101
|
Chung CC, Chan L, Chen JH, Bamodu OA, Hong CT. Neurofilament light chain level in plasma extracellular vesicles and Parkinson's disease. Ther Adv Neurol Disord 2020; 13:1756286420975917. [PMID: 33335563 PMCID: PMC7724268 DOI: 10.1177/1756286420975917] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 10/30/2020] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Neurofilament light chain (NfL) is essential for axonal maintenance and reflects neuronal damage. Extracellular vesicles (EVs), especially exosomes, secreted by cells into the blood, are emerging as novel biomedical research platforms of physiological and pathological processes. The present study investigated the possible association between plasma EV NfL and Parkinson's disease (PD). METHODS One hundred and sixteen patients with mild to moderate PD and 46 non-PD, neurological controls were recruited, and their clinical motor symptoms and cognitive function were evaluated. Plasma EVs were isolated using an exoEasy kit, and immunomagnetic reduction assay was used to assess EV NfL level. Statistical analysis was performed using SPSS 25.0, and p < 0.05 was considered significant. RESULTS The isolated plasma EVs were validated according to size and the presence of specific surface markers. Compared with the neurological control group, the levels of plasma EV NfL in patients with PD were not significantly different (PD: 9.42 ± 3.89, control: 9.53 ± 3.62 pg/mL plasma, p = 0.71). On the other hand, plasma EV NfL in patients with PD trendwise correlated with the severity of akinetic rigidity (p = 0.05). PD patients with optimal EV NfL (lowest quartile) had 6.66 ± 2.08 lower Unified Parkinson's Disease Rating Scale-III score after adjustment for age, sex, and disease duration. CONCLUSION Plasma EV NfL levels did not distinguish patients with PD from the neurological control group. The possible correlation between plasma EV NfL with the severity of motor symptoms within the PD patients, especially with akinetic rigidity, was noted. Further clinical validation of the blood EV NfL by a longitudinal follow-up study of PD patients is warranted.
Collapse
Affiliation(s)
- Chen-Chih Chung
- Department of Neurology, Shuang Ho Hospital,
Taipei Medical University, New Taipei City Department of Neurology, School
of Medicine, College of Medicine, Taipei Medical University, Taipei City
Graduate Institute of Biomedical Informatics, Taipei Medical University,
Taipei City
| | - Lung Chan
- Department of Neurology, Shuang Ho Hospital,
Taipei Medical University, New Taipei City Department of Neurology, School
of Medicine, College of Medicine, Taipei Medical University, Taipei
City
| | - Jia-Hung Chen
- Department of Neurology, Shuang Ho Hospital,
Taipei Medical University, New Taipei City
| | - Oluwaseun Adebayo Bamodu
- Department of Hematology & Oncology, Shuang
Ho Hospital, Taipei Medical University, New Taipei City Department of
Medical Research & Education, Shuang Ho Hospital, Taipei Medical
University, New Taipei City Department of Urology, Shuang Ho Hospital,
Taipei Medical University, New Taipei City
| | - Chien-Tai Hong
- Department of Neurology, Shuang Ho Hospital,
Taipei Medical University, No. 291, Zhongzheng Rd, Zhonghe District, New
Taipei City 23561 Department of Neurology, School of Medicine, College of
Medicine, Taipei Medical University, Taipei City
| |
Collapse
|
102
|
Chou SY, Chan L, Chung CC, Chiu JY, Hsieh YC, Hong CT. Altered Insulin Receptor Substrate 1 Phosphorylation in Blood Neuron-Derived Extracellular Vesicles From Patients With Parkinson's Disease. Front Cell Dev Biol 2020; 8:564641. [PMID: 33344443 PMCID: PMC7744811 DOI: 10.3389/fcell.2020.564641] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 11/12/2020] [Indexed: 01/28/2023] Open
Abstract
INTRODUCTION Diabetes increases the risk of Parkinson's disease (PD). The phosphorylation of type 1 insulin receptor substrate (IRS-1) determines the function of insulin signaling pathway. Extracellular vesicles (EVs) are emerging as biomarkers of human diseases. The present study investigated whether PD patients exert altered phosphorylation IRS-1 (p-IRS-1) inside the blood neuron-derived extracellular vesicles (NDEVs). RESEARCH DESIGN AND METHODS In total, there were 94 patients with PD and 63 healthy controls recruited and their clinical manifestations were evaluated. Blood NDEVs were isolated using the immunoprecipitation method, and Western blot analysis was conducted to assess total IRS-1, p-IRS-1, and downstream substrates level in blood NDEVs. Statistical analysis was performed using SPSS 19.0, and p < 0.05 was considered significant. RESULTS The isolated blood EVs were validated according to the presence of CD63 and HSP70, nanoparticle tracking analysis and transmission electron microscopy. NDEVs were positive with neuronal markers. PD patients exerted significantly higher level of p-IRS-1S312 in blood NDEVs than controls. In addition, the p-IRS-1S312 levels in blood NDEVs was positively associated with the severity of tremor in PD patients after adjusting of age, sex, hemoglobin A1c, and body mass index (BMI). CONCLUSION PD patients exerted altered p-IRS-1S312 in the blood NDEVs, and also correlated with the severity of tremor. These findings suggested the association between dysfunctional insulin signaling pathway with PD. The role of altered p-IRS-1S312 in blood NDEVs as a segregating biomarker of PD required further cohort study to assess the association with the progression of PD.
Collapse
Affiliation(s)
- Szu-Yi Chou
- Graduate Institute of Neural Regenerative Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
- Ph.D. Program for Neural Regenerative Medicine, College of Medical Science and Technology, Taipei Medical University and National Health Research Institutes, Taipei, Taiwan
| | - Lung Chan
- Department of Neurology, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
- Department of Neurology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Chen-Chih Chung
- Department of Neurology, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
- Department of Neurology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Graduate Institute of Biomedical Informatics, Taipei Medical University, Taipei, Taiwan
| | - Jing-Yuan Chiu
- Graduate Institute of Neural Regenerative Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Yi-Chen Hsieh
- Graduate Institute of Neural Regenerative Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
- Master Program in Applied Molecular Epidemiology, College of Public Health, Taipei Medical University, Taipei, Taiwan
| | - Chien-Tai Hong
- Department of Neurology, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
- Department of Neurology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
103
|
Arrant AE, Davis SE, Vollmer RM, Murchison CF, Mobley JA, Nana AL, Spina S, Grinberg LT, Karydas AM, Miller BL, Seeley WW, Roberson ED. Elevated levels of extracellular vesicles in progranulin-deficient mice and FTD-GRN Patients. Ann Clin Transl Neurol 2020; 7:2433-2449. [PMID: 33197149 PMCID: PMC7732244 DOI: 10.1002/acn3.51242] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 09/22/2020] [Accepted: 10/18/2020] [Indexed: 12/13/2022] Open
Abstract
OBJECTIVE The goal of this study was to investigate the effect of progranulin insufficiency on extracellular vesicles (EVs), a heterogeneous population of vesicles that may contribute to progression of neurodegenerative disease. Loss-of-function mutations in progranulin (GRN) are a major cause of frontotemporal dementia (FTD), and brains from GRN carriers with FTD (FTD-GRN) exhibit signs of lysosomal dysfunction. Lysosomal dysfunction may induce compensatory increases in secretion of exosomes, EVs secreted from the endolysosomal system, so we hypothesized that progranulin insufficiency would increase EV levels in the brain. METHODS We analyzed levels and protein contents of brain EVs from Grn-/- mice, which model the lysosomal abnormalities of FTD-GRN patients. We then measured brain EVs in FTD-GRN patients. To assess the relationship of EVs with symptomatic disease, we measured plasma EVs in presymptomatic and symptomatic GRN mutation carriers. RESULTS Grn-/- mice had elevated brain EV levels and altered EV protein contents relative to wild-type mice. These changes were age-dependent, occurring only after the emergence of pathology in Grn-/- mice. FTD-GRN patients (n = 13) had elevated brain EV levels relative to controls (n = 5). Symptomatic (n = 12), but not presymptomatic (n = 7), GRN carriers had elevated plasma EV levels relative to controls (n = 8). INTERPRETATION These data show that symptomatic FTD-GRN patients have elevated levels of brain and plasma EVs, and that this effect is modeled in the brain of Grn-/- mice after the onset of pathology. This increase in EVs could influence FTD disease progression, and provides further support for EVs as potential FTD biomarkers.
Collapse
Affiliation(s)
- Andrew E. Arrant
- Departments of Neurology and NeurobiologyCenter for Neurodegeneration and Experimental TherapeuticsAlzheimer’s Disease CenterEvelyn F. McKnight Brain InstituteUniversity of Alabama at BirminghamBirminghamAlabamaUSA
| | - Skylar E. Davis
- Departments of Neurology and NeurobiologyCenter for Neurodegeneration and Experimental TherapeuticsAlzheimer’s Disease CenterEvelyn F. McKnight Brain InstituteUniversity of Alabama at BirminghamBirminghamAlabamaUSA
| | - Rachael M. Vollmer
- Departments of Neurology and NeurobiologyCenter for Neurodegeneration and Experimental TherapeuticsAlzheimer’s Disease CenterEvelyn F. McKnight Brain InstituteUniversity of Alabama at BirminghamBirminghamAlabamaUSA
| | - Charles F. Murchison
- Departments of Neurology and NeurobiologyCenter for Neurodegeneration and Experimental TherapeuticsAlzheimer’s Disease CenterEvelyn F. McKnight Brain InstituteUniversity of Alabama at BirminghamBirminghamAlabamaUSA
- Department of BiostatisticsUniversity of Alabama at BirminghamBirminghamAlabamaUSA
| | - James A. Mobley
- Department of SurgeryUniversity of Alabama at BirminghamBirminghamAlabamaUSA
| | - Alissa L. Nana
- Department of NeurologyMemory and Aging CenterUCSF Weill Institute for NeurosciencesUniversity of California, San FranciscoSan FranciscoCaliforniaUSA
| | - Salvatore Spina
- Department of NeurologyMemory and Aging CenterUCSF Weill Institute for NeurosciencesUniversity of California, San FranciscoSan FranciscoCaliforniaUSA
| | - Lea T. Grinberg
- Department of NeurologyMemory and Aging CenterUCSF Weill Institute for NeurosciencesUniversity of California, San FranciscoSan FranciscoCaliforniaUSA
- Department of PathologyUniversity of California, San FranciscoSan FranciscoCaliforniaUSA
| | - Anna M. Karydas
- Department of NeurologyMemory and Aging CenterUCSF Weill Institute for NeurosciencesUniversity of California, San FranciscoSan FranciscoCaliforniaUSA
| | - Bruce L. Miller
- Department of NeurologyMemory and Aging CenterUCSF Weill Institute for NeurosciencesUniversity of California, San FranciscoSan FranciscoCaliforniaUSA
| | - William W. Seeley
- Department of NeurologyMemory and Aging CenterUCSF Weill Institute for NeurosciencesUniversity of California, San FranciscoSan FranciscoCaliforniaUSA
- Department of PathologyUniversity of California, San FranciscoSan FranciscoCaliforniaUSA
| | - Erik D. Roberson
- Departments of Neurology and NeurobiologyCenter for Neurodegeneration and Experimental TherapeuticsAlzheimer’s Disease CenterEvelyn F. McKnight Brain InstituteUniversity of Alabama at BirminghamBirminghamAlabamaUSA
| |
Collapse
|
104
|
Vandendriessche C, Bruggeman A, Van Cauwenberghe C, Vandenbroucke RE. Extracellular Vesicles in Alzheimer's and Parkinson's Disease: Small Entities with Large Consequences. Cells 2020; 9:cells9112485. [PMID: 33203181 PMCID: PMC7696752 DOI: 10.3390/cells9112485] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2020] [Revised: 11/09/2020] [Accepted: 11/10/2020] [Indexed: 02/07/2023] Open
Abstract
Alzheimer’s disease (AD) and Parkinson’s disease (PD) are incurable, devastating neurodegenerative disorders characterized by the formation and spreading of protein aggregates throughout the brain. Although the exact spreading mechanism is not completely understood, extracellular vesicles (EVs) have been proposed as potential contributors. Indeed, EVs have emerged as potential carriers of disease-associated proteins and are therefore thought to play an important role in disease progression, although some beneficial functions have also been attributed to them. EVs can be isolated from a variety of sources, including biofluids, and the analysis of their content can provide a snapshot of ongoing pathological changes in the brain. This underlines their potential as biomarker candidates which is of specific relevance in AD and PD where symptoms only arise after considerable and irreversible neuronal damage has already occurred. In this review, we discuss the known beneficial and detrimental functions of EVs in AD and PD and we highlight their promising potential to be used as biomarkers in both diseases.
Collapse
Affiliation(s)
- Charysse Vandendriessche
- VIB Center for Inflammation Research, VIB, 9052 Ghent, Belgium; (C.V.); (A.B.); (C.V.C.)
- Department of Biomedical Molecular Biology, Ghent University, 9000 Ghent, Belgium
| | - Arnout Bruggeman
- VIB Center for Inflammation Research, VIB, 9052 Ghent, Belgium; (C.V.); (A.B.); (C.V.C.)
- Department of Biomedical Molecular Biology, Ghent University, 9000 Ghent, Belgium
- Department of Neurology, Ghent University Hospital, 9000 Ghent, Belgium
| | - Caroline Van Cauwenberghe
- VIB Center for Inflammation Research, VIB, 9052 Ghent, Belgium; (C.V.); (A.B.); (C.V.C.)
- Department of Biomedical Molecular Biology, Ghent University, 9000 Ghent, Belgium
| | - Roosmarijn E. Vandenbroucke
- VIB Center for Inflammation Research, VIB, 9052 Ghent, Belgium; (C.V.); (A.B.); (C.V.C.)
- Department of Biomedical Molecular Biology, Ghent University, 9000 Ghent, Belgium
- Correspondence: ; Tel.: +32-9-3313730
| |
Collapse
|
105
|
Mullins RJ, Mustapic M, Chia CW, Carlson O, Gulyani S, Tran J, Li Y, Mattson MP, Resnick S, Egan JM, Greig NH, Kapogiannis D. A Pilot Study of Exenatide Actions in Alzheimer's Disease. Curr Alzheimer Res 2020; 16:741-752. [PMID: 31518224 DOI: 10.2174/1567205016666190913155950] [Citation(s) in RCA: 89] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Revised: 07/10/2019] [Accepted: 08/16/2019] [Indexed: 12/24/2022]
Abstract
BACKGROUND Strong preclinical evidence suggests that exenatide, a glucagon-like peptide-1 (GLP- 1) receptor agonist used for treating type 2 diabetes, is neuroprotective and disease-modifying in Alzheimer's Disease (AD). OBJECTIVE We performed an 18-month double-blind randomized placebo-controlled Phase II clinical trial to assess the safety and tolerability of exenatide and explore treatment responses for clinical, cognitive, and biomarker outcomes in early AD. METHOD Eighteen participants with high probability AD based on cerebrospinal fluid (CSF) biomarkers completed the entire study prior to its early termination by the sponsor; partial outcomes were available for twentyone. RESULTS Exenatide was safe and well-tolerated, showing an expectedly higher incidence of nausea and decreased appetite compared to placebo and decreasing glucose and GLP-1 during Oral Glucose Tolerance Tests. Exenatide treatment produced no differences or trends compared to placebo for clinical and cognitive measures, MRI cortical thickness and volume, or biomarkers in CSF, plasma, and plasma neuronal extracellular vesicles (EV) except for a reduction of Aβ42 in EVs. CONCLUSION The positive finding of lower EV Aβ42 supports emerging evidence that plasma neuronal EVs provide an effective platform for demonstrating biomarker responses in clinical trials in AD. The study was underpowered due to early termination and therefore we cannot draw any firm conclusions. However, the analysis of secondary outcomes shows no trends in support of the hypothesis that exenatide is diseasemodifying in clinical AD, and lowering EV Aβ42 in and of itself may not improve cognitive outcomes in AD.
