1
|
Bramble MS, Fourcassié V, Vashist N, Roux-Dalvai F, Zhou Y, Bumoko G, Kasendue ML, Spencer D, Musasa Hanshi-Hatuhu H, Kambale-Mastaki V, Manalo RVM, Mohammed A, McIlwain DR, Cunningham G, Summar M, Boivin MJ, Caldovic L, Vilain E, Mumba-Ngoyi D, Tshala-Katumbay D, Droit A. Glutathione peroxidase 3 is a potential biomarker for konzo. Nat Commun 2024; 15:7811. [PMID: 39242582 PMCID: PMC11379914 DOI: 10.1038/s41467-024-52136-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Accepted: 08/27/2024] [Indexed: 09/09/2024] Open
Abstract
Konzo is a neglected paralytic neurological disease associated with food (cassava) poisoning that affects the world's poorest children and women of childbearing ages across regions of sub-Saharan Africa. Despite understanding the dietary factors that lead to konzo, the molecular markers and mechanisms that trigger this disease remain unknown. To identify potential protein biomarkers associated with a disease status, plasma was collected from two independent Congolese cohorts, a discovery cohort (n = 60) and validation cohort (n = 204), sampled 10 years apart and subjected to multiple high-throughput assays. We identified that Glutathione Peroxidase 3 (GPx3), a critical plasma-based antioxidant enzyme, was the sole protein examined that was both significantly and differentially abundant between affected and non-affected participants in both cohorts, with large reductions observed in those affected with konzo. Our findings raise the notion that reductions in key antioxidant mechanisms may be the biological risk factor for the development of konzo, particularly those mediated through pathways involving the glutathione peroxidase family.
Collapse
Affiliation(s)
- Matthew S Bramble
- Center for Genetic Medicine Research, Children's Research Institute, Children's National Hospital, Washington, DC, USA.
- Department of Genomics and Precision Medicine, The George Washington University of Medicine and Health Sciences, Washington, DC, USA.
| | - Victor Fourcassié
- Computational Biology Laboratory and The Proteomics Platform, CHU de Québec - Université Laval Research Center, Québec City, QC, Canada
| | - Neerja Vashist
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, UCLA, Los Angeles, CA, USA
| | - Florence Roux-Dalvai
- Computational Biology Laboratory and The Proteomics Platform, CHU de Québec - Université Laval Research Center, Québec City, QC, Canada
| | - Yun Zhou
- Center for Genetic Medicine Research, Children's Research Institute, Children's National Hospital, Washington, DC, USA
| | - Guy Bumoko
- Department of Neurology, Kinshasa University, Kinshasa, Democratic Republic of the Congo
| | - Michel Lupamba Kasendue
- Institut National de Recherche Biomédicale (INRB), Kinshasa, Democratic Republic of the Congo
| | - D'Andre Spencer
- Center for Genetic Medicine Research, Children's Research Institute, Children's National Hospital, Washington, DC, USA
| | - Hilaire Musasa Hanshi-Hatuhu
- Department of Neurology, Kinshasa University, Kinshasa, Democratic Republic of the Congo
- Institut National de Recherche Biomédicale (INRB), Kinshasa, Democratic Republic of the Congo
| | - Vincent Kambale-Mastaki
- Institut National de Recherche Biomédicale (INRB), Kinshasa, Democratic Republic of the Congo
| | - Rafael Vincent M Manalo
- Biological Models Laboratory, Department of Biochemistry and Molecular Biology, College of Medicine, University of the Philippines, Manila, Ermita, Manila, Philippines
| | - Aliyah Mohammed
- Center for Genetic Medicine Research, Children's Research Institute, Children's National Hospital, Washington, DC, USA
| | - David R McIlwain
- Department of Microbiology and Immunology, University of Nevada, Reno School of Medicine, Reno, NV, USA
| | - Gary Cunningham
- Center for Genetic Medicine Research, Children's Research Institute, Children's National Hospital, Washington, DC, USA
| | - Marshall Summar
- Center for Genetic Medicine Research, Children's Research Institute, Children's National Hospital, Washington, DC, USA
| | - Michael J Boivin
- Departments of Psychiatry and Neurology & Ophthalmology, Michigan State University, East Lansing, MI, USA
| | - Ljubica Caldovic
- Center for Genetic Medicine Research, Children's Research Institute, Children's National Hospital, Washington, DC, USA
- Department of Genomics and Precision Medicine, The George Washington University of Medicine and Health Sciences, Washington, DC, USA
| | - Eric Vilain
- Institute for Clinical and Translational Science, University of California, Irvine, CA, USA
| | - Dieudonne Mumba-Ngoyi
- Institut National de Recherche Biomédicale (INRB), Kinshasa, Democratic Republic of the Congo
| | - Desire Tshala-Katumbay
- Institut National de Recherche Biomédicale (INRB), Kinshasa, Democratic Republic of the Congo.
- Department of Neurology, Oregon Health & Science University, Portland, OR, USA.
| | - Arnaud Droit
- Computational Biology Laboratory and The Proteomics Platform, CHU de Québec - Université Laval Research Center, Québec City, QC, Canada.
| |
Collapse
|
2
|
Tian L, Zhao S, Ding F, Zhang R. ITIH4 reversed the effects of thrombin on VSMCs stiffness via JNK and ERK signaling pathway. Exp Cell Res 2024; 441:114189. [PMID: 39069151 DOI: 10.1016/j.yexcr.2024.114189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 06/23/2024] [Accepted: 07/26/2024] [Indexed: 07/30/2024]
Abstract
Vascular smooth muscle cell (VSMCs) is one of the important cell types in artery. VSMCs stiffening may regulate vascular stiffness and contribute to the development of vulnerable plaques. Thrombin, an enzyme in coagulation system, is involved in pathological processes of atherosclerosis. Inter-alpha-trypsin inhibitor heavy chain 4 (ITIH4) plays an important role in regulating inflammation and may have cardiovascular protective effect. Therefore, the elucidation of the mechanisms underlying ITIH4-mediated VSMCs stiffening helps to provide new ideas and potential targets for the diagnosis and treatment of atherosclerosis. In this study, we used specific ITIH4 expression vector and siRNA methods to transfect VSMCs. Our results found that ITIH4 expression increased VSMCs stiffness, meanwhile, ITIH4 siRNA decreased VSMCs stiffness. ITIH4 increased acetylated α-tubulin and inhibited ERK1/2 and JNK, but not P38 MAPK. ERK inhibitor (PD98059) or JNK inhibitor (SP600125) treatment increased acetylated α-tubulin expression and cell stiffness in VSMCs. ITIH4 was downregulated by thrombin treatment, ITIH4 partly reversed the effect of thrombin on acetylated α-tubulin and VSMCs stiffness. These results indicated that ITIH4 regulated acetylated α-tubulin expression in VSMCs and was against the effects of thrombin on VSMCs stiffness. JNK and ERK signaling pathways were proved to participate in this process.
