1
|
Jensen C, Maarup S, Poulsen HS, Hasselbalch B, Karsdal M, Svane IM, Lassen U, Willumsen N. Degradation fragments of Tau and type IV collagen as serum biomarkers in patients with recurrent glioblastoma treated with nivolumab and bevacizumab. Clin Transl Oncol 2024:10.1007/s12094-024-03775-z. [PMID: 39499486 DOI: 10.1007/s12094-024-03775-z] [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: 07/19/2024] [Accepted: 10/18/2024] [Indexed: 11/07/2024]
Abstract
PURPOSE There is an unmet need for new treatment options and biomarkers for patients with glioblastoma (GBM). Here we investigated three non-invasive biomarkers: type VI collagen degraded by granzyme B (C4G) and matrix metalloproteases (C4M), respectively, and ADAM10-degraded Tau (Tau-A). METHODS Biomarker levels in pre- and on-treatment serum samples from patients with recurrent GBM (n = 39) treated with nivolumab and bevacizumab (NCT03890952) were compared to healthy levels (n = 22) and associated with overall survival (OS) outcome (median cutpoint). Longitudinal changes in biomarkers were investigated by a Mixed-effects analysis. RESULTS Tau-A (p < 0.0001) and C4G (p = 0.005), but not C4M (p = 0.106), were increased in patients. High Tau-A and C4G associated with improved OS (Tau-A: HR = 0.41, 95%CI = 0.20-0.86, C4G: HR = 0.47, 95%CI = 0.24-0.94). Only C4G increased with treatment (p = 0.024-0.005). CONCLUSIONS Tau-A and C4G are elevated in serum from patients with recurrent GBM and prognostic for OS. If validated, these biomarkers could be applied to clinical trials.
Collapse
Affiliation(s)
| | - Simone Maarup
- The DCCC Brain Tumor Center, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
- Department of Oncology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Hans Skovgaard Poulsen
- The DCCC Brain Tumor Center, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
- Department of Oncology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Benedikte Hasselbalch
- The DCCC Brain Tumor Center, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
- Department of Oncology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | | | - Inge Marie Svane
- National Center for Cancer Immune Therapy, CCIT-DK, Copenhagen University Hospital, Herlev, Denmark
- Department of Oncology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Ulrik Lassen
- The DCCC Brain Tumor Center, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
- Department of Oncology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | | |
Collapse
|
2
|
Islam M, Shen F, Regmi D, Du D. Therapeutic strategies for tauopathies and drug repurposing as a potential approach. Biochem Pharmacol 2022; 198:114979. [PMID: 35219701 PMCID: PMC9159505 DOI: 10.1016/j.bcp.2022.114979] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 02/03/2022] [Accepted: 02/21/2022] [Indexed: 11/26/2022]
Abstract
Tauopathies are neurodegenerative diseases characterized by the deposition of abnormal tau in the brain. To date, there are no disease-modifying therapies approved by the U.S. Food and Drug Administration (US FDA) for the treatment of tauopathies. In the past decades, extensive efforts have been provided to develop disease-modifying therapies to treat tauopathies. Specifically, exploring existing drugs with the intent of repurposing for the treatment of tauopathies affords a reasonable alternative to discover potent drugs for treating these formidable diseases. Drug repurposing will not only reduce formulation and development stage effort and cost but will also take a key advantage of the established toxicological studies, which is one of the main causes of clinical trial failure of new molecules. In this review, we provide an overview of the current treatment strategies for tauopathies and the recent progress in drug repurposing as an alternative approach to treat tauopathies.
Collapse
Affiliation(s)
- Majedul Islam
- Department of Chemistry and Biochemistry, Florida Atlantic University, Boca Raton, FL 33431, United States.
| | - Fengyun Shen
- Department of Chemistry and Biochemistry, Florida Atlantic University, Boca Raton, FL 33431, United States
| | - Deepika Regmi
- Department of Chemistry and Biochemistry, Florida Atlantic University, Boca Raton, FL 33431, United States
| | - Deguo Du
- Department of Chemistry and Biochemistry, Florida Atlantic University, Boca Raton, FL 33431, United States.
| |
Collapse
|
3
|
Chen YD, Huang PY, Chiang CS, Huang YS, Tang SC. Generation and Role of Calpain-Cleaved 17-kDa Tau Fragment in Acute Ischemic Stroke. Mol Neurobiol 2021; 58:5814-5825. [PMID: 34414533 DOI: 10.1007/s12035-021-02519-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Accepted: 08/03/2021] [Indexed: 01/04/2023]
Abstract
Stroke is the leading cause of permanent disability and death in the world. The therapy for acute stroke is still limited due to the complex mechanisms underlying stroke-induced neuronal death. The generation of a 17-kDa neurotoxic tau fragment was reported in Alzheimer's disease but it has not been well studied in stroke. In this study, we observed the accumulation of 17-kDa tau fragment in cultured primary neurons and media after oxygen-glucose deprivation/reperfusion (OGD/R) treatment that could be diminished by the presence of a calpain inhibitor. This calpain-mediated proteolytic tau fragment was also detected in brain tissues from middle cerebral artery occlusion-injured rats and acute ischemic stroke patients receiving strokectomy, and human plasma samples collected within 48 h after the onset of stroke. The mass spectrometry analysis of this 17-kDa fragment identified 2 peptide sequences containing 195-224 amino acids of tau, which agrees with the previously reported tau45-230 or tau125-230 as the calpain-cleaved tau fragment. Ectopic expression of tau45-230-GFP but not tau125-230-GFP in cultured neurons induced the formation of tortuous processes without evident cell death. In summary, the 17-kDa tau fragment is a novel stroke biomarker and may play a pathophysiological role to affect post-stroke neuronal health.