Collapse
Affiliation(s)
- Roger J Mullins
- Laboratory of Neurosciences, National Institute on Aging/National Institutes of Health (NIA/NIH), 3001 S. Hanover St, NM531, Baltimore, MD, 21225, United States
| | - Maja Mustapic
- Laboratory of Neurosciences, National Institute on Aging/National Institutes of Health (NIA/NIH), 3001 S. Hanover St, NM531, Baltimore, MD, 21225, United States
| | - Chee W Chia
- Laboratory of Clinical Investigation, National Institute on Aging/National Institutes of Health (NIA/NIH), 3001 S. Hanover St, NM531, Baltimore, MD, 21225, United States
| | - Olga Carlson
- Laboratory of Clinical Investigation, National Institute on Aging/National Institutes of Health (NIA/NIH), 3001 S. Hanover St, NM531, Baltimore, MD, 21225, United States
| | - Seema Gulyani
- Laboratory of Neurosciences, National Institute on Aging/National Institutes of Health (NIA/NIH), 3001 S. Hanover St, NM531, Baltimore, MD, 21225, United States
| | - Joyce Tran
- Laboratory of Neurosciences, National Institute on Aging/National Institutes of Health (NIA/NIH), 3001 S. Hanover St, NM531, Baltimore, MD, 21225, United States
| | - Yazhou Li
- Translational Gerontology Branch, National Institute on Aging/National Institutes of Health (NIA/NIH), 3001 S. Hanover St, NM531, Baltimore, MD, 21225, United States
| | - Mark P Mattson
- Laboratory of Neurosciences, National Institute on Aging/National Institutes of Health (NIA/NIH), 3001 S. Hanover St, NM531, Baltimore, MD, 21225, United States
| | - Susan Resnick
- Laboratory of Behavioral Neuroscience, Intramural Research Program, National Institute on Aging/National Institutes of Health (NIA/NIH), 3001 S. Hanover St, NM531, Baltimore, MD, 21225, United States
| | - Josephine M Egan
- Laboratory of Clinical Investigation, National Institute on Aging/National Institutes of Health (NIA/NIH), 3001 S. Hanover St, NM531, Baltimore, MD, 21225, United States
| | - Nigel H Greig
- Translational Gerontology Branch, National Institute on Aging/National Institutes of Health (NIA/NIH), 3001 S. Hanover St, NM531, Baltimore, MD, 21225, United States
| | - Dimitrios Kapogiannis
- Laboratory of Neurosciences, National Institute on Aging/National Institutes of Health (NIA/NIH), 3001 S. Hanover St, NM531, Baltimore, MD, 21225, United States
| |
Collapse
|
106
|
Tunset ME, Haslene-Hox H, Van Den Bossche T, Vaaler AE, Sulheim E, Kondziella D. Extracellular vesicles in patients in the acute phase of psychosis and after clinical improvement: an explorative study. PeerJ 2020; 8:e9714. [PMID: 32995075 PMCID: PMC7501784 DOI: 10.7717/peerj.9714] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Accepted: 07/23/2020] [Indexed: 12/28/2022] Open
Abstract
Extracellular vesicles (EVs) are cell-derived structures that transport proteins, lipids and nucleic acids between cells, thereby affecting the phenotype of the recipient cell. As the content of EVs reflects the status of the originating cell, EVs can have potential as biomarkers. Identifying EVs, including their cells of origin and their cargo, may provide insights in the pathophysiology of psychosis. Here, we present an in-depth analysis and proteomics of EVs from peripheral blood in patients (n = 25) during and after the acute phase of psychosis. Concentration and protein content of EVs in psychotic patients were twofold higher than in 25 age- and sex-matched healthy controls (p < 0.001 for both concentration and protein content), and the diameter of EVs was larger in patients (p = 0.02). Properties of EVs did not differ significantly in blood sampled during and after the acute psychotic episode. Proteomic analyses on isolated EVs from individual patients revealed 1,853 proteins, whereof 45 were brain-elevated proteins. Of these, five proteins involved in regulation of plasticity of glutamatergic synapses were significantly different in psychotic patients compared to controls; neurogranin (NRGN), neuron-specific calcium-binding protein hippocalcin (HPCA), kalirin (KALRN), beta-adducin (ADD2) and ankyrin-2 (ANK2). To summarize, our results show that peripheral EVs in psychotic patients are different from those in healthy controls and point at alterations on the glutamatergic system. We suggest that EVs allow investigation of blood-borne brain-originating biological material and that their role as biomarkers in patients with psychotic disorders is worthy of further exploration.
Collapse
Affiliation(s)
- Mette Elise Tunset
- Department of Østmarka- Division of Mental Healthcare, St. Olavs University Hospital, Trondheim, Norway.,Department of Mental Health- Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Hanne Haslene-Hox
- Department of Biotechnology and Nanomedicine, SINTEF, Trondheim, Norway
| | - Tim Van Den Bossche
- VIB - UGent Center for Medical Biotechnology, VIB, Ghent, Belgium.,Department of Biomolecular Medicine, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | - Arne Einar Vaaler
- Department of Østmarka- Division of Mental Healthcare, St. Olavs University Hospital, Trondheim, Norway.,Department of Mental Health- Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Einar Sulheim
- Department of Biotechnology and Nanomedicine, SINTEF, Trondheim, Norway.,Department of Physics, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Daniel Kondziella
- Department of Neurology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark.,Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
107
|
The Potential of Liquid Biopsy of the Brain Using Blood Extracellular Vesicles: The First Step Toward Effective Neuroprotection Against Neurodegenerative Diseases. Mol Diagn Ther 2020; 24:703-713. [PMID: 32975732 DOI: 10.1007/s40291-020-00493-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/02/2020] [Indexed: 10/23/2022]
Abstract
Early diagnosis and biomarker-based ante-mortem tests are essential in efforts against the development of neurodegenerative diseases and can be considered primary neuroprotective measures. Blood is the ideal biofluid for a routine ante-mortem screening test. However, biomarker discovery in the blood is particularly difficult because of interference from factors both intrinsic and extrinsic to blood with the detection of hallmark neurodegenerative biomarkers, such as the pathological prion protein, amyloid-β, and others. Blood extracellular vesicles (EVs), such as exosomes, are cell-derived vesicles released into the blood from all parts of the body (including the brain and spinal cord). They are an enriched source of neural-derived EVs containing neurodegenerative biomarkers that mirror (in the blood) the condition present in the brain. The feasibility of using, and the reliability of, neural-derived blood EVs (NDBEVs) as a method of diagnosing Alzheimer disease and other neurodegenerative diseases has been assessed in strong proof-of-concept studies. Results from these studies strongly suggest that NDBEVs might represent the right strategy for specific, reliable, and early diagnosis of neurodegenerative diseases. Based on these results, NDBEVs might enable the creation of an ante-mortem blood test (liquid biopsy of the brain) for neurodegenerative diseases. This would enormously accelerate the therapy of neurodegenerative diseases. This review highlights the powerful potential of liquid biopsy of the brain using NDBEVs for early diagnosis and treatment of neurodegenerative diseases, and the challenges and limitations related to the identification of clinically applicable EV (exosomal) biomarkers using blood are discussed.
Collapse
|
108
|
Chung CC, Huang PH, Chan L, Chen JH, Chien LN, Hong CT. Plasma Exosomal Brain-Derived Neurotrophic Factor Correlated with the Postural Instability and Gait Disturbance-Related Motor Symptoms in Patients with Parkinson's Disease. Diagnostics (Basel) 2020; 10:E684. [PMID: 32932791 PMCID: PMC7555255 DOI: 10.3390/diagnostics10090684] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 09/08/2020] [Accepted: 09/09/2020] [Indexed: 12/15/2022] Open
Abstract
Brain-derived neurotrophic factor (BDNF) is an essential neurotrophin, responsible for neuronal development, function, and survival. Assessments of peripheral blood BDNF in patients with Parkinson's disease (PD) previously yielded inconsistent results. Plasma exosomes can carry BDNF, so this study investigated the role of plasma exosomal BDNF level as a biomarker of PD. A total of 114 patients with mild to moderate PD and 42 non-PD controls were recruited, and their clinical presentations were evaluated. Plasma exosomes were isolated with exoEasy Maxi Kits, and enzyme-linked immunosorbent assay was used to assess plasma exosomal BDNF levels. Statistical analysis was performed using SPSS version 19.0, and findings were considered significant at p < 0.05. The analysis revealed no significant differences in plasma exosomal BDNF levels between patients with PD and controls. Patients with PD with low plasma exosomal BDNF levels (in the lowest quartile) exhibited a significant association with daily activity dysfunction but not with cognition/mood or overall motor symptoms as assessed using the Unified Parkinson's Disease Rating Scale (UPDRS). Investigation of UPDRS part III subitems revealed that low plasma exosomal BDNF level was significantly associated with increased motor severity of postural instability and gait disturbance (PIGD)-associated symptoms (rising from a chair, gait, and postural stability) after adjustment for age and sex. In conclusion, although plasma exosomal BDNF level could not distinguish patients with PD from controls, the association with PIGD symptoms in patients with PD may indicate its potential role as a biomarker. Follow-up studies should investigate the association between plasma exosomal BDNF levels and changes in clinical symptoms.
Collapse
Affiliation(s)
- Chen Chih Chung
- Department of Neurology, Shuang Ho Hospital, Taipei Medical University, Zhongzheng Rd, Zhonghe District, New Taipei City 23561, Taiwan; (C.C.C.); (L.C.); (J.-H.C.)
- Department of Neurology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
- Graduate Institute of Biomedical Informatics, Taipei Medical University, Taipei 11031, Taiwan
| | - Pai Hao Huang
- Department of Neurology, Cathay General Hospital, Taipei 106, Taiwan;
| | - Lung Chan
- Department of Neurology, Shuang Ho Hospital, Taipei Medical University, Zhongzheng Rd, Zhonghe District, New Taipei City 23561, Taiwan; (C.C.C.); (L.C.); (J.-H.C.)
- Department of Neurology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Jia-Hung Chen
- Department of Neurology, Shuang Ho Hospital, Taipei Medical University, Zhongzheng Rd, Zhonghe District, New Taipei City 23561, Taiwan; (C.C.C.); (L.C.); (J.-H.C.)
| | - Li-Nien Chien
- School of Health Care Administration, College of Management, Taipei Medical University, 250 Wuxing Street, Taipei 11031, Taiwan
| | - Chien Tai Hong
- Department of Neurology, Shuang Ho Hospital, Taipei Medical University, Zhongzheng Rd, Zhonghe District, New Taipei City 23561, Taiwan; (C.C.C.); (L.C.); (J.-H.C.)
- Department of Neurology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
| |
Collapse
|
109
|
Mustapic M, Tran J, Craft S, Kapogiannis D. Extracellular Vesicle Biomarkers Track Cognitive Changes Following Intranasal Insulin in Alzheimer's Disease. J Alzheimers Dis 2020; 69:489-498. [PMID: 30958348 DOI: 10.3233/jad-180578] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND Insulin resistance is implicated in Alzheimer's disease (AD), whereas intranasal insulin is an experimental treatment in clinical trials. We previously proposed insulin signaling mediators in plasma neuronal-enriched extracellular vesicles (EVs) as biomarkers of brain insulin resistance. OBJECTIVE We sought to demonstrate the capacity of neuronal-enriched EV biomarkers to demonstrate target engagement in response to intranasal insulin and their ability to track treatment-associated cognitive changes in AD. METHODS We isolated neuronal-enriched EVs from plasma samples of participants with amnestic mild cognitive impairment or probable AD involved in a 4-month duration placebo-controlled clinical trial of 20 or 40 IU intranasal insulin. We measured insulin signaling mediators as biomarkers and examined treatment-associated changes and their relationship with cognitive performance (ADAS-Cog). RESULTS There were no EV biomarker changes from baseline in any of the treatment groups. In participants treated with 20 IU insulin, EV biomarkers of insulin resistance (pS312-IRS-1, pY-IRS-1) showed strong positive correlations with ADAS-Cog changes, especially in ApoE ɛ4 non-carriers. CONCLUSION Neuronal EV biomarkers of insulin resistance (pS312-IRS-1, pY-IRS-1) were associated with cognitive changes in response to low dose intranasal insulin suggesting engagement of the insulin cascade in neurons of origin.
Collapse
Affiliation(s)
- Maja Mustapic
- Laboratory of Neurosciences, Intramural Research Program, National Institute on Aging, NIH, Baltimore, MD, USA
| | - Joyce Tran
- Laboratory of Neurosciences, Intramural Research Program, National Institute on Aging, NIH, Baltimore, MD, USA
| | - Suzanne Craft
- Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Dimitrios Kapogiannis
- Laboratory of Neurosciences, Intramural Research Program, National Institute on Aging, NIH, Baltimore, MD, USA
| |
Collapse
|
110
|
Ausó E, Gómez-Vicente V, Esquiva G. Biomarkers for Alzheimer's Disease Early Diagnosis. J Pers Med 2020; 10:E114. [PMID: 32899797 PMCID: PMC7563965 DOI: 10.3390/jpm10030114] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 08/27/2020] [Accepted: 09/01/2020] [Indexed: 12/11/2022] Open
Abstract
Alzheimer's disease (AD) is the most common cause of dementia, affecting the central nervous system (CNS) through the accumulation of intraneuronal neurofibrillary tau tangles (NFTs) and β-amyloid plaques. By the time AD is clinically diagnosed, neuronal loss has already occurred in many brain and retinal regions. Therefore, the availability of early and reliable diagnosis markers of the disease would allow its detection and taking preventive measures to avoid neuronal loss. Current diagnostic tools in the brain, such as magnetic resonance imaging (MRI), positron emission tomography (PET) imaging, and cerebrospinal fluid (CSF) biomarkers (Aβ and tau) detection are invasive and expensive. Brain-secreted extracellular vesicles (BEVs) isolated from peripheral blood have emerged as novel strategies in the study of AD, with enormous potential as a diagnostic evaluation of therapeutics and treatment tools. In addition; similar mechanisms of neurodegeneration have been demonstrated in the brain and the eyes of AD patients. Since the eyes are more accessible than the brain, several eye tests that detect cellular and vascular changes in the retina have also been proposed as potential screening biomarkers. The aim of this study is to summarize and discuss several potential markers in the brain, eye, blood, and other accessible biofluids like saliva and urine, and correlate them with earlier diagnosis and prognosis to identify individuals with mild symptoms prior to dementia.
Collapse
Affiliation(s)
| | | | - Gema Esquiva
- Department of Optics, Pharmacology and Anatomy, University of Alicante, 03690 Alicante, Spain; (E.A.); (V.G.-V.)
| |
Collapse
|
111
|
Kellar D, Craft S. Brain insulin resistance in Alzheimer's disease and related disorders: mechanisms and therapeutic approaches. Lancet Neurol 2020; 19:758-766. [PMID: 32730766 DOI: 10.1016/s1474-4422(20)30231-3] [Citation(s) in RCA: 478] [Impact Index Per Article: 95.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Revised: 05/30/2020] [Accepted: 06/15/2020] [Indexed: 12/16/2022]
|
112
|
Goetzl EJ. Advancing medicine for Alzheimer's disease: A plasma neural exosome platform. FASEB J 2020; 34:13079-13084. [PMID: 32856798 DOI: 10.1096/fj.202001655] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 07/27/2020] [Indexed: 12/20/2022]
Abstract
Enrichment of neurally derived extracellular vesicles of several cell-types from plasma for protein quantification longitudinally in living patients with Alzheimer's disease has permitted the development of a tentative temporal framework of initiating events, progression mechanisms, and amplification processes. Interactions of beta-amyloid peptides with an elevated level of their normal prion protein dendritic receptor and of phospho-tau species with their synaptogyrin-3 synaptic vesicle receptor replace excessive production and accumulation of neuropathic proteins as the major initiating events. Synaptic dysfunction and microvascular angiopathy are confirmed as early progression mechanisms of decreased neuronal network connectivity, hypoxia, altered blood-brain barrier, and neurocellular degeneration. Neurally derived extracellular vesicle protein abnormalities also reveal a range of later amplification processes that encompasses insulin resistance, lysosomal defects, decreased survival factors, increased reactive oxygen species, and excessive neuroinflammation. New potential therapeutic targets also are suggested as well as the likely timing of their pathogenic engagement.
Collapse
Affiliation(s)
- Edward J Goetzl
- Department of Medicine, University of California Medical Center, San Francisco, CA, USA.,Geriatric Research Center, Campus for Jewish Living, San Francisco, CA, USA
| |
Collapse
|
113
|
Lakshmi S, Essa MM, Hartman RE, Guillemin GJ, Sivan S, Elumalai P. Exosomes in Alzheimer's Disease: Potential Role as Pathological Mediators, Biomarkers and Therapeutic Targets. Neurochem Res 2020; 45:2553-2559. [PMID: 32840760 DOI: 10.1007/s11064-020-03111-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 08/09/2020] [Accepted: 08/13/2020] [Indexed: 12/19/2022]
Abstract
The concept of exosomes has been progressively changed from the status of cellular trashcans to multitasking organelles involved in many processes, including internalization, transport and transfer of macromolecules such as proteins, lipids and nucleic acids. While underpinning the mechanisms behind neurodegeneration and neuronal loss, exosomes were shown to be involved in carrying pathological misfolded proteins, propagation of β-amyloid protein and hyper-phosphorylated tau proteins across the brain that ultimately leads to the onset of Alzheimer's disease (AD), the most prevailing multifactorial neurodegenerative disorder. A potential novel therapeutic role of exosomes in AD intervention is suggested by their ability to increase Aβ clearance. This review aims to highlight the important pathological mechanisms as well as therapeutic strategies involving exosomes towards AD prevention.
