipid species. Several existing publications suggest αS effects on lipids and vice versa, as indicated by arrows. DG diglyceride, ER endoplasmic reticulum, FA fatty acid, LD, lipid droplet, TG triglyceride. ![]()
Collapse
|
28
|
Abstract
Noradrenergic system of brain supplies the neurotransmitter noradrenalin throughout the brain through widespread efferent projections and play pivotal role in cognitive activities and could be involve in motor and non-motor symptoms of Parkinson's disease (PD) pathology. Profound loss of noradrenergic pathways has been reported in both Parkinson's and Alzheimer's disease (AD) pathology however their employment in therapeutics is still scarce. Therefore the present review is providing the various aspects for involvement on noradrenergic pathways in PD and AD pathology as well as the imaging of locus coeruleus as indicative diagnostic marker for disease. The present review is describing about the role of tiny nucleus locus coeruleus located noradrenergic pathways in said pathologies and discussing the past research as well as lacunas in this regard.
Collapse
Affiliation(s)
- Sarika Singh
- Toxicology and Experimental Medicine Division, CDRI-CSIR, Lucknow, UP, India
| |
Collapse
|
29
|
Dixit A, Mehta R, Singh AK. Proteomics in Human Parkinson's Disease: Present Scenario and Future Directions. Cell Mol Neurobiol 2019; 39:901-915. [PMID: 31190159 PMCID: PMC11457823 DOI: 10.1007/s10571-019-00700-9] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Accepted: 06/04/2019] [Indexed: 12/26/2022]
Abstract
Parkinson's disease (PD) is an age-related, threatening neurodegenerative disorder with no reliable treatment till date. Identification of specific and reliable biomarker is a major challenge for disease diagnosis and designing effective therapeutic strategy against it. PD pathology at molecular level involves abnormal expression and function of several proteins, including alpha-synuclein. These proteins affect the normal functioning of neurons through various post-translational modifications and interaction with other cellular components. The role of protein anomalies during PD pathogenesis can be better understood by the application of proteomics approach. A number of proteomic studies conducted on brain tissue, blood, and cerebrospinal fluid of PD patients have identified a wide array of protein alterations underlying disease pathogenesis. However, these studies are limited by the types of brain regions or biofluids utilized in the research. For a complete understanding of PD mechanism and discovery of reliable protein biomarkers, it is essential to analyze the proteome of different PD-associated brain regions and easily accessible biofluids such as saliva and urine. The present review summarizes the major advances in the field of PD research in humans utilizing proteomic techniques. Moreover, potential samples for proteomic analysis and limitations associated with the analyses of different types of samples have also been discussed.
Collapse
Affiliation(s)
- Anubhuti Dixit
- Amity Institute of Neuropsychology and Neurosciences, Amity University, Sector-125, Noida, Uttar Pradesh, 201313, India.
| | - Rachna Mehta
- Amity Institute of Neuropsychology and Neurosciences, Amity University, Sector-125, Noida, Uttar Pradesh, 201313, India
| | - Abhishek Kumar Singh
- Amity Institute of Neuropsychology and Neurosciences, Amity University, Sector-125, Noida, Uttar Pradesh, 201313, India
| |
Collapse
|
30
|
Fanning S, Haque A, Imberdis T, Baru V, Barrasa MI, Nuber S, Termine D, Ramalingam N, Ho GPH, Noble T, Sandoe J, Lou Y, Landgraf D, Freyzon Y, Newby G, Soldner F, Terry-Kantor E, Kim TE, Hofbauer HF, Becuwe M, Jaenisch R, Pincus D, Clish CB, Walther TC, Farese RV, Srinivasan S, Welte MA, Kohlwein SD, Dettmer U, Lindquist S, Selkoe D. Lipidomic Analysis of α-Synuclein Neurotoxicity Identifies Stearoyl CoA Desaturase as a Target for Parkinson Treatment. Mol Cell 2019; 73:1001-1014.e8. [PMID: 30527540 PMCID: PMC6408259 DOI: 10.1016/j.molcel.2018.11.028] [Citation(s) in RCA: 183] [Impact Index Per Article: 30.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Revised: 09/05/2018] [Accepted: 11/19/2018] [Indexed: 01/08/2023]
Abstract
In Parkinson's disease (PD), α-synuclein (αS) pathologically impacts the brain, a highly lipid-rich organ. We investigated how alterations in αS or lipid/fatty acid homeostasis affect each other. Lipidomic profiling of human αS-expressing yeast revealed increases in oleic acid (OA, 18:1), diglycerides, and triglycerides. These findings were recapitulated in rodent and human neuronal models of αS dyshomeostasis (overexpression; patient-derived triplication or E46K mutation; E46K mice). Preventing lipid droplet formation or augmenting OA increased αS yeast toxicity; suppressing the OA-generating enzyme stearoyl-CoA-desaturase (SCD) was protective. Genetic or pharmacological SCD inhibition ameliorated toxicity in αS-overexpressing rat neurons. In a C. elegans model, SCD knockout prevented αS-induced dopaminergic degeneration. Conversely, we observed detrimental effects of OA on αS homeostasis: in human neural cells, excess OA caused αS inclusion formation, which was reversed by SCD inhibition. Thus, monounsaturated fatty acid metabolism is pivotal for αS-induced neurotoxicity, and inhibiting SCD represents a novel PD therapeutic approach.