Collapse
Affiliation(s)
- Ying-Da Chen
- Stroke Center and Department of Neurology, National Taiwan University Hospital, Taipei, 100, Taiwan.,Institute of Biomedical Sciences, Academia Sinica, Taipei, 11529, Taiwan
| | - Po-Yuan Huang
- Stroke Center and Department of Neurology, National Taiwan University Hospital, Taipei, 100, Taiwan
| | - Chien-Sung Chiang
- Stroke Center and Department of Neurology, National Taiwan University Hospital, Taipei, 100, Taiwan
| | - Yi-Shuian Huang
- Institute of Biomedical Sciences, Academia Sinica, Taipei, 11529, Taiwan. .,Neuroscience Program of Academia Sinica, Academia Sinica, Taipei, 11529, Taiwan.
| | - Sung-Chun Tang
- Stroke Center and Department of Neurology, National Taiwan University Hospital, Taipei, 100, Taiwan.
| |
Collapse
|
4
|
Sun L, Bhawal R, Xu H, Chen H, Anderson ET, Haroutunian V, Cross AC, Zhang S, Gibson GE. The human brain acetylome reveals that decreased acetylation of mitochondrial proteins associates with Alzheimer's disease. J Neurochem 2021; 158:282-296. [PMID: 33905124 DOI: 10.1111/jnc.15377] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 04/19/2021] [Indexed: 11/28/2022]
Abstract
Metabolic changes that correlate to cognitive changes are well-known in Alzheimer's disease (AD). Metabolism is often linked to functional changes in proteins by post-translational modifications. The importance of the regulation of transcription by acetylation is well documented. Advanced mass spectrometry reveals hundreds of acetylated proteins in multiple tissues, but the acetylome of human brain, its functional significance, and the changes with disease are unknown. Filling this gap is critical for understanding the pathophysiology and development of therapies. To fill this gap, we assessed the human brain acetylome in human brain and its changes with AD. More than 5% of the 4,442 proteins from the human brain global proteome were acetylated. Acetylated proteins were primarily found in the cytosol (148), mitochondria (100), nucleus (91), and plasma membrane (58). The comparison of the brain acetylome in controls to that of patients with AD revealed striking and selective differences in terms of its abundances of acetylated peptides/sites. Acetylation of 18 mitochondrial proteins decreased, while acetylation of two cytosolic proteins, tau and GFAP, increased. Our experiments demonstrate that acetylation at some specific lysine sites alters enzyme function. The results indicate that general activation of de-acetylases (i.e., sirtuins) is not an appropriate therapeutic approach for AD.
Collapse
Affiliation(s)
- Lidan Sun
- Brain and Mind Research Institute, Weill Cornell Medicine, Burke Neurological Institute, White Plains, NY, USA.,Integrated Medicine Research Center for Neurological Rehabilitation, College of Medicine, Jiaxing University, Jiaxing, China
| | - Ruchika Bhawal
- Proteomics and Metabolomics Facility, Institute of Biotechnology, Cornell University, Ithaca, NY, USA
| | - Hui Xu
- Brain and Mind Research Institute, Weill Cornell Medicine, Burke Neurological Institute, White Plains, NY, USA
| | - Huanlian Chen
- Brain and Mind Research Institute, Weill Cornell Medicine, Burke Neurological Institute, White Plains, NY, USA
| | - Elizabeth T Anderson
- Proteomics and Metabolomics Facility, Institute of Biotechnology, Cornell University, Ithaca, NY, USA
| | - Vahrum Haroutunian
- Department of Psychiatry, The Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Mental Illness Research, Education and Clinical Center, James J Peters VA Medical Center, Bronx, NY, USA.,Department of Neuroscience, The Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Abigail C Cross
- Brain and Mind Research Institute, Weill Cornell Medicine, Burke Neurological Institute, White Plains, NY, USA
| | - Sheng Zhang
- Proteomics and Metabolomics Facility, Institute of Biotechnology, Cornell University, Ithaca, NY, USA
| | - Gary E Gibson
- Brain and Mind Research Institute, Weill Cornell Medicine, Burke Neurological Institute, White Plains, NY, USA
| |
Collapse
|
5
|
