1
|
Almasoudi SH, Al-Kuraishy HM, Al-Gareeb AI, Eliwa D, Alexiou A, Papadakis M, Batiha GES. Role of mitogen-activated protein kinase inhibitors in Alzheimer's disease: Rouge of brain kinases. Brain Res Bull 2025; 224:111296. [PMID: 40073950 DOI: 10.1016/j.brainresbull.2025.111296] [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: 10/08/2024] [Revised: 03/03/2025] [Accepted: 03/07/2025] [Indexed: 03/14/2025]
Abstract
Alzheimer's disease (AD) is the chief cause of dementia and related mortality worldwide due to progressive accumulation of amyloid peptide (Aβ) and hyperphosphorylated tau protein. These neuropathological changes lead to cognitive impairment and memory dysfunction. Notably, most Food drug Administration (FDA) approved anti-AD medications such as tacrine and donepezil are engaged with symptomatic relief of cognitive impairment but do not reverse the underlying AD neuropathology. Therefore, searching for new anti-AD is advisable. It has been shown that the inflammatory signaling pathways such as mitogen-activated protein kinases (MAPK) are intricate with the Aβ and tau protein neuropathology in AD. In addition, inhibition of brain MAPK plays a critical role in mitigating cognitive dysfunction in early-onset AD. Though, the fundamental mechanisms for the beneficial effects of MAPK inhibitors were not fully explained. Therefore, this review aims to discuss the potential molecular mechanisms of MAPK inhibitors in AD.
Collapse
Affiliation(s)
- Suad Hamdan Almasoudi
- Department of Biology, College of Sciences, Umm Al-Qura University, Makkah 21955, Saudi Arabia.
| | - Hayder M Al-Kuraishy
- Department Of Clinical Pharmacology and Medicine, College Of Medicine, Mustansiriyah University, P.O. Box 14132, Baghdad, Iraq.
| | - Ali I Al-Gareeb
- Head of Jabir ibn Hayyan Medical University, P.O.Box13 Kufa, Al-Ameer Qu, Najaf, Iraq.
| | - Duaa Eliwa
- Department of Pharmacognosy, Faculty of Pharmacy, Tanta University, Tanta, Egypt.
| | - Athanasios Alexiou
- Department of Science and Engineering, Novel Global Community Educational Foundation, Hebersham, NSW 2770, Australia; AFNP Med, Wien 1030, Austria
| | - Marios Papadakis
- University Hospital Witten-Herdecke, Heusnerstrasse 40, University of Witten-Herdecke, Wuppertal 42283, Germany.
| | - Gaber El-Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhour University, Damanhour, AlBeheira 22511, Egypt.
| |
Collapse
|
2
|
Qu H, Liu Y, Connolly JJ, Mentch FD, Kao C, Hakonarson H. Risk of Alzheimer's disease in Down syndrome: Insights gained by multi-omics. Alzheimers Dement 2025; 21:e14604. [PMID: 40207399 PMCID: PMC11982707 DOI: 10.1002/alz.14604] [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: 07/16/2024] [Revised: 01/14/2025] [Accepted: 01/16/2025] [Indexed: 04/11/2025]
Abstract
Individuals with Down syndrome (DS) are highly susceptible to Alzheimer's disease (AD). The integration of genomics, transcriptomics, epigenomics, proteomics, and metabolomics enables unprecedented understanding of DS-AD, offering a detailed picture of this complex issue. The vast -omics data also present challenges that reflect the complexity of genetic information flow. These studies nonetheless reveal critical mechanisms behind AD risk, including unique observations in DS that differ from those seen in the general population and familial dominant AD. In addition, the correlations between the AD polygenic risk score and proteins related to female infertility and autoimmune thyroiditis corroborate clinical observations. Metabolomic data reveal disrupted metabolic networks, offering prospects for a dynamic score to create specialized nutritional interventions. By adopting a multidimensional perspective with integrated reductionism, the evolving landscape presents an opportunity to identify promising directions for developing precision strategies to mitigate the impact of AD in the DS population. HIGHLIGHTS: Individuals with Down syndrome (DS) are highly susceptible to Alzheimer's disease (AD). DS-AD is characterized by its polygenic nature, extending beyond chromosome 21 with significant contributions from various chromosomes. DS-AD also presents unique features that differ from those observed in the general population and familial dominant AD. Our review consolidates key findings from genomics, transcriptomics, epigenomics, proteomics, and metabolomics, providing a comprehensive view of the molecular mechanisms underlying DS-AD. We highlight promising research directions to further elucidate the pathogenesis of DS-AD.
Collapse
Affiliation(s)
- Hui‐Qi Qu
- The Center for Applied GenomicsChildren's Hospital of PhiladelphiaPhiladelphiaPennsylvaniaUSA
| | - Yichuan Liu
- The Center for Applied GenomicsChildren's Hospital of PhiladelphiaPhiladelphiaPennsylvaniaUSA
| | - John J. Connolly
- The Center for Applied GenomicsChildren's Hospital of PhiladelphiaPhiladelphiaPennsylvaniaUSA
| | - Frank D. Mentch
- The Center for Applied GenomicsChildren's Hospital of PhiladelphiaPhiladelphiaPennsylvaniaUSA
| | - Charlly Kao
- The Center for Applied GenomicsChildren's Hospital of PhiladelphiaPhiladelphiaPennsylvaniaUSA
| | - Hakon Hakonarson
- The Center for Applied GenomicsChildren's Hospital of PhiladelphiaPhiladelphiaPennsylvaniaUSA
- Department of Pediatrics, The Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- Division of Human GeneticsChildren's Hospital of PhiladelphiaPhiladelphiaPennsylvaniaUSA
- Division of Pulmonary MedicineChildren's Hospital of PhiladelphiaPhiladelphiaPennsylvaniaUSA
- Faculty of MedicineUniversity of IcelandReykjavikIceland
| |
Collapse
|
3
|
Sim MA, Doecke JD, Liew OW, Wong LL, Tan ESJ, Chan SP, Chong JRF, Cai Y, Hilal S, Venketasubramanian N, Tan BY, Alzheimer's Disease Neuroimaging Initiative, Lai MKP, Choi H, Masters CL, Richards AM, Chen CLH. Plasma proteomics for cognitive decline and dementia-A Southeast Asian cohort study. Alzheimers Dement 2025; 21:e14577. [PMID: 39998981 PMCID: PMC11854348 DOI: 10.1002/alz.14577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 12/27/2024] [Accepted: 01/12/2025] [Indexed: 02/27/2025]
Abstract
INTRODUCTION The prognostic utility of plasma proteomics for cognitive decline and dementia in a Southeast Asian population characterized by high cerebrovascular disease (CeVD) burden is underexplored. METHODS We examined this in a Singaporean memory clinic cohort of 528 subjects (n = 300, CeVD; n = 167, incident cognitive decline) followed-up for 4 years. RESULTS Of 1441 plasma proteins surveyed, a 12-protein signature significantly predicted cognitive decline (q-value < .05). Sixteen diverse biological processes were implicated in cognitive decline. Ten proteins independently predicted incident dementia (q-value < .05). A unified prediction model combining plasma proteins with clinical risk factors increased the area under the curve for outcome prediction from 0.62 to 0.85. External validation in the cerebrospinal fluid proteome of an independent Caucasian cohort replicated four of the significantly predictive plasma markers for cognitive decline namely: GFAP, NEFL, AREG, and PPY. DISCUSSION The prognostic proteins prioritized in our study provide robust signals in two different biological matrices, representing potential mechanistic targets for dementia and cognitive decline. HIGHLIGHTS A total of 1441 plasma proteins were profiled in a Singaporean memory clinic cohort. We report prognostic plasma protein signatures for cognitive decline and dementia. External validation was performed in the cerebrospinal fluid proteome of a Caucasian cohort. A concordant proteomic signature was identified across both biofluids and cohorts. Further studies are needed to explore the therapeutic implications of these proteins for dementia.
Collapse
Affiliation(s)
- Ming Ann Sim
- Departments of Pharmacology and Psychological Medicine, Memory Aging and Cognition CentreNational University of SingaporeSingaporeSingapore
- Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
- Department of AnesthesiaNational University Health SystemSingaporeSingapore
| | - James D. Doecke
- Australian E‐Health Research CentreCSIROHerstonQueenslandAustralia
| | - Oi Wah Liew
- Department of CardiologyNational University Heart CentreSingaporeSingapore
- Cardiovascular Research InstituteNational University of SingaporeSingaporeSingapore
| | - Lee Lee Wong
- Department of CardiologyNational University Heart CentreSingaporeSingapore
- Cardiovascular Research InstituteNational University of SingaporeSingaporeSingapore
| | - Eugene S. J. Tan
- Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
- Department of CardiologyNational University Heart CentreSingaporeSingapore
| | - Siew Pang Chan
- Department of CardiologyNational University Heart CentreSingaporeSingapore
- Cardiovascular Research InstituteNational University of SingaporeSingaporeSingapore
| | - Joyce R. F. Chong
- Departments of Pharmacology and Psychological Medicine, Memory Aging and Cognition CentreNational University of SingaporeSingaporeSingapore
| | - Yuan Cai
- Department of Medicine and Therapeutics, Faculty of MedicineDivision of NeurologyThe Chinese University of Hong KongMa Liu ShuiHong Kong
| | - Saima Hilal
- Saw Swee Hock School of Public HealthNational University of Singapore and National University Health SystemSingaporeSingapore
| | | | - Boon Yeow Tan
- Department of MedicineSt Luke's HospitalSingaporeSingapore
| | | | - Mitchell K. P Lai
- Departments of Pharmacology and Psychological Medicine, Memory Aging and Cognition CentreNational University of SingaporeSingaporeSingapore
| | - Hyungwon Choi
- Cardiovascular Research InstituteNational University of SingaporeSingaporeSingapore
- Department of MedicineYong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
- Singapore Lipidomics IncubatorLife Sciences InstituteNational University of SingaporeSingaporeSingapore
| | - Colin L. Masters
- The Florey Institute of Neuroscience and Mental HealthThe University of MelbourneParkvilleVictoriaAustralia
| | - Arthur Mark Richards
- Department of CardiologyNational University Heart CentreSingaporeSingapore
- Cardiovascular Research InstituteNational University of SingaporeSingaporeSingapore
- Christchurch Heart InstituteUniversity of OtagoDunedinNew Zealand
| | - Christopher L. H. Chen
- Departments of Pharmacology and Psychological Medicine, Memory Aging and Cognition CentreNational University of SingaporeSingaporeSingapore
| |
Collapse
|
4
|
Talab MJ, Valizadeh A, Tahershamsi Z, Housaindokht MR, Ranjbar B. Personalized biocorona as disease biomarker: The challenges and opportunities. Biochim Biophys Acta Gen Subj 2024; 1868:130724. [PMID: 39426758 DOI: 10.1016/j.bbagen.2024.130724] [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: 06/07/2024] [Revised: 09/22/2024] [Accepted: 10/15/2024] [Indexed: 10/21/2024]
Abstract
It is well known that when nanoparticles interact with biological fluids, a layer of proteins and biological components forms on them. This layer may alter the biological fate and efficiency of the nanomaterial. Recent studies have shown that illness states have a major impact on the structure of the biocorona, sometimes referred to as the "personalized protein corona." Physiological factors like illness, which impact the proteome and metabolome pattern and result in conformational changes in proteins, give rise to this structure of discrimination in biocorona decoration. Improving the efficiency of precise platforms for developing new molecular biomarkers for accurate illness diagnosis is vitally necessary. The biocorona pattern's discrimination may be a diagnostic tool for designing biosensors. As a result, in this review, we summarize the most current studies on the relationship between physiological conditions and the variety of biocorona patterns that influence the biological responses of nanosystems. The biocorona pattern's flexibility may provide new research directions and be utilized to create nanoparticle-based therapeutic and diagnostic products suited to certain physiological situations.
Collapse
Affiliation(s)
- Mahtab Jahanshah Talab
- Department of Biophysics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Ali Valizadeh
- Department of Biophysics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Zahra Tahershamsi
- Department of Biophysics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Mohammad Reza Housaindokht
- Biophysical Chemistry Laboratory, Department of Chemistry, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran.
| | - Bijan Ranjbar
- Department of Biophysics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran.
| |
Collapse
|
5
|
Sita G, Graziosi A, Corrieri C, Ghelli L, Angelini S, Cortelli P, Hrelia P, Morroni F. The Unfolded Protein Response in a Murine Model of Alzheimer's Disease: Looking for Predictors. Int J Mol Sci 2023; 24:16200. [PMID: 38003389 PMCID: PMC10671834 DOI: 10.3390/ijms242216200] [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: 09/25/2023] [Revised: 11/03/2023] [Accepted: 11/09/2023] [Indexed: 11/26/2023] Open
Abstract
Alzheimer's disease (AD) represents the most frequent type of dementia worldwide, and aging is the most important risk factor for the sporadic form of the pathology. The endoplasmic reticulum (ER), the main cellular actor involved in proteostasis, appears significantly compromised in AD due to the accumulation of the β-amyloid (Aβ) protein and the phosphorylated Tau protein. Increasing protein misfolding activates a specific cellular response known as Unfolded Protein Response (UPR), which orchestrates the recovery of ER function. The aim of the present study was to investigate the role of UPR in a murine model of AD induced by intracerebroventricular (i.c.v.) injection of Aβ1-42 oligomers at 3 or 18 months. The oligomer injection in aged animals induced memory impairment, oxidative stress, and the depletion of glutathione reserve. Furthermore, the RNA sequencing and the bioinformatic analysis performed showed the enrichment of several pathways involved in neurodegeneration and protein regulations. The analysis highlighted the significant dysregulation of the protein kinase RNA-like ER kinase (PERK), inositol-requiring protein 1α (IRE1α) and activating transcription factor 6 (ATF-6). In turn, ER stress affected the PI3K/Akt/Gsk3β and MAPK/ERK pathways, highlighting Mapkapk5 as a potential marker, whose regulation could lead to the definition of new pharmacological and neuroprotective strategies to counteract AD.
