1
|
Zhou Y, Dong W, Wang L, Ren S, Wei W, Wu G. Cystatin C Attenuates Perihematomal Secondary Brain Injury by Inhibiting the Cathepsin B/NLRP3 Signaling Pathway in a Rat Model of Intracerebral Hemorrhage. Mol Neurobiol 2024; 61:9646-9662. [PMID: 38676809 DOI: 10.1007/s12035-024-04195-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2023] [Accepted: 04/19/2024] [Indexed: 04/29/2024]
Abstract
Secondary brain injury (SBI) is a noticeable contributor to the high mortality and morbidity rates associated with intracerebral hemorrhage (ICH), and effective treatment options remain limited. Cystatin C (CysC) emerges as a novel candidate for SBI intervention. The therapeutic effects and underlying mechanisms of CysC in mitigating SBI following ICH were explored in the current research. An in vivo ICH rat model was established by injecting autologous blood into the right caudate nucleus. Western blotting (WB) was utilized to assess the levels of CysC, cathepsin B (CTSB), and the NLRP3 inflammasome. Subsequently, the ICH rat model was treated with exogenous CysC supplementation or CysC knockdown plasmids. Various parameters, including Evans blue (EB) extravasation, brain water content, and neurological function in rats, were examined. RT-qPCR and WB were employed to determine the expression levels of CTSB and the NLRP3 inflammasome. The co-expression of CTSB, CysC, and NLRP3 inflammasome with GFAP, NeuN, and Iba1 was assessed through double-labeled immunofluorescence. The interaction between CysC and CTSB was investigated using double-labeled immunofluorescence and co-immunoprecipitation. The findings revealed an elevation of CysC expression level, particularly at 24 h after ICH. Exogenous CysC supplementation alleviated severe brain edema, neurological deficit scores, and EB extravasation induced by ICH. Conversely, CysC knockdown produced opposite effects. The expression levels of CTSB and the NLRP3 inflammasome were significantly risen following ICH, and exogenous CysC supplement attenuated their expression levels. Double-labeled immunofluorescence illustrated that CysC, CTSB, and the NLRP3 inflammasome were predominantly expressed in microglial cells, and the interaction between CysC and CTSB was evidenced. CysC exhibited potential in ameliorating SBI following ICH via effectively suppressing the activation of the NLRP3 inflammasome mediated by CTSB specifically in microglial cells. These findings underscore the prospective therapeutic efficacy of CysC in the treatment of ICH-induced complications.
Collapse
Affiliation(s)
- Yongfang Zhou
- Department of Emergency, Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, China
| | - Wentao Dong
- Department of Emergency, Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, China
| | - Likun Wang
- Department of Emergency, Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, China
| | - Siying Ren
- Department of Emergency, Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, China
| | - Weiqing Wei
- Department of Emergency, Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, China
| | - Guofeng Wu
- Department of Emergency, Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, China.
| |
Collapse
|
2
|
Rad A, Weigl L, Steinecker-Frohnwieser B, Stadlmayr S, Millesi F, Haertinger M, Borger A, Supper P, Semmler L, Wolf S, Naghilou A, Weiss T, Kress HG, Radtke C. Nuclear Magnetic Resonance Treatment Induces ßNGF Release from Schwann Cells and Enhances the Neurite Growth of Dorsal Root Ganglion Neurons In Vitro. Cells 2024; 13:1544. [PMID: 39329728 PMCID: PMC11430304 DOI: 10.3390/cells13181544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 09/05/2024] [Accepted: 09/08/2024] [Indexed: 09/28/2024] Open
Abstract
Peripheral nerve regeneration depends on close interaction between neurons and Schwann cells (SCs). After nerve injury, SCs produce growth factors and cytokines that are crucial for axon re-growth. Previous studies revealed the supernatant of SCs exposed to nuclear magnetic resonance therapy (NMRT) treatment to increase survival and neurite formation of rat dorsal root ganglion (DRG) neurons in vitro. The aim of this study was to identify factors involved in transferring the observed NMRT-induced effects to SCs and consequently to DRG neurons. Conditioned media of NMRT-treated (CM NMRT) and untreated SCs (CM CTRL) were tested by beta-nerve growth factor (ßNGF) ELISA and multiplex cytokine panels to profile secreted factors. The expression of nociceptive transient receptor potential vanilloid 1 (TRPV1) channels was assessed and the intracellular calcium response in DRG neurons to high-potassium solution, capsaicin or adenosine triphosphate was measured mimicking noxious stimuli. NMRT induced the secretion of ßNGF and pro-regenerative-signaling factors. Blocking antibody experiments confirmed ßNGF as the main factor responsible for neurotrophic/neuritogenic effects of CM NMRT. The TRPV1 expression or sensitivity to specific stimuli was not altered, whereas the viability of cultured DRG neurons was increased. Positive effects of CM NMRT supernatant on DRG neurons are primarily mediated by increased ßNGF levels.
Collapse
Affiliation(s)
- Anda Rad
- Research Laboratory of the Department of Plastic, Reconstructive and Aesthetic Surgery, Medical University of Vienna, Waehringerstrasse 18-20, 1090 Vienna, Austria
| | - Lukas Weigl
- Clinical Department of Special Anesthesia and Pain Therapy, Department of Anaesthesia, Intensive Care Medicine and Pain Medicine, Medical University of Vienna, Waehringerstrasse 18-20, 1090 Vienna, Austria
| | | | - Sarah Stadlmayr
- Research Laboratory of the Department of Plastic, Reconstructive and Aesthetic Surgery, Medical University of Vienna, Waehringerstrasse 18-20, 1090 Vienna, Austria
| | - Flavia Millesi
- Research Laboratory of the Department of Plastic, Reconstructive and Aesthetic Surgery, Medical University of Vienna, Waehringerstrasse 18-20, 1090 Vienna, Austria
| | - Maximilian Haertinger
- Research Laboratory of the Department of Plastic, Reconstructive and Aesthetic Surgery, Medical University of Vienna, Waehringerstrasse 18-20, 1090 Vienna, Austria
| | - Anton Borger
- Research Laboratory of the Department of Plastic, Reconstructive and Aesthetic Surgery, Medical University of Vienna, Waehringerstrasse 18-20, 1090 Vienna, Austria
| | - Paul Supper
- Research Laboratory of the Department of Plastic, Reconstructive and Aesthetic Surgery, Medical University of Vienna, Waehringerstrasse 18-20, 1090 Vienna, Austria
| | - Lorenz Semmler
- Research Laboratory of the Department of Plastic, Reconstructive and Aesthetic Surgery, Medical University of Vienna, Waehringerstrasse 18-20, 1090 Vienna, Austria
| | - Sonja Wolf
- Research Laboratory of the Department of Plastic, Reconstructive and Aesthetic Surgery, Medical University of Vienna, Waehringerstrasse 18-20, 1090 Vienna, Austria
| | - Aida Naghilou
- Research Laboratory of the Department of Plastic, Reconstructive and Aesthetic Surgery, Medical University of Vienna, Waehringerstrasse 18-20, 1090 Vienna, Austria
- Medical Systems Biophysics and Bioengineering, Leiden Academic Centre for Drug Research, Leiden University, 2333 CC Leiden, The Netherlands
| | - Tamara Weiss
- Research Laboratory of the Department of Plastic, Reconstructive and Aesthetic Surgery, Medical University of Vienna, Waehringerstrasse 18-20, 1090 Vienna, Austria
| | - Hans G Kress
- Clinical Department of Special Anesthesia and Pain Therapy, Department of Anaesthesia, Intensive Care Medicine and Pain Medicine, Medical University of Vienna, Waehringerstrasse 18-20, 1090 Vienna, Austria
- Ludwig Boltzmann Institute for Arthritis and Rehabilitation, Hofmanning 214, 8962 Groebming, Austria
| | - Christine Radtke
- Research Laboratory of the Department of Plastic, Reconstructive and Aesthetic Surgery, Medical University of Vienna, Waehringerstrasse 18-20, 1090 Vienna, Austria
| |
Collapse
|
3
|
Jurczak P, Fayad N, Benard M, Czaplewska P, Hildebrandt N. Monomer-Dimer Equilibrium of Human Cystatin C During Internalization Into Cancer Cells. Chembiochem 2024; 25:e202400226. [PMID: 38761032 DOI: 10.1002/cbic.202400226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 04/26/2024] [Accepted: 05/17/2024] [Indexed: 05/20/2024]
Abstract
Human cystatin C (hCC) is a physiologically important protein that serves as intra- and extracellular cysteine proteinase inhibitor in homeostasis. However, in pathological states it dimerizes and further oligomerizes accumulating into a toxic amyloid. HCC forms an active monomer in the extracellular space and becomes an inactive dimer when internalized in cellular organelles. However, hCC cell penetration and its oligomeric state during this process are not well understood. To determine if and how the oligomeric state influences hCC transmembrane migration, we investigated the internalization of the hCC wild type protein as well as three different mutants, which exclusively exist in the monomeric or multimeric state into HeLa cells via confocal fluorescence microscopy. Our results showed that the preferred pathway was endocytosis and that the oligomeric state did not significantly influence the internalization because both monomeric and dimeric hCC migrated into HeLa cells. Considering the differences of the active monomeric and the passive dimeric states of hCC, our findings contribute to a better understanding of the intra and extra cellular functions of hCC and their interaction with cysteine proteases.
Collapse
Affiliation(s)
- Przemyslaw Jurczak
- Laboratory of Mass Spectrometry, Intercollegiate Faculty of Biotechnology, University of Gdansk, Abrahama 58, Gdańsk, 80-307, Poland
- Laboratoire COBRA (UMR6014 & FR3038), Université de Rouen Normandie, CNRS, INSA, Normandie Université, Rouen, 76000, France
| | - Nour Fayad
- Laboratoire COBRA (UMR6014 & FR3038), Université de Rouen Normandie, CNRS, INSA, Normandie Université, Rouen, 76000, France
| | - Magalie Benard
- PRIMACEN, Univ Rouen Normandie, INSERM, CNRS, HeRacLeS US51 UAR2026, Rouen, 76000, France
| | - Paulina Czaplewska
- Laboratory of Mass Spectrometry, Intercollegiate Faculty of Biotechnology, University of Gdansk, Abrahama 58, Gdańsk, 80-307, Poland
| | - Niko Hildebrandt
- Department of Chemistry, Seoul National University, Seoul, 08826, South Korea
- Department of Engineering Physics, McMaster University, 1280 Main Street West, Hamilton, L8S4 L7, Canada
| |
Collapse
|
4
|
Kim Y, D'Acunzo P, Levy E. Biogenesis and secretion of mitovesicles, small extracellular vesicles of mitochondrial origin at the crossroads between brain health and disease. CURRENT OPINION IN PHYSIOLOGY 2024; 40:100765. [PMID: 39219665 PMCID: PMC11364255 DOI: 10.1016/j.cophys.2024.100765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
In the brain, mitochondrial components are released into the extracellular space via several mechanisms, including a recently identified type of extracellular vesicles called mitovesicles. While vesiculation of neuronal mitochondria yields various intracellular types of vesicles, with either a single or a double membrane, mitovesicles secreted into the extracellular space are a unique subtype of these mitochondria-derived vesicles, with a double membrane and a specific set of mitochondrial DNA, RNA, proteins, and lipids. Based on the most relevant literature describing mitochondrial vesiculation and mitochondrial exocytosis, we propose a model for their secretion when the amphisome, a hybrid endosome-autophagosome organelle, fuses with the plasma membrane, releasing mitovesicles and exosomes into the extracellular space. In aging and neurodegenerative disorders, mitochondrial dysfunction, in association with endolysosomal abnormalities, alter mitovesicle number and content, with downstream effect on brain health.
