1
|
Beura SK, Panigrahi AR, Yadav P, Kulkarni PP, Lakhanpal V, Singh B, Singh SK. Role of Thrombosis in Neurodegenerative Diseases: An Intricate Mechanism of Neurovascular Complications. Mol Neurobiol 2024:10.1007/s12035-024-04589-4. [PMID: 39482419 DOI: 10.1007/s12035-024-04589-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Accepted: 10/23/2024] [Indexed: 11/03/2024]
Abstract
Thrombosis, the formation of blood clots in arteries or veins, poses a significant health risk by disrupting the blood flow. It can potentially lead to major cardiovascular complications such as acute myocardial infarction or ischemic stroke (arterial thrombosis) and deep vein thrombosis or pulmonary embolism (venous thrombosis). Nevertheless, over the course of several decades, researchers have observed an association between different cardiovascular events and neurodegenerative diseases, which progressively harm and impair parts of the nervous system, particularly the brain. Furthermore, thrombotic complications have been identified in numerous clinical instances of neurodegenerative diseases, particularly Alzheimer's disease, Parkinson's disease, multiple sclerosis, and Huntington's disease. Substantial research indicates that endothelial dysfunction, vascular inflammation, coagulation abnormalities, and platelet hyperactivation are commonly observed in these conditions, collectively contributing to an increased risk of thrombosis. Thrombosis can, in turn, contribute to the onset, pathogenesis, and severity of these neurological disorders. Hence, this concise review comprehensively explores the correlation between cardiovascular diseases and neurodegenerative diseases, elucidating the cellular and molecular mechanisms of thrombosis in these neurodegenerative diseases. Additionally, a detailed discussion is provided on the commonly employed antithrombotic medications in the context of these neuronal diseases.
Collapse
Affiliation(s)
- Samir Kumar Beura
- Department of Zoology, School of Basic Sciences, Central University of Punjab, Bathinda, Punjab, India, 151401
| | | | - Pooja Yadav
- Department of Zoology, School of Basic Sciences, Central University of Punjab, Bathinda, Punjab, India, 151401
| | - Paresh P Kulkarni
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44195, USA
| | - Vikas Lakhanpal
- Department of Neurology, All India Institute of Medical Sciences, Bathinda, Punjab, India, 151001
| | - Bhupinder Singh
- Department of Cardiology, All India Institute of Medical Sciences, Bathinda, Punjab, India, 151001
| | - Sunil Kumar Singh
- Department of Zoology, School of Basic Sciences, Central University of Punjab, Bathinda, Punjab, India, 151401.
- Department of Biochemistry, School of Basic Sciences, Central University of Punjab, Bathinda, Punjab, India, 151401.
| |
Collapse
|
2
|
Kell DB, Pretorius E. Proteomic Evidence for Amyloidogenic Cross-Seeding in Fibrinaloid Microclots. Int J Mol Sci 2024; 25:10809. [PMID: 39409138 PMCID: PMC11476703 DOI: 10.3390/ijms251910809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 10/01/2024] [Accepted: 10/03/2024] [Indexed: 10/20/2024] Open
Abstract
In classical amyloidoses, amyloid fibres form through the nucleation and accretion of protein monomers, with protofibrils and fibrils exhibiting a cross-β motif of parallel or antiparallel β-sheets oriented perpendicular to the fibre direction. These protofibrils and fibrils can intertwine to form mature amyloid fibres. Similar phenomena can occur in blood from individuals with circulating inflammatory molecules (and also some originating from viruses and bacteria). Such pathological clotting can result in an anomalous amyloid form termed fibrinaloid microclots. Previous proteomic analyses of these microclots have shown the presence of non-fibrin(ogen) proteins, suggesting a more complex mechanism than simple entrapment. We thus provide evidence against such a simple entrapment model, noting that clot pores are too large and centrifugation would have removed weakly bound proteins. Instead, we explore whether co-aggregation into amyloid fibres may involve axial (multiple proteins within the same fibril), lateral (single-protein fibrils contributing to a fibre), or both types of integration. Our analysis of proteomic data from fibrinaloid microclots in different diseases shows no significant quantitative overlap with the normal plasma proteome and no correlation between plasma protein abundance and their presence in fibrinaloid microclots. Notably, abundant plasma proteins like α-2-macroglobulin, fibronectin, and transthyretin are absent from microclots, while less abundant proteins such as adiponectin, periostin, and von Willebrand factor are well represented. Using bioinformatic tools, including AmyloGram and AnuPP, we found that proteins entrapped in fibrinaloid microclots exhibit high amyloidogenic tendencies, suggesting their integration as cross-β elements into amyloid structures. This integration likely contributes to the microclots' resistance to proteolysis. Our findings underscore the role of cross-seeding in fibrinaloid microclot formation and highlight the need for further investigation into their structural properties and implications in thrombotic and amyloid diseases. These insights provide a foundation for developing novel diagnostic and therapeutic strategies targeting amyloidogenic cross-seeding in blood clotting disorders.
Collapse
Affiliation(s)
- Douglas B. Kell
- Department of Biochemistry, Cell and Systems Biology, Institute of Systems, Molecular and Integrative Biology, Faculty of Health and Life Sciences, University of Liverpool, Crown St., Liverpool L69 7ZB, UK
- The Novo Nordisk Foundation Centre for Biosustainability, Building 220, Søltofts Plads 200, Technical University of Denmark, 2800 Kongens Lyngby, Denmark
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Private Bag X1 Matieland, Stellenbosch 7602, South Africa
| | - Etheresia Pretorius
- Department of Biochemistry, Cell and Systems Biology, Institute of Systems, Molecular and Integrative Biology, Faculty of Health and Life Sciences, University of Liverpool, Crown St., Liverpool L69 7ZB, UK
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Private Bag X1 Matieland, Stellenbosch 7602, South Africa
| |
Collapse
|
3
|
Balczon R, Lin MT, Voth S, Nelson AR, Schupp JC, Wagener BM, Pittet JF, Stevens T. Lung endothelium, tau, and amyloids in health and disease. Physiol Rev 2024; 104:533-587. [PMID: 37561137 PMCID: PMC11281824 DOI: 10.1152/physrev.00006.2023] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 06/26/2023] [Accepted: 08/04/2023] [Indexed: 08/11/2023] Open
Abstract
Lung endothelia in the arteries, capillaries, and veins are heterogeneous in structure and function. Lung capillaries in particular represent a unique vascular niche, with a thin yet highly restrictive alveolar-capillary barrier that optimizes gas exchange. Capillary endothelium surveys the blood while simultaneously interpreting cues initiated within the alveolus and communicated via immediately adjacent type I and type II epithelial cells, fibroblasts, and pericytes. This cell-cell communication is necessary to coordinate the immune response to lower respiratory tract infection. Recent discoveries identify an important role for the microtubule-associated protein tau that is expressed in lung capillary endothelia in the host-pathogen interaction. This endothelial tau stabilizes microtubules necessary for barrier integrity, yet infection drives production of cytotoxic tau variants that are released into the airways and circulation, where they contribute to end-organ dysfunction. Similarly, beta-amyloid is produced during infection. Beta-amyloid has antimicrobial activity, but during infection it can acquire cytotoxic activity that is deleterious to the host. The production and function of these cytotoxic tau and amyloid variants are the subject of this review. Lung-derived cytotoxic tau and amyloid variants are a recently discovered mechanism of end-organ dysfunction, including neurocognitive dysfunction, during and in the aftermath of infection.
Collapse
Affiliation(s)
- Ron Balczon
- Department of Biochemistry and Molecular Biology, University of South Alabama, Mobile, Alabama, United States
- Center for Lung Biology, University of South Alabama, Mobile, Alabama, United States
| | - Mike T Lin
- Department of Physiology and Cell Biology, University of South Alabama, Mobile, Alabama, United States
- Center for Lung Biology, University of South Alabama, Mobile, Alabama, United States
| | - Sarah Voth
- Department of Cell Biology and Physiology, Edward Via College of Osteopathic Medicine, Monroe, Louisiana, United States
| | - Amy R Nelson
- Department of Physiology and Cell Biology, University of South Alabama, Mobile, Alabama, United States
- Center for Lung Biology, University of South Alabama, Mobile, Alabama, United States
| | - Jonas C Schupp
- Pulmonary and Critical Care Medicine, Department of Internal Medicine, Yale University, New Haven, Connecticut, United States
- Department of Respiratory Medicine, Hannover Medical School, Hannover, Germany
- German Center for Lung Research (DZL), Hannover, Germany
| | - Brant M Wagener
- Department of Anesthesiology and Perioperative Medicine, University of Alabama-Birmingham, Birmingham, Alabama, United States
| | - Jean-Francois Pittet
- Department of Anesthesiology and Perioperative Medicine, University of Alabama-Birmingham, Birmingham, Alabama, United States
| | - Troy Stevens
- Department of Physiology and Cell Biology, University of South Alabama, Mobile, Alabama, United States
- Department of Internal Medicine, University of South Alabama, Mobile, Alabama, United States
- Center for Lung Biology, University of South Alabama, Mobile, Alabama, United States
| |
Collapse
|
4
|
Wang X, Li H, Sheng Y, He B, Liu Z, Li W, Yu S, Wang J, Zhang Y, Chen J, Qin L, Meng X. The function of sphingolipids in different pathogenesis of Alzheimer's disease: A comprehensive review. Biomed Pharmacother 2024; 171:116071. [PMID: 38183741 DOI: 10.1016/j.biopha.2023.116071] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2023] [Revised: 12/11/2023] [Accepted: 12/21/2023] [Indexed: 01/08/2024] Open
Abstract
Sphingolipids (SPLs) represent a highly diverse and structurally complex lipid class. The discussion of SPL metabolism-related issues is of importance in understanding the neuropathological progression of Alzheimer's disease (AD). AD is characterized by the accumulation of extracellular deposits of the amyloid β-peptide (Aβ) and intraneuronal aggregates of the microtubule-associated protein tau. Critical roles of Aβ oligomer deposited and ganglioside GM1 could be formed as "seed" from insoluble GAβ polymer in initiating the pathogenic process, while tau might also mediate SPLs and their toxicity. The interaction between ceramide and α-Synuclein (α-Syn) accelerates the aggregation of ferroptosis and exacerbates the pathogenesis of AD. For instance, reducing the levels of SPLs can mitigate α-Syn accumulation and inhibit AD progression. Meanwhile, loss of SPLs may inhibit the expression of APOE4 and confer protection against AD, while the loss of APOE4 expression also disrupts SPLs homeostasis. Moreover, the heightened activation of sphingomyelinase promotes the ferroptosis signaling pathway, leading to exacerbated AD symptoms. Ferroptosis plays a vital role in the pathological progression of AD by influencing Aβ, tau, APOE, and α-Syn. Conversely, the development of AD also exacerbates the manifestation of ferroptosis and SPLs. We are compiling the emerging techniques (Derivatization and IM-MS) of sphingolipidomics, to overcome the challenges of AD diagnosis and treatment. In this review, we examined the intricate neuro-mechanistic interactions between SPLs and Aβ, tau, α-Syn, APOE, and ferroptosis, mediating the onset of AD. Furthermore, our findings highlight the potential of targeting SPLs as underexplored avenue for devising innovative therapeutic strategies against AD.
