1
|
de Souza Ferreira LP, da Silva RA, Borges PP, Xavier LF, Scharf P, Sandri S, Oliani SM, Farsky SHP, Gil CD. Annexin A1 in neurological disorders: Neuroprotection and glial modulation. Pharmacol Ther 2025; 267:108809. [PMID: 39900227 DOI: 10.1016/j.pharmthera.2025.108809] [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: 08/24/2024] [Revised: 12/20/2024] [Accepted: 01/31/2025] [Indexed: 02/05/2025]
Abstract
Neurological disorders, such as neurodegenerative and neuroinflammatory diseases, have contributed significantly to global disability, even considering the rising life years expectations. Therefore, prevention, early diagnosis, and therapeutic alternatives have been essential to avoid the future collapse of health public systems. Annexin A1 (ANXA1), a Ca2 + -dependent protein, is a promising therapeutic candidate for neurological disorders. ANXA1, found in neurons and glia, displays roles in physiological and pathological processes. Despite ANXA1 undoubtedly maintains the blood-brain barrier (BBB) integrity, this review will focus on ANXA1 roles in neurons and glial cells. In neurons, the cytoplasmic expression of ANXA1 is associated with apoptosis, while its nuclear translocation is linked to ischemic neuronal death. Interactions with S100A11, the Tat-NTS peptide, and other molecules, modulate this translocation, suggesting potential therapeutic interventions. ANXA1 expressed on microglia modulates inflammation and efferocytosis. Post-translational modifications, such as SUMOylation, guide the role of ANXA1 in microglia polarization and neuroprotection. In addition, ANXA1 in astrocytes responds to inflammatory stimuli by influencing cytokine release. A comprehensive understanding of the intricate mechanisms of ANXA1 in neurons and glial cells reveals promising therapeutic strategies to alleviate neuronal damage in neurological diseases.
Collapse
Affiliation(s)
- Luiz Philipe de Souza Ferreira
- Structural and Functional Biology Graduate Program, Escola Paulista de Medicina, Universidade Federal de São Paulo (UNIFESP), São Paulo, SP, Brazil.
| | - Rafael André da Silva
- Biosciences Graduate Program, Instituto de Biociências, Letras e Ciências Exatas - IBILCE, Universidade Estadual Paulista (UNESP), São José do Rio Preto, SP, Brazil
| | - Pâmela Pacassa Borges
- Department of Clinical and Toxicological Analyses, Faculdade de Ciências Farmacêuticas, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Luana Filippi Xavier
- Department of Clinical and Toxicological Analyses, Faculdade de Ciências Farmacêuticas, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Pablo Scharf
- Department of Clinical and Toxicological Analyses, Faculdade de Ciências Farmacêuticas, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Silvana Sandri
- Department of Clinical and Toxicological Analyses, Faculdade de Ciências Farmacêuticas, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Sonia M Oliani
- Structural and Functional Biology Graduate Program, Escola Paulista de Medicina, Universidade Federal de São Paulo (UNIFESP), São Paulo, SP, Brazil; Biosciences Graduate Program, Instituto de Biociências, Letras e Ciências Exatas - IBILCE, Universidade Estadual Paulista (UNESP), São José do Rio Preto, SP, Brazil
| | - Sandra H P Farsky
- Department of Clinical and Toxicological Analyses, Faculdade de Ciências Farmacêuticas, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Cristiane D Gil
- Structural and Functional Biology Graduate Program, Escola Paulista de Medicina, Universidade Federal de São Paulo (UNIFESP), São Paulo, SP, Brazil; Biosciences Graduate Program, Instituto de Biociências, Letras e Ciências Exatas - IBILCE, Universidade Estadual Paulista (UNESP), São José do Rio Preto, SP, Brazil.
| |
Collapse
|
2
|
Alonso ADC, El Idrissi A, Candia R, Morozova V, Kleiman FE. Tau: More than a microtubule-binding protein in neurons. Cytoskeleton (Hoboken) 2024; 81:71-77. [PMID: 37819542 DOI: 10.1002/cm.21795] [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: 07/29/2023] [Revised: 09/15/2023] [Accepted: 09/19/2023] [Indexed: 10/13/2023]
Abstract
Tau protein was discovered as a microtubule-associated protein nearly 50 years ago, and our understanding of tau has revolved around that role. Even with tau's rise to stardom as a central player in neurodegenerative disease, therapeutic efforts have largely been targeted toward cytoskeletal changes. While some studies hinted toward non-cytoskeletal roles for tau, it is only fairly recently that these ideas have begun to receive considerable attention. Many new binding partners for tau have been identified, including DNA, RNA, RNA-binding proteins, some receptors, and other tau molecules. The diversity of tau binding partners coupled with the discovery of tau other than axonal compartments such as nucleus, dendrites, and synapses have led to the proposal of novel functions for tau in roles such as nuclear stability, cell signaling, transcriptional processing, and protein synthesis. Tau self-assembly in particular has made an impact, leading to the hypothesis that a prion-like function of hyperphosphorylated tau is central to tauopathies. With tau emerging as a multifaceted protein that operates in many parts of the cell and with many molecular partners, the field of tau biology is primed for discoveries that can provide new perspectives on both the unique biochemistry of tau and the nature of devastating neurological diseases.