Collapse
MESH Headings
- Thrombin/pharmacology
- Thrombin/metabolism
- MAP Kinase Signaling System/drug effects
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/drug effects
- Animals
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/drug effects
- Vascular Stiffness/drug effects
- Cells, Cultured
- Rats
- Humans
- Rats, Sprague-Dawley
- Peptide Hormones/metabolism
- Peptide Hormones/pharmacology
- Peptide Hormones/genetics
Collapse
Affiliation(s)
- Lei Tian
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China.
| | - Su Zhao
- Department of Pharmacy, Ruijin Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Fenghua Ding
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Ruiyan Zhang
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China.
| |
Collapse
|
3
|
Troldborg A, Godnic-Polai Z, Cervenak L, Hansen AG, Farkas H, Thiel S. Inter-α-trypsin inhibitor heavy chain 4 (ITIH4) as a compensatory protease inhibitor in hereditary angioedema. J Allergy Clin Immunol 2024; 154:468-479.e6. [PMID: 38657796 DOI: 10.1016/j.jaci.2024.03.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 03/14/2024] [Accepted: 03/29/2024] [Indexed: 04/26/2024]
Abstract
BACKGROUND Hereditary angioedema (HAE) is a genetic disorder that manifests as recurrent angioedema attacks, most frequently due to absent or reduced C1 inhibitor (C1INH) activity. C1INH is a crucial regulator of enzymatic cascades in the complement, fibrinolytic, and contact systems. Inter-α-trypsin inhibitor heavy chain 4 (ITIH4) is an abundant plasma protease inhibitor that can inhibit enzymes in the proteolytic pathways associated with HAE. Nothing is known about its role in HAE. OBJECTIVE We investigated ITIH4 activation in HAE, establishing it as a potential biomarker, and explored its involvement in HAE-associated proteolytic pathways. METHODS Specific immunoassays for noncleaved ITIH4 (intact ITIH4) and an assay detecting both intact and cleaved ITIH4 (total ITIH4) were developed. We initially tested serum samples from HAE patients (n = 20), angiotensin-converting enzyme inhibitor-induced edema patients (ACEI) (n = 20), and patients with HAE of unknown cause (HAE-UNK) (n = 20). Validation involved an extended cohort of 80 HAE patients (60 with HAE-C1INH type 1, 20 with HAE-C1INH type 2), including samples taken during attack and quiescent disease periods, as well as samples from 100 healthy controls. RESULTS In 63% of HAE patients, intact ITIH4 assay showed lower signals than total ITIH4 assay. This difference was not observed in ACEI and HAE-UNK patients. Western blot analysis confirmed cleaved ITIH4 with low intact ITIH4 samples. In serum samples lacking intact endogenous ITIH4, we observed immediate cleavage of added recombinant ITIH4, suggesting continuous enzymatic activity in the serum. Confirmatory HAE cohort analysis revealed significantly lower intact ITIH4 levels in both type 1 and type 2 HAE patients compared to controls, with consistently low intact/total ITIH4 ratios during clinical HAE attacks. CONCLUSION The disease-specific low intact ITIH4 levels highlight its unique nature in HAE. ITIH4 may exhibit compensatory mechanisms in HAE, suggesting its utility as a diagnostic and prognostic biomarker. The variations during quiescent and active disease periods raise intriguing questions about the dynamics of proteolytic pathways in HAE.
Collapse
Affiliation(s)
- Anne Troldborg
- Department of Biomedicine, Aarhus University, Aarhus, Denmark; Department of Clinical Medicine, Aarhus University, Aarhus, Denmark; Department of Rheumatology, Aarhus University Hospital, Aarhus, Denmark.
| | - Zsofia Godnic-Polai
- Department of Internal Medicine and Haematology, Hungarian Angioedema Center of Reference and Excellence, Semmelweis University, Budapest, Hungary
| | - László Cervenak
- Department of Internal Medicine and Haematology, Semmelweis University, Budapest, Hungary
| | | | - Henriette Farkas
- Department of Internal Medicine and Haematology, Hungarian Angioedema Center of Reference and Excellence, Semmelweis University, Budapest, Hungary
| | - Steffen Thiel
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| |
Collapse
|
4
|
Pottier C, Küçükali F, Baker M, Batzler A, Jenkins GD, van Blitterswijk M, Vicente CT, De Coster W, Wynants S, Van de Walle P, Ross OA, Murray ME, Faura J, Haggarty SJ, van Rooij JG, Mol MO, Hsiung GYR, Graff C, Öijerstedt L, Neumann M, Asmann Y, McDonnell SK, Baheti S, Josephs KA, Whitwell JL, Bieniek KF, Forsberg L, Heuer H, Lago AL, Geier EG, Yokoyama JS, Oddi AP, Flanagan M, Mao Q, Hodges JR, Kwok JB, Domoto-Reilly K, Synofzik M, Wilke C, Onyike C, Dickerson BC, Evers BM, Dugger BN, Munoz DG, Keith J, Zinman L, Rogaeva E, Suh E, Gefen T, Geula C, Weintraub S, Diehl-Schmid J, Farlow MR, Edbauer D, Woodruff BK, Caselli RJ, Donker Kaat LL, Huey ED, Reiman EM, Mead S, King A, Roeber S, Nana AL, Ertekin-Taner N, Knopman DS, Petersen RC, Petrucelli L, Uitti RJ, Wszolek ZK, Ramos EM, Grinberg LT, Gorno Tempini ML, Rosen HJ, Spina S, Piguet O, Grossman M, Trojanowski JQ, Keene DC, Lee-Way J, Prudlo J, Geschwind DH, Rissman RA, Cruchaga C, Ghetti B, Halliday GM, Beach TG, Serrano GE, Arzberger T, Herms J, Boxer AL, Honig LS, Vonsattel JP, Lopez OL, Kofler J, White CL, Gearing M, Glass J, Rohrer JD, Irwin DJ, Lee EB, Van Deerlin V, Castellani R, Mesulam MM, Tartaglia MC, Finger EC, Troakes C, Al-Sarraj S, Miller BL, Seelaar H, Graff-Radford NR, Boeve BF, Mackenzie IR, van Swieten JC, Seeley WW, Sleegers K, Dickson DW, Biernacka JM, Rademakers R. Deciphering Distinct Genetic Risk Factors for FTLD-TDP Pathological Subtypes via Whole-Genome Sequencing. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.06.24.24309088. [PMID: 38978643 PMCID: PMC11230325 DOI: 10.1101/2024.06.24.24309088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
Frontotemporal lobar degeneration with neuronal inclusions of the TAR DNA-binding protein 43 (FTLD-TDP) is a fatal neurodegenerative disorder with only a limited number of risk loci identified. We report our comprehensive genome-wide association study as part of the International FTLD-TDP Whole-Genome Sequencing Consortium, including 985 cases and 3,153 controls, and meta-analysis with the Dementia-seq cohort, compiled from 26 institutions/brain banks in the United States, Europe and Australia. We confirm UNC13A as the strongest overall FTLD-TDP risk factor and identify TNIP1 as a novel FTLD-TDP risk factor. In subgroup analyses, we further identify for the first time genome-wide significant loci specific to each of the three main FTLD-TDP pathological subtypes (A, B and C), as well as enrichment of risk loci in distinct tissues, brain regions, and neuronal subtypes, suggesting distinct disease aetiologies in each of the subtypes. Rare variant analysis confirmed TBK1 and identified VIPR1 , RBPJL , and L3MBTL1 as novel subtype specific FTLD-TDP risk genes, further highlighting the role of innate and adaptive immunity and notch signalling pathway in FTLD-TDP, with potential diagnostic and novel therapeutic implications.
Collapse
|
5
|
Zhang N, Liao H, Lin Z, Tang Q. Insights into the Role of Glutathione Peroxidase 3 in Non-Neoplastic Diseases. Biomolecules 2024; 14:689. [PMID: 38927092 PMCID: PMC11202029 DOI: 10.3390/biom14060689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 06/03/2024] [Accepted: 06/05/2024] [Indexed: 06/28/2024] Open
Abstract
Reactive oxygen species (ROSs) are byproducts of normal cellular metabolism and play pivotal roles in various physiological processes. Disruptions in the balance between ROS levels and the body's antioxidant defenses can lead to the development of numerous diseases. Glutathione peroxidase 3 (GPX3), a key component of the body's antioxidant system, is an oxidoreductase enzyme. GPX3 mitigates oxidative damage by catalyzing the conversion of hydrogen peroxide into water. Beyond its antioxidant function, GPX3 is vital in regulating metabolism, modulating cell growth, inducing apoptosis and facilitating signal transduction. It also serves as a significant tumor suppressor in various cancers. Recent studies have revealed aberrant expression of GPX3 in several non-neoplastic diseases, associating it with multiple pathological processes. This review synthesizes the current understanding of GPX3 expression and regulation, highlighting its extensive roles in noncancerous diseases. Additionally, this paper evaluates the potential of GPX3 as a diagnostic biomarker and explores emerging therapeutic strategies targeting this enzyme, offering potential avenues for future clinical treatment of non-neoplastic conditions.