Collapse
Affiliation(s)
- Sreeja Lakshmi
- School of Ocean Science and Technology, Kerala University of Fisheries and Ocean Studies, Kochi, Kerala, India
| | - Musthafa Mohamed Essa
- Ageing and Dementia Research Group, Sultan Qaboos University, Muscat, Oman.,Department of Food Science and Nutrition, College of Agricultural and Marine Sciences, Sultan Qaboos University, Muscat, Oman
| | - Richard E Hartman
- Department of Psychology, Loma Linda University, Loma Linda, CA, USA
| | - Gilles J Guillemin
- Faculty of Medicine and Health Sciences, Macquarie University, Sydney, Australia
| | - Sureshkumar Sivan
- School of Ocean Science and Technology, Kerala University of Fisheries and Ocean Studies, Kochi, Kerala, India
| | - Preetham Elumalai
- School of Ocean Science and Technology, Kerala University of Fisheries and Ocean Studies, Kochi, Kerala, India.
| |
Collapse
|
114
|
Gu D, Liu F, Meng M, Zhang L, Gordon ML, Wang Y, Cai L, Zhang N. Elevated matrix metalloproteinase-9 levels in neuronal extracellular vesicles in Alzheimer's disease. Ann Clin Transl Neurol 2020; 7:1681-1691. [PMID: 32790155 PMCID: PMC7480907 DOI: 10.1002/acn3.51155] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 07/14/2020] [Accepted: 07/16/2020] [Indexed: 12/31/2022] Open
Abstract
OBJECTIVE This study aimed to investigate plasma neuronally derived extracellular vesicle (NDEV) levels of core pathological markers [amyloid-β (Aβ) and phosphorylated tau] and inflammatory biomarkers, including interleukin 6 (IL-6) and matrix metalloproteinase-9 (MMP-9) in patients with Alzheimer's disease (AD). METHODS Thirty-one patients with AD and 15 cognitively normal controls (NCs) were recruited. The diagnosis of AD was supported by fluorodeoxyglucose and Pittsburgh Compound-B PET scans. Plasma extracellular vesicles were extracted, precipitated, and enriched for neuronal source by anti-L1CAM antibody absorption. Levels of Aβ42, P-T181-tau, P-S396-tau, IL-6, and MMP-9 in plasma NDEVs were quantified by enzyme-linked immunosorbent assay (ELISA). RESULTS Aβ42, P-T181-tau, and MMP-9 levels in plasma NDEVs were significantly higher in patients with AD than NCs. However, P-S396-tau and IL-6 levels in plasma NDEVs did not differ between AD patients and NCs. Moreover, there was no correlation between any of these biomarker levels and cognitive function as measured with Mini-Mental State Examination in patients with AD. CONCLUSIONS These findings provide further support that levels of core pathological markers, including Aβ42 and P-T181-tau, are elevated in plasma NDEVs of patients with AD. Furthermore, MMP-9 might play an important role in the pathogenesis of AD, and is a promising inflammatory biomarker for AD.
Collapse
Affiliation(s)
- Dongmei Gu
- Department of Clinical Laboratory, Tianjin Medical University General Hospital, Tianjin, China.,Department of Clinical Laboratory, Tianjin Medical University General Hospital Airport Hospital, Tianjin, China
| | - Fang Liu
- Department of Neurology, Tianjin Medical University General Hospital, Tianjin, China
| | - Meng Meng
- Department of Neurology, Tianjin Medical University General Hospital Airport Hospital, Tianjin, China
| | - Liling Zhang
- Department of Neurology, Heji Hospital Affiliated to Changzhi Medical College, Changzhi, Shanxi, China
| | - Marc L Gordon
- The Litwin-Zucker Research Center, The Feinstein Institutes for Medical Research, Northwell Health, 350 Community Drive, Manhasset, New York.,Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, 500 Hofstra Blvd, Hempstead, New York
| | - Ying Wang
- Department of PET-CT Diagnostic, Tianjin Medical University General Hospital, Tianjin, China
| | - Li Cai
- Department of PET-CT Diagnostic, Tianjin Medical University General Hospital, Tianjin, China
| | - Nan Zhang
- Department of Neurology, Tianjin Medical University General Hospital, Tianjin, China.,Department of Neurology, Tianjin Medical University General Hospital Airport Hospital, Tianjin, China
| |
Collapse
|
115
|
Jia L, Zhu M, Kong C, Pang Y, Zhang H, Qiu Q, Wei C, Tang Y, Wang Q, Li Y, Li T, Li F, Wang Q, Li Y, Wei Y, Jia J. Blood neuro-exosomal synaptic proteins predict Alzheimer's disease at the asymptomatic stage. Alzheimers Dement 2020; 17:49-60. [PMID: 32776690 PMCID: PMC7984076 DOI: 10.1002/alz.12166] [Citation(s) in RCA: 154] [Impact Index Per Article: 30.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2019] [Revised: 07/01/2020] [Accepted: 07/08/2020] [Indexed: 12/25/2022]
Abstract
Introduction Exosomes are an emerging candidate for biomarkers of Alzheimer's disease (AD). This study investigated whether exosomal synaptic proteins can predict AD at the asymptomatic stage. Methods We conducted a two‐stage‐sectional study (discovery stage: AD, 28; amnestic mild cognitive impairment [aMCI], 25; controls, 29; validation stage: AD, 73; aMCI, 71; controls, 72), a study including preclinical AD (160) and controls (160), and a confirmation study in familial AD (mutation carriers: 59; non‐mutation carriers: 62). Results The concentrations of growth associated protein 43 (GAP43), neurogranin, synaptosome associated protein 25 (SNAP25), and synaptotagmin 1 were lower in AD than in controls (P < .001). Exosomal biomarker levels were correlated with those in cerebrospinal fluid (R2 = 0.54–0.70). The combination of exosomal biomarkers detected AD 5 to 7 years before cognitive impairment (area under the curve = 0.87–0.89). Discussion This study revealed that exosomal GAP43, neurogranin, SNAP25, and synaptotagmin 1 act as effective biomarkers for prediction of AD 5 to 7 years before cognitive impairment.
Collapse
Affiliation(s)
- Longfei Jia
- Innovation Center for Neurological Disorders, Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Min Zhu
- Innovation Center for Neurological Disorders, Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Chaojun Kong
- Innovation Center for Neurological Disorders, Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Yana Pang
- Innovation Center for Neurological Disorders, Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Heng Zhang
- Innovation Center for Neurological Disorders, Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Qiongqiong Qiu
- Innovation Center for Neurological Disorders, Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Cuibai Wei
- Innovation Center for Neurological Disorders, Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Yi Tang
- Innovation Center for Neurological Disorders, Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Qi Wang
- Innovation Center for Neurological Disorders, Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Ying Li
- Innovation Center for Neurological Disorders, Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Tingting Li
- Innovation Center for Neurological Disorders, Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Fangyu Li
- Innovation Center for Neurological Disorders, Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Qigeng Wang
- Innovation Center for Neurological Disorders, Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Yan Li
- Innovation Center for Neurological Disorders, Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Yiping Wei
- Innovation Center for Neurological Disorders, Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Jianping Jia
- Innovation Center for Neurological Disorders, Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
116
|
Soares Martins T, Trindade D, Vaz M, Campelo I, Almeida M, Trigo G, da Cruz E Silva OAB, Henriques AG. Diagnostic and therapeutic potential of exosomes in Alzheimer's disease. J Neurochem 2020; 156:162-181. [PMID: 32618370 DOI: 10.1111/jnc.15112] [Citation(s) in RCA: 101] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 05/20/2020] [Accepted: 06/18/2020] [Indexed: 12/12/2022]
Abstract
Exosomes are small extracellular vesicles released by almost all cell types in physiological and pathological conditions. The exosomal potential to unravel disease mechanisms, or to be used as a source of biomarkers, is being explored, in particularly in the field of neurodegenerative diseases. Alzheimer's disease (AD) is the most prevalent neurodegenerative disease in the world and exosomes appear to have a relevant role in disease pathogenesis. This review summarizes the current knowledge on exosome contributions to AD as well as their use as disease biomarker resources or therapeutic targets. The most recent findings with respect to both protein and miRNA biomarker candidates for AD, herein described, highlight the state of the art in this field and encourage the use of exosomes derived from biofluids in clinical practice in the near future.
Collapse
Affiliation(s)
- Tânia Soares Martins
- Neurosciences and Signalling Group, Institute of Biomedicine (iBiMED), Department of Medical Sciences, University of Aveiro, Aveiro, Portugal
| | - Dário Trindade
- Neurosciences and Signalling Group, Institute of Biomedicine (iBiMED), Department of Medical Sciences, University of Aveiro, Aveiro, Portugal
| | - Margarida Vaz
- Neurosciences and Signalling Group, Institute of Biomedicine (iBiMED), Department of Medical Sciences, University of Aveiro, Aveiro, Portugal
| | - Inês Campelo
- Department of Medical Sciences, University of Aveiro, Aveiro, Portugal
| | - Martim Almeida
- Department of Medical Sciences, University of Aveiro, Aveiro, Portugal
| | - Guilherme Trigo
- Department of Medical Sciences, University of Aveiro, Aveiro, Portugal
| | - Odete A B da Cruz E Silva
- Neurosciences and Signalling Group, Institute of Biomedicine (iBiMED), Department of Medical Sciences, University of Aveiro, Aveiro, Portugal.,The Discovery CTR, University of Aveiro Campus, Aveiro, Portugal
| | - Ana Gabriela Henriques
- Neurosciences and Signalling Group, Institute of Biomedicine (iBiMED), Department of Medical Sciences, University of Aveiro, Aveiro, Portugal
| |
Collapse
|
117
|
Zhao A, Li Y, Yan Y, Qiu Y, Li B, Xu W, Wang Y, Liu J, Deng Y. Increased prediction value of biomarker combinations for the conversion of mild cognitive impairment to Alzheimer's dementia. Transl Neurodegener 2020; 9:30. [PMID: 32741361 PMCID: PMC7397685 DOI: 10.1186/s40035-020-00210-5] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Accepted: 07/03/2020] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Progression of mild cognitive impairment (MCI) to Alzheimer's disease (AD) dementia can be predicted by clinical features and a combination of biomarkers may increase the predictive power. In the present study, we investigated whether the combination of olfactory function and plasma neuronal-derived exosome (NDE) Aβ1-42 can best predict progression to AD dementia. METHODS 87 MCI patients were enrolled and received the cognitive assessment at 2-year and 3-year follow-up to reevaluate cognition. In the meanwhile, 80 healthy controls and 88 AD dementia patients were enrolled at baseline as well to evaluate the diagnose value in cross-section. Olfactory function was evaluated with the sniffin sticks (SS-16) and Aβ1-42 levels in NDEs were determined by ELISA. Logistic regression was performed to evaluate the risk factors for cognitive decline in MCI at 2-year and 3-year revisits. RESULTS In the cross cohort, lower SS-16 scores and higher Aβ1-42 levels in NDEs were found in MCI and AD dementia compared to healthy controls. For the longitudinal set, 8 MCI individuals developed AD dementia within 2 years, and 16 MCI individuals developed AD dementia within 3 years. The two parameter-combination of SS-16 scores and Aβ1-42 level in NDEs showed better prediction in the conversion of MCI to AD dementia at 2-year and 3-year revisit. Moreover, after a 3-year follow-up, SS-16 scores also significantly predicted the conversion to AD dementia, where lower scores were associated with a 10-fold increased risk of developing AD dementia (p = 0.006). Similarly, higher Aβ1-42 levels in NDEs in patients with MCI increased the risk of developing AD dementia by 8.5-fold (p = 0.002). CONCLUSION A combination of two biomarkers of NDEs (Aβ1-42) and SS-16 predicted the conversion of MCI to AD dementia more accurately in combination. These findings have critical implications for understanding the pathophysiology of AD dementia and for developing preventative treatments for cognitive decline.
Collapse
Affiliation(s)
- Aonan Zhao
- Department of Neurology and Institute of Neurology, Ruijin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuanyuan Li
- Department of Neurology and Institute of Neurology, Ruijin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yi Yan
- Department of Neurology and Institute of Neurology, Ruijin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yinghui Qiu
- Department of Neurology and Institute of Neurology, Ruijin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Binyin Li
- Department of Neurology and Institute of Neurology, Ruijin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wei Xu
- Department of Neurology and Institute of Neurology, Ruijin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ying Wang
- Department of Neurology and Institute of Neurology, Ruijin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jun Liu
- Department of Neurology and Institute of Neurology, Ruijin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China. .,Department of Neurology, RuiJin Hospital/LuWan Branch, School of Medicine, Shanghai Jiaotong University, Shanghai, China.
| | - Yulei Deng
- Department of Neurology and Institute of Neurology, Ruijin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China. .,Department of Neurology, RuiJin Hospital/LuWan Branch, School of Medicine, Shanghai Jiaotong University, Shanghai, China.
| |
Collapse
|
118
|
Dietary Gluten and Neurodegeneration: A Case for Preclinical Studies. Int J Mol Sci 2020; 21:ijms21155407. [PMID: 32751379 PMCID: PMC7432597 DOI: 10.3390/ijms21155407] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 07/23/2020] [Accepted: 07/24/2020] [Indexed: 12/11/2022] Open
Abstract
Although celiac disease (CD) is an autoimmune disease that primarily involves the intestinal tract, mounting evidence suggests that a sizeable number of patients exhibit neurological deficits. About 40% of the celiac patients with neurological manifestations have circulating antibodies against neural tissue transglutaminase-6 (tTG6). While early diagnosis and strict adherence to a gluten-free diet (GFD) have been recommended to prevent neurological dysfunction, better therapeutic strategies are needed to improve the overall quality of life. Dysregulation of the microbiota-gut-brain axis, presence of anti-tTG6 antibodies, and epigenetic mechanisms have been implicated in the pathogenesis. It is also possible that circulating or gut-derived extracellular structures and including biomolecular condensates and extracellular vesicles contribute to disease pathogenesis. There are several avenues for shaping the dysregulated gut homeostasis in individuals with CD, non-celiac gluten sensitivity (NCGS) and/or neurodegeneration. In addition to GFD and probiotics, nutraceuticals, such as phyto and synthetic cannabinoids, represent a new approach that could shape the host microbiome towards better prognostic outcomes. Finally, we provide a data-driven rationale for potential future pre-clinical research involving non-human primates (NHPs) to investigate the effect of nutraceuticals, such as phyto and synthetic cannabinoids, either alone or in combination with GFD to prevent/mitigate dietary gluten-induced neurodegeneration.
Collapse
|
119
|
Jiang C, Hopfner F, Katsikoudi A, Hein R, Catli C, Evetts S, Huang Y, Wang H, Ryder JW, Kuhlenbaeumer G, Deuschl G, Padovani A, Berg D, Borroni B, Hu MT, Davis JJ, Tofaris GK. Serum neuronal exosomes predict and differentiate Parkinson's disease from atypical parkinsonism. J Neurol Neurosurg Psychiatry 2020; 91:720-729. [PMID: 32273329 PMCID: PMC7361010 DOI: 10.1136/jnnp-2019-322588] [Citation(s) in RCA: 175] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Revised: 02/10/2020] [Accepted: 03/23/2020] [Indexed: 12/25/2022]
Abstract
OBJECTIVE Parkinson's disease is characterised neuropathologically by α-synuclein aggregation. Currently, there is no blood test to predict the underlying pathology or distinguish Parkinson's from atypical parkinsonian syndromes. We assessed the clinical utility of serum neuronal exosomes as biomarkers across the spectrum of Parkinson's disease, multiple system atrophy and other proteinopathies. METHODS We performed a cross-sectional study of 664 serum samples from the Oxford, Kiel and Brescia cohorts consisting of individuals with rapid eye movement sleep behavioural disorder, Parkinson's disease, dementia with Lewy bodies, multiple system atrophy, frontotemporal dementia, progressive supranuclear palsy, corticobasal syndrome and controls. Longitudinal samples were analysed from Parkinson's and control individuals. We developed poly(carboxybetaine-methacrylate) coated beads to isolate L1 cell adhesion molecule (L1CAM)-positive extracellular vesicles with characteristics of exosomes and used mass spectrometry or multiplexed electrochemiluminescence to measure exosomal proteins. RESULTS Mean neuron-derived exosomal α-synuclein was increased by twofold in prodromal and clinical Parkinson's disease when compared with multiple system atrophy, controls or other neurodegenerative diseases. With 314 subjects in the training group and 105 in the validation group, exosomal α-synuclein exhibited a consistent performance (AUC=0.86) in separating clinical Parkinson's disease from controls across populations. Exosomal clusterin was elevated in subjects with non-α-synuclein proteinopathies. Combined neuron-derived exosomal α-synuclein and clusterin measurement predicted Parkinson's disease from other proteinopathies with AUC=0.98 and from multiple system atrophy with AUC=0.94. Longitudinal sample analysis showed that exosomal α-synuclein remains stably elevated with Parkinson's disease progression. CONCLUSIONS Increased α-synuclein egress in serum neuronal exosomes precedes the diagnosis of Parkinson's disease, persists with disease progression and in combination with clusterin predicts and differentiates Parkinson's disease from atypical parkinsonism.