Collapse
Affiliation(s)
- Xinyi Wang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, 548 Binwen Road, Binjiang District, Hangzhou 310053, Zhejiang Province, PR China
| | - Huaqiang Li
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, 548 Binwen Road, Binjiang District, Hangzhou 310053, Zhejiang Province, PR China
| | - Yunjie Sheng
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, 548 Binwen Road, Binjiang District, Hangzhou 310053, Zhejiang Province, PR China
| | - Bingqian He
- Academy of Chinese Medical Science, Zhejiang Chinese Medical University, 548 Binwen Road, Binjiang District, Hangzhou 310053, Zhejiang Province, PR China
| | - Zeying Liu
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, 548 Binwen Road, Binjiang District, Hangzhou 310053, Zhejiang Province, PR China
| | - Wanli Li
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, 548 Binwen Road, Binjiang District, Hangzhou 310053, Zhejiang Province, PR China
| | - Shujie Yu
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, 548 Binwen Road, Binjiang District, Hangzhou 310053, Zhejiang Province, PR China
| | - Jiajing Wang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, 548 Binwen Road, Binjiang District, Hangzhou 310053, Zhejiang Province, PR China
| | - Yixin Zhang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, 548 Binwen Road, Binjiang District, Hangzhou 310053, Zhejiang Province, PR China
| | - Jianyu Chen
- Fujian University of Traditional Chinese Medicine, School of Pharmacy, Fuzhou, Fujian 350122, PR China.
| | - Luping Qin
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, 548 Binwen Road, Binjiang District, Hangzhou 310053, Zhejiang Province, PR China.
| | - Xiongyu Meng
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, 548 Binwen Road, Binjiang District, Hangzhou 310053, Zhejiang Province, PR China.
| |
Collapse
|
5
|
Ayyubova G. APOE4 is a Risk Factor and Potential Therapeutic Target for Alzheimer's Disease. CNS & NEUROLOGICAL DISORDERS DRUG TARGETS 2024; 23:342-352. [PMID: 36872358 DOI: 10.2174/1871527322666230303114425] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 12/20/2022] [Accepted: 01/12/2023] [Indexed: 03/07/2023]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disease, the main pathological hallmark of which is the loss of neurons, resulting in cognitive and memory impairments. Sporadic late-onset AD is a prevalent form of the disease and the apolipoprotein E4 (APOE4) genotype is the strongest predictor of the disease development. The structural variations of APOE isoforms affect their roles in synaptic maintenance, lipid trafficking, energy metabolism, inflammatory response, and BBB integrity. In the context of AD, APOE isoforms variously control the key pathological elements of the disease, including Aβ plaque formation, tau aggregation, and neuroinflammation. Taking into consideration the limited number of therapy choices that can alleviate symptoms and have little impact on the AD etiology and progression to date, the precise research strategies guided by apolipoprotein E (APOE) polymorphisms are required to assess the potential risk of age-related cognitive decline in people carrying APOE4 genotype. In this review, we summarize the evidence implicating the significance of APOE isoforms on brain functions in health and pathology with the aim to identify the possible targets that should be addressed to prevent AD manifestation in individuals with the APOE4 genotype and to explore proper treatment strategies.
Collapse
Affiliation(s)
- Gunel Ayyubova
- Department of Cytology, Embryology and Histology, Azerbaijan Medical University, Baku, Azerbaijan
| |
Collapse
|
6
|
Grobler C, van Tongeren M, Gettemans J, Kell DB, Pretorius E. Alzheimer's Disease: A Systems View Provides a Unifying Explanation of Its Development. J Alzheimers Dis 2023; 91:43-70. [PMID: 36442193 DOI: 10.3233/jad-220720] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Alzheimer's disease (AD) is a debilitating neurodegenerative disorder affecting 50 million people globally. It is characterized by the presence of extracellular senile plaques and intracellular neurofibrillary tangles, consisting of amyloid-β and hyperphosphorylated tau proteins, respectively. Despite global research efforts, there is currently no cure available, due in part to an incomplete understanding of the disease pathogenesis. Numerous possible mechanisms, or hypotheses, explaining the origins of sporadic or late-onset AD have been proposed, including the amyloid-β, inflammatory, vascular, and infectious hypotheses. However, despite ample evidence, the failure of multiple trial drugs at the clinical stage illuminates the possible pitfalls of these hypotheses. Systems biology is a strategy which aims to elucidate the interactions between parts of a whole. Using this approach, the current paper shows how the four previously mentioned hypotheses of AD pathogenesis can be intricately connected. This approach allows for seemingly contradictory evidence to be unified in a system-focused explanation of sporadic AD development. Within this view, it is seen that infectious agents, such as P. gingivalis, may play a central role. The data presented here shows that when present, P. gingivalis or its virulence factors, such as gingipains, may induce or exacerbate pathologies underlying sporadic AD. This evidence supports the view that infectious agents, and specifically P. gingivalis, may be suitable treatment targets in AD.
Collapse
Affiliation(s)
- Corlia Grobler
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Stellenbosch, South Africa
| | - Marvi van Tongeren
- Department of Biomolecular Medicine, Faculty of Medicine & Health Sciences, Ghent University, Ghent, Belgium
| | - Jan Gettemans
- Department of Biomolecular Medicine, Faculty of Medicine & Health Sciences, Ghent University, Ghent, Belgium
| | - Douglas B Kell
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Stellenbosch, South Africa.,Department of Biochemistry and Systems Biology, Institute of Systems, Molecular and Integrative Biology, Faculty of Health and Life Sciences, University of Liverpool, Liverpool, UK.,The Novo Nordisk Foundation Centre for Biosustainability, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Etheresia Pretorius
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Stellenbosch, South Africa.,Department of Biochemistry and Systems Biology, Institute of Systems, Molecular and Integrative Biology, Faculty of Health and Life Sciences, University of Liverpool, Liverpool, UK
| |
Collapse
|
7
|
Kell DB, Pretorius E. The potential role of ischaemia-reperfusion injury in chronic, relapsing diseases such as rheumatoid arthritis, Long COVID, and ME/CFS: evidence, mechanisms, and therapeutic implications. Biochem J 2022; 479:1653-1708. [PMID: 36043493 PMCID: PMC9484810 DOI: 10.1042/bcj20220154] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 08/09/2022] [Accepted: 08/10/2022] [Indexed: 02/07/2023]
Abstract
Ischaemia-reperfusion (I-R) injury, initiated via bursts of reactive oxygen species produced during the reoxygenation phase following hypoxia, is well known in a variety of acute circumstances. We argue here that I-R injury also underpins elements of the pathology of a variety of chronic, inflammatory diseases, including rheumatoid arthritis, ME/CFS and, our chief focus and most proximally, Long COVID. Ischaemia may be initiated via fibrin amyloid microclot blockage of capillaries, for instance as exercise is started; reperfusion is a necessary corollary when it finishes. We rehearse the mechanistic evidence for these occurrences here, in terms of their manifestation as oxidative stress, hyperinflammation, mast cell activation, the production of marker metabolites and related activities. Such microclot-based phenomena can explain both the breathlessness/fatigue and the post-exertional malaise that may be observed in these conditions, as well as many other observables. The recognition of these processes implies, mechanistically, that therapeutic benefit is potentially to be had from antioxidants, from anti-inflammatories, from iron chelators, and via suitable, safe fibrinolytics, and/or anti-clotting agents. We review the considerable existing evidence that is consistent with this, and with the biochemical mechanisms involved.
Collapse
Affiliation(s)
- Douglas B. Kell
- Department of Biochemistry and Systems Biology, Institute of Systems, Molecular and Integrative Biology, Faculty of Health and Life Sciences, University of Liverpool, Liverpool L69 7ZB, U.K
- The Novo Nordisk Foundation Centre for Biosustainability, Technical University of Denmark, Kemitorvet 200, 2800 Kgs Lyngby, Denmark
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Stellenbosch, Private Bag X1 Matieland 7602, South Africa
| | - Etheresia Pretorius
- Department of Biochemistry and Systems Biology, Institute of Systems, Molecular and Integrative Biology, Faculty of Health and Life Sciences, University of Liverpool, Liverpool L69 7ZB, U.K
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Stellenbosch, Private Bag X1 Matieland 7602, South Africa
| |
Collapse
|
8
|
Rossi R, Mereuta OM, Barbachan e Silva M, Molina Gil S, Douglas A, Pandit A, Gilvarry M, McCarthy R, O'Connell S, Tierney C, Psychogios K, Tsivgoulis G, Szikora I, Tatlisumak T, Rentzos A, Thornton J, Ó Broin P, Doyle KM. Potential Biomarkers of Acute Ischemic Stroke Etiology Revealed by Mass Spectrometry-Based Proteomic Characterization of Formalin-Fixed Paraffin-Embedded Blood Clots. Front Neurol 2022; 13:854846. [PMID: 35518205 PMCID: PMC9062453 DOI: 10.3389/fneur.2022.854846] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 03/25/2022] [Indexed: 12/31/2022] Open
Abstract
Background and Aims Besides the crucial role in the treatment of acute ischemic stroke (AIS), mechanical thrombectomy represents a unique opportunity for researchers to study the retrieved clots, with the possibility of unveiling biological patterns linked to stroke pathophysiology and etiology. We aimed to develop a shotgun proteomic approach to study and compare the proteome of formalin-fixed paraffin-embedded (FFPE) cardioembolic and large artery atherosclerotic (LAA) clots. Methods We used 16 cardioembolic and 15 LAA FFPE thrombi from 31 AIS patients. The thrombus proteome was analyzed by label-free quantitative liquid chromatography-tandem mass spectrometry (LC-MS/MS). MaxQuant v1.5.2.8 and Perseus v.1.6.15.0 were used for bioinformatics analysis. Protein classes were identified using the PANTHER database and the STRING database was used to predict protein interactions. Results We identified 1,581 protein groups as part of the AIS thrombus proteome. Fourteen significantly differentially abundant proteins across the two etiologies were identified. Four proteins involved in the ubiquitin-proteasome pathway, blood coagulation or plasminogen activating cascade were identified as significantly abundant in LAA clots. Ten proteins involved in the ubiquitin proteasome-pathway, cytoskeletal remodeling of platelets, platelet adhesion or blood coagulation were identified as significantly abundant in cardioembolic clots. Conclusion Our results outlined a set of 14 proteins for a proof-of-principle characterization of cardioembolic and LAA FFPE clots, advancing the proteome profile of AIS human thrombi and understanding the pathophysiology of ischemic stroke.
Collapse
Affiliation(s)
- Rosanna Rossi
- Department of Physiology and Galway Neuroscience Centre, School of Medicine, National University of Ireland, Galway, Ireland
- CÚRAM–SFI Research Centre for Medical Devices, National University of Ireland Galway, Galway, Ireland
| | - Oana Madalina Mereuta
- Department of Physiology and Galway Neuroscience Centre, School of Medicine, National University of Ireland, Galway, Ireland
- CÚRAM–SFI Research Centre for Medical Devices, National University of Ireland Galway, Galway, Ireland
| | - Mariel Barbachan e Silva
- School of Mathematical and Statistical Sciences, National University of Ireland Galway, Galway, Ireland
| | - Sara Molina Gil
- Department of Physiology and Galway Neuroscience Centre, School of Medicine, National University of Ireland, Galway, Ireland
- CÚRAM–SFI Research Centre for Medical Devices, National University of Ireland Galway, Galway, Ireland
| | - Andrew Douglas
- Department of Physiology and Galway Neuroscience Centre, School of Medicine, National University of Ireland, Galway, Ireland
- CÚRAM–SFI Research Centre for Medical Devices, National University of Ireland Galway, Galway, Ireland
| | - Abhay Pandit
- CÚRAM–SFI Research Centre for Medical Devices, National University of Ireland Galway, Galway, Ireland
| | | | | | - Shane O'Connell
- School of Mathematical and Statistical Sciences, National University of Ireland Galway, Galway, Ireland
| | - Ciara Tierney
- Department of Physiology and Galway Neuroscience Centre, School of Medicine, National University of Ireland, Galway, Ireland
- CÚRAM–SFI Research Centre for Medical Devices, National University of Ireland Galway, Galway, Ireland
| | | | - Georgios Tsivgoulis
- Second Department of Neurology, National and Kapodistrian University of Athens, “Attikon” University Hospital, Athens, Greece
| | - István Szikora
- Department of Neurointerventions, National Institute of Clinical Neurosciences, Budapest, Hungary
| | - Turgut Tatlisumak
- Department of Neurology, Sahlgrenska University Hospital, Gothenburg, Sweden
- Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Alexandros Rentzos
- Department of Interventional and Diagnostic Neuroradiology, Sahlgrenska University Hospital, University of Gothenburg, Gothenburg, Sweden
| | - John Thornton
- Department of Radiology, Royal College of Surgeons in Ireland, Beaumont Hospital, Dublin, Ireland
| | - Pilib Ó Broin
- School of Mathematical and Statistical Sciences, National University of Ireland Galway, Galway, Ireland
| | - Karen M. Doyle
- Department of Physiology and Galway Neuroscience Centre, School of Medicine, National University of Ireland, Galway, Ireland
- CÚRAM–SFI Research Centre for Medical Devices, National University of Ireland Galway, Galway, Ireland
| |
Collapse
|
9
|
Pomilio AB, Vitale AA, Lazarowski AJ. Uncommon Noninvasive Biomarkers for the Evaluation and Monitoring of the Etiopathogenesis of Alzheimer's Disease. Curr Pharm Des 2022; 28:1152-1169. [DOI: 10.2174/1381612828666220413101929] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Accepted: 02/25/2022] [Indexed: 11/22/2022]
Abstract
Background:
Alzheimer´s disease (AD) is the most widespread dementia in the world, followed by vascular dementia. Since AD is a heterogeneous disease that shows several varied phenotypes, it is not easy to make an accurate diagnosis, so it arises when the symptoms are clear and the disease is already very advanced. Therefore, it is important to find out biomarkers for AD early diagnosis that facilitate treatment or slow down the disease. Classic biomarkers are obtained from cerebrospinal fluid and plasma, along with brain imaging by positron emission tomography. Attempts have been made to discover uncommon biomarkers from other body fluids, which are addressed in this update.