Collapse
Affiliation(s)
- Giulia Sita
- Department of Pharmacy and BioTechnology—FaBiT, Alma Mater Studiorum University of Bologna, Via Irnerio 48, 40126 Bologna, Italy; (G.S.); (A.G.); (C.C.); (L.G.); (S.A.); (F.M.)
| | - Agnese Graziosi
- Department of Pharmacy and BioTechnology—FaBiT, Alma Mater Studiorum University of Bologna, Via Irnerio 48, 40126 Bologna, Italy; (G.S.); (A.G.); (C.C.); (L.G.); (S.A.); (F.M.)
| | - Camilla Corrieri
- Department of Pharmacy and BioTechnology—FaBiT, Alma Mater Studiorum University of Bologna, Via Irnerio 48, 40126 Bologna, Italy; (G.S.); (A.G.); (C.C.); (L.G.); (S.A.); (F.M.)
| | - Luca Ghelli
- Department of Pharmacy and BioTechnology—FaBiT, Alma Mater Studiorum University of Bologna, Via Irnerio 48, 40126 Bologna, Italy; (G.S.); (A.G.); (C.C.); (L.G.); (S.A.); (F.M.)
| | - Sabrina Angelini
- Department of Pharmacy and BioTechnology—FaBiT, Alma Mater Studiorum University of Bologna, Via Irnerio 48, 40126 Bologna, Italy; (G.S.); (A.G.); (C.C.); (L.G.); (S.A.); (F.M.)
| | - Pietro Cortelli
- Department of Biomedical and NeuroMotor Sciences—DiBiNeM, Alma Mater Studiorum University of Bologna, Via Altura 3, 40139 Bologna, Italy;
| | - Patrizia Hrelia
- Department of Pharmacy and BioTechnology—FaBiT, Alma Mater Studiorum University of Bologna, Via Irnerio 48, 40126 Bologna, Italy; (G.S.); (A.G.); (C.C.); (L.G.); (S.A.); (F.M.)
| | - Fabiana Morroni
- Department of Pharmacy and BioTechnology—FaBiT, Alma Mater Studiorum University of Bologna, Via Irnerio 48, 40126 Bologna, Italy; (G.S.); (A.G.); (C.C.); (L.G.); (S.A.); (F.M.)
| |
Collapse
|
6
|
Maroofian R, Efthymiou S, Suri M, Rahman F, Zaki MS, Maqbool S, Anwa N, Ruiz-Pérez VL, Yanovsky-Dagan S, Elpeleg O, Sudhakar S, Mankad K, Harel T, Houlden H. Consolidating the association of biallelic MAPKAPK5 pathogenic variants with a distinct syndromic neurodevelopmental disorder. J Med Genet 2023; 60:791-796. [PMID: 36581449 PMCID: PMC10423509 DOI: 10.1136/jmg-2022-108566] [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: 03/15/2022] [Accepted: 12/12/2022] [Indexed: 12/31/2022]
Abstract
BACKGROUND MAPK-activated protein kinase 5 (MAPKAPK5) is an essential enzyme for diverse cellular processes. Dysregulation of the pathways regulated by MAPKAPK enzymes can lead to the development of variable diseases. Recently, homozygous loss-of-function variants in MAPKAPK5 were reported in four patients from three families presenting with a recognisable neurodevelopmental disorder, so-called 'neurocardiofaciodigital' syndrome. OBJECTIVE AND METHODS In order to improve characterisation of the clinical features associated with biallelic MAPKAPK5 variants, we employed a genotype-first approach combined with reverse deep-phenotyping of three affected individuals. RESULTS In the present study, we identified biallelic loss-of-function and missense MAPKAPK5 variants in three unrelated individuals from consanguineous families. All affected individuals exhibited a syndromic neurodevelopmental disorder characterised by severe global developmental delay, intellectual disability, characteristic facial morphology, brachycephaly, digital anomalies, hair and nail defects and neuroradiological findings, including cerebellar hypoplasia and hypomyelination, as well as variable vision and hearing impairment. Additional features include failure to thrive, hypotonia, microcephaly and genitourinary anomalies without any reported congenital heart disease. CONCLUSION In this study, we consolidate the causality of loss of MAPKAPK5 function and further delineate the molecular and phenotypic spectrum associated with this new ultra-rare neurodevelopmental syndrome.
Collapse
Affiliation(s)
- Reza Maroofian
- Department of Neuromuscular Disorders, UCL Queen Square Institute of Neurology, London, UK
| | - Stephanie Efthymiou
- Department of Neuromuscular Disorders, UCL Queen Square Institute of Neurology, London, UK
| | - Mohnish Suri
- Clinical Genetics Service, Nottingham University Hospitals NHS Trust, Nottingham, UK
| | - Fatima Rahman
- Department of Developmental - Behavioral Pediatrics, University of Child Health Sciences & The Children's Hospital, Lahore, Pakistan
| | - Maha S Zaki
- Clinical Genetics Department, Human Genetics and Genome Research Institute, National Research Centre, Cairo, Egypt
| | - Shazia Maqbool
- Department of Developmental - Behavioral Pediatrics, University of Child Health Sciences & The Children's Hospital, Lahore, Pakistan
| | - Najwa Anwa
- Department of Developmental - Behavioral Pediatrics, University of Child Health Sciences & The Children's Hospital, Lahore, Pakistan
| | - Victor L Ruiz-Pérez
- Instituto de Investigaciones Biomédicas "Alberto Sols", Consejo Superior de Investigaciones Científicas (CSIC), Universidad Autónoma de Madrid (UAM), and CIBER de Enfermedades Raras (CIBERER), Madrid, Spain
| | | | - Orly Elpeleg
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
- Department of Genetics, Hadassah Medical Center, Jerusalem, Israel
| | - Sniya Sudhakar
- Department of Radiology, Great Ormond Street Hospital for Children, London, UK
| | - Kshitij Mankad
- Department of Radiology, Great Ormond Street Hospital for Children, London, UK
| | - Tamar Harel
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
- Department of Genetics, Hadassah Medical Center, Jerusalem, Israel
| | - Henry Houlden
- Department of Neuromuscular Disorders, UCL Queen Square Institute of Neurology, London, UK
| |
Collapse
|
7
|
Duggan MR, Butler L, Peng Z, Daya GN, Moghekar A, An Y, Rapp SR, Hayden KM, Shadyab AH, Natale G, Liu L, Snetselaar L, Moaddel R, Rebholz CM, Sullivan K, Ballantyne CM, Resnick SM, Ferrucci L, Walker KA. Plasma proteins related to inflammatory diet predict future cognitive impairment. Mol Psychiatry 2023; 28:1599-1609. [PMID: 36737481 PMCID: PMC10208977 DOI: 10.1038/s41380-023-01975-7] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 01/17/2023] [Accepted: 01/19/2023] [Indexed: 02/05/2023]
Abstract
Dysregulation of the immune system and dietary patterns that increase inflammation can increase the risk for cognitive decline, but the mechanisms by which inflammatory nutritional habits may affect the development of cognitive impairment in aging are not well understood. To determine whether plasma proteins linked to inflammatory diet predict future cognitive impairment, we applied high-throughput proteomic assays to plasma samples from a subset (n = 1528) of Women's Health Initiative Memory Study (WHIMS) participants (mean [SD] baseline age, 71.3 [SD 3.8] years). Results provide insights into how inflammatory nutritional patterns are associated with an immune-related proteome and identify a group of proteins (CXCL10, CCL3, HGF, OPG, CDCP1, NFATC3, ITGA11) related to future cognitive impairment over a 14-year follow-up period. Several of these inflammatory diet proteins were also associated with dementia risk across two external cohorts (ARIC, ESTHER), correlated with plasma biomarkers of Alzheimer's disease (AD) pathology (Aβ42/40) and/or neurodegeneration (NfL), and related to an MRI-defined index of neurodegenerative brain atrophy in a separate cohort (BLSA). In addition to evaluating their biological relevance, assessing their potential role in AD, and characterizing their immune-tissue/cell-specific expression, we leveraged published RNA-seq results to examine how the in vitro regulation of genes encoding these candidate proteins might be altered in response to an immune challenge. Our findings indicate how dietary patterns with higher inflammatory potential relate to plasma levels of immunologically relevant proteins and highlight the molecular mediators which predict subsequent risk for age-related cognitive impairment.
Collapse
Affiliation(s)
- Michael R Duggan
- Laboratory of Behavioral Neuroscience, National Institute on Aging, Baltimore, MD, USA
| | - Lauren Butler
- Laboratory of Behavioral Neuroscience, National Institute on Aging, Baltimore, MD, USA
| | - Zhongsheng Peng
- Laboratory of Behavioral Neuroscience, National Institute on Aging, Baltimore, MD, USA
| | - Gulzar N Daya
- Laboratory of Behavioral Neuroscience, National Institute on Aging, Baltimore, MD, USA
| | - Abhay Moghekar
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Yang An
- Laboratory of Behavioral Neuroscience, National Institute on Aging, Baltimore, MD, USA
| | - Stephen R Rapp
- Department of Social Sciences and Health Policy, Wake Forest University School of Medicine, Winston-Salem, NC, USA
- Department of Psychiatry & Behavioral Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Kathleen M Hayden
- Department of Psychiatry & Behavioral Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Aladdin H Shadyab
- Herbert Wertheim School of Public Health and Human Longevity Science, University of California, San Diego, La Jolla, CA, USA
| | - Ginny Natale
- Program in Public Health, Stony Brook University School of Medicine, Stony Brook, NY, USA
| | - Longjian Liu
- Department of Epidemiology and Biostatistics, Drexel University Dornsife School of Public Health, Philadelphia, PA, USA
| | - Linda Snetselaar
- Department of Epidemiology, University of Iowa, Iowa City, IA, USA
| | - Ruin Moaddel
- Laboratory of Clinical Investigation, National Institute on Aging, Baltimore, MD, USA
| | - Casey M Rebholz
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Kevin Sullivan
- Department of Medicine, University of Mississippi Medical Center, Jackson, MS, USA
| | - Christie M Ballantyne
- Section of Cardiovascular Research, Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Susan M Resnick
- Laboratory of Behavioral Neuroscience, National Institute on Aging, Baltimore, MD, USA
| | - Luigi Ferrucci
- Translational Gerontology Branch, National Institute on Aging, Baltimore, MD, USA
| | - Keenan A Walker
- Laboratory of Behavioral Neuroscience, National Institute on Aging, Baltimore, MD, USA.
| |
Collapse
|
8
|
Hillary RF, Gadd DA, McCartney DL, Shi L, Campbell A, Walker RM, Ritchie CW, Deary IJ, Evans KL, Nevado‐Holgado AJ, Hayward C, Porteous DJ, McIntosh AM, Lovestone S, Robinson MR, Marioni RE. Genome- and epigenome-wide studies of plasma protein biomarkers for Alzheimer's disease implicate TBCA and TREM2 in disease risk. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2022; 14:e12280. [PMID: 35475137 PMCID: PMC9019629 DOI: 10.1002/dad2.12280] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 12/03/2021] [Accepted: 12/07/2021] [Indexed: 11/24/2022]
Abstract
Introduction The levels of many blood proteins are associated with Alzheimer's disease (AD) or its pathological hallmarks. Elucidating the molecular factors that control circulating levels of these proteins may help to identify proteins associated with disease risk mechanisms. Methods Genome-wide and epigenome-wide studies (nindividuals ≤1064) were performed on plasma levels of 282 AD-associated proteins, identified by a structured literature review. Bayesian penalized regression estimated contributions of genetic and epigenetic variation toward inter-individual differences in plasma protein levels. Mendelian randomization (MR) and co-localization tested associations between proteins and disease-related phenotypes. Results Sixty-four independent genetic and 26 epigenetic loci were associated with 45 proteins. Novel findings included an association between plasma triggering receptor expressed on myeloid cells 2 (TREM2) levels and a polymorphism and cytosine-phosphate-guanine (CpG) site within the MS4A4A locus. Higher plasma tubulin-specific chaperone A (TBCA) and TREM2 levels were significantly associated with lower AD risk. Discussion Our data inform the regulation of biomarker levels and their relationships with AD.
Collapse
Affiliation(s)
- Robert F. Hillary
- Centre for Genomic and Experimental MedicineInstitute of Genetics and Cancer, University of EdinburghEdinburghUK
| | - Danni A. Gadd
- Centre for Genomic and Experimental MedicineInstitute of Genetics and Cancer, University of EdinburghEdinburghUK
| | - Daniel L. McCartney
- Centre for Genomic and Experimental MedicineInstitute of Genetics and Cancer, University of EdinburghEdinburghUK
| | - Liu Shi
- Department of PsychiatryUniversity of OxfordOxfordUK
| | - Archie Campbell
- Centre for Genomic and Experimental MedicineInstitute of Genetics and Cancer, University of EdinburghEdinburghUK
| | - Rosie M. Walker
- Centre for Genomic and Experimental MedicineInstitute of Genetics and Cancer, University of EdinburghEdinburghUK
- Centre for Clinical Brain Sciences, Chancellor's Building, 49 Little France CrescentUniversity of EdinburghEdinburghUK
| | - Craig W. Ritchie
- Edinburgh Dementia Prevention, Centre for Clinical Brain SciencesUniversity of EdinburghEdinburghUK
| | - Ian J. Deary
- Lothian Birth Cohorts, Department of PsychologyUniversity of EdinburghEdinburghUK
| | - Kathryn L. Evans
- Centre for Genomic and Experimental MedicineInstitute of Genetics and Cancer, University of EdinburghEdinburghUK
| | | | - Caroline Hayward
- MRC Human Genetics UnitInstitute of Genetics and CancerUniversity of EdinburghEdinburghUK
| | - David J. Porteous
- Centre for Genomic and Experimental MedicineInstitute of Genetics and Cancer, University of EdinburghEdinburghUK
| | - Andrew M. McIntosh
- Centre for Genomic and Experimental MedicineInstitute of Genetics and Cancer, University of EdinburghEdinburghUK
- Division of Psychiatry, Centre for Clinical Brain SciencesUniversity of EdinburghEdinburghUK
| | - Simon Lovestone
- Department of PsychiatryUniversity of OxfordOxfordUK
- NeurodegenerationJohnson and Johnson Medical LtdWokinghamUK
| | - Matthew R. Robinson
- Medical Genomics GroupInstitute of Science and Technology AustriaKlosterneuburgAustria
| | - Riccardo E. Marioni
- Centre for Genomic and Experimental MedicineInstitute of Genetics and Cancer, University of EdinburghEdinburghUK
| |
Collapse
|
9
|
Singh K, Cheung BM, Xu A. Ultrasensitive detection of blood biomarkers of Alzheimer's and Parkinson's diseases: a systematic review. Biomark Med 2021; 15:1693-1708. [PMID: 34743546 DOI: 10.2217/bmm-2021-0219] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Purpose: Neurodegenerative disorders are a global health burden with costly and invasive diagnoses relying on brain imaging technology or CSF-based biomarkers. Therefore, considerable efforts to identify blood-biomarkers for Alzheimer's (AD) and Parkinson's diseases (PD) are ongoing. Objectives: This review evaluates the blood biomarkers for AD and PD for their diagnostic value. Methods: This study systematically reviewed articles published between July 1984 and February 2021. Among 1266 papers, we selected 42 studies for a systematic review and 23 studies for meta-analysis. Results & conclusion: Our analysis highlights P-tau181, T-tau and Nfl as promising blood biomarkers for AD diagnosis. Nfl levels were consistently raised in 16 AD and three PD cohorts. P-tau181 and T-tau were also significantly increased in 12 and eight AD cohorts, respectively.