Jonesco DS, Karsdal MA, Henriksen K. The CNS-specific proteoglycan, brevican, and its ADAMTS4-cleaved fragment show differential serological levels in Alzheimer's disease, other types of dementia and non-demented controls: A cross-sectional study. PLoS One 2020; 15:e0234632. [PMID: 32559242 PMCID: PMC7304580 DOI: 10.1371/journal.pone.0234632] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Accepted: 05/29/2020] [Indexed: 02/05/2023] Open
Abstract
Evidence indicate that the brain-specific protein, brevican, is proteolytically cleaved during neurodegeneration, hence positioning fragments of brevican as potential blood biomarkers of neurodegenerative diseases, such as dementia. We aimed to develop two assays capable of detecting the brevican N-terminal (N-Brev) and the ADAMTS4-generated fragment (Brev-A), cleaved at Ser401, in serum and to perform a preliminary assessment of their diagnostic potential in dementias. Monoclonal antibodies against N-Brev and Brev-A were used to develop two ELISAs detecting each epitope. A comparison of brevican fragments in serum from individuals with AD (n = 28), other dementia (OD) (n = 41), and non-dementia-related memory complaints (NDCs) (n = 48) was conducted. Anti-N-Brev and anti-Brev-A antibodies selectively recognized their targets and dilution and spike recoveries were within limits of ±20%. Intra- and inter-assay CVs were below limits of 10% and 15%, respectively. For the N-Brev biomarker, serum from patients with OD showed significantly lower levels than those with AD (p = 0.05) and NDCs (p < 0.01). The opposite pattern was evident for Brev-A: serum levels in patients with OD were significantly higher than for AD (p = 0.04) and NDCs (p = 0.01). For both N-Brev and Brev-A, levels did not differ between AD and NDCs. The ratio of N-Brev/Brev-A resulted in increased significant differences between OD and AD (p < 0.01) and between OD and NDCs (p < 0.0001). The ratio discriminated between NDCs and OD (AUC: 0.75, 95% CI: 0.65-0.85, p < 0.0001) and between OD and AD (AUC: 0.72, 95% CI: 0.59-0.85, p < 0.01). In conclusion, we developed the first assays detecting the N-terminal of brevican as well as an ADAMTS4-cleaved fragment of brevican in blood. Differential levels of N-Brev and Brev-A between AD and OD allow for these biomarkers to possibly distinguish between different forms of dementias.
Collapse
Affiliation(s)
| | | | - Kim Henriksen
- Biomarkers & Research, Nordic Bioscience, Herlev, Denmark
| |
Collapse
|
6
|
Kim H, Lee JU, Kim S, Song S, Sim SJ. A Nanoplasmonic Biosensor for Ultrasensitive Detection of Alzheimer's Disease Biomarker Using a Chaotropic Agent. ACS Sens 2019; 4:595-602. [PMID: 30747516 DOI: 10.1021/acssensors.8b01242] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Blood-based diagnosis (hemodiagnosis) of Alzheimer's disease (AD) is emerging as a promising alternative to cerebrospinal-fluid-based methods because blood contains various kinds of AD biomarkers, including amyloid beta 1-40, 1-42, and τ (tau) protein. However, with current technology, the accuracy of the blood-plasma-based methods is relatively low compared to the traditional methods because the concentration of AD biomarkers in blood plasma is incredibly low, and diverse interference is present in blood plasma, which hinders precise detection. Here, we suggest a nanoplasmonic biosensor using gold nanorods with a chaotropic agent for precise ultrasensitive detecting of Alzheimer's disease biomarkers in human plasma. This nanoplasmonic biosensor is based on the localized surface plasmon resonance (LSPR), which is extremely sensitive to the point where it can respond to an insignificant change of the refractive index around the gold nanoparticles. Also, using guanidine hydrochloride as a chaotropic agent, we can overcome the obstacles of blood-based AD diagnostics. In more detail, this agent interrupts the network between water molecules and weakens the hydrophobic interactions between proteins, remarkably improving detection capabilities to target τ protein. By reducing the overlapping ranges between protein levels in an age-matched control and AD patients' plasma, this system can accurately diagnose AD patients. This platform also can analyze disease from mild cognitive impairment using standardized blood biomarker tau protein, which is related to Alzheimer's disease. As a result, our platform can be applied to clinical trials, and thus it has excellent potential in the medical field.