Collapse
Affiliation(s)
- Saranna Fanning
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA; Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA
| | - Aftabul Haque
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA
| | - Thibaut Imberdis
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Valeriya Baru
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA
| | | | - Silke Nuber
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Daniel Termine
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA
| | - Nagendran Ramalingam
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Gary P H Ho
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Tallie Noble
- Mira Costa College, 1 Barnard Drive, Oceanside, CA 92056, USA
| | - Jackson Sandoe
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA
| | - Yali Lou
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA
| | - Dirk Landgraf
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA
| | - Yelena Freyzon
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA
| | - Gregory Newby
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA; Department of Biology, MIT, Cambridge, MA 02139, USA
| | - Frank Soldner
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA
| | - Elizabeth Terry-Kantor
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Tae-Eun Kim
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Harald F Hofbauer
- Institute of Molecular Biosciences, BioTechMed-Graz, University of Graz, 8010 Graz, Austria
| | - Michel Becuwe
- Department of Genetics and Complex Diseases, Harvard T.H. Chan School of Public Health, 655 Huntington Avenue, Boston, MA 02115, USA
| | - Rudolf Jaenisch
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA; Department of Biology, MIT, Cambridge, MA 02139, USA
| | - David Pincus
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA
| | - Clary B Clish
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Tobias C Walther
- Department of Genetics and Complex Diseases, Harvard T.H. Chan School of Public Health, 655 Huntington Avenue, Boston, MA 02115, USA; Department of Cell Biology, Harvard Medical School, 240 Longwood Avenue, Boston, MA 02115, USA; Department of Genetics, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA; HHMI, Department of Genetics and Complex Diseases, Harvard T.H. Chan School of Public Health, 655 Huntington Avenue, Boston, MA 02115, USA
| | - Robert V Farese
- Department of Genetics and Complex Diseases, Harvard T.H. Chan School of Public Health, 655 Huntington Avenue, Boston, MA 02115, USA; Department of Cell Biology, Harvard Medical School, 240 Longwood Avenue, Boston, MA 02115, USA; Department of Genetics, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - Supriya Srinivasan
- Department of Chemical Physiology and The Dorris Neuroscience Center, 1 Barnard Drive, Oceanside, CA 92056, USA; The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Michael A Welte
- Department of Biology, University of Rochester, Rochester, NY 14627, USA
| | - Sepp D Kohlwein
- Institute of Molecular Biosciences, BioTechMed-Graz, University of Graz, 8010 Graz, Austria
| | - Ulf Dettmer
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA.
| | - Susan Lindquist
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA; Department of Biology, MIT, Cambridge, MA 02139, USA; HHMI, Department of Biology, MIT, Cambridge, MA 02139, USA
| | - Dennis Selkoe
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
31
|
Cardoso AL, Fernandes A, Aguilar-Pimentel JA, de Angelis MH, Guedes JR, Brito MA, Ortolano S, Pani G, Athanasopoulou S, Gonos ES, Schosserer M, Grillari J, Peterson P, Tuna BG, Dogan S, Meyer A, van Os R, Trendelenburg AU. Towards frailty biomarkers: Candidates from genes and pathways regulated in aging and age-related diseases. Ageing Res Rev 2018; 47:214-277. [PMID: 30071357 DOI: 10.1016/j.arr.2018.07.004] [Citation(s) in RCA: 315] [Impact Index Per Article: 45.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Revised: 07/08/2018] [Accepted: 07/10/2018] [Indexed: 12/12/2022]
Abstract
OBJECTIVE Use of the frailty index to measure an accumulation of deficits has been proven a valuable method for identifying elderly people at risk for increased vulnerability, disease, injury, and mortality. However, complementary molecular frailty biomarkers or ideally biomarker panels have not yet been identified. We conducted a systematic search to identify biomarker candidates for a frailty biomarker panel. METHODS Gene expression databases were searched (http://genomics.senescence.info/genes including GenAge, AnAge, LongevityMap, CellAge, DrugAge, Digital Aging Atlas) to identify genes regulated in aging, longevity, and age-related diseases with a focus on secreted factors or molecules detectable in body fluids as potential frailty biomarkers. Factors broadly expressed, related to several "hallmark of aging" pathways as well as used or predicted as biomarkers in other disease settings, particularly age-related pathologies, were identified. This set of biomarkers was further expanded according to the expertise and experience of the authors. In the next step, biomarkers were assigned to six "hallmark of aging" pathways, namely (1) inflammation, (2) mitochondria and apoptosis, (3) calcium homeostasis, (4) fibrosis, (5) NMJ (neuromuscular junction) and neurons, (6) cytoskeleton and hormones, or (7) other principles and an extensive literature search was performed for each candidate to explore their potential and priority as frailty biomarkers. RESULTS A total of 44 markers were evaluated in the seven categories listed above, and 19 were awarded a high priority score, 22 identified as medium priority and three were low priority. In each category high and medium priority markers were identified. CONCLUSION Biomarker panels for frailty would be of high value and better than single markers. Based on our search we would propose a core panel of frailty biomarkers consisting of (1) CXCL10 (C-X-C motif chemokine ligand 10), IL-6 (interleukin 6), CX3CL1 (C-X3-C motif chemokine ligand 1), (2) GDF15 (growth differentiation factor 15), FNDC5 (fibronectin type III domain containing 5), vimentin (VIM), (3) regucalcin (RGN/SMP30), calreticulin, (4) PLAU (plasminogen activator, urokinase), AGT (angiotensinogen), (5) BDNF (brain derived neurotrophic factor), progranulin (PGRN), (6) α-klotho (KL), FGF23 (fibroblast growth factor 23), FGF21, leptin (LEP), (7) miRNA (micro Ribonucleic acid) panel (to be further defined), AHCY (adenosylhomocysteinase) and KRT18 (keratin 18). An expanded panel would also include (1) pentraxin (PTX3), sVCAM/ICAM (soluble vascular cell adhesion molecule 1/Intercellular adhesion molecule 1), defensin α, (2) APP (amyloid beta precursor protein), LDH (lactate dehydrogenase), (3) S100B (S100 calcium binding protein B), (4) TGFβ (transforming growth factor beta), PAI-1 (plasminogen activator inhibitor 1), TGM2 (transglutaminase 2), (5) sRAGE (soluble receptor for advanced glycosylation end products), HMGB1 (high mobility group box 1), C3/C1Q (complement factor 3/1Q), ST2 (Interleukin 1 receptor like 1), agrin (AGRN), (6) IGF-1 (insulin-like growth factor 1), resistin (RETN), adiponectin (ADIPOQ), ghrelin (GHRL), growth hormone (GH), (7) microparticle panel (to be further defined), GpnmB (glycoprotein nonmetastatic melanoma protein B) and lactoferrin (LTF). We believe that these predicted panels need to be experimentally explored in animal models and frail cohorts in order to ascertain their diagnostic, prognostic and therapeutic potential.