Collapse
Affiliation(s)
- Kailash Singh
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong, China
| | - Bernard My Cheung
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong, China.,Department of Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong, China
| | - Aimin Xu
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong, China.,Department of Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong, China.,Department of Pharmacy & Pharmacology, The University of Hong Kong, Hong Kong, China
| |
Collapse
|
10
|
Ma Y, Qi Q, He Q, Gilyazova NS, Ibeanu G, Li PA. Neuroprotection by B355252 against Glutamate-Induced Cytotoxicity in Murine Hippocampal HT-22 Cells Is Associated with Activation of ERK3 Signaling Pathway. Biol Pharm Bull 2021; 44:1662-1669. [PMID: 34719643 DOI: 10.1248/bpb.b21-00158] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Glutamate differentially affects the levels extracellular signal-regulated kinase (ERK)1/2 and ERK3 and the protective effect of B355252, an aryl thiophene compound, 4-chloro-N-(naphthalen-1-ylmethyl)-5-(3-(piperazin-1-yl)phenoxy)thiophene-2-sulfonamide, is associated with suppression of ERK1/2. The objectives of this study were to further investigate the impact of B355252 on ERK3 and its downstream signaling pathways affected by glutamate exposure in the mouse hippocampal HT-22 neuronal cells. Murine hippocampal HT22 cells were incubated with glutamate and treated with B355252. Cell viability was assessed, protein levels of pERK3, ERK3, mitogen-activated protein kinase-activated protein kinase-5 (MAPKAPK-5), steroid receptor coactivator 3 (SRC-3), p-S6 and S6 were measured using Western blotting, and immunoreactivity of p-S6 was determined by immunocytochemistry. The results reveal that glutamate markedly diminished the protein levels of p-ERK3 and its downstream targets MK-5 and SRC-3 and increased p-S6, an indicator for mechanistic target of rapamycin (mTOR) activation. Conversely, treatment with B355252 protected the cells from glutamate-induced damage and prevented the glutamate-caused declines of p-ERK3, MK-5 and SRC-3 and increase of p-S6. Our study demonstrates that one of the mechanisms that glutamate mediates its cytotoxicity is through suppression of ERK3 and that B355252 rescues the cells from glutamate toxicity by reverting ERK3 level.
Collapse
Affiliation(s)
- Yanni Ma
- Institute of Clinical Pharmacology, Department of Pharmacy, General Hospital of Ningxia Medical University.,Department of Pharmaceutical Sciences, Biomanufacturing Research Institute and Technological Enterprise (BRITE), College of Health and Sciences, North Carolina Central University
| | - Qi Qi
- Department of Pharmaceutical Sciences, Biomanufacturing Research Institute and Technological Enterprise (BRITE), College of Health and Sciences, North Carolina Central University.,The Julis Chambers Biomedical Biotechnology Research Institute (BBRI), North Carolina Central University
| | - Qingping He
- Department of Pharmaceutical Sciences, Biomanufacturing Research Institute and Technological Enterprise (BRITE), College of Health and Sciences, North Carolina Central University
| | - Nailya S Gilyazova
- Department of Pharmaceutical Sciences, Biomanufacturing Research Institute and Technological Enterprise (BRITE), College of Health and Sciences, North Carolina Central University
| | - Gordon Ibeanu
- Department of Pharmaceutical Sciences, Biomanufacturing Research Institute and Technological Enterprise (BRITE), College of Health and Sciences, North Carolina Central University
| | - P Andy Li
- Department of Pharmaceutical Sciences, Biomanufacturing Research Institute and Technological Enterprise (BRITE), College of Health and Sciences, North Carolina Central University
| |
Collapse
|
11
|
Berry J, Brooks B, Genge A, Heiman-Patterson T, Appel S, Benatar M, Bowser R, Cudkowicz M, Gooch C, Shefner J, Westra J, Agnese W, Merrill C, Nelson S, Apple S. Radicava/Edaravone Findings in Biomarkers From Amyotrophic Lateral Sclerosis (REFINE-ALS): Protocol and Study Design. Neurol Clin Pract 2021; 11:e472-e479. [PMID: 34476128 PMCID: PMC8382414 DOI: 10.1212/cpj.0000000000000968] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Accepted: 09/03/2020] [Indexed: 11/15/2022]
Abstract
Objectives To identify putative biomarkers that may serve as quantifiable, biological, nonclinical measures of the pharmacodynamic effect of edaravone in amyotrophic lateral sclerosis (ALS) and to report real-world treatment outcomes. Methods This is a prospective, observational, longitudinal, multicenter (up to 40 sites) US study (Clinicaltrials.gov; NCT04259255) with at least 200 patients with ALS who will receive edaravone for 24 weeks (6 cycles; Food and Drug Administration-approved regimen). All participants must either be treatment naive for edaravone or be more than 1 month without receiving any edaravone dose before screening. Biomarker quantification and other assessments will be performed at baseline (before cycle 1) and during cycles 1, 3, and 6. Selected biomarkers of oxidative stress, inflammation, neuronal injury and death, and muscle injury, as well as biomarker discovery panels (EpiSwitch and SOMAscan), will be evaluated and, when feasible, compared with biobanked samples. Clinical efficacy assessments will include the ALS Functional Rating Scale-Revised, King's clinical staging, ALS Assessment Questionnaire-40, Appel ALS Score (Rating Scale), slow vital capacity, hand-held dynamometry and grip strength, and time to specified states of disease progression or death. DNA samples will also be collected for potential genomic evaluation. The predicted rates of progression and survival, and their potential correlations with biomarkers, will be evaluated. Adverse events related to the study will be reported. Results The study is estimated to be completed in 2022 with an interim analysis planned. Conclusions Findings may help to further the understanding of the pharmacodynamic effect of edaravone, including changes in biomarkers, in response to treatment.
Collapse
Affiliation(s)
- James Berry
- Massachusetts General Hospital (JB), Boston; Atrium Health Neurosciences Institute (BB), Carolinas Medical Center, University of North Carolina School of Medicine-Charlotte Campus; Montreal Neurological Institute and Hospital (AG), QC, Canada; Lewis Katz School of Medicine (TH-P), Temple University, Philadelphia, PA; Houston Methodist (S. Appel), TX; University of Miami (MB), FL; Barrow Neurological Institute (RB, JS), Phoenix, AZ; Harvard Medical School (MC), Boston, MA; University of South Florida (CG), Tampa; Oxford BioDynamics Inc. (JW), Wilmington, DE; and Mitsubishi Tanabe Pharma America (WA, CM, SN, S. Apple), Inc., Jersey City, NJ
| | - Benjamin Brooks
- Massachusetts General Hospital (JB), Boston; Atrium Health Neurosciences Institute (BB), Carolinas Medical Center, University of North Carolina School of Medicine-Charlotte Campus; Montreal Neurological Institute and Hospital (AG), QC, Canada; Lewis Katz School of Medicine (TH-P), Temple University, Philadelphia, PA; Houston Methodist (S. Appel), TX; University of Miami (MB), FL; Barrow Neurological Institute (RB, JS), Phoenix, AZ; Harvard Medical School (MC), Boston, MA; University of South Florida (CG), Tampa; Oxford BioDynamics Inc. (JW), Wilmington, DE; and Mitsubishi Tanabe Pharma America (WA, CM, SN, S. Apple), Inc., Jersey City, NJ
| | - Angela Genge
- Massachusetts General Hospital (JB), Boston; Atrium Health Neurosciences Institute (BB), Carolinas Medical Center, University of North Carolina School of Medicine-Charlotte Campus; Montreal Neurological Institute and Hospital (AG), QC, Canada; Lewis Katz School of Medicine (TH-P), Temple University, Philadelphia, PA; Houston Methodist (S. Appel), TX; University of Miami (MB), FL; Barrow Neurological Institute (RB, JS), Phoenix, AZ; Harvard Medical School (MC), Boston, MA; University of South Florida (CG), Tampa; Oxford BioDynamics Inc. (JW), Wilmington, DE; and Mitsubishi Tanabe Pharma America (WA, CM, SN, S. Apple), Inc., Jersey City, NJ
| | - Terry Heiman-Patterson
- Massachusetts General Hospital (JB), Boston; Atrium Health Neurosciences Institute (BB), Carolinas Medical Center, University of North Carolina School of Medicine-Charlotte Campus; Montreal Neurological Institute and Hospital (AG), QC, Canada; Lewis Katz School of Medicine (TH-P), Temple University, Philadelphia, PA; Houston Methodist (S. Appel), TX; University of Miami (MB), FL; Barrow Neurological Institute (RB, JS), Phoenix, AZ; Harvard Medical School (MC), Boston, MA; University of South Florida (CG), Tampa; Oxford BioDynamics Inc. (JW), Wilmington, DE; and Mitsubishi Tanabe Pharma America (WA, CM, SN, S. Apple), Inc., Jersey City, NJ
| | - Stanley Appel
- Massachusetts General Hospital (JB), Boston; Atrium Health Neurosciences Institute (BB), Carolinas Medical Center, University of North Carolina School of Medicine-Charlotte Campus; Montreal Neurological Institute and Hospital (AG), QC, Canada; Lewis Katz School of Medicine (TH-P), Temple University, Philadelphia, PA; Houston Methodist (S. Appel), TX; University of Miami (MB), FL; Barrow Neurological Institute (RB, JS), Phoenix, AZ; Harvard Medical School (MC), Boston, MA; University of South Florida (CG), Tampa; Oxford BioDynamics Inc. (JW), Wilmington, DE; and Mitsubishi Tanabe Pharma America (WA, CM, SN, S. Apple), Inc., Jersey City, NJ
| | - Michael Benatar
- Massachusetts General Hospital (JB), Boston; Atrium Health Neurosciences Institute (BB), Carolinas Medical Center, University of North Carolina School of Medicine-Charlotte Campus; Montreal Neurological Institute and Hospital (AG), QC, Canada; Lewis Katz School of Medicine (TH-P), Temple University, Philadelphia, PA; Houston Methodist (S. Appel), TX; University of Miami (MB), FL; Barrow Neurological Institute (RB, JS), Phoenix, AZ; Harvard Medical School (MC), Boston, MA; University of South Florida (CG), Tampa; Oxford BioDynamics Inc. (JW), Wilmington, DE; and Mitsubishi Tanabe Pharma America (WA, CM, SN, S. Apple), Inc., Jersey City, NJ
| | - Robert Bowser
- Massachusetts General Hospital (JB), Boston; Atrium Health Neurosciences Institute (BB), Carolinas Medical Center, University of North Carolina School of Medicine-Charlotte Campus; Montreal Neurological Institute and Hospital (AG), QC, Canada; Lewis Katz School of Medicine (TH-P), Temple University, Philadelphia, PA; Houston Methodist (S. Appel), TX; University of Miami (MB), FL; Barrow Neurological Institute (RB, JS), Phoenix, AZ; Harvard Medical School (MC), Boston, MA; University of South Florida (CG), Tampa; Oxford BioDynamics Inc. (JW), Wilmington, DE; and Mitsubishi Tanabe Pharma America (WA, CM, SN, S. Apple), Inc., Jersey City, NJ
| | - Merit Cudkowicz
- Massachusetts General Hospital (JB), Boston; Atrium Health Neurosciences Institute (BB), Carolinas Medical Center, University of North Carolina School of Medicine-Charlotte Campus; Montreal Neurological Institute and Hospital (AG), QC, Canada; Lewis Katz School of Medicine (TH-P), Temple University, Philadelphia, PA; Houston Methodist (S. Appel), TX; University of Miami (MB), FL; Barrow Neurological Institute (RB, JS), Phoenix, AZ; Harvard Medical School (MC), Boston, MA; University of South Florida (CG), Tampa; Oxford BioDynamics Inc. (JW), Wilmington, DE; and Mitsubishi Tanabe Pharma America (WA, CM, SN, S. Apple), Inc., Jersey City, NJ
| | - Clifton Gooch
- Massachusetts General Hospital (JB), Boston; Atrium Health Neurosciences Institute (BB), Carolinas Medical Center, University of North Carolina School of Medicine-Charlotte Campus; Montreal Neurological Institute and Hospital (AG), QC, Canada; Lewis Katz School of Medicine (TH-P), Temple University, Philadelphia, PA; Houston Methodist (S. Appel), TX; University of Miami (MB), FL; Barrow Neurological Institute (RB, JS), Phoenix, AZ; Harvard Medical School (MC), Boston, MA; University of South Florida (CG), Tampa; Oxford BioDynamics Inc. (JW), Wilmington, DE; and Mitsubishi Tanabe Pharma America (WA, CM, SN, S. Apple), Inc., Jersey City, NJ
| | - Jeremy Shefner
- Massachusetts General Hospital (JB), Boston; Atrium Health Neurosciences Institute (BB), Carolinas Medical Center, University of North Carolina School of Medicine-Charlotte Campus; Montreal Neurological Institute and Hospital (AG), QC, Canada; Lewis Katz School of Medicine (TH-P), Temple University, Philadelphia, PA; Houston Methodist (S. Appel), TX; University of Miami (MB), FL; Barrow Neurological Institute (RB, JS), Phoenix, AZ; Harvard Medical School (MC), Boston, MA; University of South Florida (CG), Tampa; Oxford BioDynamics Inc. (JW), Wilmington, DE; and Mitsubishi Tanabe Pharma America (WA, CM, SN, S. Apple), Inc., Jersey City, NJ
| | - Jurjen Westra
- Massachusetts General Hospital (JB), Boston; Atrium Health Neurosciences Institute (BB), Carolinas Medical Center, University of North Carolina School of Medicine-Charlotte Campus; Montreal Neurological Institute and Hospital (AG), QC, Canada; Lewis Katz School of Medicine (TH-P), Temple University, Philadelphia, PA; Houston Methodist (S. Appel), TX; University of Miami (MB), FL; Barrow Neurological Institute (RB, JS), Phoenix, AZ; Harvard Medical School (MC), Boston, MA; University of South Florida (CG), Tampa; Oxford BioDynamics Inc. (JW), Wilmington, DE; and Mitsubishi Tanabe Pharma America (WA, CM, SN, S. Apple), Inc., Jersey City, NJ