Collapse
Affiliation(s)
- Hanbi Kim
- Department of Chemical and Biological Engineering, Korea University, Seoul 02841, South Korea
| | - Jong Uk Lee
- Department of Chemical and Biological Engineering, Korea University, Seoul 02841, South Korea
| | - Soohyun Kim
- Department of Chemical and Biological Engineering, Korea University, Seoul 02841, South Korea
| | - Sojin Song
- Department of Chemical and Biological Engineering, Korea University, Seoul 02841, South Korea
| | - Sang Jun Sim
- Department of Chemical and Biological Engineering, Korea University, Seoul 02841, South Korea
| |
Collapse
|
7
|
Hampel H, O'Bryant SE, Molinuevo JL, Zetterberg H, Masters CL, Lista S, Kiddle SJ, Batrla R, Blennow K. Blood-based biomarkers for Alzheimer disease: mapping the road to the clinic. Nat Rev Neurol 2018; 14:639-652. [PMID: 30297701 PMCID: PMC6211654 DOI: 10.1038/s41582-018-0079-7] [Citation(s) in RCA: 440] [Impact Index Per Article: 62.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Biomarker discovery and development for clinical research, diagnostics and therapy monitoring in clinical trials have advanced rapidly in key areas of medicine - most notably, oncology and cardiovascular diseases - allowing rapid early detection and supporting the evolution of biomarker-guided, precision-medicine-based targeted therapies. In Alzheimer disease (AD), breakthroughs in biomarker identification and validation include cerebrospinal fluid and PET markers of amyloid-β and tau proteins, which are highly accurate in detecting the presence of AD-associated pathophysiological and neuropathological changes. However, the high cost, insufficient accessibility and/or invasiveness of these assays limit their use as viable first-line tools for detecting patterns of pathophysiology. Therefore, a multistage, tiered approach is needed, prioritizing development of an initial screen to exclude from these tests the high numbers of people with cognitive deficits who do not demonstrate evidence of underlying AD pathophysiology. This Review summarizes the efforts of an international working group that aimed to survey the current landscape of blood-based AD biomarkers and outlines operational steps for an effective academic-industry co-development pathway from identification and assay development to validation for clinical use.
Collapse
Affiliation(s)
- Harald Hampel
- AXA Research Fund and Sorbonne University Chair, Paris, France.
- Sorbonne University, GRC n° 21, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Paris, France.
- Brain & Spine Institute (ICM), INSERM U 1127, Paris, France.
- Institute of Memory and Alzheimer's Disease (IM2A), Department of Neurology, AP-HP, Pitié-Salpêtrière Hospital, Paris, France.
| | - Sid E O'Bryant
- University of North Texas Health Science Center, Fort Worth, TX, USA
| | - José L Molinuevo
- Barcelonaβeta Brain Research Center, Pasqual Maragall Foundation, Barcelona, Spain
| | - Henrik Zetterberg
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Department of Molecular Neuroscience, UCL Institute of Neurology, Queen Square, London, UK
- UK Dementia Research Institute at UCL, London, UK
| | - Colin L Masters
- The Florey Institute, The University of Melbourne, Melbourne, Australia
| | - Simone Lista
- AXA Research Fund and Sorbonne University Chair, Paris, France
- Sorbonne University, GRC n° 21, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Paris, France
- Brain & Spine Institute (ICM), INSERM U 1127, Paris, France
- Institute of Memory and Alzheimer's Disease (IM2A), Department of Neurology, AP-HP, Pitié-Salpêtrière Hospital, Paris, France
| | - Steven J Kiddle
- Department of Biostatistics and Health Informatics, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
- MRC Biostatistics Unit, Cambridge Institute of Public Health, University of Cambridge, Cambridge, UK
| | | | - Kaj Blennow
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden.
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden.
| |
Collapse
|
8
|
Bäckryd E, Edström S, Gerdle B, Ghafouri B. Do fragments and glycosylated isoforms of alpha-1-antitrypsin in CSF mirror spinal pathophysiological mechanisms in chronic peripheral neuropathic pain? An exploratory, discovery phase study. BMC Neurol 2018; 18:116. [PMID: 30115020 PMCID: PMC6097305 DOI: 10.1186/s12883-018-1116-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Accepted: 08/06/2018] [Indexed: 12/23/2022] Open
Abstract
Background Post-translational modifications (PTMs) generate a tremendous protein diversity from the ~ 20,000 protein-coding genes of the human genome. In chronic pain conditions, exposure to pathological processes in the central nervous system could lead to disease-specific PTMs detectable in the cerebrospinal fluid (CSF). In a previous hypothesis-generating study, we reported that seven out of 260 CSF proteins highly discriminated between neuropathic pain patients and healthy controls: one isoform of angiotensinogen (AG), two isoforms of alpha-1-antitrypsin (AT), three isoforms of haptoglobin (HG), and one isoform of pigment epithelium-derived factor (PEDF). The present study had three aims: (1) To examine the multivariate inter-correlations between all identified isoforms of these seven proteins; (2) Based on the results of the first aim, to characterize PTMs in a subset of interesting proteins; (3) To regress clinical pain data using the 260 proteins as predictors, thereby testing the hypothesis that the above-mentioned seven discriminating proteins and/or the characterized isoforms/fragments of aim (2) would be among the proteins having the highest predictive power for clinical pain data. Methods CSF samples from 11 neuropathic pain patients and 11 healthy controls were used for biochemical analysis of protein isoforms. PTM characterization was performed using enzymatic reaction assay and mass spectrometry. Multivariate data analysis (principal component analysis and orthogonal partial least square regression) was applied on the quantified protein isoforms. Results We identified 5 isoforms of AG, 18 isoforms of AT, 5 isoforms of HG, and 5 isoforms of PEDF. Fragments and glycosylated isoforms of AT were studied in depth. When regressing the pain intensity data of patients, three isoforms of AT, two isoforms of PEDF, and one isoform of angiotensinogen “reappeared” as major results, i.e., they were major findings both when comparing patients with healthy controls and when regressing pain intensity in patients. Conclusions Altered levels of fragments and/or glycosylated isoforms of alpha-1-antitrypsin might mirror pathophysiological processes in the spinal cord of neuropathic pain patients. In particular, we suggest that a putative disease-specific combination of the levels of two different N-truncated fragments of alpha-1-antitrypsin might be interesting for future CSF and/or plasma biomarker investigations in chronic neuropathic pain. Electronic supplementary material The online version of this article (10.1186/s12883-018-1116-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Emmanuel Bäckryd
- Pain and Rehabilitation Center, and Department of Medical and Health Sciences, Linköping University, Linköping, Sweden.