Collapse
Affiliation(s)
- Yohan Kim
- Center for Dementia Research, Nathan S. Kline Institute for Psychiatric Research, Orangeburg, NY 10962, USA
- Department of Psychiatry, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Pasquale D'Acunzo
- Center for Dementia Research, Nathan S. Kline Institute for Psychiatric Research, Orangeburg, NY 10962, USA
- Department of Psychiatry, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Efrat Levy
- Center for Dementia Research, Nathan S. Kline Institute for Psychiatric Research, Orangeburg, NY 10962, USA
- Department of Psychiatry, New York University Grossman School of Medicine, New York, NY 10016, USA
- Department of Biochemistry & Molecular Pharmacology, New York University Grossman School of Medicine, New York, NY 10016, USA
- NYU Neuroscience Institute, New York University Grossman School of Medicine, New York, NY 10016, USA
| |
Collapse
|
5
|
Zhukov I, Sikorska E, Orlikowska M, Górniewicz-Lorens M, Kepczynski M, Jurczak P. DPPA as a Potential Cell Membrane Component Responsible for Binding Amyloidogenic Protein Human Cystatin C. Molecules 2024; 29:3446. [PMID: 39124852 PMCID: PMC11313537 DOI: 10.3390/molecules29153446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Revised: 07/18/2024] [Accepted: 07/19/2024] [Indexed: 08/12/2024] Open
Abstract
A phospholipid bilayer is a typical structure that serves crucial functions in various cells and organelles. However, it is not unusual for it to take part in pathological processes. The cell membrane may be a binding target for amyloid-forming proteins, becoming a factor modulating the oligomerization process leading to amyloid deposition-a hallmark of amyloidogenic diseases-e.g., Alzheimer's disease. The information on the mechanisms governing the oligomerization influenced by the protein-membrane interactions is scarce. Therefore, our study aims to describe the interactions between DPPA, a cell membrane mimetic, and amyloidogenic protein human cystatin C. Circular dichroism spectroscopy and differential scanning calorimetry were used to monitor (i) the secondary structure of the human cystatin C and (ii) the phase transition temperature of the DPPA, during the protein-membrane interactions. NMR techniques were used to determine the protein fragments responsible for the interactions, and molecular dynamics simulations were applied to provide a molecular structure representing the interaction. The obtained data indicate that the protein interacts with DPPA, submerging itself into the bilayer via the AS region. Additionally, the interaction increases the content of α-helix within the protein's secondary structure and stabilizes the whole molecule against denaturation.
Collapse
Affiliation(s)
- Igor Zhukov
- Laboratory of Biological NMR, Institute of Biochemistry and Biophysics, Polish Academy of Sciences, 02-106 Warsaw, Poland;
| | - Emilia Sikorska
- Department of Organic Chemistry, Faculty of Chemistry, University of Gdansk, 80-308 Gdansk, Poland;
| | - Marta Orlikowska
- Department of Biomedical Chemistry, Faculty of Chemistry, University of Gdansk, 80-308 Gdansk, Poland;
| | - Magdalena Górniewicz-Lorens
- Faculty of Chemistry, Jagiellonian University, 30-387 Krakow, Poland; (M.G.-L.); (M.K.)
- Doctoral School of Exact and Natural Sciences, Jagiellonian University, Profesora Stanisława Łojasiewicza 11, 30-348 Krakow, Poland
| | - Mariusz Kepczynski
- Faculty of Chemistry, Jagiellonian University, 30-387 Krakow, Poland; (M.G.-L.); (M.K.)
| | - Przemyslaw Jurczak
- Laboratory of Mass Spectrometry, Intercollegiate Faculty of Biotechnology UG&MUG, University of Gdansk, 80-307 Gdansk, Poland
- Biomacromolecule Research Team, RIKEN Center for Sustainable Resource Science, Wako-shi 351-0198, Saitama, Japan
| |
Collapse
|
6
|
Sun M, Chen Z. Unveiling the Complex Role of Exosomes in Alzheimer's Disease. J Inflamm Res 2024; 17:3921-3948. [PMID: 38911990 PMCID: PMC11193473 DOI: 10.2147/jir.s466821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Accepted: 06/11/2024] [Indexed: 06/25/2024] Open
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative illness, characterized by memory loss and cognitive decline, accounting for 60-80% of dementia cases. AD is characterized by senile plaques made up of amyloid β (Aβ) protein, intracellular neurofibrillary tangles caused by hyperphosphorylation of tau protein linked with microtubules, and neuronal loss. Currently, therapeutic treatments and nanotechnological developments are effective in treating the symptoms of AD, but a cure for the illness has not yet been found. Recently, the increased study of extracellular vesicles (EVs) has led to a growing awareness of their significant involvement in neurodegenerative disorders, including AD. Exosomes are small extracellular vesicles that transport various components including messenger RNAs, non-coding RNAs, proteins, lipids, DNA, and other bioactive compounds from one cell to another, facilitating information transmission and material movement. There is growing evidence indicating that exosomes have complex functions in AD. Exosomes may have a dual role in Alzheimer's disease by contributing to neuronal death and also helping to alleviate the pathological progression of the disease. Therefore, the primary aim of this review is to outline the updated understandings on exosomes biogenesis and many functions of exosomes in the generation, conveyance, distribution, and elimination of hazardous proteins related to Alzheimer's disease. This review is intended to provide novel insights for understanding the development, specific treatment, and early detection of Alzheimer's disease.
Collapse
Affiliation(s)
- Mingyue Sun
- Department of Neurology, The Affiliated Changzhou No. 2 People’s Hospital of Nanjing Medical University, Changzhou, 213000, People’s Republic of China
| | - Zhuoyou Chen
- Department of Neurology, The Affiliated Changzhou No. 2 People’s Hospital of Nanjing Medical University, Changzhou, 213000, People’s Republic of China
| |
Collapse
|
7
|
Gui Y, Kim Y, Brenna S, Wilmes M, Zaghen G, Goulbourne CN, Kuchenbecker-Pöls L, Siebels B, Voß H, Gocke A, Schlüter H, Schweizer M, Altmeppen HC, Magnus T, Levy E, Puig B. Cystatin C loaded in brain-derived extracellular vesicles rescues synapses after ischemic insult in vitro and in vivo. Cell Mol Life Sci 2024; 81:224. [PMID: 38769196 PMCID: PMC11106054 DOI: 10.1007/s00018-024-05266-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Revised: 04/12/2024] [Accepted: 05/05/2024] [Indexed: 05/22/2024]
Abstract
Synaptic loss is an early event in the penumbra area after an ischemic stroke. Promoting synaptic preservation in this area would likely improve functional neurological recovery. We aimed to detect proteins involved in endogenous protection mechanisms of synapses in the penumbra after stroke and to analyse potential beneficial effects of these candidates for a prospective stroke treatment. For this, we performed Liquid Chromatography coupled to Mass Spectrometry (LC-MS)-based proteomics of synaptosomes isolated from the ipsilateral hemispheres of mice subjected to experimental stroke at different time points (24 h, 4 and 7 days) and compared them to sham-operated mice. Proteomic analyses indicated that, among the differentially expressed proteins between the two groups, cystatin C (CysC) was significantly increased at 24 h and 4 days following stroke, before returning to steady-state levels at 7 days, thus indicating a potential transient and intrinsic rescue mechanism attempt of neurons. When CysC was applied to primary neuronal cultures subjected to an in vitro model of ischemic damage, this treatment significantly improved the preservation of synaptic structures. Notably, similar effects were observed when CysC was loaded into brain-derived extracellular vesicles (BDEVs). Finally, when CysC contained in BDEVs was administered intracerebroventricularly to stroked mice, it significantly increased the expression of synaptic markers such as SNAP25, Homer-1, and NCAM in the penumbra area compared to the group supplied with empty BDEVs. Thus, we show that CysC-loaded BDEVs promote synaptic protection after ischemic damage in vitro and in vivo, opening the possibility of a therapeutic use in stroke patients.
Collapse
Affiliation(s)
- Yuqi Gui
- Neurology Department, Experimental Research in Stroke and Inflammation (ERSI) Group, University Medical Center Hamburg-Eppendorf (UKE), Martinistraße, 52, 20246, Hamburg, Germany
- Department of Intensive Care Unit, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Yohan Kim
- Center for Dementia Research, Nathan S. Kline Institute, Orangeburg, NY, 10962, USA
- Departments of Psychiatry, Biochemistry and Molecular Pharmacology, and the Neuroscience Institute, NYU Langone Medical Center, New York, NY, 10016, USA
| | - Santra Brenna
- Neurology Department, Experimental Research in Stroke and Inflammation (ERSI) Group, University Medical Center Hamburg-Eppendorf (UKE), Martinistraße, 52, 20246, Hamburg, Germany
| | - Maximilian Wilmes
- Neurology Department, Experimental Research in Stroke and Inflammation (ERSI) Group, University Medical Center Hamburg-Eppendorf (UKE), Martinistraße, 52, 20246, Hamburg, Germany
| | - Giorgio Zaghen
- Center for Dementia Research, Nathan S. Kline Institute, Orangeburg, NY, 10962, USA
- Departments of Psychiatry, Biochemistry and Molecular Pharmacology, and the Neuroscience Institute, NYU Langone Medical Center, New York, NY, 10016, USA
| | - Chris N Goulbourne
- Center for Dementia Research, Nathan S. Kline Institute, Orangeburg, NY, 10962, USA
| | - Lennart Kuchenbecker-Pöls
- Neurology Department, Experimental Research in Stroke and Inflammation (ERSI) Group, University Medical Center Hamburg-Eppendorf (UKE), Martinistraße, 52, 20246, Hamburg, Germany
| | - Bente Siebels
- Section for Mass Spectrometry and Proteomics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Hannah Voß
- Section for Mass Spectrometry and Proteomics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Antonia Gocke
- Section for Mass Spectrometry and Proteomics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg Eppendorf, Hamburg, Germany
| | - Hartmut Schlüter
- Section for Mass Spectrometry and Proteomics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Michaela Schweizer
- Morphology and Electron Microscopy Core Facility, Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Hermann C Altmeppen
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Tim Magnus
- Neurology Department, Experimental Research in Stroke and Inflammation (ERSI) Group, University Medical Center Hamburg-Eppendorf (UKE), Martinistraße, 52, 20246, Hamburg, Germany
| | - Efrat Levy
- Center for Dementia Research, Nathan S. Kline Institute, Orangeburg, NY, 10962, USA
- Departments of Psychiatry, Biochemistry and Molecular Pharmacology, and the Neuroscience Institute, NYU Langone Medical Center, New York, NY, 10016, USA
| | - Berta Puig
- Neurology Department, Experimental Research in Stroke and Inflammation (ERSI) Group, University Medical Center Hamburg-Eppendorf (UKE), Martinistraße, 52, 20246, Hamburg, Germany.
| |
Collapse
|
8
|
Sun G, Kropp KA, Kirchner M, Plückebaum N, Selich A, Serrero M, Dhingra A, Cabrera JR, Ritter B, Bauerfeind R, Wyler E, Landthaler M, Schambach A, Sodeik B, Mertins P, Viejo-Borbolla A. Herpes simplex virus type 1 modifies the protein composition of extracellular vesicles to promote neurite outgrowth and neuroinfection. mBio 2024; 15:e0330823. [PMID: 38275838 PMCID: PMC10865794 DOI: 10.1128/mbio.03308-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 12/18/2023] [Indexed: 01/27/2024] Open
Abstract
The highly prevalent herpes simplex virus type 1 (HSV-1) causes a range of diseases, including cold sores, blinding keratitis, and life-threatening encephalitis. HSV-1 initially replicates in epithelial cells, enters the peripheral nervous system via neurites, and establishes lifelong infection in the neuronal cell bodies. Neurites are highly dynamic structures that grow or retract in response to attractive or repulsive cues, respectively. Here, we show that infection with HSV-1, but not with a mutant virus lacking glycoprotein G (gG), reduced the repulsive effect of epithelial cells on neurite outgrowth and facilitated HSV-1 invasion of neurons. HSV-1 gG was required and sufficient to induce neurite outgrowth by modifying the protein composition of extracellular vesicles, increasing the amount of neurotrophic and neuroprotective proteins, including galectin-1. Antibodies directed against galectin-1 neutralized the capacity of extracellular vesicles released from HSV-1-infected cells to promote neurite outgrowth. Our study provides new insights into the neurotropism of HSV-1 and identifies a viral protein that modifies the protein composition of extracellular vesicles to stimulate neurite outgrowth and invasion of the nervous system.IMPORTANCEHerpes simplex virus type 1 (HSV-1) must infect neurites (or nerve endings) to establish a chronic infection in neurons. Neurites are highly dynamic structures that retract or grow in the presence of repulsive or attractive proteins. Some of these proteins are released by epithelial cells in extracellular vesicles and act upon interaction with their receptor present on neurites. We show here that HSV-1 infection of epithelial cells modulated their effect on neurites, increasing neurite growth. Mechanistically, HSV-1 glycoprotein G (gG) modifies the protein composition of extracellular vesicles released by epithelial cells, increasing the amount of attractive proteins that enhance neurite outgrowth and facilitate neuronal infection. These results could inform of therapeutic strategies to block HSV-1 induction of neurite outgrowth and, thereby, neuronal infection.