| | - Wendy Agnese
- Massachusetts General Hospital (JB), Boston; Atrium Health Neurosciences Institute (BB), Carolinas Medical Center, University of North Carolina School of Medicine-Charlotte Campus; Montreal Neurological Institute and Hospital (AG), QC, Canada; Lewis Katz School of Medicine (TH-P), Temple University, Philadelphia, PA; Houston Methodist (S. Appel), TX; University of Miami (MB), FL; Barrow Neurological Institute (RB, JS), Phoenix, AZ; Harvard Medical School (MC), Boston, MA; University of South Florida (CG), Tampa; Oxford BioDynamics Inc. (JW), Wilmington, DE; and Mitsubishi Tanabe Pharma America (WA, CM, SN, S. Apple), Inc., Jersey City, NJ
| | - Charlotte Merrill
- Massachusetts General Hospital (JB), Boston; Atrium Health Neurosciences Institute (BB), Carolinas Medical Center, University of North Carolina School of Medicine-Charlotte Campus; Montreal Neurological Institute and Hospital (AG), QC, Canada; Lewis Katz School of Medicine (TH-P), Temple University, Philadelphia, PA; Houston Methodist (S. Appel), TX; University of Miami (MB), FL; Barrow Neurological Institute (RB, JS), Phoenix, AZ; Harvard Medical School (MC), Boston, MA; University of South Florida (CG), Tampa; Oxford BioDynamics Inc. (JW), Wilmington, DE; and Mitsubishi Tanabe Pharma America (WA, CM, SN, S. Apple), Inc., Jersey City, NJ
| | - Sally Nelson
- Massachusetts General Hospital (JB), Boston; Atrium Health Neurosciences Institute (BB), Carolinas Medical Center, University of North Carolina School of Medicine-Charlotte Campus; Montreal Neurological Institute and Hospital (AG), QC, Canada; Lewis Katz School of Medicine (TH-P), Temple University, Philadelphia, PA; Houston Methodist (S. Appel), TX; University of Miami (MB), FL; Barrow Neurological Institute (RB, JS), Phoenix, AZ; Harvard Medical School (MC), Boston, MA; University of South Florida (CG), Tampa; Oxford BioDynamics Inc. (JW), Wilmington, DE; and Mitsubishi Tanabe Pharma America (WA, CM, SN, S. Apple), Inc., Jersey City, NJ
| | - Stephen Apple
- Massachusetts General Hospital (JB), Boston; Atrium Health Neurosciences Institute (BB), Carolinas Medical Center, University of North Carolina School of Medicine-Charlotte Campus; Montreal Neurological Institute and Hospital (AG), QC, Canada; Lewis Katz School of Medicine (TH-P), Temple University, Philadelphia, PA; Houston Methodist (S. Appel), TX; University of Miami (MB), FL; Barrow Neurological Institute (RB, JS), Phoenix, AZ; Harvard Medical School (MC), Boston, MA; University of South Florida (CG), Tampa; Oxford BioDynamics Inc. (JW), Wilmington, DE; and Mitsubishi Tanabe Pharma America (WA, CM, SN, S. Apple), Inc., Jersey City, NJ
| |
Collapse
|
12
|
Shimada YJ, Raita Y, Liang LW, Maurer MS, Hasegawa K, Fifer MA, Reilly MP. Comprehensive Proteomics Profiling Reveals Circulating Biomarkers of Hypertrophic Cardiomyopathy. Circ Heart Fail 2021; 14:e007849. [PMID: 34192899 DOI: 10.1161/circheartfailure.120.007849] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
BACKGROUND Hypertrophic cardiomyopathy (HCM) is caused by mutations in the genes coding for proteins essential in normal myocardial contraction. However, it remains unclear through which molecular pathways gene mutations mediate the development of HCM. The objectives were to determine plasma protein biomarkers of HCM and to reveal molecular pathways differentially regulated in HCM. METHODS We conducted a multicenter case-control study of cases with HCM and controls with hypertensive left ventricular hypertrophy. We performed plasma proteomics profiling of 1681 proteins. We performed a sparse partial least squares discriminant analysis to develop a proteomics-based discrimination model with data from 1 institution (ie, the training set). We tested the discriminative ability in independent samples from the other institution (ie, the test set). As an exploratory analysis, we executed pathway analysis of significantly dysregulated proteins. Pathways with false discovery rate <0.05 were declared positive. RESULTS The study included 266 cases and 167 controls (n=308 in the training set; n=125 in the test set). Using the proteomics-based model derived from the training set, the area under the receiver operating characteristic curve was 0.89 (95% CI, 0.83-0.94) in the test set. Pathway analysis revealed that the Ras-MAPK (mitogen-activated protein kinase) pathway, along with its upstream and downstream pathways, was upregulated in HCM. Pathways involved in inflammation and fibrosis-for example, the TGF (transforming growth factor)-β pathway-were also upregulated. CONCLUSIONS This study serves as the largest-scale investigation with the most comprehensive proteomics profiling in HCM, revealing circulating biomarkers and exhibiting both novel (eg, Ras-MAPK) and known (eg, TGF-β) pathways differentially regulated in HCM.
Collapse
Affiliation(s)
- Yuichi J Shimada
- Division of Cardiology, Department of Medicine (Y.J.S., L.W.L., M.S.M., M.P.R.), Columbia University Irving Medical Center, New York, NY.,Cardiology Division, Department of Medicine (Y.J.S., M.A.F.), Massachusetts General Hospital, Harvard Medical School, Boston
| | - Yoshihiko Raita
- Department of Emergency Medicine (Y.R., K.H.), Massachusetts General Hospital, Harvard Medical School, Boston
| | - Lusha W Liang
- Division of Cardiology, Department of Medicine (Y.J.S., L.W.L., M.S.M., M.P.R.), Columbia University Irving Medical Center, New York, NY
| | - Mathew S Maurer
- Division of Cardiology, Department of Medicine (Y.J.S., L.W.L., M.S.M., M.P.R.), Columbia University Irving Medical Center, New York, NY
| | - Kohei Hasegawa
- Department of Emergency Medicine (Y.R., K.H.), Massachusetts General Hospital, Harvard Medical School, Boston
| | - Michael A Fifer
- Cardiology Division, Department of Medicine (Y.J.S., M.A.F.), Massachusetts General Hospital, Harvard Medical School, Boston
| | - Muredach P Reilly
- Division of Cardiology, Department of Medicine (Y.J.S., L.W.L., M.S.M., M.P.R.), Columbia University Irving Medical Center, New York, NY.,Irving Institute for Clinical and Translational Research (M.P.R.), Columbia University Irving Medical Center, New York, NY
| |
Collapse
|
13
|
Er F, Goularas D. Predicting the Prognosis of MCI Patients Using Longitudinal MRI Data. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2021; 18:1164-1173. [PMID: 32813661 DOI: 10.1109/tcbb.2020.3017872] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
The aim of this study is to develop a computer-aided diagnosis system with a deep-learning approach for distinguishing "Mild Cognitive Impairment (MCI) due to Alzheimer's Disease (AD)" patients among a list of MCI patients. In this system we are using the power of longitudinal data extracted from magnetic resonance (MR). For this work, a total of 294 MCI patients were selected from the ADNI database. Among them, 125 patients developed AD during their follow-up and the rest remained stable. The proposed computer-aided diagnosis system (CAD) attempts to identify brain regions that are significant for the prediction of developing AD. The longitudinal data were constructed using a 3D Jacobian-based method aiming to track the brain differences between two consecutive follow-ups. The proposed CAD system distinguishes MCI patients who developed AD from those who remained stable with an accuracy of 87.2 percent. Moreover, it does not depend on data acquired by invasive methods or cognitive tests. This work demonstrates that the use of data in different time periods contains information that is beneficial for prognosis prediction purposes that outperform similar methods and are slightly inferior only to those systems that use invasive methods or neuropsychological tests.
Collapse
|
14
|
Platelet-derived growth factor BB: A potential diagnostic blood biomarker for differentiating bipolar disorder from major depressive disorder. J Psychiatr Res 2021; 134:48-56. [PMID: 33360224 DOI: 10.1016/j.jpsychires.2020.12.051] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2020] [Revised: 12/10/2020] [Accepted: 12/17/2020] [Indexed: 01/19/2023]
Abstract
Bipolar disorder (BD) is frequently misdiagnosed as major depressive disorder (MDD) due to overlapping depressive symptoms. This study investigated whether serum platelet-derived growth factor BB (PDGF-BB) is a differential diagnostic biomarker for BD and MDD. An initial SOMAscan proteomics assay of 1311 proteins in small samples from patients with BD and MDD and healthy controls (HCs) suggested that serum levels of PDGF-BB differed between BD and MDD. We then conducted a two-step, exploratory, cross-sectional, case-control study at our institute and five sites that included a total of 549 participants (157 with BD, 144 with MDD, and 248 HCs). Clinical symptoms were assessed using the Hamilton Depression Rating Scale and the Young Mania Rating Scale. In the initial analysis at our institute, serum PDGF-BB levels in the MDD group (n = 36) were significantly lower than those in the BD (n = 39) and HC groups (n = 36). In the multicenter study, serum PDGF-BB levels in the MDD group were again significantly lower than those in the BD and HC groups, with no significant difference between the BD and HC groups. Treatment with sodium valproate was associated with significantly lower serum PDGF-BB levels in patients with BD. After controlling for confounding factors (sex, age, body mass index, clinical severity, and valproate medication), serum PDGF-BB levels were lower in the MDD group than in the BD group regardless of mood state. Our findings suggest that serum PDGF-BB may be a potential biomarker to differentiate BD and MDD.
Collapse
|
15
|
Identification of Circular RNAs in Hypothalamus of Gilts during the Onset of Puberty. Genes (Basel) 2021; 12:genes12010084. [PMID: 33445426 PMCID: PMC7827264 DOI: 10.3390/genes12010084] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 01/06/2021] [Accepted: 01/08/2021] [Indexed: 01/09/2023] Open
Abstract
The disorders of puberty have shown negative outcomes on health of mammals, and the hypothalamus is thought to be the main regulator of puberty by releasing GnRH. Many studies show that the circular RNAs (circRNAs) might be implicated in the timing of puberty in mammals. However, the circRNAs in the hypothalamus of gilts have not been explored. To profile the changes and biological functions of circRNAs in the hypothalamus during the onset of puberty, RNA-seq was utilized to establish pre-, in-, and post-pubertal hypothalamic circRNAs profiles. In this study, the functions of hypothalamic circRNAs were enriched in the signaling pathway of neurotrophin, progesterone-mediated oocyte maturation, oocyte meiosis, insulin, ErbB, and mTOR, which have been highly suggested to be involved in the timing of puberty. Furthermore, 53 circRNAs were identified to be putative hypothalamus-specific expressed circRNAs, and some of them were exclusively expressed in the one of three pubertal stages. Moreover, 22 differentially expressed circRNAs were identified and chosen to construct the circRNA-miRNA-gene network. Moreover, 10 circRNAs were found to be driven by six puberty-related genes (ESR1, NF1, APP, ENPP2, ARNT, and DICER1). Subsequently, the expression changes of several circRNAs were confirmed by RT-qPCR. Collectively, the preliminary results of hypothalamic circRNAs provided useful information for the investigation of the molecular mechanism for the timing of puberty in gilts.
Collapse
|
16
|
Tweedie D, Karnati HK, Mullins R, Pick CG, Hoffer BJ, Goetzl EJ, Kapogiannis D, Greig NH. Time-dependent cytokine and chemokine changes in mouse cerebral cortex following a mild traumatic brain injury. eLife 2020; 9:55827. [PMID: 32804078 PMCID: PMC7473773 DOI: 10.7554/elife.55827] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Accepted: 08/14/2020] [Indexed: 02/07/2023] Open
Abstract
Traumatic brain injury (TBI) is a serious global health problem, many individuals live with TBI-related neurological dysfunction. A lack of biomarkers of TBI has impeded medication development. To identify new potential biomarkers, we time-dependently evaluated mouse brain tissue and neuronally derived plasma extracellular vesicle proteins in a mild model of TBI with parallels to concussive head injury. Mice (CD-1, 30–40 g) received a sham procedure or 30 g weight-drop and were euthanized 8, 24, 48, 72, 96 hr, 7, 14 and 30 days later. We quantified ipsilateral cortical proteins, many of which differed from sham by 8 hours post-mTBI, particularly GAS-1 and VEGF-B were increased while CXCL16 reduced, 23 proteins changed in 4 or more of the time points. Gene ontology pathways mapped from altered proteins over time related to pathological and physiological processes. Validation of proteins identified in this study may provide utility as treatment response biomarkers.