Collapse
Affiliation(s)
- Alejandra Del Carmen Alonso
- Biology Department and Center for Developmental Neuroscience, College of Staten Island, Staten Island, New York, USA
- Biology Program, The Graduate Center, The City University of New York, New York, New York, USA
| | - Abdeslem El Idrissi
- Biology Department and Center for Developmental Neuroscience, College of Staten Island, Staten Island, New York, USA
- Biology Program, The Graduate Center, The City University of New York, New York, New York, USA
| | - Robert Candia
- Biology Department and Center for Developmental Neuroscience, College of Staten Island, Staten Island, New York, USA
- Biology Program, The Graduate Center, The City University of New York, New York, New York, USA
| | - Viktoriya Morozova
- Biology Department and Center for Developmental Neuroscience, College of Staten Island, Staten Island, New York, USA
- Biology Program, The Graduate Center, The City University of New York, New York, New York, USA
- Helene Fuld College of Nursing, New York, New York, USA
| | - Frida Esther Kleiman
- Biology Program, The Graduate Center, The City University of New York, New York, New York, USA
- Chemistry Department, Hunter College, The City University of New York, New York, New York, USA
| |
Collapse
|
3
|
Merchant JP, Zhu K, Henrion MYR, Zaidi SSA, Lau B, Moein S, Alamprese ML, Pearse RV, Bennett DA, Ertekin-Taner N, Young-Pearse TL, Chang R. Predictive network analysis identifies JMJD6 and other potential key drivers in Alzheimer's disease. Commun Biol 2023; 6:503. [PMID: 37188718 PMCID: PMC10185548 DOI: 10.1038/s42003-023-04791-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Accepted: 03/31/2023] [Indexed: 05/17/2023] Open
Abstract
Despite decades of genetic studies on late-onset Alzheimer's disease, the underlying molecular mechanisms remain unclear. To better comprehend its complex etiology, we use an integrative approach to build robust predictive (causal) network models using two large human multi-omics datasets. We delineate bulk-tissue gene expression into single cell-type gene expression and integrate clinical and pathologic traits, single nucleotide variation, and deconvoluted gene expression for the construction of cell type-specific predictive network models. Here, we focus on neuron-specific network models and prioritize 19 predicted key drivers modulating Alzheimer's pathology, which we then validate by knockdown in human induced pluripotent stem cell-derived neurons. We find that neuronal knockdown of 10 of the 19 targets significantly modulates levels of amyloid-beta and/or phosphorylated tau peptides, most notably JMJD6. We also confirm our network structure by RNA sequencing in the neurons following knockdown of each of the 10 targets, which additionally predicts that they are upstream regulators of REST and VGF. Our work thus identifies robust neuronal key drivers of the Alzheimer's-associated network state which may represent therapeutic targets with relevance to both amyloid and tau pathology in Alzheimer's disease.
Collapse
Affiliation(s)
- Julie P Merchant
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Neuroscience Graduate Group, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Kuixi Zhu
- The Center for Innovation in Brain Sciences, University of Arizona, Tucson, AZ, USA
| | - Marc Y R Henrion
- Liverpool School of Tropical Medicine, Pembroke Place, Liverpool, Pembroke Place, L3 5QA, UK
- Malawi-Liverpool-Wellcome Trust Clinical Research Programme, PO Box 30096, Blantyre, Malawi
| | - Syed S A Zaidi
- The Center for Innovation in Brain Sciences, University of Arizona, Tucson, AZ, USA
| | - Branden Lau
- The Center for Innovation in Brain Sciences, University of Arizona, Tucson, AZ, USA
- Arizona Research Labs, Genetics Core, University of Arizona, Tucson, AZ, USA
| | - Sara Moein
- The Center for Innovation in Brain Sciences, University of Arizona, Tucson, AZ, USA
| | - Melissa L Alamprese
- The Center for Innovation in Brain Sciences, University of Arizona, Tucson, AZ, USA
| | - Richard V Pearse
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - David A Bennett
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - Nilüfer Ertekin-Taner
- Department of Neuroscience, Mayo Clinic Florida, Jacksonville, FL, USA
- Department of Neurology, Mayo Clinic Florida, Jacksonville, FL, USA
| | - Tracy L Young-Pearse
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA.
- Harvard Stem Cell Institute, Harvard University, Boston, MA, USA.
| | - Rui Chang
- The Center for Innovation in Brain Sciences, University of Arizona, Tucson, AZ, USA.
- Department of Neurology, University of Arizona, Tucson, AZ, USA.
- INTelico Therapeutics LLC, Tucson, AZ, USA.
- PATH Biotech LLC, Tucson, AZ, USA.
| |
Collapse
|
4
|
Dhaffouli F, Hachicha H, Abida O, Gharbi N, Elloumi N, Kanoun H, Belguith N, Marzouk S, Fakhfakh R, Sawsen F, Mnif H, Kamoun H, Bahloul Z, Masmoudi H. Annexin A1 and its receptor gene polymorphisms in systemic lupus erythematosus in the Tunisian population. Clin Rheumatol 2022; 41:1359-1369. [PMID: 35028743 DOI: 10.1007/s10067-022-06057-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Revised: 01/01/2022] [Accepted: 01/04/2022] [Indexed: 12/18/2022]
Abstract
BACKGROUND An association between ANXA1, FPR1 and FPR2 gene polymorphisms and the patho-physiology of many human diseases was suggested by numerous studies. OBJECTIVE Our study aimed to evaluate association between common polymorphisms in the 9q21.13 and 19q13.41 and susceptibility to systemic lupus erythematosus (SLE) in the Tunisian population. MATERIALS We performed a case-control study on 107 Tunisian SLE patients and 122 healthy controls to explore 9 polymorphisms of the three studied genes: rs2811226 and rs3739959 (ANXA1), rs5030880, rs1042229, rs1461765570, rs17849971, rs867228 (FPR1), rs17694990 and rs11666254 (FPR2). RESULTS Four polymorphisms were found to be linked with SLE susceptibility: rs3739959-ANXA1 > G and GG (p = 0.021, OR = 1.73 and p = 0.014, OR = 2.06 respectively), rs867228-FPR1 > TT (p = 0.014, OR = 4.59), rs11666254-FPR2 > GG (p = 0.019, OR = 8.34) and rs17694990-FPR2 > T (p = 0.05, OR = 1.506). In homogenous groups of SLE patients depending on clinical manifestations and serological results, previous associations were confirmed with a panoply of manifestations of lupus including lupus nephritis, malar rash, mouth ulceration and hypocomplementia. CONCLUSION Our study showed an association between ANXA1 > rs3739959, FPR1 > rs867228, FPR2 > rs11666254, FPR2 > rs17694990 and SLE susceptibility. Our results also showed a strong association between the two ANXA1 studied SNPs and LN which allowed us to suggest these two SNPs as biomarkers of LN development in SLE. Further research is needed to understand by which mechanism the gene variants affect susceptibility to SLE. Key Points • Lupus erythematosus is an autoimmune disease in which a panoply of factors are implicated • Annexin A1 interaction with its receptors are suggested as a target in therapy of a panoply of human disease in particular cancers • The present results highlighted the implication of Annexin A1 and its receptors gene polymorphisms in the physiopathology of lupus, in particular in the involvement of renal and cutaneous lesions.