Collapse
Affiliation(s)
- Nan Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China; (N.Z.); (H.L.)
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan 430060, China
| | - Haihan Liao
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China; (N.Z.); (H.L.)
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan 430060, China
| | - Zheng Lin
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China; (N.Z.); (H.L.)
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan 430060, China
| | - Qizhu Tang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China; (N.Z.); (H.L.)
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan 430060, China
| |
Collapse
|
6
|
Duan QQ, Wang H, Su WM, Gu XJ, Shen XF, Jiang Z, Ren YL, Cao B, Li GB, Wang Y, Chen YP. TBK1, a prioritized drug repurposing target for amyotrophic lateral sclerosis: evidence from druggable genome Mendelian randomization and pharmacological verification in vitro. BMC Med 2024; 22:96. [PMID: 38443977 PMCID: PMC10916235 DOI: 10.1186/s12916-024-03314-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Accepted: 02/23/2024] [Indexed: 03/07/2024] Open
Abstract
BACKGROUND There is a lack of effective therapeutic strategies for amyotrophic lateral sclerosis (ALS); therefore, drug repurposing might provide a rapid approach to meet the urgent need for treatment. METHODS To identify therapeutic targets associated with ALS, we conducted Mendelian randomization (MR) analysis and colocalization analysis using cis-eQTL of druggable gene and ALS GWAS data collections to determine annotated druggable gene targets that exhibited significant associations with ALS. By subsequent repurposing drug discovery coupled with inclusion criteria selection, we identified several drug candidates corresponding to their druggable gene targets that have been genetically validated. The pharmacological assays were then conducted to further assess the efficacy of genetics-supported repurposed drugs for potential ALS therapy in various cellular models. RESULTS Through MR analysis, we identified potential ALS druggable genes in the blood, including TBK1 [OR 1.30, 95%CI (1.19, 1.42)], TNFSF12 [OR 1.36, 95%CI (1.19, 1.56)], GPX3 [OR 1.28, 95%CI (1.15, 1.43)], TNFSF13 [OR 0.45, 95%CI (0.32, 0.64)], and CD68 [OR 0.38, 95%CI (0.24, 0.58)]. Additionally, we identified potential ALS druggable genes in the brain, including RESP18 [OR 1.11, 95%CI (1.07, 1.16)], GPX3 [OR 0.57, 95%CI (0.48, 0.68)], GDF9 [OR 0.77, 95%CI (0.67, 0.88)], and PTPRN [OR 0.17, 95%CI (0.08, 0.34)]. Among them, TBK1, TNFSF12, RESP18, and GPX3 were confirmed in further colocalization analysis. We identified five drugs with repurposing opportunities targeting TBK1, TNFSF12, and GPX3, namely fostamatinib (R788), amlexanox (AMX), BIIB-023, RG-7212, and glutathione as potential repurposing drugs. R788 and AMX were prioritized due to their genetic supports, safety profiles, and cost-effectiveness evaluation. Further pharmacological analysis revealed that R788 and AMX mitigated neuroinflammation in ALS cell models characterized by overly active cGAS/STING signaling that was induced by MSA-2 or ALS-related toxic proteins (TDP-43 and SOD1), through the inhibition of TBK1 phosphorylation. CONCLUSIONS Our MR analyses provided genetic evidence supporting TBK1, TNFSF12, RESP18, and GPX3 as druggable genes for ALS treatment. Among the drug candidates targeting the above genes with repurposing opportunities, FDA-approved drug-R788 and AMX served as effective TBK1 inhibitors. The subsequent pharmacological studies validated the potential of R788 and AMX for treating specific ALS subtypes through the inhibition of TBK1 phosphorylation.
Collapse
Affiliation(s)
- Qing-Qing Duan
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Institute of Brain Science and Brain-Inspired Technology, West China Hospital, Sichuan University, Sichuan, Chengdu,, 610041, China
- Rare Disease Center, West China Hospital, Sichuan University, Sichuan, Chengdu, 610041, China
| | - Han Wang
- Department of Pathophysiology, West China College of Basic Medical Sciences and Forensic Medicine, Sichuan University, Sichuan, Chengdu, 610041, China
| | - Wei-Ming Su
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Institute of Brain Science and Brain-Inspired Technology, West China Hospital, Sichuan University, Sichuan, Chengdu,, 610041, China
- Rare Disease Center, West China Hospital, Sichuan University, Sichuan, Chengdu, 610041, China
| | - Xiao-Jing Gu
- Mental Health Center, West China Hospital, Sichuan University, Sichuan, Chengdu, 610041, China
| | - Xiao-Fei Shen
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China
| | - Zheng Jiang
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Institute of Brain Science and Brain-Inspired Technology, West China Hospital, Sichuan University, Sichuan, Chengdu,, 610041, China
- Rare Disease Center, West China Hospital, Sichuan University, Sichuan, Chengdu, 610041, China
| | - Yan-Ling Ren
- Department of Pathophysiology, West China College of Basic Medical Sciences and Forensic Medicine, Sichuan University, Sichuan, Chengdu, 610041, China
| | - Bei Cao
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Institute of Brain Science and Brain-Inspired Technology, West China Hospital, Sichuan University, Sichuan, Chengdu,, 610041, China
- Rare Disease Center, West China Hospital, Sichuan University, Sichuan, Chengdu, 610041, China
| | - Guo-Bo Li
- Key Laboratory of Drug Targeting and Drug Delivery System of Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| | - Yi Wang
- Department of Pathophysiology, West China College of Basic Medical Sciences and Forensic Medicine, Sichuan University, Sichuan, Chengdu, 610041, China.
| | - Yong-Ping Chen
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
- Institute of Brain Science and Brain-Inspired Technology, West China Hospital, Sichuan University, Sichuan, Chengdu,, 610041, China.
- Rare Disease Center, West China Hospital, Sichuan University, Sichuan, Chengdu, 610041, China.
| |
Collapse
|
7
|
Wainberg M, Andrews SJ, Tripathy SJ. Shared genetic risk loci between Alzheimer's disease and related dementias, Parkinson's disease, and amyotrophic lateral sclerosis. Alzheimers Res Ther 2023; 15:113. [PMID: 37328865 PMCID: PMC10273745 DOI: 10.1186/s13195-023-01244-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Accepted: 05/16/2023] [Indexed: 06/18/2023]
Abstract
BACKGROUND Genome-wide association studies (GWAS) have indicated moderate genetic overlap between Alzheimer's disease (AD) and related dementias (ADRD), Parkinson's disease (PD) and amyotrophic lateral sclerosis (ALS), neurodegenerative disorders traditionally considered etiologically distinct. However, the specific genetic variants and loci underlying this overlap remain almost entirely unknown. METHODS We leveraged state-of-the-art GWAS for ADRD, PD, and ALS. For each pair of disorders, we examined each of the GWAS hits for one disorder and tested whether they were also significant for the other disorder, applying Bonferroni correction for the number of variants tested. This approach rigorously controls the family-wise error rate for both disorders, analogously to genome-wide significance. RESULTS Eleven loci with GWAS hits for one disorder were also associated with one or both of the other disorders: one with all three disorders (the MAPT/KANSL1 locus), five with ADRD and PD (near LCORL, CLU, SETD1A/KAT8, WWOX, and GRN), three with ADRD and ALS (near GPX3, HS3ST5/HDAC2/MARCKS, and TSPOAP1), and two with PD and ALS (near GAK/TMEM175 and NEK1). Two of these loci (LCORL and NEK1) were associated with an increased risk of one disorder but decreased risk of another. Colocalization analysis supported a shared causal variant between ADRD and PD at the CLU, WWOX, and LCORL loci, between ADRD and ALS at the TSPOAP1 locus, and between PD and ALS at the NEK1 and GAK/TMEM175 loci. To address the concern that ADRD is an imperfect proxy for AD and that the ADRD and PD GWAS have overlapping participants (nearly all of which are from the UK Biobank), we confirmed that all our ADRD associations had nearly identical odds ratios in an AD GWAS that excluded the UK Biobank, and all but one remained nominally significant (p < 0.05) for AD. CONCLUSIONS In one of the most comprehensive investigations to date of pleiotropy between neurodegenerative disorders, we identify eleven genetic risk loci shared among ADRD, PD, and ALS. These loci support lysosomal/autophagic dysfunction (GAK/TMEM175, GRN, KANSL1), neuroinflammation/immunity (TSPOAP1), oxidative stress (GPX3, KANSL1), and the DNA damage response (NEK1) as transdiagnostic processes underlying multiple neurodegenerative disorders.
