1
|
Wang J, Wang S, Li Q, Liu F, Wan Y, Liang H. Bibliometric and visual analysis of single-cell multiomics in neurodegenerative disease arrest studies. Front Neurol 2024; 15:1450663. [PMID: 39440247 PMCID: PMC11493674 DOI: 10.3389/fneur.2024.1450663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Accepted: 09/26/2024] [Indexed: 10/25/2024] Open
Abstract
Background Neurodegenerative diseases are progressive disorders that severely diminish the quality of life of patients. However, research on neurodegenerative diseases needs to be refined and deepened. Single-cell polyomics is a technique for obtaining transcriptomic, proteomic, and other information from a single cell. In recent years, the heat of single-cell multiomics as an emerging research tool for brain science has gradually increased. Therefore, the aim of this study was to analyze the current status and trends of studies related to the application of single-cell multiomics in neurodegenerative diseases through bibliometrics. Result A total of 596 publications were included in the bibliometric analysis. Between 2015 and 2022, the number of publications increased annually, with the total number of citations increasing significantly, exhibiting the fastest rate of growth between 2019 and 2022. The country/region collaboration map shows that the United States has the most publications and cumulative citations, and that China and the United States have the most collaborations. The institutions that produced the greatest number of articles were Harvard Medical School, Skupin, Alexander, and Wiendl. Among the authors, Heinz had the highest output. Mathys, H accumulated the most citations and was the authoritative author in the field. The journal Nature Communications has published the most literature in this field. A keyword analysis reveals that neurodegenerative diseases and lesions (e.g., Alzheimer's disease, amyloid beta) are the core and foundation of the field. Conversely, single-cell multiomics related research (e.g., single-cell RNA sequencing, bioinformatics) and brain nerve cells (e.g., microglia, astrocytes, neural stem cells) are the hot frontiers of this specialty. Among the references, the article "Single-cell transcriptomic analysis of Alzheimer's disease" is the most frequently cited (1,146 citations), and the article "Cell types in the mouse cortex and hippocampus revealed by single-cell RNA-seq" was the most cited article in the field. Conclusion The objective of this study is to employ bibliometric methods to visualize studies related to single-cell multiomics in neurodegenerative diseases. This will enable us to summarize the current state of research and to reveal key trends and emerging hotspots in the field.
Collapse
Affiliation(s)
- Jieyan Wang
- Department of Urology, People’s Hospital of Longhua, Shenzhen, China
| | - Shuqing Wang
- First Clinical Medical School, Southern Medical University, Guangzhou, China
| | - Qingyu Li
- First Clinical Medical School, Southern Medical University, Guangzhou, China
| | - Fei Liu
- First Clinical Medical School, Southern Medical University, Guangzhou, China
| | - Yantong Wan
- Guangdong Provincial Key Laboratory of Proteomics, Department of Pathophysiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Hui Liang
- Department of Urology, People’s Hospital of Longhua, Shenzhen, China
| |
Collapse
|
2
|
Godoy JA, Mira RG, Inestrosa NC. Intracellular effects of lithium in aging neurons. Ageing Res Rev 2024; 99:102396. [PMID: 38942199 DOI: 10.1016/j.arr.2024.102396] [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: 04/15/2024] [Revised: 06/14/2024] [Accepted: 06/25/2024] [Indexed: 06/30/2024]
Abstract
Lithium therapy received approval during the 1970s, and it has been used for its antidepressant, antimanic, and anti-suicidal effects for acute and long-term prophylaxis and treatment of bipolar disorder (BPD). These properties have been well established; however, the molecular and cellular mechanisms remain controversial. In the past few years, many studies demonstrated that at the cellular level, lithium acts as a regulator of neurogenesis, aging, and Ca2+ homeostasis. At the molecular level, lithium modulates aging by inhibiting glycogen synthase kinase-3β (GSK-3β), and the phosphatidylinositol (PI) cycle; latter, lithium specifically inhibits inositol production, acting as a non-competitive inhibitor of inositol monophosphatase (IMPase). Mitochondria and peroxisome proliferator-activated receptor γ coactivator-1α (PGC-1α) have been related to lithium activity, and its regulation is mediated by GSK-3β degradation and inhibition. Lithium also impacts Ca2+ homeostasis in the mitochondria modulating the function of the lithium-permeable mitochondrial Na+-Ca2+exchanger (NCLX), affecting Ca2+ efflux from the mitochondrial matrix to the endoplasmic reticulum (ER). A close relationship between the protease Omi, GSK-3β, and PGC-1α has also been established. The purpose of this review is to summarize some of the intracellular mechanisms related to lithium activity and how, through them, neuronal aging could be controlled.
Collapse
Affiliation(s)
- Juan A Godoy
- Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Rodrigo G Mira
- Centro de Excelencia en Biomedicina de Magallanes (CEBIMA), Escuela de Medicina, Universidad de Magallanes, Punta Arenas, Chile
| | - Nibaldo C Inestrosa
- Centro de Excelencia en Biomedicina de Magallanes (CEBIMA), Escuela de Medicina, Universidad de Magallanes, Punta Arenas, Chile; Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile.
| |
Collapse
|
3
|
Wareham LK, Baratta RO, Del Buono BJ, Schlumpf E, Calkins DJ. Collagen in the central nervous system: contributions to neurodegeneration and promise as a therapeutic target. Mol Neurodegener 2024; 19:11. [PMID: 38273335 PMCID: PMC10809576 DOI: 10.1186/s13024-024-00704-0] [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: 09/27/2023] [Accepted: 01/10/2024] [Indexed: 01/27/2024] Open
Abstract
The extracellular matrix is a richly bioactive composition of substrates that provides biophysical stability, facilitates intercellular signaling, and both reflects and governs the physiological status of the local microenvironment. The matrix in the central nervous system (CNS) is far from simply an inert scaffold for mechanical support, instead conducting an active role in homeostasis and providing broad capacity for adaptation and remodeling in response to stress that otherwise would challenge equilibrium between neuronal, glial, and vascular elements. A major constituent is collagen, whose characteristic triple helical structure renders mechanical and biochemical stability to enable bidirectional crosstalk between matrix and resident cells. Multiple members of the collagen superfamily are critical to neuronal maturation and circuit formation, axon guidance, and synaptogenesis in the brain. In mature tissue, collagen interacts with other fibrous proteins and glycoproteins to sustain a three-dimensional medium through which complex networks of cells can communicate. While critical for matrix scaffolding, collagen in the CNS is also highly dynamic, with multiple binding sites for partnering matrix proteins, cell-surface receptors, and other ligands. These interactions are emerging as critical mediators of CNS disease and injury, particularly regarding changes in matrix stiffness, astrocyte recruitment and reactivity, and pro-inflammatory signaling in local microenvironments. Changes in the structure and/or deposition of collagen impact cellular signaling and tissue biomechanics in the brain, which in turn can alter cellular responses including antigenicity, angiogenesis, gliosis, and recruitment of immune-related cells. These factors, each involving matrix collagen, contribute to the limited capacity for regeneration of CNS tissue. Emerging therapeutics that attempt to rebuild the matrix using peptide fragments, including collagen-enriched scaffolds and mimetics, hold great potential to promote neural repair and regeneration. Recent evidence from our group and others indicates that repairing protease-degraded collagen helices with mimetic peptides helps restore CNS tissue and promote neuronal survival in a broad spectrum of degenerative conditions. Restoration likely involves bolstering matrix stiffness to reduce the potential for astrocyte reactivity and local inflammation as well as repairing inhibitory binding sites for immune-signaling ligands. Facilitating repair rather than endogenous replacement of collagen degraded by disease or injury may represent the next frontier in developing therapies based on protection, repair, and regeneration of neurons in the central nervous system.
