1
|
Ustianowski Ł, Udzik J, Szostak J, Gorący A, Ustianowska K, Pawlik A. Genetic and Epigenetic Factors in Gestational Diabetes Mellitus Pathology. Int J Mol Sci 2023; 24:16619. [PMID: 38068941 PMCID: PMC10706782 DOI: 10.3390/ijms242316619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 11/14/2023] [Accepted: 11/20/2023] [Indexed: 12/18/2023] Open
Abstract
Gestational diabetes (GDM) is the carbohydrate intolerance occurring during pregnancy. The risk factors of GDM include obesity, advanced maternal age, polycystic ovary syndrome, multigravidity, a sedentary lifestyle, and pre-existing hypertension. Additionally, complex genetic and epigenetic processes are also believed to play a crucial role in the development of GDM. In this narrative review, we discuss the role of genetic and epigenetic factors in gestational diabetes mellitus pathogenesis.
Collapse
Affiliation(s)
- Łukasz Ustianowski
- Department of Physiology, Pomeranian Medical University, 70-111 Szczecin, Poland; (Ł.U.); (J.U.); (K.U.)
| | - Jakub Udzik
- Department of Physiology, Pomeranian Medical University, 70-111 Szczecin, Poland; (Ł.U.); (J.U.); (K.U.)
- Department of Cardiac Surgery, Pomeranian Medical University, 70-111 Szczecin, Poland
| | - Joanna Szostak
- Department of Experimental and Clinical Pharmacology, Pomeranian Medical University, 70-111 Szczecin, Poland;
| | - Anna Gorący
- Department of Clinical and Molecular Biochemistry, Pomeranian Medical University, 70-111 Szczecin, Poland;
| | - Klaudia Ustianowska
- Department of Physiology, Pomeranian Medical University, 70-111 Szczecin, Poland; (Ł.U.); (J.U.); (K.U.)
| | - Andrzej Pawlik
- Department of Physiology, Pomeranian Medical University, 70-111 Szczecin, Poland; (Ł.U.); (J.U.); (K.U.)
| |
Collapse
|
2
|
Rani S, Dhar SB, Khajuria A, Gupta D, Jaiswal PK, Singla N, Kaur M, Singh G, Barnwal RP. Advanced Overview of Biomarkers and Techniques for Early Diagnosis of Alzheimer's Disease. Cell Mol Neurobiol 2023; 43:2491-2523. [PMID: 36847930 PMCID: PMC11410160 DOI: 10.1007/s10571-023-01330-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 02/15/2023] [Indexed: 03/01/2023]
Abstract
The development of early non-invasive diagnosis methods and identification of novel biomarkers are necessary for managing Alzheimer's disease (AD) and facilitating effective prognosis and treatment. AD has multi-factorial nature and involves complex molecular mechanism, which causes neuronal degeneration. The primary challenges in early AD detection include patient heterogeneity and lack of precise diagnosis at the preclinical stage. Several cerebrospinal fluid (CSF) and blood biomarkers have been proposed to show excellent diagnosis ability by identifying tau pathology and cerebral amyloid beta (Aβ) for AD. Intense research endeavors are being made to develop ultrasensitive detection techniques and find potent biomarkers for early AD diagnosis. To mitigate AD worldwide, understanding various CSF biomarkers, blood biomarkers, and techniques that can be used for early diagnosis is imperative. This review attempts to provide information regarding AD pathophysiology, genetic and non-genetic factors associated with AD, several potential blood and CSF biomarkers, like neurofilament light, neurogranin, Aβ, and tau, along with biomarkers under development for AD detection. Besides, numerous techniques, such as neuroimaging, spectroscopic techniques, biosensors, and neuroproteomics, which are being explored to aid early AD detection, have been discussed. The insights thus gained would help in finding potential biomarkers and suitable techniques for the accurate diagnosis of early AD before cognitive dysfunction.
Collapse
Affiliation(s)
- Shital Rani
- Department of Biophysics, Panjab University, Chandigarh, 160014, India
| | - Sudhrita Basu Dhar
- University Institute of Pharmaceutical Sciences, Panjab University, Chandigarh, 160014, India
| | - Akhil Khajuria
- University Institute of Pharmaceutical Sciences, Panjab University, Chandigarh, 160014, India
| | - Dikshi Gupta
- JoyScore Inc., 2440 Cerritos Ave, Signal Hill, CA, 90755, USA
| | - Pradeep Kumar Jaiswal
- Department of Biochemistry and Biophysics, Texas A & M University, College Station, TX, 77843, USA
| | - Neha Singla
- Department of Biophysics, Panjab University, Chandigarh, 160014, India
| | - Mandeep Kaur
- Department of Biophysics, Panjab University, Chandigarh, 160014, India.
| | - Gurpal Singh
- University Institute of Pharmaceutical Sciences, Panjab University, Chandigarh, 160014, India.
| | | |
Collapse
|
3
|
Pestereva N, Ivleva I, Zubov A, Tikhomirova M, Karpenko M. m-Calpain is released from striatal synaptosomes. Int J Neurosci 2023; 133:215-221. [PMID: 33688783 DOI: 10.1080/00207454.2021.1901697] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Purpose of the study: We aimed to investigate whether m-calpain (a Ca2+-dependent neutral cysteine protease) is released from synaptosomes.Materials and methods: This research was carry on Wistar male rats and isolated nerve endings - synaptosomes. The synaptosomal integrity was checked by the method of measuring LDH activity. Activity of calpains was measured by the casein zymography in gel and in solution. Extracellular calpain was detected by immunoprecipitation and immunoblotting procedures Prediction of secreted proteins peptide on a protein sequence through a local version of the PrediSi tool (http://www.predisi.de). The probability of calpain isoform nonclassical secretion was analyzed by using SecretomeP (http://www.cbs.dtu.dk/services/SecretomeP2.0) software.Results: It has been shown that calcium- and time-dependent m-calpain is released from synaptosomes in an activated form or in a form capable of activation, and this process is not a result of a violation of the integrity of synaptosomes. Analysis of the probability of secretion of the small catalytic subunit of rat m-calpain along a nonclassical pathway showed a high probability of its secretion. Additionally, the release of calpain from synaptosomes revealed by us is suppressed by the addition of glyburide, an ABC transporter inhibitor, to the incubation medium. Among extracellular proteins, potential substrates of calpains are of calpains are found, for example, matrix metalloprotease-2 and -9, alpha-synuclein, etc.Conclusions: Active m-calpain is present in the media generated from striatal synaptosomes. Glyburide prevents m-calpain release from striatal synaptosomes.