Collapse
|
32
|
Seipin deficiency in mice causes loss of dopaminergic neurons via aggregation and phosphorylation of α-synuclein and neuroinflammation. Cell Death Dis 2018; 9:440. [PMID: 29670081 PMCID: PMC5906676 DOI: 10.1038/s41419-018-0471-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Revised: 02/12/2018] [Accepted: 03/09/2018] [Indexed: 12/16/2022]
Abstract
Seipin gene is originally found in type 2 congenital generalized lipodystrophy (CGL2) to involve lipid droplet formation. Recently, decrease of seipin expression is reported in substantia nigra of Parkinson’s disease patients. Dopaminergic neurons in substantia nigra pars compacta expressed the seipin protein. The objective of this study is to investigate influence of the seipin deficiency on dopaminergic neurons and motor behaviors. Neuronal seipin knockout (seipin-nKO) mice (3–12 months of age) displayed an age-related deficit in motor coordination. The number of dopaminergic neurons in seipin-nKO mice was age dependently reduced with increase in cleaved caspase-3. The levels of αSyn oligomers and oligomer phosphorylation (S129), but not αSyn monomers, were elevated in dopaminergic neurons and substantia nigra of seipin-nKO mice. The PPARγ expression in seipin-nKO mice was reduced. In seipin-nKO mice, the phosphorylation of GSK3β was increased at Tyr216 and was reduced at Ser9, which was corrected by the PPARγ agonist rosiglitazone. The increased IL-6 level in seipin-nKO mice was sensitive to rosiglitazone and GSK3β inhibitor AR-A014418. The enhanced phosphorylation of αSyn was prevented by rosiglitazone and AR-A014418, while the increase in αSyn oligomers was corrected only by rosiglitazone. The treatment of seipin-nKO mice with rosiglitazone and AR-A014418 rescued the death of dopaminergic neurons, which was accompanied by the improvement of motor coordination. Therefore, the results indicate that seipin deficiency causes an age-related loss of dopaminergic neurons and impairment of motor coordination through reducing PPARγ to enhance aggregation and phosphorylation of αSyn and neuroinflammation.
Collapse
|
33
|
Weinshenker D. Long Road to Ruin: Noradrenergic Dysfunction in Neurodegenerative Disease. Trends Neurosci 2018; 41:211-223. [PMID: 29475564 PMCID: PMC5878728 DOI: 10.1016/j.tins.2018.01.010] [Citation(s) in RCA: 200] [Impact Index Per Article: 28.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Revised: 01/25/2018] [Accepted: 01/29/2018] [Indexed: 01/09/2023]
Abstract
It has been known for decades that degeneration of the locus coeruleus (LC), the major noradrenergic nucleus in the brain, occurs in both Alzheimer's disease (AD) and Parkinson's disease (PD), but it was given scant attention. It is now recognized that hyperphosphorylated tau in the LC is the first detectable AD-like neuropathology in the human brain, α-synuclein inclusions in the LC represent an early step in PD, and experimental LC lesions exacerbate neuropathology and cognitive/behavioral deficits in animal models. The purpose of this review is to consider the causes and consequences of LC pathology, dysfunction, and degeneration, as well as their implications for early detection and treatment.
Collapse
Affiliation(s)
- David Weinshenker
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, USA.
| |
Collapse
|
34
|
Post MR, Lieberman OJ, Mosharov EV. Can Interactions Between α-Synuclein, Dopamine and Calcium Explain Selective Neurodegeneration in Parkinson's Disease? Front Neurosci 2018; 12:161. [PMID: 29593491 PMCID: PMC5861202 DOI: 10.3389/fnins.2018.00161] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2017] [Accepted: 02/27/2018] [Indexed: 12/11/2022] Open
Abstract
Several lines of evidence place alpha-synuclein (aSyn) at the center of Parkinson's disease (PD) etiology, but it is still unclear why overexpression or mutated forms of this protein affect some neuronal populations more than others. Susceptible neuronal populations in PD, dopaminergic neurons of the substantia nigra pars compacta (SNpc) and the locus coeruleus (LC), are distinguished by relatively high cytoplasmic concentrations of dopamine and calcium ions. Here we review the evidence for the multi-hit hypothesis of neurodegeneration, including recent papers that demonstrate synergistic interactions between aSyn, calcium ions and dopamine that may lead to imbalanced protein turnover and selective susceptibility of these neurons. We conclude that decreasing the levels of any one of these toxicity mediators can be beneficial for the survival of SNpc and LC neurons, providing multiple opportunities for targeted drug interventions aimed at modifying the course of PD.
Collapse
Affiliation(s)
- Michael R Post
- Departments of Psychiatry and Neurology, New York State Psychiatric Institute, Columbia University Medical Center, New York, NY, United States
| | - Ori J Lieberman
- Departments of Psychiatry and Neurology, New York State Psychiatric Institute, Columbia University Medical Center, New York, NY, United States
| | - Eugene V Mosharov
- Departments of Psychiatry and Neurology, New York State Psychiatric Institute, Columbia University Medical Center, New York, NY, United States
| |
Collapse
|
35
|
|
36
|
Scifo E, Calza G, Fuhrmann M, Soliymani R, Baumann M, Lalowski M. Recent advances in applying mass spectrometry and systems biology to determine brain dynamics. Expert Rev Proteomics 2017; 14:545-559. [PMID: 28539064 DOI: 10.1080/14789450.2017.1335200] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
INTRODUCTION Neurological disorders encompass various pathologies which disrupt normal brain physiology and function. Poor understanding of their underlying molecular mechanisms and their societal burden argues for the necessity of novel prevention strategies, early diagnostic techniques and alternative treatment options to reduce the scale of their expected increase. Areas covered: This review scrutinizes mass spectrometry based approaches used to investigate brain dynamics in various conditions, including neurodegenerative and neuropsychiatric disorders. Different proteomics workflows for isolation/enrichment of specific cell populations or brain regions, sample processing; mass spectrometry technologies, for differential proteome quantitation, analysis of post-translational modifications and imaging approaches in the brain are critically deliberated. Future directions, including analysis of cellular sub-compartments, targeted MS platforms (selected/parallel reaction monitoring) and use of mass cytometry are also discussed. Expert commentary: Here, we summarize and evaluate current mass spectrometry based approaches for determining brain dynamics in health and diseases states, with a focus on neurological disorders. Furthermore, we provide insight on current trends and new MS technologies with potential to improve this analysis.