Collapse
Affiliation(s)
- Lauren K Wareham
- Department of Ophthalmology and Visual Sciences, Vanderbilt Eye Institute , Vanderbilt University Medical Center, 1161 21st Avenue S, 37232, Nashville, TN, USA
| | - Robert O Baratta
- Stuart Therapeutics, Inc., 411 SE Osceola St, 34994, Stuart, FL, USA
| | - Brian J Del Buono
- Stuart Therapeutics, Inc., 411 SE Osceola St, 34994, Stuart, FL, USA
| | - Eric Schlumpf
- Stuart Therapeutics, Inc., 411 SE Osceola St, 34994, Stuart, FL, USA
| | - David J Calkins
- Department of Ophthalmology and Visual Sciences, Vanderbilt Eye Institute , Vanderbilt University Medical Center, 1161 21st Avenue S, 37232, Nashville, TN, USA
| |
Collapse
|
4
|
Feng J, Goedegebuure SP, Zeng A, Bi Y, Wang T, Payne P, Ding L, DeNardo D, Hawkins W, Fields RC, Li F. sc2MeNetDrug: A computational tool to uncover inter-cell signaling targets and identify relevant drugs based on single cell RNA-seq data. PLoS Comput Biol 2024; 20:e1011785. [PMID: 38181047 PMCID: PMC10796047 DOI: 10.1371/journal.pcbi.1011785] [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: 03/07/2023] [Revised: 01/18/2024] [Accepted: 12/23/2023] [Indexed: 01/07/2024] Open
Abstract
Single-cell RNA sequencing (scRNA-seq) is a powerful technology to investigate the transcriptional programs in stromal, immune, and disease cells, like tumor cells or neurons within the Alzheimer's Disease (AD) brain or tumor microenvironment (ME) or niche. Cell-cell communications within ME play important roles in disease progression and immunotherapy response and are novel and critical therapeutic targets. Though many tools of scRNA-seq analysis have been developed to investigate the heterogeneity and sub-populations of cells, few were designed for uncovering cell-cell communications of ME and predicting the potentially effective drugs to inhibit the communications. Moreover, the data analysis processes of discovering signaling communication networks and effective drugs using scRNA-seq data are complex and involve a set of critical analysis processes and external supportive data resources, which are difficult for researchers who have no strong computational background and training in scRNA-seq data analysis. To address these challenges, in this study, we developed a novel open-source computational tool, sc2MeNetDrug (https://fuhaililab.github.io/sc2MeNetDrug/). It was specifically designed using scRNA-seq data to identify cell types within disease MEs, uncover the dysfunctional signaling pathways within individual cell types and interactions among different cell types, and predict effective drugs that can potentially disrupt cell-cell signaling communications. sc2MeNetDrug provided a user-friendly graphical user interface to encapsulate the data analysis modules, which can facilitate the scRNA-seq data-based discovery of novel inter-cell signaling communications and novel therapeutic regimens.
Collapse
Affiliation(s)
- Jiarui Feng
- Institute for Informatics (I2), Washington University School of Medicine, Washington University in St. Louis, St. Louis, Missouri, United States of America
- Department of Computer Science and Engineering, Washington University in St. Louis, St. Louis, Missouri, United States of America
| | - S. Peter Goedegebuure
- Department of Surgery, Washington University in St. Louis, St. Louis, Missouri, United States of America
- Siteman Cancer Center, Washington University in St. Louis, St. Louis, Missouri, United States of America
| | - Amanda Zeng
- Department of Computer Science and Engineering, Washington University in St. Louis, St. Louis, Missouri, United States of America
| | - Ye Bi
- Department of Surgery, Washington University in St. Louis, St. Louis, Missouri, United States of America
| | - Ting Wang
- Department of Genetics, Washington University in St. Louis, St. Louis, Missouri, United States of America
- McDonnell Genome Institute, Washington University in St. Louis, St. Louis, Missouri, United States of America
| | - Philip Payne
- Institute for Informatics (I2), Washington University School of Medicine, Washington University in St. Louis, St. Louis, Missouri, United States of America
| | - Li Ding
- Siteman Cancer Center, Washington University in St. Louis, St. Louis, Missouri, United States of America
- Department of Genetics, Washington University in St. Louis, St. Louis, Missouri, United States of America
- McDonnell Genome Institute, Washington University in St. Louis, St. Louis, Missouri, United States of America
- Department of Medicine, Washington University in St. Louis, St. Louis, Missouri, United States of America
| | - David DeNardo
- Department of Surgery, Washington University in St. Louis, St. Louis, Missouri, United States of America
- Siteman Cancer Center, Washington University in St. Louis, St. Louis, Missouri, United States of America
| | - William Hawkins
- Department of Surgery, Washington University in St. Louis, St. Louis, Missouri, United States of America
- Siteman Cancer Center, Washington University in St. Louis, St. Louis, Missouri, United States of America
| | - Ryan C. Fields
- Department of Surgery, Washington University in St. Louis, St. Louis, Missouri, United States of America
- Siteman Cancer Center, Washington University in St. Louis, St. Louis, Missouri, United States of America
| | - Fuhai Li
- Institute for Informatics (I2), Washington University School of Medicine, Washington University in St. Louis, St. Louis, Missouri, United States of America
- Department of Pediatrics, Washington University School of Medicine, Washington University in St. Louis, St. Louis, Missouri, United States of America
| |
Collapse
|
5
|
Jongsma E, Goyala A, Mateos JM, Ewald CY. Removal of extracellular human amyloid beta aggregates by extracellular proteases in C. elegans. eLife 2023; 12:e83465. [PMID: 37728486 PMCID: PMC10541181 DOI: 10.7554/elife.83465] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Accepted: 09/19/2023] [Indexed: 09/21/2023] Open
Abstract
The amyloid beta (Aβ) plaques found in Alzheimer's disease (AD) patients' brains contain collagens and are embedded extracellularly. Several collagens have been proposed to influence Aβ aggregate formation, yet their role in clearance is unknown. To investigate the potential role of collagens in forming and clearance of extracellular aggregates in vivo, we created a transgenic Caenorhabditis elegans strain that expresses and secretes human Aβ1-42. This secreted Aβ forms aggregates in two distinct places within the extracellular matrix. In a screen for extracellular human Aβ aggregation regulators, we identified different collagens to ameliorate or potentiate Aβ aggregation. We show that a disintegrin and metalloprotease a disintegrin and metalloprotease 2 (ADM-2), an ortholog of ADAM9, reduces the load of extracellular Aβ aggregates. ADM-2 is required and sufficient to remove the extracellular Aβ aggregates. Thus, we provide in vivo evidence of collagens essential for aggregate formation and metalloprotease participating in extracellular Aβ aggregate removal.
Collapse
Affiliation(s)
- Elisabeth Jongsma
- Laboratory of Extracellular Matrix Regeneration, Institute of Translational Medicine, Department of Health Sciences and Technology, ETH ZürichSchwerzenbachSwitzerland
| | - Anita Goyala
- Laboratory of Extracellular Matrix Regeneration, Institute of Translational Medicine, Department of Health Sciences and Technology, ETH ZürichSchwerzenbachSwitzerland
| | - José Maria Mateos
- Center for Microscopy and Image Analysis, University of ZurichZurichSwitzerland
| | - Collin Yvès Ewald
- Laboratory of Extracellular Matrix Regeneration, Institute of Translational Medicine, Department of Health Sciences and Technology, ETH ZürichSchwerzenbachSwitzerland
| |
Collapse
|
6
|
Stevenson M, Varghese R, Hebron ML, Liu X, Ratliff N, Smith A, Turner RS, Moussa C. Inhibition of discoidin domain receptor (DDR)-1 with nilotinib alters CSF miRNAs and is associated with reduced inflammation and vascular fibrosis in Alzheimer's disease. J Neuroinflammation 2023; 20:116. [PMID: 37194065 PMCID: PMC10186647 DOI: 10.1186/s12974-023-02802-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Accepted: 05/10/2023] [Indexed: 05/18/2023] Open
Abstract
Discoidin Domain Receptor (DDR)-1 is activated by collagen. Nilotinib is a tyrosine kinase inhibitor that is FDA-approved for leukemia and potently inhibits DDR-1. Individuals diagnosed with mild-moderate Alzheimer's disease (AD) treated with nilotinib (versus placebo) for 12 months showed reduction of amyloid plaque and cerebrospinal fluid (CSF) amyloid, and attenuation of hippocampal volume loss. However, the mechanisms are unclear. Here, we explored unbiased next generation whole genome miRNA sequencing from AD patients CSF and miRNAs were matched with their corresponding mRNAs using gene ontology. Changes in CSF miRNAs were confirmed via measurement of CSF DDR1 activity and plasma levels of AD biomarkers. Approximately 1050 miRNAs are detected in the CSF but only 17 miRNAs are specifically altered between baseline and 12-month treatment with nilotinib versus placebo. Treatment with nilotinib significantly reduces collagen and DDR1 gene expression (upregulated in AD brain), in association with inhibition of CSF DDR1. Pro-inflammatory cytokines, including interleukins and chemokines are reduced along with caspase-3 gene expression. Specific genes that indicate vascular fibrosis, e.g., collagen, Transforming Growth Factors (TGFs) and Tissue Inhibitors of Metalloproteases (TIMPs) are altered by DDR1 inhibition with nilotinib. Specific changes in vesicular transport, including the neurotransmitters dopamine and acetylcholine, and autophagy genes, including ATGs, indicate facilitation of autophagic flux and cellular trafficking. Inhibition of DDR1 with nilotinib may be a safe and effective adjunct treatment strategy involving an oral drug that enters the CNS and adequately engages its target. DDR1 inhibition with nilotinib exhibits multi-modal effects not only on amyloid and tau clearance but also on anti-inflammatory markers that may reduce cerebrovascular fibrosis.