Collapse
Affiliation(s)
- Nina Pestereva
- Department of Physiology (Pavlov's), Institute of Experimental Medicine, St. Petersburg, Russia
| | - Irina Ivleva
- Department of Physiology (Pavlov's), Institute of Experimental Medicine, St. Petersburg, Russia
| | - Alexander Zubov
- Department of Physiology (Pavlov's), Institute of Experimental Medicine, St. Petersburg, Russia
| | - Maria Tikhomirova
- Institute of Molecular Life Sciences, University of Zurich, Zurich, Switzerland
| | - Marina Karpenko
- Department of Physiology (Pavlov's), Institute of Experimental Medicine, St. Petersburg, Russia.,Institute of Physics, Nanotechnology and Telecommunications, Peter the Great St. Petersburg Polytechnic University, St. Petersburg, Russia
| |
Collapse
|
4
|
Increment of CSF fractalkine-positive microvesicles preceded the spatial memory impairment in amyloid beta neurotoxicity. Cytokine 2022; 160:156050. [PMID: 36179535 DOI: 10.1016/j.cyto.2022.156050] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 09/03/2022] [Accepted: 09/09/2022] [Indexed: 11/23/2022]
Abstract
BACKGROUND Fractalkine (CX3CL1) is a key chemokine, affects neuronal cell communication and involves in Alzheimer's disease pathogenesis. Microvesicles (MVs) participate in neuronal cells' cross-talk in physiological and pathological states. Microvesicles released in cerebrospinal fluid (CSF) may provide a valuable footprint of brain changes. Little information is available regarding the release of fractalkine-positive MVs (CX3CL1+ -MVs) in the nervous system. METHODS We induced cognitive impairment by bilateral injection of amyloid-beta (Aβ) into the cerebral ventricles. We analyzed the CSF by flow cytometry in two experiments (trained and untrained) to elucidate the presence of CX3CL1+ -MVs. The hippocampal TNF-α as an inflammatory factor was assessed by immunohistochemistry. RESULTS The Aβ induced spatial memory impairment after two weeks, verified by a decrease in the escape latency in Morris water maze test. It caused an increase in the anxiety-like behaviors demonstrated by a decrease in entries into the open arms of elevated plus maze test. The Aβ increased the percent of the positive area for TNF-α staining. Histological evaluation of the hippocampus confirmed the tissue injuries. The CSF levels of CX3CL1+ -MVs, increased 2 and 7 days after Aβ injection. The Aβ increased the TNF-α staining and provided an inflammatory context to facilitate the MVs release. The rise of CX3CL1+ -MVs was transient and subsided after two weeks. Both trained and untrained experiments showed a similar rise pattern of CX3CL1+ -MVs. CONCLUSION Increase of fractalkine-positive microvesicles preceded the cognitive impairment, more studies are required to approve the CX3CL1+ -MVs as a potential biomarker in the early diagnosis of Alzheimer's disease.
Collapse
|
5
|
Yalameha B, Nejabati HR, Nouri M. Circulating microparticles as indicators of cardiometabolic risk in PCOS. Clin Chim Acta 2022; 533:63-70. [PMID: 35718107 DOI: 10.1016/j.cca.2022.06.019] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 06/14/2022] [Accepted: 06/15/2022] [Indexed: 11/17/2022]
Abstract
Polycystic ovary syndrome (PCOS), the most prevalent endocrine disturbance of the female reproductive system, is associated with several pathologic conditions, such as metabolic syndrome, obesity, diabetes, dyslipidemia, and insulin resistance, all of which are tightly connected to its progression. These factors are associated with a type of extracellular vesicle, ie, microparticles (MPs), released by shedding due to cell activation and apoptosis. Circulating MPs (cMPs) are secreted by a variety of cells, such as platelets, endothelial, leukocytes, and erythrocytes, and contain cytoplasmic substances derived from parent cells that account for their biologic activity. Current evidence has clearly shown that increased cMPs contribute to endothelial dysfunction, diabetes, hypertriglyceridemia, metabolic syndrome, cardiovascular abnormalities as well as PCOS. It has also been reported that platelet and endothelial MPs are specifically increased in PCOS thus endangering vascular health and subsequent cardiovascular disease. Given the importance of cMPs in the pathophysiology of PCOS, we review the role of cMPs in PCOS with a special focus on cardiometabolic significance.
Collapse
Affiliation(s)
- Banafsheh Yalameha
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hamid Reza Nejabati
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Mohammad Nouri
- Department of Reproductive Biology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
6
|
Moretto E, Stuart S, Surana S, Vargas JNS, Schiavo G. The Role of Extracellular Matrix Components in the Spreading of Pathological Protein Aggregates. Front Cell Neurosci 2022; 16:844211. [PMID: 35573838 PMCID: PMC9100790 DOI: 10.3389/fncel.2022.844211] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Accepted: 03/08/2022] [Indexed: 11/23/2022] Open
Abstract
Several neurodegenerative diseases are characterized by the accumulation of aggregated misfolded proteins. These pathological agents have been suggested to propagate in the brain via mechanisms similar to that observed for the prion protein, where a misfolded variant is transferred from an affected brain region to a healthy one, thereby inducing the misfolding and/or aggregation of correctly folded copies. This process has been characterized for several proteins, such as α-synuclein, tau, amyloid beta (Aβ) and less extensively for huntingtin and TDP-43. α-synuclein, tau, TDP-43 and huntingtin are intracellular proteins, and their aggregates are located in the cytosol or nucleus of neurons. They have been shown to spread between cells and this event occurs, at least partially, via secretion of these protein aggregates in the extracellular space followed by re-uptake. Conversely, Aβ aggregates are found mainly extracellularly, and their spreading occurs in the extracellular space between brain regions. Due to the inherent nature of their spreading modalities, these proteins are exposed to components of the extracellular matrix (ECM), including glycans, proteases and core matrix proteins. These ECM components can interact with or process pathological misfolded proteins, potentially changing their properties and thus regulating their spreading capabilities. Here, we present an overview of the documented roles of ECM components in the spreading of pathological protein aggregates in neurodegenerative diseases with the objective of identifying the current gaps in knowledge and stimulating further research in the field. This could potentially lead to the identification of druggable targets to slow down the spreading and/or progression of these pathologies.