Collapse
Affiliation(s)
- Enzo Scifo
- a Department of Psychiatry, and of Pharmacology and Toxicology , University of Toronto, Campbell Family Mental Health Research Institute of CAMH , Toronto , Canada
| | - Giulio Calza
- b Medicum, Meilahti Clinical Proteomics Core Facility, Biochemistry/Developmental Biology, Faculty of Medicine , FI-00014 University of Helsinki , Helsinki , Finland
| | - Martin Fuhrmann
- c Neuroimmunology and Imaging Group , German Center for Neurodegenerative Diseases (DZNE) , Bonn , Germany
| | - Rabah Soliymani
- b Medicum, Meilahti Clinical Proteomics Core Facility, Biochemistry/Developmental Biology, Faculty of Medicine , FI-00014 University of Helsinki , Helsinki , Finland
| | - Marc Baumann
- b Medicum, Meilahti Clinical Proteomics Core Facility, Biochemistry/Developmental Biology, Faculty of Medicine , FI-00014 University of Helsinki , Helsinki , Finland
| | - Maciej Lalowski
- b Medicum, Meilahti Clinical Proteomics Core Facility, Biochemistry/Developmental Biology, Faculty of Medicine , FI-00014 University of Helsinki , Helsinki , Finland
| |
Collapse
|
37
|
Xu B, Jia T, Macare C, Banaschewski T, Bokde ALW, Bromberg U, Büchel C, Cattrell A, Conrod PJ, Flor H, Frouin V, Gallinat J, Garavan H, Gowland P, Heinz A, Ittermann B, Martinot JL, Paillère Martinot ML, Nees F, Orfanos DP, Paus T, Poustka L, Smolka MN, Walter H, Whelan R, Schumann G, Desrivières S. Impact of a Common Genetic Variation Associated With Putamen Volume on Neural Mechanisms of Attention-Deficit/Hyperactivity Disorder. J Am Acad Child Adolesc Psychiatry 2017; 56:436-444.e4. [PMID: 28433093 DOI: 10.1016/j.jaac.2017.02.009] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Revised: 02/07/2017] [Accepted: 03/01/2017] [Indexed: 11/27/2022]
Abstract
OBJECTIVE In a recent genomewide association study of subcortical brain volumes, a common genetic variation at rs945270 was identified as having the strongest effect on putamen volume, a brain measurement linked to familial risk for attention-deficit/hyperactivity disorder (ADHD). To determine whether rs945270 might be a genetic determinant of ADHD, its effects on ADHD-related symptoms and neural mechanisms of ADHD, such as response inhibition and reward sensitivity, were explored. METHOD A large population sample of 1,834 14-year-old adolescents was used to test the effects of rs945270 on ADHD symptoms assessed through the Strengths and Difficulties Questionnaire and region-of-interest analyses of putamen activation by functional magnetic resonance imaging using the stop signal and monetary incentive delay tasks, assessing response inhibition and reward sensitivity, respectively. RESULTS There was a significant link between rs945270 and ADHD symptom scores, with the C allele associated with lower symptom scores, most notably hyperactivity. In addition, there were sex-specific effects of this variant on the brain. In boys, the C allele was associated with lower putamen activity during successful response inhibition, a brain response that was not associated with ADHD symptoms. In girls, putamen activation during reward anticipation increased with the number of C alleles, most significantly in the right putamen. Remarkably, right putamen activation during reward anticipation tended to negatively correlate with ADHD symptoms. CONCLUSION These results indicate that rs945270 might contribute to the genetic risk of ADHD partly through its effects on hyperactivity and reward processing in girls.
Collapse
Affiliation(s)
- Bing Xu
- Institute of Psychiatry, Psychology and Neuroscience, King's College London, UK
| | - Tianye Jia
- Institute of Psychiatry, Psychology and Neuroscience, King's College London, UK
| | - Christine Macare
- Institute of Psychiatry, Psychology and Neuroscience, King's College London, UK
| | - Tobias Banaschewski
- Clinical Faculty Mannheim, Central Institute of Mental Health, University of Heidelberg, Germany
| | - Arun L W Bokde
- Discipline of Psychiatry, School of Medicine and the Trinity College Institute of Neurosciences, Trinity College Dublin, Ireland
| | - Uli Bromberg
- University Medical Centre Hamburg-Eppendorf, Germany
| | | | - Anna Cattrell
- Institute of Psychiatry, Psychology and Neuroscience, King's College London, UK
| | - Patricia J Conrod
- Université de Montreal, Centre Hospitalier Universitaire Sainte-Justine, Montreal, Canada
| | - Herta Flor
- Central Institute of Mental Health, Medical Faculty Mannheim, Germany
| | - Vincent Frouin
- Neurospin, Commissariat à l'Energie Atomique, CEA-Saclay Center, Paris, France
| | - Jürgen Gallinat
- University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | | | - Penny Gowland
- Sir Peter Mansfield Imaging Centre School of Physics and Astronomy, University of Nottingham, UK
| | - Andreas Heinz
- Campus Charité Mitte, Charité, Universitätsmedizin Berlin, Germany
| | - Bernd Ittermann
- Physikalisch-Technische Bundesanstalt, Braunschweig and Berlin, Germany
| | - Jean-Luc Martinot
- Institut National de la Santé et de la Recherche Médicale, INSERM Unit 1000 Neuroimaging and Psychiatry, University Paris Sud, University Paris Descartes, Sorbonne Paris Cité and Maison de Solenn, Paris
| | - Marie-Laure Paillère Martinot
- Institut National de la Santé et de la Recherche Médicale, INSERM Unit 1000 Neuroimaging and Psychiatry, University Paris Sud, University Paris Descartes, Sorbonne Paris Cité and Maison de Solenn, Paris; Maison de Solenn, Cochin Hospital, Paris
| | - Frauke Nees
- Central Institute of Mental Health, Medical Faculty Mannheim, Germany
| | | | - Tomáš Paus
- Rotman Research Institute, Baycrest and the University of Toronto, Canada
| | - Luise Poustka
- Clinical Faculty Mannheim, Central Institute of Mental Health, University of Heidelberg, Germany
| | | | - Henrik Walter
- Campus Charité Mitte, Charité, Universitätsmedizin Berlin, Germany
| | | | - Gunter Schumann
- Institute of Psychiatry, Psychology and Neuroscience, King's College London, UK
| | - Sylvane Desrivières
- Institute of Psychiatry, Psychology and Neuroscience, King's College London, UK.