Collapse
Affiliation(s)
- Fatma Dhaffouli
- Research Laboratoy LR18/SP12 "Autoimmunity, Cancer And Immunogenetics", Habib Bourguiba University Hospital, University of Sfax, Sfax, Tunisia.
| | - Hend Hachicha
- Research Laboratoy LR18/SP12 "Autoimmunity, Cancer And Immunogenetics", Habib Bourguiba University Hospital, University of Sfax, Sfax, Tunisia
- Department of Immunology, Habib Bourguiba University Hospital, University of Sfax, Sfax, Tunisia
| | - Olfa Abida
- Research Laboratoy LR18/SP12 "Autoimmunity, Cancer And Immunogenetics", Habib Bourguiba University Hospital, University of Sfax, Sfax, Tunisia
| | - Nourhene Gharbi
- Research Laboratory of Human Molecular Genetics, Hedi Chaker University Hospital of Sfax, Sfax, Tunisia
| | - Nesrine Elloumi
- Research Laboratoy LR18/SP12 "Autoimmunity, Cancer And Immunogenetics", Habib Bourguiba University Hospital, University of Sfax, Sfax, Tunisia
| | - Houda Kanoun
- Research Laboratory of Human Molecular Genetics, Hedi Chaker University Hospital of Sfax, Sfax, Tunisia
| | - Neila Belguith
- Research Laboratory of Human Molecular Genetics, Hedi Chaker University Hospital of Sfax, Sfax, Tunisia
- Department of Congenital and Hereditary Diseases, Charles Nicolle Hospital, Tunis, Tunisia
| | - Sameh Marzouk
- Department of Internal Medicine, Hedi Chaker University Hospital, Sfax, Tunisia
| | - Raouia Fakhfakh
- Research Laboratoy LR18/SP12 "Autoimmunity, Cancer And Immunogenetics", Habib Bourguiba University Hospital, University of Sfax, Sfax, Tunisia
| | - Feki Sawsen
- Research Laboratoy LR18/SP12 "Autoimmunity, Cancer And Immunogenetics", Habib Bourguiba University Hospital, University of Sfax, Sfax, Tunisia
- Department of Immunology, Habib Bourguiba University Hospital, University of Sfax, Sfax, Tunisia
| | - Hela Mnif
- The Regional Blood Transfusion Center of Sfax, Sfax, Tunisia
| | - Hassen Kamoun
- Research Laboratory of Human Molecular Genetics, Hedi Chaker University Hospital of Sfax, Sfax, Tunisia
| | - Zouhir Bahloul
- Department of Internal Medicine, Hedi Chaker University Hospital, Sfax, Tunisia
| | - Hatem Masmoudi
- Research Laboratoy LR18/SP12 "Autoimmunity, Cancer And Immunogenetics", Habib Bourguiba University Hospital, University of Sfax, Sfax, Tunisia
- Department of Immunology, Habib Bourguiba University Hospital, University of Sfax, Sfax, Tunisia
| |
Collapse
|
5
|
Jiang Y, Ma H, Zhang Q, Shi J, Gao Y, Sun C, Zhang W. Integrative analyses reveal RNA regulatory network in Ti particles induced inflammation. EUR J INFLAMM 2021. [DOI: 10.1177/20587392211044863] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Introduction Wear particles induced inflammatory osteolysis is the most important initiating factors in the mechanism of aseptic loosening. However, the molecular network changes in this process remain largely elusive. Methods Here, we performed whole transcriptome analysis using Ti particles induced RAW264.7 cell model to identify specific genes and pathways. Results Sequencing results totally identified 159 mRNAs, 96 lncRNAs, 31 circRNAs, and 12 miRNAs were significantly differently expressed. Of these, we selected two of each RNA for qRT-PCR validation and the results were highly consistent with the RNA-seq data. GSEA analysis shows that upregulated gene sets were related to the three classical inflammation pathway, cytokine–cytokine receptor interaction, TNF, and NF-kappa B signaling pathway. The enriched genes included not only IL-1β and TNF- α, which were independently verified before sequencing, but also other inflammatory osteolysis-related genes such as Mmp9, Fas, and Ccl2. Co-differentially expressed RNAs were employed to construct the ceRNA co-regulatory network. Conclusion: The results revealed that 4 lncRNAs and 2 circRNAs formed a regulatory network to simultaneously regulate miR-3065-3p targeting Myo18a. The present study helps to comprehensively understand the molecular mechanisms and regulatory interaction networks during early inflammatory response.
Collapse
Affiliation(s)
- Yonghui Jiang
- Center for Reproductive Medicine, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Huanzhi Ma
- Department of Orthopedics, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Qin Zhang
- Department of Orthopedics, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Jun Shi
- Department of Orthopedics, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Yutong Gao
- Department of Orthopedics, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Chengliang Sun
- Department of Orthopedics, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Wei Zhang
- Department of Orthopedics, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| |
Collapse
|
6
|
Cheng J, Liu HP, Lin WY, Tsai FJ. Machine learning compensates fold-change method and highlights oxidative phosphorylation in the brain transcriptome of Alzheimer's disease. Sci Rep 2021; 11:13704. [PMID: 34211065 PMCID: PMC8249453 DOI: 10.1038/s41598-021-93085-z] [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: 10/19/2020] [Accepted: 06/18/2021] [Indexed: 02/06/2023] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder causing 70% of dementia cases. However, the mechanism of disease development is still elusive. Despite the availability of a wide range of biological data, a comprehensive understanding of AD's mechanism from machine learning (ML) is so far unrealized, majorly due to the lack of needed data density. To harness the AD mechanism's knowledge from the expression profiles of postmortem prefrontal cortex samples of 310 AD and 157 controls, we used seven predictive operators or combinations of RapidMiner Studio operators to establish predictive models from the input matrix and to assign a weight to each attribute. Besides, conventional fold-change methods were also applied as controls. The identified genes were further submitted to enrichment analysis for KEGG pathways. The average accuracy of ML models ranges from 86.30% to 91.22%. The overlap ratio of the identified genes between ML and conventional methods ranges from 19.7% to 21.3%. ML exclusively identified oxidative phosphorylation genes in the AD pathway. Our results highlighted the deficiency of oxidative phosphorylation in AD and suggest that ML should be considered as complementary to the conventional fold-change methods in transcriptome studies.