Collapse
Affiliation(s)
- Edoardo Moretto
- Institute of Neuroscience, National Research Council, CNR, Milan, Italy
- UK Dementia Research Institute, University College London, London, United Kingdom
- Department of Neuromuscular Diseases, Queen Square Institute of Neurology, University College London, London, United Kingdom
- *Correspondence: Edoardo Moretto,
| | - Skye Stuart
- UK Dementia Research Institute, University College London, London, United Kingdom
- Department of Neuromuscular Diseases, Queen Square Institute of Neurology, University College London, London, United Kingdom
| | - Sunaina Surana
- UK Dementia Research Institute, University College London, London, United Kingdom
- Department of Neuromuscular Diseases, Queen Square Institute of Neurology, University College London, London, United Kingdom
- UCL Queen Square Motor Neuron Disease Centre, University College London, London, United Kingdom
| | - Jose Norberto S. Vargas
- UK Dementia Research Institute, University College London, London, United Kingdom
- Department of Neuromuscular Diseases, Queen Square Institute of Neurology, University College London, London, United Kingdom
- UCL Queen Square Motor Neuron Disease Centre, University College London, London, United Kingdom
| | - Giampietro Schiavo
- UK Dementia Research Institute, University College London, London, United Kingdom
- Department of Neuromuscular Diseases, Queen Square Institute of Neurology, University College London, London, United Kingdom
- UCL Queen Square Motor Neuron Disease Centre, University College London, London, United Kingdom
- Giampietro Schiavo,
| |
Collapse
|
7
|
The Reduction of Tau Hyperphosphorylation by Cornel Iridoid Glycosides Is Mediated by Their Influence on Calpain Activity. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:9213046. [PMID: 35096120 PMCID: PMC8794656 DOI: 10.1155/2022/9213046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 11/11/2021] [Accepted: 12/08/2021] [Indexed: 11/18/2022]
Abstract
Alzheimer’s disease (AD) is the most common type of dementia, and the abnormal hyperphosphorylation of the tau protein is the main component of its pathogenesis. Calpain was found to be abnormally activated in neurofibrillary tangles (NFTs) in a previous report. Cornel iridoid glycosides (CIG) have been reported to reduce the hyperphosphorylation of tau protein. Nevertheless, the role of calpain in the reduction tau hyperphosphorylation by CIG remains unclear. In the present study, we investigated the effect of CIG on calpain activity through in vitro and in vivo experiments. Western blotting results suggested that CIG decreased the phosphorylation of tau at Ser 404 and Ser 262 sites in P301S mice. Moreover, CIG inhibited the activity of calpain and glycogen synthase kinase 3β (GSK-3β) and enhanced the activity of protein phosphatase 2A (PP2A) both in vivo and in vitro. CIG also inhibited the activation of PP2A and reduced the GSK-3β activity caused by the calpain activator dibucaine. In addition, the main components of CIG, morroniside and loganin, play an equivalent role in reducing calpain activity, as the effect of their combined use is equivalent to that of CIG. The abovementioned findings revealed that CIG improved PP2A activity and reduced GSK-3β activity by adjusting the activity of calpain 1, leading to a reduction in the phosphorylation of tau. This study highlights the remarkable therapeutic potential of CIG for managing AD.
Collapse
|
8
|
Targeted proteolytic products of τ and α-synuclein in neurodegeneration. Essays Biochem 2021; 65:905-912. [PMID: 34846537 PMCID: PMC8709889 DOI: 10.1042/ebc20210028] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 10/05/2021] [Accepted: 10/14/2021] [Indexed: 11/17/2022]
Abstract
CNS pathological inclusions comprising τ or α-synuclein (αSyn) define a spectrum of neurodegenerative diseases, and these can often present concurrently in the same individuals. The aggregation of both proteins is clearly associated with neurodegeneration and the deleterious properties of each protein is further supported by mutations in each gene (MAPT and SNCA, respectively) resulting in disease. The initiating events in most sporadic neurodegenerative diseases are still unclear but growing evidence suggests that the aberrant proteolytic cleavage of τ and αSyn results in products that can be toxic and/or initiate aggregation that can further spread by a prion-like mechanism. The accumulation of some of these cleavage products can further potentiate the progression of protein aggregation transmission and lead to their accumulation in peripheral biofluids such as cerebrospinal fluid (CSF) and blood. The future development of new tools to detect specific τ and αSyn abnormal cleavage products in peripheral biofluids could be useful biomarkers and better understand of the role of unique proteolytic activities could yield therapeutic interventions.
Collapse
|
9
|
Page MJ, Pretorius E. Platelet Behavior Contributes to Neuropathologies: A Focus on Alzheimer's and Parkinson's Disease. Semin Thromb Hemost 2021; 48:382-404. [PMID: 34624913 DOI: 10.1055/s-0041-1733960] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The functions of platelets are broad. Platelets function in hemostasis and thrombosis, inflammation and immune responses, vascular regulation, and host defense against invading pathogens, among others. These actions are achieved through the release of a wide set of coagulative, vascular, inflammatory, and other factors as well as diverse cell surface receptors involved in the same activities. As active participants in these physiological processes, platelets become involved in signaling pathways and pathological reactions that contribute to diseases that are defined by inflammation (including by pathogen-derived stimuli), vascular dysfunction, and coagulation. These diseases include Alzheimer's and Parkinson's disease, the two most common neurodegenerative diseases. Despite their unique pathological and clinical features, significant shared pathological processes exist between these two conditions, particularly relating to a central inflammatory mechanism involving both neuroinflammation and inflammation in the systemic environment, but also neurovascular dysfunction and coagulopathy, processes which also share initiation factors and receptors. This triad of dysfunction-(neuro)inflammation, neurovascular dysfunction, and hypercoagulation-illustrates the important roles platelets play in neuropathology. Although some mechanisms are understudied in Alzheimer's and Parkinson's disease, a strong case can be made for the relevance of platelets in neurodegeneration-related processes.
Collapse
Affiliation(s)
- Martin J Page
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Stellenbosch, Private Bag X1 Matieland, South Africa
| | - Etheresia Pretorius
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Stellenbosch, Private Bag X1 Matieland, South Africa
| |
Collapse
|
10
|
González-Molina LA, Villar-Vesga J, Henao-Restrepo J, Villegas A, Lopera F, Cardona-Gómez GP, Posada-Duque R. Extracellular Vesicles From 3xTg-AD Mouse and Alzheimer's Disease Patient Astrocytes Impair Neuroglial and Vascular Components. Front Aging Neurosci 2021; 13:593927. [PMID: 33679370 PMCID: PMC7933224 DOI: 10.3389/fnagi.2021.593927] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Accepted: 01/21/2021] [Indexed: 12/19/2022] Open
Abstract
Astrocytes are specialized glial cells that are essential components of the neurovascular unit (NVU) and are involved in neurodevelopment, brain maintenance and repair, and neurodegeneration. Astrocytes mediate these processes by releasing cellular mediators such as extracellular vesicles (EVs). EVs are vehicles of cell-cell communication and have been proposed as mediators of damage in AD. However, the transcellular mechanism by which Alzheimer disease (AD) astrocytes impair the function of NVU components is poorly understood. Therefore, we evaluated the effects of adult PS1-KI and 3xTg-AD astrocyte conditioned media (CM) and EVs on NVU components (neuroglia and endothelium) in vitro. Additionally, SAD and FAD astrocyte-derived EVs (A-EVs) were characterized, and we evaluated their effects on NVU in cocultured cells in vitro and on intrahippocampal CA1 cells in vivo. Surprisingly, cultured 3xTg-AD astrocytes showed increased glial fibrillary acidic protein (GFAP) reactivity compared to PS1-KI astrocytes, which denotes astrocytic hyperreactivity. CM from adult mice 3xTg-AD astrocytes increased cell-cell gaps between endothelial cells, filopodia-like dendritic protrusions in neurons and neuronal and endothelial cell death. 3xTg-AD A-EVs induced neurotoxicity and increased astrocyte GFAP reactivity. Cultured human postmortem astrocytes from AD patients also increased GFAP reactivity and EVs release. No differences in the size or number of A-EVs were detected between AD and control samples; however, both SAD and FAD A-EVs showed increased expression of the surface marker aquaporin 4. A-EVs induced cytotoxicity and astrocyte hyperactivation: specifically, FAD A-EVs induced neuroglial cytotoxicity and increased gaps between the endothelium, while SAD A-EVs mainly altered the endothelium. Similarly, both AD A-EVs increased astrocyte GS reactivity and vascular deterioration in vivo. We associated this finding with perivascular reactive astrocytes and vascular deterioration in the human AD brain. In summary, these results suggest that AD A-EVs impair neuroglial and vascular components.