Objective:
This update aims to describe recent biomarkers from minimally invasive body fluids for the patients, such as saliva, urine, eye fluid or tears.
Methods:
Biomarkers were determined in patients versus controls by single tandem mass spectrometry, and immunoassays. Metabolites were identified by nuclear magnetic resonance, and microRNAs with genome-wide high-throughput real-time polymerase chain reaction-based platforms.
Results:
Biomarkers from urine, saliva, and eye fluid were described, including peptides/proteins, metabolites, and some microRNAs. The association with AD neuroinflammation and neurodegeneration was analyzed, highlighting the contribution of matrix metalloproteinases, the immune system and microglia, as well as the vascular system.
Conclusion:
Unusual biomarkers have been developed, which distinguish each stage and progression of the disease, and are suitable for the early AD diagnosis. An outstanding relationship of biomarkers with neuroinflammation and neurodegeneration was assessed, clearing up concerns of the etiopathogenesis of AD.
Collapse
Affiliation(s)
- Alicia B. Pomilio
- Departamento de Bioquímica Clínica, Área Hematología, Hospital de Clínicas “José de San Martín”, Universidad de Buenos Aires, Av. Córdoba 2351, C1120AAF Buenos Aires, Argentina
| | - Arturo A. Vitale
- Departamento de Bioquímica Clínica, Área Hematología, Hospital de Clínicas “José de San Martín”, Universidad de Buenos Aires, Av. Córdoba 2351, C1120AAF Buenos Aires, Argentina
| | - Alberto J. Lazarowski
- Departamento de Bioquímica Clínica, Facultad de Farmacia y Bioquímica, Instituto de Fisiopatología y Bioquímica Clínica (INFIBIOC), Universidad de Buenos Aires, Córdoba 2351, C1120AAF Buenos Aires, Argentina
| |
Collapse
|
10
|
Kim Y, Kim J, Son M, Lee J, Yeo I, Choi KY, Kim H, Kim BC, Lee KH, Kim Y. Plasma protein biomarker model for screening Alzheimer disease using multiple reaction monitoring-mass spectrometry. Sci Rep 2022; 12:1282. [PMID: 35075217 PMCID: PMC8786819 DOI: 10.1038/s41598-022-05384-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Accepted: 01/11/2022] [Indexed: 12/01/2022] Open
Abstract
Alzheimer disease (AD) is a leading cause of dementia that has gained prominence in our aging society. Yet, the complexity of diagnosing AD and measuring its invasiveness poses an obstacle. To this end, blood-based biomarkers could mitigate the inconveniences that impede an accurate diagnosis. We developed models to diagnose AD and measure the severity of neurocognitive impairment using blood protein biomarkers. Multiple reaction monitoring-mass spectrometry, a highly selective and sensitive approach for quantifying targeted proteins in samples, was used to analyze blood samples from 4 AD groups: cognitive normal control, asymptomatic AD, prodromal AD), and AD dementia. Multimarker models were developed using 10 protein biomarkers and apolipoprotein E genotypes for amyloid beta and 10 biomarkers with Korean Mini-Mental Status Examination (K-MMSE) score for predicting Alzheimer disease progression. The accuracies for the AD classification model and AD progression monitoring model were 84.9% (95% CI 82.8 to 87.0) and 79.1% (95% CI 77.8 to 80.5), respectively. The models were more accurate in diagnosing AD, compared with single APOE genotypes and the K-MMSE score. Our study demonstrates the possibility of predicting AD with high accuracy by blood biomarker analysis as an alternative method of screening for AD.
Collapse
Affiliation(s)
- Yeongshin Kim
- Interdisciplinary Program of Bioengineering, Seoul National University College of Engineering, Seoul, Republic of Korea
| | - Jaenyeon Kim
- Interdisciplinary Program of Bioengineering, Seoul National University College of Engineering, Seoul, Republic of Korea
| | - Minsoo Son
- Interdisciplinary Program of Bioengineering, Seoul National University College of Engineering, Seoul, Republic of Korea
| | - Jihyeon Lee
- Department of Biomedical Engineering, Seoul National University College of Medicine, 28 Yongon-Dong, Chongno-Ku, Seoul, 110-799, Republic of Korea
| | - Injoon Yeo
- Department of Biomedical Engineering, Seoul National University College of Medicine, 28 Yongon-Dong, Chongno-Ku, Seoul, 110-799, Republic of Korea
| | - Kyu Yeong Choi
- Gwangju Alzheimer's Disease and Related Dementia Cohort Research Center and Department of Biomedical Science, Chosun University, Gwangju, 61452, Republic of Korea
| | - Hoowon Kim
- Gwangju Alzheimer's Disease and Related Dementia Cohort Research Center and Department of Biomedical Science, Chosun University, Gwangju, 61452, Republic of Korea
- Department of Neurology, Chosun University Hospital, Gwangju, 61452, Republic of Korea
| | - Byeong C Kim
- Department of Neurology, Chonnam National University Medical School, Gwangju, 61469, Republic of Korea
| | - Kun Ho Lee
- Gwangju Alzheimer's Disease and Related Dementia Cohort Research Center and Department of Biomedical Science, Chosun University, Gwangju, 61452, Republic of Korea.
- Department of Biomedical Science, Chosun University, Gwangju, 61452, Republic of Korea.
- Aging Neuroscience Research Group, Korea Brain Research Institute, Daegu, 41062, Republic of Korea.
| | - Youngsoo Kim
- Interdisciplinary Program of Bioengineering, Seoul National University College of Engineering, Seoul, Republic of Korea.
- Department of Biomedical Engineering, Seoul National University College of Medicine, 28 Yongon-Dong, Chongno-Ku, Seoul, 110-799, Republic of Korea.
| |
Collapse
|
11
|
Eyileten C, Wicik Z, Simões SN, Martins-Jr DC, Klos K, Wlodarczyk W, Assinger A, Soldacki D, Chcialowski A, Siller-Matula JM, Postula M. Thrombosis-related circulating miR-16-5p is associated with disease severity in patients hospitalised for COVID-19. RNA Biol 2022; 19:963-979. [PMID: 35938548 PMCID: PMC9361765 DOI: 10.1080/15476286.2022.2100629] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 07/06/2022] [Indexed: 11/21/2022] Open
Abstract
SARS-CoV-2 tropism for the ACE2 receptor, along with the multifaceted inflammatory reaction, is likely to drive the generalized hypercoagulable and thrombotic state seen in patients with COVID-19. Using the original bioinformatic workflow and network medicine approaches we reanalysed four coronavirus-related expression datasets and performed co-expression analysis focused on thrombosis and ACE2 related genes. We identified microRNAs (miRNAs) which play role in ACE2-related thrombosis in coronavirus infection and further, we validated the expressions of precisely selected miRNAs-related to thrombosis (miR-16-5p, miR-27a-3p, let-7b-5p and miR-155-5p) in 79 hospitalized COVID-19 patients and 32 healthy volunteers by qRT-PCR. Consequently, we aimed to unravel whether bioinformatic prioritization could guide selection of miRNAs with a potential of diagnostic and prognostic biomarkers associated with disease severity in patients hospitalized for COVID-19. In bioinformatic analysis, we identified EGFR, HSP90AA1, APP, TP53, PTEN, UBC, FN1, ELAVL1 and CALM1 as regulatory genes which could play a pivotal role in COVID-19 related thrombosis. We also found miR-16-5p, miR-27a-3p, let-7b-5p and miR-155-5p as regulators in the coagulation and thrombosis process. In silico predictions were further confirmed in patients hospitalized for COVID-19. The expression levels of miR-16-5p and let-7b in COVID-19 patients were lower at baseline, 7-days and 21-day after admission compared to the healthy controls (p < 0.0001 for all time points for both miRNAs). The expression levels of miR-27a-3p and miR-155-5p in COVID-19 patients were higher at day 21 compared to the healthy controls (p = 0.007 and p < 0.001, respectively). A low baseline miR-16-5p expression presents predictive utility in assessment of the hospital length of stay or death in follow-up as a composite endpoint (AUC:0.810, 95% CI, 0.71-0.91, p < 0.0001) and low baseline expression of miR-16-5p and diabetes mellitus are independent predictors of increased length of stay or death according to a multivariate analysis (OR: 9.417; 95% CI, 2.647-33.506; p = 0.0005 and OR: 6.257; 95% CI, 1.049-37.316; p = 0.044, respectively). This study enabled us to better characterize changes in gene expression and signalling pathways related to hypercoagulable and thrombotic conditions in COVID-19. In this study we identified and validated miRNAs which could serve as novel, thrombosis-related predictive biomarkers of the COVID-19 complications, and can be used for early stratification of patients and prediction of severity of infection development in an individual.Abbreviations: ACE2, angiotensin-converting enzyme 2AF, atrial fibrillationAPP, Amyloid Beta Precursor ProteinaPTT, activated partial thromboplastin timeAUC, Area under the curveAβ, amyloid betaBMI, body mass indexCAD, coronary artery diseaseCALM1, Calmodulin 1 geneCaM, calmodulinCCND1, Cyclin D1CI, confidence intervalCOPD, chronic obstructive pulmonary diseaseCOVID-19, Coronavirus disease 2019CRP, C-reactive proteinCV, CardiovascularCVDs, cardiovascular diseasesDE, differentially expressedDM, diabetes mellitusEGFR, Epithelial growth factor receptorELAVL1, ELAV Like RNA Binding Protein 1FLNA, Filamin AFN1, Fibronectin 1GEO, Gene Expression OmnibushiPSC-CMs, Human induced pluripotent stem cell-derived cardiomyocytesHSP90AA1, Heat Shock Protein 90 Alpha Family Class A Member 1Hsp90α, heat shock protein 90αICU, intensive care unitIL, interleukinIQR, interquartile rangelncRNAs, long non-coding RNAsMI, myocardial infarctionMiRNA, MiR, microRNAmRNA, messenger RNAncRNA, non-coding RNANERI, network-medicine based integrative approachNF-kB, nuclear factor kappa-light-chain-enhancer of activated B cellsNPV, negative predictive valueNXF, nuclear export factorPBMCs, Peripheral blood mononuclear cellsPCT, procalcitoninPPI, Protein-protein interactionsPPV, positive predictive valuePTEN, phosphatase and tensin homologqPCR, quantitative polymerase chain reactionROC, receiver operating characteristicSARS-CoV-2, severe acute respiratory syndrome coronavirus 2SD, standard deviationTLR4, Toll-like receptor 4TM, thrombomodulinTP53, Tumour protein P53UBC, Ubiquitin CWBC, white blood cells.