Collapse
Affiliation(s)
- David Tweedie
- Translational Gerontology Branch, Intramural Research Program, National Institute on Aging, NIH, Baltimore, United States
| | - Hanuma Kumar Karnati
- Translational Gerontology Branch, Intramural Research Program, National Institute on Aging, NIH, Baltimore, United States
| | - Roger Mullins
- Laboratory of Clinical Investigation, Intramural Research Program, National Institute on Aging, NIH, Baltimore, United States
| | - Chaim G Pick
- Department of Anatomy and Anthropology, Sackler School of Medicine, Sylvan Adams Sports Institute, and Dr. Miriam and SheldonG. Adelson Chair and Center for the Biology of Addictive Diseases, Tel Aviv University, Tel Aviv, Israel
| | - Barry J Hoffer
- Department of Neurosurgery, Case Western Reserve University School of Medicine, Cleveland, United States
| | - Edward J Goetzl
- Department of Medicine, University of California Medical Center, San Francisco, San Francisco, United States
| | - Dimitrios Kapogiannis
- Laboratory of Clinical Investigation, Intramural Research Program, National Institute on Aging, NIH, Baltimore, United States
| | - Nigel H Greig
- Translational Gerontology Branch, Intramural Research Program, National Institute on Aging, NIH, Baltimore, United States
| |
Collapse
|
17
|
Tanaka T, Lavery R, Varma V, Fantoni G, Colpo M, Thambisetty M, Candia J, Resnick SM, Bennett DA, Biancotto A, Bandinelli S, Ferrucci L. Plasma proteomic signatures predict dementia and cognitive impairment. ALZHEIMER'S & DEMENTIA (NEW YORK, N. Y.) 2020; 6:e12018. [PMID: 32607407 PMCID: PMC7210784 DOI: 10.1002/trc2.12018] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Accepted: 02/27/2020] [Indexed: 12/03/2022]
Abstract
INTRODUCTION Biomarker discovery of dementia and cognitive impairment is important to gather insight into mechanisms underlying the pathogenesis of these conditions. METHODS In 997 adults from the InCHIANTI study, we assessed the association of 1301 plasma proteins with dementia and cognitive impairment. Validation was conducted in two Alzheimer's disease (AD) case-control studies as well as endophenotypes of AD including cognitive decline, brain amyloid burden, and brain volume. RESULTS We identified four risk proteins that were significantly associated with increased odds (peptidase inhibitor 3 (PI3), trefoil factor 3 (TFF3), pregnancy associated plasma protein A (PAPPA), agouti-related peptide (AGRP)) and two protective proteins (myostatin (MSTN), integrin aVb5 (ITGAV/ITGB5)) with decreased odds of baseline cognitive impairment or dementia. Of these, four proteins (MSTN, PI3, TFF3, PAPPA) were associated cognitive decline in subjects that were cognitively normal at baseline. ITGAV/ITGB5 was associated with lower brain amyloid burden, MSTN and ITGAV/ITGB5 were associated with larger brain volume and slower brain atrophy, and PI3, PAPPA, and AGRP were associated with smaller brain volume and/or faster brain atrophy. DISCUSSION These proteins may be useful as non-invasive biomarkers of dementia and cognitive impairment.
Collapse
Affiliation(s)
- Toshiko Tanaka
- Translational Gerontology BranchNational Institute on AgingNIHBaltimoreMaryland
| | - Robert Lavery
- Translational Gerontology BranchNational Institute on AgingNIHBaltimoreMaryland
| | - Vijay Varma
- Laboratory of Behavioral NeuroscienceNational Institute on AgingNIHBaltimoreMaryland
| | - Giovanna Fantoni
- National Institute on Aging (NIA)Intramural Research Program (IRP)Clinical Research Core (CRC)
| | - Marco Colpo
- Geriatric UnitAzienda Sanitaria di FirenzeFlorenceItaly
| | - Madhav Thambisetty
- Laboratory of Behavioral NeuroscienceNational Institute on AgingNIHBaltimoreMaryland
| | - Julian Candia
- Laboratory of Human CarcinogenesisCenter for Cancer ResearchNational Cancer InstituteNIHBethesdaMaryland
| | - Susan M. Resnick
- Laboratory of Behavioral NeuroscienceNational Institute of AgingBaltimoreMaryland
| | - David A. Bennett
- Rush Alzheimer's Disease CenterRush University Medical CenterChicagoIllinois
| | - Angelique Biancotto
- Precision Immunology, Immunology and Inflammation Research Therapeutic AreaSanofiCambridgeMAUSA
| | | | - Luigi Ferrucci
- Translational Gerontology BranchNational Institute on AgingNIHBaltimoreMaryland
| |
Collapse
|
18
|
Martin TC, Ilieva KM, Visconti A, Beaumont M, Kiddle SJ, Dobson RJB, Mangino M, Lim EM, Pezer M, Steves CJ, Bell JT, Wilson SG, Lauc G, Roederer M, Walsh JP, Spector TD, Karagiannis SN. Dysregulated Antibody, Natural Killer Cell and Immune Mediator Profiles in Autoimmune Thyroid Diseases. Cells 2020; 9:E665. [PMID: 32182948 PMCID: PMC7140647 DOI: 10.3390/cells9030665] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 03/04/2020] [Accepted: 03/05/2020] [Indexed: 12/12/2022] Open
Abstract
The pathogenesis of autoimmune thyroid diseases (AITD) is poorly understood and the association between different immune features and the germline variants involved in AITD are yet unclear. We previously observed systemic depletion of IgG core fucosylation and antennary α1,2 fucosylation in peripheral blood mononuclear cells in AITD, correlated with anti-thyroid peroxidase antibody (TPOAb) levels. Fucose depletion is known to potentiate strong antibody-mediated NK cell activation and enhanced target antigen-expressing cell killing. In autoimmunity, this may translate to autoantibody-mediated immune cell recruitment and attack of self-antigen expressing normal tissues. Hence, we investigated the crosstalk between immune cell traits, secreted proteins, genetic variants and the glycosylation patterns of serum IgG, in a multi-omic and cross-sectional study of 622 individuals from the TwinsUK cohort, 172 of whom were diagnosed with AITD. We observed associations between two genetic variants (rs505922 and rs687621), AITD status, the secretion of Desmoglein-2 protein, and the profile of two IgG N-glycan traits in AITD, but further studies need to be performed to better understand their crosstalk in AITD. On the other side, enhanced afucosylated IgG was positively associated with activatory CD335- CD314+ CD158b+ NK cell subsets. Increased levels of the apoptosis and inflammation markers Caspase-2 and Interleukin-1α positively associated with AITD. Two genetic variants associated with AITD, rs1521 and rs3094228, were also associated with altered expression of the thyrocyte-expressed ligands known to recognize the NK cell immunoreceptors CD314 and CD158b. Our analyses reveal a combination of heightened Fc-active IgG antibodies, effector cells, cytokines and apoptotic signals in AITD, and AITD genetic variants associated with altered expression of thyrocyte-expressed ligands to NK cell immunoreceptors. Together, TPOAb responses, dysregulated immune features, germline variants associated with immunoactivity profiles, are consistent with a positive autoreactive antibody-dependent NK cell-mediated immune response likely drawn to the thyroid gland in AITD.
Collapse
Affiliation(s)
- Tiphaine C. Martin
- Department of Twin Research and Genetic Epidemiology, King’s College, London SE1 7EH, UK; (A.V.); (M.B.); (M.M.); (C.J.S.); (J.T.B.); (S.G.W.); (T.D.S.)
- School of Biomedical Sciences, University of Western Australia, Crawley, WA 6009, Australia
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Kristina M. Ilieva
- St John’s Institute of Dermatology, School of Basic & Medical Biosciences, King’s College London, Guy’s Hospital, London SE1 9RT, UK; (K.M.I.); (S.N.K.)
- Breast Cancer Now Research Unit, School of Cancer & Pharmaceutical Sciences, King’s College London, Guy’s Cancer Centre, London SE1 9RT, UK
| | - Alessia Visconti
- Department of Twin Research and Genetic Epidemiology, King’s College, London SE1 7EH, UK; (A.V.); (M.B.); (M.M.); (C.J.S.); (J.T.B.); (S.G.W.); (T.D.S.)
| | - Michelle Beaumont
- Department of Twin Research and Genetic Epidemiology, King’s College, London SE1 7EH, UK; (A.V.); (M.B.); (M.M.); (C.J.S.); (J.T.B.); (S.G.W.); (T.D.S.)
| | - Steven J. Kiddle
- Department of Biostatistics and Health Informatics, Institute of Psychiatry, Psychology and Neuroscience, King’s College, London SE5 8AF, UK; (S.J.K.); (R.J.B.D.)
- MRC Biostatistics Unit, University of Cambridge, Cambridge CB2 0SR, UK
| | - Richard J. B. Dobson
- Department of Biostatistics and Health Informatics, Institute of Psychiatry, Psychology and Neuroscience, King’s College, London SE5 8AF, UK; (S.J.K.); (R.J.B.D.)
- Health Data Research UK (HDR UK), London Institute of Health Informatics, University College London, London NW1 2DA, UK
| | - Massimo Mangino
- Department of Twin Research and Genetic Epidemiology, King’s College, London SE1 7EH, UK; (A.V.); (M.B.); (M.M.); (C.J.S.); (J.T.B.); (S.G.W.); (T.D.S.)
- NIHR Biomedical Research Centre at Guy’s and St. Thomas’s NHS Foundation Trust, London SE1 9RT, UK
| | - Ee Mun Lim
- Department of Endocrinology and Diabetes, Sir Charles Gairdner Hospital, Nedlands, WA 6009, Australia; (E.M.L.); (J.P.W.)
- Medical School, The University of Western Australia, Crawley, WA 6009, Australia
- PathWest Laboratory Medicine, QEII Medical Centre, Nedlands, WA 6009, Australia
| | - Marija Pezer
- Genos, Glycoscience Research Laboratory, 10000 Zagreb, Croatia; (M.P.); (G.L.)
| | - Claire J. Steves
- Department of Twin Research and Genetic Epidemiology, King’s College, London SE1 7EH, UK; (A.V.); (M.B.); (M.M.); (C.J.S.); (J.T.B.); (S.G.W.); (T.D.S.)
| | - Jordana T. Bell
- Department of Twin Research and Genetic Epidemiology, King’s College, London SE1 7EH, UK; (A.V.); (M.B.); (M.M.); (C.J.S.); (J.T.B.); (S.G.W.); (T.D.S.)
| | - Scott G. Wilson
- Department of Twin Research and Genetic Epidemiology, King’s College, London SE1 7EH, UK; (A.V.); (M.B.); (M.M.); (C.J.S.); (J.T.B.); (S.G.W.); (T.D.S.)
- School of Biomedical Sciences, University of Western Australia, Crawley, WA 6009, Australia
- Department of Endocrinology and Diabetes, Sir Charles Gairdner Hospital, Nedlands, WA 6009, Australia; (E.M.L.); (J.P.W.)
| | - Gordan Lauc
- Genos, Glycoscience Research Laboratory, 10000 Zagreb, Croatia; (M.P.); (G.L.)
- Faculty of Pharmacy and Biochemistry, University of Zagreb, 10000 Zagreb, Croatia
| | - Mario Roederer
- ImmunoTechnology Section, Vaccine Research Center, NIAID, NIH, Bethesda, MD 20892, USA;
| | - John P. Walsh
- Department of Endocrinology and Diabetes, Sir Charles Gairdner Hospital, Nedlands, WA 6009, Australia; (E.M.L.); (J.P.W.)
- Medical School, The University of Western Australia, Crawley, WA 6009, Australia
| | - Tim D. Spector
- Department of Twin Research and Genetic Epidemiology, King’s College, London SE1 7EH, UK; (A.V.); (M.B.); (M.M.); (C.J.S.); (J.T.B.); (S.G.W.); (T.D.S.)
| | - Sophia N. Karagiannis
- St John’s Institute of Dermatology, School of Basic & Medical Biosciences, King’s College London, Guy’s Hospital, London SE1 9RT, UK; (K.M.I.); (S.N.K.)
- Breast Cancer Now Research Unit, School of Cancer & Pharmaceutical Sciences, King’s College London, Guy’s Cancer Centre, London SE1 9RT, UK
| |
Collapse
|
19
|
Cuvelliez M, Vandewalle V, Brunin M, Beseme O, Hulot A, de Groote P, Amouyel P, Bauters C, Marot G, Pinet F. Circulating proteomic signature of early death in heart failure patients with reduced ejection fraction. Sci Rep 2019; 9:19202. [PMID: 31844116 PMCID: PMC6914779 DOI: 10.1038/s41598-019-55727-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Accepted: 12/03/2019] [Indexed: 12/11/2022] Open
Abstract
Heart failure (HF) remains a main cause of mortality worldwide. Risk stratification of patients with systolic chronic HF is critical to identify those who may benefit from advanced HF therapies. The aim of this study is to identify plasmatic proteins that could predict the early death (within 3 years) of HF patients with reduced ejection fraction hospitalized in CHRU de Lille. The subproteome targeted by an aptamer-based technology, the Slow Off-rate Modified Aptamer (SOMA) scan assay of 1310 proteins, was profiled in blood samples from 168 HF patients, and 203 proteins were significantly modulated between patients who died of cardiovascular death and patients who were alive after 3 years of HF evaluation (Wilcoxon test, FDR 5%). A molecular network was built using these 203 proteins, and the resulting network contained 2281 molecules assigned to 34 clusters annotated to biological pathways by Gene Ontology. This network model highlighted extracellular matrix organization as the main mechanism involved in early death in HF patients. In parallel, an adaptive Least Absolute Shrinkage and Selection Operator (LASSO) was performed on these 203 proteins, and six proteins were selected as candidates to predict early death in HF patients: complement C3, cathepsin S and F107B were decreased and MAPK5, MMP1 and MMP7 increased in patients who died of cardiovascular causes compared with patients living 3 years after HF evaluation. This proteomic signature of 6 circulating plasma proteins allows the identification of systolic HF patients with a risk of early death.