Collapse
Affiliation(s)
- Luis Alfonso González-Molina
- Área de Neurobiología Celular y Molecular, Grupo de Neurociencias de Antioquia, Universidad de Antioquia, Medellin, Colombia.,Facultad de Ciencias Exactas y Naturales, Instituto de Biología, Universidad de Antioquia, Medellin, Colombia
| | - Juan Villar-Vesga
- Área de Neurobiología Celular y Molecular, Grupo de Neurociencias de Antioquia, Universidad de Antioquia, Medellin, Colombia.,Facultad de Ciencias Exactas y Naturales, Instituto de Biología, Universidad de Antioquia, Medellin, Colombia
| | - Julián Henao-Restrepo
- Área de Neurobiología Celular y Molecular, Grupo de Neurociencias de Antioquia, Universidad de Antioquia, Medellin, Colombia.,Facultad de Ciencias Exactas y Naturales, Instituto de Biología, Universidad de Antioquia, Medellin, Colombia
| | - Andrés Villegas
- Neurobank, Neuroscience Group of Antioquia, Faculty of Medicine, SIU, University of Antioquia, Medellin, Colombia
| | - Francisco Lopera
- Neurobank, Neuroscience Group of Antioquia, Faculty of Medicine, SIU, University of Antioquia, Medellin, Colombia
| | - Gloria Patricia Cardona-Gómez
- Área de Neurobiología Celular y Molecular, Grupo de Neurociencias de Antioquia, Universidad de Antioquia, Medellin, Colombia
| | - Rafael Posada-Duque
- Área de Neurobiología Celular y Molecular, Grupo de Neurociencias de Antioquia, Universidad de Antioquia, Medellin, Colombia.,Facultad de Ciencias Exactas y Naturales, Instituto de Biología, Universidad de Antioquia, Medellin, Colombia
| |
Collapse
|
11
|
Muruzheva ZM, Traktirov DS, Zubov AS, Pestereva NS, Tikhomirova MS, Karpenko MN. Calpain activity in plasma of patients with essential tremor and Parkinson's disease: a pilot study. Neurol Res 2020; 43:314-320. [PMID: 33729106 DOI: 10.1080/01616412.2020.1854004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Background: Essential tremor (ET) and Parkinson's disease (PD) are the two most common movement disorders in adults with similar clinical symptoms, which is hinting towards existence of coincident pathogenesis steps.Objectives: The objective of this report is to characterize the relationship between ET and PD severity and the activity of calcium-dependent proteases calpain in plasma.Methods: The study enrolled 12 volunteers for each condition: ET, PD, healthy. We evaluated the stage of PD on the H&Y scale in patients with PD, and the severity of tremor in patients with ET on the FTMS scale. IL-1β, TNFα, IL6, IL10 were determined in plasma using ELISA. Calpain activity was measured using fluorescent substrate and zymography methods.Results: We demonstrated that the activity of calpains in plasma of patients with PD and ET increased 5.1 and 4.3 times, respectively. The increase of calpain activity in plasma of PD patients correlated with the content of IL-1β, for ET such a connection was not found. At the advanced stages of PD calpain activity in plasma was significantly higher than that of the PD group at the early stage, and this increase was mediated by the increase in m-calpain activity. The increase in the tremor severity in ET did not lead to an increase in the activity of calpains in plasma.Conclusions: We observed general increase in the activity of calpains in plasma of both PD and ET patients that hints towards presence of the common steps in the pathogenesis of these diseases.
Collapse
Affiliation(s)
- Zamira M Muruzheva
- I.P. Pavlov Department of Physiology, Institute of Experimental Medicine, Saint Petersburg, Russia
| | - Dmitry S Traktirov
- I.P. Pavlov Department of Physiology, Institute of Experimental Medicine, Saint Petersburg, Russia
| | - Alexander S Zubov
- I.P. Pavlov Department of Physiology, Institute of Experimental Medicine, Saint Petersburg, Russia
| | - Nina S Pestereva
- I.P. Pavlov Department of Physiology, Institute of Experimental Medicine, Saint Petersburg, Russia
| | - Maria S Tikhomirova
- I.P. Pavlov Department of Physiology, Institute of Experimental Medicine, Saint Petersburg, Russia
| | - Marina N Karpenko
- I.P. Pavlov Department of Physiology, Institute of Experimental Medicine, Saint Petersburg, Russia
| |
Collapse
|
12
|
Villar-Vesga J, Henao-Restrepo J, Voshart DC, Aguillon D, Villegas A, Castaño D, Arias-Londoño JD, Zuhorn IS, Ribovski L, Barazzuol L, Cardona-Gómez GP, Posada-Duque R. Differential Profile of Systemic Extracellular Vesicles From Sporadic and Familial Alzheimer's Disease Leads to Neuroglial and Endothelial Cell Degeneration. Front Aging Neurosci 2020; 12:587989. [PMID: 33281599 PMCID: PMC7705379 DOI: 10.3389/fnagi.2020.587989] [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: 07/27/2020] [Accepted: 10/09/2020] [Indexed: 01/01/2023] Open
Abstract
Evidence suggests that extracellular vesicles (EVs) act as mediators and biomarkers of neurodegenerative diseases. Two distinct forms of Alzheimer disease (AD) are known: a late-onset sporadic form (SAD) and an early-onset familial form (FAD). Recently, neurovascular dysfunction and altered systemic immunological components have been linked to AD neurodegeneration. Therefore, we characterized systemic-EVs from postmortem SAD and FAD patients and evaluated their effects on neuroglial and endothelial cells. We found increase CLN-5 spots with vesicular morphology in the abluminal portion of vessels from SAD patients. Both forms of AD were associated with larger and more numerous systemic EVs. Specifically, SAD patients showed an increase in endothelial- and leukocyte-derived EVs containing mitochondria; in contrast, FAD patients showed an increase in platelet-derived EVs. We detected a differential protein composition for SAD- and FAD-EVs associated with the coagulation cascade, inflammation, and lipid-carbohydrate metabolism. Using mono- and cocultures (endothelium-astrocytes-neurons) and human cortical organoids, we showed that AD-EVs induced cytotoxicity. Both forms of AD featured decreased neuronal branches area and astrocytic hyperreactivity, but SAD-EVs led to greater endothelial detrimental effects than FAD-EVs. In addition, FAD- and SAD-EVs affected calcium dynamics in a cortical organoid model. Our findings indicate that the phenotype of systemic AD-EVs is differentially defined by the etiopathology of the disease (SAD or FAD), which results in a differential alteration of the NVU cells implied in neurodegeneration.