Collapse
Affiliation(s)
- Michael Wainberg
- Centre for Addiction and Mental Health, 250 College Street, Toronto, M5T 1R8, Canada
| | - Shea J Andrews
- Department of Psychiatry & Behavioral Sciences, University of California San Francisco, San Francisco, 94143, USA
| | - Shreejoy J Tripathy
- Centre for Addiction and Mental Health, 250 College Street, Toronto, M5T 1R8, Canada.
- Institute of Medical Sciences, University of Toronto, Toronto, M5S 1A8, Canada.
- Department of Psychiatry, University of Toronto, Toronto, M5T 1R8, Canada.
- Department of Physiology, University of Toronto, Toronto, M5S 1A8, Canada.
| |
Collapse
|
8
|
PGK1 modulates balance between pro- and anti-inflammatory cytokines by interacting with ITI-H4. Biomed Pharmacother 2023; 161:114437. [PMID: 36841032 DOI: 10.1016/j.biopha.2023.114437] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 02/16/2023] [Accepted: 02/21/2023] [Indexed: 02/27/2023] Open
Abstract
Inter-α-trypsin inhibitor heavy chain 4 (ITI-H4) is one of the acute phase proteins and is mainly related with inflammatory diseases such as bacterial bloodstream infection and recurrent pregnancy loss (RPL). In a previous study, ITI-H4 was reported to be cleaved by kallikrein B1 (KLKB1) and its cleaved form induces the imbalance between pro- and anti-inflammatory cytokines. Therefore, in this study, putative substrates of ITI-H4 were isolated by immunoprecipitation and matrix-assisted laser desorption/ionization time-of-flight mass spectrometry (MALDI-TOF/MS) analysis. Of those, phosphoglycerate kinase 1 (PGK1) was found to be a binding protein of ITI-H4. PGK1 increases the level of ITI-H4 expression and blocks the cleavage of ITI-H4 mediated by KLKB1. It also inhibits pro-inflammatory response by inhibiting the JAK2/STAT3 signaling pathway. Therefore, PGK1, a novel binding partner of ITI-H4, is expected to have cellular functions in the pathogenesis of ITI-H4-related inflammatory diseases.
Collapse
|
9
|
Restuadi R, Steyn FJ, Kabashi E, Ngo ST, Cheng FF, Nabais MF, Thompson MJ, Qi T, Wu Y, Henders AK, Wallace L, Bye CR, Turner BJ, Ziser L, Mathers S, McCombe PA, Needham M, Schultz D, Kiernan MC, van Rheenen W, van den Berg LH, Veldink JH, Ophoff R, Gusev A, Zaitlen N, McRae AF, Henderson RD, Wray NR, Giacomotto J, Garton FC. Functional characterisation of the amyotrophic lateral sclerosis risk locus GPX3/TNIP1. Genome Med 2022; 14:7. [PMID: 35042540 PMCID: PMC8767698 DOI: 10.1186/s13073-021-01006-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Accepted: 11/30/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Amyotrophic lateral sclerosis (ALS) is a complex, late-onset, neurodegenerative disease with a genetic contribution to disease liability. Genome-wide association studies (GWAS) have identified ten risk loci to date, including the TNIP1/GPX3 locus on chromosome five. Given association analysis data alone cannot determine the most plausible risk gene for this locus, we undertook a comprehensive suite of in silico, in vivo and in vitro studies to address this. METHODS The Functional Mapping and Annotation (FUMA) pipeline and five tools (conditional and joint analysis (GCTA-COJO), Stratified Linkage Disequilibrium Score Regression (S-LDSC), Polygenic Priority Scoring (PoPS), Summary-based Mendelian Randomisation (SMR-HEIDI) and transcriptome-wide association study (TWAS) analyses) were used to perform bioinformatic integration of GWAS data (Ncases = 20,806, Ncontrols = 59,804) with 'omics reference datasets including the blood (eQTLgen consortium N = 31,684) and brain (N = 2581). This was followed up by specific expression studies in ALS case-control cohorts (microarray Ntotal = 942, protein Ntotal = 300) and gene knockdown (KD) studies of human neuronal iPSC cells and zebrafish-morpholinos (MO). RESULTS SMR analyses implicated both TNIP1 and GPX3 (p < 1.15 × 10-6), but there was no simple SNP/expression relationship. Integrating multiple datasets using PoPS supported GPX3 but not TNIP1. In vivo expression analyses from blood in ALS cases identified that lower GPX3 expression correlated with a more progressed disease (ALS functional rating score, p = 5.5 × 10-3, adjusted R2 = 0.042, Beffect = 27.4 ± 13.3 ng/ml/ALSFRS unit) with microarray and protein data suggesting lower expression with risk allele (recessive model p = 0.06, p = 0.02 respectively). Validation in vivo indicated gpx3 KD caused significant motor deficits in zebrafish-MO (mean difference vs. control ± 95% CI, vs. control, swim distance = 112 ± 28 mm, time = 1.29 ± 0.59 s, speed = 32.0 ± 2.53 mm/s, respectively, p for all < 0.0001), which were rescued with gpx3 expression, with no phenotype identified with tnip1 KD or gpx3 overexpression. CONCLUSIONS These results support GPX3 as a lead ALS risk gene in this locus, with more data needed to confirm/reject a role for TNIP1. This has implications for understanding disease mechanisms (GPX3 acts in the same pathway as SOD1, a well-established ALS-associated gene) and identifying new therapeutic approaches. Few previous examples of in-depth investigations of risk loci in ALS exist and a similar approach could be applied to investigate future expected GWAS findings.
