1
|
Hunter TR, Santos LE, Tovar-Moll F, De Felice FG. Alzheimer's disease biomarkers and their current use in clinical research and practice. Mol Psychiatry 2025; 30:272-284. [PMID: 39232196 DOI: 10.1038/s41380-024-02709-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Revised: 08/14/2024] [Accepted: 08/19/2024] [Indexed: 09/06/2024]
Abstract
While blood-based tests are readily available for various conditions, including cardiovascular diseases, type 2 diabetes, and common cancers, Alzheimer's disease (AD) and other neurodegenerative diseases lack an early blood-based screening test that can be used in primary care. Major efforts have been made towards the investigation of approaches that may lead to minimally invasive, cost-effective, and reliable tests capable of measuring brain pathological status. Here, we review past and current technologies developed to investigate biomarkers of AD, including novel blood-based approaches and the more established cerebrospinal fluid and neuroimaging biomarkers of disease. The utility of blood as a source of AD-related biomarkers in both clinical practice and interventional trials is discussed, supported by a comprehensive list of clinical trials for AD drugs and interventions that list biomarkers as primary or secondary endpoints. We highlight that identifying individuals in early preclinical AD using blood-based biomarkers will improve clinical trials and the optimization of therapeutic treatments as they become available. Lastly, we discuss challenges that remain in the field and address new approaches being developed, such as the examination of cargo packaged within extracellular vesicles of neuronal origin isolated from peripheral blood.
Collapse
Affiliation(s)
- Tai R Hunter
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON, Canada
| | - Luis E Santos
- D'Or Institute for Research and Education, Rio de Janeiro, RJ, Brazil.
| | | | - Fernanda G De Felice
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON, Canada.
- D'Or Institute for Research and Education, Rio de Janeiro, RJ, Brazil.
- Centre for Neuroscience Studies and Department of Psychiatry, Queen's University, Kingston, ON, Canada.
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil.
| |
Collapse
|
2
|
Yuan X, Li W, Yan Q, Ou Y, Long Q, Zhang P. Biomarkers of mature neuronal differentiation and related diseases. Future Sci OA 2024; 10:2410146. [PMID: 39429212 PMCID: PMC11497955 DOI: 10.1080/20565623.2024.2410146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Accepted: 09/16/2024] [Indexed: 10/22/2024] Open
Abstract
The nervous system regulates perception, cognition and behavioral responses by serving as the body's primary communication system for receiving, regulating and transmitting information. Neurons are the fundamental structures and units of the nervous system. Their differentiation and maturation processes rely on the expression of specific biomarkers. Neuron-specific intracellular markers can be used to determine the degree of neuronal maturation. Neuronal cytoskeletal proteins dictate the shape and structure of neurons, while synaptic plasticity and signaling processes are intricately associated with neuronal synaptic markers. Furthermore, abnormal expression levels of biomarkers can serve as diagnostic indicators for nervous system diseases. This article reviews the markers of mature neuronal differentiation and their relationship with nervous system diseases.
Collapse
Affiliation(s)
- Xiaodong Yuan
- Department of Neurology, Kailuan General Hospital Affiliated to North China University of Science & Technology, Tangshan, Hebei Province, 063000, China
- Hebei Provincial Key Laboratory of Neurobiological Function, Department of Neurology, Tangshan, Hebei Province, 063000, China
| | - Wen Li
- Department of Neurology, Kailuan General Hospital Affiliated to North China University of Science & Technology, Tangshan, Hebei Province, 063000, China
- Hebei Provincial Key Laboratory of Neurobiological Function, Department of Neurology, Tangshan, Hebei Province, 063000, China
| | - Qi Yan
- Department of Neurology, Kailuan General Hospital Affiliated to North China University of Science & Technology, Tangshan, Hebei Province, 063000, China
- Hebei Provincial Key Laboratory of Neurobiological Function, Department of Neurology, Tangshan, Hebei Province, 063000, China
| | - Ya Ou
- Department of Neurology, Kailuan General Hospital Affiliated to North China University of Science & Technology, Tangshan, Hebei Province, 063000, China
- Hebei Provincial Key Laboratory of Neurobiological Function, Department of Neurology, Tangshan, Hebei Province, 063000, China
| | - Qingxi Long
- Department of Neurology, Kailuan General Hospital Affiliated to North China University of Science & Technology, Tangshan, Hebei Province, 063000, China
- Hebei Provincial Key Laboratory of Neurobiological Function, Department of Neurology, Tangshan, Hebei Province, 063000, China