Collapse
Affiliation(s)
- Juan Villar-Vesga
- Neuroscience Group of Antioquia, Cellular and Molecular Neurobiology Area, Faculty of Medicine, Sede de Investigación Universitaria, University of Antioquia, Medellín, Colombia.,Institute of Biology, Faculty of Exact and Natural Sciences, University of Antioquia, Medellín, Colombia
| | - Julián Henao-Restrepo
- Neuroscience Group of Antioquia, Cellular and Molecular Neurobiology Area, Faculty of Medicine, Sede de Investigación Universitaria, University of Antioquia, Medellín, Colombia.,Institute of Biology, Faculty of Exact and Natural Sciences, University of Antioquia, Medellín, Colombia
| | - Daniëlle C Voshart
- Department of Radiation Oncology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands.,Section of Molecular Cell Biology, Department of Biomedical Sciences of Cells and Systems, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - David Aguillon
- Neurobank, Neuroscience Group of Antioquia, Faculty of Medicine, Sede de Investigación Universitaria, University of Antioquia, Medellín, Colombia
| | - Andrés Villegas
- Neurobank, Neuroscience Group of Antioquia, Faculty of Medicine, Sede de Investigación Universitaria, University of Antioquia, Medellín, Colombia
| | - Diana Castaño
- Grupo de Inmunología Celular e Inmunogenética, Instituto de Investigaciones Médicas, Facultad de Medicina, Universidad de Antioquia, Medellín, Colombia
| | | | - Inge S Zuhorn
- Department of Biomedical Engineering, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Laís Ribovski
- Department of Biomedical Engineering, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Lara Barazzuol
- Department of Radiation Oncology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands.,Section of Molecular Cell Biology, Department of Biomedical Sciences of Cells and Systems, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Gloria P Cardona-Gómez
- Neuroscience Group of Antioquia, Cellular and Molecular Neurobiology Area, Faculty of Medicine, Sede de Investigación Universitaria, University of Antioquia, Medellín, Colombia
| | - Rafael Posada-Duque
- Neuroscience Group of Antioquia, Cellular and Molecular Neurobiology Area, Faculty of Medicine, Sede de Investigación Universitaria, University of Antioquia, Medellín, Colombia.,Institute of Biology, Faculty of Exact and Natural Sciences, University of Antioquia, Medellín, Colombia
| |
Collapse
|
13
|
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
|
14
|
Jensen LR, Garrett L, Hölter SM, Rathkolb B, Rácz I, Adler T, Prehn C, Hans W, Rozman J, Becker L, Aguilar-Pimentel JA, Puk O, Moreth K, Dopatka M, Walther DJ, von Bohlen und Halbach V, Rath M, Delatycki M, Bert B, Fink H, Blümlein K, Ralser M, Van Dijck A, Kooy F, Stark Z, Müller S, Scherthan H, Gecz J, Wurst W, Wolf E, Zimmer A, Klingenspor M, Graw J, Klopstock T, Busch D, Adamski J, Fuchs H, Gailus-Durner V, de Angelis MH, von Bohlen und Halbach O, Ropers HH, Kuss AW. A mouse model for intellectual disability caused by mutations in the X-linked 2′‑O‑methyltransferase Ftsj1 gene. Biochim Biophys Acta Mol Basis Dis 2019; 1865:2083-2093. [DOI: 10.1016/j.bbadis.2018.12.011] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Revised: 12/06/2018] [Accepted: 12/10/2018] [Indexed: 01/13/2023]
|
15
|
Ahmad F, Das D, Kommaddi RP, Diwakar L, Gowaikar R, Rupanagudi KV, Bennett DA, Ravindranath V. Isoform-specific hyperactivation of calpain-2 occurs presymptomatically at the synapse in Alzheimer's disease mice and correlates with memory deficits in human subjects. Sci Rep 2018; 8:13119. [PMID: 30177812 PMCID: PMC6120938 DOI: 10.1038/s41598-018-31073-6] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Accepted: 08/06/2018] [Indexed: 12/31/2022] Open
Abstract
Calpain hyperactivation is implicated in late-stages of neurodegenerative diseases including Alzheimer's disease (AD). However, calpains are also critical for synaptic function and plasticity, and hence memory formation and learning. Since synaptic deficits appear early in AD pathogenesis prior to appearance of overt disease symptoms, we examined if localized dysregulation of calpain-1 and/or 2 contributes to early synaptic dysfunction in AD. Increased activity of synaptosomal calpain-2, but not calpain-1 was observed in presymptomatic 1 month old APPswe/PS1ΔE9 mice (a mouse model of AD) which have no evident pathological or behavioural hallmarks of AD and persisted up to 10 months of age. However, total cellular levels of calpain-2 remained unaffected. Moreover, synaptosomal calpain-2 was hyperactivated in frontal neocortical tissue samples of post-mortem brains of AD-dementia subjects and correlated significantly with decline in tests for cognitive and memory functions, and increase in levels of β-amyloid deposits in brain. We conclude that isoform-specific hyperactivation of calpain-2, but not calpain-1 occurs at the synapse early in the pathogenesis of AD potentially contributing to the deregulation of synaptic signaling in AD. Our findings would be important in paving the way for potential therapeutic strategies for amelioration of cognitive deficits observed in ageing-related dementia disorders like AD.
Collapse
Affiliation(s)
- Faraz Ahmad
- 0000 0001 0482 5067grid.34980.36Centre for Neuroscience, Indian Institute of Science, Bangalore, 560012 India
| | - Debajyoti Das
- 0000 0001 0482 5067grid.34980.36Centre for Neuroscience, Indian Institute of Science, Bangalore, 560012 India
| | - Reddy Peera Kommaddi
- 0000 0001 0482 5067grid.34980.36Centre for Neuroscience, Indian Institute of Science, Bangalore, 560012 India
| | - Latha Diwakar
- 0000 0001 0482 5067grid.34980.36Centre for Neuroscience, Indian Institute of Science, Bangalore, 560012 India
| | - Ruturaj Gowaikar
- 0000 0001 0482 5067grid.34980.36Centre for Neuroscience, Indian Institute of Science, Bangalore, 560012 India
| | - Khader Valli Rupanagudi
- 0000 0001 0482 5067grid.34980.36Centre for Neuroscience, Indian Institute of Science, Bangalore, 560012 India
| | - David A. Bennett
- 0000 0001 0705 3621grid.240684.cRush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, IL 60612 USA
| | - Vijayalakshmi Ravindranath
- Centre for Neuroscience, Indian Institute of Science, Bangalore, 560012, India. .,Centre for Brain Research, Indian Institute of Science, Bangalore, 560012, India.