Collapse
Affiliation(s)
- Max Stevenson
- Translational Neurotherapeutics Program, Laboratory for Dementia and Parkinsonism, Department of Neurology, Georgetown University Medical Center, Building D, Room 265, 4000 Reservoir Rd, NW, Washington, DC, 20057, USA
| | - Rency Varghese
- Genomics and Epigenomics Shared Resource, Department of Oncology, Georgetown University Medical Center, Building D, 4000 Reservoir Rd, NW, Washington, DC, 20057, USA
| | - Michaeline L Hebron
- Translational Neurotherapeutics Program, Laboratory for Dementia and Parkinsonism, Department of Neurology, Georgetown University Medical Center, Building D, Room 265, 4000 Reservoir Rd, NW, Washington, DC, 20057, USA
| | - Xiaoguang Liu
- Translational Neurotherapeutics Program, Laboratory for Dementia and Parkinsonism, Department of Neurology, Georgetown University Medical Center, Building D, Room 265, 4000 Reservoir Rd, NW, Washington, DC, 20057, USA
| | - Nick Ratliff
- Translational Neurotherapeutics Program, Laboratory for Dementia and Parkinsonism, Department of Neurology, Georgetown University Medical Center, Building D, Room 265, 4000 Reservoir Rd, NW, Washington, DC, 20057, USA
| | - Amelia Smith
- Translational Neurotherapeutics Program, Laboratory for Dementia and Parkinsonism, Department of Neurology, Georgetown University Medical Center, Building D, Room 265, 4000 Reservoir Rd, NW, Washington, DC, 20057, USA
| | - R Scott Turner
- Memory Disorders Program, Department of Neurology, Georgetown University Medical Center, 4000 Reservoir Rd, NW, Washington, DC, 20057, USA
| | - Charbel Moussa
- Translational Neurotherapeutics Program, Laboratory for Dementia and Parkinsonism, Department of Neurology, Georgetown University Medical Center, Building D, Room 265, 4000 Reservoir Rd, NW, Washington, DC, 20057, USA.
| |
Collapse
|
7
|
The 90 plus: longevity and COVID-19 survival. Mol Psychiatry 2022; 27:1936-1944. [PMID: 35136227 DOI: 10.1038/s41380-022-01461-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 01/11/2022] [Accepted: 01/20/2022] [Indexed: 11/08/2022]
Abstract
The world population is getting older and studies aiming to enhance our comprehension of the underlying mechanisms responsible for health span are of utmost interest for longevity and as a measure for health care. In this review, we summarized previous genetic association studies (GWAS) and next-generation sequencing (NGS) of elderly cohorts. We also present the updated hypothesis for the aging process, together with the factors associated with healthy aging. We discuss the relevance of studying older individuals and build databanks to characterize the presence and resistance against late-onset disorders. The identification of about 2 million novel variants in our cohort of more than 1000 elderly Brazilians illustrates the importance of studying highly admixed populations of non-European ancestry. Finally, the ascertainment of nonagenarians and particularly of centenarians who were recovered from COVID-19 or remained asymptomatic opens new avenues of research aiming to enhance our comprehension of biological mechanisms associated with resistance against pathogens.
Collapse
|
8
|
Knoedler JR, Inoue S, Bayless DW, Yang T, Tantry A, Davis CH, Leung NY, Parthasarathy S, Wang G, Alvarado M, Rizvi AH, Fenno LE, Ramakrishnan C, Deisseroth K, Shah NM. A functional cellular framework for sex and estrous cycle-dependent gene expression and behavior. Cell 2022; 185:654-671.e22. [PMID: 35065713 PMCID: PMC8956134 DOI: 10.1016/j.cell.2021.12.031] [Citation(s) in RCA: 65] [Impact Index Per Article: 32.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 11/22/2021] [Accepted: 12/20/2021] [Indexed: 01/05/2023]
Abstract
Sex hormones exert a profound influence on gendered behaviors. How individual sex hormone-responsive neuronal populations regulate diverse sex-typical behaviors is unclear. We performed orthogonal, genetically targeted sequencing of four estrogen receptor 1-expressing (Esr1+) populations and identified 1,415 genes expressed differentially between sexes or estrous states. Unique subsets of these genes were distributed across all 137 transcriptomically defined Esr1+ cell types, including estrous stage-specific ones, that comprise the four populations. We used differentially expressed genes labeling single Esr1+ cell types as entry points to functionally characterize two such cell types, BNSTprTac1/Esr1 and VMHvlCckar/Esr1. We observed that these two cell types, but not the other Esr1+ cell types in these populations, are essential for sex recognition in males and mating in females, respectively. Furthermore, VMHvlCckar/Esr1 cell type projections are distinct from those of other VMHvlEsr1 cell types. Together, projection and functional specialization of dimorphic cell types enables sex hormone-responsive populations to regulate diverse social behaviors.
Collapse
Affiliation(s)
- Joseph R Knoedler
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305, USA
| | - Sayaka Inoue
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305, USA
| | - Daniel W Bayless
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305, USA
| | - Taehong Yang
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305, USA
| | - Adarsh Tantry
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305, USA
| | - Chung-Ha Davis
- Neurosciences Graduate Program, Stanford University, Stanford, CA 94305, USA
| | - Nicole Y Leung
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305, USA
| | | | - Grace Wang
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305, USA
| | - Maricruz Alvarado
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305, USA
| | - Abbas H Rizvi
- Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY 10027, USA
| | - Lief E Fenno
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305, USA
| | | | - Karl Deisseroth
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305, USA; Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - Nirao M Shah
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305, USA; Department of Neurobiology, Stanford University, Stanford, CA 94305, USA.
| |
Collapse
|
9
|
Neuronal Phenotype of col4a1 and col25a1: An Intriguing Hypothesis in Vertebrates Brain Aging. Int J Mol Sci 2022; 23:ijms23031778. [PMID: 35163698 PMCID: PMC8836537 DOI: 10.3390/ijms23031778] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 01/25/2022] [Accepted: 01/27/2022] [Indexed: 12/11/2022] Open
Abstract
Collagens are the most abundant proteins in vertebrates and constitute the major components of the extracellular matrix. Collagens play an important and multifaceted role in the development and functioning of the nervous system and undergo structural remodeling and quantitative modifications during aging. Here, we investigated the age-dependent regulation of col4a1 and col25a1 in the brain of the short-lived vertebrate Nothobranchius furzeri, a powerful model organism for aging research due to its natural fast-aging process and further characterized typical hallmarks of brain aging in this species. We showed that col4a1 and col25a1 are relatively well conserved during vertebrate evolution, and their expression significantly increases in the brain of N. furzeri upon aging. Noteworthy, we report that both col4a1 and col25a1 are expressed in cells with a neuronal phenotype, unlike what has already been documented in mammalian brain, in which only col25a1 is considered a neuronal marker, whereas col4a1 seems to be expressed only in endothelial cells. Overall, our findings encourage further investigation on the role of col4a1 and col25a1 in the biology of the vertebrate brain as well as the onset of aging and neurodegenerative diseases.