Collapse
Affiliation(s)
- Ceren Eyileten
- Department of Experimental and Clinical Pharmacology, Medical University of Warsaw, Center for Preclinical Research and Technology CEPT, Warsaw, Poland
- Genomics Core Facility, Centre of New Technologies, University of Warsaw, Warsaw, Poland
| | - Zofia Wicik
- Department of Experimental and Clinical Pharmacology, Medical University of Warsaw, Center for Preclinical Research and Technology CEPT, Warsaw, Poland
- Center for Mathematics, Computing and Cognition, Federal University of ABC, Santo AndréBrazil
| | - Sérgio N. Simões
- Department of Informatics, Federal Institute of Espírito Santo, Serra, Brazil
| | - David C. Martins-Jr
- Center for Mathematics, Computing and Cognition, Federal University of ABC, Santo AndréBrazil
| | - Krzysztof Klos
- Department of Infectious Diseases and Allergology - Military Institute of Medicine, Warsaw, Poland
| | - Wojciech Wlodarczyk
- Department of Infectious Diseases and Allergology - Military Institute of Medicine, Warsaw, Poland
| | - Alice Assinger
- Department of Vascular Biology and Thrombosis Research, Center of Physiology and Pharmacology, Medical University of Vienna, Austria
| | - Dariusz Soldacki
- Department of Clinical Immunology, Medical University of Warsaw, Warsaw, Poland
| | - Andrzej Chcialowski
- Department of Infectious Diseases and Allergology - Military Institute of Medicine, Warsaw, Poland
| | - Jolanta M. Siller-Matula
- Department of Experimental and Clinical Pharmacology, Medical University of Warsaw, Center for Preclinical Research and Technology CEPT, Warsaw, Poland
- Department of Internal Medicine II, Division of Cardiology, Medical University of Vienna, Vienna, Austria
| | - Marek Postula
- Department of Experimental and Clinical Pharmacology, Medical University of Warsaw, Center for Preclinical Research and Technology CEPT, Warsaw, Poland
| |
Collapse
|
12
|
McLarnon JG. A Leaky Blood–Brain Barrier to Fibrinogen Contributes to Oxidative Damage in Alzheimer’s Disease. Antioxidants (Basel) 2021; 11:antiox11010102. [PMID: 35052606 PMCID: PMC8772934 DOI: 10.3390/antiox11010102] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 12/26/2021] [Accepted: 12/28/2021] [Indexed: 12/15/2022] Open
Abstract
The intactness of blood–brain barrier (BBB) is compromised in Alzheimer’s disease (AD). Importantly, evidence suggests that the perturbation and abnormalities appearing in BBB can manifest early in the progression of the disease. The disruption of BBB allows extravasation of the plasma protein, fibrinogen, to enter brain parenchyma, eliciting immune reactivity and response. The presence of amyloid-β (Aβ) peptide leads to the formation of abnormal aggregates of fibrin resistant to degradation. Furthermore, Aβ deposits act on the contact system of blood coagulation, altering levels of thrombin, fibrin clots and neuroinflammation. The neurovascular unit (NVU) comprises an ensemble of brain cells which interact with infiltrating fibrinogen. In particular, interaction of resident immune cell microglia with fibrinogen, fibrin and Aβ results in the production of reactive oxygen species (ROS), a neurotoxic effector in AD brain. Overall, fibrinogen infiltration through a leaky BBB in AD animal models and in human AD tissue is associated with manifold abnormalities including persistent fibrin aggregation and clots, microglial-mediated production of ROS and diminished viability of neurons and synaptic connectivity. An objective of this review is to better understand how processes associated with BBB leakiness to fibrinogen link vascular pathology with neuronal and synaptic damage in AD.
Collapse
Affiliation(s)
- James G McLarnon
- Department of Anesthesiology, Pharmacology and Therapeutics, Faculty of Medicine, The University of British Columbia, 2176 Health Sciences Mall, Vancouver, BC V6T1Z3, Canada
| |
Collapse
|
13
|
VanDusen KW, Li YJ, Cai V, Hall A, Hiles S, Thompson JW, Moseley MA, Cooter M, Acker L, Levy JH, Ghadimi K, Quiñones QJ, Devinney MJ, Chung S, Terrando N, Moretti EW, Browndyke JN, Mathew JP, Berger M. Cerebrospinal Fluid Proteome Changes in Older Non-Cardiac Surgical Patients with Postoperative Cognitive Dysfunction. J Alzheimers Dis 2021; 80:1281-1297. [PMID: 33682719 PMCID: PMC8052629 DOI: 10.3233/jad-201544] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Background: Postoperative cognitive dysfunction (POCD), a syndrome of cognitive deficits occurring 1–12 months after surgery primarily in older patients, is associated with poor postoperative outcomes. POCD is hypothesized to result from neuroinflammation; however, the pathways involved remain unclear. Unbiased proteomic analyses have been used to identify neuroinflammatory pathways in multiple neurologic diseases and syndromes but have not yet been applied to POCD. Objective: To utilize unbiased mass spectrometry-based proteomics to identify potential neuroinflammatory pathways underlying POCD. Methods: Unbiased LC-MS/MS proteomics was performed on immunodepleted cerebrospinal fluid (CSF) samples obtained before, 24 hours after, and 6 weeks after major non-cardiac surgery in older adults who did (n = 8) or did not develop POCD (n = 6). Linear mixed models were used to select peptides and proteins with intensity differences for pathway analysis. Results: Mass spectrometry quantified 8,258 peptides from 1,222 proteins in > 50%of patient samples at all three time points. Twelve peptides from 11 proteins showed differences in expression over time between patients with versus without POCD (q < 0.05), including proteins previously implicated in neurodegenerative disease pathophysiology. Additionally, 283 peptides from 182 proteins were identified with trend-level differences (q < 0.25) in expression over time between these groups. Among these, pathway analysis revealed that 50 were from 17 proteins mapping to complement and coagulation pathways (q = 2.44*10–13). Conclusion: These data demonstrate the feasibility of performing unbiased mass spectrometry on perioperative CSF samples to identify pathways associated with POCD. Additionally, they provide hypothesis-generating evidence for CSF complement and coagulation pathway changes in patients with POCD.
Collapse
Affiliation(s)
- Keith W VanDusen
- Department of Anesthesiology, Duke University Medical Center, Durham, NC, USA
| | - Yi-Ju Li
- Department of Biostatistics and Bioinformatics, Duke University, Durham, NC, USA.,Duke Molecular Physiology Institute, Duke University, Durham, NC, USA
| | - Victor Cai
- Department of Anesthesiology, Duke University Medical Center, Durham, NC, USA
| | - Ashley Hall
- Department of Anesthesiology, Duke University Medical Center, Durham, NC, USA
| | - Sarah Hiles
- Duke Center for Genomic and Computational Biology, Duke University, Durham, NC, USA
| | - J Will Thompson
- Duke Center for Genomic and Computational Biology, Duke University, Durham, NC, USA
| | - M Arthur Moseley
- Duke Center for Genomic and Computational Biology, Duke University, Durham, NC, USA
| | - Mary Cooter
- Department of Anesthesiology, Duke University Medical Center, Durham, NC, USA
| | - Leah Acker
- Department of Anesthesiology, Duke University Medical Center, Durham, NC, USA
| | - Jerrold H Levy
- Department of Anesthesiology, Duke University Medical Center, Durham, NC, USA
| | - Kamrouz Ghadimi
- Department of Anesthesiology, Duke University Medical Center, Durham, NC, USA
| | - Quintin J Quiñones
- Department of Anesthesiology, Duke University Medical Center, Durham, NC, USA
| | - Michael J Devinney
- Department of Anesthesiology, Duke University Medical Center, Durham, NC, USA
| | - Stacey Chung
- Department of Anesthesiology, Duke University Medical Center, Durham, NC, USA
| | - Niccolò Terrando
- Department of Anesthesiology, Duke University Medical Center, Durham, NC, USA
| | - Eugene W Moretti
- Department of Anesthesiology, Duke University Medical Center, Durham, NC, USA
| | - Jeffrey N Browndyke
- Department of Psychiatry & Behavioral Sciences, Division of Geriatric Behavioral Health, Duke University Medical Center, Durham, NC, USA.,Duke Institute for Brain Sciences, Duke University, Durham, NC, USA.,Center for Cognitive Neuroscience, Duke University Medical Center, Durham, NC, USA
| | - Joseph P Mathew
- Department of Anesthesiology, Duke University Medical Center, Durham, NC, USA
| | - Miles Berger
- Department of Anesthesiology, Duke University Medical Center, Durham, NC, USA.,Department of Anesthesiology, Duke University Medical Center, Durham, NC, USA.,Department of Anesthesiology, Duke University Medical Center, Durham, NC, USA.,Department of Anesthesiology, Duke University Medical Center, Durham, NC, USA
| | | |
Collapse
|
14
|
Rahman MM, Lendel C. Extracellular protein components of amyloid plaques and their roles in Alzheimer's disease pathology. Mol Neurodegener 2021; 16:59. [PMID: 34454574 PMCID: PMC8400902 DOI: 10.1186/s13024-021-00465-0] [Citation(s) in RCA: 83] [Impact Index Per Article: 27.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2020] [Accepted: 06/11/2021] [Indexed: 12/20/2022] Open
Abstract
Alzheimer's disease (AD) is pathologically defined by the presence of fibrillar amyloid β (Aβ) peptide in extracellular senile plaques and tau filaments in intracellular neurofibrillary tangles. Extensive research has focused on understanding the assembly mechanisms and neurotoxic effects of Aβ during the last decades but still we only have a brief understanding of the disease associated biological processes. This review highlights the many other constituents that, beside Aβ, are accumulated in the plaques, with the focus on extracellular proteins. All living organisms rely on a delicate network of protein functionality. Deposition of significant amounts of certain proteins in insoluble inclusions will unquestionably lead to disturbances in the network, which may contribute to AD and copathology. This paper provide a comprehensive overview of extracellular proteins that have been shown to interact with Aβ and a discussion of their potential roles in AD pathology. Methods that can expand the knowledge about how the proteins are incorporated in plaques are described. Top-down methods to analyze post-mortem tissue and bottom-up approaches with the potential to provide molecular insights on the organization of plaque-like particles are compared. Finally, a network analysis of Aβ-interacting partners with enriched functional and structural key words is presented.
Collapse
Affiliation(s)
- M Mahafuzur Rahman
- Department of Chemistry, KTH Royal Institute of Technology, SE-100 44, Stockholm, Sweden.
| | - Christofer Lendel
- Department of Chemistry, KTH Royal Institute of Technology, SE-100 44, Stockholm, Sweden.
| |
Collapse
|
15
|
Amelianchik A, Merkel J, Palanisamy P, Kaneki S, Hyatt E, Norris EH. The protective effect of early dietary fat consumption on Alzheimer's disease-related pathology and cognitive function in mice. ALZHEIMER'S & DEMENTIA (NEW YORK, N. Y.) 2021; 7:e12173. [PMID: 34084889 PMCID: PMC8144936 DOI: 10.1002/trc2.12173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Revised: 03/24/2021] [Accepted: 04/05/2021] [Indexed: 11/12/2022]
Abstract
INTRODUCTION It has been suggested that obesity may influence Alzheimer's disease (AD) pathogenesis, yet the numerous publications on this topic have inconsistent results and conclusions. METHODS Our study examined the effect of varying the timing of high-fat diet (HFD) consumption on AD-related pathology and cognition in transgenic Tg6799 AD mice. RESULTS HFD feeding starting at or before 3 months of age, prior to severe AD pathology, had protective effects in AD mice: reduced extracellular amyloid beta (Aβ) deposition, decreased fibrinogen extravasation into the brain parenchyma, and improved cognitive function. However, delaying HFD consumption until 6 months of age, when AD pathology is ubiquitous, reduced these protective effects in AD mice. DISCUSSION Overall, we demonstrate that the timeline of HFD consumption may play an important role in how dietary fats affect AD pathogenesis and cognitive function.