| | - Pingshu Zhang
- Department of Neurology, Kailuan General Hospital Affiliated to North China University of Science & Technology, Tangshan, Hebei Province, 063000, China
- Hebei Provincial Key Laboratory of Neurobiological Function, Department of Neurology, Tangshan, Hebei Province, 063000, China
| |
Collapse
|
3
|
Agliardi C, Guerini FR, Zanzottera M, Bolognesi E, Caputo D, Rovaris M, Clerici M. Increased concentrations of P2X7R in oligodendrocyte derived extracellular vesicles of Multiple sclerosis patients. Neurobiol Dis 2024; 199:106601. [PMID: 38996986 DOI: 10.1016/j.nbd.2024.106601] [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: 05/14/2024] [Revised: 06/25/2024] [Accepted: 07/09/2024] [Indexed: 07/14/2024] Open
Abstract
Activation of the purinergic receptor P2X7 (P2X7R) is believed to be deleterious in autoimmune diseases and it was hypothesized to play a role in the pathogenesis of MS. P2X7R is an ATP-gated non-selective cationic channel; its activation can be driven by high concentrations of ATP and leads to the generation of large, cytolytic conductance pores. P2X7R activation can also result in apoptosis as a consequence of the activation of the caspase cascade via P2X7R-dependent stimulation of the NLRP3 inflammasome. We measured P2X7R in oligodendrocyte derived extracellular vesicles (ODEVs) in MS patients and in healthy subjects. Sixty-eight MS patients (50 relapsing-remitting, RR-MS, 18 primary progressive, PP-MS) and 57 healthy controls (HC) were enrolled. ODEVs were enriched from serum by a double step immunoaffinity method using an anti OMGp (oligodendrocyte myelin glycoprotein) antibody. P2X7R concentration was measured in ODEVs lysates by ELISA. One-way Anova test showed that P2X7R in ODEVs is significantly higher in PP-MS (mean: 1742.89 pg/mL) compared both to RR-MS (mean: 1277.33 pg/mL) (p < 0.001) and HC (mean: 879.79 pg/mL) (p < 0.001). Comparison between RR-MS and HC was also statistically significant (p < 0.001). Pearson's correlations showed that P2RX7 in ODEVs was positively correlated with EDSS (p = 0.002, r = 0.38, 0.15-0.57 95% CI) and MSSS (p = 0.004, r = 0.34, 0.12-0.54 95% CI) scores, considering MS patients together (PP-MS + RR-MS) and with disease duration in PP-MS group (p = 0.02, r = 0.53, 0.09-0.80 95% CI). Results suggest that ODEVs-associated P2X7R levels could be a biomarker for MS.
Collapse
Affiliation(s)
- Cristina Agliardi
- IRCCS Fondazione Don Carlo Gnocchi ONLUS, Via Capecelatro 66, Milan, Italy
| | | | - Milena Zanzottera
- IRCCS Fondazione Don Carlo Gnocchi ONLUS, Via Capecelatro 66, Milan, Italy
| | | | - Domenico Caputo
- IRCCS Fondazione Don Carlo Gnocchi ONLUS, Via Capecelatro 66, Milan, Italy
| | - Marco Rovaris
- IRCCS Fondazione Don Carlo Gnocchi ONLUS, Via Capecelatro 66, Milan, Italy
| | - Mario Clerici
- IRCCS Fondazione Don Carlo Gnocchi ONLUS, Via Capecelatro 66, Milan, Italy; Pathophysiology and Transplantation Department, University of Milan, via Francesco Sforza 35, Milan, Italy
| |
Collapse
|
4
|
Gonçalves MO, Di Iorio JF, Marin GV, Meneghetti P, Negreiros NGS, Torrecilhas AC. Extracellular vesicles. CURRENT TOPICS IN MEMBRANES 2024; 94:1-31. [PMID: 39370203 DOI: 10.1016/bs.ctm.2024.06.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/08/2024]
Abstract
Cells, pathogens, and other systems release extracellular vesicles (EVs). The particles promote intercellular communication and contain proteins, lipids, RNA and DNA. Initially considered to be cellular waste in the twentieth century, EVs were becoming recognized for their function in biological communication and control. EVs are divided into many subtypes: exosomes, microvesicles, and apoptotic bodies. Exosomes form in the late endosome/multivesicular body and are released when the compartments fuse with the plasma membrane. Microvesicles are generated by direct budding of the plasma membrane, whereas apoptotic bodies are formed after cellular apoptosis. The new guideline for EVs that describes alternate nomenclature for EVs. The particles modulate the immune response by affecting both innate and adaptive immunity, and their specific the structure allows them to be used as biomarkers to diagnose a variety of diseases. EVs have a wide range of applications, for example, delivery systems for medications and genetic therapies because of their ability to convey specific cellular material. In anti-tumor therapy, EVs deliver therapeutic chemicals to tumor cells. The EVs promote transplant compatibility and reduce organ rejection. Host-parasite interactions, therapeutic and diagnostic for cancer, cardiovascular disease, cardiac tissue regeneration, and the treatment of neurological diseases such as Alzheimer's and Parkinson's. The study of EVs keeps on expanding, revealing new functions and beneficial options. EVs have the potential to change drug delivery, diagnostics, and specific therapeutics, creating a new frontier in biomedical.