Collapse
|
10
|
Martens DS, Thijs L, Latosinska A, Trenson S, Siwy J, Zhang ZY, Wang C, Beige J, Vlahou A, Janssens S, Mischak H, Nawrot TS, Staessen JA. Urinary peptidomic profiles to address age-related disabilities: a prospective population study. THE LANCET. HEALTHY LONGEVITY 2021; 2:e690-e703. [PMID: 34766101 PMCID: PMC8566278 DOI: 10.1016/s2666-7568(21)00226-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND The Global Burden of Diseases, Injuries, and Risk Factors Study 2019 called for innovation in addressing age-related disabilities. Our study aimed to identify and validate a urinary peptidomic profile (UPP) differentiating healthy from unhealthy ageing in the general population, to test the UPP predictor in independent patient cohorts, and to search for targetable molecular pathways underlying age-related chronic diseases. METHODS In this prospective population study, we used data from participants in the Flemish Study on Environment, Genes and Health Outcomes (FLEMENGHO), done in northern Belgium from 1985 to 2019, and invited participants to a follow-up examination in 2005-10. Participants were eligible if their address was within 15 km of the examination centre and if they had not withdrawn consent in any of the previous examination cycles (1985-2004). All participants (2005-10) were also invited to an additional follow-up examination in 2009-13. Participants who took part in both the 2005-10 follow-up examination and in the additional 2009-13 follow-up visit constituted the derivation dataset, which included their 2005-10 data, and the time-shifted internal validation dataset, which included their 2009-13 data. The remaining participants who only had 2005-10 data constituted the synchronous internal validation dataset. Participants were excluded from analyses if they were incapacitated, had not undergone UPP, or had either missing or outlying (three SDs greater than the mean of all consenting participants) values of body-mass index, plasma glucose, or serum creatinine. The UPP was assessed by capillary electrophoresis coupled with mass spectrometry. The multidimensional UPP signature reflecting ageing was generated from the derivation dataset and validated in the time-shifted internal validation dataset and the synchronous validation dataset. It was further validated in patients with diabetes, COVID-19, or chronic kidney disease (CKD). In FLEMENGHO, the mortality endpoints were all-cause, cardiovascular, and non-cardiovascular mortality; other endpoints were fatal or non-fatal cancer and musculoskeletal disorders. Molecular pathway exploration was done using the Reactome and Kyoto Encyclopedia of Genes and Genomes databases. FINDINGS 778 individuals (395 [51%] women and 383 [49%] men; aged 16·2-82·1 years; mean age 50·9 years [SD 15·8]) from the FLEMENGHO cohort had a follow-up examination between 2005 and 2010, of whom 559 participants had a further follow-up from Oct 28, 2009, to March 19, 2013, and made up the derivation (2005-10) and time-shifted internal validation (2009-13) datasets. 219 were examined once and constituted the synchronous internal validation dataset (2005-10). With correction for multiple testing and multivariable adjustment, chronological age was associated with 210 sequenced peptides mainly showing downregulation of collagen fragments. The trained model relating chronological age to UPP, derived by elastic net regression, included 54 peptides from 17 proteins. The UPP-age prediction model explained 76·3% (r=0·87) of chronological age in the derivation dataset, 54·4% (r=0·74) in the time-shifted validation dataset, and 65·3% (r=0·81) in the synchronous internal validation dataset. Compared with chronological age, the predicted UPP-age was greater in patients with diabetes (chronological age 50·8 years [SE 0·37] vs UPP-age 56·9 years [0·30]), COVID‑19 (53·2 years [1·80] vs 58·5 years [1·67]), or CKD (54·6 years [0·97] vs 62·3 years [0·85]; all p<0·0001). In the FLEMENGHO cohort, independent of chronological age, UPP-age was significantly associated with various risk markers related to cardiovascular, metabolic, and renal disease, inflammation, and medication use. Over a median of 12·4 years (IQR 10·8-13·2), total mortality, cardiovascular mortality, and osteoporosis in the population was associated with UPP-age independent of chronological age, with hazard ratios per 10 year increase in UPP-age of 1·54 (95% CI 1·22-1·95) for total mortality, 1·72 (1·20-2·47) for cardiovascular mortality, and 1·40 (1·06-1·85) for osteoporosis and fractures. The most relevant molecular pathways informed by the proteins involved deregulation of collagen biology and extracellular matrix maintenance. INTERPRETATION The UPP signature indicative of ageing reflects fibrosis and extracellular matrix remodelling and was associated with risk factors and adverse health outcomes in the population and with accelerated ageing in patients. Innovation in addressing disability should shift focus from the ontology of diseases to shared disease mechanisms, in particular ageing-related fibrotic degeneration. FUNDING European Research Council, Ministry of the Flemish Community, OMRON Healthcare.
Collapse
Affiliation(s)
- Dries S Martens
- Centre for Environmental Sciences, Hasselt University, Hasselt, Belgium
| | - Lutgarde Thijs
- Research Unit Hypertension and Cardiovascular Epidemiology, KU Leuven Department of Cardiovascular Diseases, University of Leuven, Leuven, Belgium
| | | | - Sander Trenson
- Division of Cardiology, Sint-Jan Hospital, Bruges, Belgium
| | | | - Zhen-Yu Zhang
- Research Unit Hypertension and Cardiovascular Epidemiology, KU Leuven Department of Cardiovascular Diseases, University of Leuven, Leuven, Belgium
| | - Congrong Wang
- Centre for Environmental Sciences, Hasselt University, Hasselt, Belgium
| | - Joachim Beige
- Martin Luther University of Halle-Wittenberg, Halle (Saale), Germany
| | - Antonia Vlahou
- Systems Biology Center, Biomedical Research Foundation, Academy of Athens, Athens, Greece
| | - Stefan Janssens
- Division of Cardiology, University Hospitals Leuven, Leuven, Belgium
| | - Harald Mischak
- Mosaiques-Diagnostics, Hannover, Germany
- Institute of Cardiovascular and Medical Sciences, Glasgow, UK
| | - Tim S Nawrot
- Centre for Environmental Sciences, Hasselt University, Hasselt, Belgium
- Research Unit Environment and Health, Department of Public Health and Primary Care, University of Leuven, Leuven, Belgium
| | - Jan A Staessen
- Biomedical Sciences Group, Faculty of Medicine, University of Leuven, Leuven, Belgium
- Alliance for the Promotion of Preventive Medicine, Mechelen, Belgium
| |
Collapse
|
11
|
Borden EA, Furey M, Gattone NJ, Hambardikar VD, Liang XH, Scoma ER, Abou Samra A, D-Gary LR, Dennis DJ, Fricker D, Garcia C, Jiang Z, Khan SA, Kumarasamy D, Kuppala H, Ringrose S, Rosenheim EJ, Van Exel K, Vudhayagiri HS, Zhang J, Zhang Z, Guitart-Mampel M, Urquiza P, Solesio ME. Is there a link between inorganic polyphosphate (polyP), mitochondria, and neurodegeneration? Pharmacol Res 2021; 163:105211. [PMID: 33010423 PMCID: PMC7855267 DOI: 10.1016/j.phrs.2020.105211] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 09/08/2020] [Accepted: 09/15/2020] [Indexed: 12/14/2022]
Abstract
Mitochondrial dysfunction - including increased apoptosis, calcium and protein dyshomeostasis within the organelle, and dysfunctional bioenergetics and oxidative status - is a common, early feature in all the major neurodegenerative diseases, including Alzheimer's Disease (AD) and Parkinson's Disease (PD). However, the exact molecular mechanisms that drive the organelle to dysfunction and ultimately to failure in these conditions are still not well described. Different authors have shown that inorganic polyphosphate (polyP), an ancient and well-conserved molecule, plays a key role in the regulation of mitochondrial physiology under basal conditions. PolyP, which is present in all studied organisms, is composed of chains of orthophosphates linked together by highly energetic phosphoanhydride bonds, similar to those found in ATP. This polymer shows a ubiquitous distribution, even if a high co-localization with mitochondria has been reported. It has been proposed that polyP might be an alternative to ATP for cellular energy storage in different organisms, as well as the implication of polyP in the regulation of many of the mitochondrial processes affected in AD and PD, including protein and calcium homeostasis. Here, we conduct a comprehensive review and discussion of the bibliography available regarding the role of polyP in the mitochondrial dysfunction present in AD and PD. Taking into account the data presented in this review, we postulate that polyP could be a valid, innovative and, plausible pharmacological target against mitochondrial dysfunction in AD and PD. However, further research should be conducted to better understand the exact role of polyP in neurodegeneration, as well as the metabolism of the polymer, and the effect of different lengths of polyP on cellular and mitochondrial physiology.