Collapse
Affiliation(s)
- Jack Cheng
- grid.254145.30000 0001 0083 6092Graduate Institute of Integrated Medicine, College of Chinese Medicine, China Medical University, Taichung, 40402 Taiwan ,grid.411508.90000 0004 0572 9415Department of Medical Research, China Medical University Hospital, Taichung, 40447 Taiwan
| | - Hsin-Ping Liu
- grid.254145.30000 0001 0083 6092Graduate Institute of Acupuncture Science, College of Chinese Medicine, China Medical University, Taichung, 40402 Taiwan
| | - Wei-Yong Lin
- grid.254145.30000 0001 0083 6092Graduate Institute of Integrated Medicine, College of Chinese Medicine, China Medical University, Taichung, 40402 Taiwan ,grid.411508.90000 0004 0572 9415Department of Medical Research, China Medical University Hospital, Taichung, 40447 Taiwan ,grid.254145.30000 0001 0083 6092Brain Diseases Research Center, China Medical University, Taichung, 40402 Taiwan
| | - Fuu-Jen Tsai
- grid.411508.90000 0004 0572 9415Department of Medical Research, China Medical University Hospital, Taichung, 40447 Taiwan ,grid.254145.30000 0001 0083 6092School of Chinese Medicine, China Medical University, Taichung, 40402 Taiwan ,grid.252470.60000 0000 9263 9645Department of Medical Laboratory and Biotechnology, Asia University, Taichung, 41354 Taiwan ,grid.254145.30000 0001 0083 6092Division of Pediatric Genetics, Children’s Hospital of China Medical University, Taichung, 40447 Taiwan
| |
Collapse
|
7
|
Grewal T, Rentero C, Enrich C, Wahba M, Raabe CA, Rescher U. Annexin Animal Models-From Fundamental Principles to Translational Research. Int J Mol Sci 2021; 22:ijms22073439. [PMID: 33810523 PMCID: PMC8037771 DOI: 10.3390/ijms22073439] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 03/18/2021] [Accepted: 03/24/2021] [Indexed: 02/07/2023] Open
Abstract
Routine manipulation of the mouse genome has become a landmark in biomedical research. Traits that are only associated with advanced developmental stages can now be investigated within a living organism, and the in vivo analysis of corresponding phenotypes and functions advances the translation into the clinical setting. The annexins, a family of closely related calcium (Ca2+)- and lipid-binding proteins, are found at various intra- and extracellular locations, and interact with a broad range of membrane lipids and proteins. Their impacts on cellular functions has been extensively assessed in vitro, yet annexin-deficient mouse models generally develop normally and do not display obvious phenotypes. Only in recent years, studies examining genetically modified annexin mouse models which were exposed to stress conditions mimicking human disease often revealed striking phenotypes. This review is the first comprehensive overview of annexin-related research using animal models and their exciting future use for relevant issues in biology and experimental medicine.
Collapse
Affiliation(s)
- Thomas Grewal
- School of Pharmacy, Faculty of Medicine and Health, University of Sydney, Sydney, NSW 2006, Australia;
- Correspondence: (T.G.); (U.R.); Tel.: +61-(0)2-9351-8496 (T.G.); +49-(0)251-83-52121 (U.R.)
| | - Carles Rentero
- Departament de Biomedicina, Unitat de Biologia Cel·lular, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, 08036 Barcelona, Spain; (C.R.); (C.E.)
- Centre de Recerca Biomèdica CELLEX, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
| | - Carlos Enrich
- Departament de Biomedicina, Unitat de Biologia Cel·lular, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, 08036 Barcelona, Spain; (C.R.); (C.E.)
- Centre de Recerca Biomèdica CELLEX, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
| | - Mohamed Wahba
- School of Pharmacy, Faculty of Medicine and Health, University of Sydney, Sydney, NSW 2006, Australia;
| | - Carsten A. Raabe
- Research Group Regulatory Mechanisms of Inflammation, Center for Molecular Biology of Inflammation (ZMBE) and Cells in Motion Interfaculty Center (CiM), Institute of Medical Biochemistry, University of Muenster, 48149 Muenster, Germany;
| | - Ursula Rescher
- Research Group Regulatory Mechanisms of Inflammation, Center for Molecular Biology of Inflammation (ZMBE) and Cells in Motion Interfaculty Center (CiM), Institute of Medical Biochemistry, University of Muenster, 48149 Muenster, Germany;
- Correspondence: (T.G.); (U.R.); Tel.: +61-(0)2-9351-8496 (T.G.); +49-(0)251-83-52121 (U.R.)
| |
Collapse
|
8
|
Espregueira Themudo G, Leerschool AR, Rodriguez-Proano C, Christiansen SL, Andersen JD, Busch JR, Christensen MR, Banner J, Morling N. Targeted exon sequencing in deceased schizophrenia patients in Denmark. Int J Legal Med 2019; 134:135-147. [PMID: 31773318 DOI: 10.1007/s00414-019-02212-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Accepted: 11/13/2019] [Indexed: 12/26/2022]
Abstract
Schizophrenia patients have higher mortality rates and lower life expectancy than the general population. However, forensic investigations of their deaths often fail to determine the cause of death, hindering prevention. As schizophrenia is a highly heritable condition and given recent advances in our understanding of the genetics of schizophrenia, it is now possible to investigate how genetic factors may contribute to mortality. We made use of findings from genome-wide association studies (GWAS) to design a targeted panel (PsychPlex) for sequencing of exons of 451 genes near index single nucleotide polymorphisms (SNPs) identified with GWAS. We sequenced the DNA of 95 deceased schizophrenia patients included in SURVIVE, a prospective, autopsy-based study of mentally ill persons in Denmark. We compared the allele frequencies of 1039 SNPs in these cases with the frequencies of 2000 Danes without psychiatric diseases and calculated their deleteriousness (CADD) scores. For 81 SNPs highly associated with schizophrenia and CADD scores above 15, expression profiles in the Genotype-Tissue Expression (GTEx) Project indicated that these variants were in exons, whose expressions are increased in several types of brain tissues, particularly in the cerebellum. Molecular pathway analysis indicated the involvement of 163 different pathways. As for rare SNP variants, most variants were scored as either benign or likely benign with an average of 17 variants of unknown significance per individual and no pathogenic variant. Our results highlight the potential of DNA sequencing of an exon panel to discover genetic factors that may be involved in the development of schizophrenia.