| | - Sofia Edström
- Pain and Rehabilitation Center, and Department of Medical and Health Sciences, Linköping University, Linköping, Sweden
| | - Björn Gerdle
- Pain and Rehabilitation Center, and Department of Medical and Health Sciences, Linköping University, Linköping, Sweden
| | - Bijar Ghafouri
- Pain and Rehabilitation Center, and Department of Medical and Health Sciences, Linköping University, Linköping, Sweden
| |
Collapse
|
9
|
Quinn JP, Corbett NJ, Kellett KAB, Hooper NM. Tau Proteolysis in the Pathogenesis of Tauopathies: Neurotoxic Fragments and Novel Biomarkers. J Alzheimers Dis 2018; 63:13-33. [PMID: 29630551 PMCID: PMC5900574 DOI: 10.3233/jad-170959] [Citation(s) in RCA: 108] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/31/2018] [Indexed: 12/11/2022]
Abstract
With predictions showing that 131.5 million people worldwide will be living with dementia by 2050, an understanding of the molecular mechanisms underpinning disease is crucial in the hunt for novel therapeutics and for biomarkers to detect disease early and/or monitor disease progression. The metabolism of the microtubule-associated protein tau is altered in different dementias, the so-called tauopathies. Tau detaches from microtubules, aggregates into oligomers and neurofibrillary tangles, which can be secreted from neurons, and spreads through the brain during disease progression. Post-translational modifications exacerbate the production of both oligomeric and soluble forms of tau, with proteolysis by a range of different proteases being a crucial driver. However, the impact of tau proteolysis on disease progression has been overlooked until recently. Studies have highlighted that proteolytic fragments of tau can drive neurodegeneration in a fragment-dependent manner as a result of aggregation and/or transcellular propagation. Proteolytic fragments of tau have been found in the cerebrospinal fluid and plasma of patients with different tauopathies, providing an opportunity to develop these fragments as novel disease progression biomarkers. A range of therapeutic strategies have been proposed to halt the toxicity associated with proteolysis, including reducing protease expression and/or activity, selectively inhibiting protease-substrate interactions, and blocking the action of the resulting fragments. This review highlights the importance of tau proteolysis in the pathogenesis of tauopathies, identifies putative sites during tau fragment-mediated neurodegeneration that could be targeted therapeutically, and discusses the potential use of proteolytic fragments of tau as biomarkers for different tauopathies.
Collapse
Affiliation(s)
- James P. Quinn
- Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - Nicola J. Corbett
- Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - Katherine A. B. Kellett
- Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - Nigel M. Hooper
- Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| |
Collapse
|
10
|
Platelet phosphorylated TDP-43: an exploratory study for a peripheral surrogate biomarker development for Alzheimer's disease. Future Sci OA 2017; 3:FSO238. [PMID: 29134122 PMCID: PMC5674277 DOI: 10.4155/fsoa-2017-0090] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Accepted: 07/28/2017] [Indexed: 12/13/2022] Open
Abstract
Aim: Alzheimer's disease (AD) and other forms of dementia create a noncurable disease population in world's societies. To develop a blood-based biomarker is important so that the remedial or disease-altering therapeutic intervention for AD patients would be available at the early stage. Materials & methods: TDP-43 levels were analyzed in postmortem brain tissue and platelets of AD and control subjects. Results: We observed an increased TDP-43 (<60%) in postmortem AD brain regions and similar trends were also observed in patient's platelets. Conclusion: Platelet TDP-43 could be used as a surrogate biomarker that is measurable, reproducible and sensitive for screening the patients with some early clinical signs of AD and can be used to monitor disease prognosis. In this study, we explore to identify an Alzheimer's disease (AD)-selective phospho-specific antibody that recognizes the diseased form of TDP-43 protein in patient's blood-derived platelets. Our results suggest that selective antiphosphorylated TDP-43 antibody discriminates AD from non-demented controls and patients with amyotrophic lateral sclerosis. Therefore, platelet screening with a selective antibody could potentially be a useful tool for diagnostic purposes for AD.