Collapse
Affiliation(s)
- Anna Amelianchik
- Patricia and John Rosenwald Laboratory of Neurobiology and GeneticsThe Rockefeller UniversityNew YorkUSA
| | - Jonathan Merkel
- Patricia and John Rosenwald Laboratory of Neurobiology and GeneticsThe Rockefeller UniversityNew YorkUSA
- Paul Flechsig Institute of Brain ResearchLeipzig UniversityLeipzigGermany
| | - Premkumar Palanisamy
- Patricia and John Rosenwald Laboratory of Neurobiology and GeneticsThe Rockefeller UniversityNew YorkUSA
| | - Shigeru Kaneki
- Patricia and John Rosenwald Laboratory of Neurobiology and GeneticsThe Rockefeller UniversityNew YorkUSA
| | - Emily Hyatt
- Patricia and John Rosenwald Laboratory of Neurobiology and GeneticsThe Rockefeller UniversityNew YorkUSA
| | - Erin H. Norris
- Patricia and John Rosenwald Laboratory of Neurobiology and GeneticsThe Rockefeller UniversityNew YorkUSA
| |
Collapse
|
16
|
Stetter C, Lopez-Caperuchipi S, Hopp-Krämer S, Bieber M, Kleinschnitz C, Sirén AL, Albert-Weißenberger C. Amelioration of Cognitive and Behavioral Deficits after Traumatic Brain Injury in Coagulation Factor XII Deficient Mice. Int J Mol Sci 2021; 22:4855. [PMID: 34063730 PMCID: PMC8124758 DOI: 10.3390/ijms22094855] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Accepted: 05/01/2021] [Indexed: 11/23/2022] Open
Abstract
Based on recent findings that show that depletion of factor XII (FXII) leads to better posttraumatic neurological recovery, we studied the effect of FXII-deficiency on post-traumatic cognitive and behavioral outcomes in female and male mice. In agreement with our previous findings, neurological deficits on day 7 after weight-drop traumatic brain injury (TBI) were significantly reduced in FXII-/- mice compared to wild type (WT) mice. Also, glycoprotein Ib (GPIb)-positive platelet aggregates were more frequent in brain microvasculature of WT than FXII-/- mice 3 months after TBI. Six weeks after TBI, memory for novel object was significantly reduced in both female and male WT but not in FXII-/- mice compared to sham-operated mice. In the setting of automated home-cage monitoring of socially housed mice in IntelliCages, female WT mice but not FXII-/- mice showed decreased exploration and reacted negatively to reward extinction one month after TBI. Since neuroendocrine stress after TBI might contribute to trauma-induced cognitive dysfunction and negative emotional contrast reactions, we measured peripheral corticosterone levels and the ration of heart, lung, and spleen weight to bodyweight. Three months after TBI, plasma corticosterone levels were significantly suppressed in both female and male WT but not in FXII-/- mice, while the relative heart weight increased in males but not in females of both phenotypes when compared to sham-operated mice. Our results indicate that FXII deficiency is associated with efficient post-traumatic behavioral and neuroendocrine recovery.
Collapse
Affiliation(s)
- Christian Stetter
- Department of Neurosurgery, University Hospital of Würzburg, Josef-Schneider-Str. 11, 97080 Würzburg, Germany; (C.S.); (S.L.-C.); (S.H.-K.); (C.A.-W.)
| | - Simon Lopez-Caperuchipi
- Department of Neurosurgery, University Hospital of Würzburg, Josef-Schneider-Str. 11, 97080 Würzburg, Germany; (C.S.); (S.L.-C.); (S.H.-K.); (C.A.-W.)
| | - Sarah Hopp-Krämer
- Department of Neurosurgery, University Hospital of Würzburg, Josef-Schneider-Str. 11, 97080 Würzburg, Germany; (C.S.); (S.L.-C.); (S.H.-K.); (C.A.-W.)
- Department of Neurology, University Hospital of Würzburg, Josef-Schneider-Str. 11, 97080 Würzburg, Germany; (M.B.); (C.K.)
| | - Michael Bieber
- Department of Neurology, University Hospital of Würzburg, Josef-Schneider-Str. 11, 97080 Würzburg, Germany; (M.B.); (C.K.)
| | - Christoph Kleinschnitz
- Department of Neurology, University Hospital of Würzburg, Josef-Schneider-Str. 11, 97080 Würzburg, Germany; (M.B.); (C.K.)
- Department of Neurology and Center for Translational and Behavioral Neurosciences (C-TNBS), University Hospital of Essen, Hufelandstr. 55, 45147 Essen, Germany
| | - Anna-Leena Sirén
- Department of Neurosurgery, University Hospital of Würzburg, Josef-Schneider-Str. 11, 97080 Würzburg, Germany; (C.S.); (S.L.-C.); (S.H.-K.); (C.A.-W.)
| | - Christiane Albert-Weißenberger
- Department of Neurosurgery, University Hospital of Würzburg, Josef-Schneider-Str. 11, 97080 Würzburg, Germany; (C.S.); (S.L.-C.); (S.H.-K.); (C.A.-W.)
- Institute for Physiology, Department for Neurophysiology, Julius-Maximilians-University Würzburg, Röntgenring 9, 97070 Würzburg, Germany
| |
Collapse
|
17
|
Tanaka T, Hirai S, Hosokawa M, Saito T, Sakuma H, Saido T, Hasegawa M, Okado H. Early-life stress induces the development of Alzheimer's disease pathology via angiopathy. Exp Neurol 2021; 337:113552. [PMID: 33309748 DOI: 10.1016/j.expneurol.2020.113552] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 11/25/2020] [Accepted: 12/06/2020] [Indexed: 01/06/2023]
Abstract
Alzheimer's disease (AD), a progressive neurodegenerative disorder, is a major societal, scientific, and economic problem. Several early-life factors associated with an increased risk for the clinical diagnosis of AD have recently been identified. In the present study, we investigated the involvement of early-life stress in the pathogenesis of AD using heterozygous amyloid precursor protein (APP) mutant mice (AppNL-G-F/wt) and wild-type (Appwt/wt) mice. We found that maternal-separated Appwt/wt mice showed narrowing of vessels and decreased pericyte coverage of capillaries in the prefrontal cortex, while maternal-separated AppNL-G-F/wt mice additionally showed the impairment of cognitive function, earlier formation of Aβ plaques, increased vessel-associated microglia, and disruption of the blood-brain barrier. Substantial activation of microglia was detected in the maternal-separated AppNL-G-F/wt mice and maternal-separated Appwt/wt mice. At an early stage, morphological changes and inflammatory responses were observed in the microglia of the maternal-separated AppNL-G-F/wt mice and maternal-separated Appwt/wt mice, and morphological changes in the microglia were observed in the non-maternal-separated AppNL-G-F/wt mice. Microglia activation induced by maternal separation in combination with the APP mutation may impair the vascular system, leading to AD progression. These findings therefore suggest that maternal separation results in the early induction of AD-related pathology via angiopathy.
Collapse
Affiliation(s)
- Tomoko Tanaka
- Laboratory of Neural Development, Department of Psychiatry & Behavioral Science, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Shinobu Hirai
- Laboratory of Neural Development, Department of Psychiatry & Behavioral Science, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Masato Hosokawa
- Dementia Research Project, Department of Brain & Neuroscience, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Takashi Saito
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, Saitama, Japan; Department of Neurocognitive Science, Institute of Brain Science, Nagoya City University Graduate School of Medical Science, Aichi, Japan
| | - Hiroshi Sakuma
- Child brain Project, Department of Brain & Neuroscience, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Takaomi Saido
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, Saitama, Japan
| | - Masato Hasegawa
- Dementia Research Project, Department of Brain & Neuroscience, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Haruo Okado
- Laboratory of Neural Development, Department of Psychiatry & Behavioral Science, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan.
| |
Collapse
|
18
|
Phochantachinda S, Chantong B, Reamtong O, Chatchaisak D. Change in the plasma proteome associated with canine cognitive dysfunction syndrome (CCDS) in Thailand. BMC Vet Res 2021; 17:60. [PMID: 33514370 PMCID: PMC7845120 DOI: 10.1186/s12917-021-02744-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Accepted: 01/01/2021] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Canine cognitive dysfunction syndrome (CCDS) is a progressive neurodegenerative disorder found in senior dogs. Due to the lack of biological markers, CCDS is commonly underdiagnosed. The aim of this study was to identify potential plasma biomarkers using proteomics techniques and to increase our understanding of the pathogenic mechanism of the disease. Plasma amyloid beta 42 (Aβ42) has been seen to be a controversial biomarker for CCDS. Proteomics analysis was performed for protein identification and quantification. RESULTS Within CCDS, ageing, and adult dogs, 87 proteins were identified specific to Canis spp. in the plasma samples. Of 87 proteins, 48 and 41 proteins were changed in the ageing and adult groups, respectively. Several distinctly expressed plasma proteins identified in CCDS were involved in complement and coagulation cascades and the apolipoprotein metabolism pathway. Plasma Aβ42 levels considerably overlapped within the CCDS and ageing groups. In the adult group, the Aβ42 level was low compared with that in the other groups. Nevertheless, plasma Aβ42 did not show a correlation with the Canine Cognitive Dysfunction Rating scale (CCDR) score in the CCDS group (p = 0.131, R2 = 0.261). CONCLUSIONS Our present findings suggest that plasma Aβ42 does not show potential for use as a diagnostic biomarker in CCDS. The nano-LC-MS/MS data revealed that the predictive underlying mechanism of CCDS was the co-occurrence of inflammation-mediated acute phase response proteins and complement and coagulation cascades that partly functioned by apolipoproteins and lipid metabolism. Some of the differentially expressed proteins may serve as potential predictor biomarkers along with Aβ42 in plasma for improved CCDS diagnosis. Further study in larger population-based cohort study is required in validation to define the correlation between protein expression and the pathogenesis of CCDS.
Collapse
Affiliation(s)
- Sataporn Phochantachinda
- Faculty of Veterinary Science, Mahidol University, Salaya, Phutthamonthon, Nakorn Pathom, 73170, Thailand
| | - Boonrat Chantong
- Department of Pre-Clinical and Applied Animal Science, Faculty of Veterinary Science, Mahidol University, Salaya, Phutthamonthon, Nakorn Pathom, 73170, Thailand
| | - Onrapak Reamtong
- Department of Molecular Tropical Medicine and Genetics, Faculty of Tropical Medicine, Mahidol University, Phaya Thai, Ratchathewi, Bangkok, 10400, Thailand
| | - Duangthip Chatchaisak
- Department of Clinical Sciences and Public Health, Faculty of Veterinary Science, Mahidol University, Salaya, Phutthamonthon, Nakorn Pathom, 73170, Thailand.
| |
Collapse
|
19
|
Levin RA, Carnegie MH, Celermajer DS. Pulse Pressure: An Emerging Therapeutic Target for Dementia. Front Neurosci 2020; 14:669. [PMID: 32670015 PMCID: PMC7327093 DOI: 10.3389/fnins.2020.00669] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Accepted: 06/02/2020] [Indexed: 12/11/2022] Open
Abstract
Elevated pulse pressure can cause blood-brain barrier dysfunction and subsequent adverse neurological changes that may drive or contribute to the development of dementia with age. In short, elevated pulse pressure dysregulates cerebral endothelial cells and increases cellular production of oxidative and inflammatory molecules. The resulting cerebral microvascular damage, along with excessive pulsatile mechanical force, can induce breakdown of the blood-brain barrier, which in turn triggers brain cell impairment and death. We speculate that elevated pulse pressure may also reduce the efficacy of other therapeutic strategies for dementia. For instance, BACE1 inhibitors and anti-amyloid-β biologics reduce amyloid-β deposits in the brain that are thought to be a cause of Alzheimer’s disease, the most prevalent form of dementia. However, upregulation of oxidative and inflammatory molecules and increased amyloid-β secretion by cerebral endothelial cells exposed to elevated pulse pressure may hinder cognitive improvements with these drugs. Additionally, stem or progenitor cell therapy has the potential to repair blood-brain barrier damage, but chronic oxidative and inflammatory stress due to elevated pulse pressure can inhibit stem and progenitor cell regeneration. Finally, we discuss current efforts to repurpose blood pressure medications to prevent or treat dementia. We propose that new drugs or devices should be developed to safely reduce elevated pulse pressure specifically to the brain. Such novel technologies may alleviate an entire downstream pathway of cellular dysfunction, oxidation, inflammation, and amyloidogenesis, thereby preventing pulse-pressure-induced cognitive decline. Furthermore, these technologies may also enhance efficacy of other dementia therapeutics when used in combination.