Collapse
Affiliation(s)
- Cheng Jiang
- Nuffield Department of Clinical Neurosciences, University of Oxford, John Radcliffe Hospital, Oxford, UK
| | - Franziska Hopfner
- Nuffield Department of Clinical Neurosciences, University of Oxford, John Radcliffe Hospital, Oxford, UK.,Department of Neurology, Christian-Albrechts-University, Kiel, Germany
| | - Antigoni Katsikoudi
- Nuffield Department of Clinical Neurosciences, University of Oxford, John Radcliffe Hospital, Oxford, UK
| | - Robert Hein
- Department of Chemistry, Physical and Theoretical Chemistry Laboratory, University of Oxford, Oxford, United Kingdom
| | - Candan Catli
- Department of Chemistry, Physical and Theoretical Chemistry Laboratory, University of Oxford, Oxford, United Kingdom
| | - Samuel Evetts
- Nuffield Department of Clinical Neurosciences, University of Oxford, John Radcliffe Hospital, Oxford, UK.,Oxford Parkinson's Disease Centre, Oxford, United Kingdom
| | - Yongzhi Huang
- Nuffield Department of Surgical Sciences, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
| | - Hong Wang
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana, USA
| | - John W Ryder
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana, USA
| | | | - Guenther Deuschl
- Department of Neurology, Christian-Albrechts-University, Kiel, Germany
| | - Alessandro Padovani
- Department of Clinical and Experimental Sciences, Neurology Unit, University of Brescia, Brescia, Italy
| | - Daniela Berg
- Department of Neurology, Christian-Albrechts-University, Kiel, Germany
| | - Barbara Borroni
- Department of Clinical and Experimental Sciences, Neurology Unit, University of Brescia, Brescia, Italy
| | - Michele T Hu
- Nuffield Department of Clinical Neurosciences, University of Oxford, John Radcliffe Hospital, Oxford, UK.,Oxford Parkinson's Disease Centre, Oxford, United Kingdom
| | - Jason J Davis
- Department of Chemistry, Physical and Theoretical Chemistry Laboratory, University of Oxford, Oxford, United Kingdom
| | - George K Tofaris
- Nuffield Department of Clinical Neurosciences, University of Oxford, John Radcliffe Hospital, Oxford, UK
| |
Collapse
|
120
|
Lim CZJ, Natalia A, Sundah NR, Shao H. Biomarker Organization in Circulating Extracellular Vesicles: New Applications in Detecting Neurodegenerative Diseases. ACTA ACUST UNITED AC 2020; 4:e1900309. [PMID: 32597034 DOI: 10.1002/adbi.201900309] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Revised: 05/23/2020] [Indexed: 12/12/2022]
Abstract
Neurodegenerative diseases are heterogeneous disorders characterized by a progressive loss of function and/or death of nerve cells, leading to severe cognitive and functional decline. Due to the complex pathology, early detection and intervention are critical to the development of successful treatments; however, current diagnostic approaches are limited to subjective, late-stage clinical findings. Extracellular vesicles (EVs) have recently emerged as a promising circulating biomarker for neurodegenerative diseases. Actively released by diverse cells, EVs are nanoscale membrane vesicles. They abound in blood, readily cross the blood-brain barrier, and carry diverse molecular cargoes in different organizational states: these molecular cargoes are inherited from the parent cells or bound to the EV membrane through surface associations. Specifically, EVs have been found to be associated with several important pathogenic proteins of neurodegenerative diseases, and their involvement could alter disease progression. This article provides an overview of EVs as circulating biomarkers of neurodegenerative diseases and introduces new technological advances to characterize the biophysical properties of EV-associated biomarkers for accurate, blood-based detection of neurodegenerative diseases.
Collapse
Affiliation(s)
- Carine Z J Lim
- Department of Biomedical Engineering, Faculty of Engineering, National University of Singapore, Singapore, 117583, Singapore.,Institute for Health Innovation and Technology, National University of Singapore, Singapore, 117599, Singapore
| | - Auginia Natalia
- Department of Biomedical Engineering, Faculty of Engineering, National University of Singapore, Singapore, 117583, Singapore.,Institute for Health Innovation and Technology, National University of Singapore, Singapore, 117599, Singapore
| | - Noah R Sundah
- Department of Biomedical Engineering, Faculty of Engineering, National University of Singapore, Singapore, 117583, Singapore.,Institute for Health Innovation and Technology, National University of Singapore, Singapore, 117599, Singapore
| | - Huilin Shao
- Department of Biomedical Engineering, Faculty of Engineering, National University of Singapore, Singapore, 117583, Singapore.,Institute for Health Innovation and Technology, National University of Singapore, Singapore, 117599, Singapore.,Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore, 138673, Singapore.,Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore
| |
Collapse
|
121
|
Eren E, Hunt JFV, Shardell M, Chawla S, Tran J, Gu J, Vogt NM, Johnson SC, Bendlin BB, Kapogiannis D. Extracellular vesicle biomarkers of Alzheimer's disease associated with sub-clinical cognitive decline in late middle age. Alzheimers Dement 2020; 16:1293-1304. [PMID: 32588967 PMCID: PMC7984100 DOI: 10.1002/alz.12130] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Revised: 04/08/2020] [Accepted: 05/17/2020] [Indexed: 12/12/2022]
Abstract
Introduction Neuronal extracellular vesicle (nEV) tau and insulin signaling biomarkers may detect preclinical Alzheimer's disease and age‐associated cognitive decline. Methods This case‐control study used repeated serum samples from 73 cognitively declining and 73 stable Wisconsin Registry for Alzheimer's Prevention participants (62.4 ± 6.3 years old). We immunocaptured nEVs; measured tau and insulin signaling biomarkers; and examined biomarker differences by group, their performance in group classification in training and test datasets (97, 49 individuals, respectively), and whether they predict cognitive performance change. Results Declining compared to stable individuals showed higher baseline total, p231‐, and p181‐tau with older age and higher annualized change for p‐IR and p‐IGF‐1R. Combining biomarkers classified decliners with 94% area under the curve (AUC), 86.0% sensitivity and 86.7% specificity, in training data, and 75% AUC, 71.4% sensitivity, and 77.3% specificity, in test data. Insulin biomarkers predicted cognitive performance change prospectively. Discussion Combining nEV biomarkers can identify individuals with age‐associated cognitive decline.
Collapse
Affiliation(s)
- Erden Eren
- Laboratory of Clinical InvestigationIntramural Research ProgramNational Institute on AgingNIHBaltimoreMarylandUSA
| | - Jack F. V. Hunt
- Wisconsin Alzheimer's Disease Research CenterUniversity of WisconsinMadisonWisconsinUSA
| | | | - Sahil Chawla
- Laboratory of Clinical InvestigationIntramural Research ProgramNational Institute on AgingNIHBaltimoreMarylandUSA
| | - Joyce Tran
- Laboratory of Clinical InvestigationIntramural Research ProgramNational Institute on AgingNIHBaltimoreMarylandUSA
| | - Jeffrey Gu
- Laboratory of Clinical InvestigationIntramural Research ProgramNational Institute on AgingNIHBaltimoreMarylandUSA
| | - Nick M. Vogt
- Wisconsin Alzheimer's Disease Research CenterUniversity of WisconsinMadisonWisconsinUSA
| | - Sterling C. Johnson
- Wisconsin Alzheimer's Disease Research CenterUniversity of WisconsinMadisonWisconsinUSA
- Geriatric Research Education and Clinical Center of the Wm. S. MiddletonMemorial Veterans HospitalMadisonWisconsinUSA
| | - Barbara B. Bendlin
- Wisconsin Alzheimer's Disease Research CenterUniversity of WisconsinMadisonWisconsinUSA
| | - Dimitrios Kapogiannis
- Laboratory of Clinical InvestigationIntramural Research ProgramNational Institute on AgingNIHBaltimoreMarylandUSA
| |
Collapse
|
122
|
Spinelli M, Fusco S, Grassi C. Brain insulin resistance impairs hippocampal plasticity. VITAMINS AND HORMONES 2020; 114:281-306. [PMID: 32723548 DOI: 10.1016/bs.vh.2020.04.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Nutrient-related signals have been demonstrated to influence brain development and cognitive functions. In particular, insulin signaling has been shown to impact on molecular cascades underlying hippocampal plasticity, learning and memory. Alteration of brain insulin signaling interferes with the maintenance of neural stem cell niche and neuronal activity in the hippocampus. Brain insulin resistance is also emerging as key factor causing the cognitive impairment observed in metabolic and neurodegenerative diseases. Here, we review the molecular mechanisms involved in the insulin modulation of both adult neurogenesis and synaptic activity in the hippocampus. We also summarize the effects of altered insulin sensitivity on hippocampal plasticity. Finally, we reassume the experimental and epidemiological evidence highlighting the critical role of brain insulin resistance at the crossroad between type 2 diabetes and Alzheimer's disease.
Collapse
Affiliation(s)
- Matteo Spinelli
- Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Salvatore Fusco
- Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, Italy; Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy.
| | - Claudio Grassi
- Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, Italy; Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy.
| |
Collapse
|
123
|
Bhargava P, Nogueras-Ortiz C, Kim S, Delgado-Peraza F, Calabresi PA, Kapogiannis D. Synaptic and complement markers in extracellular vesicles in multiple sclerosis. Mult Scler 2020; 27:509-518. [PMID: 32669030 DOI: 10.1177/1352458520924590] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
BACKGROUND Synaptic loss is a feature of multiple sclerosis pathology that can be seen even in normal-appearing gray matter. Opsonization of synapses with complement components may underlie pathologic synapse loss. OBJECTIVE We sought to determine whether circulating neuronal-enriched and astrocytic-enriched extracellular vesicles (NEVs and AEVs) provide biomarkers reflecting complement-mediated synaptic loss in multiple sclerosis. METHODS From plasma of 61 people with multiple sclerosis (46 relapsing-remitting multiple sclerosis (RRMS) and 15 progressive MS) and 31 healthy controls, we immunocaptured L1CAM + NEVs and GLAST + AEVs. We measured pre- and post-synaptic proteins synaptopodin and synaptophysin in NEVs and complement components (C1q, C3, C3b/iC3b, C4, C5, C5a, C9, Factor B, and Factor H) in AEVs, total circulating EVs, and neat plasma. RESULTS We found lower levels of NEV synaptopodin and synaptophysin in MS compared to controls (p < 0.0001 for both). In AEVs, we found higher levels of multiple complement cascade components in people with MS compared to controls; these differences were not noted in total EVs or neat plasma. Strikingly, there were strong inverse correlations between NEV synaptic proteins and multiple AEV complement components in MS, but not in controls. CONCLUSION Circulating EVs could identify synaptic loss in MS and suggest a link between astrocytic complement production and synaptic loss.
Collapse
Affiliation(s)
- Pavan Bhargava
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Carlos Nogueras-Ortiz
- Laboratory of Clinical Investigation, National Institutes of Aging, Baltimore, MD, USA
| | - Sol Kim
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | | - Peter A Calabresi
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Dimitrios Kapogiannis
- Laboratory of Clinical Investigation, National Institutes of Aging, Baltimore, MD, USA/Biomedical Research Center, National Institute on Aging, National Institute of Health, Baltimore, MD, USA/ Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
124
|
Li CJ, Fang QH, Liu ML, Lin JN. Current understanding of the role of Adipose-derived Extracellular Vesicles in Metabolic Homeostasis and Diseases: Communication from the distance between cells/tissues. Am J Cancer Res 2020; 10:7422-7435. [PMID: 32642003 PMCID: PMC7330853 DOI: 10.7150/thno.42167] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2019] [Accepted: 05/26/2020] [Indexed: 12/22/2022] Open
Abstract
Extracellular vesicles (EVs) including exosomes, microvesicles (MVs), and apoptotic bodies, are small membrane vesicular structures that are released during cell activation, senescence, or programmed cell death, including apoptosis, necroptosis, and pyroptosis. EVs serve as novel mediators for long-distance cell-to-cell communications and can transfer various bioactive molecules, such as encapsulated cytokines and genetic information from their parental cells to distant target cells. In the context of obesity, adipocyte-derived EVs are implicated in metabolic homeostasis serving as novel adipokines. In particular, EVs released from brown adipose tissue or adipose-derived stem cells may help control the remolding of white adipose tissue towards browning and maintaining metabolic homeostasis. Interestingly, EVs may even serve as mediators for the transmission of metabolic dysfunction across generations. Also, EVs have been recognized as novel modulators in various metabolic disorders, including insulin resistance, diabetes mellitus, and non-alcoholic fatty liver disease. In this review, we summarize the latest progress from basic and translational studies regarding the novel effects of EVs on metabolic diseases. We also discuss EV-mediated cross-talk between adipose tissue and other organs/tissues that are relevant to obesity and metabolic diseases, as well as the relevant mechanisms, providing insight into the development of new therapeutic strategies in obesity and metabolic diseases.
Collapse
|
125
|
Serpente M, Fenoglio C, D'Anca M, Arcaro M, Sorrentino F, Visconte C, Arighi A, Fumagalli GG, Porretti L, Cattaneo A, Ciani M, Zanardini R, Benussi L, Ghidoni R, Scarpini E, Galimberti D. MiRNA Profiling in Plasma Neural-Derived Small Extracellular Vesicles from Patients with Alzheimer's Disease. Cells 2020; 9:cells9061443. [PMID: 32531989 PMCID: PMC7349735 DOI: 10.3390/cells9061443] [Citation(s) in RCA: 82] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 06/04/2020] [Accepted: 06/08/2020] [Indexed: 02/06/2023] Open
Abstract
Small extracellular vesicles (EVs) are able to pass from the central nervous system (CNS) into peripheral blood and contain molecule markers of their parental origin. The aim of our study was to isolate and characterize total and neural-derived small EVs (NDEVs) and their micro RNA (miRNA) cargo in Alzheimer's disease (AD) patients. Small NDEVs were isolated from plasma in a population consisting of 40 AD patients and 40 healthy subjects (CTRLs) using high throughput Advanced TaqMan miRNA OpenArrays®, which enables the simultaneous determination of 754 miRNAs. MiR-23a-3p, miR-223-3p, miR-100-3p and miR-190-5p showed a significant dysregulation in small NDEVs from AD patients as compared with controls (1.16 ± 0.49 versus 7.54 ± 2.5, p = 0.026; 9.32 ± 2.27 versus 0.66 ± 0.18, p <0.0001; 0.069 ± 0.01 versus 0.5 ± 0.1, p < 0.0001 and 2.9 ± 1.2 versus 1.93 ± 0.9, p < 0.05, respectively). A further validation analysis confirmed that miR-23a-3p, miR-223-3p and miR-190a-5p levels in small NDEVs from AD patients were significantly upregulated as compared with controls (p = 0.008; p = 0.016; p = 0.003, respectively) whereas miR-100-3p levels were significantly downregulated (p = 0.008). This is the first study that carries out the comparison between total plasma small EV population and NDEVs, demonstrating the presence of a specific AD NDEV miRNA signature.