Collapse
Affiliation(s)
- Mariana Ottaiano Gonçalves
- Laboratório de Imunologia Celular e Bioquímica de Fungos e Protozoários, Departamento de Ciências Farmacêuticas, Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil
| | - Juliana Fortes Di Iorio
- Laboratório de Imunologia Celular e Bioquímica de Fungos e Protozoários, Departamento de Ciências Farmacêuticas, Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil
| | - Gabriela Villa Marin
- Laboratório de Imunologia Celular e Bioquímica de Fungos e Protozoários, Departamento de Ciências Farmacêuticas, Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil
| | - Paula Meneghetti
- Laboratório de Imunologia Celular e Bioquímica de Fungos e Protozoários, Departamento de Ciências Farmacêuticas, Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil
| | - Náthani Gabrielly Silva Negreiros
- Laboratório de Imunologia Celular e Bioquímica de Fungos e Protozoários, Departamento de Ciências Farmacêuticas, Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil
| | - Ana Claudia Torrecilhas
- Laboratório de Imunologia Celular e Bioquímica de Fungos e Protozoários, Departamento de Ciências Farmacêuticas, Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil.
| |
Collapse
|
5
|
Monteleone MC, Billi SC, Abarzúa-Catalán L, Henzi R, Fernández EM, Kaehne T, Wyneken U, Brocco MA. Bulk serum extracellular vesicles from stressed mice show a distinct proteome and induce behavioral and molecular changes in naive mice. PLoS One 2024; 19:e0308976. [PMID: 39146369 PMCID: PMC11326636 DOI: 10.1371/journal.pone.0308976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Accepted: 08/02/2024] [Indexed: 08/17/2024] Open
Abstract
Chronic stress can trigger several pathologies including mood disorders for which no clear diagnostic molecular markers have been established yet. Attractive biomarker sources are extracellular vesicles (EVs). Evs are released by cells in health and disease and contain genetic material, proteins and lipids characteristic of the cell state. Here we show that Evs recovered from the blood of animals exposed to a repeated interrupted stress protocol (RIS) have a different protein profile compared to those obtained from control animals. Proteomic analysis indicated that proteins differentially present in bulk serum Evs from stressed animals were implicated in metabolic and inflammatory pathways and several of them were previously related to psychiatric disorders. Interestingly, these serum Evs carry brain-enriched proteins including the stress-responsive neuronal protein M6a. Then, we used an in-utero electroporation strategy to selectively overexpress M6a-GFP in brain neurons and found that M6a-GFP could also be detected in bulk serum Evs suggesting a neuronal origin. Finally, to determine if these Evs could have functional consequences, we administered Evs from control and RIS animals intranasally to naïve mice. Animals receiving stress EVs showed changes in behavior and brain M6a levels similar to those observed in physically stressed animals. Such changes could therefore be attributed, or at least in part, to EV protein transfer. Altogether these findings show that EVs may participate in stress signaling and propose proteins carried by EVs as a valuable source of biomarkers for stress-induced diseases.
Collapse
Affiliation(s)
- Melisa C Monteleone
- Instituto de Investigaciones Biotecnológicas, Universidad Nacional de San Martín (UNSAM)-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), San Martín, Argentina
- Escuela de Bio y Nanotecnologías (EByN), Universidad Nacional de San Martín, San Martín, Argentina
| | - Silvia C Billi
- Instituto de Investigaciones Biotecnológicas, Universidad Nacional de San Martín (UNSAM)-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), San Martín, Argentina
- Escuela de Bio y Nanotecnologías (EByN), Universidad Nacional de San Martín, San Martín, Argentina
| | - Lorena Abarzúa-Catalán
- Facultad de Medicina, Centro de Investigación e Innovación Biomédica CiiB, Universidad de los Andes, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
| | - Roberto Henzi
- Facultad de Medicina, Centro de Investigación e Innovación Biomédica CiiB, Universidad de los Andes, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
| | - Eliana M Fernández
- Instituto de Investigaciones Biotecnológicas, Universidad Nacional de San Martín (UNSAM)-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), San Martín, Argentina
- Escuela de Bio y Nanotecnologías (EByN), Universidad Nacional de San Martín, San Martín, Argentina
| | - Thilo Kaehne
- Institute of Experimental Internal Medicine, Medical School, Otto von Guericke University Magdeburg, Magdeburg, Germany
| | - Ursula Wyneken
- Facultad de Medicina, Centro de Investigación e Innovación Biomédica CiiB, Universidad de los Andes, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
| | - Marcela A Brocco
- Instituto de Investigaciones Biotecnológicas, Universidad Nacional de San Martín (UNSAM)-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), San Martín, Argentina
- Escuela de Bio y Nanotecnologías (EByN), Universidad Nacional de San Martín, San Martín, Argentina
| |
Collapse
|
6
|
Yao PJ, Manolopoulos A, Eren E, Rivera SM, Hessl DR, Hagerman R, Martinez‐Cerdeno V, Tassone F, Kapogiannis D. Mitochondrial dysfunction in brain tissues and Extracellular Vesicles Fragile X-associated tremor/ataxia syndrome. Ann Clin Transl Neurol 2024; 11:1420-1429. [PMID: 38717724 PMCID: PMC11187838 DOI: 10.1002/acn3.52040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 01/30/2024] [Accepted: 02/24/2024] [Indexed: 06/20/2024] Open
Abstract
OBJECTIVE Mitochondrial impairments have been implicated in the pathogenesis of Fragile X-associated tremor/ataxia syndrome (FXTAS) based on analysis of mitochondria in peripheral tissues and cultured cells. We sought to assess whether mitochondrial abnormalities present in postmortem brain tissues of patients with FXTAS are also present in plasma neuron-derived extracellular vesicles (NDEVs) from living carriers of fragile X messenger ribonucleoprotein1 (FMR1) gene premutations at an early asymptomatic stage of the disease continuum. METHODS We utilized postmortem frozen cerebellar and frontal cortex samples from a cohort of eight patients with FXTAS and nine controls and measured the quantity and activity of the mitochondrial proteins complex IV and complex V. In addition, we evaluated the same measures in isolated plasma NDEVs by selective immunoaffinity capture targeting L1CAM from a separate cohort of eight FMR1 premutation carriers and four age-matched controls. RESULTS Lower complex IV and V quantity and activity were observed in the cerebellum of FXTAS patients compared to controls, without any differences in total mitochondrial content. No patient-control differences were observed in the frontal cortex. In NDEVs, FMR1 premutation carriers compared to controls had lower activity of Complex IV and Complex V, but higher Complex V quantity. INTERPRETATION Quantitative and functional abnormalities in mitochondrial electron transport chain complexes IV and V seen in the cerebellum of patients with FXTAS are also manifest in plasma NDEVs of FMR1 premutation carriers. Plasma NDEVs may provide further insights into mitochondrial pathologies in this syndrome and could potentially lead to the development of biomarkers for predicting symptomatic FXTAS among premutation carriers and disease monitoring.