Collapse
Affiliation(s)
- Guorong Sun
- Institute of Virology, Hannover Medical School, Hannover, Germany
| | | | - Marieluise Kirchner
- Proteomics platform, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC) and Berlin Institute of Health (BIH), Berlin, Germany
| | - Nina Plückebaum
- Institute of Virology, Hannover Medical School, Hannover, Germany
| | - Anton Selich
- Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany
| | - Manutea Serrero
- Institute of Virology, Hannover Medical School, Hannover, Germany
| | - Akshay Dhingra
- Institute of Virology, Hannover Medical School, Hannover, Germany
| | - Jorge Rubén Cabrera
- Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas—Universidad Autónoma de Madrid, Madrid, Spain
| | - Birgit Ritter
- Institute of Virology, Hannover Medical School, Hannover, Germany
| | - Rudolf Bauerfeind
- Research Core Unit for Laser Microscopy, Hannover Medical School, Hannover, Germany
| | - Emanuel Wyler
- Berlin Institute for Medical Systems Biology (BIMSB), Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Markus Landthaler
- Berlin Institute for Medical Systems Biology (BIMSB), Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Institute for Biology, Humboldt University of Berlin, Berlin, Germany
| | - Axel Schambach
- Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany
| | - Beate Sodeik
- Institute of Virology, Hannover Medical School, Hannover, Germany
- Cluster of Excellence-Resolving Infection Susceptibility (RESIST, EXC 2155), Hannover Medical School, Hannover, Germany
| | - Philipp Mertins
- Proteomics platform, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC) and Berlin Institute of Health (BIH), Berlin, Germany
| | - Abel Viejo-Borbolla
- Institute of Virology, Hannover Medical School, Hannover, Germany
- Cluster of Excellence-Resolving Infection Susceptibility (RESIST, EXC 2155), Hannover Medical School, Hannover, Germany
| |
Collapse
|
9
|
Du Y, Li Z. Cystatin C alleviates unconjugated bilirubin-induced neurotoxicity by promoting bilirubin clearance from neurocytes via exosomes, dependent on hepatocyte UGT1A1 activity. Transl Neurosci 2024; 15:20220357. [PMID: 39434773 PMCID: PMC11491770 DOI: 10.1515/tnsci-2022-0357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 09/08/2024] [Accepted: 09/23/2024] [Indexed: 10/23/2024] Open
Abstract
There is an urgent need to identify effective drugs for the treatment of nerve injury caused by unconjugated bilirubin (UCB). Our previous research found that cystatin C (CST3) alleviates UCB-induced neurotoxicity by promoting autophagy in nerve cells, but that autophagy inhibitors did not completely inhibit the effects of CST3. This study investigated whether CST3 could alleviate the neurotoxicity of UCB by promoting the secretion and transport of exosomes containing UCB to the liver for metabolism. It demonstrated that hyperbilirubinemia mice treated with CST3 had a higher number of serum exosomes than those in hyperbilirubinemia mice treated with phosphate-buffered saline. CST3-mediated protection against UCB-induced damage was abolished when autophagy and extracellular vesicle inhibitors were used in combination. The number of exosomes in the CST3 overexpression group was higher than that in the control group. Molecular docking experiments showed that UCB and CST3 had high docking score (-8.2). These results suggest that UCB may be excreted from cells by exosomes, and CST3 may promote this process by binding to UCB and entering the exosomes. We demonstrated that the effect of CST3 relied on liver cells with normal UDP-glucuronyl transferase1A1 (UGT1A1) activity in a coculture system of HT22 and L02 cells. CST3 levels were lower in exosomes secreted by L02 cells than in those secreted by human umbilical vein endothelial cells (HUVECs), whereas CST3 levels were higher in the culture supernatants of L02 cells than in the culture supernatants of HUVECs. This suggests that UCB exosomes in L02 cells may be released and internalized by CST3 and that UCB is then processed by UGT1A1 to conjugate UCB, thus reducing its toxicity. These results suggest that CST3 might alleviate UCB-induced neurotoxicity by promoting the clearance of UCB from cells via exosomes and that these effects are dependent on UGT1A1 activity in liver cells.
Collapse
Affiliation(s)
- Yating Du
- Department of Anesthesiology, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong-An Road, Xi-Cheng District, Beijing, 100050, People’s Republic of China
| | - Zhenkun Li
- Beijing Clinical Research Institute, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong-An Road, Xi-Cheng District, Beijing, 100050, People’s Republic of China
| |
Collapse
|
10
|
Tin A, Fohner AE, Yang Q, Brody JA, Davies G, Yao J, Liu D, Caro I, Lindbohm JV, Duggan MR, Meirelles O, Harris SE, Gudmundsdottir V, Taylor AM, Henry A, Beiser AS, Shojaie A, Coors A, Fitzpatrick AL, Langenberg C, Satizabal CL, Sitlani CM, Wheeler E, Tucker-Drob EM, Bressler J, Coresh J, Bis JC, Candia J, Jennings LL, Pietzner M, Lathrop M, Lopez OL, Redmond P, Gerszten RE, Rich SS, Heckbert SR, Austin TR, Hughes TM, Tanaka T, Emilsson V, Vasan RS, Guo X, Zhu Y, Tzourio C, Rotter JI, Walker KA, Ferrucci L, Kivimäki M, Breteler MMB, Cox SR, Debette S, Mosley TH, Gudnason VG, Launer LJ, Psaty BM, Seshadri S, Fornage M. Identification of circulating proteins associated with general cognitive function among middle-aged and older adults. Commun Biol 2023; 6:1117. [PMID: 37923804 PMCID: PMC10624811 DOI: 10.1038/s42003-023-05454-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 10/12/2023] [Indexed: 11/06/2023] Open
Abstract
Identifying circulating proteins associated with cognitive function may point to biomarkers and molecular process of cognitive impairment. Few studies have investigated the association between circulating proteins and cognitive function. We identify 246 protein measures quantified by the SomaScan assay as associated with cognitive function (p < 4.9E-5, n up to 7289). Of these, 45 were replicated using SomaScan data, and three were replicated using Olink data at Bonferroni-corrected significance. Enrichment analysis linked the proteins associated with general cognitive function to cell signaling pathways and synapse architecture. Mendelian randomization analysis implicated higher levels of NECTIN2, a protein mediating viral entry into neuronal cells, with higher Alzheimer's disease (AD) risk (p = 2.5E-26). Levels of 14 other protein measures were implicated as consequences of AD susceptibility (p < 2.0E-4). Proteins implicated as causes or consequences of AD susceptibility may provide new insight into the potential relationship between immunity and AD susceptibility as well as potential therapeutic targets.
Collapse
Grants
- N01 HC095163 NHLBI NIH HHS
- RC2 HL102419 NHLBI NIH HHS
- HHSN268201500003C NHLBI NIH HHS
- UH3 NS100605 NINDS NIH HHS
- R01 HL103612 NHLBI NIH HHS
- 75N92020D00002 NHLBI NIH HHS
- U01 HL096812 NHLBI NIH HHS
- MC_UU_00006/1 Medical Research Council
- UF1 NS125513 NINDS NIH HHS
- 75N92020D00005 NHLBI NIH HHS
- N01AG12100 NIA NIH HHS
- N01HC95160 NHLBI NIH HHS
- R01 AG054076 NIA NIH HHS
- R01 HL120393 NHLBI NIH HHS
- BB/F019394/1 Biotechnology and Biological Sciences Research Council
- RF1 AG059421 NIA NIH HHS
- R01 HL131136 NHLBI NIH HHS
- N01 HC095168 NHLBI NIH HHS
- UL1 RR025005 NCRR NIH HHS
- R01 AG015928 NIA NIH HHS
- HHSN268201800004I NHLBI NIH HHS
- U01 HL080295 NHLBI NIH HHS
- N01HC95163 NHLBI NIH HHS
- N01 AG012100 NIA NIH HHS
- HHSN268201500001C NHLBI NIH HHS
- UL1 TR001079 NCATS NIH HHS
- N01 HC085082 NHLBI NIH HHS
- U01 HL096917 NHLBI NIH HHS
- R01 HL059367 NHLBI NIH HHS
- U01 HL130114 NHLBI NIH HHS
- HHSN268200800007C NHLBI NIH HHS
- R01 HL085251 NHLBI NIH HHS
- N01HC95169 NHLBI NIH HHS
- R01 NS087541 NINDS NIH HHS
- 75N92020D00001 NHLBI NIH HHS
- R01 HL086694 NHLBI NIH HHS
- R01 AG054628 NIA NIH HHS
- U01 HL096902 NHLBI NIH HHS
- R01 HL087652 NHLBI NIH HHS
- N01 HC095162 NHLBI NIH HHS
- U01 HG004402 NHGRI NIH HHS
- N01HC95164 NHLBI NIH HHS
- N01 HC085086 NHLBI NIH HHS
- N01HC55222 NHLBI NIH HHS
- R01 AG049607 NIA NIH HHS
- R01 AG065596 NIA NIH HHS
- N01 HC095165 NHLBI NIH HHS
- N01HC95162 NHLBI NIH HHS
- MR/R024227/1 Medical Research Council
- N01HC85086 NHLBI NIH HHS
- 75N92020D00003 NHLBI NIH HHS
- R01 HL105756 NHLBI NIH HHS
- N01HC95168 NHLBI NIH HHS
- N01 HC095169 NHLBI NIH HHS
- HHSN268201800003I NHLBI NIH HHS
- P30 DK063491 NIDDK NIH HHS
- HHSN268201800007I NHLBI NIH HHS
- HHSN268201700002C NHLBI NIH HHS
- R01 AG066524 NIA NIH HHS
- RF1 AG063507 NIA NIH HHS
- HHSN268201200036C NHLBI NIH HHS
- R01 HL144483 NHLBI NIH HHS
- HHSN268201800001C NHLBI NIH HHS
- HHSN268201700001I NHLBI NIH HHS
- R01 AG056477 NIA NIH HHS
- HHSN268201700004I NHLBI NIH HHS
- N01HC95165 NHLBI NIH HHS
- N01 HC095159 NHLBI NIH HHS
- U01 AG058589 NIA NIH HHS
- N01HC95159 NHLBI NIH HHS
- N01 HC095161 NHLBI NIH HHS
- HHSN268201500001I NHLBI NIH HHS
- R01 AG058969 NIA NIH HHS
- HHSN271201200022C NIDA NIH HHS
- N01 HC025195 NHLBI NIH HHS
- N01HC95161 NHLBI NIH HHS
- UL1 TR001420 NCATS NIH HHS
- 75N92020D00004 NHLBI NIH HHS
- U01 HL096814 NHLBI NIH HHS
- P30 AG066509 NIA NIH HHS
- R01 HL132320 NHLBI NIH HHS
- 75N92020D00007 NHLBI NIH HHS
- P30 AG066546 NIA NIH HHS
- R01 AG033040 NIA NIH HHS
- MR/S011676/1 Medical Research Council
- U01 AG052409 NIA NIH HHS
- HHSN268201500003I NHLBI NIH HHS
- K01 AG071689 NIA NIH HHS
- 75N92021D00006 NHLBI NIH HHS
- R01 AG026307 NIA NIH HHS
- R01 AG020098 NIA NIH HHS
- HHSN268201700005C NHLBI NIH HHS
- HHSN268201700001C NHLBI NIH HHS
- N01HC85082 NHLBI NIH HHS
- HHSN268201700003C NHLBI NIH HHS
- N01 HC095166 NHLBI NIH HHS
- N01HC95167 NHLBI NIH HHS
- N01HC85083 NHLBI NIH HHS
- UH2 NS100605 NINDS NIH HHS
- N01HC25195 NHLBI NIH HHS
- 75N92019D00031 NHLBI NIH HHS
- U01 HL096899 NHLBI NIH HHS
- HHSN268201700004C NHLBI NIH HHS
- UL1 TR000040 NCATS NIH HHS
- HHSN268201700002I NHLBI NIH HHS
- HHSN268201700005I NHLBI NIH HHS
- P30 AG072947 NIA NIH HHS
- R01 AG025941 NIA NIH HHS
- Chief Scientist Office
- 75N92020D00006 NHLBI NIH HHS
- N01HC95166 NHLBI NIH HHS
- R01 AG023629 NIA NIH HHS
- R01 HL087641 NHLBI NIH HHS
- N01HC85079 NHLBI NIH HHS
- N01 HC085080 NHLBI NIH HHS
- UL1 TR001881 NCATS NIH HHS
- N01 HC095167 NHLBI NIH HHS
- HHSN268201800005I NHLBI NIH HHS
- N01HC85080 NHLBI NIH HHS
- HHSN268201700003I NHLBI NIH HHS
- HHSN268201800006I NHLBI NIH HHS
- N01 HC095164 NHLBI NIH HHS
- N01HC85081 NHLBI NIH HHS
- N01 HC095160 NHLBI NIH HHS