Collapse
Affiliation(s)
- Maria Serpente
- Department of Pathophysiology and Transplantation, Dino Ferrari Center, University of Milan, 20122 Milan, Italy
| | - Chiara Fenoglio
- Department of Pathophysiology and Transplantation, Dino Ferrari Center, University of Milan, 20122 Milan, Italy
| | - Marianna D'Anca
- Department of Pathophysiology and Transplantation, Dino Ferrari Center, University of Milan, 20122 Milan, Italy
| | - Marina Arcaro
- Neurodegenerative Diseases Unit, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Federica Sorrentino
- Neurodegenerative Diseases Unit, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, 20122 Milan, Italy
- Department of Biomedical, Surgical and Dental Sciences, Dino Ferrari Center, CRC Molecular Basis of Neuro-Psycho-Geriatrics Diseases, University of Milan, 20122 Milan, Italy
| | - Caterina Visconte
- Neurodegenerative Diseases Unit, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Andrea Arighi
- Neurodegenerative Diseases Unit, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Giorgio G Fumagalli
- Neurodegenerative Diseases Unit, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Laura Porretti
- Flow Cytometry Service, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Alessandra Cattaneo
- Department of Transfusion Medicine and Haematology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Miriam Ciani
- Molecular Markers Laboratory, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, 25125 Brescia, Italy
| | - Roberta Zanardini
- Molecular Markers Laboratory, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, 25125 Brescia, Italy
| | - Luisa Benussi
- Molecular Markers Laboratory, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, 25125 Brescia, Italy
| | - Roberta Ghidoni
- Molecular Markers Laboratory, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, 25125 Brescia, Italy
| | - Elio Scarpini
- Neurodegenerative Diseases Unit, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, 20122 Milan, Italy
- Department of Biomedical, Surgical and Dental Sciences, Dino Ferrari Center, CRC Molecular Basis of Neuro-Psycho-Geriatrics Diseases, University of Milan, 20122 Milan, Italy
| | - Daniela Galimberti
- Neurodegenerative Diseases Unit, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, 20122 Milan, Italy
- Department of Biomedical, Surgical and Dental Sciences, Dino Ferrari Center, CRC Molecular Basis of Neuro-Psycho-Geriatrics Diseases, University of Milan, 20122 Milan, Italy
| |
Collapse
|
126
|
Zhang N, Gu D, Meng M, Gordon ML. TDP-43 Is Elevated in Plasma Neuronal-Derived Exosomes of Patients With Alzheimer's Disease. Front Aging Neurosci 2020; 12:166. [PMID: 32581773 PMCID: PMC7287025 DOI: 10.3389/fnagi.2020.00166] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Accepted: 05/14/2020] [Indexed: 12/14/2022] Open
Abstract
Background Recently, TDP-43 has been recognized as a common proteinopathy in the “oldest old” and a neuropathological comorbidity in patients with Alzheimer’s disease (AD). However, since it has a low concentration in cerebrospinal fluid, the presence of TDP-43 in AD is rarely investigated in vivo. Methods Twenty-four patients with amyloid PET confirmed AD and 15 healthy controls (HCs) were included in this study. TDP-43 level in plasma neuronal-derived exosomes (NDEs) was measured by enzyme-linked immunosorbent assay. Results TDP-43 level was elevated in patients with AD compared with HCs (median 1.08 ng/ml, IQR 0.72–1.37 ng/ml vs. median 0.66 ng/ml, IQR 0.48–0.76 ng/ml, P = 0.002). There was no correlation between TDP-43 level and cognitive function, neuropsychiatric symptoms or APOE genotype in patients with AD. Conclusion This study demonstrated increased TDP-43 accumulation in AD patients by examining plasma NDEs, which may provide a window into the effects of TDP-43 on AD progression.
Collapse
Affiliation(s)
- Nan Zhang
- Department of Neurology, Tianjin Medical University General Hospital, Tianjin Neurological Institute, Tianjin, China.,Department of Neurology, Tianjin Medical University General Hospital Airport Hospital, Tianjin, China
| | - Dongmei Gu
- Department of Clinical Laboratory Medicine, Tianjin Medical University General Hospital, Tianjin, China
| | - Meng Meng
- Department of Neurology, Tianjin Medical University General Hospital Airport Hospital, Tianjin, China
| | - Marc L Gordon
- The Litwin-Zucker Research Center, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, United States.,Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, United States
| |
Collapse
|
127
|
Cheng L, Vella LJ, Barnham KJ, McLean C, Masters CL, Hill AF. Small RNA fingerprinting of Alzheimer's disease frontal cortex extracellular vesicles and their comparison with peripheral extracellular vesicles. J Extracell Vesicles 2020; 9:1766822. [PMID: 32922692 PMCID: PMC7448944 DOI: 10.1080/20013078.2020.1766822] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Alzheimer’s disease is a progressive neurodegenerative disorder, with the strongest disease-associated changes observed at clinical or end-stage disease. Transcriptomic deregulation of miRNA expression can spread via “horizontal” RNA transfer through extracellular vesicles (EVs) to act in conjunction with proteins, leading to changes in mRNA, which can provide early signals to indicate forthcoming neuropathological changes in the brain. Here, we analysed the small RNA content, in particular, miRNA, contained in brain-derived EVs isolated from the frontal cortex of Alzheimer’s subjects (n = 8) and neurological control subjects (n = 9). Brain-derived EVs were found to contain an upregulation of disease-associated miRNA. RNA species from brain-derived EVs were correlated with miRNA profiles obtained from matching total brain homogenate. These results provide a blueprint into the biological pathways potentially effected during disease that may be assisted by brain-derived EV RNA horizontal transfer.We also correlated the miRNA changes in the brain with those detected in peripheral EVs collected from serum of Alzheimer’s disease patients (n = 23, and healthy controls, n = 23) and revealed a panel of miRNA that could be used as a liquid brain biopsy. Overall, our study provides the first interrogation of the small RNA contents in brain-derived EVs and how they could be used to understand the early pathological changes in Alzheimer’s disease which will benefit the development of an early diagnostic blood test.
Collapse
Affiliation(s)
- Lesley Cheng
- The Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Victoria, Australia
| | - Laura J Vella
- Florey Institute of Neuroscience, The University of Melbourne, Victoria, Australia
| | - Kevin J Barnham
- Florey Institute of Neuroscience, The University of Melbourne, Victoria, Australia
| | - Catriona McLean
- Florey Institute of Neuroscience, The University of Melbourne, Victoria, Australia.,Victorian Brain Bank, Florey Institute of Neurosciences, Victoria, Australia
| | - Colin L Masters
- Florey Institute of Neuroscience, The University of Melbourne, Victoria, Australia
| | - Andrew F Hill
- The Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Victoria, Australia
| |
Collapse
|
128
|
Song Z, Xu Y, Deng W, Zhang L, Zhu H, Yu P, Qu Y, Zhao W, Han Y, Qin C. Brain Derived Exosomes Are a Double-Edged Sword in Alzheimer's Disease. Front Mol Neurosci 2020; 13:79. [PMID: 32547364 PMCID: PMC7274346 DOI: 10.3389/fnmol.2020.00079] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Accepted: 04/22/2020] [Indexed: 12/12/2022] Open
Abstract
Brain derived exosomes (BDEs) are extracellular nanovesicles that are collectively released by all cell lineages of the central nervous system and contain cargo from their original cells. They are emerging as key mediators of communication and waste management among neurons, glial cells and connective tissue during both physiological and pathological conditions in the brain. We review the rapidly growing frontier of BDEs biology in recent years including the involvement of exosomes in neuronal development, maintenance and communication through their multiple signaling functions. Particularly, we highlight the important role of exosomes in Alzheimer's disease (AD), both as a pathogenic agent and as a disease biomarker. Our understanding of such unique nanovesicles may offer not only answers about the (patho) physiological course in AD and associated neurodegenerative diseases but also ideal methods to develop these vesicles as vehicles for drug delivery or as tools to monitor brain diseases in a non-invasive manner because crossing the blood brain barrier is an inherent capability of exosomes. BDEs have potential as biomarkers and as therapeutic tools for AD and related brain disorders in the near future.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Chuan Qin
- Key Laboratory of Human Disease Comparative Medicine, Chinese Ministry of Health, Beijing Key Laboratory for Animal Models of Emerging and Remerging Infectious Diseases, Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences, Comparative Medicine Center, Peking Union Medical College, Beijing, China
| |
Collapse
|
129
|
Sáez T, Toledo F, Sobrevia L. Extracellular Vesicles and Insulin Resistance: A Potential Interaction in Vascular Dysfunction. Curr Vasc Pharmacol 2020; 17:491-497. [PMID: 30277159 DOI: 10.2174/1570161116666181002095745] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Revised: 09/11/2018] [Accepted: 09/11/2018] [Indexed: 12/19/2022]
Abstract
Insulin resistance plays a key role in cardiovascular complications associated with diabetes mellitus and hypertensive disorders. In states of insulin resistance several circulating factors may contribute to a defective insulin sensitivity in different tissues, including the vasculature. One of these factors influencing the vascular insulin resistance are the extracellular vesicles. The extracellular vesicles include exosomes, microvesicles, and apoptotic bodies which are released to the circulation by different vascular cells. Since the cargo of extracellular vesicles seems to be altered in metabolic complications associated with insulin resistance, these vesicles may be candidates contributing to vascular insulin resistance. Despite the studies linking insulin resistance signalling pathways with the vascular effect of extracellular vesicles, the involvement of these structures in vascular insulin resistance is a phenomenon that remains unclear.
Collapse
Affiliation(s)
- Tamara Sáez
- Department of Obstetrics and Gynecology, University of Alberta, Edmonton T6G 2S2, AB, Canada.,Women and Children's Health Research Institute, University of Alberta, Edmonton T6G 2S2, AB, Canada
| | - Fernando Toledo
- Department of Basic Sciences, Faculty of Sciences, Bio-Bio University, Chillan 3780000, Chile.,Cellular and Molecular Physiology Laboratory (CMPL), Department of Obstetrics, Division of Obstetrics and Gynaecology, School of Medicine, Faculty of Medicine, Pontifical Catholic University of Chile, Santiago 8330024, Chile
| | - Luis Sobrevia
- Cellular and Molecular Physiology Laboratory (CMPL), Department of Obstetrics, Division of Obstetrics and Gynaecology, School of Medicine, Faculty of Medicine, Pontifical Catholic University of Chile, Santiago 8330024, Chile.,Department of Physiology, Faculty of Pharmacy, University of Sevilla, Seville E-41012, Spain.,University of Queensland Centre for Clinical Research (UQCCR), Faculty of Medicine and Biomedical Sciences, University of Queensland, Herston, QLD 4029, Queensland, Australia
| |
Collapse
|
130
|
Qin T, Prins S, Groeneveld GJ, Van Westen G, de Vries HE, Wong YC, Bischoff LJ, de Lange EC. Utility of Animal Models to Understand Human Alzheimer's Disease, Using the Mastermind Research Approach to Avoid Unnecessary Further Sacrifices of Animals. Int J Mol Sci 2020; 21:ijms21093158. [PMID: 32365768 PMCID: PMC7247586 DOI: 10.3390/ijms21093158] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 04/27/2020] [Accepted: 04/28/2020] [Indexed: 12/18/2022] Open
Abstract
To diagnose and treat early-stage (preclinical) Alzheimer’s disease (AD) patients, we need body-fluid-based biomarkers that reflect the processes that occur in this stage, but current knowledge on associated processes is lacking. As human studies on (possible) onset and early-stage AD would be extremely expensive and time-consuming, we investigate the potential value of animal AD models to help to fill this knowledge gap. We provide a comprehensive overview of processes associated with AD pathogenesis and biomarkers, current knowledge on AD-related biomarkers derived from on human and animal brains and body fluids, comparisons of biomarkers obtained in human AD and frequently used animal AD models, and emerging body-fluid-based biomarkers. In human studies, amyloid beta (Aβ), hyperphosphorylated tau (P-tau), total tau (T-tau), neurogranin, SNAP-25, glial fibrillary acidic protein (GFAP), YKL-40, and especially neurofilament light (NfL) are frequently measured. In animal studies, the emphasis has been mostly on Aβ. Although a direct comparison between human (familial and sporadic) AD and (mostly genetic) animal AD models cannot be made, still, in brain, cerebrospinal fluid (CSF), and blood, a majority of similar trends are observed for human AD stage and animal AD model life stage. This indicates the potential value of animal AD models in understanding of the onset and early stage of AD. Moreover, animal studies can be smartly designed to provide mechanistic information on the interrelationships between the different AD processes in a longitudinal fashion and may also include the combinations of different conditions that may reflect comorbidities in human AD, according to the Mastermind Research approach.
Collapse
Affiliation(s)
- Tian Qin
- Predictive Pharmacology, Division of Systems Biomedicine and Pharmacology, Leiden Academic Centre of Drug Research, Leiden University, 2333 CC Leiden, The Netherlands; (T.Q.); (L.J.M.B.)
| | - Samantha Prins
- Centre for Human Drug Research (CHDR), 2333 CL Leiden, The Netherlands; (S.P.); (G.J.G.)
| | - Geert Jan Groeneveld
- Centre for Human Drug Research (CHDR), 2333 CL Leiden, The Netherlands; (S.P.); (G.J.G.)
| | - Gerard Van Westen
- Computational Drug Discovery, Division of Drug Discovery and Safety, Leiden Academic Centre of Drug Research, Leiden University, 2333 CC Leiden, The Netherlands;
| | - Helga E. de Vries
- Neuro-immunology research group, Department of Molecular Cell Biology and Immunology, Amsterdam Neuroscience, Amsterdam UMC, 1081 HZ Amsterdam, The Netherlands;
| | - Yin Cheong Wong
- Advanced Modelling and Simulation, UCB Celltech, Slough SL1 3WE, UK;
| | - Luc J.M. Bischoff
- Predictive Pharmacology, Division of Systems Biomedicine and Pharmacology, Leiden Academic Centre of Drug Research, Leiden University, 2333 CC Leiden, The Netherlands; (T.Q.); (L.J.M.B.)
| | - Elizabeth C.M. de Lange
- Predictive Pharmacology, Division of Systems Biomedicine and Pharmacology, Leiden Academic Centre of Drug Research, Leiden University, 2333 CC Leiden, The Netherlands; (T.Q.); (L.J.M.B.)
- Correspondence: ; Tel.: +31-71-527-6330
| |
Collapse
|
131
|
Type 3 Diabetes and Its Role Implications in Alzheimer's Disease. Int J Mol Sci 2020; 21:ijms21093165. [PMID: 32365816 PMCID: PMC7246646 DOI: 10.3390/ijms21093165] [Citation(s) in RCA: 185] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2020] [Revised: 04/21/2020] [Accepted: 04/28/2020] [Indexed: 12/13/2022] Open
Abstract
The exact connection between Alzheimer’s disease (AD) and type 2 diabetes is still in debate. However, poorly controlled blood sugar may increase the risk of developing Alzheimer’s. This relationship is so strong that some have called Alzheimer’s “diabetes of the brain” or “type 3 diabetes (T3D)”. Given more recent studies continue to indicate evidence linking T3D with AD, this review aims to demonstrate the relationship between T3D and AD based on the fact that both the processing of amyloid-β (Aβ) precursor protein toxicity and the clearance of Aβ are attributed to impaired insulin signaling, and that insulin resistance mediates the dysregulation of bioenergetics and progress to AD. Furthermore, insulin-related therapeutic strategies are suggested to succeed in the development of therapies for AD by slowing down their progressive nature or even halting their future complications.
Collapse
|
132
|
Ashton NJ, Hye A, Rajkumar AP, Leuzy A, Snowden S, Suárez-Calvet M, Karikari TK, Schöll M, La Joie R, Rabinovici GD, Höglund K, Ballard C, Hortobágyi T, Svenningsson P, Blennow K, Zetterberg H, Aarsland D. An update on blood-based biomarkers for non-Alzheimer neurodegenerative disorders. Nat Rev Neurol 2020; 16:265-284. [PMID: 32322100 DOI: 10.1038/s41582-020-0348-0] [Citation(s) in RCA: 122] [Impact Index Per Article: 24.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/17/2020] [Indexed: 01/11/2023]
Abstract
Cerebrospinal fluid analyses and neuroimaging can identify the underlying pathophysiology at the earliest stage of some neurodegenerative disorders, but do not have the scalability needed for population screening. Therefore, a blood-based marker for such pathophysiology would have greater utility in a primary care setting and in eligibility screening for clinical trials. Rapid advances in ultra-sensitive assays have enabled the levels of pathological proteins to be measured in blood samples, but research has been predominantly focused on Alzheimer disease (AD). Nonetheless, proteins that were identified as potential blood-based biomarkers for AD, for example, amyloid-β, tau, phosphorylated tau and neurofilament light chain, are likely to be relevant to other neurodegenerative disorders that involve similar pathological processes and could also be useful for the differential diagnosis of clinical symptoms. This Review outlines the neuropathological, clinical, molecular imaging and cerebrospinal fluid features of the most common neurodegenerative disorders outside the AD continuum and gives an overview of the current status of blood-based biomarkers for these disorders.