Collapse
Affiliation(s)
- Pamela J. Yao
- Laboratory of Clinical Investigation, Intramural Research ProgramNational Institute on Aging, National Institutes of HealthBaltimoreMarylandUSA
| | - Apostolos Manolopoulos
- Laboratory of Clinical Investigation, Intramural Research ProgramNational Institute on Aging, National Institutes of HealthBaltimoreMarylandUSA
| | - Erden Eren
- Laboratory of Clinical Investigation, Intramural Research ProgramNational Institute on Aging, National Institutes of HealthBaltimoreMarylandUSA
| | - Susan Michelle Rivera
- Department of PsychologyUniversity of MarylandCollege ParkMarylandUSA
- MIND InstituteUniversity of California, Davis, Medical CenterSacramentoCaliforniaUSA
| | - David R. Hessl
- MIND InstituteUniversity of California, Davis, Medical CenterSacramentoCaliforniaUSA
- Department of Psychiatry and Behavioral SciencesUniversity of California, Davis, School of MedicineSacramentoCaliforniaUSA
| | - Randi Hagerman
- MIND InstituteUniversity of California, Davis, Medical CenterSacramentoCaliforniaUSA
- Department of PediatricsUniversity of California, Davis, School of MedicineSacramentoCaliforniaUSA
| | - Veronica Martinez‐Cerdeno
- MIND InstituteUniversity of California, Davis, Medical CenterSacramentoCaliforniaUSA
- Department of Pathology and Laboratory MedicineUniversity of California, Davis, School of MedicineSacramentoCaliforniaUSA
- Institute for Pediatric Regenerative Medicine at Shriners Hospitals for Children Northern CaliforniaSacramentoCaliforniaUSA
| | - Flora Tassone
- MIND InstituteUniversity of California, Davis, Medical CenterSacramentoCaliforniaUSA
- Department of Biochemistry and Molecular MedicineUniversity of California, Davis, School of MedicineSacramentoCaliforniaUSA
| | - Dimitrios Kapogiannis
- Laboratory of Clinical Investigation, Intramural Research ProgramNational Institute on Aging, National Institutes of HealthBaltimoreMarylandUSA
| |
Collapse
|
7
|
Nogueras‐Ortiz CJ, Eren E, Yao P, Calzada E, Dunn C, Volpert O, Delgado‐Peraza F, Mustapic M, Lyashkov A, Rubio FJ, Vreones M, Cheng L, You Y, Hill AF, Ikezu T, Eitan E, Goetzl EJ, Kapogiannis D. Single-extracellular vesicle (EV) analyses validate the use of L1 Cell Adhesion Molecule (L1CAM) as a reliable biomarker of neuron-derived EVs. J Extracell Vesicles 2024; 13:e12459. [PMID: 38868956 PMCID: PMC11170079 DOI: 10.1002/jev2.12459] [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: 10/31/2023] [Revised: 04/15/2024] [Accepted: 05/10/2024] [Indexed: 06/14/2024] Open
Abstract
Isolation of neuron-derived extracellular vesicles (NDEVs) with L1 Cell Adhesion Molecule (L1CAM)-specific antibodies has been widely used to identify blood biomarkers of CNS disorders. However, full methodological validation requires demonstration of L1CAM in individual NDEVs and lower levels or absence of L1CAM in individual EVs from other cells. Here, we used multiple single-EV techniques to establish the neuronal origin and determine the abundance of L1CAM-positive EVs in human blood. L1CAM epitopes of the ectodomain are shown to be co-expressed on single-EVs with the neuronal proteins β-III-tubulin, GAP43, and VAMP2, the levels of which increase in parallel with the enrichment of L1CAM-positive EVs. Levels of L1CAM-positive EVs carrying the neuronal proteins VAMP2 and β-III-tubulin range from 30% to 63%, in contrast to 0.8%-3.9% of L1CAM-negative EVs. Plasma fluid-phase L1CAM does not bind to single-EVs. Our findings support the use of L1CAM as a target for isolating plasma NDEVs and leveraging their cargo to identify biomarkers reflecting neuronal function.