- The ARIC study has been funded in whole or in part with Federal funds from the National Heart, Lung, and Blood Institute, National Institutes of Health, Department of Health and Human Services (contract numbers HHSN268201700001I, HHSN268201700002I, HHSN268201700003I, HHSN268201700004I and HHSN268201700005I), R01HL087641, R01HL059367 and R01HL086694; National Human Genome Research Institute contract U01HG004402; and National Institutes of Health contract HHSN268200625226C. Funding was also supported by 5RC2HL102419, R01NS087541 and R01HL131136. Neurocognitive data were collected by U01 2U01HL096812, 2U01HL096814, 2U01HL096899, 2U01HL096902, 2U01HL096917 from the NIH (NHLBI, NINDS, NIA and NIDCD). Infrastructure was partly supported by Grant Number UL1RR025005, a component of the National Institutes of Health and NIH Roadmap for Medical Research. This Cardiovascular Heath Study (CHS) research was supported by NHLBI contracts HHSN268201200036C, HHSN268200800007C, HHSN268201800001C, N01HC55222, N01HC85079, N01HC85080, N01HC85081, N01HC85082, N01HC85083, N01HC85086, 75N92021D00006; and NHLBI grants U01HL080295, R01HL087652, R01HL105756, R01HL103612, R01HL120393, R01HL085251, R01HL144483, and U01HL130114 with additional contribution from the National Institute of Neurological Disorders and Stroke (NINDS). Additional support was provided through R01AG023629, R01AG15928, and R01AG20098 from the National Institute on Aging (NIA). AEF is supported by K01AG071689. The Framingham Heart Study is conducted and supported by the National Heart, Lung, and Blood Institute (NHLBI) in collaboration with Boston University (Contract No. N01-HC-25195, HHSN268201500001I and 75N92019D00031). This work was also supported by grant R01AG063507, R01AG054076, R01AG049607, R01AG059421, R01AG033040, R01AG066524, P30AG066546, U01 AG052409, U01 AG058589 from from the National Institute on Aging and R01 AG017950, UH2/3 NS100605, UF1 NS125513 from National Institute of Neurological Disorders and Stroke and R01HL132320. AGES has been funded by NIA contracts N01-AG012100 and HSSN271201200022C, NIH Grant No. 1R01AG065596-01A1, Hjartavernd (the Icelandic Heart Association), and the Althingi (the Icelandic Parliament). M. R. Duggan, T. Tanaka, J. Candia, K. A. Walker, L. Ferrucci, L.J. Launer, O. Meirelles are funded by the National Institute on Aging Intramural Research Program. This study was funded, in part, by the National Institute on Aging Intramural Research Program. The Coronary Artery Risk Development in Young Adults Study (CARDIA) is supported by contracts HHSN268201800003I, HHSN268201800004I, HHSN268201800005I, HHSN268201800006I, and HHSN268201800007I from the National Heart, Lung, and Blood Institute (NHLBI). The LBC1921 was supported by the UK’s Biotechnology and Biological Sciences Research Council (BBSRC), The Royal Society, and The Chief Scientist Office of the Scottish Government. Genotyping was funded by the BBSRC (BB/F019394/1). LBC1936 is supported by the Biotechnology and Biological Sciences Research Council, and the Economic and Social Research Council [BB/W008793/1], Age UK (Disconnected Mind project), and the University of Edinburgh. Genotyping was funded by the BBSRC (BB/F019394/1). The Olink® Neurology Proteomics assay was supported by a National Institutes of Health (NIH) research grant R01AG054628. Phenotype harmonization, data management, sample-identity QC, and general study coordination, were provided by the TOPMed Data Coordinating Center (3R01HL-120393-02S1), and TOPMed MESA Multi-Omics (HHSN2682015000031/HSN26800004). The MESA projects are conducted and supported by the National Heart, Lung, and Blood Institute (NHLBI) in collaboration with MESA investigators. Support for the Multi-Ethnic Study of Atherosclerosis (MESA) projects are conducted and supported by the National Heart, Lung, and Blood Institute (NHLBI) in collaboration with MESA investigators. Support for MESA is provided by contracts 75N92020D00001, HHSN268201500003I, N01-HC-95159, 75N92020D00005, N01-HC-95160, 75N92020D00002, N01-HC-95161, 75N92020D00003, N01-HC-95162, 75N92020D00006, N01-HC-95163, 75N92020D00004, N01-HC-95164, 75N92020D00007, N01-HC-95165, N01-HC-95166, N01-HC-95167, N01-HC-95168, N01-HC-95169, UL1-TR-000040, UL1-TR-001079, UL1-TR-001420, UL1TR001881, DK063491, and R01HL105756. The Three City (3C) Study is conducted under a partnership agreement among the Institut National de la Santé et de la Recherche Médicale (INSERM), the University of Bordeaux, and Sanofi-Aventis. The Fondation pour la Recherche Médicale funded the preparation and initiation of the study. The 3C Study is also supported by the Caisse Nationale Maladie des Travailleurs Salariés, Direction Générale de la Santé, Mutuelle Générale de l’Education Nationale (MGEN), Institut de la Longévité, Conseils Régionaux of Aquitaine and Bourgogne, Fondation de France, and Ministry of Research–INSERM Programme “Cohortes et collections de données biologiques.” Ilana Caro received a grant from the EUR digital public health. This PhD program is supported within the framework of the PIA3 (Investment for the future). Project reference 17-EURE-0019.
Collapse
Affiliation(s)
- Adrienne Tin
- Memory Impairment and Neurodegenerative Dementia (MIND) Center, University of Mississippi Medical Center, Jackson, MS, USA.
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA.
| | - Alison E Fohner
- Department of Epidemiology, University of Washington, Seattle, WA, USA.
- Institute for Public Health Genetics, University of Washington, Seattle, WA, USA.
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA.
| | - Qiong Yang
- Department of Biostatistics, Boston University, Boston, MA, USA
| | - Jennifer A Brody
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Gail Davies
- Lothian Birth Cohorts, Department of Psychology, University of Edinburgh, 7 George Square, Edinburgh, EH8 9JZ, UK
| | - Jie Yao
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Dan Liu
- Population Health Sciences, German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Ilana Caro
- University of Bordeaux, Institut National de la Santé et de la Recherche Médicale (INSERM), Bordeaux Population Health Research Center, UMR 1219, CHU Bordeaux, Bordeaux, France
| | - Joni V Lindbohm
- Broad Institute of the Massachusetts Institute of Technology and Harvard University, The Klarman Cell Observatory, Cambridge, MA, USA
- Clinicum, Department of Public Health, University of Helsinki, Helsinki, Finland
- Department of Epidemiology and Public Health, University College London, London, UK
| | - Michael R Duggan
- Laboratory of Behavioral Neuroscience, National Institute on Aging, Baltimore, MD, USA
| | - Osorio Meirelles
- National Institute on Aging, National Institutes of Health, Laboratory of Epidemiology and Population Science, Bethesda, MD, USA
| | - Sarah E Harris
- Lothian Birth Cohorts, Department of Psychology, University of Edinburgh, 7 George Square, Edinburgh, EH8 9JZ, UK
| | - Valborg Gudmundsdottir
- Faculty of Medicine, University of Iceland, Reykjavik, Iceland
- Icelandic Heart Association, Kopavogur, Iceland
| | - Adele M Taylor
- Lothian Birth Cohorts, Department of Psychology, University of Edinburgh, 7 George Square, Edinburgh, EH8 9JZ, UK
| | - Albert Henry
- Institute of Cardiovascular Science, University of London, London, UK
| | - Alexa S Beiser
- Department of Biostatistics, Boston University, Boston, MA, USA
- Framingham Heart Study, Framingham, MA, USA
| | - Ali Shojaie
- Department of Biostatistics, University of Washington, Seattle, WA, USA
| | - Annabell Coors
- Population Health Sciences, German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Annette L Fitzpatrick
- Department of Epidemiology, University of Washington, Seattle, WA, USA
- Departments of Family Medicine, University of Washington, Seattle, WA, USA
| | - Claudia Langenberg
- Precision Healthcare Institute, Queen Mary University of London, London, UK
- MRC Epidemiology Unit, University of Cambridge, Cambridge, UK
- Computational Medicine, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Claudia L Satizabal
- Framingham Heart Study, Framingham, MA, USA
- Department of Population Health Sciences and Glenn Biggs Institute for Alzheimer's & Neurodegenerative Diseases, UT Health San Antonio, San Antonio, TX, USA
- Department of Neurology, Boston University School of Medicine, Boston, MA, USA
| | - Colleen M Sitlani
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Eleanor Wheeler
- MRC Epidemiology Unit, University of Cambridge, Cambridge, UK
| | | | - Jan Bressler
- Human Genetics Center, School of Public Health, University of Texas Health Science Center at Houston, Houston, TX, USA
| | | | - Joshua C Bis
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Julián Candia
- Translational Gerontology Branch, National Institute on Aging, Baltimore, MD, USA
| | - Lori L Jennings
- Novartis Institutes for Biomedical Research, 22 Windsor Street, Cambridge, MA, USA
| | - Maik Pietzner
- Precision Healthcare Institute, Queen Mary University of London, London, UK
- MRC Epidemiology Unit, University of Cambridge, Cambridge, UK
- Computational Medicine, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
| | | | - Oscar L Lopez
- Departments of Neurology and Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | - Paul Redmond
- Lothian Birth Cohorts, Department of Psychology, University of Edinburgh, 7 George Square, Edinburgh, EH8 9JZ, UK
| | - Robert E Gerszten
- Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Stephen S Rich
- Center for Public Health Genomics, Department of Public Health Sciences, University of Virginia, Charlottesville, VA, USA
| | - Susan R Heckbert
- Department of Epidemiology, University of Washington, Seattle, WA, USA
| | - Thomas R Austin
- Department of Epidemiology, University of Washington, Seattle, WA, USA
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Timothy M Hughes
- Department of Internal Medicine, Section of Gerontology and Geriatric Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
- Department of Epidemiology and Prevention, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Toshiko Tanaka
- Translational Gerontology Branch, National Institute on Aging, Baltimore, MD, USA
| | - Valur Emilsson
- Faculty of Medicine, University of Iceland, Reykjavik, Iceland
- Icelandic Heart Association, Kopavogur, Iceland
| | - Ramachandran S Vasan
- Framingham Heart Study, Framingham, MA, USA
- University of Texas School of Public Health in San Antonio, San Antonio, TX, USA
- University of Texas Health Sciences Center, San Antonio, TX, USA
| | - Xiuqing Guo
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Yineng Zhu
- Department of Biostatistics, Boston University, Boston, MA, USA
| | - Christophe Tzourio
- University of Bordeaux, Institut National de la Santé et de la Recherche Médicale (INSERM), Bordeaux Population Health Research Center, UMR 1219, CHU Bordeaux, Bordeaux, France
| | - Jerome I Rotter
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Keenan A Walker
- Laboratory of Behavioral Neuroscience, National Institute on Aging, Baltimore, MD, USA
| | - Luigi Ferrucci
- Translational Gerontology Branch, National Institute on Aging, Baltimore, MD, USA
| | - Mika Kivimäki
- UCL Brain Sciences, University College London, London, UK
- Clinicum, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Monique M B Breteler
- Population Health Sciences, German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- Institute for Medical Biometry, Informatics and Epidemiology (IMBIE), Faculty of Medicine, University of Bonn, Bonn, Germany
| | - Simon R Cox
- Lothian Birth Cohorts, Department of Psychology, University of Edinburgh, 7 George Square, Edinburgh, EH8 9JZ, UK
| | - Stephanie Debette
- University of Bordeaux, Institut National de la Santé et de la Recherche Médicale (INSERM), Bordeaux Population Health Research Center, UMR 1219, CHU Bordeaux, Bordeaux, France
- Department of Neurology, Institute for Neurodegenerative Diseases, CHU de Bordeaux, Bordeaux, France
| | - Thomas H Mosley
- Memory Impairment and Neurodegenerative Dementia (MIND) Center, University of Mississippi Medical Center, Jackson, MS, USA
| | | | - Lenore J Launer
- Laboratory of Epidemiology and Population Science, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Bruce M Psaty
- Department of Epidemiology, University of Washington, Seattle, WA, USA
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
- Department of Health Systems and Population Health, University of Washington, Seattle, WA, USA
| | - Sudha Seshadri
- Framingham Heart Study, Framingham, MA, USA
- Department of Population Health Sciences and Glenn Biggs Institute for Alzheimer's & Neurodegenerative Diseases, UT Health San Antonio, San Antonio, TX, USA
| | - Myriam Fornage
- Human Genetics Center, School of Public Health, University of Texas Health Science Center at Houston, Houston, TX, USA
- Institute of Molecular Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, USA
| |
Collapse
|
11
|
Bei J, Miranda-Morales EG, Gan Q, Qiu Y, Husseinzadeh S, Liew JY, Chang Q, Krishnan B, Gaitas A, Yuan S, Felicella M, Qiu WQ, Fang X, Gong B. Circulating exosomes from Alzheimer's disease suppress VE-cadherin expression and induce barrier dysfunction in recipient brain microvascular endothelial cell. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.03.535441. [PMID: 37066187 PMCID: PMC10103966 DOI: 10.1101/2023.04.03.535441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/19/2023]
Abstract