Collapse
Affiliation(s)
- Emily A Borden
- Department of Biology, College of Arts and Sciences, Rutgers University, NJ, USA
| | - Matthew Furey
- Department of Biology, College of Arts and Sciences, Rutgers University, NJ, USA
| | - Nicholas J Gattone
- Department of Biology, College of Arts and Sciences, Rutgers University, NJ, USA
| | | | - Xiao Hua Liang
- Department of Biology, College of Arts and Sciences, Rutgers University, NJ, USA
| | - Ernest R Scoma
- Department of Biology, College of Arts and Sciences, Rutgers University, NJ, USA
| | - Antonella Abou Samra
- Department of Biology, College of Arts and Sciences, Rutgers University, NJ, USA
| | - LaKeshia R D-Gary
- Department of Biology, College of Arts and Sciences, Rutgers University, NJ, USA
| | - Dayshaun J Dennis
- Department of Biology, College of Arts and Sciences, Rutgers University, NJ, USA
| | - Daniel Fricker
- Department of Biology, College of Arts and Sciences, Rutgers University, NJ, USA
| | - Cindy Garcia
- Department of Biology, College of Arts and Sciences, Rutgers University, NJ, USA
| | - ZeCheng Jiang
- Department of Biology, College of Arts and Sciences, Rutgers University, NJ, USA
| | - Shariq A Khan
- Department of Biology, College of Arts and Sciences, Rutgers University, NJ, USA
| | | | - Hasmitha Kuppala
- Department of Biology, College of Arts and Sciences, Rutgers University, NJ, USA
| | - Savannah Ringrose
- Department of Biology, College of Arts and Sciences, Rutgers University, NJ, USA
| | - Evan J Rosenheim
- Department of Biology, College of Arts and Sciences, Rutgers University, NJ, USA
| | - Kimberly Van Exel
- Center for Computational and Integrative Biology, Rutgers University, NJ, USA
| | | | - Jiarui Zhang
- Center for Computational and Integrative Biology, Rutgers University, NJ, USA
| | - Zhaowen Zhang
- Department of Biology, College of Arts and Sciences, Rutgers University, NJ, USA
| | | | - Pedro Urquiza
- Department of Biology, College of Arts and Sciences, Rutgers University, NJ, USA
| | - Maria E Solesio
- Department of Biology, College of Arts and Sciences, Rutgers University, NJ, USA; Center for Computational and Integrative Biology, Rutgers University, NJ, USA.
| |
Collapse
|
12
|
Hashimoto T, Fujii D, Naka Y, Kashiwagi-Hakozaki M, Matsuo Y, Matsuura Y, Wakabayashi T, Iwatsubo T. Collagenous Alzheimer amyloid plaque component impacts on the compaction of amyloid-β plaques. Acta Neuropathol Commun 2020; 8:212. [PMID: 33287899 PMCID: PMC7720522 DOI: 10.1186/s40478-020-01075-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Accepted: 11/11/2020] [Indexed: 12/31/2022] Open
Abstract
Massive deposition of amyloid β peptides (Aβ) as senile plaques (SP) characterizes the brain pathology of Alzheimer’s disease (AD). SPs exhibit a variety of morphologies, although little is known about the SP components that determine their morphology. Collagenous Alzheimer amyloid plaque component (CLAC) is one of the major non-Aβ proteinaceous components of SP amyloid in AD brains. Here we show that overexpression of CLAC precursor (CLAC-P) in the brains of APP transgenic mice results in a significant remodeling of amyloid pathology, i.e., reduction in diffuse-type amyloid plaques and an increase in compact plaques laden with thioflavin S-positive amyloid cores. In vivo microdialysis revealed a significant decrease in Aβ in the brain interstitial fluid of CLAC-P/APP double transgenic mice compared with APP transgenic mice. These findings implicate CLAC in the compaction of Aβ in amyloid plaques and the brain dynamics of Aβ.
Collapse
|
13
|
Romero-Ortuno R, Kenny RA, McManus R. Collagens and elastin genetic variations and their potential role in aging-related diseases and longevity in humans. Exp Gerontol 2019; 129:110781. [PMID: 31740390 DOI: 10.1016/j.exger.2019.110781] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2019] [Revised: 11/11/2019] [Accepted: 11/12/2019] [Indexed: 12/17/2022]
Abstract
Collagens and elastin are 'building blocks' of tissues and extracellular matrix. Mutations in these proteins cause severe congenital syndromes. Adverse genetic variations may accelerate the aging process in adults contributing to premature morbidity, disability and/or mortality. Favorable variants may contribute to longevity and/or healthy aging, but this is much less studied. We reviewed the association between variation in the genes of collagens and elastin and premature aging, accelerated aging, age-related diseases and/or frailty; and the association between genetic variation in those and longevity and/or healthy aging in humans. A systematic search was conducted in MEDLINE and other online databases (OMIM, Genetics Home Reference, Orphanet, ClinVar). Results suggest that genetic variants lead to aging phenotypes of known congenital disease, but also to association with common age-related diseases in adults without known congenital disease. This may be due to the variable penetrance and expressivity of many variants. Some collagen variants have been associated with longevity or healthy aging. A limitation is that most studies had <1000 participants and their criterion for statistical significance was p < 0.05. Results highlight the importance of adopting a lifecourse approach to the study of the genomics of aging. Gerontology can help with new methodologies that operationalize biological aging.
Collapse
Affiliation(s)
- Roman Romero-Ortuno
- Trinity College Dublin, Discipline of Medical Gerontology, Mercer's Institute for Successful Ageing, St James's Hospital, Dublin 8, Ireland; The Irish Longitudinal Study on Ageing (TILDA), Trinity College Dublin, Dublin, Ireland..
| | - Rose Anne Kenny
- Trinity College Dublin, Discipline of Medical Gerontology, Mercer's Institute for Successful Ageing, St James's Hospital, Dublin 8, Ireland; The Irish Longitudinal Study on Ageing (TILDA), Trinity College Dublin, Dublin, Ireland
| | - Ross McManus
- Trinity College Dublin, Trinity Translational Medicine Institute, Trinity Centre for Health Sciences, St James's Hospital, Dublin 8, Ireland
| |
Collapse
|
14
|
Exploring the roles of MACIT and multiplexin collagens in stem cells and cancer. Semin Cancer Biol 2019; 62:134-148. [PMID: 31479735 DOI: 10.1016/j.semcancer.2019.08.033] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Revised: 08/20/2019] [Accepted: 08/30/2019] [Indexed: 02/07/2023]
Abstract
The extracellular matrix (ECM) is ubiquitously involved in neoplastic transformation, tumour growth and metastatic dissemination, and the interplay between tumour and stromal cells and the ECM is now considered crucial for the formation of a tumour-supporting microenvironment. The 28 different collagens (Col) form a major ECM protein family and display extraordinary functional diversity in tissue homeostasis as well as in pathological conditions, with functions ranging from structural support for tissues to regulatory binding activities and storage of biologically active cryptic domains releasable through ECM proteolysis. Two subfamilies of collagens, namely the plasma membrane-associated collagens with interrupted triple-helices (MACITs, including ColXIII, ColXXIII and ColXXV) and the basement membrane-associated collagens with multiple triple-helix domains with interruptions (multiplexins, including ColXV and ColXVIII), have highly interesting regulatory functions in tissue and organ development, as well as in various diseases, including cancer. An increasing, albeit yet sparse, data suggest that these collagens play crucial roles in conveying regulatory signals from the extracellular space to cells. We summarize here the current knowledge about MACITs and multiplexins as regulators of stemness and oncogenic processes, as well as their roles in influencing cell fate decisions in healthy and cancerous tissues. In addition, we present a bioinformatic analysis of the impacts of MACITs and multiplexins transcript levels on the prognosis of patients representing a wide array of malignant diseases, to aid future diagnostic and therapeutic efforts.