Collapse
Affiliation(s)
- Carlos J Nogueras‐Ortiz
- Laboratory of Clinical Investigation, Intramural Research ProgramNational Institute on Aging, National Institutes of Health (NIA/NIH)BaltimoreMarylandUSA
| | - Erden Eren
- Laboratory of Clinical Investigation, Intramural Research ProgramNational Institute on Aging, National Institutes of Health (NIA/NIH)BaltimoreMarylandUSA
| | - Pamela Yao
- Laboratory of Clinical Investigation, Intramural Research ProgramNational Institute on Aging, National Institutes of Health (NIA/NIH)BaltimoreMarylandUSA
| | - Elizabeth Calzada
- Laboratory of Clinical Investigation, Intramural Research ProgramNational Institute on Aging, National Institutes of Health (NIA/NIH)BaltimoreMarylandUSA
| | - Christopher Dunn
- Flow Cytometry Unit, Intramural Research ProgramNational Institute on Aging, National Institutes of Health (NIA/NIH)BaltimoreMarylandUSA
| | | | - Francheska Delgado‐Peraza
- Laboratory of Clinical Investigation, Intramural Research ProgramNational Institute on Aging, National Institutes of Health (NIA/NIH)BaltimoreMarylandUSA
| | - Maja Mustapic
- Laboratory of Clinical Investigation, Intramural Research ProgramNational Institute on Aging, National Institutes of Health (NIA/NIH)BaltimoreMarylandUSA
| | - Alexey Lyashkov
- Laboratory of Clinical Investigation, Intramural Research ProgramNational Institute on Aging, National Institutes of Health (NIA/NIH)BaltimoreMarylandUSA
| | - F Javier Rubio
- Neuronal Ensembles in Addiction Section, Behavioral Neuroscience Research BranchIntramural Research Program/National Institute on Drug Abuse/National Institutes of HealthBaltimoreMarylandUSA
| | - Michael Vreones
- Laboratory of Clinical Investigation, Intramural Research ProgramNational Institute on Aging, National Institutes of Health (NIA/NIH)BaltimoreMarylandUSA
| | - Lesley Cheng
- La Trobe Institute for Molecular ScienceLa Trobe UniversityBundooraVictoriaAustralia
| | - Yang You
- Department of NeuroscienceMayo ClinicJacksonvilleFloridaUSA
- Department of Pharmacology and Experimental TherapeuticsBoston University School of MedicineBostonMassachusettsUSA
| | - Andrew F Hill
- La Trobe Institute for Molecular ScienceLa Trobe UniversityBundooraVictoriaAustralia
- Institute for Health and SportVictoria UniversityMelbourneVictoriaAustralia
| | - Tsuneya Ikezu
- Department of NeuroscienceMayo ClinicJacksonvilleFloridaUSA
- Department of Pharmacology and Experimental TherapeuticsBoston University School of MedicineBostonMassachusettsUSA
| | | | - Edward J Goetzl
- Department of MedicineUniversity of CaliforniaSan FranciscoCaliforniaUSA
- San Francisco Campus for Jewish LivingSan FranciscoCaliforniaUSA
| | - Dimitrios Kapogiannis
- Laboratory of Clinical Investigation, Intramural Research ProgramNational Institute on Aging, National Institutes of Health (NIA/NIH)BaltimoreMarylandUSA
- Department of NeurologyJohns Hopkins School of MedicineBaltimoreMarylandUSA
| |
Collapse
|
8
|
Krokidis MG, Pucha KA, Mustapic M, Exarchos TP, Vlamos P, Kapogiannis D. Lipidomic Analysis of Plasma Extracellular Vesicles Derived from Alzheimer's Disease Patients. Cells 2024; 13:702. [PMID: 38667317 PMCID: PMC11049154 DOI: 10.3390/cells13080702] [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: 02/16/2024] [Revised: 03/31/2024] [Accepted: 04/14/2024] [Indexed: 04/28/2024] Open
Abstract
Analysis of blood-based indicators of brain health could provide an understanding of early disease mechanisms and pinpoint possible intervention strategies. By examining lipid profiles in extracellular vesicles (EVs), secreted particles from all cells, including astrocytes and neurons, and circulating in clinical samples, important insights regarding the brain's composition can be gained. Herein, a targeted lipidomic analysis was carried out in EVs derived from plasma samples after removal of lipoproteins from individuals with Alzheimer's disease (AD) and healthy controls. Differences were observed for selected lipid species of glycerolipids (GLs), glycerophospholipids (GPLs), lysophospholipids (LPLs) and sphingolipids (SLs) across three distinct EV subpopulations (all-cell origin, derived by immunocapture of CD9, CD81 and CD63; neuronal origin, derived by immunocapture of L1CAM; and astrocytic origin, derived by immunocapture of GLAST). The findings provide new insights into the lipid composition of EVs isolated from plasma samples regarding specific lipid families (MG, DG, Cer, PA, PC, PE, PI, LPI, LPE, LPC), as well as differences between AD and control individuals. This study emphasizes the crucial role of plasma EV lipidomics analysis as a comprehensive approach for identifying biomarkers and biological targets in AD and related disorders, facilitating early diagnosis and potentially informing novel interventions.