Background Blood-brain barrier (BBB) breakdown is a component of the progression and pathology of Alzheimer's disease (AD). BBB dysfunction is primarily caused by reduced or disorganized tight junction or adherens junction proteins of brain microvascular endothelial cell (BMEC). While there is growing evidence of tight junction disruption in BMECs in AD, the functional role of adherens junctions during BBB dysfunction in AD remains unknown. Exosomes secreted from senescent cells have unique characteristics and contribute to modulating the phenotype of recipient cells. However, it remains unknown if and how these exosomes cause BMEC dysfunction in AD. Objectives This study aimed to investigate the potential roles of AD circulating exosomes and their RNA cargos in brain endothelial dysfunction in AD. Methods We isolated exosomes from sera of five cases of AD compared with age- and sex-matched cognitively normal controls using size-exclusion chromatography technology. We validated the qualities and particle sizes of isolated exosomes with nanoparticle tracking analysis and atomic force microscopy. We measured the biomechanical natures of the endothelial barrier of BMECs, the lateral binding forces between live BMECs, using fluidic force miscopy. We visualized the paracellular expressions of the key adherens junction protein VE-cadherin in BMEC cultures and a 3D BBB model that employs primary human BMECs and pericytes with immunostaining and evaluated them using confocal microscopy. We also examined the VE-cadherin signal in brain tissues from five cases of AD and five age- and sex-matched cognitively normal controls. Results We found that circulating exosomes from AD patients suppress the paracellular expression levels of VE-cadherin and impair the barrier function of recipient BMECs. Immunostaining analysis showed that AD circulating exosomes damage VE-cadherin integrity in a 3D model of microvascular tubule formation. We found that circulating exosomes in AD weaken the BBB depending on the RNA cargos. In parallel, we observed that microvascular VE-cadherin expression is diminished in AD brains compared to normal controls. Conclusion Using in vitro and ex vivo models, our study illustrates that circulating exosomes from AD patients play a significant role in mediating the damage effect on adherens junction of recipient BMEC of the BBB in an exosomal RNA-dependent manner. This suggests a novel mechanism of peripheral senescent exosomes for AD risk.
Collapse
|
12
|
Bei J, Miranda-Morales EG, Gan Q, Qiu Y, Husseinzadeh S, Liew JY, Chang Q, Krishnan B, Gaitas A, Yuan S, Felicella M, Qiu WQ, Fang X, Gong B. Circulating Exosomes from Alzheimer's Disease Suppress Vascular Endothelial-Cadherin Expression and Induce Barrier Dysfunction in Recipient Brain Microvascular Endothelial Cell. J Alzheimers Dis 2023; 95:869-885. [PMID: 37661885 DOI: 10.3233/jad-230347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/05/2023]
Abstract
BACKGROUND Blood-brain barrier (BBB) breakdown is a crucial aspect of Alzheimer's disease (AD) progression. Dysfunction in BBB is primarily caused by impaired tight junction and adherens junction proteins in brain microvascular endothelial cells (BMECs). The role of adherens junctions in AD-related BBB dysfunction remains unclear. Exosomes from senescent cells have unique characteristics and contribute to modulating the phenotype of recipient cells. However, it remains unknown if and how these exosomes cause BMEC dysfunction in AD. OBJECTIVE This study aimed to investigate the impact of AD circulating exosomes on brain endothelial dysfunction. METHODS Exosomes were isolated from sera of AD patients and age- and sex-matched cognitively normal controls using size-exclusion chromatography. The study measured the biomechanical nature of BMECs' endothelial barrier, the lateral binding forces between live BMECs. Paracellular expressions of the key adherens junction protein vascular endothelial (VE)-cadherin were visualized in BMEC cultures and a 3D BBB model using human BMECs and pericytes. VE-cadherin signals were also examined in brain tissues from AD patients and normal controls. RESULTS Circulating exosomes from AD patients reduced VE-cadherin expression levels and impaired barrier function in recipient BMECs. Immunostaining analysis demonstrated that AD exosomes damaged VE-cadherin integrity in a 3D microvascular tubule formation model. The study found that AD exosomes weakened BBB integrity depending on their RNA content. Additionally, diminished microvascular VE-cadherin expression was observed in AD brains compared to controls. CONCLUSION These findings highlight the significant role of circulating exosomes from AD patients in damaging adherens junctions of recipient BMECs, dependent on exosomal RNA.
Collapse
Affiliation(s)
- Jiani Bei
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, USA
| | - Ernesto G Miranda-Morales
- Department of Neurology, Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, Galveston, TX, USA
| | - Qini Gan
- Alzheimer's Disease Center, Boston University School of Medicine, Boston, MA, USA
| | - Yuan Qiu
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, USA
| | - Sorosh Husseinzadeh
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, USA
| | - Jia Yi Liew
- Department of Neurobiology, University of Texas Medical Branch, Galveston, TX, USA
| | - Qing Chang
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, USA
| | - Balaji Krishnan
- Department of Neurology, Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, Galveston, TX, USA
| | - Angelo Gaitas
- The Estelle and Daniel Maggin Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Subo Yuan
- Department of Neurobiology, University of Texas Medical Branch, Galveston, TX, USA
| | - Michelle Felicella
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, USA
| | - Wei Qiao Qiu
- Alzheimer's Disease Center, Boston University School of Medicine, Boston, MA, USA
| | - Xiang Fang
- Department of Neurology, Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, Galveston, TX, USA
| | - Bin Gong
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, USA
| |
Collapse
|
13
|
Cano A, Ettcheto M, Bernuz M, Puerta R, Esteban de Antonio E, Sánchez-López E, Souto EB, Camins A, Martí M, Pividori MI, Boada M, Ruiz A. Extracellular vesicles, the emerging mirrors of brain physiopathology. Int J Biol Sci 2023; 19:721-743. [PMID: 36778117 PMCID: PMC9910004 DOI: 10.7150/ijbs.79063] [Citation(s) in RCA: 23] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Accepted: 11/01/2022] [Indexed: 01/11/2023] Open
Abstract
Extracellular vesicles are secreted by a wide variety of cells, and their primary functions include intercellular communication, immune responses, human reproduction, and synaptic plasticity. Their molecular cargo reflects the physiological processes that their cells of origin are undergoing. Thus, many studies have suggested that extracellular vesicles could be a promising biomarker tool for many diseases, mainly due to their biological relevance and easy accessibility to a broad range of body fluids. Moreover, since their biological composition leads them to cross the blood-brain barrier bidirectionally, growing evidence points to extracellular vesicles as emerging mirrors of brain diseases processes. In this regard, this review explores the biogenesis and biological functions of extracellular vesicles, their role in different physiological and pathological processes, their potential in clinical practice, and the recent outstanding studies about the role of exosomes in major human brain diseases, such as Alzheimer's disease (AD), Parkinson's disease (PD), multiple sclerosis (MS), amyotrophic lateral sclerosis (ALS), or brain tumors.
Collapse
Affiliation(s)
- Amanda Cano
- Ace Alzheimer Center Barcelona - International University of Catalunya (UIC), Barcelona, Spain.,Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Madrid, Spain.,Institute of Nanoscience and Nanotechnology (IN2UB), Barcelona, Spain.,Department of Pharmacy, Pharmaceutical Technology and Physical Chemistry, Faculty of Pharmacy and Food Sciences, University of Barcelona, Spain
| | - Miren Ettcheto
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Madrid, Spain.,Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Sciences, University of Barcelona, Spain
| | - Mireia Bernuz
- Biosensing and Bioanalysis Group, Institut de Biotecnologia i de Biomedicina (IBB-UAB), Mòdul B Parc de Recerca UAB, Campus Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain.,Grup de Sensors i Biosensors, Departament de Química, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain
| | - Raquel Puerta
- Ace Alzheimer Center Barcelona - International University of Catalunya (UIC), Barcelona, Spain
| | | | - Elena Sánchez-López
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Madrid, Spain.,Institute of Nanoscience and Nanotechnology (IN2UB), Barcelona, Spain.,Department of Pharmacy, Pharmaceutical Technology and Physical Chemistry, Faculty of Pharmacy and Food Sciences, University of Barcelona, Spain.,Unit of Synthesis and Biomedical Applications of Peptides, IQAC-CSIC, 08034 Barcelona, Spain
| | - Eliana B Souto
- Department of Pharmaceutical Technology, Faculty of Pharmacy, University of Porto, Porto, Portugal.,REQUIMTE/UCIBIO, Faculty of Pharmacy, University of Porto, Porto, Portugal
| | - Antonio Camins
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Madrid, Spain.,Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Sciences, University of Barcelona, Spain
| | - Mercè Martí
- Biosensing and Bioanalysis Group, Institut de Biotecnologia i de Biomedicina (IBB-UAB), Mòdul B Parc de Recerca UAB, Campus Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain
| | - María Isabel Pividori
- Biosensing and Bioanalysis Group, Institut de Biotecnologia i de Biomedicina (IBB-UAB), Mòdul B Parc de Recerca UAB, Campus Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain.,Grup de Sensors i Biosensors, Departament de Química, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain
| | - Mercè Boada
- Ace Alzheimer Center Barcelona - International University of Catalunya (UIC), Barcelona, Spain.,Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Agustín Ruiz
- Ace Alzheimer Center Barcelona - International University of Catalunya (UIC), Barcelona, Spain.,Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| |
Collapse
|
14
|
Isolation of mitochondria-derived mitovesicles and subpopulations of microvesicles and exosomes from brain tissues. Nat Protoc 2022; 17:2517-2549. [PMID: 35962195 PMCID: PMC9633367 DOI: 10.1038/s41596-022-00719-1] [Citation(s) in RCA: 48] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Accepted: 04/29/2022] [Indexed: 11/08/2022]
Abstract
Extracellular vesicles (EVs) are nanoscale vesicles secreted into the extracellular space by all cell types, including neurons and astrocytes in the brain. EVs play pivotal roles in physiological and pathophysiological processes such as waste removal, cell-to-cell communication and transport of either protective or pathogenic material into the extracellular space. Here we describe a detailed protocol for the reliable and consistent isolation of EVs from both murine and human brains, intended for anyone with basic laboratory experience and performed in a total time of 27 h. The method includes a mild extracellular matrix digestion of the brain tissue, a series of filtration and centrifugation steps to purify EVs and an iodixanol-based high-resolution density step gradient that fractionates different EV populations, including mitovesicles, a newly identified type of EV of mitochondrial origin. We also report detailed downstream protocols for the characterization and analysis of brain EV preparations using nanotrack analysis, electron microscopy and western blotting, as well as for measuring mitovesicular ATP kinetics. Furthermore, we compared this novel iodixanol-based high-resolution density step gradient to the previously described sucrose-based gradient. Although the yield of total EVs recovered was similar, the iodixanol-based gradient better separated distinct EV species as compared with the sucrose-based gradient, including subpopulations of microvesicles, exosomes and mitovesicles. This technique allows quantitative, highly reproducible analyses of brain EV subtypes under normal physiological processes and pathological brain conditions, including neurodegenerative diseases such as Alzheimer's disease and Parkinson's disease.