| |
Collapse
|
16
|
Wu K, Cai Y. The SNP43 (G/A) polymorphism in CAPN10 gene confers an increased risk of cognitive impairment in cerebral small vessel disease. J Clin Lab Anal 2018; 32:e22615. [PMID: 30014550 DOI: 10.1002/jcla.22615] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2018] [Accepted: 06/12/2018] [Indexed: 11/07/2022] Open
Abstract
BACKGROUND Cognitive impairment, significantly reducing processing speed and executive function, is the critical consequence of cerebral small vessel disease (SVD), in which genetic variations have been studied. In this study, we explore the role of SNP43 (G/A) and SNP63 (C/T) polymorphism in the CAPN10 on cognitive impairment process in cerebral SVD. METHODS Cerebral SVD patients (n = 224) and healthy controls (n = 187) were recruited. The relationship between frequency distribution of SNP43 (G/A) and SNP63 (T/C) genotype and allele in CAPN10 gene, and cognitive impairment was examined. The independent risk factors for cognitive impairment in SVD were determined by logistic regression analysis. RESULTS Accordingly, the frequency distribution of genotype and allele at SNP43 (G/A) was significantly different between cerebral SVD patients and healthy controls. Cerebral SVD patients with GG genotype were more susceptible for cognitive impairment, whereas cerebral SVD patients with GA + AA genotype were less possible to suffer from cognitive impairment, compared with those with GG genotype. And also, cerebral SVD does not include SNP63 (C/T) to associate with cognitive impairment, and SNP43 (G/A), total cholesterol, triglyceride, low-density lipoprotein, and high-density lipoprotein were independent risk factors for cognitive impairment in SVD. CONCLUSION Our study provides evidence that SNP43 (G/A) in the CAPN10 gene increases the risk of cognitive impairment in SVD patients. Besides it is proven that, patients with G allele are more susceptible to suffer from cerebral SVD with worse cognitive impairment.
Collapse
Affiliation(s)
- Kai Wu
- Department of Rehabilitation, Xiangya Hospital, Central South University, Changsha, China
| | - Ying Cai
- Department of Rehabilitation, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
17
|
Ma XH, Duan WJ, Mo YS, Chen JL, Li S, Zhao W, Yang L, Mi SQ, Mao XL, Wang H, Wang Q. Neuroprotective effect of paeoniflorin on okadaic acid-induced tau hyperphosphorylation via calpain/Akt/GSK-3β pathway in SH-SY5Y cells. Brain Res 2018; 1690:1-11. [PMID: 29596798 DOI: 10.1016/j.brainres.2018.03.022] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2017] [Revised: 03/15/2018] [Accepted: 03/16/2018] [Indexed: 12/13/2022]
Abstract
Abnormal phosphorylation of tau, one of the most common symptoms of dementia, has become increasingly important in the study of the etiology and development of Alzheimer's disease. Paeoniflorin, the main bioactive component of herbaceous peony, is a monoterpene glycoside, which has been reported to exert beneficial effects on neurodegenerative disease. However, the effect of paeoniflorin on tauopathies remains ambiguous. SH-SY5Y cells were treated with okadaic acid (OA) for 8 h to induce tau phosphorylation and no cell death was observed. Optical microscopy results showed that paeoniflorin ameliorated okadaic acid induced morphological changes, including cell swelling and synapsis shortening. Western blotting data illustrated that paeoniflorin reversed okadaic acid induced tau hyperphosphorylation, which was enhanced by inhibiting the activities of calpain, Akt and GSK-3β. Transmission electron microscopy results showed that paeoniflorin alone can reduce the number of autophagosomes and stabilize the microtubule structure. In addition, calpastain and paeoniflorin enhance the effect of paeoniflorin on stabilizing microtubules. In addition, calpastain markedly enhanced the effect of paeoniflorin on reversing okadaic acid-lowered fluorescence intensity of both MAP-2 and β III-tubulin, two microtubule-associated proteins. This study shows that paeoniflorin protected SH-SY5Y cells against okadaic acid assault by interfering with the calpain/Akt/GSK-3β-related pathways, in which autophagy might be involved. Besides, paeoniflorin is found to relieve the stress response of the microtubule structure system caused by okadaic acid treatment. The results presented in this study suggest that paeoniflorin potentially plays an important role in tauopathies.
Collapse
Affiliation(s)
- Xiao-Hui Ma
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Wen-Jun Duan
- College of Pharmacy, Jinan University, Guangzhou 510080, China
| | - You-Sheng Mo
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Jun-Li Chen
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Shi Li
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Wei Zhao
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Lei Yang
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Sui-Qing Mi
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Xin-Liang Mao
- Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
| | - Hong Wang
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou 510405, China.
| | - Qi Wang
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou 510405, China.
| |
Collapse
|
18
|
Hosseinzadeh S, Noroozian M, Mortaz E, Mousavizadeh K. Plasma microparticles in Alzheimer's disease: The role of vascular dysfunction. Metab Brain Dis 2018; 33:293-299. [PMID: 29209923 DOI: 10.1007/s11011-017-0149-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2017] [Accepted: 11/14/2017] [Indexed: 11/26/2022]
Abstract
Cerebrovascular lesions, a potent stimulus for endothelial cell activation, trigger cognitive and degenerative changes and contribute to pathology of Alzheimer's disease (AD). Circulating microparticles (MPs) are actively involved in the pathogenesis of AD and cerebrovascular diseases, which share common vascular risk factors. We examined the plasma changes of endothelial MPs (EMPs) and platelet MPs (PMPs) in AD patients with vascular risk factors. The plasma Annexin V+ CD 41a- CD144+ EMPs and Annexin V+ CD41a+ CD144- PMPs of 37 patients with AD, with or without vascular risk factors (hypertension, diabetes, dyslipidemia, stroke, coronary artery disease, and smoking), and 10 age-matched controls were quantified by flow cytometry. Pearson correlation analysis used to evaluate the linear relationship between variables. Significantly higher plasma levels of EMPs were observed in AD patients with vascular risk factors as compared to the patients without vascular risk factors [Mean Difference (MD): 2587.80, 95% confidence interval (CI) 770.30-4404.80], and control subjects (MD: 4990.60, 95% CI, 3054.40-6926.79). Significant correlations were found between circulating EMPs, total MPs, and PMPs. There were no significant correlations between plasma levels of EMPs/ PMPs, and cognitive decline indices. Circulating EMP levels are influenced by AD disease status, and plasma levels of MPs and PMPs are associated with vascular risk factors in patients with AD. EMP phenotyping, as cellular biomarkers of vascular injury/dysfunction, and their effects on cerebral perfusion, and cognitive decline should be further investigated. Graphical abstract Vascular endothelial cell activation results in release of endothelial-derived microparticles (EMPs), which contributing to vascular dysfunction and cognitive decline.