Collapse
Affiliation(s)
- Restuadi Restuadi
- Institute for Molecular Bioscience, The University of Queensland, QLD, Brisbane, 4072, Australia
| | - Frederik J Steyn
- School of Biomedical Sciences, The University of Queensland, QLD, Brisbane, 4072, Australia
- Department of Neurology, Royal Brisbane and Women's Hospital, QLD, Brisbane, 4029, Australia
- Centre for Clinical Research, The University of Queensland, QLD, Brisbane, 4019, Australia
| | - Edor Kabashi
- Imagine Institute, Institut National de la Santé et de la Recherche Médicale (INSERM) Unité 1163, Paris Descartes Université, 75015, Paris, France
- Sorbonne Université, Université Pierre et Marie Curie (UPMC), Université de Paris 06, INSERM Unité 1127, Centre National de la Recherche Scientifique (CNRS) Unité Mixte de Recherche 7225, Institut du Cerveau et de la Moelle Épinière (ICM), 75013, Paris, France
| | - Shyuan T Ngo
- Centre for Clinical Research, The University of Queensland, QLD, Brisbane, 4019, Australia
- Queensland Brain Institute, The University of Queensland, QLD, Brisbane, 4072, Australia
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, QLD, Brisbane, 4072, Australia
| | - Fei-Fei Cheng
- Institute for Molecular Bioscience, The University of Queensland, QLD, Brisbane, 4072, Australia
| | - Marta F Nabais
- Institute for Molecular Bioscience, The University of Queensland, QLD, Brisbane, 4072, Australia
- University of Exeter Medical School, RILD Building, RD&E Hospital Wonford, Barrack Road, Exeter, EX2 5DW, UK
| | - Mike J Thompson
- Department of Computer Science, University of California Los Angeles, Los Angeles, CA, USA
- Department of Bioinformatics, University of California Los Angeles, Los Angeles, CA, USA
| | - Ting Qi
- Institute for Molecular Bioscience, The University of Queensland, QLD, Brisbane, 4072, Australia
| | - Yang Wu
- Institute for Molecular Bioscience, The University of Queensland, QLD, Brisbane, 4072, Australia
| | - Anjali K Henders
- Institute for Molecular Bioscience, The University of Queensland, QLD, Brisbane, 4072, Australia
| | - Leanne Wallace
- Institute for Molecular Bioscience, The University of Queensland, QLD, Brisbane, 4072, Australia
| | - Chris R Bye
- Florey Institute for Neuroscience and Mental Health, University of Melbourne, Melbourne, VIC, 3052, Australia
| | - Bradley J Turner
- Florey Institute for Neuroscience and Mental Health, University of Melbourne, Melbourne, VIC, 3052, Australia
| | - Laura Ziser
- Institute for Molecular Bioscience, The University of Queensland, QLD, Brisbane, 4072, Australia
| | - Susan Mathers
- Calvary Health Care Bethlehem, Parkdale, VIC, 3195, Australia
| | - Pamela A McCombe
- Department of Neurology, Royal Brisbane and Women's Hospital, QLD, Brisbane, 4029, Australia
- Centre for Clinical Research, The University of Queensland, QLD, Brisbane, 4019, Australia
| | - Merrilee Needham
- Fiona Stanley Hospital, Perth, WA, 6150, Australia
- Notre Dame University, Fremantle, WA, 6160, Australia
- Institute for Immunology and Infectious Diseases, Murdoch University, Perth, WA, 6150, Australia
| | - David Schultz
- Department of Neurology, Flinders Medical Centre, Bedford Park, SA, 5042, Australia
| | - Matthew C Kiernan
- Brain & Mind Centre, University of Sydney, Institute of Clinical Neurosciences, Royal Prince Alfred Hospital, Sydney, NSW, 2006, Australia
| | - Wouter van Rheenen
- Department of Neurology, University Medical Center Utrecht Brain Center, Utrecht University, Utrecht, The Netherlands
| | - Leonard H van den Berg
- Department of Neurology, University Medical Center Utrecht Brain Center, Utrecht University, Utrecht, The Netherlands
| | - Jan H Veldink
- Department of Neurology, University Medical Center Utrecht Brain Center, Utrecht University, Utrecht, The Netherlands
| | - Roel Ophoff
- Department of Computer Science, University of California Los Angeles, Los Angeles, CA, USA
- Department of Bioinformatics, University of California Los Angeles, Los Angeles, CA, USA
| | - Alexander Gusev
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA
- Division of Genetics, Brigham and Women's Hospital, Boston, MA, USA
| | - Noah Zaitlen
- Department of Computer Science, University of California Los Angeles, Los Angeles, CA, USA
- Department of Bioinformatics, University of California Los Angeles, Los Angeles, CA, USA
- Department of Neurology, University of California Los Angeles, Los Angeles, CA, 90095, USA
- Department of Medicine, University of California San Francisco, San Francisco, CA, 94158, USA
| | - Allan F McRae
- Institute for Molecular Bioscience, The University of Queensland, QLD, Brisbane, 4072, Australia
| | - Robert D Henderson
- Department of Neurology, Royal Brisbane and Women's Hospital, QLD, Brisbane, 4029, Australia
- Centre for Clinical Research, The University of Queensland, QLD, Brisbane, 4019, Australia
- Queensland Brain Institute, The University of Queensland, QLD, Brisbane, 4072, Australia
| | - Naomi R Wray
- Institute for Molecular Bioscience, The University of Queensland, QLD, Brisbane, 4072, Australia
- Queensland Brain Institute, The University of Queensland, QLD, Brisbane, 4072, Australia
| | - Jean Giacomotto
- Queensland Brain Institute, The University of Queensland, QLD, Brisbane, 4072, Australia
- Queensland Centre for Mental Health Research, West Moreton Hospital and Health Service, Wacol, QLD, 4076, Australia
| | - Fleur C Garton
- Institute for Molecular Bioscience, The University of Queensland, QLD, Brisbane, 4072, Australia.
| |
Collapse
|
10
|
Soler L, Szczubiał M, Dąbrowski R, Płusa A, Bochniarz M, Brodzki P, Lampreave F, Piñeiro M. Measurement of ITIH4 and Hp levels in bitches with pyometra using newly developed ELISA methods. Vet Immunol Immunopathol 2021; 235:110221. [PMID: 33730638 DOI: 10.1016/j.vetimm.2021.110221] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 02/22/2021] [Accepted: 02/27/2021] [Indexed: 12/01/2022]
Abstract
Measurement of acute phase proteins (APPs) as biomarkers in canine medicine is in increasing demand. In the present study, the development and validation of two ELISA methods for the quantification of canine inter-alpha-trypsin inhibitor heavy chain 4 (ITIH4) and haptoglobin (Hp) are shown. The adequate imprecision and accuracy and wide analytical range make the developed methods appropriate to quantify ITIH4 and Hp in serum samples. The inter- and intra-assay CVs were lower than 10 %, and the assays maintained linearity under dilution and showed analytical equivalence with the method of radial immunodiffusion. The measurement of CRP, Hp and ITIH4 in sera from bitches affected by pyometra allowed us to determine that ITIH4 behaves as a moderate APP in dogs. The group of bitches affected by pyometra showed very high levels of CRP (147 ± 91 mg/L), corresponding to a strong inflammatory process, which resulted in a moderate increase in the concentrations of Hp (7 times) and ITIH4 (3 times) compared to the control group.
Collapse
Affiliation(s)
- Lourdes Soler
- Acuvet Biotech, C/Bari 25 dpdo, Zaragoza, 50197, Spain.
| | - Marek Szczubiał
- Department and Clinic of Animal Reproduction, Faculty of Veterinary Medicine, University of Life Sciences, Gleboka 30, Lublin, 20-612, Poland
| | - Roman Dąbrowski
- Department and Clinic of Animal Reproduction, Faculty of Veterinary Medicine, University of Life Sciences, Gleboka 30, Lublin, 20-612, Poland
| | - Anna Płusa
- Department and Clinic of Animal Reproduction, Faculty of Veterinary Medicine, University of Life Sciences, Gleboka 30, Lublin, 20-612, Poland
| | - Mariola Bochniarz
- Department and Clinic of Animal Reproduction, Faculty of Veterinary Medicine, University of Life Sciences, Gleboka 30, Lublin, 20-612, Poland
| | - Piotr Brodzki
- Department and Clinic of Animal Reproduction, Faculty of Veterinary Medicine, University of Life Sciences, Gleboka 30, Lublin, 20-612, Poland
| | - Fermín Lampreave
- Departamento de Bioquímica y Biología Molecular y Celular, Facultad de Ciencias, Universidad de Zaragoza, Pedro Cerbuna 12, Zaragoza, 50009, Spain
| | | |
Collapse
|
11
|
Li CY, Yang TM, Ou RW, Wei QQ, Shang HF. Genome-wide genetic links between amyotrophic lateral sclerosis and autoimmune diseases. BMC Med 2021; 19:27. [PMID: 33541344 PMCID: PMC7863260 DOI: 10.1186/s12916-021-01903-y] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Accepted: 01/05/2021] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Epidemiological and clinical studies have suggested comorbidity between amyotrophic lateral sclerosis (ALS) and autoimmune disorders. However, little is known about their shared genetic architecture. METHODS To examine the relation between ALS and 10 autoimmune diseases, including asthma, celiac disease (CeD), Crohn's disease (CD), inflammatory bowel disease (IBD), multiple sclerosis (MS), psoriasis, rheumatoid arthritis (RA), systemic lupus erythematosus (SLE), type 1 diabetes (T1D), and ulcerative colitis (UC), and identify shared risk loci, we first estimated the genetic correlation using summary statistics from genome-wide association studies, and then analyzed the genetic enrichment leveraging the conditional false discovery rate statistical method. RESULTS We identified a significant positive genetic correlation between ALS and CeD, MS, RA, and SLE, as well as a significant negative genetic correlation between ALS and IBD, UC, and CD. Robust genetic enrichment was observed between ALS and CeD and MS, and moderate enrichment was found between ALS and UC and T1D. Thirteen shared genetic loci were identified, among which five were suggestively significant in another ALS GWAS, namely rs3828599 (GPX3), rs3849943 (C9orf72), rs7154847 (G2E3), rs6571361 (SCFD1), and rs9903355 (GGNBP2). By integrating cis-expression quantitative trait loci analyses in Braineac and GTEx, we further identified GGNBP2, ATXN3, and SLC9A8 as novel ALS risk genes. Functional enrichment analysis indicated that the shared risk genes were involved in four pathways including membrane trafficking, vesicle-mediated transport, ER to Golgi anterograde transport, and transport to the Golgi and subsequent modification. CONCLUSIONS Our findings demonstrate a specific genetic correlation between ALS and autoimmune diseases and identify shared risk loci, including three novel ALS risk genes. These results provide a better understanding for the pleiotropy of ALS and have implications for future therapeutic trials.