Collapse
|
15
|
Sex Differentially Alters Secretion of Brain Extracellular Vesicles During Aging: A Potential Mechanism for Maintaining Brain Homeostasis. Neurochem Res 2022; 47:3428-3439. [PMID: 35904699 PMCID: PMC9546961 DOI: 10.1007/s11064-022-03701-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 06/27/2022] [Accepted: 07/15/2022] [Indexed: 11/06/2022]
Abstract
Extracellular vesicles (EVs) in the brain play a role in neuronal homeostasis by removing intracellular material and regulating cell-to-cell communication. Given that sex and aging differentially modulate brain networks, we investigated sex-dependent differences in EV levels and content in the brain during aging. EVs were isolated from the brains of 3, 6, 12, 18, and 24 month-old female and male C57BL/6 J mice, and the levels of different EV species determined. While the number of plasma membrane-derived microvesicles and a subset of late endosomes-derived exosomes increased with age in the brain of female mice, no significant changes were seen in males. Mitochondria-derived mitovesicles in the brain increased during aging in both sexes, a change that may reflect aging-dependent alterations in mitochondrial function. These findings reveal enhanced turnover during aging in female brains, suggesting a mechanism for advantageous successful female brain aging and sex-depending different susceptibility to age-related neurodegenerative diseases.
Collapse
|
16
|
Longobardi A, Nicsanu R, Bellini S, Squitti R, Catania M, Tiraboschi P, Saraceno C, Ferrari C, Zanardini R, Binetti G, Di Fede G, Benussi L, Ghidoni R. Cerebrospinal Fluid EV Concentration and Size Are Altered in Alzheimer’s Disease and Dementia with Lewy Bodies. Cells 2022; 11:cells11030462. [PMID: 35159272 PMCID: PMC8834088 DOI: 10.3390/cells11030462] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 01/21/2022] [Accepted: 01/28/2022] [Indexed: 02/06/2023] Open
Abstract
Alzheimer’s disease (AD), dementia with Lewy bodies (DLB) and frontotemporal dementia (FTD) represent the three major neurodegenerative dementias characterized by abnormal brain protein accumulation. In this study, we investigated extracellular vesicles (EVs) and neurotrophic factors in the cerebrospinal fluid (CSF) of 120 subjects: 36 with AD, 30 with DLB, 34 with FTD and 20 controls. Specifically, CSF EVs were analyzed by Nanoparticle Tracking Analysis and neurotrophic factors were measured with ELISA. We found higher EV concentration and lower EV size in AD and DLB groups compared to the controls. Classification tree analysis demonstrated EV size as the best parameter able to discriminate the patients from the controls (96.7% vs. 3.3%, respectively). The diagnostic performance of the EV concentration/size ratio resulted in a fair discrimination level with an area under the curve of 0.74. Moreover, the EV concentration/size ratio was associated with the p-Tau181/Aβ42 ratio in AD patients. In addition, we described altered levels of cystatin C and progranulin in the DLB and AD groups. We did not find any correlation between neurotrophic factors and EV parameters. In conclusion, the results of this study suggest a common involvement of the endosomal pathway in neurodegenerative dementias, giving important insight into the molecular mechanisms underlying these pathologies.
Collapse
Affiliation(s)
- Antonio Longobardi
- Molecular Markers Laboratory, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, 25125 Brescia, Italy; (A.L.); (R.N.); (S.B.); (R.S.); (C.S.); (R.Z.); (L.B.)
| | - Roland Nicsanu
- Molecular Markers Laboratory, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, 25125 Brescia, Italy; (A.L.); (R.N.); (S.B.); (R.S.); (C.S.); (R.Z.); (L.B.)
| | - Sonia Bellini
- Molecular Markers Laboratory, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, 25125 Brescia, Italy; (A.L.); (R.N.); (S.B.); (R.S.); (C.S.); (R.Z.); (L.B.)
| | - Rosanna Squitti
- Molecular Markers Laboratory, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, 25125 Brescia, Italy; (A.L.); (R.N.); (S.B.); (R.S.); (C.S.); (R.Z.); (L.B.)
| | - Marcella Catania
- Neurology 5 and Neuropathology Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milan, Italy; (M.C.); (P.T.); (G.D.F.)
| | - Pietro Tiraboschi
- Neurology 5 and Neuropathology Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milan, Italy; (M.C.); (P.T.); (G.D.F.)
| | - Claudia Saraceno
- Molecular Markers Laboratory, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, 25125 Brescia, Italy; (A.L.); (R.N.); (S.B.); (R.S.); (C.S.); (R.Z.); (L.B.)
| | - Clarissa Ferrari
- Service of Statistics, 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; (A.L.); (R.N.); (S.B.); (R.S.); (C.S.); (R.Z.); (L.B.)
| | - Giuliano Binetti
- MAC Memory Clinic and Molecular Markers Laboratory, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, 25125 Brescia, Italy;
| | - Giuseppe Di Fede
- Neurology 5 and Neuropathology Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milan, Italy; (M.C.); (P.T.); (G.D.F.)
| | - Luisa Benussi
- Molecular Markers Laboratory, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, 25125 Brescia, Italy; (A.L.); (R.N.); (S.B.); (R.S.); (C.S.); (R.Z.); (L.B.)
| | - Roberta Ghidoni
- Molecular Markers Laboratory, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, 25125 Brescia, Italy; (A.L.); (R.N.); (S.B.); (R.S.); (C.S.); (R.Z.); (L.B.)
- Correspondence: ; Tel.: +39-030-3501725
| |
Collapse
|
17
|
Limongi T, Susa F, Marini M, Allione M, Torre B, Pisano R, di Fabrizio E. Lipid-Based Nanovesicular Drug Delivery Systems. NANOMATERIALS (BASEL, SWITZERLAND) 2021; 11:3391. [PMID: 34947740 PMCID: PMC8707227 DOI: 10.3390/nano11123391] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 12/07/2021] [Accepted: 12/13/2021] [Indexed: 12/14/2022]
Abstract
In designing a new drug, considering the preferred route of administration, various requirements must be fulfilled. Active molecules pharmacokinetics should be reliable with a valuable drug profile as well as well-tolerated. Over the past 20 years, nanotechnologies have provided alternative and complementary solutions to those of an exclusively pharmaceutical chemical nature since scientists and clinicians invested in the optimization of materials and methods capable of regulating effective drug delivery at the nanometer scale. Among the many drug delivery carriers, lipid nano vesicular ones successfully support clinical candidates approaching such problems as insolubility, biodegradation, and difficulty in overcoming the skin and biological barriers such as the blood-brain one. In this review, the authors discussed the structure, the biochemical composition, and the drug delivery applications of lipid nanovesicular carriers, namely, niosomes, proniosomes, ethosomes, transferosomes, pharmacosomes, ufasomes, phytosomes, catanionic vesicles, and extracellular vesicles.
Collapse
|
18
|
Jahić Mujkić A, Tušek Žnidarič M, Berbić S, Žerovnik E. Synergy of the Inhibitory Action of Polyphenols Plus Vitamin C on Amyloid Fibril Formation: Case Study of Human Stefin B. Antioxidants (Basel) 2021; 10:1471. [PMID: 34573102 PMCID: PMC8464686 DOI: 10.3390/antiox10091471] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 09/10/2021] [Accepted: 09/13/2021] [Indexed: 12/06/2022] Open
Abstract
In order to study how polyphenols and vitamin C (vitC) together affect protein aggregation to amyloid fibrils, we performed similar in vitro studies as before using stefin B as a model and a potentially amyloid-forming protein (it aggregates upon overexpression, under stressful conditions and some progressive myoclonus epilepsy of tape 1-EPM1-missense mutations). In addition to the chosen polyphenol, this time, we added a proven antioxidant concentration of 0.5 mM vitC into the fibrillation mixture and varied concentrations of resveratrol, quercetin, and curcumin. Synergy with vitC was observed with curcumin and quercetin.
Collapse
Affiliation(s)
- Alma Jahić Mujkić
- Department of Biochemistry and Molecular and Structural Biology, Jožef Stefan Institute, Jamova 39, 1000 Ljubljana, Slovenia
- Department of Biochemistry, Faculty of Pharmacy, University of Tuzla, Univerzitetska 1, 75000 Tuzla, Bosnia and Herzegovina; (A.J.M.); (S.B.)
| | - Magda Tušek Žnidarič
- Department of Biotechnology and Systems Biology, National Institute of Biology, Večna pot 111, 1000 Ljubljana, Slovenia;
| | - Selma Berbić
- Department of Biochemistry, Faculty of Pharmacy, University of Tuzla, Univerzitetska 1, 75000 Tuzla, Bosnia and Herzegovina; (A.J.M.); (S.B.)
| | - Eva Žerovnik
- Department of Biochemistry and Molecular and Structural Biology, Jožef Stefan Institute, Jamova 39, 1000 Ljubljana, Slovenia
- Jožef Stefan International Postgraduate School, Jamova 39, 1000 Ljubljana, Slovenia
| |
Collapse
|
19
|
Zheng X, She HD, Zhang QX, Si T, Wu KS, Xiao YX. Cystatin C predicts the risk of incident cerebrovascular disease in the elderly: A meta-analysis on survival date studies. Medicine (Baltimore) 2021; 100:e26617. [PMID: 34260548 PMCID: PMC8284707 DOI: 10.1097/md.0000000000026617] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2019] [Revised: 05/26/2021] [Accepted: 06/22/2021] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Stroke is the third leading cause of global year of life lost in all-age and second-ranked cause of disability adjusted life years in middle-aged and elder population. Therefore, it is critical to study the relationship between vascular-related risk factors and cerebrovascular diseases. Several cross-sectional studies have shown that Cystatin C (Cys C) is an independent risk factor for cerebrovascular diseases and levels of Cys C are significantly higher in stroke patients than in healthy individuals. In this meta-analysis, we introduce a Cox proportional hazards model to evaluate the causality between Cys C and the risk of cerebrovascular accident in the elderly. METHODS We searched PubMed, EMBASE, the Web of Science, and the Cochrane Library from 1985 to 2019 for studies on the relationship between serum Cys C and incidence stroke with Cox proportional hazards models. We conducted a subgroup analysis of the selected studies to determine a connection between atherosclerosis and stroke. Finally, 7 research studies, including 26,768 patients without a history of cerebrovascular, were studied. RESULTS After comparing the maximum and minimum Cys C levels, the hazard ratio for all types of stroke, including ischemic and hemorrhagic stroke, was 1.18 (95% confidence interval 1.04-1.31) with moderate heterogeneity (I2 = 43.0%; P = .119) in a fixed-effect model after pooled adjustment for other potential risk factors. In the subgroup analysis, the hazard ratio and 95% confidence interval for Cys C stratified by atherosclerosis was 1.85 (0.97-2.72). As shown in Egger linear regression test, there was no distinct publication bias (P = .153). CONCLUSION Increased serum Cys C is significantly associated with future stroke events in the elderly, especially in patients with carotid atherosclerosis. Thus, serum levels of Cys C could serve as a predicted biomarker for stroke attack.