Collapse
Affiliation(s)
- Soheila Hosseinzadeh
- Neuroscience Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
- Department of Physiology, School of Medicine, Babol University of Medical Sciences, Babol, Iran
| | - Maryam Noroozian
- Memory and Behavioral Neurology Division, Department of Psychiatry, Roozbeh Hospital, Tehran University of Medical Sciences, Tehran, 1333795914, Iran.
| | - Esmaeil Mortaz
- Department of Immunology, National Research Institute of Tuberculosis and Lung Diseases (NRITLD), Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Kazem Mousavizadeh
- Cellular and Molecular Research Center and Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
19
|
Microglia-derived extracellular vesicles in Alzheimer's Disease: A double-edged sword. Biochem Pharmacol 2018; 148:184-192. [PMID: 29305855 DOI: 10.1016/j.bcp.2017.12.020] [Citation(s) in RCA: 99] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Accepted: 12/22/2017] [Indexed: 01/15/2023]
Abstract
Extracellular vesicles (EVs), based on their origin or size, can be classified as apoptotic bodies, microvesicles (MVs)/microparticles (MPs), and exosomes. EVs are one of the new emerging modes of communication between cells that are providing new insights into the pathophysiology of several diseases. EVs released from activated or apoptotic cells contain specific proteins (signaling molecules, receptors, integrins, cytokines), bioactive lipids, nucleic acids (mRNA, miRNA, small non coding RNAs, DNA) from their progenitor cells. In the brain, EVs contribute to intercellular communication through their basal release and uptake by surrounding cells, or release into the cerebrospinal fluid (CSF) and blood. In the central nervous system (CNS), EVs have been suggested as potential carriers in the intercellular delivery of misfolded proteins associated to neurodegenerative disorders, such as tau and amyloid β in Alzheimer's Disease (AD), α-synuclein in Parkinson's Disease (PD), superoxide dismutase (SOD)1 in amyotrophic lateral sclerosis and huntingtin in Huntington's Disease. Multiple studies indicate that EVs are involved in the pathogenesis of AD, although their role has not been completely elucidated. The focus of this review is to analyze the new emerging role of EVs in AD progression, paying particular attention to microglia EVs. Recent data show that microglia are the first myeloid cells to be activated during neuroinflammation. Microglial EVs in fact, could have both a beneficial and a detrimental action in AD. The study of EVs may provide specific, precise information regarding the AD transition stage that may offer possibilities to intervene in order to retain cognition. In chronic neurodegenerative diseases EVs could be a novel biomarker to monitor the progression of the pathology and also represent a new therapeutical approach to CNS diseases.
Collapse
|
20
|
Fruzangohar M, Ebrahimie E, Adelson DL. A novel hypothesis-unbiased method for Gene Ontology enrichment based on transcriptome data. PLoS One 2017; 12:e0170486. [PMID: 28199395 PMCID: PMC5310883 DOI: 10.1371/journal.pone.0170486] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2016] [Accepted: 01/05/2017] [Indexed: 11/23/2022] Open
Abstract
Gene Ontology (GO) classification of statistically significantly differentially expressed genes is commonly used to interpret transcriptomics data as a part of functional genomic analysis. In this approach, all significantly expressed genes contribute equally to the final GO classification regardless of their actual expression levels. Gene expression levels can significantly affect protein production and hence should be reflected in GO term enrichment. Genes with low expression levels can also participate in GO term enrichment through cumulative effects. In this report, we have introduced a new GO enrichment method that is suitable for multiple samples and time series experiments that uses a statistical outlier test to detect GO categories with special patterns of variation that can potentially identify candidate biological mechanisms. To demonstrate the value of our approach, we have performed two case studies. Whole transcriptome expression profiles of Salmonella enteritidis and Alzheimer's disease (AD) were analysed in order to determine GO term enrichment across the entire transcriptome instead of a subset of differentially expressed genes used in traditional GO analysis. Our result highlights the key role of inflammation related functional groups in AD pathology as granulocyte colony-stimulating factor receptor binding, neuromedin U binding, and interleukin were remarkably upregulated in AD brain when all using all of the gene expression data in the transcriptome. Mitochondrial components and the molybdopterin synthase complex were identified as potential key cellular components involved in AD pathology.
Collapse
Affiliation(s)
- Mario Fruzangohar
- School of Biological Sciences, The University of Adelaide, Adelaide, South Australia, Australia
- School of Agriculture, Food and Wine, The University of Adelaide, Adelaide, South Australia, Australia
| | - Esmaeil Ebrahimie
- Australian Centre for Antimicrobial Resistance Ecology, School of Animal and Veterinary Sciences, The University of Adelaide, Adelaide, South Australia, Australia
- School of Medicine, Faculty of Health Sciences, The University of Adelaide, Adelaide, Australia
- School of Information Technology and Mathematical Sciences, Division of Information Technology, Engineering and the Environment, University of South Australia, Adelaide, Australia
- School of Biological Sciences, Faculty of Science and Engineering, Flinders University, Adelaide, Australia
| | - David L. Adelson
- School of Biological Sciences, The University of Adelaide, Adelaide, South Australia, Australia
- Zhendong Australia – China Centre for Molecular Chinese Medicine, The University of Adelaide, Adelaide, South Australia, Australia
| |
Collapse
|
21
|
Dinkins MB, Wang G, Bieberich E. Sphingolipid-Enriched Extracellular Vesicles and Alzheimer's Disease: A Decade of Research. J Alzheimers Dis 2017; 60:757-768. [PMID: 27662306 PMCID: PMC5360538 DOI: 10.3233/jad-160567] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Extracellular vesicles (EVs), particularly exosomes, have emerged in the last 10 years as a new player in the progression of Alzheimer's disease (AD) with high potential for being useful as a diagnostic and treatment tool. Exosomes and other EVs are enriched with the sphingolipid ceramide as well as other more complex glycosphingolipids such as gangliosides. At least a subpopulation of exosomes requires neutral sphingomyelinase activity for their biogenesis and secretion. As ceramide is often elevated in AD, exosome secretion may be affected as well. Here, we review the available data showing that exosomes regulate the aggregation and clearance of amyloid-beta (Aβ) and discuss the differences in data from laboratories regarding Aβ binding, induction of aggregation, and glial clearance. We also summarize available data on the role of exosomes in extracellular tau propagation, AD-related exosomal mRNA/miRNA cargo, and the use of exosomes as biomarker and gene therapy vehicles for diagnosis and potential treatment.
Collapse
Affiliation(s)
- Michael B. Dinkins
- Department of Neuroscience and Regenerative Medicine, The Medical College of Georgia, Augusta University, Augusta, Georgia, 30912, USA
| | - Guanghu Wang
- Department of Neuroscience and Regenerative Medicine, The Medical College of Georgia, Augusta University, Augusta, Georgia, 30912, USA
| | - Erhard Bieberich
- Department of Neuroscience and Regenerative Medicine, The Medical College of Georgia, Augusta University, Augusta, Georgia, 30912, USA
| |
Collapse
|
22
|
Peng X, Xing P, Li X, Qian Y, Song F, Bai Z, Han G, Lei H. Towards Personalized Intervention for Alzheimer's Disease. GENOMICS PROTEOMICS & BIOINFORMATICS 2016; 14:289-297. [PMID: 27693548 PMCID: PMC5093853 DOI: 10.1016/j.gpb.2016.01.006] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Revised: 01/14/2016] [Accepted: 01/31/2016] [Indexed: 01/17/2023]
Abstract
Alzheimer's disease (AD) remains to be a grand challenge for the international community despite over a century of exploration. A key factor likely accounting for such a situation is the vast heterogeneity in the disease etiology, which involves very complex and divergent pathways. Therefore, intervention strategies shall be tailored for subgroups of AD patients. Both demographic and in-depth information is needed for patient stratification. The demographic information includes primarily APOE genotype, age, gender, education, environmental exposure, life style, and medical history, whereas in-depth information stems from genome sequencing, brain imaging, peripheral biomarkers, and even functional assays on neurons derived from patient-specific induced pluripotent cells (iPSCs). Comprehensive information collection, better understanding of the disease mechanisms, and diversified strategies of drug development would help with more effective intervention in the foreseeable future.