Collapse
Affiliation(s)
- Chun Yu Li
- Department of Neurology, Laboratory of Neurodegenerative Disorders, National Clinical Research Center for Geriatric, West China Hospital, Sichuan University, Chengdu, China
| | - Tian Mi Yang
- Department of Neurology, Laboratory of Neurodegenerative Disorders, National Clinical Research Center for Geriatric, West China Hospital, Sichuan University, Chengdu, China
| | - Ru Wei Ou
- Department of Neurology, Laboratory of Neurodegenerative Disorders, National Clinical Research Center for Geriatric, West China Hospital, Sichuan University, Chengdu, China
| | - Qian Qian Wei
- Department of Neurology, Laboratory of Neurodegenerative Disorders, National Clinical Research Center for Geriatric, West China Hospital, Sichuan University, Chengdu, China
| | - Hui Fang Shang
- Department of Neurology, Laboratory of Neurodegenerative Disorders, National Clinical Research Center for Geriatric, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
12
|
Pihl R, Jensen RK, Poulsen EC, Jensen L, Hansen AG, Thøgersen IB, Dobó J, Gál P, Andersen GR, Enghild JJ, Thiel S. ITIH4 acts as a protease inhibitor by a novel inhibitory mechanism. SCIENCE ADVANCES 2021; 7:7/2/eaba7381. [PMID: 33523981 PMCID: PMC7793589 DOI: 10.1126/sciadv.aba7381] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Accepted: 11/16/2020] [Indexed: 05/02/2023]
Abstract
Inter-α-inhibitor heavy chain 4 (ITIH4) is a poorly characterized plasma protein that is proteolytically processed in multiple pathological conditions. However, no biological function of ITIH4 has been identified. Here, we show that ITIH4 is cleaved by several human proteases within a protease-susceptible region, enabling ITIH4 to function as a protease inhibitor. This is exemplified by its inhibition of mannan-binding lectin-associated serine protease-1 (MASP-1), MASP-2, and plasma kallikrein, which are key proteases for intravascular host defense. Mechanistically, ITIH4 acts as bait that, upon cleavage, forms a noncovalent, inhibitory complex with the executing protease that depends on the ITIH4 von Willebrand factor A domain. ITIH4 inhibits the MASPs by sterically preventing larger protein substrates from accessing their active sites, which remain accessible and fully functional toward small substrates. Thus, we demonstrate that ITIH4 functions as a protease inhibitor by a previously undescribed inhibitory mechanism.
Collapse
Affiliation(s)
- Rasmus Pihl
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Rasmus K Jensen
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| | - Emil C Poulsen
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| | - Lisbeth Jensen
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | | | - Ida B Thøgersen
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| | - József Dobó
- Institute of Enzymology, Research Centre for Natural Sciences, Budapest, Hungary
| | - Péter Gál
- Institute of Enzymology, Research Centre for Natural Sciences, Budapest, Hungary
| | - Gregers R Andersen
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| | - Jan J Enghild
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| | - Steffen Thiel
- Department of Biomedicine, Aarhus University, Aarhus, Denmark.
| |
Collapse
|
13
|
Soler L, García N, Andrés M, Armengol R, Lampreave F, Alava MA, Piñeiro M. Development and validation of an ELISA for the quantification of bovine ITIH4 in serum and milk. Vet Immunol Immunopathol 2019; 217:109922. [PMID: 31450165 DOI: 10.1016/j.vetimm.2019.109922] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Revised: 07/31/2019] [Accepted: 08/07/2019] [Indexed: 12/12/2022]
Abstract
Inter alpha trypsin inhibitor heavy chain 4 (ITIH4) is a serum protein belonging to the Inter alpha trypsin inhibitor (ITI) family, which was previously characterized by our group as a new APP in cattle. This protein was firstly described in pigs where is known to be a major acute phase protein, also denominated Pig-MAP. Increases of ITIH4 of up to 12 times the pre-infection values were previously reported in the serum of heifers with experimentally induced summer mastitis. ITIH4 was detected in the milk of cows with mastitis by western blot, but the method previously used to quantify this protein, radial immunodiffusion, was not sensitive enough to quantify it in milk samples. In this study we developed an ELISA method which allows the quantification of bovine ITIH4 in serum and milk samples. Previously developed antibodies were used to perform the assay, including anti bovine ITIH4 polyclonal antibodies and a monoclonal antibody against pig ITIH4 that also recognizes the bovine homologous protein. The ELISA developed showed an adequate precision, with inter and intra- assay coefficients of variation lower than 10% for serum and milk samples. The assay keeps linearity under dilution for both serum and milk samples. A good agreement was observed between the values measured by ELISA and radial immunodiffusion in serum samples.
Collapse
Affiliation(s)
- Lourdes Soler
- Acuvet Biotech, C/ Bari 25 dpdo, 50197, Zaragoza, Spain; Departamento de Bioquímica y Biología Molecular y Celular, Facultad de Ciencias, Universidad de Zaragoza, Pedro Cerbuna 12, 50009, Zaragoza, Spain
| | - Natalia García
- Departamento de Bioquímica y Biología Molecular y Celular, Facultad de Ciencias, Universidad de Zaragoza, Pedro Cerbuna 12, 50009, Zaragoza, Spain
| | - Marta Andrés
- Departamento de Bioquímica y Biología Molecular y Celular, Facultad de Ciencias, Universidad de Zaragoza, Pedro Cerbuna 12, 50009, Zaragoza, Spain
| | - Ramón Armengol
- Departament de Ciència Animal, ETSEA, University of Lleida, Av. Alcalde Rovira Roure 191, E25198, Lleida, Spain
| | - Fermín Lampreave
- Departamento de Bioquímica y Biología Molecular y Celular, Facultad de Ciencias, Universidad de Zaragoza, Pedro Cerbuna 12, 50009, Zaragoza, Spain
| | - María A Alava
- Departamento de Bioquímica y Biología Molecular y Celular, Facultad de Ciencias, Universidad de Zaragoza, Pedro Cerbuna 12, 50009, Zaragoza, Spain
| | | |
Collapse
|
14
|
Oxidative Stress in Neurodegenerative Diseases: From a Mitochondrial Point of View. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:2105607. [PMID: 31210837 PMCID: PMC6532273 DOI: 10.1155/2019/2105607] [Citation(s) in RCA: 298] [Impact Index Per Article: 59.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Accepted: 04/15/2019] [Indexed: 12/12/2022]
Abstract
Age is the main risk factor for a number of human diseases, including neurodegenerative disorders such as Alzheimer's disease, Parkinson's disease, and amyotrophic lateral sclerosis, which increasing numbers of elderly individuals suffer. These pathological conditions are characterized by progressive loss of neuron cells, compromised motor or cognitive functions, and accumulation of abnormally aggregated proteins. Mitochondrial dysfunction is one of the main features of the aging process, particularly in organs requiring a high-energy source such as the heart, muscles, brain, or liver. Neurons rely almost exclusively on the mitochondria, which produce the energy required for most of the cellular processes, including synaptic plasticity and neurotransmitter synthesis. The brain is particularly vulnerable to oxidative stress and damage, because of its high oxygen consumption, low antioxidant defenses, and high content of polyunsaturated fats very prone to be oxidized. Thus, it is not surprising the importance of protecting systems, including antioxidant defenses, to maintain neuronal integrity and survival. Here, we review the role of mitochondrial oxidative stress in the aging process, with a specific focus on neurodegenerative diseases. Understanding the molecular mechanisms involving mitochondria and oxidative stress in the aging and neurodegeneration may help to identify new strategies for improving the health and extending lifespan.