Collapse
Affiliation(s)
- Gonçalo Espregueira Themudo
- Section of Forensic Genetics, Department of Forensic Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark. .,CIIMAR - Interdisciplinary Centre of Marine and Environmental Research of the University of Porto, Terminal de Cruzeiros do Porto de Leixões, Avenida General Norton de Matos, S/N, 4450-208, Matosinhos, Portugal.
| | - Anna-Roos Leerschool
- Section of Forensic Genetics, Department of Forensic Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.,Department of Complex Genetics, Maastricht University, PO Box 616 6200, MD, Maastricht, The Netherlands
| | - Carla Rodriguez-Proano
- Section of Forensic Genetics, Department of Forensic Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.,Clinical Laboratory, Ambulatory Clinical Surgical Center and Day Hospital "El Batán", Quito, Ecuador
| | - Sofie Lindgren Christiansen
- Section of Forensic Genetics, Department of Forensic Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jeppe Dyrberg Andersen
- Section of Forensic Genetics, Department of Forensic Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Johannes Rødbro Busch
- Section of Forensic Pathology, Department of Forensic Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Martin Roest Christensen
- Section of Forensic Pathology, Department of Forensic Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jytte Banner
- Section of Forensic Pathology, Department of Forensic Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Niels Morling
- Section of Forensic Genetics, Department of Forensic Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
9
|
Fraga VG, Magalhães CA, Loures CDMG, de Souza LC, Guimarães HC, Zauli DAG, Carvalho MDG, Ferreira CN, Caramelli P, de Sousa LP, Gomes KB. Inflammatory and Pro-resolving Mediators in Frontotemporal Dementia and Alzheimer's Disease. Neuroscience 2019; 421:123-135. [PMID: 31654714 DOI: 10.1016/j.neuroscience.2019.09.008] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Revised: 09/07/2019] [Accepted: 09/10/2019] [Indexed: 01/06/2023]
Abstract
Chronic inflammation contributes to neuronal death in Alzheimer's disease (AD) and frontotemporal dementia (FTD). Here we evaluated inflammatory and pro-resolving mediators in AD and behavioural variant of FTD (bvFTD) patients compared with controls, since neuroinflamamtion is a common feature in both diseases. Ninety-eight subjects were included in this study, divided into AD (n = 32), bvFTD (n = 30), and control (n = 36) groups. The levels of hsCRP, IL-1β, IL-6, TNF, and TGF-β1, as well as annexin A1 (AnxA1) and lipoxin A4 (LXA4) were measured in blood and cerebrospinal fluid (CSF). The expression profile of AnxA1 was evaluated in peripheral blood mononuclear cells (PBMCs) as well the distribution of ANXA1 rs2611228 polymorphism. We found reduced peripheral levels of hsCRP and TNF in AD compared with bvFTD patients and controls, and increased levels of TGF-β1 in AD compared to controls. Moreover, reduced plasma levels of AnxA1 were observed in bvFTD compared to AD and controls. There was a significant cleavage of AnxA1 in PBMCs in both dementia groups. The results suggest differential regulation of inflammatory and pro-resolving mediators in bvFTD and AD, while AnxA1 cleavage may impair pro-resolving mechanisms in both groups.
Collapse
Affiliation(s)
- Vanêssa Gomes Fraga
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Carolina Antunes Magalhães
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Cristina de Mello Gomide Loures
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Leonardo Cruz de Souza
- Departamento de Clínca Médica, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Henrique Cerqueira Guimarães
- Departamento de Clínca Médica, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | | | - Maria das Graças Carvalho
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | | | - Paulo Caramelli
- Departamento de Clínca Médica, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Lirlândia Pires de Sousa
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Karina Braga Gomes
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil.
| |
Collapse
|
10
|
Baquero J, Varriano S, Ordonez M, Kuczaj P, Murphy MR, Aruggoda G, Lundine D, Morozova V, Makki AE, Alonso ADC, Kleiman FE. Nuclear Tau, p53 and Pin1 Regulate PARN-Mediated Deadenylation and Gene Expression. Front Mol Neurosci 2019; 12:242. [PMID: 31749682 PMCID: PMC6843027 DOI: 10.3389/fnmol.2019.00242] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Accepted: 09/20/2019] [Indexed: 12/17/2022] Open
Abstract
While nuclear tau plays a role in DNA damage response (DDR) and chromosome relaxation, the mechanisms behind these functions are not fully understood. Here, we show that tau forms complex(es) with factors involved in nuclear mRNA processing such as tumor suppressor p53 and poly(A)-specific ribonuclease (PARN) deadenylase. Tau induces PARN activity in different cellular models during DDR, and this activation is further increased by p53 and inhibited by tau phosphorylation at residues implicated in neurological disorders. Tau's binding factor Pin1, a mitotic regulator overexpressed in cancer and depleted in Alzheimer's disease (AD), also plays a role in the activation of nuclear deadenylation. Tau, Pin1 and PARN target the expression of mRNAs deregulated in AD and/or cancer. Our findings identify novel biological roles of tau and toxic effects of hyperphosphorylated-tau. We propose a model in which factors involved in cancer and AD regulate gene expression by interactions with the mRNA processing machinery, affecting the transcriptome and suggesting insights into alternative mechanisms for the initiation and/or developments of these diseases.