Collapse
Affiliation(s)
- Nicholas J Ashton
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden.,Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden.,Department of Old Age Psychiatry, Institute of Psychiatry, Psychology & Neuroscience, Maurice Wohl Clinical Neuroscience Institute, King's College London, London, UK.,NIHR Biomedical Research Centre for Mental Health & Biomedical Research Unit for Dementia at South London & Maudsley NHS Foundation, London, UK
| | - Abdul Hye
- Department of Old Age Psychiatry, Institute of Psychiatry, Psychology & Neuroscience, Maurice Wohl Clinical Neuroscience Institute, King's College London, London, UK.,NIHR Biomedical Research Centre for Mental Health & Biomedical Research Unit for Dementia at South London & Maudsley NHS Foundation, London, UK
| | - Anto P Rajkumar
- Department of Old Age Psychiatry, Institute of Psychiatry, Psychology & Neuroscience, Maurice Wohl Clinical Neuroscience Institute, King's College London, London, UK.,NIHR Biomedical Research Centre for Mental Health & Biomedical Research Unit for Dementia at South London & Maudsley NHS Foundation, London, UK.,Institute of Mental Health, University of Nottingham, Nottingham, UK
| | - Antoine Leuzy
- Clinical Memory Research Unit, Lund University, Malmö, Sweden
| | - Stuart Snowden
- Core Metabolomics and Lipidomics Laboratory, Metabolic Research Laboratories, Institute of Metabolic Science, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - Marc Suárez-Calvet
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden.,Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Catalonia, Spain.,Department of Neurology, Hospital del Mar, Barcelona, Catalonia, Spain
| | - Thomas K Karikari
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Michael Schöll
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden.,Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden.,Clinical Memory Research Unit, Lund University, Malmö, Sweden.,Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK
| | - Renaud La Joie
- Memory and Aging Center, University of California, San Francisco, San Francisco, CA, USA
| | - Gil D Rabinovici
- Memory and Aging Center, University of California, San Francisco, San Francisco, CA, USA
| | - Kina Höglund
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden.,Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Disease Research, Neurogeriatrics Division, Karolinska Institutet, Novum, Huddinge, Stockholm, Sweden
| | | | - Tibor Hortobágyi
- Department of Old Age Psychiatry, Institute of Psychiatry, Psychology & Neuroscience, Maurice Wohl Clinical Neuroscience Institute, King's College London, London, UK.,MTA-DE Cerebrovascular and Neurodegenerative Research Group, Department of Neurology, University of Debrecen, Debrecen, Hungary
| | - Per Svenningsson
- Department of Old Age Psychiatry, Institute of Psychiatry, Psychology & Neuroscience, Maurice Wohl Clinical Neuroscience Institute, King's College London, London, UK.,Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden.,Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden.,Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden.,Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, UK.,UK Dementia Research Institute at UCL, London, UK
| | - Dag Aarsland
- Department of Old Age Psychiatry, Institute of Psychiatry, Psychology & Neuroscience, Maurice Wohl Clinical Neuroscience Institute, King's College London, London, UK. .,NIHR Biomedical Research Centre for Mental Health & Biomedical Research Unit for Dementia at South London & Maudsley NHS Foundation, London, UK. .,Centre for Age-Related Medicine, Stavanger University Hospital, Stavanger, Norway.
| |
Collapse
|
133
|
Nguyen TT, Ta QTH, Nguyen TTD, Le TT, Vo VG. Role of Insulin Resistance in the Alzheimer's Disease Progression. Neurochem Res 2020; 45:1481-1491. [PMID: 32314178 DOI: 10.1007/s11064-020-03031-0] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 04/05/2020] [Accepted: 04/09/2020] [Indexed: 02/06/2023]
|
134
|
Upadhya R, Zingg W, Shetty S, Shetty AK. Astrocyte-derived extracellular vesicles: Neuroreparative properties and role in the pathogenesis of neurodegenerative disorders. J Control Release 2020; 323:225-239. [PMID: 32289328 DOI: 10.1016/j.jconrel.2020.04.017] [Citation(s) in RCA: 158] [Impact Index Per Article: 31.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 04/08/2020] [Accepted: 04/09/2020] [Indexed: 12/11/2022]
Abstract
Extracellular vesicles (EVs) released by neural cells play an essential role in brain homeostasis and the crosstalk between neural cells and the periphery. EVs are diverse, nano-sized vesicles, which transport proteins, nucleic acids, and lipids between cells over short and long expanses and hence are proficient for modulating the target cells. EVs released from neural cells are implicated in synaptic plasticity, neuron-glia interface, neuroprotection, neuroregeneration, and the dissemination of neuropathological molecules. This review confers the various properties of EVs secreted by astrocytes and their potential role in health and disease with a focus on evolving concepts. Naïve astrocytes shed EVs containing a host of neuroprotective compounds, which include fibroblast growth factor-2, vascular endothelial growth factor, and apolipoprotein-D. Stimulated astrocytes secrete EVs with neuroprotective molecules including heat shock proteins, synapsin 1, unique microRNAs, and glutamate transporters. Well-characterized astrocyte-derived EVs (ADEVs) generated in specific culture conditions and ADEVs that are engineered to carry the desired miRNAs or proteins are likely useful for treating brain injury and neurogenerative diseases. On the other hand, in conditions such as Alzheimer's disease (AD), stroke, Parkinson's disease, Amyotrophic lateral sclerosis (ALS), and other neuroinflammatory conditions, EVs released by activated astrocytes appear to mediate or exacerbate the pathological processes. The examples include ADEVs spreading the dysregulated complement system in AD, mediating motoneuron toxicity in ALS, and stimulating peripheral leukocyte migration into the brain in inflammatory conditions. Strategies restraining the release of EVs by activated astrocytes or modulating the composition of ADEVs are likely beneficial for treating neurodegenerative diseases. Also, periodic analyses of ADEVs in the blood is useful for detecting astrocyte-specific biomarkers in different neurological conditions and for monitoring disease progression and remission with distinct therapeutic approaches.
Collapse
Affiliation(s)
- Raghavendra Upadhya
- Institute for Regenerative Medicine, Department of Molecular and Cellular Medicine, Texas A&M University College of Medicine, College Station, TX, USA
| | - Winston Zingg
- Institute for Regenerative Medicine, Department of Molecular and Cellular Medicine, Texas A&M University College of Medicine, College Station, TX, USA
| | - Siddhant Shetty
- Institute for Regenerative Medicine, Department of Molecular and Cellular Medicine, Texas A&M University College of Medicine, College Station, TX, USA
| | - Ashok K Shetty
- Institute for Regenerative Medicine, Department of Molecular and Cellular Medicine, Texas A&M University College of Medicine, College Station, TX, USA.
| |
Collapse
|
135
|
Mansur RB, Delgado-Peraza F, Subramaniapillai M, Lee Y, Iacobucci M, Rodrigues N, Rosenblat JD, Brietzke E, Cosgrove VE, Kramer NE, Suppes T, Raison CL, Chawla S, Nogueras-Ortiz C, McIntyre RS, Kapogiannis D. Extracellular Vesicle Biomarkers Reveal Inhibition of Neuroinflammation by Infliximab in Association with Antidepressant Response in Adults with Bipolar Depression. Cells 2020; 9:cells9040895. [PMID: 32268604 PMCID: PMC7226726 DOI: 10.3390/cells9040895] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 03/28/2020] [Accepted: 03/30/2020] [Indexed: 12/26/2022] Open
Abstract
Accumulating evidence suggests that neuroinflammation is involved in bipolar disorder (BD) pathogenesis. The tumor necrosis factor-alpha (TNF-α) antagonist infliximab was recently reported to improve depressive symptoms in a subpopulation of individuals with BD and history of childhood maltreatment. To explore the mechanistic mediators of infliximab's effects, we investigated its engagement with biomarkers of cellular response to inflammation derived from plasma extracellular vesicles enriched for neuronal origin (NEVs). We hypothesized that infliximab, compared to placebo, would decrease TNF-α receptors (TNFRs) and nuclear factor-kappa B (NF-κB) pathway signaling biomarkers, and that history of childhood abuse would moderate infliximab's effects. We immunocaptured NEVs from plasma samples collected at baseline and at weeks 2, 6, and 12 (endpoint) from 55 participants of this clinical trial and measured NEV biomarkers using immunoassays. A subset of participants (n = 27) also underwent whole-brain magnetic resonance imaging at baseline and endpoint. Childhood physical abuse moderated treatment by time interactions for TNFR1 (χ2 = 9.275, p = 0.026), NF-κB (χ2 = 13.825, p = 0.003), and inhibitor of NF-κB (IκBα) (χ2 = 7.990, p = 0.046), indicating that higher levels of physical abuse were associated with larger biomarker decreases over time. Moreover, the antidepressant response to infliximab was moderated by TNFR1 (χ2 = 7.997, p = 0.046). In infliximab-treated participants, reductions in TNFR1 levels were associated with improvement of depressive symptoms, an effect not detected in the placebo group. Conversely, reductions in TNFR1 levels were associated with increased global cortical thickness in infliximab- (r = -0.581, p = 0.029), but not placebo-treated, patients (r = 0.196, p = 0.501). In conclusion, we report that NEVs revealed that infliximab engaged the TNFR/NF-κB neuro-inflammatory pathway in individuals with BD, in a childhood trauma-dependent manner, which was associated with clinical response and brain structural changes.
Collapse
Affiliation(s)
- Rodrigo B. Mansur
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, ON M5T 2S8, Canada; (M.S.); (Y.L.); (M.I.); (N.R.); (J.D.R.); (E.B.); (R.S.M.)
- Department of Psychiatry, University of Toronto, Toronto, ON M5T 2S8, Canada
- Correspondence: ; Tel.: +4166-0358-00; Fax: +4166-0353-68
| | - Francheska Delgado-Peraza
- Laboratory of Clinical Investigation, Intramural Research Program, National Institute on Aging, National Institutes of Health (NIA/NIH), Baltimore, MD 20892, USA; (F.D.-P.); (S.C.); (C.N.-O.); (D.K.)
| | - Mehala Subramaniapillai
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, ON M5T 2S8, Canada; (M.S.); (Y.L.); (M.I.); (N.R.); (J.D.R.); (E.B.); (R.S.M.)
| | - Yena Lee
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, ON M5T 2S8, Canada; (M.S.); (Y.L.); (M.I.); (N.R.); (J.D.R.); (E.B.); (R.S.M.)
- Institute of Medical Science, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Michelle Iacobucci
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, ON M5T 2S8, Canada; (M.S.); (Y.L.); (M.I.); (N.R.); (J.D.R.); (E.B.); (R.S.M.)
| | - Nelson Rodrigues
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, ON M5T 2S8, Canada; (M.S.); (Y.L.); (M.I.); (N.R.); (J.D.R.); (E.B.); (R.S.M.)
| | - Joshua D. Rosenblat
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, ON M5T 2S8, Canada; (M.S.); (Y.L.); (M.I.); (N.R.); (J.D.R.); (E.B.); (R.S.M.)
- Department of Psychiatry, University of Toronto, Toronto, ON M5T 2S8, Canada
| | - Elisa Brietzke
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, ON M5T 2S8, Canada; (M.S.); (Y.L.); (M.I.); (N.R.); (J.D.R.); (E.B.); (R.S.M.)
- Kingston General Hospital, Providence Care Hospital, Department of Psychiatry, Queen’s University School of Medicine, Kingston, ON K7L 4X3, Canada
| | - Victoria E. Cosgrove
- Department of Psychiatry & Behavioral Sciences, Stanford University, School of Medicine, Palo Alto, CA 94304, USA; (V.E.C.); (N.E.K.); (T.S.)
| | - Nicole E. Kramer
- Department of Psychiatry & Behavioral Sciences, Stanford University, School of Medicine, Palo Alto, CA 94304, USA; (V.E.C.); (N.E.K.); (T.S.)
| | - Trisha Suppes
- Department of Psychiatry & Behavioral Sciences, Stanford University, School of Medicine, Palo Alto, CA 94304, USA; (V.E.C.); (N.E.K.); (T.S.)
| | - Charles L. Raison
- School of Human Ecology, University of Wisconsin-Madison, Madison, WI 53706, USA;
- Department of Psychiatry, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 30322, USA
| | - Sahil Chawla
- Laboratory of Clinical Investigation, Intramural Research Program, National Institute on Aging, National Institutes of Health (NIA/NIH), Baltimore, MD 20892, USA; (F.D.-P.); (S.C.); (C.N.-O.); (D.K.)
| | - Carlos Nogueras-Ortiz
- Laboratory of Clinical Investigation, Intramural Research Program, National Institute on Aging, National Institutes of Health (NIA/NIH), Baltimore, MD 20892, USA; (F.D.-P.); (S.C.); (C.N.-O.); (D.K.)
| | - Roger S. McIntyre
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, ON M5T 2S8, Canada; (M.S.); (Y.L.); (M.I.); (N.R.); (J.D.R.); (E.B.); (R.S.M.)
- Department of Psychiatry, University of Toronto, Toronto, ON M5T 2S8, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Dimitrios Kapogiannis
- Laboratory of Clinical Investigation, Intramural Research Program, National Institute on Aging, National Institutes of Health (NIA/NIH), Baltimore, MD 20892, USA; (F.D.-P.); (S.C.); (C.N.-O.); (D.K.)
| |
Collapse
|
136
|
Saint-Pol J, Gosselet F, Duban-Deweer S, Pottiez G, Karamanos Y. Targeting and Crossing the Blood-Brain Barrier with Extracellular Vesicles. Cells 2020; 9:cells9040851. [PMID: 32244730 PMCID: PMC7226770 DOI: 10.3390/cells9040851] [Citation(s) in RCA: 303] [Impact Index Per Article: 60.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 03/23/2020] [Accepted: 03/31/2020] [Indexed: 12/18/2022] Open
Abstract
The blood–brain barrier (BBB) is one of the most complex and selective barriers in the human organism. Its role is to protect the brain and preserve the homeostasis of the central nervous system (CNS). The central elements of this physical and physiological barrier are the endothelial cells that form a monolayer of tightly joined cells covering the brain capillaries. However, as endothelial cells regulate nutrient delivery and waste product elimination, they are very sensitive to signals sent by surrounding cells and their environment. Indeed, the neuro-vascular unit (NVU) that corresponds to the assembly of extracellular matrix, pericytes, astrocytes, oligodendrocytes, microglia and neurons have the ability to influence BBB physiology. Extracellular vesicles (EVs) play a central role in terms of communication between cells. The NVU is no exception, as each cell can produce EVs that could help in the communication between cells in short or long distances. Studies have shown that EVs are able to cross the BBB from the brain to the bloodstream as well as from the blood to the CNS. Furthermore, peripheral EVs can interact with the BBB leading to changes in the barrier’s properties. This review focuses on current knowledge and potential applications regarding EVs associated with the BBB.
Collapse
Affiliation(s)
- Julien Saint-Pol
- Laboratoire de la Barrière Hémato-Encéphalique (LBHE), UR 2465, University Artois, F-62300 Lens, France; (F.G.); (S.D.-D.); (Y.K.)
- Correspondence: ; Tel.: +33-3-2179-1746
| | - Fabien Gosselet
- Laboratoire de la Barrière Hémato-Encéphalique (LBHE), UR 2465, University Artois, F-62300 Lens, France; (F.G.); (S.D.-D.); (Y.K.)
| | - Sophie Duban-Deweer
- Laboratoire de la Barrière Hémato-Encéphalique (LBHE), UR 2465, University Artois, F-62300 Lens, France; (F.G.); (S.D.-D.); (Y.K.)
| | - Gwënaël Pottiez
- Caprion Biosciences Inc., 141, Avenue du Président-Kennedy Suite 5650, Montréal, QC H2X3Y7, Canada;
| | - Yannis Karamanos
- Laboratoire de la Barrière Hémato-Encéphalique (LBHE), UR 2465, University Artois, F-62300 Lens, France; (F.G.); (S.D.-D.); (Y.K.)
| |
Collapse
|
137
|
Chawla S, Gulyani S, Allen RP, Earley CJ, Li X, Van Zijl P, Kapogiannis D. Extracellular vesicles reveal abnormalities in neuronal iron metabolism in restless legs syndrome. Sleep 2020; 42:5415757. [PMID: 30895312 DOI: 10.1093/sleep/zsz079] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2018] [Revised: 02/19/2019] [Indexed: 12/31/2022] Open
Abstract
STUDY OBJECTIVES Determine abnormalities in levels of iron-management proteins in neuronal origin-enriched extracellular vesicles (nEVs) in restless legs syndrome (RLS). METHODS We used immunoprecipitation for neuronal marker L1CAM to isolate nEVs from the serum of 20 participants with RLS from a study including magnetic resonance imaging (MRI) determinations of iron deposition in the substantia nigra and hematologic parameters and 28 age- and sex-matched Controls. RESULTS RLS compared with Control participants showed higher levels of nEV total ferritin but similar levels of transferrin receptor and ferroportin. Western blot analysis showed that heavy- but not light-chain ferritin was increased in nEVs of RLS compared with Control participants. In RLS but not Control participants, nEV total ferritin was positively correlated with systemic iron parameters; the two groups also differed in the relation of nEV total ferritin to MRI measures of iron deposition in substantia nigra. CONCLUSIONS Given the neuronal origin and diversity of EV cargo, nEVs provide an important platform for exploring the underlying pathophysiology and possible biomarkers of RLS.