Collapse
Affiliation(s)
- Rachel A Levin
- The Brain Protection Company, Sydney, NSW, Australia.,M.H. Carnegie & Co., Sydney, NSW, Australia
| | - Mark H Carnegie
- The Brain Protection Company, Sydney, NSW, Australia.,M.H. Carnegie & Co., Sydney, NSW, Australia
| | - David S Celermajer
- The Brain Protection Company, Sydney, NSW, Australia.,The Heart Research Institute, Sydney, NSW, Australia
| |
Collapse
|
20
|
Rehiman SH, Lim SM, Neoh CF, Majeed ABA, Chin AV, Tan MP, Kamaruzzaman SB, Ramasamy K. Proteomics as a reliable approach for discovery of blood-based Alzheimer's disease biomarkers: A systematic review and meta-analysis. Ageing Res Rev 2020; 60:101066. [PMID: 32294542 DOI: 10.1016/j.arr.2020.101066] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 04/03/2020] [Accepted: 04/03/2020] [Indexed: 02/08/2023]
Abstract
In order to gauge the impact of proteomics in discovery of Alzheimer's disease (AD) blood-based biomarkers, this study had systematically reviewed articles published between 1984-2019. Articles that fulfilled the inclusion criteria were assessed for risk of bias. A meta-analysis was performed for replicable candidate biomarkers (CB). Of the 1651 articles that were identified, 17 case-control and two cohort studies, as well as three combined case-control and longitudinal designs were shortlisted. A total of 207 AD and mild cognitive impairment (MCI) CB were discovered, with 48 reported in >2 studies. This review highlights six CB, namely alpha-2-macroglobulin (α2M)ps, pancreatic polypeptide (PP)ps, apolipoprotein A-1 (ApoA-1)ps, afaminp, insulin growth factor binding protein-2 (IGFBP-2)ps and fibrinogen-γ-chainp, all of which exhibited consistent pattern of regulation in >three independent cohorts. They are involved in AD pathogenesis via amyloid-beta (Aβ), neurofibrillary tangles, diabetes and cardiovascular diseases (CVD). Meta-analysis indicated that ApoA-1ps was significantly downregulated in AD (SMD = -1.52, 95% CI: -1.89, -1.16, p < 0.00001), with low inter-study heterogeneity (I2 = 0%, p = 0.59). α2Mps was significantly upregulated in AD (SMD = 0.83, 95% CI: 0.05, 1.62, p = 0.04), with moderate inter-study heterogeneity (I2 = 41%, p = 0.19). Both CB are involved in Aβ formation. These findings provide important insights into blood-based AD biomarkers discovery via proteomics.
Collapse
|
21
|
Lu Y, Fang Z, Zeng T, Li M, Chen Q, Zhang H, Zhou Q, Hu Y, Chen L, Su S. Chronic hepatitis B: dynamic change in Traditional Chinese Medicine syndrome by dynamic network biomarkers. Chin Med 2019; 14:52. [PMID: 31768187 PMCID: PMC6873721 DOI: 10.1186/s13020-019-0275-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Accepted: 11/04/2019] [Indexed: 02/06/2023] Open
Abstract
Background In traditional Chinese medicine (TCM) clinical practice, TCM syndromes help to understand human homeostasis and guide individualized treatment. However, the TCM syndrome changes with disease progression, of which the scientific basis and mechanism remain unclear. Methods To demonstrate the underlying mechanism of dynamic changes in the TCM syndrome, we applied a dynamic network biomarker (DNB) algorithm to obtain the DNBs of changes in the TCM syndrome, based on the transcriptomic data of patients with chronic hepatitis B and typical TCM syndromes, including healthy controls and patients with liver-gallbladder dampness-heat syndrome (LGDHS), liver-depression spleen-deficiency syndrome (LDSDS), and liver-kidney yin-deficiency syndrome (LKYDS). The DNB model exploits collective fluctuations and correlations of the observed genes, then diagnoses the critical state. Results Our results showed that the DNBs of TCM syndromes were comprised of 52 genes and the tipping point occurred at the LDSDS stage. Meanwhile, there were numerous differentially expressed genes between LGDHS and LKYDS, which highlighted the drastic changes before and after the tipping point, implying the 52 DNBs could serve as early-warning signals of the upcoming change in the TCM syndrome. Next, we validated DNBs by cytokine profiling and isobaric tags for relative and absolute quantitation (iTRAQ). The results showed that PLG (plasminogen) and coagulation factor XII (F12) were significantly expressed during the progression of TCM syndrome from LGDHS to LKYDS. Conclusions This study provides a scientific understanding of changes in the TCM syndrome. During this process, the cytokine system was involved all the time. The DNBs PLG and F12 were confirmed to significantly change during TCM-syndrome progression and indicated a potential value of DNBs in auxiliary diagnosis of TCM syndrome in CHB. Trial registration Identifier: NCT03189992. Registered on June 4, 2017. Retrospectively registered (http://www.clinicaltrials.gov)
Collapse
Affiliation(s)
- Yiyu Lu
- 1Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203 China
| | - Zhaoyuan Fang
- 2Key Laboratory of Systems Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Shanghai Institute of Biological Sciences, Chinese Academy of Sciences, Shanghai, 200031 China
| | - Tao Zeng
- 2Key Laboratory of Systems Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Shanghai Institute of Biological Sciences, Chinese Academy of Sciences, Shanghai, 200031 China
| | - Meiyi Li
- 5Minhang Branch, Zhongshan Hospital, Fudan University/Institute of Fudan-Minhang Academic Health System, Minhang Hospital, Fudan University, Shanghai, 201199 China
| | - Qilong Chen
- 1Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203 China
| | - Hui Zhang
- 1Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203 China
| | - Qianmei Zhou
- 1Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203 China
| | - Yiyang Hu
- 4Institute of Liver Disease, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203 China
| | - Luonan Chen
- 2Key Laboratory of Systems Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Shanghai Institute of Biological Sciences, Chinese Academy of Sciences, Shanghai, 200031 China.,3CAS Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming, 650223 China
| | - Shibing Su
- 1Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203 China
| |
Collapse
|
22
|
Liu Y, Chen H, Zhao K, He W, Lin S, He J. High levels of plasma fibrinogen are related to post-stroke cognitive impairment. Brain Behav 2019; 9:e01391. [PMID: 31475471 PMCID: PMC6790326 DOI: 10.1002/brb3.1391] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2018] [Revised: 06/30/2019] [Accepted: 07/28/2019] [Indexed: 01/01/2023] Open
Abstract
INTRODUCTION Studies have shown that high levels of the fibrinogen (FIB) are related to cognitive deficits. However, the relationship between fibrinogen and cognitive deficit after stroke remains unclear. Therefore, we explored the relationship between plasma fibrinogen and post-stroke cognitive impairment (PSCI). METHODS This study is carried out in the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China. A total of 210 patients with acute ischemic stroke were enrolled in this study. Ultimately, 134 patients completed 3-month follow-up. Blood samples were collected at hospital admission. Cognitive function was evaluated 3 months after stroke. All patients underwent the Mini-Mental State Examination (MMSE) after 3 months. RESULTS Higher levels of fibrinogen were observed in patients with post-stroke cognitive impairment compared with the non-PSCI group (p < .001). Additionally, elevated plasma fibrinogen levels were independently associated with PSCI (odds ratio [OR] = 2.000, 95% CI 1.062-3.770 p = .032). The plasma fibrinogen levels were negatively correlated with the 3-month MMSE scores (r = -.171, p = .048). In a multivariate linear regression, FIB was negatively associated with the 3-month MMSE scores after adjustment for the other variables (β = -0.782, p = .035). CONCLUSION High levels of plasma fibrinogen were associated with the presence and severity of PSCI.
Collapse
Affiliation(s)
- Yuntao Liu
- Department of Neurology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Huijun Chen
- Department of Neurology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Kai Zhao
- Department of Neurology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Weilei He
- Department of Neurology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Shasha Lin
- Department of Neurology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jincai He
- Department of Neurology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
23
|
Cai Z, Qiao PF, Wan CQ, Cai M, Zhou NK, Li Q. Role of Blood-Brain Barrier in Alzheimer's Disease. J Alzheimers Dis 2019; 63:1223-1234. [PMID: 29782323 DOI: 10.3233/jad-180098] [Citation(s) in RCA: 208] [Impact Index Per Article: 41.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The blood-brain barrier (BBB) is involved in the pathogenesis of Alzheimer's disease (AD). BBB is a highly selective semipermeable structural and chemical barrier which ensures a stable internal environment of the brain and prevents foreign objects invading the brain tissue. BBB dysfunction induces the failure of Aβ transport from brain to the peripheral circulation across the BBB. Especially, decreased levels of LRP-1 (low density lipoprotein receptor-related protein 1) and increased levels of RAGE (receptor for advanced glycation endproducts) at the BBB can cause the failure of Aβ transport. The pathogenesis of AD is related to the BBB structural components, including pericytes, astrocytes, vascular endothelial cells, and tight junctions. BBB dysfunction will trigger neuroinflammation and oxidative stress, then enhance the activity of β-secretase and γ-secretase, and finally promote Aβ generation. A progressive accumulation of Aβ in brain and BBB dysfunction may become a feedback loop that gives rise to cognitive impairment and the onset of dementia. The correlation between BBB dysfunction and tau pathology has been well-reported. Therefore, regulating BBB function may be a new therapeutic target for treating AD.
Collapse
Affiliation(s)
- Zhiyou Cai
- Department of Neurology, Chongqing General Hospital, Chongqing, Chongqing, China
| | - Pei-Feng Qiao
- Department of Neurology, Chongqing General Hospital, Chongqing, Chongqing, China
| | - Cheng-Qun Wan
- Department of Neurology, Chongqing General Hospital, Chongqing, Chongqing, China
| | - Min Cai
- Department of Neurology, Chongqing General Hospital, Chongqing, Chongqing, China
| | - Nan-Kai Zhou
- Department of Neurology, Chongqing General Hospital, Chongqing, Chongqing, China
| | - Qin Li
- Department of Neurology, Chongqing General Hospital, Chongqing, Chongqing, China
| |
Collapse
|
24
|
Soria J, Mirshahi S, Mirshahi SQ, Varin R, Pritchard LL, Soria C, Mirshahi M. Fibrinogen αC domain: Its importance in physiopathology. Res Pract Thromb Haemost 2019; 3:173-183. [PMID: 31011701 PMCID: PMC6462745 DOI: 10.1002/rth2.12183] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Accepted: 11/22/2018] [Indexed: 12/17/2022] Open
Abstract
ABSTRACT Fibrinogen, involved in coagulation, is a soluble protein composed of two sets of disulfide-bridged Aα, Bβ, and γ-chains. In this review, we present the clinical implications of the αC domain of the molecule in Alzheimer's disease, hereditary renal amyloidosis and a number of thrombotic and hemorrhagic disorders. In Alzheimer's disease, amyloid beta peptide (Aβ) is increased and binds to the αC domain of normal fibrinogen, triggering increased fibrin(ogen) deposition in patients' brain parenchyma. In hereditary renal amyloidosis, fibrinogen is abnormal, with mutations located in the fibrinogen αC domain. The mutant αC domain derived from fibrinogen degradation folds incorrectly so that, in time, aggregates form, leading to amyloid deposits in the kidneys. In these patients, no thrombotic tendency has been observed. Abnormal fibrinogens with either a point mutation in the αC domain or a frameshift mutation resulting in absence of a part of the αC domain are often associated with either thrombotic events or bleeding. Mutation of an amino acid into cysteine (as in fibrinogens Dusart and Caracas V) or a frameshift mutation yielding an unpaired cysteine in the αC domain is often responsible for thrombotic events. Covalent binding of albumin to the unpaired cysteine via a disulphide bridge leads to decreased accessibility to the fibrinolytic enzymes, hence formation of poorly degradable fibrin clots, which explains the high incidence of thrombosis. In contrast, anomalies due to a frameshift mutation in the αC connector of the molecule, provoking deletion of a great part of the αC domain, are associated with bleeding.