Collapse
Affiliation(s)
- Xing Peng
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China; Cunji Medical School, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Peiqi Xing
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China; Cunji Medical School, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xiuhui Li
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China; Cunji Medical School, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Ying Qian
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China; Cunji Medical School, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Fuhai Song
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China; Cunji Medical School, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zhouxian Bai
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China; Cunji Medical School, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Guangchun Han
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China
| | - Hongxing Lei
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China; Cunji Medical School, University of Chinese Academy of Sciences, Beijing 100049, China; Center of Alzheimer's Disease, Beijing Institute for Brain Disorders, Beijing 100053, China.
| |
Collapse
|
23
|
Kurbatskaya K, Phillips EC, Croft CL, Dentoni G, Hughes MM, Wade MA, Al-Sarraj S, Troakes C, O’Neill MJ, Perez-Nievas BG, Hanger DP, Noble W. Upregulation of calpain activity precedes tau phosphorylation and loss of synaptic proteins in Alzheimer's disease brain. Acta Neuropathol Commun 2016; 4:34. [PMID: 27036949 PMCID: PMC4818436 DOI: 10.1186/s40478-016-0299-2] [Citation(s) in RCA: 95] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2016] [Accepted: 03/15/2016] [Indexed: 12/05/2022] Open
Abstract
Alterations in calcium homeostasis are widely reported to contribute to synaptic degeneration and neuronal loss in Alzheimer’s disease. Elevated cytosolic calcium concentrations lead to activation of the calcium-sensitive cysteine protease, calpain, which has a number of substrates known to be abnormally regulated in disease. Analysis of human brain has shown that calpain activity is elevated in AD compared to controls, and that calpain-mediated proteolysis regulates the activity of important disease-associated proteins including the tau kinases cyclin-dependent kinase 5 and glycogen kinase synthase-3. Here, we sought to investigate the likely temporal association between these changes during the development of sporadic AD using Braak staged post-mortem brain. Quantification of protein amounts in these tissues showed increased activity of calpain-1 from Braak stage III onwards in comparison to controls, extending previous findings that calpain-1 is upregulated at end-stage disease, and suggesting that activation of calcium-sensitive signalling pathways are sustained from early stages of disease development. Increases in calpain-1 activity were associated with elevated activity of the endogenous calpain inhibitor, calpastatin, itself a known calpain substrate. Activation of the tau kinases, glycogen-kinase synthase-3 and cyclin-dependent kinase 5 were also found to occur in Braak stage II-III brain, and these preceded global elevations in tau phosphorylation and the loss of post-synaptic markers. In addition, we identified transient increases in total amyloid precursor protein and pre-synaptic markers in Braak stage II-III brain, that were lost by end stage Alzheimer's disease, that may be indicative of endogenous compensatory responses to the initial stages of neurodegeneration. These findings provide insight into the molecular events that underpin the progression of Alzheimer's disease, and further highlight the rationale for investigating novel treatment strategies that are based on preventing abnormal calcium homeostasis or blocking increases in the activity of calpain or important calpain substrates.
Collapse
|
24
|
Matsumoto SE, Motoi Y, Ishiguro K, Tabira T, Kametani F, Hasegawa M, Hattori N. The twenty-four KDa C-terminal tau fragment increases with aging in tauopathy mice: implications of prion-like properties. Hum Mol Genet 2015; 24:6403-16. [PMID: 26374846 DOI: 10.1093/hmg/ddv351] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2015] [Accepted: 08/26/2015] [Indexed: 12/21/2022] Open
Abstract
The truncated tau protein is a component of the neurofibrillary tangles found in the brains with tauopathies. However, the molecular mechanisms by which the truncated tau fragment causes neurodegeneration remain unknown. Tau pathology was recently suggested to spread through intercellular propagation, and required the formation of 'prion-like' species. We herein identified a new fragment of the tau protein that consisted of four binding domains and a C-terminal tail (Tau-CTF24), but lacked the N-terminal projection domain, and found that it increased with aging in tauopathy model mice (Tg601). Tau-CTF24-like fragments were also present in human brains with tauopathies. A mass spectroscopic analysis revealed that Tau-CTF24 was cleaved behind R242. The digestion of full-length tau (Tau-FL) by calpain produced Tau-CTF24 in vitro and calpain activity increased in old Tg601. Recombinant Tau-CTF24 accelerated heparin-induced aggregation and lost the ability to promote microtubule assembly. When insoluble tau from diseased brains or aggregated recombinant tau was introduced as seeds into SH-SY5Y cells, a larger amount of insoluble tau was formed in cells overexpressing Tau-CTF24 than in those overexpressing Tau-FL. Furthermore, lysates containing the Tau-CTF24 inclusion propagated to naive tau-expressing cells more efficiently than those containing the Tau-FL inclusion. Immunoblot and confocal microscopic analyses revealed that aggregated Tau-CTF24 bound to cells more rapidly and abundantly than aggregated Tau-FL. Our results suggest that Tau-CTF24 contributes to neurodegeneration by enhancing prion-like propagation as well as deteriorating the mechanisms involved in microtubule function.
Collapse
Affiliation(s)
- Shin-Ei Matsumoto
- Department of Diagnosis, Prevention and Treatment of Dementia, Department of Neurology, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 116-0013, Japan and
| | - Yumiko Motoi
- Department of Diagnosis, Prevention and Treatment of Dementia, Department of Neurology, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 116-0013, Japan and
| | - Koichi Ishiguro
- Department of Neurology, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 116-0013, Japan and
| | - Takeshi Tabira
- Department of Diagnosis, Prevention and Treatment of Dementia
| | - Fuyuki Kametani
- Department of Neuropathology and Cell Biology, Tokyo Metropolitan Institute of Medical Science, Setagaya-ku, Tokyo 156-8506, Japan
| | - Masato Hasegawa
- Department of Neuropathology and Cell Biology, Tokyo Metropolitan Institute of Medical Science, Setagaya-ku, Tokyo 156-8506, Japan
| | - Nobutaka Hattori
- Department of Diagnosis, Prevention and Treatment of Dementia, Department of Neurology, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 116-0013, Japan and
| |
Collapse
|