Collapse
Affiliation(s)
- Xin Zheng
- Department of Neurology, The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong
| | - Hong-da She
- Department of Neurology, The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong
| | - Qiao-xin Zhang
- Department of Neurology, The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong
| | - Tong Si
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing
| | - Ku-sheng Wu
- Department of Public and preventive medicine, Shantou University Medical College, Shantou, Guangdong, China
| | - Ying-xiu Xiao
- Department of Neurology, The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong
| |
Collapse
|
20
|
Landfield Q, Saito M, Hashim A, Canals-Baker S, Sershen H, Levy E, Saito M. Cocaine Induces Sex-Associated Changes in Lipid Profiles of Brain Extracellular Vesicles. Neurochem Res 2021; 46:2909-2922. [PMID: 34245421 PMCID: PMC8490334 DOI: 10.1007/s11064-021-03395-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 07/01/2021] [Accepted: 07/03/2021] [Indexed: 11/27/2022]
Abstract
Cocaine is a highly addictive stimulant with diverse effects on physiology. Recent studies indicate the involvement of extracellular vesicles (EVs) secreted by neural cells in the cocaine addiction process. It is hypothesized that cocaine affects secretion levels of EVs and their cargos, resulting in modulation of synaptic transmission and plasticity related to addiction physiology and pathology. Lipids present in EVs are important for EV formation and for intercellular lipid exchange that may trigger physiological and pathological responses, including neuroplasticity, neurotoxicity, and neuroinflammation. Specific lipids are highly enriched in EVs compared to parent cells, and recent studies suggest the involvement of various lipids in drug-induced synaptic plasticity during the development and maintenance of addiction processes. Therefore, we examined interstitial small EVs isolated from the brain of mice treated with either saline or cocaine, focusing on the effects of cocaine on the lipid composition of EVs. We demonstrate that 12 days of noncontingent repeated cocaine (10 mg/kg) injections to mice, which induce locomotor sensitization, cause lipid composition changes in brain EVs of male mice as compared with saline-injected controls. The most prominent change is the elevation of GD1a ganglioside in brain EVs of males. However, cocaine does not affect the EV lipid profiles of the brain in female mice. Understanding the relationship between lipid composition in EVs and vulnerability to cocaine addiction may provide insight into novel targets for therapies for addiction.
Collapse
Affiliation(s)
- Qwynn Landfield
- Division of Neurochemistry, Nathan S. Kline Institute for Psychiatric Research, 140 Old Orangeburg Rd., Orangeburg, NY, 10962, USA
| | - Mitsuo Saito
- Division of Neurochemistry, Nathan S. Kline Institute for Psychiatric Research, 140 Old Orangeburg Rd., Orangeburg, NY, 10962, USA
| | - Audrey Hashim
- Division of Neurochemistry, Nathan S. Kline Institute for Psychiatric Research, 140 Old Orangeburg Rd., Orangeburg, NY, 10962, USA
| | - Stefanie Canals-Baker
- Division of Neurochemistry, Nathan S. Kline Institute for Psychiatric Research, 140 Old Orangeburg Rd., Orangeburg, NY, 10962, USA
| | - Henry Sershen
- Division of Neurochemistry, Nathan S. Kline Institute for Psychiatric Research, 140 Old Orangeburg Rd., Orangeburg, NY, 10962, USA
- Department of Psychiatry, New York University School of Medicine, New York, NY, USA
| | - Efrat Levy
- Department of Psychiatry, New York University School of Medicine, New York, NY, USA
- Center for Dementia Research, Nathan S. Kline Institute for Psychiatric Research, Orangeburg, NY, USA
- NYU Neuroscience Institute, New York University School of Medicine, New York, NY, USA
- Department of Biochemistry & Molecular Pharmacology, New York University School of Medicine, New York, NY, USA
| | - Mariko Saito
- Division of Neurochemistry, Nathan S. Kline Institute for Psychiatric Research, 140 Old Orangeburg Rd., Orangeburg, NY, 10962, USA.
- Department of Psychiatry, New York University School of Medicine, New York, NY, USA.
| |
Collapse
|
21
|
Zhang T, Ma S, Lv J, Wang X, Afewerky HK, Li H, Lu Y. The emerging role of exosomes in Alzheimer's disease. Ageing Res Rev 2021; 68:101321. [PMID: 33727157 DOI: 10.1016/j.arr.2021.101321] [Citation(s) in RCA: 68] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Revised: 01/20/2021] [Accepted: 03/08/2021] [Indexed: 02/07/2023]
Abstract
Alzheimer's disease (AD), manifested by memory loss and a decline in cognitive functions, is the most prevalent neurodegenerative disease accounting for 60-80 % of dementia cases. But, to-date, there is no effective treatment available to slow or stop the progression of AD. Exosomes are small extracellular vesicles that carry constituents, such as functional messenger RNAs, non-coding RNAs, proteins, lipids, DNA, and other bioactive substances of their source cells. In the brain, exosomes are likely to be sourced by almost all cell types and involve in cell communication to regulate cellular functions. The yet, accumulated evidence on the roles of exosomes and their constituents in the AD pathological process suggests their significance as additional biomarkers and therapeutic targets for AD. This review summarizes the current reported research findings on exosomes roles in the pathogenesis, diagnosis, and treatment of AD.
Collapse
|
22
|
Longobardi A, Benussi L, Nicsanu R, Bellini S, Ferrari C, Saraceno C, Zanardini R, Catania M, Di Fede G, Squitti R, Binetti G, Ghidoni R. Plasma Extracellular Vesicle Size and Concentration Are Altered in Alzheimer's Disease, Dementia With Lewy Bodies, and Frontotemporal Dementia. Front Cell Dev Biol 2021; 9:667369. [PMID: 34046409 PMCID: PMC8148014 DOI: 10.3389/fcell.2021.667369] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Accepted: 03/31/2021] [Indexed: 12/14/2022] Open
Abstract
Alzheimer's disease (AD), frontotemporal dementia (FTD), and dementia with Lewy bodies (DLB) are the three major neurodegenerative dementias. In this study, we provide evidence that an alteration in extracellular vesicles (EVs) release is common across the three most common neurodegenerative dementias, AD, DLB, and FTD. Specifically, we analyzed plasma EVs in three groups of patients affected by AD, DLB, and FTD, and we found a significant reduction in EVs concentration and larger EVs size in all patient groups. We then investigated whether the loss of neurotrophic factors is also a common pathogenic mechanism among FTD, DLB, and AD, and if levels of neurotrophic factors might affect EVs release. Plasma levels of progranulin and cystatin C (CysC) were partially altered; however, taking together all variables significantly associated with the diagnostic groups only EVs size and concentration were able to distinguish patients from controls. The diagnostic performance of these two EVs parameters together (ratio) was high, with a sensitivity of 83.3% and a specificity of 86.7%, able to distinguish patients from controls but not to differentiate the different forms of dementias. Among the candidate neurotrophic factors, only CysC levels were associated with EVs concentration. Our study suggests that an alteration in the intercellular communication mediated by EVs might be a common molecular pathway underlying neurodegenerative dementias. The identification of shared disease mechanisms is of pivotal importance to develop treatments to delay disease progression. To this aim, further studies investigating plasma EVs size and concentration as early biomarkers of dementia are required.
Collapse
Affiliation(s)
- Antonio Longobardi
- Molecular Markers Laboratory, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| | - Luisa Benussi
- Molecular Markers Laboratory, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| | - Roland Nicsanu
- Molecular Markers Laboratory, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| | - Sonia Bellini
- Molecular Markers Laboratory, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| | - Clarissa Ferrari
- Service of Statistics, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| | - Claudia Saraceno
- Molecular Markers Laboratory, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| | - Roberta Zanardini
- Molecular Markers Laboratory, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| | - Marcella Catania
- Neurology 5/Neuropathology Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Giuseppe Di Fede
- Neurology 5/Neuropathology Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Rosanna Squitti
- Molecular Markers Laboratory, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| | - Giuliano Binetti
- Molecular Markers Laboratory, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy.,MAC-Memory Clinic, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| | - Roberta Ghidoni
- Molecular Markers Laboratory, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| |
Collapse
|
23
|
Cerebellar Kv3.3 potassium channels activate TANK-binding kinase 1 to regulate trafficking of the cell survival protein Hax-1. Nat Commun 2021; 12:1731. [PMID: 33741962 PMCID: PMC7979925 DOI: 10.1038/s41467-021-22003-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2020] [Accepted: 02/22/2021] [Indexed: 02/06/2023] Open
Abstract
Mutations in KCNC3, which encodes the Kv3.3 potassium channel, cause degeneration of the cerebellum, but exactly how the activity of an ion channel is linked to the survival of cerebellar neurons is not understood. Here, we report that Kv3.3 channels bind and stimulate Tank Binding Kinase 1 (TBK1), an enzyme that controls trafficking of membrane proteins into multivesicular bodies, and that this stimulation is greatly increased by a disease-causing Kv3.3 mutation. TBK1 activity is required for the binding of Kv3.3 to its auxiliary subunit Hax-1, which prevents channel inactivation with depolarization. Hax-1 is also an anti-apoptotic protein required for survival of cerebellar neurons. Overactivation of TBK1 by the mutant channel leads to the loss of Hax-1 by its accumulation in multivesicular bodies and lysosomes, and also stimulates exosome release from neurons. This process is coupled to activation of caspases and increased cell death. Our studies indicate that Kv3.3 channels are directly coupled to TBK1-dependent biochemical pathways that determine the trafficking of cellular constituents and neuronal survival. How the activity of the neuronal Kv3.3 voltage-dependent channel is regulated is unclear. Here, the authors show that the known Kv3.3 channel complex with Hax1, which affects spinal cerebellar ataxia, regulates the enzyme Tank Binding Kinase 1, modulating survival of cerebellar neurons.
Collapse
|
24
|
D'Acunzo P, Pérez-González R, Kim Y, Hargash T, Miller C, Alldred MJ, Erdjument-Bromage H, Penikalapati SC, Pawlik M, Saito M, Saito M, Ginsberg SD, Neubert TA, Goulbourne CN, Levy E. Mitovesicles are a novel population of extracellular vesicles of mitochondrial origin altered in Down syndrome. SCIENCE ADVANCES 2021; 7:7/7/eabe5085. [PMID: 33579698 PMCID: PMC7880603 DOI: 10.1126/sciadv.abe5085] [Citation(s) in RCA: 135] [Impact Index Per Article: 45.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Accepted: 12/23/2020] [Indexed: 05/03/2023]
Abstract
Mitochondrial dysfunction is an established hallmark of aging and neurodegenerative disorders such as Down syndrome (DS) and Alzheimer's disease (AD). Using a high-resolution density gradient separation of extracellular vesicles (EVs) isolated from murine and human DS and diploid control brains, we identify and characterize a previously unknown population of double-membraned EVs containing multiple mitochondrial proteins distinct from previously described EV subtypes, including microvesicles and exosomes. We term these newly identified mitochondria-derived EVs "mitovesicles." We demonstrate that brain-derived mitovesicles contain a specific subset of mitochondrial constituents and that their levels and cargo are altered during pathophysiological processes where mitochondrial dysfunction occurs, including in DS. The development of a method for the selective isolation of mitovesicles paves the way for the characterization in vivo of biological processes connecting EV biology and mitochondria dynamics and for innovative therapeutic and diagnostic strategies.