Collapse
|
11
|
Lin NH, Messing A, Perng MD. Characterization of a panel of monoclonal antibodies recognizing specific epitopes on GFAP. PLoS One 2017; 12:e0180694. [PMID: 28700643 PMCID: PMC5503259 DOI: 10.1371/journal.pone.0180694] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2017] [Accepted: 06/20/2017] [Indexed: 11/19/2022] Open
Abstract
Alexander disease (AxD) is a neurodegenerative disease caused by heterozygous mutations in the GFAP gene, which encodes the major intermediate filament protein of astrocytes. This disease is characterized by the accumulation of cytoplasmic protein aggregates, known as Rosenthal fibers. Antibodies specific to GFAP could provide invaluable tools to facilitate studies of the normal biology of GFAP and to elucidate the pathologic role of this IF protein in disease. While a large number of antibodies to GFAP are available, few if any of them have defined epitopes. Here we described the characterization of a panel of commonly used anti-GFAP antibodies, which recognized epitopes at regions extending across the rod domain of GFAP. We show that all of the antibodies are useful for immunoblotting and immunostaining, and identify a subset that preferentially recognized human GFAP. Using these antibodies, we demonstrate the presence of biochemically modified forms of GFAP in brains of human AxD patients and mouse AxD models. These data suggest that this panel of anti-GFAP antibodies will be useful for studies of animal and cell-based models of AxD and related diseases in which cytoskeletal defects associated with GFAP modifications occur.
Collapse
Affiliation(s)
- Ni-Hsuan Lin
- Institute of Molecular Medicine, College of Life Sciences, National Tsing Hua University, Hsinchu, Taiwan
| | - Albee Messing
- Waisman Center, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Ming-Der Perng
- Institute of Molecular Medicine, College of Life Sciences, National Tsing Hua University, Hsinchu, Taiwan
- * E-mail:
| |
Collapse
|
12
|
A Single-Use, In Vitro Biosensor for the Detection of T-Tau Protein, A Biomarker of Neuro-Degenerative Disorders, in PBS and Human Serum Using Differential Pulse Voltammetry (DPV). BIOSENSORS-BASEL 2017; 7:bios7010010. [PMID: 28218731 PMCID: PMC5371783 DOI: 10.3390/bios7010010] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Revised: 02/08/2017] [Accepted: 02/14/2017] [Indexed: 12/24/2022]
Abstract
A single-use, in vitro biosensor for the detection of T-Tau protein in phosphate-buffer saline (PBS) and undiluted human serum was designed, manufactured, and tested. Differential pulse voltammetry (DPV) served as the transduction mechanism. This biosensor consisted of three electrodes: working, counter, and reference electrodes fabricated on a PET sheet. Both working and counter electrodes were thin gold film, 10 nm in thickness. Laser ablation technique was used to define the size and structure of the biosensor. The biosensor was produced using cost-effective roll-to-roll process. Self-assembled monolayers (SAM) of 3-mercaptopropionic acid (MPA) were employed to covalently immobilize the anti-T-Tau (T-Tau antibody) on the gold working electrode. A carbodiimide conjugation approach using N-(3-dimethylaminopropyl)-N’-ethylcarbodiimide hydrochloride (EDC) and N–hydroxysuccinimide (NHS) cross-linked anti-T-Tau to the carboxylic groups on one end of the MPA. A T-Tau protein ladder with six isoforms was used in this study. The anti-T-Tau concentration used was 500,000 pg/mL. The T-Tau protein concentration ranged from 1000 pg/mL to 100,000 pg/mL. DPV measurements showed excellent responses, with a good calibration curve. Thus, a practical tool for simple detection of T-Tau protein, a biomarker of neuro-degenerative disorders, has been successfully developed. This tool could also be extended to detect other biomarkers for neuro-degenerative disorders, such as P-Tau protein and β-amyloid 42.
Collapse
|
13
|
Giacomelli C, Daniele S, Martini C. Potential biomarkers and novel pharmacological targets in protein aggregation-related neurodegenerative diseases. Biochem Pharmacol 2017; 131:1-15. [PMID: 28159621 DOI: 10.1016/j.bcp.2017.01.017] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Accepted: 01/26/2017] [Indexed: 10/20/2022]
Abstract
The aggregation of specific proteins plays a pivotal role in the etiopathogenesis of several neurodegenerative diseases (NDs). β-Amyloid (Aβ) peptide-containing plaques and intraneuronal neurofibrillary tangles composed of hyperphosphorylated protein tau are the two main neuropathological lesions in Alzheimer's disease. Meanwhile, Parkinson's disease is defined by the presence of intraneuronal inclusions (Lewy bodies), in which α-synuclein (α-syn) has been identified as a major protein component. The current literature provides considerable insights into the mechanisms underlying oligomeric-related neurodegeneration, as well as the relationship between protein aggregation and ND, thus facilitating the development of novel putative biomarkers and/or pharmacological targets. Recently, α-syn, tau and Aβ have been shown to interact each other or with other "pathological proteins" to form toxic heteroaggregates. These latest findings are overcoming the concept that each neurodegenerative disease is related to the misfolding of a single specific protein. In this review, potential opportunities and pharmacological approaches targeting α-syn, tau and Aβ and their oligomeric forms are highlighted with examples from recent studies. Protein aggregation as a biomarker of NDs, in both the brain and peripheral fluids, is deeply explored. Finally, the relationship between biomarker establishment and assessment and their use as diagnostics or therapeutic targets are discussed.