Collapse
Affiliation(s)
- Jorge Baquero
- Chemistry Department, Hunter College and Biochemistry Program, The Graduate Center, The City University of New York, New York, NY, United States
| | - Sophia Varriano
- Chemistry Department, Hunter College and Biochemistry Program, The Graduate Center, The City University of New York, New York, NY, United States
| | - Martha Ordonez
- Chemistry Department, Hunter College and Biochemistry Program, The Graduate Center, The City University of New York, New York, NY, United States
| | - Pawel Kuczaj
- Chemistry Department, Hunter College and Biochemistry Program, The Graduate Center, The City University of New York, New York, NY, United States
| | - Michael R. Murphy
- Chemistry Department, Hunter College and Biochemistry Program, The Graduate Center, The City University of New York, New York, NY, United States
| | - Gamage Aruggoda
- Chemistry Department, Hunter College and Biochemistry Program, The Graduate Center, The City University of New York, New York, NY, United States
| | - Devon Lundine
- Chemistry Department, Hunter College and Biochemistry Program, The Graduate Center, The City University of New York, New York, NY, United States
| | - Viktoriya Morozova
- Department of Biology and Center for Developmental Neuroscience, College of Staten Island, Graduate Center, The City University of New York, Staten Island, NY, United States
| | - Ali Elhadi Makki
- Department of Biology and Center for Developmental Neuroscience, College of Staten Island, Graduate Center, The City University of New York, Staten Island, NY, United States
| | - Alejandra del C. Alonso
- Department of Biology and Center for Developmental Neuroscience, College of Staten Island, Graduate Center, The City University of New York, Staten Island, NY, United States
| | - Frida E. Kleiman
- Chemistry Department, Hunter College and Biochemistry Program, The Graduate Center, The City University of New York, New York, NY, United States
| |
Collapse
|
11
|
Single nucleotide polymorphisms in piRNA-pathway genes: an insight into genetic determinants of human diseases. Mol Genet Genomics 2019; 295:1-12. [DOI: 10.1007/s00438-019-01612-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Accepted: 09/22/2019] [Indexed: 12/23/2022]
|
12
|
Gao S, Casey AE, Sargeant TJ, Mäkinen VP. Genetic variation within endolysosomal system is associated with late-onset Alzheimer’s disease. Brain 2018; 141:2711-2720. [DOI: 10.1093/brain/awy197] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2018] [Accepted: 06/19/2018] [Indexed: 12/20/2022] Open
Abstract
AbstractLate-onset Alzheimer’s disease is the most common dementia type, yet no treatment exists to stop the neurodegeneration. Evidence from monogenic lysosomal diseases, neuronal pathology and experimental models suggest that autophagic and endolysosomal dysfunction may contribute to neurodegeneration by disrupting the degradation of potentially neurotoxic molecules such as amyloid-β and tau. However, it is uncertain how well the evidence from rare disorders and experimental models capture causal processes in common forms of dementia, including late-onset Alzheimer’s disease. For this reason, we set out to investigate if autophagic and endolysosomal genes were enriched for genetic variants that convey increased risk of Alzheimer’s disease; such a finding would provide population-based support for the endolysosomal hypothesis of neurodegeneration. We quantified the collective genetic associations between the endolysosomal system and Alzheimer’s disease in three genome-wide associations studies (combined n = 62 415). We used the Mergeomics pathway enrichment algorithm that incorporates permutations of the full hierarchical cascade of SNP-gene-pathway to estimate enrichment. We used a previously published collection of 891 autophagic and endolysosomal genes (denoted as AphagEndoLyso, and derived from the Lysoplex sequencing platform) as a proxy for cellular processes related to autophagy, endocytosis and lysosomal function. We also investigated a subset of 142 genes of the 891 that have been implicated in Mendelian diseases (MenDisLyso). We found that both gene sets were enriched for genetic Alzheimer’s associations: an enrichment score 3.67 standard deviations from the null model (P = 0.00012) was detected for AphagEndoLyso, and a score 3.36 standard deviations from the null model (P = 0.00039) was detected for MenDisLyso. The high enrichment score was specific to the AphagEndoLyso gene set (stronger than 99.7% of other tested pathways) and to Alzheimer’s disease (stronger than all other tested diseases). The APOE locus explained most of the MenDisLyso signal (1.16 standard deviations after APOE removal, P = 0.12), but the AphagEndoLyso signal was less affected (3.35 standard deviations after APOE removal, P = 0.00040). Additional sensitivity analyses further indicated that the AphagEndoLyso Gene Set contained an aggregate genetic association that comprised a combination of subtle genetic signals in multiple genes. We also observed an enrichment of Parkinson’s disease signals for MenDisLyso (3.25 standard deviations) and for AphagEndoLyso (3.95 standard deviations from the null model), and a brain-specific pattern of gene expression for AphagEndoLyso in the Gene Tissue Expression Project dataset. These results provide evidence that a diffuse aggregation of genetic perturbations to the autophagy and endolysosomal system may mediate late-onset Alzheimer’s risk in human populations.
Collapse
Affiliation(s)
- Song Gao
- Heart Health Theme, South Australian Health and Medical Research Institute, Adelaide, South Australia
- School of Biological Sciences, University of Adelaide, Adelaide, South Australia
| | - Aaron E Casey
- Heart Health Theme, South Australian Health and Medical Research Institute, Adelaide, South Australia
- School of Biological Sciences, University of Adelaide, Adelaide, South Australia
| | - Tim J Sargeant
- Hopwood Centre for Neurobiology, Nutrition and Metabolism Theme, South Australian Health and Medical Research Institute, Adelaide, South Australia
| | - Ville-Petteri Mäkinen
- Heart Health Theme, South Australian Health and Medical Research Institute, Adelaide, South Australia
- School of Biological Sciences, University of Adelaide, Adelaide, South Australia
- Computational Medicine, Faculty of Medicine, University of Oulu and Biocenter Oulu, Oulu, Finland
| |
Collapse
|
13
|
Huang X, Liu H, Li X, Guan L, Li J, Tellier LCAM, Yang H, Wang J, Zhang J. Revealing Alzheimer's disease genes spectrum in the whole-genome by machine learning. BMC Neurol 2018; 18:5. [PMID: 29320986 PMCID: PMC5763548 DOI: 10.1186/s12883-017-1010-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Accepted: 12/21/2017] [Indexed: 11/23/2022] Open
Abstract
Background Alzheimer’s disease (AD) is an important, progressive neurodegenerative disease, with a complex genetic architecture. A key goal of biomedical research is to seek out disease risk genes, and to elucidate the function of these risk genes in the development of disease. For this purpose, expanding the AD-associated gene set is necessary. In past research, the prediction methods for AD related genes has been limited in their exploration of the target genome regions. We here present a genome-wide method for AD candidate genes predictions. Methods We present a machine learning approach (SVM), based upon integrating gene expression data with human brain-specific gene network data, to discover the full spectrum of AD genes across the whole genome. Results We classified AD candidate genes with an accuracy and the area under the receiver operating characteristic (ROC) curve of 84.56% and 94%. Our approach provides a supplement for the spectrum of AD-associated genes extracted from more than 20,000 genes in a genome wide scale. Conclusions In this study, we have elucidated the whole-genome spectrum of AD, using a machine learning approach. Through this method, we expect for the candidate gene catalogue to provide a more comprehensive annotation of AD for researchers. Electronic supplementary material The online version of this article (10.1186/s12883-017-1010-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Xiaoyan Huang