Collapse
|
15
|
Liguori M, Nuzziello N, Licciulli F, Consiglio A, Simone M, Viterbo RG, Creanza TM, Ancona N, Tortorella C, Margari L, Grillo G, Giordano P, Liuni S, Trojano M. Combined microRNA and mRNA expression analysis in pediatric multiple sclerosis: an integrated approach to uncover novel pathogenic mechanisms of the disease. Hum Mol Genet 2019; 27:66-79. [PMID: 29087462 DOI: 10.1093/hmg/ddx385] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2017] [Accepted: 10/23/2017] [Indexed: 12/12/2022] Open
Abstract
Multiple sclerosis (MS) is a complex disease of the CNS that usually affects young adults, although 3-5% of cases are diagnosed in childhood and adolescence (hence called pediatric MS, PedMS). Genetic predisposition, among other factors, seems to contribute to the risk of the onset, in pediatric as in adult ages, but few studies have investigated the genetic 'environmentally naïve' load of PedMS. The main goal of this study was to identify circulating markers (miRNAs), target genes (mRNAs) and functional pathways associated with PedMS; we also verified the impact of miRNAs on clinical features, i.e. disability and cognitive performances. The investigation was performed in 19 PedMS and 20 pediatric controls (PCs) using a High-Throughput Next-generation Sequencing (HT-NGS) approach followed by an integrated bioinformatics/biostatistics analysis. Twelve miRNAs were significantly upregulated (let-7a-5p, let-7b-5p, miR-25-3p, miR-125a-5p, miR-942-5p, miR-221-3p, miR-652-3p, miR-182-5p, miR-185-5p, miR-181a-5p, miR-320a, miR-99b-5p) and 1 miRNA was downregulated (miR-148b-3p) in PedMS compared with PCs. The interactions between the significant miRNAs and their targets uncovered predicted genes (i.e. TNFSF13B, TLR2, BACH2, KLF4) related to immunological functions, as well as genes involved in autophagy-related processes (i.e. ATG16L1, SORT1, LAMP2) and ATPase activity (i.e. ABCA1, GPX3). No significant molecular profiles were associated with any PedMS demographic/clinical features. Both miRNAs and mRNA expressions predicted the phenotypes (PedMS-PC) with an accuracy of 92% and 91%, respectively. In our view, this original strategy of contemporary miRNA/mRNA analysis may help to shed light in the genetic background of the disease, suggesting further molecular investigations in novel pathogenic mechanisms.
Collapse
Affiliation(s)
- Maria Liguori
- National Research Council of Italy, Department of Biomedicine, Institute of Biomedical Technologies, Bari Section, 70125 Bari, Italy
| | - Nicoletta Nuzziello
- National Research Council of Italy, Department of Biomedicine, Institute of Biomedical Technologies, Bari Section, 70125 Bari, Italy.,Department of Basic Sciences, Neurosciences and Sense Organs, University of Bari, 70125 Bari, Italy
| | - Flavio Licciulli
- National Research Council of Italy, Department of Biomedicine, Institute of Biomedical Technologies, Bari Section, 70125 Bari, Italy
| | - Arianna Consiglio
- National Research Council of Italy, Department of Biomedicine, Institute of Biomedical Technologies, Bari Section, 70125 Bari, Italy
| | - Marta Simone
- Department of Basic Sciences, Neurosciences and Sense Organs, University of Bari, 70125 Bari, Italy
| | - Rosa Gemma Viterbo
- Department of Basic Sciences, Neurosciences and Sense Organs, University of Bari, 70125 Bari, Italy
| | - Teresa Maria Creanza
- National Research Council of Italy, Department of Engineering, ICT and Technology for Energy and Transportation, Institute of Intelligent Systems for Automation, 70125 Bari, Italy
| | - Nicola Ancona
- National Research Council of Italy, Department of Engineering, ICT and Technology for Energy and Transportation, Institute of Intelligent Systems for Automation, 70125 Bari, Italy
| | - Carla Tortorella
- Department of Basic Sciences, Neurosciences and Sense Organs, University of Bari, 70125 Bari, Italy.,Department of Neurosciences, San Camillo Forlanini Hospital, 00185 Rome, Italy
| | - Lucia Margari
- Department of Basic Sciences, Neurosciences and Sense Organs, University of Bari, 70125 Bari, Italy
| | - Giorgio Grillo
- National Research Council of Italy, Department of Biomedicine, Institute of Biomedical Technologies, Bari Section, 70125 Bari, Italy
| | - Paola Giordano
- General Paediatric Unit "B. Trambusti", Azienda Policlinico-Giovanni XXIII and University of Bari, 70125 Bari, Italy
| | - Sabino Liuni
- National Research Council of Italy, Department of Biomedicine, Institute of Biomedical Technologies, Bari Section, 70125 Bari, Italy
| | - Maria Trojano
- Department of Basic Sciences, Neurosciences and Sense Organs, University of Bari, 70125 Bari, Italy
| |
Collapse
|
16
|
Cross-ethnic meta-analysis identifies association of the GPX3-TNIP1 locus with amyotrophic lateral sclerosis. Nat Commun 2017; 8:611. [PMID: 28931804 PMCID: PMC5606989 DOI: 10.1038/s41467-017-00471-1] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Accepted: 06/30/2017] [Indexed: 01/22/2023] Open
Abstract
Cross-ethnic genetic studies can leverage power from differences in disease epidemiology and population-specific genetic architecture. In particular, the differences in linkage disequilibrium and allele frequency patterns across ethnic groups may increase gene-mapping resolution. Here we use cross-ethnic genetic data in sporadic amyotrophic lateral sclerosis (ALS), an adult-onset, rapidly progressing neurodegenerative disease. We report analyses of novel genome-wide association study data of 1,234 ALS cases and 2,850 controls. We find a significant association of rs10463311 spanning GPX3-TNIP1 with ALS (p = 1.3 × 10−8), with replication support from two independent Australian samples (combined 576 cases and 683 controls, p = 1.7 × 10−3). Both GPX3 and TNIP1 interact with other known ALS genes (SOD1 and OPTN, respectively). In addition, GGNBP2 was identified using gene-based analysis and summary statistics-based Mendelian randomization analysis, although further replication is needed to confirm this result. Our results increase our understanding of genetic aetiology of ALS. Amyotrophic lateral sclerosis (ALS) is a rapidly progressing neurodegenerative disease. Here, Wray and colleagues identify association of the GPX3-TNIP1 locus with ALS using cross-ethnic meta-analyses.