Collapse
Affiliation(s)
- Pasquale D'Acunzo
- Center for Dementia Research, Nathan S. Kline Institute for Psychiatric Research, Orangeburg, NY 10962, USA
- Department of Psychiatry, New York University School of Medicine, New York, NY 10016, USA
| | - Rocío Pérez-González
- Center for Dementia Research, Nathan S. Kline Institute for Psychiatric Research, Orangeburg, NY 10962, USA
- Department of Psychiatry, New York University School of Medicine, New York, NY 10016, USA
| | - Yohan Kim
- Center for Dementia Research, Nathan S. Kline Institute for Psychiatric Research, Orangeburg, NY 10962, USA
- Department of Psychiatry, New York University School of Medicine, New York, NY 10016, USA
| | - Tal Hargash
- Center for Dementia Research, Nathan S. Kline Institute for Psychiatric Research, Orangeburg, NY 10962, USA
| | - Chelsea Miller
- Center for Dementia Research, Nathan S. Kline Institute for Psychiatric Research, Orangeburg, NY 10962, USA
| | - Melissa J Alldred
- Center for Dementia Research, Nathan S. Kline Institute for Psychiatric Research, Orangeburg, NY 10962, USA
- Department of Psychiatry, New York University School of Medicine, New York, NY 10016, USA
| | - Hediye Erdjument-Bromage
- Department of Cell Biology, New York University School of Medicine, New York, NY 10016, USA
- Kimmel Center for Biology and Medicine at the Skirball Institute, New York University School of Medicine, New York, NY 10016, USA
| | - Sai C Penikalapati
- Center for Dementia Research, Nathan S. Kline Institute for Psychiatric Research, Orangeburg, NY 10962, USA
| | - Monika Pawlik
- Center for Dementia Research, Nathan S. Kline Institute for Psychiatric Research, Orangeburg, NY 10962, USA
| | - Mitsuo Saito
- Department of Psychiatry, New York University School of Medicine, New York, NY 10016, USA
- Division of Analytical Psychopharmacology, Nathan S. Kline Institute for Psychiatric Research, Orangeburg, NY 10962, USA
| | - Mariko Saito
- Department of Psychiatry, New York University School of Medicine, New York, NY 10016, USA
- Division of Neurochemistry, Nathan S. Kline Institute for Psychiatric Research, Orangeburg, NY 10962, USA
| | - Stephen D Ginsberg
- Center for Dementia Research, Nathan S. Kline Institute for Psychiatric Research, Orangeburg, NY 10962, USA
- Department of Psychiatry, New York University School of Medicine, New York, NY 10016, USA
- NYU Neuroscience Institute, New York University School of Medicine, New York, NY 10016, USA
- Department of Neuroscience and Physiology, New York University School of Medicine, New York, NY 10016, USA
| | - Thomas A Neubert
- Department of Cell Biology, New York University School of Medicine, New York, NY 10016, USA
- Kimmel Center for Biology and Medicine at the Skirball Institute, New York University School of Medicine, New York, NY 10016, USA
| | - Chris N Goulbourne
- Center for Dementia Research, Nathan S. Kline Institute for Psychiatric Research, Orangeburg, NY 10962, USA
| | - Efrat Levy
- Center for Dementia Research, Nathan S. Kline Institute for Psychiatric Research, Orangeburg, NY 10962, USA.
- Department of Psychiatry, New York University School of Medicine, New York, NY 10016, USA
- NYU Neuroscience Institute, New York University School of Medicine, New York, NY 10016, USA
- Department of Biochemistry & Molecular Pharmacology, New York University School of Medicine, New York, NY 10016, USA
| |
Collapse
|
25
|
Huang Y, Cheng L, Turchinovich A, Mahairaki V, Troncoso JC, Pletniková O, Haughey NJ, Vella LJ, Hill AF, Zheng L, Witwer KW. Influence of species and processing parameters on recovery and content of brain tissue-derived extracellular vesicles. J Extracell Vesicles 2020; 9:1785746. [PMID: 32944174 PMCID: PMC7480582 DOI: 10.1080/20013078.2020.1785746] [Citation(s) in RCA: 72] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Extracellular vesicles (EVs) are involved in a wide range of physiological and pathological processes by shuttling material out of and between cells. Tissue EVs may thus lend insights into disease mechanisms and also betray disease when released into easily accessed biological fluids. Since brain-derived EVs (bdEVs) and their cargo may serve as biomarkers of neurodegenerative diseases, we evaluated modifications to a published, rigorous protocol for separation of EVs from brain tissue and studied effects of processing variables on quantitative and qualitative outcomes. To this end, size exclusion chromatography (SEC) and sucrose density gradient ultracentrifugation were compared as final separation steps in protocols involving stepped ultracentrifugation. bdEVs were separated from brain tissues of human, macaque, and mouse. Effects of tissue perfusion and a model of post-mortem interval (PMI) before final bdEV separation were probed. MISEV2018-compliant EV characterization was performed, and both small RNA and protein profiling were done. We conclude that the modified, SEC-employing protocol achieves EV separation efficiency roughly similar to a protocol using gradient density ultracentrifugation, while decreasing operator time and, potentially, variability. The protocol appears to yield bdEVs of higher purity for human tissues compared with those of macaque and, especially, mouse, suggesting opportunities for optimization. Where possible, perfusion should be performed in animal models. The interval between death/tissue storage/processing and final bdEV separation can also affect bdEV populations and composition and should thus be recorded for rigorous reporting. Finally, different populations of EVs obtained through the modified method reported herein display characteristic RNA and protein content that hint at biomarker potential. To conclude, this study finds that the automatable and increasingly employed technique of SEC can be applied to tissue EV separation, and also reveals more about the importance of species-specific and technical considerations when working with tissue EVs. These results are expected to enhance the use of bdEVs in revealing and understanding brain disease.
Collapse
Affiliation(s)
- Yiyao Huang
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Lesley Cheng
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Australia
| | - Andrey Turchinovich
- Molecular Epidemiology, German Cancer Research Center DKFZ, Heidelberg, Germany.,SciBerg e.Kfm, Mannheim, Germany
| | - Vasiliki Mahairaki
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Juan C Troncoso
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Olga Pletniková
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Norman J Haughey
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Laura J Vella
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Australia.,Department of Surgery, The University of Melbourne, the Royal Melbourne Hospital, Parkville, Australia
| | - Andrew F Hill
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Australia
| | - Lei Zheng
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Kenneth W Witwer
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
26
|
Extracellular Vesicles in Cell Biology and Medicine. Sci Rep 2020; 10:8667. [PMID: 32439963 PMCID: PMC7242379 DOI: 10.1038/s41598-020-65826-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Accepted: 05/11/2020] [Indexed: 12/19/2022] Open
|
27
|
Teixeira FG, Vilaça-Faria H, Domingues AV, Campos J, Salgado AJ. Preclinical Comparison of Stem Cells Secretome and Levodopa Application in a 6-Hydroxydopamine Rat Model of Parkinson's Disease. Cells 2020; 9:cells9020315. [PMID: 32012897 PMCID: PMC7072263 DOI: 10.3390/cells9020315] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 01/22/2020] [Accepted: 01/22/2020] [Indexed: 12/18/2022] Open
Abstract
Parkinson's Disease (PD) is characterized by the massive loss of dopaminergic neurons, leading to the appearance of several motor impairments. Current pharmacological treatments, such as the use of levodopa, are yet unable to cure the disease. Therefore, there is a need for novel strategies, particularly those that can combine in an integrated manner neuroprotection and neuroregeneration properties. In vitro and in vivo models have recently revealed that the secretome of mesenchymal stem cells (MSCs) holds a promising potential for treating PD, given its effects on neural survival, proliferation, differentiation. In the present study, we aimed to access the impact of human bone marrow MSCs (hBM-MSCs) secretome in 6-hydroxydopamine (6-OHDA) PD model when compared to levodopa administration, by addressing animals' motor performance, and substantia nigra (SN), and striatum (STR) histological parameters by tyrosine hydroxylase (TH) expression. Results revealed that hBM-MSCs secretome per se appears to be a modulator of the dopaminergic system, enhancing TH-positive cells expression (e.g., dopaminergic neurons) and terminals both in the SN and STR when compared to the untreated group 6-OHDA. Such finding was positively correlated with a significant amelioration of the motor outcomes of 6-OHDA PD animals (assessed by the staircase test). Thus, the present findings support hBM-MSCs secretome administration as a potential therapeutic tool in treating PD, and although we suggest candidate molecules (Trx1, SEMA7A, UCHL1, PEDF, BDNF, Clusterin, SDF-1, CypA, CypB, Cys C, VEGF, DJ-1, Gal-1, GDNF, CDH2, IL-6, HSP27, PRDX1, UBE3A, MMP-2, and GDN) and possible mechanisms of hBM-MSCs secretome-mediated effects, further detailed studies are needed to carefully and clearly define which players may be responsible for its therapeutic actions. By doing so, it will be reasonable to presume that potential treatments that can, per se, or in combination modulate or slow PD may lead to a rational design of new therapeutic or adjuvant strategies for its functional modeling and repair.
Collapse
Affiliation(s)
- Fábio G. Teixeira
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057 Braga, Portugal; (H.V.-F.); (J.C.)
- ICVS/3B’s Associate Lab, PT Government Associated Lab, 4806-909 Braga/Guimarães, Portugal
- Correspondence: (F.G.T.); (A.J.S.); Tel.: +351-253-60-48-71 (F.G.T.); +351-253-60-49-47 (A.J.S.)
| | - Helena Vilaça-Faria
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057 Braga, Portugal; (H.V.-F.); (J.C.)
- ICVS/3B’s Associate Lab, PT Government Associated Lab, 4806-909 Braga/Guimarães, Portugal
| | - Ana V. Domingues
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057 Braga, Portugal; (H.V.-F.); (J.C.)
- ICVS/3B’s Associate Lab, PT Government Associated Lab, 4806-909 Braga/Guimarães, Portugal
| | - Jonas Campos
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057 Braga, Portugal; (H.V.-F.); (J.C.)
- ICVS/3B’s Associate Lab, PT Government Associated Lab, 4806-909 Braga/Guimarães, Portugal
| | - António J. Salgado
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057 Braga, Portugal; (H.V.-F.); (J.C.)
- ICVS/3B’s Associate Lab, PT Government Associated Lab, 4806-909 Braga/Guimarães, Portugal
- Correspondence: (F.G.T.); (A.J.S.); Tel.: +351-253-60-48-71 (F.G.T.); +351-253-60-49-47 (A.J.S.)
| |
Collapse
|
28
|
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: 2.4] [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
|
29
|
Mathews PM, Levy E. Exosome Production Is Key to Neuronal Endosomal Pathway Integrity in Neurodegenerative Diseases. Front Neurosci 2019; 13:1347. [PMID: 31911768 PMCID: PMC6920185 DOI: 10.3389/fnins.2019.01347] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Accepted: 11/29/2019] [Indexed: 12/28/2022] Open
Abstract
Dysfunction of the endosomal–lysosomal system is a prominent pathogenic factor in Alzheimer’s disease (AD) and other neurodevelopmental and neurodegenerative disorders. We and others have extensively characterized the neuronal endosomal pathway pathology that results from either triplication of the amyloid-β precursor protein (APP) gene in Down syndrome (DS) or from expression of the apolipoprotein E ε4 allele (APOE4), the greatest genetic risk factor for late-onset AD. More recently brain exosomes, extracellular vesicles that are generated within and released from endosomal compartments, have been shown to be altered in DS and by APOE4 expression. In this review, we discuss the emerging data arguing for an interdependence between exosome production and endosomal pathway integrity in the brain. In vitro and in vivo studies indicate that altered trafficking through the endosomal pathway or compromised cargo turnover within lysosomes can affect the production, secretion, and content of exosomes. Conversely, exosome biogenesis can affect the endosomal–lysosomal system. Indeed, we propose that efficient exosome release helps to modulate flux through the neuronal endosomal pathway by decompressing potential “traffic jams.” Exosome secretion may have the added benefit of unburdening the neuron’s lysosomal system by delivering endosomal–lysosomal material into the extracellular space, where other cell types may contribute to the degradation of neuronal debris. Thus, maintaining robust neuronal exosome production may prevent or mitigate endosomal and lysosomal abnormalities linked to aging and neurodegenerative diseases. While the current evidence suggests that the exosomal system in the brain can be modulated both by membrane lipid composition and the expression of key proteins that contribute to the formation and secretion of exosomes, how exosomal pathway-regulatory elements sense and respond to perturbations in the endosomal pathway is not well understood. Based upon findings from the extensively studied DS and APOE4 models, we propose that enhanced neuronal exosome secretion can be a protective response, reducing pathological disruption of the endosomal–lysosomal system in disease-vulnerable neurons. Developing therapeutic approaches that help to maintain or enhance neuronal exosome biogenesis and release may be beneficial in a range of disorders of the central nervous system.
Collapse
Affiliation(s)
- Paul M Mathews
- Center for Dementia Research, The Nathan S. Kline Institute for Psychiatric Research, Orangeburg, NY, United States.,Department of Psychiatry, New York University Langone Health, New York, NY, United States.,NYU Neuroscience Institute, New York University Langone Health, New York, NY, United States
| | - Efrat Levy
- Center for Dementia Research, The Nathan S. Kline Institute for Psychiatric Research, Orangeburg, NY, United States.,Department of Psychiatry, New York University Langone Health, New York, NY, United States.,NYU Neuroscience Institute, New York University Langone Health, New York, NY, United States.,Department of Biochemistry and Molecular Pharmacology, New York University Langone Health, New York, NY, United States
| |
Collapse
|