- BGI Education Center, University of Chinese Academy of Sciences, Shenzhen, 518083, China.,BGI-Shenzhen, Shenzhen, 518083, China.,China National GeneBank, BGI-Shenzhen, Shenzhen, 518120, China
| | - Hankui Liu
- BGI-Shenzhen, Shenzhen, 518083, China.,China National GeneBank, BGI-Shenzhen, Shenzhen, 518120, China
| | - Xinming Li
- College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Liping Guan
- BGI-Shenzhen, Shenzhen, 518083, China.,China National GeneBank, BGI-Shenzhen, Shenzhen, 518120, China
| | - Jiankang Li
- BGI-Shenzhen, Shenzhen, 518083, China.,China National GeneBank, BGI-Shenzhen, Shenzhen, 518120, China
| | - Laurent Christian Asker M Tellier
- BGI-Shenzhen, Shenzhen, 518083, China.,China National GeneBank, BGI-Shenzhen, Shenzhen, 518120, China.,Department of Biology, Bioinformatics, University of Copenhagen, Copenhagen, Denmark
| | - Huanming Yang
- BGI-Shenzhen, Shenzhen, 518083, China.,James D. Watson Institute of Genome Sciences, Hangzhou, 310058, China
| | - Jian Wang
- BGI-Shenzhen, Shenzhen, 518083, China.,James D. Watson Institute of Genome Sciences, Hangzhou, 310058, China
| | - Jianguo Zhang
- BGI-Shenzhen, Shenzhen, 518083, China. .,China National GeneBank, BGI-Shenzhen, Shenzhen, 518120, China. .,Shenzhen Key Lab of Neurogenomics, BGI-Shenzhen, Shenzhen, 518120, China.
| |
Collapse
|
14
|
Perucci LO, Sugimoto MA, Gomes KB, Dusse LM, Teixeira MM, Sousa LP. Annexin A1 and specialized proresolving lipid mediators: promoting resolution as a therapeutic strategy in human inflammatory diseases. Expert Opin Ther Targets 2017; 21:879-896. [PMID: 28786708 DOI: 10.1080/14728222.2017.1364363] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
INTRODUCTION The timely resolution of inflammation is essential to restore tissue homeostasis and to avoid chronic inflammatory diseases. Resolution of inflammation is an active process modulated by various proresolving mediators, including annexin A1 (AnxA1) and specialized proresolving lipid mediators (SPMs), which counteract excessive inflammatory responses and stimulate proresolving mechanisms. Areas covered: The protective effects of AnxA1 and SPMs have been extensively explored in pre-clinical animal models. However, studies investigating the function of these molecules in human diseases are just emerging. This review highlights recent advances on the role of proresolving mediators, and pharmacological opportunities of promoting resolution pathways in preclinical models and patients with various human diseases. Expert opinion: Dysregulation or 'failure' in proresolving mechanisms might be involved in the pathogenesis of chronic inflammatory diseases. Altered levels of proresolving mediators were found in a wide range of human diseases. In some cases, AnxA1 and SPMs are up-regulated in human blood and tissues but fail to engage in proresolving signaling and, hence, to regulate excessive inflammation. Thus, the new concept of 'resolution pharmacology' could be applied to compensate deficiency of endogenous proresolving mediators' generation and/or possible failures in the engagement of resolution pathways observed in many chronic inflammatory diseases.
Collapse
Affiliation(s)
- Luiza Oliveira Perucci
- a Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia , Universidade Federal de Minas Gerais , Belo Horizonte , Minas Gerais , Brazil.,b Programa de Pós-Graduação em Análises Clínicas e Toxicológicas , Universidade Federal de Minas Gerais , Belo Horizonte , Minas Gerais , Brazil
| | - Michelle Amantéa Sugimoto
- a Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia , Universidade Federal de Minas Gerais , Belo Horizonte , Minas Gerais , Brazil.,c Programa de Pós-Graduação em Ciências Farmacêuticas , Universidade Federal de Minas Gerais , Belo Horizonte , Minas Gerais , Brazil
| | - Karina Braga Gomes
- a Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia , Universidade Federal de Minas Gerais , Belo Horizonte , Minas Gerais , Brazil.,b Programa de Pós-Graduação em Análises Clínicas e Toxicológicas , Universidade Federal de Minas Gerais , Belo Horizonte , Minas Gerais , Brazil
| | - Luci Maria Dusse
- a Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia , Universidade Federal de Minas Gerais , Belo Horizonte , Minas Gerais , Brazil.,b Programa de Pós-Graduação em Análises Clínicas e Toxicológicas , Universidade Federal de Minas Gerais , Belo Horizonte , Minas Gerais , Brazil.,c Programa de Pós-Graduação em Ciências Farmacêuticas , Universidade Federal de Minas Gerais , Belo Horizonte , Minas Gerais , Brazil
| | - Mauro Martins Teixeira
- d Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas , Universidade Federal de Minas Gerais , Belo Horizonte , Minas Gerais , Brazil
| | - Lirlândia Pires Sousa
- a Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia , Universidade Federal de Minas Gerais , Belo Horizonte , Minas Gerais , Brazil.,b Programa de Pós-Graduação em Análises Clínicas e Toxicológicas , Universidade Federal de Minas Gerais , Belo Horizonte , Minas Gerais , Brazil.,c Programa de Pós-Graduação em Ciências Farmacêuticas , Universidade Federal de Minas Gerais , Belo Horizonte , Minas Gerais , Brazil
| |
Collapse
|
15
|
Grewal T, Wason SJ, Enrich C, Rentero C. Annexins - insights from knockout mice. Biol Chem 2017; 397:1031-53. [PMID: 27318360 DOI: 10.1515/hsz-2016-0168] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Accepted: 06/14/2016] [Indexed: 12/23/2022]
Abstract
Annexins are a highly conserved protein family that bind to phospholipids in a calcium (Ca2+) - dependent manner. Studies with purified annexins, as well as overexpression and knockdown approaches identified multiple functions predominantly linked to their dynamic and reversible membrane binding behavior. However, most annexins are found at multiple locations and interact with numerous proteins. Furthermore, similar membrane binding characteristics, overlapping localizations and shared interaction partners have complicated identification of their precise functions. To gain insight into annexin function in vivo, mouse models deficient of annexin A1 (AnxA1), A2, A4, A5, A6 and A7 have been generated. Interestingly, with the exception of one study, all mice strains lacking one or even two annexins are viable and develop normally. This suggested redundancy within annexins, but examining these knockout (KO) strains under stress conditions revealed striking phenotypes, identifying underlying mechanisms specific for individual annexins, often supporting Ca2+ homeostasis and membrane transport as central for annexin biology. Conversely, mice lacking AnxA1 or A2 show extracellular functions relevant in health and disease that appear independent of membrane trafficking or Ca2+ signaling. This review will summarize the mechanistic insights gained from studies utilizing mouse models lacking members of the annexin family.