Collapse
|
15
|
Aberrant Expression of Collagen Gene Family in the Brain Regions of Male Mice with Behavioral Psychopathologies Induced by Chronic Agonistic Interactions. BIOMED RESEARCH INTERNATIONAL 2019; 2019:7276389. [PMID: 31183373 PMCID: PMC6512038 DOI: 10.1155/2019/7276389] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/13/2018] [Revised: 03/04/2019] [Accepted: 03/27/2019] [Indexed: 11/17/2022]
Abstract
Chronic agonistic interactions promote the development of experimental psychopathologies in animals: a depression-like state in chronically defeated mice and the pathology of aggressive behavior in the mice with repeated wins. The abundant research data indicate that such psychopathological states are associated with significant molecular and cellular changes in the brain. This paper aims to study the influence of a 20-day period of agonistic interactions on the expression patterns of collagen family genes encoding the proteins which are basic components of extracellular matrix (ECM) in different brain regions of mice using the RNA-Seq database. Most of differentially expressed collagen genes were shown to be upregulated in the hypothalamus and striatum of chronically aggressive and defeated mice and in the hippocampus of defeated mice, whereas downregulation of collagen genes was demonstrated in the ventral tegmental areas in both experimental groups. Aberrant expression of collagen genes induced by chronic agonistic interactions may be indicative of specific ECM defects in the brain regions of mice with alternative social experience. This is the first study demonstrating remodeling of ECM under the development of experimental disorders.
Collapse
|
16
|
Kelley AR, Perry G, Bach SBH. Characterization of Proteins Present in Isolated Senile Plaques from Alzheimer's Diseased Brains by MALDI-TOF MS with MS/MS. ACS Chem Neurosci 2018; 9:708-714. [PMID: 29301075 DOI: 10.1021/acschemneuro.7b00445] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
The increase of insoluble senile plaques in the brain is a primary hallmark of Alzheimer's disease. The usefulness of matrix-assisted laser desorption/ionization time-of-flight mass spectrometry (MALDI-TOF MS) with tandem MS for the characterization of senile plaques from AD brains and the relevance of the components identified to furthering AD research using MS is discussed. Thirty-three components were reproducibly observed within tryptic aliquots of senile plaques from two different AD brains after sample preparation optimization. Additionally, this is one of the first accounts of LIFT being utilized for the direct sequencing of peptides from isolated senile plaques. While many of the species observed coisolated within senile plaques have been linked to AD etiology, if only speculatively, this is the first instance that many of them have been demonstrated to be a part of the plaques themselves. This work is the first step in determining the potential roles that the species may have in the aggregation or proliferation of the plaques.
Collapse
Affiliation(s)
- Andrea R. Kelley
- Department of Chemistry, University of Texas at San Antonio, One UTSA Circle, San Antonio, Texas 78249, United States
| | - George Perry
- College of Sciences, University of Texas at San Antonio, One UTSA Circle, San Antonio, Texas 78249, United States
| | - Stephan B. H. Bach
- Department of Chemistry, University of Texas at San Antonio, One UTSA Circle, San Antonio, Texas 78249, United States
| |
Collapse
|
17
|
Giri M, Shah A, Upreti B, Rai JC. Unraveling the genes implicated in Alzheimer's disease. Biomed Rep 2017; 7:105-114. [PMID: 28781776 DOI: 10.3892/br.2017.927] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Accepted: 05/29/2017] [Indexed: 12/17/2022] Open
Abstract
Alzheimer's disease (AD) is a heterogeneous neurodegenerative disorder and it is the most common form of dementia in the elderly. Early onset AD is caused by mutations in three genes: Amyloid-β precursor protein, presenilin 1 (PSEN1) and PSEN2. Late onset AD (LOAD) is complex and apolipoprotein E is the only unanimously accepted genetic risk factor for its development. Various genes implicated in AD have been identified using advanced genetic technologies, however, there are many additional genes that remain unidentified. The present review highlights the genetics of early and LOAD and summarizes the genes involved in different signaling pathways. This may provide insight into neurodegenerative disease research and will facilitate the development of effective strategies to combat AD.
Collapse
Affiliation(s)
- Mohan Giri
- National Center for Rheumatic Diseases, Ratopul, Kathmandu 44600, Nepal
| | - Abhilasha Shah
- National Center for Rheumatic Diseases, Ratopul, Kathmandu 44600, Nepal
| | - Bibhuti Upreti
- National Center for Rheumatic Diseases, Ratopul, Kathmandu 44600, Nepal
| | | |
Collapse
|
18
|
Matveev SV, Spielmann HP, Metts BM, Chen J, Onono F, Zhu H, Scheff SW, Walker LC, LeVine H. A distinct subfraction of Aβ is responsible for the high-affinity Pittsburgh compound B-binding site in Alzheimer's disease brain. J Neurochem 2014; 131:356-68. [PMID: 24995708 DOI: 10.1111/jnc.12815] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2014] [Revised: 06/18/2014] [Accepted: 06/29/2014] [Indexed: 12/27/2022]
Abstract
The positron emission tomography (PET) ligand (11) C-labeled Pittsburgh compound B (PIB) is used to image β-amyloid (Aβ) deposits in the brains of living subjects with the intent of detecting early stages of Alzheimer's disease (AD). However, deposits of human-sequence Aβ in amyloid precursor protein transgenic mice and non-human primates bind very little PIB. The high stoichiometry of PIB:Aβ binding in human AD suggests that the PIB-binding site may represent a particularly pathogenic entity and/or report local pathologic conditions. In this study, (3) H-PIB was employed to track purification of the PIB-binding site in > 90% yield from frontal cortical tissue of autopsy-diagnosed AD subjects. The purified PIB-binding site comprises a distinct, highly insoluble subfraction of the Aβ in AD brain with low buoyant density because of the sodium dodecyl sulfate-resistant association with a limited subset of brain proteins and lipids with physical properties similar to lipid rafts and to a ganglioside:Aβ complex in AD and Down syndrome brain. Both the protein and lipid components are required for PIB binding. Elucidation of human-specific biological components and pathways will be important in guiding improvement of the animal models for AD and in identifying new potential therapeutic avenues. A lipid-associated subpopulation of Aβ accounts for the high-affinity binding of Pittsburgh compound B (PIB) in Alzheimer's disease brain. Mass spectrometry of the isolated PIB-binding site from frontal cortex identified Aβ peptides and a set of plaque-associated proteins in AD but not age-matched normal brain. The PIB-binding site may represent a particularly pathogenic entity and/or report local pathologic conditions.
Collapse
Affiliation(s)
- Sergey V Matveev
- Center on Aging, University of Kentucky, Lexington, Kentucky, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Abstract
Chemical synapses allow neurons to perform complex computations and regulate other systems of the body. At a chemical synapse, pre- and postsynaptic sites are separated by a small space (the synaptic cleft) and surrounded by astrocytes. The basement membrane (BM), a sheetlike, specialized extracellular matrix (ECM), is found ubiquitously in the PNS. It has become clear that the ECMs not only play a structural role but also serve as barriers and filters in the PNS and CNS. Moreover, proteoglycans and tenascin family proteins in the ECM regulate synapse formation and synaptic plasticity. Although CNS synapses lack the BMs, recent results indicate that the BM-associated collagens are also present in the CNS synaptic cleft and affect synaptogenesis in both the CNS and the PNS. The C1q domain-containing family proteins are important components of the CNS synaptic cleft in regulating synapse formation, maintenance, and the pruning process. The ECM is regarded as a crucial component of the tetrapartite synapse, consisting of pre- and postsynaptic neurons, astrocyte, and ECM.