Collapse
Affiliation(s)
- Marios G. Krokidis
- Laboratory of Bioinformatics and Human Electrophysiology, Department of Informatics, Ionian University, 49100 Corfu, Greece; (M.G.K.); (T.P.E.); (P.V.)
| | - Krishna A. Pucha
- Laboratory of Clinical Investigation, National Institute on Aging, National Institutes of Health (NIA/NIH), Baltimore, MD 21224, USA; (K.A.P.); (M.M.)
| | - Maja Mustapic
- Laboratory of Clinical Investigation, National Institute on Aging, National Institutes of Health (NIA/NIH), Baltimore, MD 21224, USA; (K.A.P.); (M.M.)
| | - Themis P. Exarchos
- Laboratory of Bioinformatics and Human Electrophysiology, Department of Informatics, Ionian University, 49100 Corfu, Greece; (M.G.K.); (T.P.E.); (P.V.)
| | - Panagiotis Vlamos
- Laboratory of Bioinformatics and Human Electrophysiology, Department of Informatics, Ionian University, 49100 Corfu, Greece; (M.G.K.); (T.P.E.); (P.V.)
| | - Dimitrios Kapogiannis
- Laboratory of Clinical Investigation, National Institute on Aging, National Institutes of Health (NIA/NIH), Baltimore, MD 21224, USA; (K.A.P.); (M.M.)
| |
Collapse
|
9
|
Li X, Chen J, Yang Y, Cai H, Ao Z, Xing Y, Li K, Yang K, Wallace A, Friend J, Lee LP, Wang N, Guo F. Extracellular vesicles-based point-of-care testing for the diagnosis and monitoring of Alzheimer's disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.31.587511. [PMID: 38617279 PMCID: PMC11014472 DOI: 10.1101/2024.03.31.587511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/16/2024]
Abstract
Alzheimer's disease (AD) is a debilitating condition that affects millions of people worldwide. One promising strategy for detecting and monitoring AD early on is using extracellular vesicles (EVs)-based point-of-care testing; however, diagnosing AD using EVs poses a challenge due to the low abundance of EV-biomarkers. Here, we present a fully integrated organic electrochemical transistor (OECT) that enables high accuracy, speed, and convenience in the detection of EVs from AD patients. We incorporated self-aligned acoustoelectric enhancement of EVs on a chip that rapidly propels, enriches, and specifically binds EVs to the OECT detection area. With our enhancement of pre-concentration, we increased the sensitivity to a limit of detection of 500 EV particles/μL and reduced the required detection time to just two minutes. We also tested the sensor on an AD mouse model to monitor AD progression, examined mouse Aβ EVs at different time courses, and compared them with intraneuronal Aβ cumulation using MRI. This innovative technology has the potential to diagnose Alzheimer's and other neurodegenerative diseases accurately and quickly, enabling monitoring of disease progression and treatment response.
Collapse
|
10
|
Chang J, Feng Z, Li Y, Lv H, Liu S, Luo Y, Hao N, Zhao L, Liu J. Mesenchymal stem cell-derived extracellular vesicles: A novel promising neuroprotective agent for Alzheimer's disease. Biochem Pharmacol 2024; 222:116064. [PMID: 38373595 DOI: 10.1016/j.bcp.2024.116064] [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: 11/27/2023] [Revised: 01/25/2024] [Accepted: 02/16/2024] [Indexed: 02/21/2024]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disease characterized by progressive loss of neurons in the brain. However, there are no effective drugs for AD. Mesenchymal stem cell-derived extracellular vesicles (MSCs-EVs), as a new mediator of intercellular communication, are associated with low immunogenicity, low risk of tumor formation, and good safety profile. Therefore, MSCs-EVs may be a safe and attractive cell-free nanotherapeutics, offering a new perspective for AD treatment. Although preclinical studies have demonstrated that MSCs-EVs have significant neuroprotective effects, the underlying mechanism is unclear. This study aimed to: outline the diagnostic and delivery roles of MSCs-EVs for AD treatment; summarize the optimal sources and delivery methods of MSCs-EVs; provide a comprehensive review on the neuroprotective mechanisms of MSCs-EVs; explore how to enhance the neuroprotective effects of MSCs-EVs; and discuss the limitations and potential of their translation to the clinic. Therefore, this study may provide a more precise theoretical reference and practical basis for clinical research of MSCs-EVs.