| | | |
Collapse
|
38
|
Halbgebauer S, Öckl P, Wirth K, Steinacker P, Otto M. Protein biomarkers in Parkinson's disease: Focus on cerebrospinal fluid markers and synaptic proteins. Mov Disord 2016; 31:848-60. [PMID: 27134134 DOI: 10.1002/mds.26635] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Revised: 03/06/2016] [Accepted: 03/09/2016] [Indexed: 01/06/2023] Open
Abstract
Despite extensive research, to date, no validated biomarkers for PD have been found. This review seeks to summarize studies approaching the detection of biomarker candidates for PD and introduce promising ones in more detail, with special attention to synaptic proteins. To this end, we performed a PubMed search and included studies using proteomic tools (2-dimensional difference in gel electrophoresis and/or mass spectrometry) for the comparison of samples from PD and control patients. We found 27 studies reporting more than 500 differentially expressed proteins in which a total of 28 were detected in 2 and 17 in 3 or more independent studies, including posttranslationally modified proteins. In addition, of these 500 proteins, 25 were found to be brain specific, and 14 were enriched in synapses. Special attention was given to the applicability of the biomarker regarding sampling procedures, that is, using CSF/serum material for diagnosis. Furthermore, presynaptic proteins involved in vesicle membrane fusion seem to be interesting candidates for future analyses. Nonetheless, even though such promising biomarker candidates for PD exist, validation of these biomarkers in large-scale clinical studies is necessary to evaluate the diagnostic potential. © 2016 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
| | - Patrick Öckl
- Department of Neurology, University of Ulm, Ulm, Germany
| | | | | | - Markus Otto
- Department of Neurology, University of Ulm, Ulm, Germany
| |
Collapse
|
39
|
Ruiz-Riquelme A, Sánchez-Iglesias S, Rábano A, Guillén-Navarro E, Domingo-Jiménez R, Ramos A, Rosa I, Senra A, Nilsson P, García Á, Araújo-Vilar D, Requena JR. Larger aggregates of mutant seipin in Celia's Encephalopathy, a new protein misfolding neurodegenerative disease. Neurobiol Dis 2015; 83:44-53. [PMID: 26282322 DOI: 10.1016/j.nbd.2015.08.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2015] [Revised: 07/18/2015] [Accepted: 08/12/2015] [Indexed: 11/19/2022] Open
Abstract
Celia's Encephalopathy (MIM #615924) is a recently discovered fatal neurodegenerative syndrome associated with a new BSCL2 mutation (c.985C>T) that results in an aberrant isoform of seipin (Celia seipin). This mutation is lethal in both homozygosity and compounded heterozygosity with a lipodystrophic BSCL2 mutation, resulting in a progressive encephalopathy with fatal outcomes at ages 6-8. Strikingly, heterozygous carriers are asymptomatic, conflicting with the gain of toxic function attributed to this mutation. Here we report new key insights about the molecular pathogenic mechanism of this new syndrome. Intranuclear inclusions containing mutant seipin were found in brain tissue from a homozygous patient suggesting a pathogenic mechanism similar to other neurodegenerative diseases featuring brain accumulation of aggregated, misfolded proteins. Sucrose gradient distribution showed that mutant seipin forms much larger aggregates as compared with wild type (wt) seipin, indicating an impaired oligomerization. On the other hand, the interaction between wt and Celia seipin confirmed by coimmunoprecipitation (CoIP) assays, together with the identification of mixed oligomers in sucrose gradient fractionation experiments can explain the lack of symptoms in heterozygous carriers. We propose that the increased aggregation and subsequent impaired oligomerization of Celia seipin leads to cell death. In heterozygous carriers, wt seipin might prevent the damage caused by mutant seipin through its sequestration into harmless mixed oligomers.
Collapse
Affiliation(s)
- Alejandro Ruiz-Riquelme
- CIMUS Biomedical Research Institute, University of Santiago de Compostela-IDIS, 15782 Santiago de Compostela, Spain
| | - Sofía Sánchez-Iglesias
- CIMUS Biomedical Research Institute, University of Santiago de Compostela-IDIS, 15782 Santiago de Compostela, Spain
| | - Alberto Rábano
- Neuropathology Department and Tissue Bank, Fundación CIEN, 28031 Madrid, Spain
| | - Encarna Guillén-Navarro
- Section of Medical Genetics Dysmorphology, Division of Pediatrics, Hospital Clínico Universitario Virgen de la Arrixaca, IMIB-Arrixaca, 30120 Murcia, Spain; UCAM-Catholic University of Murcia, CIBERER-ISCIII, Madrid, Spain
| | - Rosario Domingo-Jiménez
- Section of Neuropediatrics, Division of Pediatrics, Hospital Clínico Universitario Virgen de la Arrixaca, IMIB-Arrixaca, 30120 Murcia, Spain; CIBERER-ISCIII, Madrid, Spain
| | - Adriana Ramos
- CIMUS Biomedical Research Institute, University of Santiago de Compostela-IDIS, 15782 Santiago de Compostela, Spain
| | - Isaac Rosa
- CIMUS Biomedical Research Institute, University of Santiago de Compostela-IDIS, 15782 Santiago de Compostela, Spain; Department of Pharmacology, University of Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Ana Senra
- CIMUS Biomedical Research Institute, University of Santiago de Compostela-IDIS, 15782 Santiago de Compostela, Spain
| | - Peter Nilsson
- Affinity Proteomics, SciLifeLab, School of Biotechnology, KTH - Royal Institute of Technology, SE 171-21 Stockholm, Sweden
| | - Ángel García
- CIMUS Biomedical Research Institute, University of Santiago de Compostela-IDIS, 15782 Santiago de Compostela, Spain; Department of Pharmacology, University of Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - David Araújo-Vilar
- CIMUS Biomedical Research Institute, University of Santiago de Compostela-IDIS, 15782 Santiago de Compostela, Spain; Department of Medicine, University of Santiago de Compostela, 15782 Santiago de Compostela, Spain.
| | - Jesús R Requena
- CIMUS Biomedical Research Institute, University of Santiago de Compostela-IDIS, 15782 Santiago de Compostela, Spain; Department of Medicine, University of Santiago de Compostela, 15782 Santiago de Compostela, Spain.
| |
Collapse
|
40
|
Lithgow BJ, Shoushtarian M. Parkinson's disease: disturbed vestibular function and levodopa. J Neurol Sci 2015; 353:49-58. [PMID: 25899315 DOI: 10.1016/j.jns.2015.03.050] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2014] [Revised: 03/19/2015] [Accepted: 03/31/2015] [Indexed: 11/15/2022]
Abstract
Evidence indicates Levodopa effects central postural control. As electrophysiological postural control biomarkers, sensory oto-acoustic features were extracted from Electrovestibulography (EVestG) data to identify 20 healthy age and gender matched individuals as Controls from 20 PD subjects before (PDlowmed) and 18 after (PDmed) morning doses of Levodopa. EVestG data was collected using a single tilt stimulus applied in the pitch plane. The extracted features were based on the measured firing pattern, interval histogram and the shape of the average field potential response. An unbiased cross validated classification accuracy of 88%, 88% and 79% was achieved using combinations of 2 features for separating PDlowmed from control, control from PD (combined PDlowmed and PDmed), and PDlowmed from PDmed groups respectively. One feature showed significant correlations (p<0.05) with the Modified Hoehn and Yahr PD staging scale. The results indicate disturbed vestibular function is observed in both the PDmed and PDlowmed conditions, and these are separable. The implication is that Levodopa may also affect peripheral as well as central postural control.