Collapse
Affiliation(s)
- Anne Heikkinen
- Oulu Center for Cell-Matrix Research, Biocenter Oulu and Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland
| | - Taina Pihlajaniemi
- Oulu Center for Cell-Matrix Research, Biocenter Oulu and Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland
| | - Andreas Faissner
- Department of Cell Morphology and Molecular Neurobiology, Ruhr-University, Bochum, Germany
| | - Michisuke Yuzaki
- Department of Physiology, School of Medicine, Keio University, Tokyo, Japan.
| |
Collapse
|
20
|
Parmar AS, Nunes AM, Baum J, Brodsky B. A peptide study of the relationship between the collagen triple-helix and amyloid. Biopolymers 2012; 97:795-806. [PMID: 22806499 DOI: 10.1002/bip.22070] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Type XXV collagen, or collagen-like amyloidogenic component, is a component of amyloid plaques, and recent studies suggest this collagen affects amyloid fibril elongation and has a genetic association with Alzheimer's disease. The relationship between the collagen triple helix and amyloid fibrils was investigated by studying peptide models, including a very stable triple helical peptide (Pro-Hyp-Gly)₁₀ , an amyloidogenic peptide GNNQQNY, and a hybrid peptide where the GNNQQNY sequence was incorporated between (GPO)(n) domains. Circular dichroism and nuclear magnetic resonance (NMR) spectroscopy showed the GNNQQNY peptide formed a random coil structure, whereas the hybrid peptide contained a central disordered GNNQQNY region transitioning to triple-helical ends. Light scattering confirmed the GNNQQNY peptide had a high propensity to form amyloid fibrils, whereas amyloidogenesis was delayed in the hybrid peptide. NMR data suggested the triple-helix constraints on the GNNQQNY sequence within the hybrid peptide may disfavor the conformational change necessary for aggregation. Independent addition of a triple-helical peptide to the GNNQQNY peptide under aggregating conditions delayed nucleation and amyloid fibril growth. The inhibition of amyloid nucleation depended on the Gly-Xaa-Yaa sequence and required the triple-helix conformation. The inhibitory effect of the collagen triple-helix on an amyloidogenic sequence, when in the same molecule or when added separately, suggests Type XXV collagen, and possibly other collagens, may play a role in regulating amyloid fibril formation.
Collapse
Affiliation(s)
- Avanish S Parmar
- Department of Biomedical Engineering, Tufts University, Medford, MA, USA
| | | | | | | |
Collapse
|
21
|
Li D, Zhao H, Kranzler HR, Oslin D, Anton RF, Farrer LA, Gelernter J. Association of COL25A1 with comorbid antisocial personality disorder and substance dependence. Biol Psychiatry 2012; 71:733-40. [PMID: 22297151 PMCID: PMC3548659 DOI: 10.1016/j.biopsych.2011.12.011] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2011] [Revised: 12/12/2011] [Accepted: 12/13/2011] [Indexed: 10/14/2022]
Abstract
BACKGROUND Antisocial personality disorder (ASPD) is a psychiatric disorder characterized by a long-term pattern of manipulating, exploiting, or violating the rights of others. METHODS Subjects ascertained for genetic studies of substance dependence (SD) and diagnosed with ASPD and comorbid SD were included in a two-stage genetic association study. In the discovery stage, 627 single nucleotide polymorphisms (SNPs) located in 179 candidate genes for addiction were analyzed in a case-control cohort and family-based cohort. The significant findings were replicated in an independent case-control cohort. RESULTS One SNP, rs13134663, in the collagen XXV alpha 1 gene (COL25A1) was significantly associated with ASPD in both African Americans and European Americans (smallest p values were .0002 and .0004, respectively). There was also evidence of association with the same SNP in independent samples of African American and European American cases and control subjects (p = .035 and .033, respectively). Analysis of the combined set of case-control subjects yielded an allelic p value of 9 × 10(-6) with odds ratio (95% confidence interval) of 1.3 (1.16, 1.47) (smallest p = 1 × 10(-7); Bonferroni threshold p = .00012). CONCLUSIONS The COL25A1 gene, located at chromosome 4q25, encodes the collagen-like Alzheimer amyloid plaque component precursor, a type II transmembrane protein specifically expressed in neurons; it co-localizes with amyloid β in senile plaques in Alzheimer disease brains. This SNP maps to the transcription factor binding site and is conserved in 17 vertebrates, including mice and rats. Our findings suggest that COL25A1 may be associated with ASPD, especially in the context of SD.
Collapse
Affiliation(s)
- Dawei Li
- Department of Psychiatry, School of Medicine, Yale University, New Haven, Connecticut
| | - Hongyu Zhao
- Department of Epidemiology and Public Health, School of Medicine, Yale University, New Haven, Connecticut,Department of Genetics, School of Medicine, Yale University, New Haven, Connecticut
| | - Henry R. Kranzler
- Department of Psychiatry, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania,VISN 4 MIRECC, Philadelphia VAMC, Philadelphia, Pennsylvania
| | - David Oslin
- Department of Psychiatry, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania,VISN 4 MIRECC, Philadelphia VAMC, Philadelphia, Pennsylvania
| | - Raymond F Anton
- Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, South Carolina
| | - Lindsay A Farrer
- Departments of Medicine (Biomedical Genetics), Neurology, Ophthalmology, Genetics & Genomics, Biostatistics, and Epidemiology, Boston University Schools of Medicine and Public Health, Boston, Massachusetts
| | - Joel Gelernter
- Department of Psychiatry, School of Medicine, Yale University, New Haven, Connecticut,Department of Genetics, School of Medicine, Yale University, New Haven, Connecticut,VA Connecticut Healthcare Center, West Haven, Connecticut,Department of Neurobiology, Yale University School of Medicine, New Haven, Connecticut,To whom correspondence should be addressed: Joel Gelernter, Department of Psychiatry, Yale University School of Medicine, VA CT Healthcare Center, 950 Campbell Avenue, 116A2, West Haven, CT 06516.
| |
Collapse
|
22
|
Wexler EM, Rosen E, Lu D, Osborn GE, Martin E, Raybould H, Geschwind DH. Genome-wide analysis of a Wnt1-regulated transcriptional network implicates neurodegenerative pathways. Sci Signal 2012; 4:ra65. [PMID: 21971039 DOI: 10.1126/scisignal.2002282] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Wnt proteins are critical to mammalian brain development and function. The canonical Wnt signaling pathway involves the stabilization and nuclear translocation of β-catenin; however, Wnt also signals through alternative, noncanonical pathways. To gain a systems-level, genome-wide view of Wnt signaling, we analyzed Wnt1-stimulated changes in gene expression by transcriptional microarray analysis in cultured human neural progenitor (hNP) cells at multiple time points over a 72-hour time course. We observed a widespread oscillatory-like pattern of changes in gene expression, involving components of both the canonical and the noncanonical Wnt signaling pathways. A higher-order, systems-level analysis that combined independent component analysis, waveform analysis, and mutual information-based network construction revealed effects on pathways related to cell death and neurodegenerative disease. Wnt effectors were tightly clustered with presenilin1 (PSEN1) and granulin (GRN), which cause dominantly inherited forms of Alzheimer's disease and frontotemporal dementia (FTD), respectively. We further explored a potential link between Wnt1 and GRN and found that Wnt1 decreased GRN expression by hNPs. Conversely, GRN knockdown increased WNT1 expression, demonstrating that Wnt and GRN reciprocally regulate each other. Finally, we provided in vivo validation of the in vitro findings by analyzing gene expression data from individuals with FTD. These unbiased and genome-wide analyses provide evidence for a connection between Wnt signaling and the transcriptional regulation of neurodegenerative disease genes.