Collapse
Affiliation(s)
- Jun Chang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Zihang Feng
- School of Nursing, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yujiao Li
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Honglin Lv
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Shuzhen Liu
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Yongyin Luo
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Nan Hao
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Lan Zhao
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China.
| | - Jianwei Liu
- Tianjin University of Traditional Chinese Medicine, Tianjin, China.
| |
Collapse
|
11
|
Kumar A, Nader MA, Deep G. Emergence of Extracellular Vesicles as "Liquid Biopsy" for Neurological Disorders: Boom or Bust. Pharmacol Rev 2024; 76:199-227. [PMID: 38351075 PMCID: PMC10877757 DOI: 10.1124/pharmrev.122.000788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 11/11/2023] [Accepted: 11/27/2023] [Indexed: 02/16/2024] Open
Abstract
Extracellular vesicles (EVs) have emerged as an attractive liquid biopsy approach in the diagnosis and prognosis of multiple diseases and disorders. The feasibility of enriching specific subpopulations of EVs from biofluids based on their unique surface markers has opened novel opportunities to gain molecular insight from various tissues and organs, including the brain. Over the past decade, EVs in bodily fluids have been extensively studied for biomarkers associated with various neurological disorders, such as Alzheimer's disease, Parkinson's disease, schizophrenia, bipolar disorder, major depressive disorders, substance use disorders, human immunodeficiency virus-associated neurocognitive disorder, and cancer/treatment-induced neurodegeneration. These studies have focused on the isolation and cargo characterization of either total EVs or brain cells, such as neuron-, astrocyte-, microglia-, oligodendrocyte-, pericyte-, and endothelial-derived EVs from biofluids to achieve early diagnosis and molecular characterization and to predict the treatment and intervention outcomes. The findings of these studies have demonstrated that EVs could serve as a repetitive and less invasive source of valuable molecular information for these neurological disorders, supplementing existing costly neuroimaging techniques and relatively invasive measures, like lumbar puncture. However, the initial excitement surrounding blood-based biomarkers for brain-related diseases has been tempered by challenges, such as lack of central nervous system specificity in EV markers, lengthy protocols, and the absence of standardized procedures for biological sample collection, EV isolation, and characterization. Nevertheless, with rapid advancements in the EV field, supported by improved isolation methods and sensitive assays for cargo characterization, brain cell-derived EVs continue to offer unparallel opportunities with significant translational implications for various neurological disorders. SIGNIFICANCE STATEMENT: Extracellular vesicles present a less invasive liquid biopsy approach in the diagnosis and prognosis of various neurological disorders. Characterizing these vesicles in biofluids holds the potential to yield valuable molecular information, thereby significantly impacting the development of novel biomarkers for various neurological disorders. This paper has reviewed the methodology employed to isolate extracellular vesicles derived from various brain cells in biofluids, their utility in enhancing the molecular understanding of neurodegeneration, and the potential challenges in this research field.
Collapse
Affiliation(s)
- Ashish Kumar
- Departments of Cancer Biology (A.K., G.D.), Physiology and Pharmacology (M.A.N.), Radiology (M.A.N.), and Center for Addiction Research (M.A.N., G.D.), Wake Forest University School of Medicine, Winston-Salem, North Carolina; Atrium Health Wake Forest Baptist Comprehensive Cancer Center, Winston-Salem, North Carolina (G.D.); and Sticht Center for Healthy Aging and Alzheimer's Prevention, Wake Forest School of Medicine, Winston-Salem, North Carolina (G.D.)
| | - Michael A Nader
- Departments of Cancer Biology (A.K., G.D.), Physiology and Pharmacology (M.A.N.), Radiology (M.A.N.), and Center for Addiction Research (M.A.N., G.D.), Wake Forest University School of Medicine, Winston-Salem, North Carolina; Atrium Health Wake Forest Baptist Comprehensive Cancer Center, Winston-Salem, North Carolina (G.D.); and Sticht Center for Healthy Aging and Alzheimer's Prevention, Wake Forest School of Medicine, Winston-Salem, North Carolina (G.D.)
| | - Gagan Deep
- Departments of Cancer Biology (A.K., G.D.), Physiology and Pharmacology (M.A.N.), Radiology (M.A.N.), and Center for Addiction Research (M.A.N., G.D.), Wake Forest University School of Medicine, Winston-Salem, North Carolina; Atrium Health Wake Forest Baptist Comprehensive Cancer Center, Winston-Salem, North Carolina (G.D.); and Sticht Center for Healthy Aging and Alzheimer's Prevention, Wake Forest School of Medicine, Winston-Salem, North Carolina (G.D.)