Collapse
Affiliation(s)
- Brian J Lithgow
- Monash Alfred Psychiatry Research Centre, Monash University Central Clinical School and the Alfred Hospital, 4th Floor, 607St Kilda Rd, Melbourne, Victoria, Australia 3004; Diagnostic and Neurosignal Processing Research Laboratory, Monash University, Wellington Rd, Clayton, Victoria, Australia 3180; Diagnostic and Neurosignal Processing Research Laboratory, University of Manitoba, Riverview Health Centre, 1 Morley St, Winnipeg, MB, Canada R3L 2P4.
| | - Mehrnaz Shoushtarian
- Diagnostic and Neurosignal Processing Research Laboratory, Monash University, Wellington Rd, Clayton, Victoria, Australia 3180.
| |
Collapse
|
41
|
Fernández-Irigoyen J, Zelaya MV, Perez-Valderrama E, Santamaría E. New insights into the human brain proteome: Protein expression profiling of deep brain stimulation target areas. J Proteomics 2015; 127:395-405. [PMID: 25845585 DOI: 10.1016/j.jprot.2015.03.032] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2014] [Revised: 02/09/2015] [Accepted: 03/19/2015] [Indexed: 12/30/2022]
Abstract
UNLABELLED Deep brain stimulation (DBS) is a neurosurgical procedure that provides therapeutic benefits for movement and affective disorders. The nucleus basalis of Meynert (NBM) and substantia nigra (SN) are considered target areas to apply DBS. Even though the degeneration of NBM and SN underlies the cognitive decline observed in neurological diseases, the protein knowledge derived from both areas is scarce. We have characterized the proteome present in both subcortical brain areas using the Triple TOF 5600 mass spectrometer, identifying 2775 and 3469 proteoforms in NBM and SN respectively. Data mining of MS-generated proteomic data have revealed that: i) 675 proteins tend to localize to synaptic ending, ii) 61% of the global dataset is also present in human CSF and/or plasma, and iii) 894 proteins have not been previously identified in healthy brain by MS. The correlation of NBM and SN proteomic expression profiles with human brain transcriptome data available at Allen Brain Atlas has revealed protein evidence for 250 genes considered with brain-wide expression and 112 genes with region-specific expression in human brain. In addition, protein datasets have been classified according to their chromosomal origin, increasing the current proteome coverage in healthy human brain. BIOLOGICAL SIGNIFICANCE The nucleus basalis of Meynert and substantia nigra are brain areas of clinical interest to apply the deep brain stimulation (DBS) technology in neurosurgery. Our proteomic characterization has revealed 675 proteins involved in the regulation of synaptic transmission, electrical machinery, and neurotransmitter release in both DBS target areas. Moreover, 2599 identified proteins present capacity to be secreted to the CSF and plasma. Our data contribute to a further step towards the characterization of the anatomical atlas of the human brain proteome, detecting 652 proteins that are common between different basal ganglia structures. This article is part of a Special Issue entitled: HUPO 2014.
Collapse
Affiliation(s)
- Joaquín Fernández-Irigoyen
- ProteoRed-ISCIII, Proteomics Unit, Clinical Neuroproteomics Group, Navarrabiomed, Fundación Miguel Servet, Instituto de Investigación Sanitaria de Navarra (IDISNA), 31008 Pamplona, Spain
| | - María Victoria Zelaya
- ProteoRed-ISCIII, Proteomics Unit, Clinical Neuroproteomics Group, Navarrabiomed, Fundación Miguel Servet, Instituto de Investigación Sanitaria de Navarra (IDISNA), 31008 Pamplona, Spain; Neurological Tissue Bank, Navarrabiomed, Fundación Miguel Servet, 31008 Pamplona, Spain
| | - Estela Perez-Valderrama
- ProteoRed-ISCIII, Proteomics Unit, Clinical Neuroproteomics Group, Navarrabiomed, Fundación Miguel Servet, Instituto de Investigación Sanitaria de Navarra (IDISNA), 31008 Pamplona, Spain
| | - Enrique Santamaría
- ProteoRed-ISCIII, Proteomics Unit, Clinical Neuroproteomics Group, Navarrabiomed, Fundación Miguel Servet, Instituto de Investigación Sanitaria de Navarra (IDISNA), 31008 Pamplona, Spain.
| |
Collapse
|
42
|
Fernandez-Irigoyen J, Labarga A, Zabaleta A, de Morentin XM, Perez-Valderrama E, Zelaya MV, Santamaria E. Toward defining the anatomo-proteomic puzzle of the human brain: An integrative analysis. Proteomics Clin Appl 2015; 9:796-807. [PMID: 25418211 DOI: 10.1002/prca.201400127] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2014] [Revised: 10/17/2014] [Accepted: 11/18/2014] [Indexed: 01/18/2023]
Abstract
The human brain is exceedingly complex, constituted by billions of neurons and trillions of synaptic connections that, in turn, define ∼900 neuroanatomical subdivisions in the adult brain (Hawrylycz et al. An anatomically comprehensive atlas of the human brain transcriptome. Nature 2012, 489, 391-399). The human brain transcriptome has revealed specific regional transcriptional signatures that are regulated in a spatiotemporal manner, increasing the complexity of the structural and molecular organization of this organ (Kang et al. Spatio-temporal transcriptome of the human brain. Nature 2011, 478, 483-489). During the last decade, neuroproteomics has emerged as a powerful approach to profile neural proteomes using shotgun-based MS, providing complementary information about protein content and function at a global level. Here, we revise recent proteome profiling studies performed in human brain, with special emphasis on proteome mapping of anatomical macrostructures, specific subcellular compartments, and cerebrospinal fluid. Moreover, we have performed an integrative functional analysis of the protein compilation derived from these large-scale human brain proteomic studies in order to obtain a comprehensive view of human brain biology. Finally, we also discuss the potential contribution of our meta-analysis to the Chromosome-centric Human Proteome Project initiative.