Collapse
Affiliation(s)
- Chiara Giacomelli
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy
| | - Simona Daniele
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy
| | - Claudia Martini
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy.
| |
Collapse
|
14
|
Grewal R, Haghighi M, Huang S, Smith AG, Cao C, Lin X, Lee DC, Teten N, Hill AM, Selenica MLB. Identifying biomarkers of dementia prevalent among amnestic mild cognitively impaired ethnic female patients. ALZHEIMERS RESEARCH & THERAPY 2016; 8:43. [PMID: 27756387 PMCID: PMC5067885 DOI: 10.1186/s13195-016-0211-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Accepted: 09/13/2016] [Indexed: 12/22/2022]
Abstract
Background There is a need to investigate biomarkers that are indicative of the progression of dementia in ethnic patient populations. The disparity of information in these populations has been the focus of many clinical and academic centers, including ours, to contribute to a higher success rate in clinical trials. In this study, we have investigated plasma biomarkers in amnestic mild cognitively impaired (aMCI) female patient cohorts in the context of ethnicity and cognitive status. Method A panel of 12 biomarkers involved in the progression of brain pathology, inflammation, and cardiovascular disorders were investigated in female cohorts of African American, Hispanic, and White aMCI patients. Both biochemical and algorithmic analyses were applied to correlate biomarker levels measured during the early stages of the disease for each ethnicity. Results We report elevated plasma Aβ40, Aβ42, YKL-40, and cystatin C levels in the Hispanic cohort at early aMCI status. In addition, elevated plasma Aβ40 levels were associated with the aMCI status in both White and African American patient cohorts by the decision tree algorithm. Eotaxin-1 levels, as determined by the decision tree algorithm and biochemically measured total tau levels, were associated with the aMCI status in the African American cohort. Conclusions Overall, our data displayed novel differences in the plasma biomarkers of the aMCI female cohorts where the plasma levels of several biomarkers distinguished between each ethnicity at an early aMCI stage. Identification of these plasma biomarkers encourages new areas of investigation among aMCI ethnic populations, including larger patient cohorts and longitudinal study designs. Electronic supplementary material The online version of this article (doi:10.1186/s13195-016-0211-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Rinko Grewal
- Byrd Alzheimer's Institute, University of South Florida, 4001 E. Fletcher Ave, Tampa, FL, 33613, USA.,Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, 12901 Bruce B. Downs Blvd, Tampa, FL, 33612, USA
| | - Mona Haghighi
- Department of Industrial and Systems Engineering, University of Washington, 3900 Northeast Stevens Way, Seattle, WA, 98195, USA
| | - Shuai Huang
- Department of Industrial and Systems Engineering, University of Washington, 3900 Northeast Stevens Way, Seattle, WA, 98195, USA.,School of Aging Studies, University of South Florida, 4202 E Fowler Ave, Tampa, FL, 33620, USA
| | - Amanda G Smith
- Byrd Alzheimer's Institute, University of South Florida, 4001 E. Fletcher Ave, Tampa, FL, 33613, USA.,Department of Psychiatry and Behavioral Medicine, College of Medicine, University of South Florida, 3515 E Fletcher Ave, Tampa, FL, 33613, USA
| | - Chuanhai Cao
- Byrd Alzheimer's Institute, University of South Florida, 4001 E. Fletcher Ave, Tampa, FL, 33613, USA.,Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, 12901 Bruce B. Downs Blvd, Tampa, FL, 33612, USA
| | - Xiaoyang Lin
- Byrd Alzheimer's Institute, University of South Florida, 4001 E. Fletcher Ave, Tampa, FL, 33613, USA.,Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, 12901 Bruce B. Downs Blvd, Tampa, FL, 33612, USA
| | - Daniel C Lee
- Byrd Alzheimer's Institute, University of South Florida, 4001 E. Fletcher Ave, Tampa, FL, 33613, USA.,Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, 12901 Bruce B. Downs Blvd, Tampa, FL, 33612, USA
| | - Nancy Teten
- Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, 12901 Bruce B. Downs Blvd, Tampa, FL, 33612, USA
| | - Angela M Hill
- Byrd Alzheimer's Institute, University of South Florida, 4001 E. Fletcher Ave, Tampa, FL, 33613, USA.,Department of Pharmacotherapeutics and Clinical Research, College of Pharmacy, University of South Florida, 12901 Bruce B. Downs Blvd, Tampa, FL, 33612, USA
| | - Maj-Linda B Selenica
- Byrd Alzheimer's Institute, University of South Florida, 4001 E. Fletcher Ave, Tampa, FL, 33613, USA. .,Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, 12901 Bruce B. Downs Blvd, Tampa, FL, 33612, USA.