Collapse
Affiliation(s)
- Sahil Chawla
- Laboratory of Neurosciences, Intramural Research Program, National Institute on Aging, NIH, Baltimore, MD
| | - Seema Gulyani
- Johns Hopkins Sleep Disorders Center, Johns Hopkins Bayview Medical Center, Baltimore, MD
| | - Richard P Allen
- Department of Neurology, The Johns Hopkins Center for Restless Legs Syndrome, Johns Hopkins Bayview Medical Center, Baltimore, MD
| | - Christopher J Earley
- Department of Neurology, The Johns Hopkins Center for Restless Legs Syndrome, Johns Hopkins Bayview Medical Center, Baltimore, MD
| | - Xu Li
- Department of Radiology and Radiological Science, Division of MR Research, Johns Hopkins Medicine, Baltimore, MD
| | - Peter Van Zijl
- Department of Radiology and Radiological Science, Division of MR Research, Johns Hopkins Medicine, Baltimore, MD
| | - Dimitrios Kapogiannis
- Laboratory of Neurosciences, Intramural Research Program, National Institute on Aging, NIH, Baltimore, MD
| |
Collapse
|
138
|
Schou AS, Nielsen JE, Askeland A, Jørgensen MM. Extracellular vesicle-associated proteins as potential biomarkers. Adv Clin Chem 2020; 99:1-48. [PMID: 32951635 DOI: 10.1016/bs.acc.2020.02.011] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Every cell in the body secretes extracellular vesicles (EVs) possibly as cellular signaling components and these cell-derivatives can be found in multiple numbers in biological fluids. EVs have in the scientific field received great attention in relation to pathophysiology and disease diagnostics. Altered protein expressions associated with circulating EVs in diseased individuals can serve as biomarkers for different disease states. This capacity paves the way for non-invasive screening tools and early diagnostic markers. However, no isolation method of EVs has been acknowledged as the "golden standard," thus reproducibility of the studies remains inadequate. Increasing interest in EV proteins as disease biomarkers could give rise to more scientific knowledge with diagnostic applicability. In this chapter, studies of proteins believed to be associated with EVs within cancer, autoimmunity, metabolic and neurodegenerative diseases have been outlined.
Collapse
Affiliation(s)
- Anne Sophie Schou
- Department of Clinical Immunology, Aalborg University Hospital, Aalborg, Denmark; Research Unit for Molecular Medicine, Department of Clinical Medicine, Aarhus University and Aarhus University Hospital, Aarhus, Denmark
| | - Jonas Ellegaard Nielsen
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark; Department of Clinical Biochemistry, Aalborg University Hospital, Aalborg, Denmark
| | - Anders Askeland
- Department of Clinical Biochemistry, Aalborg University Hospital, Aalborg, Denmark
| | - Malene Møller Jørgensen
- Department of Clinical Immunology, Aalborg University Hospital, Aalborg, Denmark; Department of Clinical Medicine, Aalborg University, Aalborg, Denmark.
| |
Collapse
|
139
|
Badhwar A, Haqqani AS. Biomarker potential of brain-secreted extracellular vesicles in blood in Alzheimer's disease. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2020; 12:e12001. [PMID: 32211497 PMCID: PMC7085285 DOI: 10.1002/dad2.12001] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Revised: 10/17/2019] [Accepted: 11/01/2019] [Indexed: 02/06/2023]
Abstract
INTRODUCTION Brain cells secrete extracellular microvesicles (EVs) that cross the blood-brain barrier. Involved in cell-to-cell communication, EVs contain surface markers and a biologically active cargo of molecules specific to their tissue (and cell) of origin, reflecting the tissue or cell's physiological state. Isolation of brain-secreted EVs (BEVs) from blood provides a minimally invasive way to sample components of brain tissue in Alzheimer's disease (AD), and is considered a form of "liquid biopsy." METHODS We performed a comprehensive review of the PubMed literature to assess the biomarker and therapeutic potential of blood-isolated BEVs in AD. RESULTS We summarize methods used for BEV isolation, validation, and novel biomarker discovery, as well as provide insights from 26 studies in humans on the biomarker potential in AD of four cell-specific BEVs isolated from blood: neuron-, neural precursor-, astrocyte-, and brain vasculature-derived BEVs. Of these, neuron-derived BEVs has been investigated on several fronts, and these include levels of amyloid-β and tau proteins, as well as synaptic proteins. In addition, we provide a synopsis of the current landscape of BEV-based evaluation/monitoring of AD therapeutics based on two published trials and a review of registered clinical trials. DISCUSSION Blood-isolated BEVs have emerged as a novel player in the study of AD, with enormous potential as a diagnostic, evaluation of therapeutics, and treatment tool. The literature has largely concentrated on neuron-derived BEVs in the blood in AD. Given the multifactorial pathophysiology of AD, additional studies, in neuron-derived and other brain cell-specific BEVs are warranted to establish BEVs as a robust blood-based biomarker of AD.
Collapse
Affiliation(s)
- AmanPreet Badhwar
- Centre de recherche de l'Institut universitaire de gériatrie de MontréalUniversity of MontrealMontrealQuebecCanada
| | - Arsalan S. Haqqani
- Human Health Therapeutics Research CentreNational Research CouncilOttawaOntarioCanada
| |
Collapse
|
140
|
Acute Insulin Resistance and Rapid Alterations in Neuronal Derived Blood Exosome Concentration After Branched Endovascular Aortic Aneurysm Repair. Eur J Vasc Endovasc Surg 2020; 59:457-463. [DOI: 10.1016/j.ejvs.2019.10.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2019] [Revised: 09/26/2019] [Accepted: 10/11/2019] [Indexed: 12/23/2022]
|
141
|
Athauda D, Gulyani S, Karnati HK, Li Y, Tweedie D, Mustapic M, Chawla S, Chowdhury K, Skene SS, Greig NH, Kapogiannis D, Foltynie T. Utility of Neuronal-Derived Exosomes to Examine Molecular Mechanisms That Affect Motor Function in Patients With Parkinson Disease: A Secondary Analysis of the Exenatide-PD Trial. JAMA Neurol 2020; 76:420-429. [PMID: 30640362 DOI: 10.1001/jamaneurol.2018.4304] [Citation(s) in RCA: 172] [Impact Index Per Article: 34.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Importance Exenatide, a glucagon-like peptide 1 agonist used in type 2 diabetes, was recently found to have beneficial effects on motor function in a randomized, placebo-controlled trial in Parkinson disease (PD). Accumulating evidence suggests that impaired brain insulin and protein kinase B (Akt) signaling play a role in PD pathogenesis; however, exploring the extent to which drugs engage with putative mechnisms in vivo remains a challenge. Objective To assess whether participants in the Exenatide-PD trial have augmented activity in brain insulin and Akt signaling pathways. Design, Setting, and Participants Serum samples were collected from 60 participants in the single-center Exenatide-PD trial (June 18, 2014, to June 16, 2016), which compared patients with moderate PD randomized to 2 mg of exenatide once weekly or placebo for 48 weeks followed by a 12-week washout period. Serum extracellular vesicles, including exosomes, were extracted, precipitated, and enriched for neuronal source by anti-L1 cell adhesion molecule antibody absorption, and proteins of interest were evaluated using electrochemiluminescence assays. Statistical analysis was performed from May 1, 2017, to August 31, 2017. Main Outcomes and Measures The main outcome was augmented brain insulin signaling that manifested as a change in tyrosine phosphorylated insulin receptor substrate 1 within neuronal extracellular vesicles at the end of 48 weeks of exenatide treatment. Additional outcome measures were changes in other insulin receptor substrate proteins and effects on protein expression in the Akt and mitogen-activated protein kinase pathways. Results Sixty patients (mean [SD] age, 59.9 [8.4] years; 43 [72%] male) participated in the study: 31 in the exenatide group and 29 in the placebo group (data from 1 patient in the exenatide group were excluded). Patients treated with exenatide had augmented tyrosine phosphorylation of insulin receptor substrate 1 at 48 weeks (0.27 absorbance units [AU]; 95% CI, 0.09-0.44 AU; P = .003) and 60 weeks (0.23 AU; 95% CI, 0.05-0.41 AU; P = .01) compared with patients receiving placebo. Exenatide-treated patients had elevated expression of downstream substrates, including total Akt (0.35 U/mL; 95% CI, 0.16-0.53 U/mL; P < .001) and phosphorylated mechanistic target of rapamycin (mTOR) (0.22 AU; 95% CI, 0.04-0.40 AU; P = .02). Improvements in Movement Disorders Society Unified Parkinson's Disease Rating Scale part 3 off-medication scores were associated with levels of total mTOR (F4,50 = 5.343, P = .001) and phosphorylated mTOR (F4,50 = 4.384, P = .04). Conclusions and Relevance The results of this study are consistent with target engagement of brain insulin, Akt, and mTOR signaling pathways by exenatide and provide a mechanistic context for the clinical findings of the Exenatide-PD trial. This study suggests the potential of using exosome-based biomarkers as objective measures of target engagement in clinical trials using drugs that target neuronal pathways.
Collapse
Affiliation(s)
- Dilan Athauda
- Department of Clinical and Movement Neurosciences, University College London Institute of Neurology, The National Hospital for Neurology and Neurosurgery, London, United Kingdom
| | - Seema Gulyani
- Laboratory of Neurosciences, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, Maryland
| | - Hanuma Kumar Karnati
- Translational Gerontology Branch, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, Maryland
| | - Yazhou Li
- Translational Gerontology Branch, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, Maryland
| | - David Tweedie
- Translational Gerontology Branch, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, Maryland
| | - Maja Mustapic
- Laboratory of Neurosciences, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, Maryland
| | - Sahil Chawla
- Laboratory of Neurosciences, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, Maryland
| | - Kashfia Chowdhury
- University College London Comprehensive Clinical Trials Unit, London, United Kingdom
| | - Simon S Skene
- University College London Comprehensive Clinical Trials Unit, London, United Kingdom.,School of Biosciences and Medicine, University of Surrey, Kent, United Kingdom
| | - Nigel H Greig
- Translational Gerontology Branch, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, Maryland
| | - Dimitrios Kapogiannis
- Laboratory of Neurosciences, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, Maryland
| | - Thomas Foltynie
- Department of Clinical and Movement Neurosciences, University College London Institute of Neurology, The National Hospital for Neurology and Neurosurgery, London, United Kingdom
| |
Collapse
|
142
|
Li Y, Wang K, Zhang P, Huang J, Liu Y, Wang Z, Lu Y, Tan S, Yang F, Tan Y. Pyrosequencing analysis of IRS1 methylation levels in schizophrenia with tardive dyskinesia. Mol Med Rep 2020; 21:1702-1708. [PMID: 32319643 PMCID: PMC7057828 DOI: 10.3892/mmr.2020.10984] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Accepted: 01/17/2020] [Indexed: 12/16/2022] Open
Abstract
Tardive dyskinesia (TD) is a serious side effect of certain antipsychotic medications that are used to treat schizophrenia (SCZ) and other mental illnesses. The methylation status of the insulin receptor substrate 1 (IRS1) gene is reportedly associated with SCZ; however, no study, to the best of the authors' knowledge, has focused on the quantitative DNA methylation levels of the IRS1 gene using pyrosequencing in SCZ with or without TD. The present study aimed to quantify DNA methylation levels of 4 CpG sites in the IRS1 gene using a Chinese sample including SCZ patients with TD and without TD (NTD) and healthy controls (HCs). The general linear model (GLM) was used to detect DNA methylation levels among the 3 proposed groups (TD vs. NTD vs. HC). Mean DNA methylation levels of 4 CpG sites demonstrated normal distribution. Pearson's correlation analysis did not reveal any significant correlations between the DNA methylation levels of the 4 CpG sites and the severity of SCZ. GLM revealed significant differences between the 3 groups for CpG site 1 and the average of the 4 CpG sites (P=0.0001 and P=0.0126, respectively). Furthermore, the TD, NTD and TD + NTD groups demonstrated lower methylation levels in CpG site 1 (P=0.0003, P<0.0001 and P<0.0001, respectively) and the average of 4 CpG sites (P=0.0176, P=0.0063 and P=0.003, respectively) compared with the HC group. The results revealed that both NTD and TD patients had significantly decreased DNA methylation levels compared with healthy controls, which indicated a significant association between the DNA methylation levels of the IRS1 gene with SCZ and TD.
Collapse
Affiliation(s)
- Yanli Li
- Beijing Huilongguan Hospital, Peking University Huilongguan Clinical Medical School, Beijing 100096, P.R. China
| | - Kesheng Wang
- Department of Family and Community Health, School of Nursing, Health Sciences Center, West Virginia University, Morgantown, WV 26506, USA
| | - Ping Zhang
- Beijing Huilongguan Hospital, Peking University Huilongguan Clinical Medical School, Beijing 100096, P.R. China
| | - Junchao Huang
- Beijing Huilongguan Hospital, Peking University Huilongguan Clinical Medical School, Beijing 100096, P.R. China
| | - Ying Liu
- Department of Biostatistics and Epidemiology, College of Public Health, East Tennessee State University, Johnson City, TN 37614, USA
| | - Zhiren Wang
- Beijing Huilongguan Hospital, Peking University Huilongguan Clinical Medical School, Beijing 100096, P.R. China
| | - Yongke Lu
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25755, USA
| | - Shuping Tan
- Beijing Huilongguan Hospital, Peking University Huilongguan Clinical Medical School, Beijing 100096, P.R. China
| | - Fude Yang
- Beijing Huilongguan Hospital, Peking University Huilongguan Clinical Medical School, Beijing 100096, P.R. China
| | - Yunlong Tan
- Beijing Huilongguan Hospital, Peking University Huilongguan Clinical Medical School, Beijing 100096, P.R. China
| |
Collapse
|
143
|
Tschuschke M, Kocherova I, Bryja A, Mozdziak P, Angelova Volponi A, Janowicz K, Sibiak R, Piotrowska-Kempisty H, Iżycki D, Bukowska D, Antosik P, Shibli JA, Dyszkiewicz-Konwińska M, Kempisty B. Inclusion Biogenesis, Methods of Isolation and Clinical Application of Human Cellular Exosomes. J Clin Med 2020; 9:jcm9020436. [PMID: 32041096 PMCID: PMC7074492 DOI: 10.3390/jcm9020436] [Citation(s) in RCA: 139] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2019] [Revised: 01/18/2020] [Accepted: 01/30/2020] [Indexed: 12/12/2022] Open
Abstract
Exosomes are a heterogenous subpopulation of extracellular vesicles 30–150 nm in range and of endosome-derived origin. We explored the exosome formation through different systems, including the endosomal sorting complex required for transport (ESCRT) and ESCRT-independent system, looking at the mechanisms of release. Different isolation techniques and specificities of exosomes from different tissues and cells are also discussed. Despite more than 30 years of research that followed their definition and indicated their important role in cellular physiology, the exosome biology is still in its infancy with rapidly growing interest. The reasons for the rapid increase in interest with respect to exosome biology is because they provide means of intercellular communication and transmission of macromolecules between cells, with a potential role in the development of diseases. Moreover, they have been investigated as prognostic biomarkers, with a potential for further development as diagnostic tools for neurodegenerative diseases and cancer. The interest grows further with the fact that exosomes were reported as useful vectors for drugs.
Collapse
Affiliation(s)
- Max Tschuschke
- Department of Anatomy, Poznan University of Medical Sciences, 60-781 Poznań, Poland; (M.T.); (I.K.); (A.B.); (K.J.); (M.D.-K.)
| | - Ievgeniia Kocherova
- Department of Anatomy, Poznan University of Medical Sciences, 60-781 Poznań, Poland; (M.T.); (I.K.); (A.B.); (K.J.); (M.D.-K.)
| | - Artur Bryja
- Department of Anatomy, Poznan University of Medical Sciences, 60-781 Poznań, Poland; (M.T.); (I.K.); (A.B.); (K.J.); (M.D.-K.)
| | - Paul Mozdziak
- Physiology Graduate Program, North Carolina State University, Raleigh, NC 27695, USA;
| | - Ana Angelova Volponi
- Centre for Craniofacial and Regenerative Biology, Faculty for Dentistry, Oral and Craniofacial Sciences, King’s College University of London, London SE1 9RT, UK;
| | - Krzysztof Janowicz
- Department of Anatomy, Poznan University of Medical Sciences, 60-781 Poznań, Poland; (M.T.); (I.K.); (A.B.); (K.J.); (M.D.-K.)
- The School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen AB25 2ZD, UK
| | - Rafał Sibiak
- Division of Reproduction, Department of Obstetrics, Gynecology, and Gynecologic Oncology, Poznan University of Medical Sciences, 60-535 Poznan, Poland;
| | | | - Dariusz Iżycki
- Department of Cancer Immunology, Poznan University of Medical Sciences, 61-866 Poznań, Poland;
| | - Dorota Bukowska
- Department of Diagnostics and Clinical Sciences, Nicolaus Copernicus University in Torun, 87-100 Toruń, Poland;
| | - Paweł Antosik
- Department of Veterinary Surgery, Nicolaus Copernicus University in Torun, 87-100 Toruń, Poland;
| | - Jamil A. Shibli
- Department of Periodontology and Oral Implantology, Dental Research Division, University of Guarulhos, Guarulhos 07030-010, Brazil;
| | - Marta Dyszkiewicz-Konwińska
- Department of Anatomy, Poznan University of Medical Sciences, 60-781 Poznań, Poland; (M.T.); (I.K.); (A.B.); (K.J.); (M.D.-K.)