Collapse
Affiliation(s)
- Marie Cuvelliez
- Univ. Lille, CHU Lille, Inserm, Institut Pasteur de Lille, U1167 - RID-AGE - Facteurs de risque et déterminants moléculaires des maladies liées au vieillissement, F-59000, Lille, France.,FHU REMOD-HF, Lille, France
| | - Vincent Vandewalle
- Univ. Lille, CHU Lille, Inria Lille Nord-Europe, EA2694 - MODAL - MOdels for Data Analysis and Learning, F-59000, Lille, France.,Univ. Lille, « Institut Français de Bioinformatique », « Billille- plateforme de bioinformatique et bioanalyse de Lille », F-59000, Lille, France
| | - Maxime Brunin
- Univ. Lille, « Institut Français de Bioinformatique », « Billille- plateforme de bioinformatique et bioanalyse de Lille », F-59000, Lille, France
| | - Olivia Beseme
- Univ. Lille, CHU Lille, Inserm, Institut Pasteur de Lille, U1167 - RID-AGE - Facteurs de risque et déterminants moléculaires des maladies liées au vieillissement, F-59000, Lille, France.,FHU REMOD-HF, Lille, France
| | - Audrey Hulot
- Univ. Lille, « Institut Français de Bioinformatique », « Billille- plateforme de bioinformatique et bioanalyse de Lille », F-59000, Lille, France
| | - Pascal de Groote
- Univ. Lille, CHU Lille, Inserm, Institut Pasteur de Lille, U1167 - RID-AGE - Facteurs de risque et déterminants moléculaires des maladies liées au vieillissement, F-59000, Lille, France.,FHU REMOD-HF, Lille, France
| | - Philippe Amouyel
- Univ. Lille, CHU Lille, Inserm, Institut Pasteur de Lille, U1167 - RID-AGE - Facteurs de risque et déterminants moléculaires des maladies liées au vieillissement, F-59000, Lille, France
| | - Christophe Bauters
- Univ. Lille, CHU Lille, Inserm, Institut Pasteur de Lille, U1167 - RID-AGE - Facteurs de risque et déterminants moléculaires des maladies liées au vieillissement, F-59000, Lille, France.,FHU REMOD-HF, Lille, France
| | - Guillemette Marot
- Univ. Lille, CHU Lille, Inria Lille Nord-Europe, EA2694 - MODAL - MOdels for Data Analysis and Learning, F-59000, Lille, France.,Univ. Lille, « Institut Français de Bioinformatique », « Billille- plateforme de bioinformatique et bioanalyse de Lille », F-59000, Lille, France
| | - Florence Pinet
- Univ. Lille, CHU Lille, Inserm, Institut Pasteur de Lille, U1167 - RID-AGE - Facteurs de risque et déterminants moléculaires des maladies liées au vieillissement, F-59000, Lille, France. .,FHU REMOD-HF, Lille, France.
| |
Collapse
|
20
|
Peña-Bautista C, Baquero M, Vento M, Cháfer-Pericás C. Omics-based Biomarkers for the Early Alzheimer Disease Diagnosis and Reliable Therapeutic Targets Development. Curr Neuropharmacol 2019; 17:630-647. [PMID: 30255758 PMCID: PMC6712290 DOI: 10.2174/1570159x16666180926123722] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Revised: 08/31/2018] [Accepted: 09/19/2018] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND Alzheimer's disease (AD), the most common cause of dementia in adulthood, has great medical, social, and economic impact worldwide. Available treatments result in symptomatic relief, and most of them are indicated from the early stages of the disease. Therefore, there is an increasing body of research developing accurate and early diagnoses, as well as diseasemodifying therapies. OBJECTIVE Advancing the knowledge of AD physiopathological mechanisms, improving early diagnosis and developing effective treatments from omics-based biomarkers. METHODS Studies using omics technologies to detect early AD, were reviewed with a particular focus on the metabolites/lipids, micro-RNAs and proteins, which are identified as potential biomarkers in non-invasive samples. RESULTS This review summarizes recent research on metabolomics/lipidomics, epigenomics and proteomics, applied to early AD detection. Main research lines are the study of metabolites from pathways, such as lipid, amino acid and neurotransmitter metabolisms, cholesterol biosynthesis, and Krebs and urea cycles. In addition, some microRNAs and proteins (microglobulins, interleukins), related to a common network with amyloid precursor protein and tau, have been also identified as potential biomarkers. Nevertheless, the reproducibility of results among studies is not good enough and a standard methodological approach is needed in order to obtain accurate information. CONCLUSION The assessment of metabolomic/lipidomic, epigenomic and proteomic changes associated with AD to identify early biomarkers in non-invasive samples from well-defined participants groups will potentially allow the advancement in the early diagnosis and improvement of therapeutic interventions.
Collapse
Affiliation(s)
| | | | | | - Consuelo Cháfer-Pericás
- Address correspondence to this author at the Health Research Institute La Fe, Avda de Fernando Abril Martorell, 106; 46026 Valencia, Spain;Tel: +34 96 124 66 61; Fax: + 34 96 124 57 46; E-mail:
| |
Collapse
|
21
|
Hugon J, Mouton-Liger F, Cognat E, Dumurgier J, Paquet C. Blood-Based Kinase Assessments in Alzheimer's Disease. Front Aging Neurosci 2018; 10:338. [PMID: 30487744 PMCID: PMC6246745 DOI: 10.3389/fnagi.2018.00338] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Accepted: 10/05/2018] [Indexed: 12/11/2022] Open
Abstract
Alzheimer’s disease (AD) is marked by memory disturbances followed by aphasia, apraxia and agnosia. Brain lesions include the accumulation of the amyloid peptide in extracellular plaques, neurofibrillary tangles with abnormally phosphorylated tau protein and synaptic and neuronal loss. New findings have suggested that brain lesions could occur one or two decades before the first clinical signs. This asymptomatic preclinical phase could be an opportunity to put in place a secondary prevention but the detection of these brain lesions can only be achieved so far by cerebrospinal fluid (CSF) evaluation or molecular amyloid and tau PET imaging. There is an urgent need to find out simple and easily accessible new biomarkers to set up an efficient screening in adult and aging population. Neuropathological and biochemical studies have revealed that abnormal accumulations of potentially toxic kinases are present in the brains of AD patients. Kinase activation leads to abnormal tau phosphorylation, amyloid production, apoptosis and neuroinflammation. Increased levels of these kinases are present in the CSF of mild cognitive impairment (MCI) and AD patients. Over the last years the search for abnormal kinase levels was performed in the blood of patients. Glycogen synthase kinase 3 (GSK 3), protein kinase R (PKR), mamalian target of rapamycin (mTOR), dual specificity tyrosine-phosphorylation-regulated kinase 1A (DIRK1A), c-Jun N-terminal kinase (JNK), protein 70 kD ribosomal protein S6 kinase (P70S6K), ERK2 and other kinase concentrations were evaluated and abnormal levels were found in many studies. For example, GSK3 levels are increased in MCI and AD patients. PKR levels are also augmented in peripheral blood mononuclear cells (PBMC) of AD patients. In the future, the assessment of several blood kinase levels in large cohorts of patients will be needed to confirm the usefulness of this test at an early phase of the disease.
Collapse
Affiliation(s)
- Jacques Hugon
- Center of Cognitive Neurology, Lariboisiere Fernand-Widal Hospital, APHP, University Paris Diderot, Paris, France.,INSERM U 942, Paris, France
| | - François Mouton-Liger
- Center of Cognitive Neurology, Lariboisiere Fernand-Widal Hospital, APHP, University Paris Diderot, Paris, France.,INSERM U 942, Paris, France
| | - Emmanuel Cognat
- Center of Cognitive Neurology, Lariboisiere Fernand-Widal Hospital, APHP, University Paris Diderot, Paris, France.,INSERM U 942, Paris, France
| | - Julien Dumurgier
- Center of Cognitive Neurology, Lariboisiere Fernand-Widal Hospital, APHP, University Paris Diderot, Paris, France.,INSERM U 942, Paris, France
| | - Claire Paquet
- Center of Cognitive Neurology, Lariboisiere Fernand-Widal Hospital, APHP, University Paris Diderot, Paris, France.,INSERM U 942, Paris, France
| |
Collapse
|
22
|
Patel V, Dwivedi AK, Deodhar S, Mishra I, Cistola DP. Aptamer-based search for correlates of plasma and serum water T 2: implications for early metabolic dysregulation and metabolic syndrome. Biomark Res 2018; 6:28. [PMID: 30237882 PMCID: PMC6142358 DOI: 10.1186/s40364-018-0143-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Accepted: 08/29/2018] [Indexed: 12/13/2022] Open
Abstract
Background Metabolic syndrome is a cluster of abnormalities that increases the risk for type 2 diabetes and atherosclerosis. Plasma and serum water T2 from benchtop nuclear magnetic resonance relaxometry are early, global and practical biomarkers for metabolic syndrome and its underlying abnormalities. In a prior study, water T2 was analyzed against ~ 130 strategically selected proteins and metabolites to identify associations with insulin resistance, inflammation and dyslipidemia. In the current study, the analysis was broadened ten-fold using a modified aptamer (SOMAmer) library, enabling an unbiased search for new proteins correlated with water T2 and thus, metabolic health. Methods Water T2 measurements were recorded using fasting plasma and serum from non-diabetic human subjects. In parallel, plasma samples were analyzed using a SOMAscan assay that employed modified DNA aptamers to determine the relative concentrations of 1310 proteins. A multi-step statistical analysis was performed to identify the biomarkers most predictive of water T2. The steps included Spearman rank correlation, followed by principal components analysis with variable clustering, random forests for biomarker selection, and regression trees for biomarker ranking. Results The multi-step analysis unveiled five new proteins most predictive of water T2: hepatocyte growth factor, receptor tyrosine kinase FLT3, bone sialoprotein 2, glucokinase regulatory protein and endothelial cell-specific molecule 1. Three of the five strongest predictors of water T2 have been previously implicated in cardiometabolic diseases. Hepatocyte growth factor has been associated with incident type 2 diabetes, and endothelial cell specific molecule 1, with atherosclerosis in subjects with diabetes. Glucokinase regulatory protein plays a critical role in hepatic glucose uptake and metabolism and is a drug target for type 2 diabetes. By contrast, receptor tyrosine kinase FLT3 and bone sialoprotein 2 have not been previously associated with metabolic conditions. In addition to the five most predictive biomarkers, the analysis unveiled other strong correlates of water T2 that would not have been identified in a hypothesis-driven biomarker search. Conclusions The identification of new proteins associated with water T2 demonstrates the value of this approach to biomarker discovery. It provides new insights into the metabolic significance of water T2 and the pathophysiology of metabolic syndrome. Electronic supplementary material The online version of this article (10.1186/s40364-018-0143-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Vipulkumar Patel
- 1Nanoparticle Diagnostics Laboratory, Institute for Cardiovascular & Metabolic Diseases, University of North Texas Health Science Center, Fort Worth, TX 76107 USA.,2Center of Emphasis in Diabetes & Metabolism, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, TX 79905 USA
| | - Alok K Dwivedi
- 3Division of Biostatistics & Epidemiology, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, TX 79905 USA
| | - Sneha Deodhar
- 1Nanoparticle Diagnostics Laboratory, Institute for Cardiovascular & Metabolic Diseases, University of North Texas Health Science Center, Fort Worth, TX 76107 USA
| | - Ina Mishra
- 1Nanoparticle Diagnostics Laboratory, Institute for Cardiovascular & Metabolic Diseases, University of North Texas Health Science Center, Fort Worth, TX 76107 USA.,2Center of Emphasis in Diabetes & Metabolism, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, TX 79905 USA
| | - David P Cistola
- 1Nanoparticle Diagnostics Laboratory, Institute for Cardiovascular & Metabolic Diseases, University of North Texas Health Science Center, Fort Worth, TX 76107 USA.,2Center of Emphasis in Diabetes & Metabolism, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, TX 79905 USA
| |
Collapse
|
23
|
Curran AM, Scott-Boyer MP, Kaput J, Ryan MF, Drummond E, Gibney ER, Gibney MJ, Roche HM, Brennan L. A proteomic signature that reflects pancreatic beta-cell function. PLoS One 2018; 13:e0202727. [PMID: 30161145 PMCID: PMC6117012 DOI: 10.1371/journal.pone.0202727] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Accepted: 08/08/2018] [Indexed: 01/08/2023] Open
Abstract
AIM Proteomics has the potential to enhance early identification of beta-cell dysfunction, in conjunction with monitoring the various stages of type 2 diabetes onset. The most routine method of assessing pancreatic beta-cell function is an oral glucose tolerance test, however this method is time consuming and carries a participant burden. The objectives of this research were to identify protein signatures and pathways related to pancreatic beta-cell function in fasting blood samples. METHODS Beta-cell function measures were calculated for MECHE study participants who completed an oral glucose tolerance test and had proteomic data (n = 100). Information on 1,129 protein levels was obtained using the SOMAscan assay. Receiver operating characteristic curves were used to assess discriminatory ability of proteins of interest. Subsequent in vitro experiments were performed using the BRIN-BD11 pancreatic beta-cell line. Replication of findings were achieved in a second human cohort where possible. RESULTS Twenty-two proteins measured by aptamer technology were significantly associated with beta-cell function/HOMA-IR while 17 proteins were significantly associated with the disposition index (p ≤ 0.01). Receiver operator characteristic curves determined the protein panels to have excellent discrimination between low and high beta-cell function. Linear regression analysis determined that beta-endorphin and IL-17F have strong associations with beta-cell function/HOMA-IR, β = 0.039 (p = 0.005) and β = -0.027 (p = 0.013) respectively. Calcineurin and CRTAM were strongly associated with the disposition index (β = 0.005 and β = 0.005 respectively, p = 0.012). In vitro experiments confirmed that IL-17F modulated insulin secretion in the BRIN-BD11 cell line, with the lower concentration of 10 ng/mL significantly increasing glucose stimulated insulin secretion (p = 0.043). CONCLUSIONS Early detection of compromised beta-cell function could allow for implementation of nutritional and lifestyle interventions before progression to type 2 diabetes.