Collapse
|
16
|
McArthur S, Loiola RA, Maggioli E, Errede M, Virgintino D, Solito E. The restorative role of annexin A1 at the blood-brain barrier. Fluids Barriers CNS 2016; 13:17. [PMID: 27655189 PMCID: PMC5031267 DOI: 10.1186/s12987-016-0043-0] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Accepted: 09/12/2016] [Indexed: 12/20/2022] Open
Abstract
Annexin A1 is a potent anti-inflammatory molecule that has been extensively studied in the peripheral immune system, but has not as yet been exploited as a therapeutic target/agent. In the last decade, we have undertaken the study of this molecule in the central nervous system (CNS), focusing particularly on the primary interface between the peripheral body and CNS: the blood-brain barrier. In this review, we provide an overview of the role of this molecule in the brain, with a particular emphasis on its functions in the endothelium of the blood-brain barrier, and the protective actions the molecule may exert in neuroinflammatory, neurovascular and metabolic disease. We focus on the possible new therapeutic avenues opened up by an increased understanding of the role of annexin A1 in the CNS vasculature, and its potential for repairing blood-brain barrier damage in disease and aging.
Collapse
Affiliation(s)
- Simon McArthur
- Department of Biomedical Sciences, Faculty of Science and Technology, University of Westminster, London, UK
| | - Rodrigo Azevedo Loiola
- William Harvey Research Institute, School of Medicine and Dentistry, Queen Mary University, London, UK
| | - Elisa Maggioli
- William Harvey Research Institute, School of Medicine and Dentistry, Queen Mary University, London, UK
| | - Mariella Errede
- Department of Basic Medical Sciences, Neuroscience and Sensory Organs, Bari University School of Medicine, Bari, Italy
| | - Daniela Virgintino
- Department of Basic Medical Sciences, Neuroscience and Sensory Organs, Bari University School of Medicine, Bari, Italy
| | - Egle Solito
- William Harvey Research Institute, School of Medicine and Dentistry, Queen Mary University, London, UK
| |
Collapse
|
17
|
Gao H, Tao Y, He Q, Song F, Saffen D. Functional enrichment analysis of three Alzheimer's disease genome-wide association studies identities DAB1 as a novel candidate liability/protective gene. Biochem Biophys Res Commun 2015; 463:490-5. [PMID: 26028559 DOI: 10.1016/j.bbrc.2015.05.044] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2015] [Accepted: 05/02/2015] [Indexed: 11/17/2022]
Abstract
To explore genetic contributions of Alzheimer's disease (AD) at the level of biological terms and pathways, we analyzed three Caucasian population-based genome-wide association study datasets (TGEN_ND, GeneADA and NIA_LOAD) using the Database for Annotation, Visualization and Integrated Discovery (DAVID). This analysis identified 4 annotation terms ("Fibronectin type III-like fold," "Cell adhesion," "Cell motion" and "Ig-like-C2-type 3") and 17 genes that associated with AD susceptibility in two or more of the GWAS datasets. Ten of these genes, have previously been identified as candidate AD liability genes in genetic association studies (AGT, COL11A1) or encode proteins that function in biological systems or pathways previously implicated in AD (BARHL2, CSF3R, DAB1, HMCN1, LEPR, PTPRF, PXDN, TNR). Among these, DAB1 (Dab, reelin signal transducer, homolog 1) was of particular interest, since it encodes a protein that functions downstream from reelin, a signaling pathway previously identified as protective in AD. Multiple linear regression analysis of correlations between brain DAB1 mRNA expression and SNP genotype using data from the "BrainCloud" database identified five SNPs within the DAB1 locus that correlated with mRNA expression in human dorsolateral prefrontal cortex. Analysis of predicted levels of DAB1 mRNA expression based on genotype combinations present in AD cases and controls vs. the log10-transformed odds ratios for AD diagnosis, revealed statistically significant correlations in one of the GWAS datasets (GenADA), with high DAB1 mRNA expression correlating with AD protection. Multidimensional scaling (MDS) analysis of cases and controls in the three GWAS, revealed genetic differences between GenADA and TGEN_ND/NIA_LOAD, which were similar to each other. To our knowledge, this study is the first to provide genetic evidence for DAB1 as a candidate AD liability/protection gene, although the strength of the contribution of DAB1 may differ among populations.
Collapse
Affiliation(s)
- Hui Gao
- Department of Cellular and Genetic Medicine, School of Basic Medical Sciences, China; Institutes of Brain Science, Fudan University, Shanghai, China
| | - Yu Tao
- Department of Cellular and Genetic Medicine, School of Basic Medical Sciences, China; Institutes of Brain Science, Fudan University, Shanghai, China
| | - Qin He
- Department of Cellular and Genetic Medicine, School of Basic Medical Sciences, China; Institutes of Brain Science, Fudan University, Shanghai, China
| | - Fan Song
- Department of Cellular and Genetic Medicine, School of Basic Medical Sciences, China
| | - David Saffen
- Department of Cellular and Genetic Medicine, School of Basic Medical Sciences, China; Institutes of Brain Science, Fudan University, Shanghai, China; State Key Laboratory for Medical Neurobiology Fudan University, 130 Dong'an Road, Shanghai 200032, China.
| |
Collapse
|