| |
Collapse
|
15
|
Herbert J, Lucassen PJ. Depression as a risk factor for Alzheimer's disease: Genes, steroids, cytokines and neurogenesis - What do we need to know? Front Neuroendocrinol 2016; 41:153-71. [PMID: 26746105 DOI: 10.1016/j.yfrne.2015.12.001] [Citation(s) in RCA: 91] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Revised: 12/23/2015] [Accepted: 12/27/2015] [Indexed: 01/18/2023]
Abstract
Depression (MDD) is prodromal to, and a component of, Alzheimer's disease (AD): it may also be a trigger for incipient AD. MDD is not a unitary disorder, so there may be particular subtypes of early life MDD that pose independent high risks for later AD, though the identification of these subtypes is problematical. There may either be a common pathological event underlying both MDD and AD, or MDD may sensitize the brain to a second event ('hit') that precipitates AD. MDD may also accelerate brain ageing, including altered DNA methylation, increased cortisol but decreasing DHEA and thus the risk for AD. So far, genes predicting AD (e.g. APOEε4) are not risk factors for MDD, and those implicated in MDD (e.g. SLC6A4) are not risks for AD, so a common genetic predisposition looks unlikely. There is as yet no strong indication that an epigenetic event occurs during some forms of MDD that predisposes to later AD, though the evidence is limited. Glucocorticoids (GCs) are disturbed in some cases of MDD and in AD. GCs have marked degenerative actions on the hippocampus, a site of early β-amyloid deposition, and rare genetic variants of GC-regulating enzymes (e.g. 11β-HSD) predispose to AD. GCs also inhibit hippocampal neurogenesis and plasticity, and thus episodic memory, a core symptom of AD. Disordered GCs in MDD may inhibit neurogenesis, but the contribution of diminished neurogenesis to the onset or progression of AD is still debated. GCs and cytokines also reduce BDNF, implicated in both MDD and AD and hippocampal neurogenesis, reinforcing the notion that those cases of MDD with disordered GCs may be a risk for AD. Cytokines, including IL1β, IL6 and TNFα, are increased in the blood in some cases of MDD. They also reduce hippocampal neurogenesis, and increased cytokines are a known risk for later AD. Inflammatory changes occur in both MDD and AD (e.g. raised CRP, TNFα). Both cytokines and GCs can have pro-inflammatory actions in the brain. Inflammation (e.g. microglial activation) may be a common link, but this has not been systematically investigated. We lack substantial, rigorous and comprehensive follow-up studies to better identify possible subtypes of MDD that may represent a major predictor for later AD. This would enable specific interventions during critical episodes of these subtypes of MDD that should reduce this substantial risk.
Collapse
Affiliation(s)
- Joe Herbert
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, UK.
| | - Paul J Lucassen
- Swammerdam Institute for Life Sciences, Center for Neuroscience, University of Amsterdam, The Netherlands
| |
Collapse
|
16
|
Grabrucker AM, Ruozi B, Belletti D, Pederzoli F, Forni F, Vandelli MA, Tosi G. Nanoparticle transport across the blood brain barrier. Tissue Barriers 2016; 4:e1153568. [PMID: 27141426 DOI: 10.1080/21688370.2016.1153568] [Citation(s) in RCA: 117] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2015] [Revised: 02/02/2016] [Accepted: 02/04/2016] [Indexed: 01/13/2023] Open
Abstract
While the role of the blood-brain barrier (BBB) is increasingly recognized in the (development of treatments targeting neurodegenerative disorders, to date, few strategies exist that enable drug delivery of non-BBB crossing molecules directly to their site of action, the brain. However, the recent advent of Nanomedicines may provide a potent tool to implement CNS targeted delivery of active compounds. Approaches for BBB crossing are deeply investigated in relation to the pathology: among the main important diseases of the CNS, this review focuses on the application of nanomedicines to neurodegenerative disorders (Alzheimer, Parkinson and Huntington's Disease) and to other brain pathologies as epilepsy, infectious diseases, multiple sclerosis, lysosomal storage disorders, strokes.
Collapse
Affiliation(s)
- Andreas M Grabrucker
- WG Molecular Analysis of Synaptopathies, Neurology Dept, Neurocenter of Ulm University , Ulm, Germany
| | - Barbara Ruozi
- Pharmaceutical Technology, Te.Far.T.I. Group, Department of Life Sciences, University of Modena and Reggio Emilia ; Modena, Italy
| | - Daniela Belletti
- Pharmaceutical Technology, Te.Far.T.I. Group, Department of Life Sciences, University of Modena and Reggio Emilia ; Modena, Italy
| | - Francesca Pederzoli
- Pharmaceutical Technology, Te.Far.T.I. Group, Department of Life Sciences, University of Modena and Reggio Emilia ; Modena, Italy
| | - Flavio Forni
- Pharmaceutical Technology, Te.Far.T.I. Group, Department of Life Sciences, University of Modena and Reggio Emilia ; Modena, Italy
| | - Maria Angela Vandelli
- Pharmaceutical Technology, Te.Far.T.I. Group, Department of Life Sciences, University of Modena and Reggio Emilia ; Modena, Italy
| | - Giovanni Tosi
- Pharmaceutical Technology, Te.Far.T.I. Group, Department of Life Sciences, University of Modena and Reggio Emilia ; Modena, Italy
| |
Collapse
|