Collapse
Affiliation(s)
- Joaquín Fernandez-Irigoyen
- Clinical Neuroproteomics Group, Proteomics Unit, Proteored-ISCIII, Navarrabiomed, Fundación Miguel Servet, Pamplona, Spain
| | - Alberto Labarga
- Bioinformatics Unit, Navarrabiomed, Fundación Miguel Servet, Pamplona, Spain
| | - Aintzane Zabaleta
- Biofunctional Nanomaterials Laboratory, CIC Biomagune, San Sebastian, Spain
| | - Xabier Martínez de Morentin
- Clinical Neuroproteomics Group, Proteomics Unit, Proteored-ISCIII, Navarrabiomed, Fundación Miguel Servet, Pamplona, Spain
| | - Estela Perez-Valderrama
- Clinical Neuroproteomics Group, Proteomics Unit, Proteored-ISCIII, Navarrabiomed, Fundación Miguel Servet, Pamplona, Spain
| | | | - Enrique Santamaria
- Clinical Neuroproteomics Group, Proteomics Unit, Proteored-ISCIII, Navarrabiomed, Fundación Miguel Servet, Pamplona, Spain
| |
Collapse
|
43
|
Licker V, Burkhard PR. Proteomics as a new paradigm to tackle Parkinson’s disease research challenges. TRANSLATIONAL PROTEOMICS 2014. [DOI: 10.1016/j.trprot.2014.08.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
|
44
|
Gupta MK, Jayaram S, Madugundu AK, Chavan S, Advani J, Pandey A, Thongboonkerd V, Sirdeshmukh R. Chromosome-centric Human Proteome Project: Deciphering Proteins Associated with Glioma and Neurodegenerative Disorders on Chromosome 12. J Proteome Res 2014; 13:3178-90. [PMID: 24804578 DOI: 10.1021/pr500023p] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Affiliation(s)
- Manoj Kumar Gupta
- Institute
of Bioinformatics, International
Tech Park, Bangalore 560066, India
- Manipal University, Madhav Nagar, Manipal 576104, India
| | - Savita Jayaram
- Institute
of Bioinformatics, International
Tech Park, Bangalore 560066, India
- Manipal University, Madhav Nagar, Manipal 576104, India
| | - Anil K. Madugundu
- Institute
of Bioinformatics, International
Tech Park, Bangalore 560066, India
| | - Sandip Chavan
- Institute
of Bioinformatics, International
Tech Park, Bangalore 560066, India
- Manipal University, Madhav Nagar, Manipal 576104, India
| | - Jayshree Advani
- Institute
of Bioinformatics, International
Tech Park, Bangalore 560066, India
| | - Akhilesh Pandey
- Institute
of Bioinformatics, International
Tech Park, Bangalore 560066, India
- McKusick-Nathans
Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, 21205 United States
| | | | - Ravi Sirdeshmukh
- Institute
of Bioinformatics, International
Tech Park, Bangalore 560066, India
- Mazumdar
Shaw Centre for Translational Research, Narayana Health, Bangalore 560099, India
| |
Collapse
|
45
|
Regucalcin as a potential biomarker for metabolic and neuronal diseases. Mol Cell Biochem 2014; 391:157-66. [DOI: 10.1007/s11010-014-1998-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2013] [Accepted: 02/21/2014] [Indexed: 01/13/2023]
|
46
|
Licker V, Turck N, Kövari E, Burkhardt K, Côte M, Surini-Demiri M, Lobrinus JA, Sanchez JC, Burkhard PR. Proteomic analysis of human substantia nigra identifies novel candidates involved in Parkinson's disease pathogenesis. Proteomics 2014; 14:784-94. [DOI: 10.1002/pmic.201300342] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2013] [Revised: 12/13/2013] [Accepted: 12/15/2013] [Indexed: 11/09/2022]
Affiliation(s)
- Virginie Licker
- Neuroproteomics Group; University Medical Center; Faculty of Medicine; Geneva University; Geneva Switzerland
- Translational Biomarker Group; University Medical Center; Faculty of Medicine; Geneva University; Geneva Switzerland
| | - Natacha Turck
- Translational Biomarker Group; University Medical Center; Faculty of Medicine; Geneva University; Geneva Switzerland
| | - Enikö Kövari
- Department of Psychiatry; Geneva University Hospitals; Geneva Switzerland
| | - Karim Burkhardt
- Department of Pathology; Geneva University Hospitals; Geneva Switzerland
| | - Mélanie Côte
- Neuroproteomics Group; University Medical Center; Faculty of Medicine; Geneva University; Geneva Switzerland
- Translational Biomarker Group; University Medical Center; Faculty of Medicine; Geneva University; Geneva Switzerland
| | | | | | - Jean-Charles Sanchez
- Translational Biomarker Group; University Medical Center; Faculty of Medicine; Geneva University; Geneva Switzerland
| | - Pierre R. Burkhard
- Neuroproteomics Group; University Medical Center; Faculty of Medicine; Geneva University; Geneva Switzerland
- Translational Biomarker Group; University Medical Center; Faculty of Medicine; Geneva University; Geneva Switzerland
- Department of Neurology; Geneva University Hospitals; Geneva Switzerland
| |
Collapse
|
47
|
Marques R, Maia CJ, Vaz C, Correia S, Socorro S. The diverse roles of calcium-binding protein regucalcin in cell biology: from tissue expression and signalling to disease. Cell Mol Life Sci 2014; 71:93-111. [PMID: 23519827 PMCID: PMC11113322 DOI: 10.1007/s00018-013-1323-3] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2012] [Revised: 02/21/2013] [Accepted: 03/07/2013] [Indexed: 02/06/2023]
Abstract
Regucalcin (RGN) is a calcium (Ca(2+))-binding protein widely expressed in vertebrate and invertebrate species, which is also known as senescence marker protein 30, due to its molecular weight (33 kDa) and a characteristically diminished expression with the aging process. RGN regulates intracellular Ca(2+) homeostasis and the activity of several proteins involved in intracellular signalling pathways, namely, kinases, phosphatases, phosphodiesterase, nitric oxide synthase and proteases, which highlights its importance in cell biology. In addition, RGN has cytoprotective effects reducing intracellular levels of oxidative stress, also playing a role in the control of cell survival and apoptosis. Multiple factors have been identified regulating the cell levels of RGN transcripts and protein, and an altered expression pattern of this interesting protein has been found in cases of reproductive disorders, neurodegenerative diseases and cancer. Moreover, RGN is a serum-secreted protein, and its levels have been correlated with the stage of disease, which strongly suggests the usefulness of this protein as a potential biomarker for monitoring disease onset and progression. The present review aims to discuss the available information concerning RGN expression and function in distinct cell types and tissues, integrating cellular and molecular mechanisms in the context of normal and pathological conditions. Insight into the cellular actions of RGN will be a key step towards deepening the knowledge of the biology of several human diseases.