Collapse
Affiliation(s)
- Eric M Wexler
- Department of Psychiatry, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA 90024, USA.
| | | | | | | | | | | | | |
Collapse
|
23
|
A role for collagen XXIII in cancer cell adhesion, anchorage-independence and metastasis. Oncogene 2011; 31:2362-72. [PMID: 21963851 DOI: 10.1038/onc.2011.406] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Collagen XXIII is a transmembrane collagen previously shown to be upregulated in metastatic prostate cancer that has been used as a tissue and fluid biomarker for non-small cell lung cancer and prostate cancer. To determine whether collagen XXIII facilitates cancer cell metastasis in vivo and to establish a function for collagen XXIII in cancer progression, collagen XXIII knockdown cells were examined for alterations in in vivo metastasis as well as in vitro cell adhesion. In experimental and spontaneous xenograft models of metastasis, H460 cells expressing collagen XXIII shRNA formed fewer lung metastases than control cells. Loss of collagen XXIII in H460 cells also impaired cell adhesion, anchorage-independent growth and cell seeding to the lung, but did not affect cell proliferation. Corroborating a role for collagen XXIII in cell adhesion, overexpression of collagen XXIII in H1299 cells, which do not express endogenous collagen XXIII, enhanced cell adhesion. Consequent reduction in OB-cadherin, alpha-catenin, gamma-catenin, beta-catenin, vimentin and galectin-3 protein expression was also observed in response to loss of collagen XXIII. This study suggests a potential role for collagen XXIII in mediating metastasis by facilitating cell-cell and cell-matrix adhesion as well as anchorage-independent cell growth.
Collapse
|
24
|
Wang KS, Liu X, Zhang Q, Aragam N, Pan Y. Genome-wide association analysis of age at onset in schizophrenia in a European-American sample. Am J Med Genet B Neuropsychiatr Genet 2011; 156B:671-80. [PMID: 21688384 DOI: 10.1002/ajmg.b.31209] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2010] [Accepted: 05/26/2011] [Indexed: 12/19/2022]
Abstract
We performed a genome-wide association analysis to identify genetic variants influencing age at onset (AAO) and examine gene × gender interactions for AAO in schizophrenia (SCZ) using a European-American sample (1,162 cases). Linear regression model in PLINK was used to test for associations with AAO while the GxE option was chosen to test for the influence of gene × gender interactions. The most significant association with AAO was observed with SNP rs7819815 (P = 3.10×10(-7)) at 8q24.22. The next best signal was at 4q25 in COL25A1 gene (rs17039583, P = 4.30×10(-6)) and the third region was at 4p16.1 (rs17407555, P = 4.56×10(-6) , near RAF1P1, and rs4697924, P = 1.23×10(-5) within WDR1 gene). Conditional analysis on chromosome 4 indicated that 4p16.1 and 4q25 loci were independent. Furthermore, 2 SNPs (rs16834822 and rs16834824) at 1q43 in RYR2 showed strong associations in the female sample (P = 2.10×10(-6) and 2.33×10(-6) , respectively) and strong gene × gender interactions in influencing AAO (P = 9.23×10(-7) and 1.15×10(-6) , respectively) while the second best region showing gene × gender interaction was at 7q22.3 (rs179863, P = 2.33×10(-6) ). Using an independent sample of 1,068 cases, we could not replicate the associations for above top SNPs; however, we found nominal significance associations for their flanking SNPs (P < 0.05). These findings provide evidence of several genetic variants influencing AAO of SCZ.
Collapse
Affiliation(s)
- Ke-Sheng Wang
- Department of Biostatistics and Epidemiology, College of Public Health, East Tennessee State University, Johnson City, 37614-1700, USA.
| | | | | | | | | |
Collapse
|
25
|
Abstract
Collagens are the most abundant proteins in mammals. The collagen family comprises 28 members that contain at least one triple-helical domain. Collagens are deposited in the extracellular matrix where most of them form supramolecular assemblies. Four collagens are type II membrane proteins that also exist in a soluble form released from the cell surface by shedding. Collagens play structural roles and contribute to mechanical properties, organization, and shape of tissues. They interact with cells via several receptor families and regulate their proliferation, migration, and differentiation. Some collagens have a restricted tissue distribution and hence specific biological functions.
Collapse
Affiliation(s)
- Sylvie Ricard-Blum
- Institut de Biologie et Chimie des Protéines, UMR 5086 CNRS, Université Lyon 1, Lyon, 69367, France.
| |
Collapse
|
26
|
Vandenbroeck K, Urcelay E, Comabella M. IFN-beta pharmacogenomics in multiple sclerosis. Pharmacogenomics 2010; 11:1137-48. [PMID: 20712530 DOI: 10.2217/pgs.10.108] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Multiple sclerosis (MS) is a condition of the CNS marked by inflammation and neurodegeneration. Interferon (IFN)-beta was the first, and still is the main, immunomodulatory treatment for MS. Its clinical efficacy is limited, and a proportion of patients, ranging between 20-55%, do not respond to the therapy. Identification and subsequently, implementation in the clinic of biomarkers predictive for individual therapeutic response would facilitate improved patient care in addition to ensuring a more rational provision of this therapy. In this article, we summarize the main findings from studies addressing the pharmacogenomics of clinical response to IFN-beta in MS by either whole-genome association scans, candidate gene or transcriptomics studies. Whole-genome DNA association screens have revealed a high representation of brain-specific genes, and have hinted toward both extracellular ligand-gated ion channels and type I IFNs pathway genes as important categories of genetic IFN-beta response modifiers. One hit, glypican 5 (GPC5), was recently replicated in an independent study of IFN-beta responsiveness. Recent RNA transcriptomics studies have revealed the occurrence of a pre-existing type I IFN gene-expression signature, composed of genes that are predominantly induced by type I IFNs, as a potential contributing feature of poor response to therapy. Thus, while the outlines of a complex polygenic mechanism are gradually being uncovered, the main challenges for the near future will reside in the robust validation of identified response-modifying genes as well as in the decipherment of the mechanistic relationships between these genes and clinical response to IFN-beta.
Collapse
Affiliation(s)
- Koen Vandenbroeck
- Neurogenomiks Group, Universidad del País Vasco (UPV/EHU), Leioa, Spain.
| | | | | |
Collapse
|
27
|
Tong Y, Xu Y, Scearce-Levie K, Ptácek LJ, Fu YH. COL25A1 triggers and promotes Alzheimer's disease-like pathology in vivo. Neurogenetics 2009; 11:41-52. [PMID: 19548013 PMCID: PMC2807601 DOI: 10.1007/s10048-009-0201-5] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2009] [Accepted: 05/28/2009] [Indexed: 12/19/2022]
Abstract
Collagen XXV alpha 1 (COL25A1) is a collagenous type II transmembrane protein purified from senile plaques of Alzheimer's disease (AD) brains. COL25A1 alleles have been associated with increased risk for AD in a Swedish population. COL25A1 is specifically expressed in neurons and binds to aggregated Abeta in vitro. However, its contribution to the pathogenesis of AD and in vivo function are unknown. Here, we report that over-expression of COL25A1 in transgenic mice increases p35/p25 and beta-site APP-cleaving enzyme 1 (BACE1) levels, facilitates intracellular aggregation and extracellular matrix deposits of Abeta, and causes synaptophysin loss and astrocyte activation. COL25A1 mice displayed reduced anxiety-like behavior in elevated plus maze and open field tests and significantly slower swimming speed in Morris water maze. In stable cell lines, motifs in noncollagenous domains of COL25A1 were important for the induction of BACE1 expression. These findings demonstrate that COL25A1 leads to AD-like pathology in vivo. Modulation of COL25A1 function may represent an alternative therapeutic intervention for AD.
Collapse
Affiliation(s)
- Ying Tong
- Department of Neurology, University of California San Francisco, 1550 Fourth Street, Rock Hall Rm548, San Francisco, CA, 94158, USA
| | | | | | | | | |
Collapse
|