Collapse
Affiliation(s)
- Aoife M. Curran
- Institute of Food and Health, UCD School of Agriculture and Food Science, University College Dublin, Belfield, Ireland University College Dublin, Dublin, Republic of Ireland
- Food for Health Ireland (FHI), University College Dublin, Belfield, Ireland University College Dublin, Dublin, Republic of Ireland
| | - Marie Pier Scott-Boyer
- The Microsoft Research – University of Trento Centre for Computational and Systems Biology (COSBI), Rovereto, Italy
| | - Jim Kaput
- Nestlé Institute of Health Sciences, Lausanne, Switzerland
| | - Miriam F. Ryan
- Institute of Food and Health, UCD School of Agriculture and Food Science, University College Dublin, Belfield, Ireland University College Dublin, Dublin, Republic of Ireland
| | - Elaine Drummond
- Institute of Food and Health, UCD School of Agriculture and Food Science, University College Dublin, Belfield, Ireland University College Dublin, Dublin, Republic of Ireland
- Food for Health Ireland (FHI), University College Dublin, Belfield, Ireland University College Dublin, Dublin, Republic of Ireland
| | - Eileen R. Gibney
- Institute of Food and Health, UCD School of Agriculture and Food Science, University College Dublin, Belfield, Ireland University College Dublin, Dublin, Republic of Ireland
- Food for Health Ireland (FHI), University College Dublin, Belfield, Ireland University College Dublin, Dublin, Republic of Ireland
| | - Michael J. Gibney
- Institute of Food and Health, UCD School of Agriculture and Food Science, University College Dublin, Belfield, Ireland University College Dublin, Dublin, Republic of Ireland
- Food for Health Ireland (FHI), University College Dublin, Belfield, Ireland University College Dublin, Dublin, Republic of Ireland
| | - Helen M. Roche
- Institute of Food and Health, UCD School of Agriculture and Food Science, University College Dublin, Belfield, Ireland University College Dublin, Dublin, Republic of Ireland
- Food for Health Ireland (FHI), University College Dublin, Belfield, Ireland University College Dublin, Dublin, Republic of Ireland
- Nutrigenomics Research Group, UCD Conway Institute of Biomolecular and Biomedical Research and UCD Institute of Food and Health, School of Public Health, Physiotherapy and Sports Science, University College Dublin, Belfield, Dublin, Republic of Ireland
| | - Lorraine Brennan
- Institute of Food and Health, UCD School of Agriculture and Food Science, University College Dublin, Belfield, Ireland University College Dublin, Dublin, Republic of Ireland
- Food for Health Ireland (FHI), University College Dublin, Belfield, Ireland University College Dublin, Dublin, Republic of Ireland
- * E-mail:
| |
Collapse
|
24
|
Long-term moderate exercise enhances specific proteins that constitute neurotrophin signaling pathway: A TMT-based quantitative proteomic analysis of rat plasma. J Proteomics 2018; 185:39-50. [PMID: 29953961 DOI: 10.1016/j.jprot.2018.06.017] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Revised: 06/15/2018] [Accepted: 06/17/2018] [Indexed: 12/15/2022]
Abstract
Physical exercise has been reported to increase neurotrophin in brain tissues as hippocampus as well as increased neurotrophic level peripherally in blood plasma and might have an effect on/or affect molecular processes of energy metabolism (and homeostasis). In this study, using quantitative proteomic analysis, we obtained a plasma protein profile from the rat with long-term moderate exercise. A total of 752 proteins were identified in the plasma. Among them, 54 proteins were significant up-regulated and 47 proteins were down-regulated in the plasma of exercise group compared with the control group. Bioinformatic analyses showed that these altered proteins are widely involved in multiple biological processes, molecular functions and cellular components, which connect with 11 signaling pathways. Interestingly, 5 up-regulated proteins Rap1b, PTPN11, ARHGDIA, Cdc42 and YWHAE, confirmed by Western blots, are involved in the neurotrophin signaling pathway which shows the lowest P value among the identified pathways. Further analyses showed that the 5 neurotrophin-signaling-pathway-related proteins participate in two important protein-protein interaction networks associated to cell survival and apoptosis, axonal development, synapse formation and plasticity. This study provides an exercise-induced plasma protein profile, suggesting that long-term exercise enhances the proteins involved in neurotrophin signaling pathway which may contribute to health benefit. SIGNIFICANCE Physical activity contributes to myriad benefits on body health across the lifespan. The changes in plasma proteins after chronic moderate exercise may be used as biomarkers for health and may also play important roles in increase of cardiovascular fitness, enhancement of immune competence, prevention of obesity, decrease of risk for neurological disorders, cancer, stroke, diabetes and other metabolic disorders. Using a TMT-based proteomic method, this study identified 101 altered proteins in the plasma of rats after long-term moderate treadmill running, which may provide novel biomarkers for further investigation of the underlying mechanism of physical exercise. We confirmed that exercise enhances 5 proteins of the neurotrophin signaling pathway that may contribute to health benefits.
Collapse
|
25
|
Lista S, Zetterberg H, O'Bryant SE, Blennow K, Hampel H. Evolving Relevance of Neuroproteomics in Alzheimer's Disease. Methods Mol Biol 2018; 1598:101-115. [PMID: 28508359 DOI: 10.1007/978-1-4939-6952-4_5] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
Abstract
Substantial progress in the understanding of the biology of Alzheimer's disease (AD) has been achieved over the past decades. The early detection and diagnosis of AD and other age-related neurodegenerative diseases, however, remain a challenging scientific frontier. Therefore, the comprehensive discovery (relating to all individual, converging or diverging biochemical disease mechanisms), development, validation, and qualification of standardized biological markers with diagnostic and prognostic functions with a precise performance profile regarding specificity, sensitivity, and positive and negative predictive value are warranted.Methodological innovations in the area of exploratory high-throughput technologies, such as sequencing, microarrays, and mass spectrometry-based analyses of proteins/peptides, have led to the generation of large global molecular datasets from a multiplicity of biological systems, such as biological fluids, cells, tissues, and organs. Such methodological progress has shifted the attention to the execution of hypothesis-independent comprehensive exploratory analyses (opposed to the classical hypothesis-driven candidate approach), with the aim of fully understanding the biological systems in physiology and disease as a whole. The systems biology paradigm integrates experimental biology with accurate and rigorous computational modelling to describe and foresee the dynamic features of biological systems. The use of dynamically evolving technological platforms, including mass spectrometry, in the area of proteomics has enabled to rush the process of biomarker discovery and validation for refining significantly the diagnosis of AD. Currently, proteomics-which is part of the systems biology paradigm-is designated as one of the dominant matured sciences needed for the effective exploratory discovery of prospective biomarker candidates expected to play an effective role in aiding the early detection, diagnosis, prognosis, and therapy development in AD.
Collapse
Affiliation(s)
- Simone Lista
- AXA Research Fund & UPMC Chair, Paris, France. .,Sorbonne Universités, Université Pierre et Marie Curie (UPMC) Paris 06, Inserm, CNRS, Institut du cerveau et dela moelle (ICM), Département de Neurologie, Institut de la Mémoire et de la Maladie d'Alzheimer (IM2A), HôpitalPitié-Salpêtrière, Boulevard de l'hôpital, F-75013, Paris, France.
| | - Henrik Zetterberg
- Clinical Neurochemistry Laboratory, Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden.,Department of Molecular Neuroscience, UCL Institute of Neurology, London, UK
| | - Sid E O'Bryant
- Institute for Healthy Aging, University of North Texas Health Science Center, Fort Worth, TX, USA
| | - Kaj Blennow
- Clinical Neurochemistry Laboratory, Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden.,The Torsten Söderberg Professorship in Medicine at the Royal Swedish Academy of Sciences, Stockholm, Sweden
| | - Harald Hampel
- AXA Research Fund & UPMC Chair, Paris, France.,Sorbonne Universités, Université Pierre et Marie Curie, Paris 06, Institut de la Mémoire et de la Maladie d'Alzheimer (IM2A) & Institut du Cerveau et de la Moelle Épinière (ICM), Paris, France; Département de Neurologie, Hôpital de la Pitié-Salpêtrière, Boulevard de l'hôpital, F-75013, Paris, France
| |
Collapse
|
26
|
Wilson EN, Abela AR, Do Carmo S, Allard S, Marks AR, Welikovitch LA, Ducatenzeiler A, Chudasama Y, Cuello AC. Intraneuronal Amyloid Beta Accumulation Disrupts Hippocampal CRTC1-Dependent Gene Expression and Cognitive Function in a Rat Model of Alzheimer Disease. ACTA ACUST UNITED AC 2018; 27:1501-1511. [PMID: 26759481 DOI: 10.1093/cercor/bhv332] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Abstract
In Alzheimer disease (AD), the accumulation of amyloid beta (Aβ) begins decades before cognitive symptoms and progresses from intraneuronal material to extracellular plaques. To date, however, the precise mechanism by which the early buildup of Aβ peptides leads to cognitive dysfunction remains unknown. Here, we investigate the impact of the early Aβ accumulation on temporal and frontal lobe dysfunction. We compared the performance of McGill-R-Thy1-APP transgenic AD rats with wild-type littermate controls on a visual discrimination task using a touchscreen operant platform. Subsequently, we conducted studies to establish the biochemical and molecular basis for the behavioral alterations. It was found that the presence of intraneuronal Aβ caused a severe associative learning deficit in the AD rats. This coincided with reduced nuclear translocation and genomic occupancy of the CREB co-activator, CRTC1, and decreased production of synaptic plasticity-associated transcripts Arc, c-fos, Egr1, and Bdnf. Thus, blockade of CRTC1-dependent gene expression in the early, preplaque phase of AD-like pathology provides a molecular basis for the cognitive deficits that figure so prominently in early AD.
Collapse
Affiliation(s)
- Edward N Wilson
- Department of Pharmacology and Therapeutics, McGill University, Montreal, QCCanadaH3G 1Y6
| | - Andrew R Abela
- Department of Psychology, McGill University, Montreal, QCCanadaH3A 1B1
| | - Sonia Do Carmo
- Department of Pharmacology and Therapeutics, McGill University, Montreal, QCCanadaH3G 1Y6
| | - Simon Allard
- Department of Pharmacology and Therapeutics, McGill University, Montreal, QCCanadaH3G 1Y6
| | - Adam R Marks
- Department of Pharmacology and Therapeutics, McGill University, Montreal, QCCanadaH3G 1Y6
| | - Lindsay A Welikovitch
- Department of Pharmacology and Therapeutics, McGill University, Montreal, QCCanadaH3G 1Y6
| | - Adriana Ducatenzeiler
- Department of Pharmacology and Therapeutics, McGill University, Montreal, QCCanadaH3G 1Y6
| | - Yogita Chudasama
- Department of Psychology, McGill University, Montreal, QC Canada H3A 1B1.,Current address: National Institute of Mental Health, National Institutes of Health, Bethesda, MD 20892, USA
| | - A Claudio Cuello
- Department of Pharmacology and Therapeutics, McGill University, Montreal, QC Canada H3G 1Y6.,Department of Anatomy and Cell Biology.,Department of Neurology and Neurosurgery, McGill University, Montreal, QC Canada
| |
Collapse
|
27
|
Stivaros S, Garg S, Tziraki M, Cai Y, Thomas O, Mellor J, Morris AA, Jim C, Szumanska-Ryt K, Parkes LM, Haroon HA, Montaldi D, Webb N, Keane J, Castellanos FX, Silva AJ, Huson S, Williams S, Gareth Evans D, Emsley R, Green J. Randomised controlled trial of simvastatin treatment for autism in young children with neurofibromatosis type 1 (SANTA). Mol Autism 2018; 9:12. [PMID: 29484149 PMCID: PMC5824534 DOI: 10.1186/s13229-018-0190-z] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Accepted: 01/12/2018] [Indexed: 11/24/2022] Open
Abstract
Background Neurofibromatosis 1 (NF1) is a monogenic model for syndromic autism. Statins rescue the social and cognitive phenotype in animal knockout models, but translational trials with subjects > 8 years using cognition/behaviour outcomes have shown mixed results. This trial breaks new ground by studying statin effects for the first time in younger children with NF1 and co-morbid autism and by using multiparametric imaging outcomes. Methods A single-site triple-blind RCT of simvastatin vs. placebo was done. Assessment (baseline and 12-week endpoint) included peripheral MAPK assay, awake magnetic resonance imaging spectroscopy (MRS; GABA and glutamate+glutamine (Glx)), arterial spin labelling (ASL), apparent diffusion coefficient (ADC), resting state functional MRI, and autism behavioural outcomes (Aberrant Behaviour Checklist and Clinical Global Impression). Results Thirty subjects had a mean age of 8.1 years (SD 1.8). Simvastatin was well tolerated. The amount of imaging data varied by test. Simvastatin treatment was associated with (i) increased frontal white matter MRS GABA (t(12) = - 2.12, p = .055), GABA/Glx ratio (t(12) = - 2.78, p = .016), and reduced grey nuclei Glx (ANCOVA p < 0.05, Mann-Whitney p < 0.01); (ii) increased ASL perfusion in ventral diencephalon (Mann-Whitney p < 0.01); and (iii) decreased ADC in cingulate gyrus (Mann-Whitney p < 0.01). Machine-learning classification of imaging outcomes achieved 79% (p < .05) accuracy differentiating groups at endpoint against chance level (64%, p = 0.25) at baseline. Three of 12 (25%) simvastatin cases compared to none in placebo met 'clinical responder' criteria for behavioural outcome. Conclusions We show feasibility of peripheral MAPK assay and autism symptom measurement, but the study was not powered to test effectiveness. Multiparametric imaging suggests possible simvastatin effects in brain areas previously associated with NF1 pathophysiology and the social brain network. Trial registration EU Clinical Trial Register (EudraCT) 2012-005742-38 (www.clinicaltrialsregister.eu).
Collapse
Affiliation(s)
- Stavros Stivaros
- Academic Unit of Paediatric Radiology, Royal Manchester Children’s Hospital, Central Manchester University Hospitals NHS Foundation Trust, Manchester Academic Health Sciences Centre, Manchester, UK
- Division of Informatics, Imaging and Data Sciences, School of Health Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - Shruti Garg
- Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester University NHS Foundation Trust, Greater Manchester Mental Health NHS Foundation Trust, Room 3.311, Jean McFarlane Building, Oxford Road, Manchester, M13 9PL UK
| | - Maria Tziraki
- Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - Ying Cai
- Departments of Neurobiology, Psychiatry and Biobehavioral Sciences and Psychology, Integrative Center for Learning and Memory, Brain Research Institute, Brain Research Institute, University of California, California, LA 90095 USA
| | - Owen Thomas
- Academic Unit of Radiology, Salford Royal Foundation NHS Trust, Manchester Academic Health Sciences Centre, Manchester, UK
| | - Joseph Mellor
- Computer Science, University of Manchester, Manchester, UK
| | - Andrew A. Morris
- Manchester University NHS Foundation Trust, Manchester Academic Health Sciences Centre, Manchester, UK
| | - Carly Jim
- Manchester Metropolitan University, Manchester, UK
| | - Karolina Szumanska-Ryt
- Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - Laura M Parkes
- Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - Hamied A. Haroon
- Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - Daniela Montaldi
- Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - Nicholas Webb
- Department of Paediatric Nephrology, Royal Manchester Children’s Hospital, Manchester University NHS Foundation Trust, Academic Health Sciences Centre, Manchester, UK
| | - John Keane
- Computer Science, University of Manchester, Manchester, UK
| | - Francisco X. Castellanos
- Hassenfeld Children’s Hospital at NYU Langone, Nathan S. Kline Institute for Psychiatric Research, New York, USA
| | - Alcino J. Silva
- Departments of Neurobiology, Psychiatry and Biobehavioral Sciences and Psychology, Integrative Center for Learning and Memory, Brain Research Institute, Brain Research Institute, University of California, California, LA 90095 USA
| | - Sue Huson
- Manchester Centre for Genomic Medicine, St Mary’s Hospital, Manchester University NHS Foundation Trust, Academic Health Sciences Centre, Manchester, UK
| | - Stephen Williams
- Division of Informatics, Imaging and Data Sciences, School of Health Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - D. Gareth Evans
- Manchester Centre for Genomic Medicine, St Mary’s Hospital, Manchester University NHS Foundation Trust, Academic Health Sciences Centre, Manchester, UK
| | - Richard Emsley
- Centre for Biostatistics, School of Health Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Jonathan Green
- Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester University NHS Foundation Trust, Greater Manchester Mental Health NHS Foundation Trust, Room 3.311, Jean McFarlane Building, Oxford Road, Manchester, M13 9PL UK
| |
Collapse
|
28
|
Kiddle SJ, Voyle N, Dobson RJB. A Blood Test for Alzheimer's Disease: Progress, Challenges, and Recommendations. J Alzheimers Dis 2018; 64:S289-S297. [PMID: 29614671 PMCID: PMC6010156 DOI: 10.3233/jad-179904] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Ever since the discovery of APOEɛ4 around 25 years ago, researchers have been excited about the potential of a blood test for Alzheimer's disease (AD). Since then researchers have looked for genetic, protein, metabolite, and/or gene expression markers of AD and related phenotypes. However, no blood test for AD is yet being used in the clinical setting. We first review the trends and challenges in AD blood biomarker research, before giving our personal recommendations to help researchers overcome these challenges. While some degree of consistency and replication has been seen across independent studies, several high-profile studies have seemingly failed to replicate. Partly due to academic incentives, there is a reluctance in the field to report predictive ability, to publish negative findings, and to independently replicate the work of others. If this can be addressed, then we will know sooner whether a blood test for AD or related phenotypes with clinical utility can be developed.