Collapse
|
17
|
Soler L, García N, Unzueta A, Piñeiro M, Álava M, Lampreave F. Purification and determination of C-reactive protein and inter-α-trypsin inhibitor heavy chain 4 in dogs after major surgery through generation of specific antibodies. Vet Immunol Immunopathol 2016; 179:26-31. [DOI: 10.1016/j.vetimm.2016.07.010] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Revised: 07/12/2016] [Accepted: 07/20/2016] [Indexed: 12/12/2022]
|
18
|
Niedzielska E, Smaga I, Gawlik M, Moniczewski A, Stankowicz P, Pera J, Filip M. Oxidative Stress in Neurodegenerative Diseases. Mol Neurobiol 2016; 53:4094-4125. [PMID: 26198567 PMCID: PMC4937091 DOI: 10.1007/s12035-015-9337-5] [Citation(s) in RCA: 496] [Impact Index Per Article: 62.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2015] [Accepted: 07/01/2015] [Indexed: 12/12/2022]
Abstract
The pathophysiologies of neurodegenerative diseases, including amyotrophic lateral sclerosis (ALS), Parkinson's disease (PD), and Alzheimer's disease (AD), are far from being fully explained. Oxidative stress (OS) has been proposed as one factor that plays a potential role in the pathogenesis of neurodegenerative disorders. Clinical and preclinical studies indicate that neurodegenerative diseases are characterized by higher levels of OS biomarkers and by lower levels of antioxidant defense biomarkers in the brain and peripheral tissues. In this article, we review the current knowledge regarding the involvement of OS in neurodegenerative diseases, based on clinical trials and animal studies. In addition, we analyze the effects of the drug-induced modulation of oxidative balance, and we explore pharmacotherapeutic strategies for OS reduction.
Collapse
Affiliation(s)
- Ewa Niedzielska
- Department of Toxicology, Chair of Toxicology, Faculty of Pharmacy, Jagiellonian University, Medical College, Medyczna 9, 30-688, Kraków, Poland
| | - Irena Smaga
- Department of Toxicology, Chair of Toxicology, Faculty of Pharmacy, Jagiellonian University, Medical College, Medyczna 9, 30-688, Kraków, Poland
| | - Maciej Gawlik
- Department of Toxicology, Chair of Toxicology, Faculty of Pharmacy, Jagiellonian University, Medical College, Medyczna 9, 30-688, Kraków, Poland
| | - Andrzej Moniczewski
- Department of Toxicology, Chair of Toxicology, Faculty of Pharmacy, Jagiellonian University, Medical College, Medyczna 9, 30-688, Kraków, Poland
| | - Piotr Stankowicz
- Department of Toxicology, Chair of Toxicology, Faculty of Pharmacy, Jagiellonian University, Medical College, Medyczna 9, 30-688, Kraków, Poland
| | - Joanna Pera
- Department of Neurology, Faculty of Medicine, Jagiellonian University, Medical College, Botaniczna 3, 31-503, Krakow, Poland
| | - Małgorzata Filip
- Department of Toxicology, Chair of Toxicology, Faculty of Pharmacy, Jagiellonian University, Medical College, Medyczna 9, 30-688, Kraków, Poland.
- Laboratory of Drug Addiction Pharmacology, Institute of Pharmacology, Polish Academy of Sciences, Smętna 12, 31-343, Kraków, Poland.
| |
Collapse
|
19
|
Huang J, Luo G, Zhang Z, Wang X, Ju Z, Qi C, Zhang Y, Wang C, Li R, Li J, Yin W, Xu Y, Moisá SJ, Loor JJ, Zhong J. iTRAQ-proteomics and bioinformatics analyses of mammary tissue from cows with clinical mastitis due to natural infection with Staphylococci aureus. BMC Genomics 2014; 15:839. [PMID: 25273983 PMCID: PMC4198675 DOI: 10.1186/1471-2164-15-839] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2014] [Accepted: 09/26/2014] [Indexed: 12/12/2022] Open
Abstract
Background Proteomics and bioinformatics may help us better understand the biological adaptations occurring during bovine mastitis. This systems approach also could help identify biomarkers for monitoring clinical and subclinical mastitis. The aim of the present study was to use isobaric tags for relative and absolute quantification (iTRAQ) to screen potential proteins associated with mastitis at late infectious stage. Results Healthy and mastitic cows’ mammary gland tissues were analyzed using iTRAQ combined with two-dimensional liquid chromatography-tandem mass spectrometry (2D-LC-MS/MS). Bioinformatics analyses of differentially expressed proteins were performed by means of Gene Ontology, metabolic pathways, transcriptional regulation networks using Blast2GO software, the Dynamic Impact Approach and Ingenuity Pathway Analysis. At a false discovery rate of 5%, a total of 768 proteins were identified from 6,499 peptides, which were matched with 15,879 spectra. Compared with healthy mammary gland tissue, 36 proteins were significantly up-regulated (>1.5-fold) while 19 were significantly down-regulated (<0.67-fold) in response to mastitis due to natural infections with Staphylococci aureus. Up-regulation of collagen, type I, alpha 1 (COL1A1) and inter-alpha (Globulin) inhibitor H4 (ITIH4) in the mastitis-infected tissue was confirmed by Western blotting and Immunohistochemistry. Conclusion This paper is the first to show the protein expression in the late response to a mastitic pathogen, thus, revealing mechanisms associated with host tissue damage. The bioinformatics analyses highlighted the effects of mastitis on proteins such as collagen, fibrinogen, fibronectin, casein alpha and heparan sulfate proteoglycan 2. Our findings provide additional clues for further studies of candidate genes for mastitis susceptibility. The up-regulated expression of COL1A1 and ITIH4 in the mastitic mammary gland may be associated with tissue damage and repair during late stages of infection. Electronic supplementary material The online version of this article (doi:10.1186/1471-2164-15-839) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jinming Huang
- Dairy Cattle Research Center, Shandong Academy of Agricultural Sciences, No,159 North of Industry Road, Jinan, Shandong 250131, China.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Tanaka H, Hara H. [Aiming to overcome ALS - the biomarkers for ALS]. Nihon Yakurigaku Zasshi 2014; 143:313. [PMID: 24919559 DOI: 10.1254/fpj.143.313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
|
21
|
Lu W, Wan X, Liu B, Rong X, Zhu L, Li P, Li J, Wang L, Cui L, Wang X. Specific changes of serum proteins in Parkinson's disease patients. PLoS One 2014; 9:e95684. [PMID: 24769800 PMCID: PMC4000217 DOI: 10.1371/journal.pone.0095684] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2013] [Accepted: 03/28/2014] [Indexed: 12/12/2022] Open
Abstract
The aim of this study is to identify and validate protein change in the serum from PD patients. We used serum samples from 21 PD patients and 20 age-matched normal people as control to conduct a comparative proteomic study. We performed 2-DE and analyzed the differentially expressed protein spots by LC-MS/MS. In PD group 13 spots were shown to be differentially expressed compared to control group. They were identified as 6 proteins. Among these, 3 proteins were confirmed by Western blot analysis. It showed that the frequency of fibrinogen γ-chain (FGG) appeared 70% in PD, which could not be detected in control group. The protein of inter-alpha-trypsin inhibitor heavy chain H4 (ITI-H4) was found to exist two forms in serum. The full size (120 kDa) of the protein was increased and the fragmented ITI-H4 (35 kDa) was decreased in PD group. The ratio of full size ITI-H4 to fragmented ITI-H4 in PD patients was 3.85±0.29-fold higher than in control group. Furthermore, fragmented Apo A-IV (∼26 kDa) was mainly detected in control group, while it was rare to be found in PD group. Above findings might be useful for diagnosis of PD. When the expressions of FGG and 120 kDa ITI-H4 are increase, as well as ∼26 kDa Apo A-IV disappear would provide strong evidence for PD.
Collapse
Affiliation(s)
- Wenwen Lu
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Department of Pharmacology, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Xinhua Wan
- Department of Neurology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Bin Liu
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Department of Pharmacology, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Xianfang Rong
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Department of Pharmacology, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Lei Zhu
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Department of Pharmacology, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Pingping Li
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Department of Pharmacology, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Jiang Li
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Department of Pharmacology, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Ling Wang
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Department of Pharmacology, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Liying Cui
- Department of Neurology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
- * E-mail: (LC); (XW)
| | - Xiaoliang Wang
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Department of Pharmacology, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
- * E-mail: (LC); (XW)
| |
Collapse
|