Collapse
Affiliation(s)
- Ricardo Marques
- CICS-UBI, Health Sciences Research Centre, University of Beira Interior, Covilhã, Portugal
| | - Cláudio J. Maia
- CICS-UBI, Health Sciences Research Centre, University of Beira Interior, Covilhã, Portugal
| | - Cátia Vaz
- CICS-UBI, Health Sciences Research Centre, University of Beira Interior, Covilhã, Portugal
| | - Sara Correia
- CICS-UBI, Health Sciences Research Centre, University of Beira Interior, Covilhã, Portugal
| | - Sílvia Socorro
- CICS-UBI, Health Sciences Research Centre, University of Beira Interior, Covilhã, Portugal
| |
Collapse
|
48
|
Pedunculopontine Cholinergic Cell Loss in Hallucinating Parkinson Disease Patients but Not in Dementia With Lewy Bodies Patients. J Neuropathol Exp Neurol 2013; 72:1162-70. [DOI: 10.1097/nen.0000000000000014] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
|
49
|
Yin Y, Yu G, Chen Y, Jiang S, Wang M, Jin Y, Lan X, Liang Y, Sun H. Genome-wide transcriptome and proteome analysis on different developmental stages of Cordyceps militaris. PLoS One 2012; 7:e51853. [PMID: 23251642 PMCID: PMC3522581 DOI: 10.1371/journal.pone.0051853] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2012] [Accepted: 11/13/2012] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Cordyceps militaris, an ascomycete caterpillar fungus, has been used as a traditional Chinese medicine for many years owing to its anticancer and immunomodulatory activities. Currently, artificial culturing of this beneficial fungus has been widely used and can meet the market, but systematic molecular studies on the developmental stages of cultured C. militaris at transcriptional and translational levels have not been determined. METHODOLOGY/PRINCIPAL FINDINGS We utilized high-throughput Illumina sequencing to obtain the transcriptomes of C. militaris mycelium and fruiting body. All clean reads were mapped to C. militaris genome and most of the reads showed perfect coverage. Alternative splicing and novel transcripts were predicted to enrich the database. Gene expression analysis revealed that 2,113 genes were up-regulated in mycelium and 599 in fruiting body. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis were performed to analyze the genes with expression differences. Moreover, the putative cordycepin metabolism difference between different developmental stages was studied. In addition, the proteome data of mycelium and fruiting body were obtained by one-dimensional gel electrophoresis (1-DGE) coupled with nano-electrospray ionization liquid chromatography tandem mass spectrometry (nESI-LC-MS/MS). 359 and 214 proteins were detected from mycelium and fruiting body respectively. GO, KEGG and Cluster of Orthologous Groups (COG) analysis were further conducted to better understand their difference. We analyzed the amounts of some noteworthy proteins in these two samples including lectin, superoxide dismutase, glycoside hydrolase and proteins involved in cordycepin metabolism, providing important information for further protein studies. CONCLUSIONS/SIGNIFICANCE The results reveal the difference in gene expression between the mycelium and fruiting body of artificially cultivated C. militaris by transcriptome and proteome analysis. Our study provides an effective resource for the further developmental and medicinal research of this promising fungus.
Collapse
Affiliation(s)
- Yalin Yin
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, Hubei Province, People’s Republic of China
| | - Guojun Yu
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, Hubei Province, People’s Republic of China
| | - Yijie Chen
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, Hubei Province, People’s Republic of China
| | - Shuai Jiang
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, Hubei Province, People’s Republic of China
| | - Man Wang
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, Hubei Province, People’s Republic of China
| | - Yanxia Jin
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, Hubei Province, People’s Republic of China
| | - Xianqing Lan
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, Hubei Province, People’s Republic of China
| | - Yi Liang
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, Hubei Province, People’s Republic of China
- Department of Clinical Immunology, Guangdong Medical College, Dongguan, People’s Republic of China
| | - Hui Sun
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, Hubei Province, People’s Republic of China
- Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan, People’s Republic of China
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery (Wuhan University), Ministry of Education, Wuhan, People’s Republic of China
- * E-mail:
| |
Collapse
|
50
|
Jellinger KA. Interaction between pathogenic proteins in neurodegenerative disorders. J Cell Mol Med 2012; 16:1166-83. [PMID: 22176890 PMCID: PMC3823071 DOI: 10.1111/j.1582-4934.2011.01507.x] [Citation(s) in RCA: 94] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2011] [Accepted: 12/16/2011] [Indexed: 12/21/2022] Open
Abstract
The misfolding and progressive aggregation of specific proteins in selective regions of the nervous system is a seminal occurrence in many neurodegenerative disorders, and the interaction between pathological/toxic proteins to cause neurodegeneration is a hot topic of current neuroscience research. Despite clinical, genetic and experimental differences, increasing evidence indicates considerable overlap between synucleinopathies, tauopathies and other protein-misfolding diseases. Inclusions, often characteristic hallmarks of these disorders, suggest interactions of pathological proteins enganging common downstream pathways. Novel findings that have shifted our understanding in the role of pathologic proteins in the pathogenesis of Alzheimer, Parkinson, Huntington and prion diseases, have confirmed correlations/overlaps between these and other neurodegenerative disorders. Emerging evidence, in addition to synergistic effects of tau protein, amyloid-β, α-synuclein and other pathologic proteins, suggests that prion-like induction and spreading, involving secreted proteins, are major pathogenic mechanisms in various neurodegenerative diseases, depending on genetic backgrounds and environmental factors. The elucidation of the basic molecular mechanisms underlying the interaction and spreading of pathogenic proteins, suggesting a dualism or triad of neurodegeneration in protein-misfolding disorders, is a major challenge for modern neuroscience, to provide a deeper insight into their pathogenesis as a basis of effective diagnosis and treatment.
Collapse
|