Collapse
Affiliation(s)
- Steven J. Kiddle
- MRC Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, UK, SE5 8AF
- MRC Biostatistics Unit, Cambridge Biomedical Campus, Cambridge Institute of Public Health, Forvie Site, Robinson Way, Cambridge CB2 0SR, UK
| | - Nicola Voyle
- MRC Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, UK, SE5 8AF
| | - Richard JB Dobson
- MRC Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, UK, SE5 8AF
- NIHR Biomedical Research Centre for Mental Health and Biomedical Research Unit for Dementia at South London and Maudsley NHS Foundation, London, UK, SE5 8AF
- Farr Institute of Health Informatics Research, UCL Institute of Health Informatics, University College London, London WC1E 6BT, UK
| |
Collapse
|
29
|
Corbo C, Molinaro R, Tabatabaei M, Farokhzad OC, Mahmoudi M. Personalized protein corona on nanoparticles and its clinical implications. Biomater Sci 2017; 5:378-387. [PMID: 28133653 PMCID: PMC5592724 DOI: 10.1039/c6bm00921b] [Citation(s) in RCA: 212] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
It is now well understood that once in contact with biological fluids, nanoscale objects lose their original identity and acquire a new biological character, referred to as a protein corona. The protein corona changes many of the physicochemical properties of nanoparticles, including size, surface charge, and aggregation state. These changes, in turn, affect the biological fate of nanoparticles, including their pharmacokinetics, biodistribution, and therapeutic efficacy. It is progressively being accepted that even slight variations in the composition of a protein source (e.g., plasma and serum) can substantially change the composition of the corona formed on the surface of the exact same nanoparticles. Recently it has been shown that the protein corona is strongly affected by the patient's specific disease. Therefore, the same nanomaterial incubated with plasma proteins of patients with different pathologies adsorb protein coronas with different compositions, giving rise to the concept of personalized protein corona. Herein, we review this concept along with recent advances on the topic, with a particular focus on clinical relevance.
Collapse
Affiliation(s)
- Claudia Corbo
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| | - Roberto Molinaro
- Center for Biomimetic Medicine, Houston Methodist Research Institute, Houston, TX, USA
| | - Mateen Tabatabaei
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| | - Omid C Farokhzad
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA. and King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Morteza Mahmoudi
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA. and Nanotechnology Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
30
|
Counts SE, Ikonomovic MD, Mercado N, Vega IE, Mufson EJ. Biomarkers for the Early Detection and Progression of Alzheimer's Disease. Neurotherapeutics 2017; 14:35-53. [PMID: 27738903 PMCID: PMC5233625 DOI: 10.1007/s13311-016-0481-z] [Citation(s) in RCA: 122] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The recent failures of potential disease-modifying drugs for Alzheimer's disease (AD) may reflect the fact that the enrolled participants in clinical trials are already too advanced to derive a clinical benefit. Thus, well-validated biomarkers for the early detection and accurate diagnosis of the preclinical stages of AD will be crucial for therapeutic advancement. The combinatorial use of biomarkers derived from biological fluids, such as cerebrospinal fluid (CSF), with advanced molecular imaging and neuropsychological testing may eventually achieve the diagnostic sensitivity and specificity necessary to identify people in the earliest stages of the disease when drug modification is most likely possible. In this regard, positive amyloid or tau tracer retention on positron emission tomography imaging, low CSF concentrations of the amyloid-β 1-42 peptide, high CSF concentrations in total tau and phospho-tau, mesial temporal lobe atrophy on magnetic resonance imaging, and temporoparietal/precuneus hypometabolism or hypoperfusion on 18F-fluorodeoxyglucose positron emission tomography have all emerged as biomarkers for the progression to AD. However, the ultimate AD biomarker panel will likely involve the inclusion of novel CSF and blood biomarkers more precisely associated with confirmed pathophysiologic mechanisms to improve its reliability for detecting preclinical AD. This review highlights advancements in biological fluid and imaging biomarkers that are moving the field towards achieving the goal of a preclinical detection of AD.
Collapse
Affiliation(s)
- Scott E Counts
- Department of Translational Science and Molecular Medicine, Michigan State University, Grand Rapids, MI, USA
- Department of Family Medicine, Michigan State University, Grand Rapids, MI, USA
- Hauenstein Neuroscience Center, Mercy Health Saint Mary's Hospital, Grand Rapids, MI, USA
| | - Milos D Ikonomovic
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
- Geriatric Research Education and Clinical Center, VA Pittsburgh Healthcare System, Pittsburgh, PA, USA
| | - Natosha Mercado
- Department of Translational Science and Molecular Medicine, Michigan State University, Grand Rapids, MI, USA
| | - Irving E Vega
- Department of Translational Science and Molecular Medicine, Michigan State University, Grand Rapids, MI, USA
| | - Elliott J Mufson
- Department of Neurobiology and Neurology, Barrow Neurological Institute, Phoenix, AZ, USA.
| |
Collapse
|
31
|
Paterson RW, Heywood WE, Heslegrave AJ, Magdalinou NK, Andreasson U, Sirka E, Bliss E, Slattery CF, Toombs J, Svensson J, Johansson P, Fox NC, Zetterberg H, Mills K, Schott JM. A targeted proteomic multiplex CSF assay identifies increased malate dehydrogenase and other neurodegenerative biomarkers in individuals with Alzheimer's disease pathology. Transl Psychiatry 2016; 6:e952. [PMID: 27845782 PMCID: PMC5314115 DOI: 10.1038/tp.2016.194] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2016] [Accepted: 07/31/2016] [Indexed: 12/12/2022] Open
Abstract
Alzheimer's disease (AD) is the most common cause of dementia. Biomarkers are required to identify individuals in the preclinical phase, explain phenotypic diversity, measure progression and estimate prognosis. The development of assays to validate candidate biomarkers is costly and time-consuming. Targeted proteomics is an attractive means of quantifying novel proteins in cerebrospinal and other fluids, and has potential to help overcome this bottleneck in biomarker development. We used a previously validated multiplexed 10-min, targeted proteomic assay to assess 54 candidate cerebrospinal fluid (CSF) biomarkers in two independent cohorts comprising individuals with neurodegenerative dementias and healthy controls. Individuals were classified as 'AD' or 'non-AD' on the basis of their CSF T-tau and amyloid Aβ1-42 profile measured using enzyme-linked immunosorbent assay; biomarkers of interest were compared using univariate and multivariate analyses. In all, 35/31 individuals in Cohort 1 and 46/36 in Cohort 2 fulfilled criteria for AD/non-AD profile CSF, respectively. After adjustment for multiple comparisons, five proteins were elevated significantly in AD CSF compared with non-AD CSF in both cohorts: malate dehydrogenase; total APOE; chitinase-3-like protein 1 (YKL-40); osteopontin and cystatin C. In an independent multivariate orthogonal projection to latent structures discriminant analysis (OPLS-DA), these proteins were also identified as major contributors to the separation between AD and non-AD in both cohorts. Independent of CSF Aβ1-42 and tau, a combination of these biomarkers differentiated AD and non-AD with an area under curve (AUC)=0.88. This targeted proteomic multiple reaction monitoring (MRM)-based assay can simultaneously and rapidly measure multiple candidate CSF biomarkers. Applying this technique to AD we demonstrate differences in proteins involved in glucose metabolism and neuroinflammation that collectively have potential clinical diagnostic utility.
Collapse
Affiliation(s)
- R W Paterson
- Dementia Research Centre, Institute of Neurology, University College London, London, UK
| | - W E Heywood
- Centre for Translational Omics, Genetics and Genomic Medicine Programme, Institute of Child Health, University College London, London, UK
| | - A J Heslegrave
- Department of Molecular Neuroscience, Institute of Neurology, University College London, London, UK
| | - N K Magdalinou
- Lila Weston Institute, University College London Institute of Neurology, London, UK
| | - U Andreasson
- Clinical Neurochemistry Laboratory, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at University of Gothenburg, Sahlgrenska University Hospital, Mölndal, Sweden
| | - E Sirka
- Centre for Translational Omics, Genetics and Genomic Medicine Programme, Institute of Child Health, University College London, London, UK
| | - E Bliss
- Centre for Translational Omics, Genetics and Genomic Medicine Programme, Institute of Child Health, University College London, London, UK
| | - C F Slattery
- Dementia Research Centre, Institute of Neurology, University College London, London, UK
| | - J Toombs
- Department of Molecular Neuroscience, Institute of Neurology, University College London, London, UK
| | - J Svensson
- Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Endocrinology, Skaraborg Central Hospital, Skövde, Sweden
| | - P Johansson
- Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Neuropsychiatry, Skaraborg Central Hospital, Falköping, Sweden
| | - N C Fox
- Dementia Research Centre, Institute of Neurology, University College London, London, UK
| | - H Zetterberg
- Department of Molecular Neuroscience, Institute of Neurology, University College London, London, UK
- Clinical Neurochemistry Laboratory, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at University of Gothenburg, Sahlgrenska University Hospital, Mölndal, Sweden
| | - K Mills
- Dementia Research Centre, Institute of Neurology, University College London, London, UK
- Centre for Translational Omics, Genetics and Genomic Medicine Programme, Institute of Child Health, University College London, London, UK
| | - J M Schott
- Dementia Research Centre, Institute of Neurology, University College London, London, UK
| |
Collapse
|
32
|
Proteomic study of hepatocellular carcinoma using a novel modified aptamer-based array (SOMAscan™) platform. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2016; 1865:434-443. [PMID: 27663888 DOI: 10.1016/j.bbapap.2016.09.011] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 06/24/2016] [Revised: 08/17/2016] [Accepted: 09/19/2016] [Indexed: 12/12/2022]
Abstract
Vascular invasion is a pathological hallmark of hepatocellular carcinoma (HCC), associated with poor prognosis; it is strongly related to the early recurrence and poor survival after curative resection. In order to determine the proteomic backgrounds of HCC carcinogenesis and vascular invasion, we employed a novel modified aptamer-based array (SOMAscan) platform. SOMAscan is based on the Slow Off-rate Modified Aptamers (SOMAmers), which rely on the natural 3D folding of single-stranded DNA-based protein affinity reagents. Currently, the expression level of 1129 proteins can be assessed quantitatively. Correlation matrix analysis showed that the overall proteomic features captured by SOMAscan differ between tumor and non-tumor tissues. Non-tumor tissues were shown to have more homogeneous proteome backgrounds than tumor tissues. A comparative study identified 68 proteins with differential expression between tumor and non-tumor tissues, together with eight proteins associated with vascular invasion. Gene Ontology analysis showed that the extracellular space and extracellular region proteins were predominantly detected. Network analysis revealed the linkage of seven proteins, AKT1, MDM2, PTEN, FGF1, MAPK8, PRKCB, and FN1, which were categorized as the components of "Pathways in cancer" in pathway analysis. The results of SOMAscan analysis were not concordant with those obtained by western blotting; only the determined FN1 levels were concordant between the two platforms. We demonstrated that the proteome captured by SOMAscan includes the proteins relevant to carcinogenesis and vascular invasion in HCC. The identified proteins may serve as candidates for the future studies of disease mechanisms and clinical applications.
Collapse
|
33
|
Liebert AD, Chow RT, Bicknell BT, Varigos E. Neuroprotective Effects Against POCD by Photobiomodulation: Evidence from Assembly/Disassembly of the Cytoskeleton. J Exp Neurosci 2016; 10:1-19. [PMID: 26848276 PMCID: PMC4737522 DOI: 10.4137/jen.s33444] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Revised: 12/09/2015] [Accepted: 12/15/2015] [Indexed: 02/07/2023] Open
Abstract
Postoperative cognitive dysfunction (POCD) is a decline in memory following anaesthesia and surgery in elderly patients. While often reversible, it consumes medical resources, compromises patient well-being, and possibly accelerates progression into Alzheimer's disease. Anesthetics have been implicated in POCD, as has neuroinflammation, as indicated by cytokine inflammatory markers. Photobiomodulation (PBM) is an effective treatment for a number of conditions, including inflammation. PBM also has a direct effect on microtubule disassembly in neurons with the formation of small, reversible varicosities, which cause neural blockade and alleviation of pain symptoms. This mimics endogenously formed varicosities that are neuroprotective against damage, toxins, and the formation of larger, destructive varicosities and focal swellings. It is proposed that PBM may be effective as a preconditioning treatment against POCD; similar to the PBM treatment, protective and abscopal effects that have been demonstrated in experimental models of macular degeneration, neurological, and cardiac conditions.
Collapse
Affiliation(s)
| | - Roberta T. Chow
- Brain and Mind Institute, University of Sydney, Sydney, NSW, Australia
| | | | | |
Collapse
|