- Department of Biomaterials and Experimental Dentistry, Poznan University of Medical Sciences, 61-701 Poznan, Poland
| | - Bartosz Kempisty
- Department of Anatomy, Poznan University of Medical Sciences, 60-781 Poznań, Poland; (M.T.); (I.K.); (A.B.); (K.J.); (M.D.-K.)
- Department of Histology and Embryology, Poznan University of Medical Sciences, 60-781 Poznań, Poland
- Department of Obstetrics and Gynaecology, University Hospital and Masaryk University, 601 77 Brno, Czech Republic
- Institute of Veterinary Medicine, Nicolaus Copernicus University in Toruń, 87-100 Toruń, Poland
- Correspondence: ; Tel.: +48-6185-464-18; Fax: +48-6185-464-40
| |
Collapse
|
144
|
Johnson PH, Weinreb NJ, Cloyd JC, Tuite PJ, Kartha RV. GBA1 mutations: Prospects for exosomal biomarkers in α-synuclein pathologies. Mol Genet Metab 2020; 129:35-46. [PMID: 31761523 PMCID: PMC7002237 DOI: 10.1016/j.ymgme.2019.10.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 08/03/2019] [Accepted: 10/12/2019] [Indexed: 12/13/2022]
Abstract
The discovery that patients with Gaucher Disease (GD), a rare lysosomal storage disorder, were developing symptoms similar to Parkinson's disease (PD) led to investigation of the relationship between the two seemingly unrelated pathologies. GD, an autosomal recessive disorder, is the result of a biallelic mutation in the gene GBA1, which encodes for the enzyme glucocerebrosidase (GCase). Since the observation of its relation to PD, GBA1 mutations have become recognized as the most common genetic risk factor for development of synucleinopathies such as PD and dementia with Lewy bodies. Although the exact mechanism by which GBA1 mutations promote PD is unknown, current understanding suggests that impaired GCase inhibits lysosomal activity and decreases the overall ability of the cell to degrade proteins, specifically the neuronal protein α-synuclein. Decreased elimination of α-synuclein can lead to its abnormal accumulation and aggregation, an important component of PD development. Further understanding of how decreased GCase activity increases risk for α-synuclein pathology can assist with the development of clinical biomarkers for early detection of synucleinopathies, as well as promote novel treatments tailored for people with a GBA1 mutation. Historically, α-synuclein has not been a reliable biomarker for PD. However, recent research on α-synuclein content within exosomes, which are small vesicles released by cells that carry specific cellular cargo, has yielded encouraging results. Moreover, decreased GCase activity has been shown to influence exosomal contents. Exosomes have emerged as a promising new avenue for the identification of novel biomarkers and therapeutic targets aimed at improving neuronal GCase function and limiting the development of synucleinopathies.
Collapse
Affiliation(s)
- Parker H Johnson
- Center for Orphan Drug Research, Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, MN 55455, United States of America
| | - Neal J Weinreb
- Department of Human Genetics and Medicine (Hematology), Leonard Miller School of Medicine of University of Miami, Miami, FL, United States of America
| | - James C Cloyd
- Center for Orphan Drug Research, Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, MN 55455, United States of America; Department of Neurology, University of Minnesota, Minneapolis, MN 55455, United States of America
| | - Paul J Tuite
- Department of Neurology, University of Minnesota, Minneapolis, MN 55455, United States of America
| | - Reena V Kartha
- Center for Orphan Drug Research, Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, MN 55455, United States of America.
| |
Collapse
|
145
|
Kapogiannis D, Avgerinos KI. Brain glucose and ketone utilization in brain aging and neurodegenerative diseases. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2020; 154:79-110. [PMID: 32739015 PMCID: PMC9989941 DOI: 10.1016/bs.irn.2020.03.015] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
To meet its high energy demands, the brain mostly utilizes glucose. However, the brain has evolved to exploit additional fuels, such as ketones, especially during prolonged fasting. With aging and neurodegenerative diseases (NDDs), the brain becomes inefficient at utilizing glucose due to changes in glia and neurons that involve glucose transport, glycolytic and Krebs cycle enzyme activities, and insulin signaling. Positron emission tomography and magnetic resonance spectroscopy studies have identified glucose metabolism abnormalities in aging, Alzheimer's disease (AD) and other NDDs in vivo. Despite glucose hypometabolism, brain cells can utilize ketones efficiently, thereby providing a rationale for the development of therapeutic ketogenic interventions in AD and other NDDs. This review compares available ketogenic interventions and discusses the potential of the potent oral Ketone Ester for future therapeutic use in AD and other NDDs characterized by inefficient glucose utilization.
Collapse
Affiliation(s)
- Dimitrios Kapogiannis
- Laboratory of Clinical Investigation, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, MD, United States.
| | - Konstantinos I Avgerinos
- Laboratory of Clinical Investigation, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, MD, United States
| |
Collapse
|
146
|
Cell-to-Cell Communication in Learning and Memory: From Neuro- and Glio-Transmission to Information Exchange Mediated by Extracellular Vesicles. Int J Mol Sci 2019; 21:ijms21010266. [PMID: 31906013 PMCID: PMC6982255 DOI: 10.3390/ijms21010266] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Revised: 12/14/2019] [Accepted: 12/28/2019] [Indexed: 02/06/2023] Open
Abstract
Most aspects of nervous system development and function rely on the continuous crosstalk between neurons and the variegated universe of non-neuronal cells surrounding them. The most extraordinary property of this cellular community is its ability to undergo adaptive modifications in response to environmental cues originating from inside or outside the body. Such ability, known as neuronal plasticity, allows long-lasting modifications of the strength, composition and efficacy of the connections between neurons, which constitutes the biochemical base for learning and memory. Nerve cells communicate with each other through both wiring (synaptic) and volume transmission of signals. It is by now clear that glial cells, and in particular astrocytes, also play critical roles in both modes by releasing different kinds of molecules (e.g., D-serine secreted by astrocytes). On the other hand, neurons produce factors that can regulate the activity of glial cells, including their ability to release regulatory molecules. In the last fifteen years it has been demonstrated that both neurons and glial cells release extracellular vesicles (EVs) of different kinds, both in physiologic and pathological conditions. Here we discuss the possible involvement of EVs in the events underlying learning and memory, in both physiologic and pathological conditions.
Collapse
|
147
|
Tian S, Jia W, Lu M, Zhao J, Sun X. Dual-specificity tyrosine phosphorylation-regulated kinase 1A ameliorates insulin resistance in neurons by up-regulating IRS-1 expression. J Biol Chem 2019; 294:20164-20176. [PMID: 31723029 PMCID: PMC6937568 DOI: 10.1074/jbc.ra119.010809] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Revised: 11/10/2019] [Indexed: 11/06/2022] Open
Abstract
Insulin resistance in the brain is a pathological mechanism that is shared between Alzheimer's disease (AD) and type 2 diabetes mellitus (T2DM). Although aberrant expression and phosphorylation of insulin receptor substrate 1 (IRS-1) contribute to insulin resistance, the underlying mechanism remains elusive. In this study, we used several approaches, including adeno-associated virus-based protein overexpression, immunoblotting, immunoprecipitation, immunohistochemistry, and in situ proximal ligation assays, to investigate the function of dual-specificity tyrosine phosphorylation-regulated kinase 1A (DYRK1A) in IRS-1 regulation and the downstream insulin signaling in neurons. We found that DYRK1A overexpression up-regulated IRS-1 expression by slowing turnover of the IRS-1 protein. We further observed that DYRK1A directly interacted with IRS-1 and phosphorylated IRS-1's multiple serine residues. Of note, DYRK1A and IRS-1 were coordinately up-regulated in the prefrontal cortex of db/db mice brain. Furthermore, DYRK1A overexpression ameliorated chronic high insulin-induced insulin resistance in SH-SY5Y cells as well as in primary rat neurons. These findings suggest that DYRK1A protects against insulin resistance in the brain by elevating IRS-1 expression.
Collapse
Affiliation(s)
- Shijiao Tian
- Department of Neurology, Qilu Hospital of Shandong University, No. 107 Wenhuaxi Rd., 250012 Jinan, China
- Brain Research Institute, Qilu Hospital of Shandong University, No. 107 Wenhuaxi Rd., 250012 Jinan, China
| | - Wenming Jia
- NHC Key Laboratory of Otorhinolaryngology, Chinese Ministry of Health, Qilu Hospital of Shandong University, No. 44 Wenhuaxi Rd., 250012 Jinan, China
| | - Mei Lu
- Department of Geriatrics, Qilu Hospital of Shandong University, No. 107 Wenhuaxi Rd., 250012 Jinan, China
| | - Juan Zhao
- NHC Key Laboratory of Otorhinolaryngology, Chinese Ministry of Health, Qilu Hospital of Shandong University, No. 44 Wenhuaxi Rd., 250012 Jinan, China
| | - Xiulian Sun
- Brain Research Institute, Qilu Hospital of Shandong University, No. 107 Wenhuaxi Rd., 250012 Jinan, China
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission, Qilu Hospital of Shandong University, No. 107 West Wenhua Rd., Jinan, 250012 Shandong Province, China
| |
Collapse
|
148
|
Yuan Q, Li XD, Zhang SM, Wang HW, Wang YL. Extracellular vesicles in neurodegenerative diseases: Insights and new perspectives. Genes Dis 2019; 8:124-132. [PMID: 33997159 PMCID: PMC8099685 DOI: 10.1016/j.gendis.2019.12.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2019] [Revised: 11/24/2019] [Accepted: 12/05/2019] [Indexed: 12/11/2022] Open
Abstract
Extracellular vesicles (EVs) are vesicle-like substances released by eukaryotic cells. Based on their origin and size, EVs are mainly divided into exosomes, microvesicles and apoptotic bodies, and they are secreted by eukaryotic cells under physiological and pathological conditions. EVs are enriched with nucleic acids, proteins and other factors. EVs can regulate the function of adjacent and distant cells, and they are even involved in the pathogenesis of diseases. They contain proteins associated with the pathogenesis of neurodegenerative diseases (NDs), such as the α-synuclein (α-syn) and tau proteins, which suggest potential roles for EVs as biomarkers and carriers of drugs and other therapeutic molecules that can cross the blood–brain barrier to treat NDs. In this review, we summarized the function of EVs in the pathogenesis of different NDs and related advances in EVs as diagnostic biomarkers and treatments for diseases.
Collapse
Affiliation(s)
- Qian Yuan
- Department of Neurology, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, 450014, PR China
| | - Xiao-Dong Li
- Department of Neurology, Zhengzhou Central Hospital, Zhengzhou, Henan Province, 450014, PR China
| | - Si-Miao Zhang
- Department of Neurology, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, 450014, PR China
| | - Hong-Wei Wang
- Department of Medicine, The University of Chicago, IL, 60637, USA
| | - Yun-Liang Wang
- Department of Neurology, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, 450014, PR China.,Department of Neurology, The 960th Hospital of Chinese PLA, Zibo, Shandong Province, 255300, PR China
| |
Collapse
|
149
|
Cha DJ, Mengel D, Mustapic M, Liu W, Selkoe DJ, Kapogiannis D, Galasko D, Rissman RA, Bennett DA, Walsh DM. miR-212 and miR-132 Are Downregulated in Neurally Derived Plasma Exosomes of Alzheimer's Patients. Front Neurosci 2019; 13:1208. [PMID: 31849573 PMCID: PMC6902042 DOI: 10.3389/fnins.2019.01208] [Citation(s) in RCA: 142] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Accepted: 10/25/2019] [Indexed: 12/14/2022] Open
Abstract
It was recently discovered that brain cells release extracellular vesicles (EV) which can pass from brain into blood. These findings raise the possibility that brain-derived EV’s present in blood can be used to monitor disease processes occurring in the cerebrum. Since the levels of certain micro-RNAs (miRNAs) have been reported to be altered in Alzheimer’s disease (AD) brain, we sought to assess miRNA dysregulation in AD brain tissue and to determine if these changes were reflected in neural EVs isolated from blood of subjects with AD. To this end, we employed high-content miRNA arrays to search for differences in miRNAs in RNA pools from brain tissue of AD (n = 5), high pathological control (HPC) (n = 5), or cognitively intact pathology-free controls (n = 5). Twelve miRNAs were altered by >1.5-fold in AD compared to controls, and six of these were also changed compared to HPCs. Analysis of hits in brain extracts from 11 AD, 7 HPCs and 9 controls revealed a similar fold difference in these six miRNAs, with three showing statistically significant group differences and one with a strong trend toward group differences. Thereafter, we focused on the four miRNAs that showed group differences and measured their content in neurally derived blood EVs isolated from 63 subjects: 16 patients with early stage dementia and a CSF Aβ42+ tau profile consistent with AD, 16 individuals with mild cognitive impairment (MCI) and an AD CSF profile, and 31 cognitively intact controls with normal CSF Aβ42+ tau levels. ROC analysis indicated that measurement of miR-132-3p in neurally-derived plasma EVs showed good sensitivity and specificity to diagnose AD, but did not effectively separate individuals with AD-MCI from controls. Moreover, when we measured the levels of a related miRNA, miR-212, we found that this miRNA was also decreased in neural EVs from AD patients compared to controls. Our results suggest that measurement of miR-132 and miR-212 in neural EVs should be further investigated as a diagnostic aid for AD and as a potential theragnostic.
Collapse
Affiliation(s)
- Diana J Cha
- Laboratory for Neurodegenerative Disease Research, Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| | - David Mengel
- Laboratory for Neurodegenerative Disease Research, Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States.,Department of Neurodegenerative Diseases, Hertie-Institute for Clinical Brain Research and Center of Neurology, University of Tübingen, Tübingen, Germany.,German Center for Neurodegenerative Diseases, University of Tübingen, Tübingen, Germany
| | - Maja Mustapic
- Laboratory of Neurosciences, National Institute on Aging, National Institutes of Health, Baltimore, MD, United States
| | - Wen Liu
- Laboratory for Neurodegenerative Disease Research, Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| | - Dennis J Selkoe
- Laboratory for Neurodegenerative Disease Research, Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| | - Dimitrios Kapogiannis
- Laboratory of Neurosciences, National Institute on Aging, National Institutes of Health, Baltimore, MD, United States
| | - Douglas Galasko
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, United States
| | - Robert A Rissman
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, United States.,VA San Diego Healthcare System, La Jolla, CA, United States
| | - David A Bennett
- Rush Alzheimer's Disease Center, Rush Medical College, Chicago, IL, United States
| | - Dominic M Walsh
- Laboratory for Neurodegenerative Disease Research, Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States.,Alzheimer's Disease and Dementia Research Unit, Biogen Inc., Cambridge, MA, United States
| |
Collapse
|
150
|
Winston CN, Goetzl EJ, Baker LD, Vitiello MV, Rissman RA. Growth Hormone-Releasing Hormone Modulation of Neuronal Exosome Biomarkers in Mild Cognitive Impairment. J Alzheimers Dis 2019; 66:971-981. [PMID: 30372675 DOI: 10.3233/jad-180302] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Age-related changes in cognition are linked to decreased expression of somatotropins, GHRH and IGF-1. Mild cognitive impairment (MCI) and Alzheimer's disease (AD) are heterogeneous conditions. The loss of GHRH signaling in the brain may be mechanistically involved in AD pathogenesis. The consequent need to identify AD at an early and perhaps more treatable stage has fueled research into blood-based, exosome biomarkers. Plasma exosomes from participants enrolled in a randomized, double-blind, placebo-controlled 20-week trial of GHRH administration, were isolated, precipitated, and enriched by immuno-absorption with anti-L1CAM antibody (neural adhesion protein) from adults with MCI and age-matched, cognitively normal controls (CNC). Extracted protein cargo from neuronally-derived exosomes (NDEs) were assessed by ELISAs for protein levels implicated in AD neuropathology and for synaptic proteins altered by AD. Plasma NDE concentrations of Aβ1-42 were significantly increased while plasma NDE concentrations of NRGN, synaptophysin, synaptotagmin, and synaptopodin were significantly decreased in patients with MCI, independent of GHRH treatment. Plasma NDE concentrations of ptau-S396 and GAP43 were not affected by cognitive status (CNC versus MCI) or by GHRH treatment. Aβ1-42, neurogranin (NRGN), synaptophysin, synaptotagmin, and synaptopodin demonstrated the highest diagnostic accuracy for distinguishing between CNC and MCI patients, while synaptophysin and synaptotagmin demonstrated moderate accuracy in distinguishing between placebo-treated and GHRH-treated, MCI patients.
Collapse
|