Collapse
Affiliation(s)
- Jeannette Soria
- Laboratoire de recherche en Onco‐HématologieHôtel Dieu de ParisParisFrance
- INSERM U 965‐ CARTHôpital LariboisièreParisFrance
| | - Shahsoltan Mirshahi
- INSERM U 965‐ CARTHôpital LariboisièreParisFrance
- Diagnostica StagoGennevilliersFrance
| | | | - Remi Varin
- Faculté de Médecine et de PharmacieRouenFrance
| | - Linda L. Pritchard
- Laboratoire de recherche en Onco‐HématologieHôtel Dieu de ParisParisFrance
| | - Claudine Soria
- Laboratoire de recherche en Onco‐HématologieHôtel Dieu de ParisParisFrance
| | - Massoud Mirshahi
- Laboratoire de recherche en Onco‐HématologieHôtel Dieu de ParisParisFrance
- INSERM U 965‐ CARTHôpital LariboisièreParisFrance
| |
Collapse
|
25
|
Abstract
BACKGROUND The growing body of evidence indicating the heterogeneity of Alzheimer's disease (AD), coupled with disappointing clinical studies directed at a fit-for-all therapy, suggest that the development of a single magic cure suitable for all cases may not be possible. This calls for a shift in paradigm where targeted treatment is developed for specific AD subpopulations that share distinct genetic or pathological properties. Apolipoprotein E4 (apoE4), the most prevalent genetic risk factor of AD, is expressed in more than half of AD patients and is thus an important possible AD therapeutic target. REVIEW This review focuses initially on the pathological effects of apoE4 in AD, as well as on the corresponding cellular and animal models and the suggested cellular and molecular mechanisms which mediate them. The second part of the review focuses on recent apoE4-targeted (from the APOE gene to the apoE protein and its interactors) therapeutic approaches that have been developed in animal models and are ready to be translated to human. Further, the issue of whether the pathological effects of apoE4 are due to loss of protective function or due to gain of toxic function is discussed herein. It is possible that both mechanisms coexist, with certain constituents of the apoE4 molecule and/or its downstream signaling mediating a toxic effect, while others are associated with a loss of protective function. CONCLUSION ApoE4 is a promising AD therapeutic target that remains understudied. Recent studies are now paving the way for effective apoE4-directed AD treatment approaches.
Collapse
|
26
|
Page MJ, Thomson GJA, Nunes JM, Engelbrecht AM, Nell TA, de Villiers WJS, de Beer MC, Engelbrecht L, Kell DB, Pretorius E. Serum amyloid A binds to fibrin(ogen), promoting fibrin amyloid formation. Sci Rep 2019; 9:3102. [PMID: 30816210 PMCID: PMC6395759 DOI: 10.1038/s41598-019-39056-x] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Accepted: 01/16/2019] [Indexed: 12/15/2022] Open
Abstract
Complex associations exist between inflammation and thrombosis, with the inflammatory state tending to promote coagulation. Fibrinogen, an acute phase protein, has been shown to interact with the amyloidogenic ß-amyloid protein of Alzheimer’s disease. However, little is known about the association between fibrinogen and serum amyloid A (SAA), a highly fibrillogenic protein that is one of the most dramatically changing acute phase reactants in the circulation. To study the role of SAA in coagulation and thrombosis, in vitro experiments were performed where purified human SAA, in concentrations resembling a modest acute phase response, was added to platelet-poor plasma (PPP) and whole blood (WB), as well as purified and fluorescently labelled fibrinogen. Results from thromboelastography (TEG) suggest that SAA causes atypical coagulation with a fibrin(ogen)-mediated increase in coagulation, but a decreased platelet/fibrin(ogen) interaction. In WB scanning electron microscopy analysis, SAA mediated red blood cell (RBC) agglutination, platelet activation and clumping, but not platelet spreading. Following clot formation in PPP, the presence of SAA increased amyloid formation of fibrin(ogen) as determined both with auto-fluorescence and with fluorogenic amyloid markers, under confocal microcopy. SAA also binds to fibrinogen, as determined with a fluorescent-labelled SAA antibody and correlative light electron microscopy (CLEM). The data presented here indicate that SAA can affect coagulation by inducing amyloid formation in fibrin(ogen), as well as by propelling platelets to a more prothrombotic state. The discovery of these multiple and complex effects of SAA on coagulation invite further mechanistic analyses.
Collapse
Affiliation(s)
- Martin J Page
- Department of Physiological Sciences, Stellenbosch University, Stellenbosch Private Bag X1 Matieland, 7602, Stellenbosch, South Africa
| | - Greig J A Thomson
- Department of Physiological Sciences, Stellenbosch University, Stellenbosch Private Bag X1 Matieland, 7602, Stellenbosch, South Africa
| | - J Massimo Nunes
- Department of Physiological Sciences, Stellenbosch University, Stellenbosch Private Bag X1 Matieland, 7602, Stellenbosch, South Africa
| | - Anna-Mart Engelbrecht
- Department of Physiological Sciences, Stellenbosch University, Stellenbosch Private Bag X1 Matieland, 7602, Stellenbosch, South Africa
| | - Theo A Nell
- Department of Physiological Sciences, Stellenbosch University, Stellenbosch Private Bag X1 Matieland, 7602, Stellenbosch, South Africa
| | - Willem J S de Villiers
- Department of Physiological Sciences, Stellenbosch University, Stellenbosch Private Bag X1 Matieland, 7602, Stellenbosch, South Africa.,Department of Internal Medicine, Stellenbosch University, Stellenbosch, South Africa
| | - Maria C de Beer
- Department of Physiology, Saha Cardiovascular Research Center and Barnstable Brown Diabetes Center, University of Kentucky, Lexington, KY, USA
| | - Lize Engelbrecht
- Fluorescence Microscopy Unit, Central Analytical Facilities, Stellenbosch University, Stellenbosch, South Africa
| | - Douglas B Kell
- Department of Physiological Sciences, Stellenbosch University, Stellenbosch Private Bag X1 Matieland, 7602, Stellenbosch, South Africa. .,School of Chemistry, The University of Manchester, 131 Princess St, MANCHESTER M1 7DN, Lancs, UK. .,The Manchester Institute of Biotechnology, The University of Manchester, 131 Princess St, MANCHESTER M1 7DN, Lancs, UK. .,Department of Biochemistry, Institute of integrative Biology, Biosciences Building., University of Liverpool, Crown St, Liverpool, L69 7ZB, UK.
| | - Etheresia Pretorius
- Department of Physiological Sciences, Stellenbosch University, Stellenbosch Private Bag X1 Matieland, 7602, Stellenbosch, South Africa.
| |
Collapse
|
27
|
Ashton NJ, Nevado-Holgado AJ, Barber IS, Lynham S, Gupta V, Chatterjee P, Goozee K, Hone E, Pedrini S, Blennow K, Schöll M, Zetterberg H, Ellis KA, Bush AI, Rowe CC, Villemagne VL, Ames D, Masters CL, Aarsland D, Powell J, Lovestone S, Martins R, Hye A. A plasma protein classifier for predicting amyloid burden for preclinical Alzheimer's disease. SCIENCE ADVANCES 2019; 5:eaau7220. [PMID: 30775436 PMCID: PMC6365111 DOI: 10.1126/sciadv.aau7220] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Accepted: 12/19/2018] [Indexed: 05/03/2023]
Abstract
A blood-based assessment of preclinical disease would have huge potential in the enrichment of participants for Alzheimer's disease (AD) therapeutic trials. In this study, cognitively unimpaired individuals from the AIBL and KARVIAH cohorts were defined as Aβ negative or Aβ positive by positron emission tomography. Nontargeted proteomic analysis that incorporated peptide fractionation and high-resolution mass spectrometry quantified relative protein abundances in plasma samples from all participants. A protein classifier model was trained to predict Aβ-positive participants using feature selection and machine learning in AIBL and independently assessed in KARVIAH. A 12-feature model for predicting Aβ-positive participants was established and demonstrated high accuracy (testing area under the receiver operator characteristic curve = 0.891, sensitivity = 0.78, and specificity = 0.77). This extensive plasma proteomic study has unbiasedly highlighted putative and novel candidates for AD pathology that should be further validated with automated methodologies.
Collapse
Affiliation(s)
- Nicholas J. Ashton
- King’s College London, Institute of Psychiatry, Psychology and Neuroscience, Maurice Wohl Institute Clinical Neuroscience Institute, London, UK
- NIHR Biomedical Research Centre for Mental Health and Biomedical Research Unit for Dementia at South London and Maudsley NHS Foundation, London, UK
- Department of Psychiatry and Neurochemistry, Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden
| | | | | | - Steven Lynham
- Proteomics Core Facility, James Black Centre, King’s College, London, UK
| | - Veer Gupta
- School of Medical Sciences, Edith Cowan University, Joondalup, WA, Australia
- Cooperative Research Centre for Mental Health, Carlton South, VIC, Australia
- School of Medicine, Faculty of Health, Deakin University, 3220 VIC, Australia
| | - Pratishtha Chatterjee
- School of Medical Sciences, Edith Cowan University, Joondalup, WA, Australia
- KaRa Institute of Neurological Diseases, Macquarie Park, NSW, Australia
- Department of Biomedical Sciences, Macquarie University, 2109, NSW, Australia
| | - Kathryn Goozee
- KaRa Institute of Neurological Diseases, Macquarie Park, NSW, Australia
- Department of Biomedical Sciences, Macquarie University, 2109, NSW, Australia
- Clinical Research Department, Anglicare, Sydney, NSW, Australia
- School of Psychiatry and Clinical Neurosciences, University of Western Australia, WA, Australia
| | - Eugene Hone
- School of Medical Sciences, Edith Cowan University, Joondalup, WA, Australia
- Cooperative Research Centre for Mental Health, Carlton South, VIC, Australia
| | - Steve Pedrini
- School of Medical Sciences, Edith Cowan University, Joondalup, WA, Australia
- Cooperative Research Centre for Mental Health, Carlton South, VIC, Australia
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Michael Schöll
- Department of Psychiatry and Neurochemistry, Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Molecular Neuroscience, UCL Institute of Neurology, Queen Square, London, UK
- UK Dementia Research Institute at UCL, London, UK
| | - Kathryn A. Ellis
- Academic Unit for Psychiatry of Old Age, St. George’s Hospital, University of Melbourne, VIC, Australia
| | - Ashley I. Bush
- Cooperative Research Centre for Mental Health, Carlton South, VIC, Australia
- The Florey Institute, University of Melbourne, VIC, Australia
| | - Christopher C. Rowe
- Department of Molecular Imaging and Therapy, Austin Health, Heidelberg, VIC, Australia
| | - Victor L. Villemagne
- Department of Molecular Imaging and Therapy, Austin Health, Heidelberg, VIC, Australia
| | - David Ames
- Academic Unit for Psychiatry of Old Age, St. George’s Hospital, University of Melbourne, VIC, Australia
- National Ageing Research Institute, Parkville, VIC, Australia
| | | | - Dag Aarsland
- King’s College London, Institute of Psychiatry, Psychology and Neuroscience, Maurice Wohl Institute Clinical Neuroscience Institute, London, UK
- NIHR Biomedical Research Centre for Mental Health and Biomedical Research Unit for Dementia at South London and Maudsley NHS Foundation, London, UK
- Centre for Age-Related Medicine, Stavanger University Hospital, Stavanger, Norway
| | - John Powell
- King’s College London, Institute of Psychiatry, Psychology and Neuroscience, Maurice Wohl Institute Clinical Neuroscience Institute, London, UK
- NIHR Biomedical Research Centre for Mental Health and Biomedical Research Unit for Dementia at South London and Maudsley NHS Foundation, London, UK
| | | | - Ralph Martins
- School of Medical Sciences, Edith Cowan University, Joondalup, WA, Australia
- Cooperative Research Centre for Mental Health, Carlton South, VIC, Australia
- KaRa Institute of Neurological Diseases, Macquarie Park, NSW, Australia
- Department of Biomedical Sciences, Macquarie University, 2109, NSW, Australia
| | - Abdul Hye
- King’s College London, Institute of Psychiatry, Psychology and Neuroscience, Maurice Wohl Institute Clinical Neuroscience Institute, London, UK
- NIHR Biomedical Research Centre for Mental Health and Biomedical Research Unit for Dementia at South London and Maudsley NHS Foundation, London, UK
- Corresponding author.
| |
Collapse
|
28
|
Zheng C, Xu R. The Alzheimer's comorbidity phenome: mining from a large patient database and phenome-driven genetics prediction. JAMIA Open 2018; 2:131-138. [PMID: 30944912 PMCID: PMC6434979 DOI: 10.1093/jamiaopen/ooy050] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Revised: 10/23/2018] [Accepted: 12/05/2018] [Indexed: 01/08/2023] Open
Abstract
Objective Alzheimer’s disease (AD) is a severe neurodegenerative disorder and has become a global public health problem. Intensive research has been conducted for AD. But the pathophysiology of AD is still not elucidated. Disease comorbidity often associates diseases with overlapping patterns of genetic markers. This may inform a common etiology and suggest essential protein targets. US Food and Drug Administration (FDA) Adverse Event Reporting System (FAERS) collects large-scale postmarketing surveillance data that provide a unique opportunity to investigate disease co-occurrence pattern. We aim to construct a heterogeneous network that integrates disease comorbidity network (DCN) from FAERS with protein–protein interaction (PPI) to prioritize the AD risk genes using network-based ranking algorithm. Materials and Methods We built a DCN based on indication data from FAERS using association rule mining. DCN was further integrated with PPI network. We used random walk with restart ranking algorithm to prioritize AD risk genes. Results We evaluated the performance of our approach using AD risk genes curated from genetic association studies. Our approach achieved an area under a receiver operating characteristic curve of 0.770. Top 500 ranked genes achieved 5.53-fold enrichment for known AD risk genes as compared to random expectation. Pathway enrichment analysis using top-ranked genes revealed that two novel pathways, ERBB and coagulation pathways, might be involved in AD pathogenesis. Conclusion We innovatively leveraged FAERS, a comprehensive data resource for FDA postmarket drug safety surveillance, for large-scale AD comorbidity mining. This exploratory study demonstrated the potential of disease-comorbidities mining from FAERS in AD genetics discovery.
Collapse
Affiliation(s)
- Chunlei Zheng
- Department of Population and Quantitative Health Sciences, Institute of Computational Biology, School of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| | - Rong Xu
- Department of Population and Quantitative Health Sciences, Institute of Computational Biology, School of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| |
Collapse
|
29
|
Hara Y, McKeehan N, Fillit HM. Translating the biology of aging into novel therapeutics for Alzheimer disease. Neurology 2018; 92:84-93. [PMID: 30530798 PMCID: PMC6340342 DOI: 10.1212/wnl.0000000000006745] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Accepted: 08/23/2018] [Indexed: 12/11/2022] Open
Abstract
Aging is the leading risk factor for most chronic illnesses of old age, including Alzheimer disease (AD), a progressive neurodegenerative disease with currently no therapies that prevent, slow, or halt disease progression. Like other chronic diseases of old age, the progressive pathology of AD begins decades before the onset of symptoms. Many decades of research in biological gerontology have revealed common processes that are relevant to understanding why the aging brain is vulnerable to AD. In this review, we frame the development of novel therapeutics for AD in the context of biological gerontology. The many therapies currently in development based on biological gerontology principles provide promise for the development of a new generation of therapeutics to prevent and treat AD.
Collapse
Affiliation(s)
- Yuko Hara
- From the Alzheimer's Drug Discovery Foundation, New York, NY
| | | | - Howard M Fillit
- From the Alzheimer's Drug Discovery Foundation, New York, NY.
| |
Collapse
|
30
|
Kell DB, Pretorius E. No effects without causes: the Iron Dysregulation and Dormant Microbes hypothesis for chronic, inflammatory diseases. Biol Rev Camb Philos Soc 2018; 93:1518-1557. [PMID: 29575574 PMCID: PMC6055827 DOI: 10.1111/brv.12407] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Revised: 02/12/2018] [Accepted: 02/15/2018] [Indexed: 12/11/2022]
Abstract
Since the successful conquest of many acute, communicable (infectious) diseases through the use of vaccines and antibiotics, the currently most prevalent diseases are chronic and progressive in nature, and are all accompanied by inflammation. These diseases include neurodegenerative (e.g. Alzheimer's, Parkinson's), vascular (e.g. atherosclerosis, pre-eclampsia, type 2 diabetes) and autoimmune (e.g. rheumatoid arthritis and multiple sclerosis) diseases that may appear to have little in common. In fact they all share significant features, in particular chronic inflammation and its attendant inflammatory cytokines. Such effects do not happen without underlying and initially 'external' causes, and it is of interest to seek these causes. Taking a systems approach, we argue that these causes include (i) stress-induced iron dysregulation, and (ii) its ability to awaken dormant, non-replicating microbes with which the host has become infected. Other external causes may be dietary. Such microbes are capable of shedding small, but functionally significant amounts of highly inflammagenic molecules such as lipopolysaccharide and lipoteichoic acid. Sequelae include significant coagulopathies, not least the recently discovered amyloidogenic clotting of blood, leading to cell death and the release of further inflammagens. The extensive evidence discussed here implies, as was found with ulcers, that almost all chronic, infectious diseases do in fact harbour a microbial component. What differs is simply the microbes and the anatomical location from and at which they exert damage. This analysis offers novel avenues for diagnosis and treatment.
Collapse
Affiliation(s)
- Douglas B. Kell
- School of ChemistryThe University of Manchester, 131 Princess StreetManchesterLancsM1 7DNU.K.
- The Manchester Institute of BiotechnologyThe University of Manchester, 131 Princess StreetManchesterLancsM1 7DNU.K.
- Department of Physiological SciencesStellenbosch University, Stellenbosch Private Bag X1Matieland7602South Africa
| | - Etheresia Pretorius
- Department of Physiological SciencesStellenbosch University, Stellenbosch Private Bag X1Matieland7602South Africa
| |
Collapse
|
31
|
Chen LC, Tseng HJ, Liu CY, Huang YY, Yen CC, Weng JR, Lu YL, Hou WC, Lin TE, Pan IH, Huang KK, Huang WJ, Hsu KC. Design of Diarylheptanoid Derivatives as Dual Inhibitors Against Class IIa Histone Deacetylase and β-amyloid Aggregation. Front Pharmacol 2018; 9:708. [PMID: 30018556 PMCID: PMC6037852 DOI: 10.3389/fphar.2018.00708] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Accepted: 06/12/2018] [Indexed: 01/11/2023] Open
Abstract
Alzheimer’s disease (AD) is a progressive neurodegenerative disorder with multiple etiologies. Beta-amyloid (Aβ) self-aggregation and overexpression of class IIa histone deacetylases (HDACs) are strongly implicated with AD pathogenesis. In this study, a series of novel diarylheptanoid derivatives were designed, synthesized and evaluated for use as dual Aβ self-aggregation and class IIa HDAC inhibitors. Among these compounds, 4j, 5c, and 5e displayed effective inhibitions for Aβ self-aggregation, HDAC5 activity and HDAC7 activity with IC50 values of <10 μM. The compounds contain three common features: (1) a catechol or pyrogallol moiety, (2) a carbonyl linker and (3) an aromatic ring that can function as an HDAC cap and create hydrophobic interactions with Aβ1-42. Furthermore, compounds 4j, 5c, and 5e showed no significant cytotoxicity to human neuroblastoma SH-SY5Y cells and also exhibited neuroprotective effect against H2O2-induced toxicity. Overall, these promising in vitro data highlighted compounds 4j, 5c, and 5e as lead compounds that are worthy for further investigation.
Collapse
Affiliation(s)
- Liang-Chieh Chen
- Graduate Institute of Pharmacognosy, College of Pharmacy, Taipei Medical University, Taipei, Taiwan.,School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, Taiwan
| | - Hui-Ju Tseng
- Ph.D. Program in Biotechnology Research and Development, College of Pharmacy, Taipei Medical University, Taipei, Taiwan
| | - Chang-Yi Liu
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Yun-Yi Huang
- Graduate Institute of Pharmacognosy, College of Pharmacy, Taipei Medical University, Taipei, Taiwan
| | - Cheng-Chung Yen
- Graduate Institute of Pharmacognosy, College of Pharmacy, Taipei Medical University, Taipei, Taiwan
| | - Jing-Ru Weng
- Department of Marine Technology and Resources, College of Marine Sciences, National Sun Yat-sen University, Kaohsiung, Taiwan
| | - Yeh-Lin Lu
- Graduate Institute of Pharmacognosy, College of Pharmacy, Taipei Medical University, Taipei, Taiwan.,School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, Taiwan
| | - Wen-Chi Hou
- Graduate Institute of Pharmacognosy, College of Pharmacy, Taipei Medical University, Taipei, Taiwan
| | - Tony E Lin
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - I-Horng Pan
- Herbal Medicinal Product Division, Industrial Technology Research Institute, Hsinchu, Taiwan
| | - Kuo-Kuei Huang
- Herbal Medicinal Product Division, Industrial Technology Research Institute, Hsinchu, Taiwan
| | - Wei-Jan Huang
- Graduate Institute of Pharmacognosy, College of Pharmacy, Taipei Medical University, Taipei, Taiwan.,Ph.D. Program in Biotechnology Research and Development, College of Pharmacy, Taipei Medical University, Taipei, Taiwan.,Ph.D. Program for the Clinical Drug Discovery from Botanical Herbs, College of Pharmacy, Taipei Medical University, Taipei, Taiwan.,School of Pharmacy, National Defense Medical Center, Taipei, Taiwan
| | - Kai-Cheng Hsu
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
32
|
Singh PK, Kawasaki M, Berk-Rauch HE, Nishida G, Yamasaki T, Foley MA, Norris EH, Strickland S, Aso K, Ahn HJ. Aminopyrimidine Class Aggregation Inhibitor Effectively Blocks Aβ-Fibrinogen Interaction and Aβ-Induced Contact System Activation. Biochemistry 2018; 57:1399-1409. [PMID: 29394041 DOI: 10.1021/acs.biochem.7b01214] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Accumulating evidence suggests that fibrinogen, a key protein in the coagulation cascade, plays an important role in circulatory dysfunction in Alzheimer's disease (AD). Previous work has shown that the interaction between fibrinogen and β-amyloid (Aβ), a hallmark pathological protein in AD, induces plasmin-resistant abnormal blood clots, delays fibrinolysis, increases inflammation, and aggravates cognitive function in mouse models of AD. Since Aβ oligomers have a much stronger affinity for fibrinogen than Aβ monomers, we tested whether amyloid aggregation inhibitors could block the Aβ-fibrinogen interaction and found that some Aβ aggregation inhibitors showed moderate inhibitory efficacy against this interaction. We then modified a hit compound so that it not only showed a strong inhibitory efficacy toward the Aβ-fibrinogen interaction but also retained its potency toward the Aβ42 aggregation inhibition process. Furthermore, our best hit compound, TDI-2760, modulated Aβ42-induced contact system activation, a pathological condition observed in some AD patients, in addition to inhibiting the Aβ-fibrinogen interaction and Aβ aggregation. Thus, TDI-2760 has the potential to lessen vascular abnormalities as well as Aβ aggregation-driven pathology in AD.
Collapse
Affiliation(s)
- Pradeep K Singh
- Patricia and John Rosenwald Laboratory of Neurobiology and Genetics, The Rockefeller University , New York, New York 10065, United States
| | - Masanori Kawasaki
- Tri-Institutional Therapeutics Discovery Institute , New York, New York 10021, United States
| | - Hanna E Berk-Rauch
- Patricia and John Rosenwald Laboratory of Neurobiology and Genetics, The Rockefeller University , New York, New York 10065, United States
| | - Goushi Nishida
- Tri-Institutional Therapeutics Discovery Institute , New York, New York 10021, United States
| | - Takeshi Yamasaki
- Tri-Institutional Therapeutics Discovery Institute , New York, New York 10021, United States
| | - Michael A Foley
- Tri-Institutional Therapeutics Discovery Institute , New York, New York 10021, United States
| | - Erin H Norris
- Patricia and John Rosenwald Laboratory of Neurobiology and Genetics, The Rockefeller University , New York, New York 10065, United States
| | - Sidney Strickland
- Patricia and John Rosenwald Laboratory of Neurobiology and Genetics, The Rockefeller University , New York, New York 10065, United States
| | - Kazuyoshi Aso
- Tri-Institutional Therapeutics Discovery Institute , New York, New York 10021, United States
| | - Hyung Jin Ahn
- Patricia and John Rosenwald Laboratory of Neurobiology and Genetics, The Rockefeller University , New York, New York 10065, United States
| |
Collapse
|