| |
Collapse
|
12
|
Manolopoulos A, Delgado-Peraza F, Mustapic M, Pucha KA, Nogueras-Ortiz C, Daskalopoulos A, Knight DD, Leoutsakos JM, Oh ES, Lyketsos CG, Kapogiannis D. Comparative assessment of Alzheimer's disease-related biomarkers in plasma and neuron-derived extracellular vesicles: a nested case-control study. Front Mol Biosci 2023; 10:1254834. [PMID: 37828917 PMCID: PMC10565036 DOI: 10.3389/fmolb.2023.1254834] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 09/12/2023] [Indexed: 10/14/2023] Open
Abstract
Introduction: Alzheimer's disease (AD) is currently defined according to biomarkers reflecting the core underlying neuropathological processes: Aβ deposition, Tau, and neurodegeneration (ATN). The soluble phase of plasma and plasma neuron-derived extracellular vesicles (NDEVs) are increasingly being investigated as sources of biomarkers. The aim of this study was to examine the comparative biomarker potential of these two biofluids, as well as the association between respective biomarkers. Methods: We retrospectively identified three distinct diagnostic groups of 44 individuals who provided samples at baseline and at a mean of 3.1 years later; 14 were cognitively unimpaired at baseline and remained so (NRM-NRM), 13 had amnestic MCI that progressed to AD dementia (MCI-DEM) and 17 had AD dementia at both timepoints (DEM-DEM). Plasma NDEVs were isolated by immunoaffinity capture targeting the neuronal markers L1CAM, GAP43, and NLGN3. In both plasma and NDEVs, we assessed ATN biomarkers (Aβ42, Aβ40, total Tau, P181-Tau) alongside several other exploratory markers. Results: The Aβ42/Aβ40 ratio in plasma and NDEVs was lower in MCI-DEM than NRM-NRM at baseline and its levels in NDEVs decreased over time in all three groups. Similarly, plasma and NDEV-associated Aβ42 was lower in MCI-DEM compared to NRM-NRM at baseline and its levels in plasma decreased over time in DEM-DEM. For NDEV-associated proBDNF, compared to NRM-NRM, its levels were lower in MCI-DEM and DEM-DEM at baseline, and they decreased over time in the latter group. No group differences were found for other exploratory markers. NDEV-associated Aβ42/Aβ40 ratio and proBDNF achieved the highest areas under the curve (AUCs) for discriminating between diagnostic groups, while proBDNF was positively associated with Mini-Mental State Examination (MMSE) score. No associations were found between the two biofluids for any assessed marker. Discussion: The soluble phase of plasma and plasma NDEVs demonstrate distinct biomarker profiles both at a single time point and longitudinally. The lack of association between plasma and NDEV measures indicates that the two types of biofluids demonstrate distinct biomarker signatures that may be attributable to being derived through different biological processes. NDEV-associated proBDNF may be a useful biomarker for AD diagnosis and monitoring.
Collapse
Affiliation(s)
- Apostolos Manolopoulos
- Intramural Research Program, Laboratory of Clinical Investigation, National Institute on Aging, Baltimore, MD, United States
| | - Francheska Delgado-Peraza
- Intramural Research Program, Laboratory of Clinical Investigation, National Institute on Aging, Baltimore, MD, United States
| | - Maja Mustapic
- Intramural Research Program, Laboratory of Clinical Investigation, National Institute on Aging, Baltimore, MD, United States
| | - Krishna Ananthu Pucha
- Intramural Research Program, Laboratory of Clinical Investigation, National Institute on Aging, Baltimore, MD, United States
| | - Carlos Nogueras-Ortiz
- Intramural Research Program, Laboratory of Clinical Investigation, National Institute on Aging, Baltimore, MD, United States
| | - Alexander Daskalopoulos
- Intramural Research Program, Laboratory of Clinical Investigation, National Institute on Aging, Baltimore, MD, United States
| | - De’Larrian DeAnté Knight
- Intramural Research Program, Laboratory of Clinical Investigation, National Institute on Aging, Baltimore, MD, United States
| | - Jeannie-Marie Leoutsakos
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD, United States
- Richman Family Precision Medicine Center of Excellence in Alzheimer’s Disease, Johns Hopkins School of Medicine, Baltimore, MD, United States
| | - Esther S. Oh
- Richman Family Precision Medicine Center of Excellence in Alzheimer’s Disease, Johns Hopkins School of Medicine, Baltimore, MD, United States
- Division of Geriatric Medicine and Gerontology, Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD, United States
| | - Constantine G. Lyketsos
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD, United States
- Richman Family Precision Medicine Center of Excellence in Alzheimer’s Disease, Johns Hopkins School of Medicine, Baltimore, MD, United States
| | - Dimitrios Kapogiannis
- Intramural Research Program, Laboratory of Clinical Investigation, National Institute on Aging, Baltimore, MD, United States
| |
Collapse
|
13
|
Stajano D, Lombino FL, Schweizer M, Glatzel M, Saftig P, Gromova KV, Kneussel M. Tetraspanin 15 depletion impairs extracellular vesicle docking at target neurons. JOURNAL OF EXTRACELLULAR BIOLOGY 2023; 2:e113. [PMID: 38938373 PMCID: PMC11080857 DOI: 10.1002/jex2.113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 08/11/2023] [Accepted: 08/30/2023] [Indexed: 06/29/2024]
Abstract
Neurons in the central nervous system release extracellular vesicles (EVs) and exosomes in response to synaptic activity to regulate physiological processes at target neurons. The intercellular transfer of proteins, mRNAs, lipids or metabolites through EVs potentially modulates the structure and function of neurons and circuits. Whereas the biogenesis of EVs, their release from donor cells, and their molecular composition have been studied extensively, the critical factors and mechanisms regulating EV interactions with target cells are incompletely understood. Here, we identified tetraspanin 15 (Tspan15) as a component of tumor susceptibility gene 101 protein (TSG101)- and CD81-positive EV fractions. Tspan15 fluorescent fusion proteins were released from donor cells and interacted with target cells together with the exosomal marker CD63. EVs collected from wildtype cortical neurons (WT-EVs) underwent similar association with target neurons derived from either wildtype (+/+) or Tspan15 knockout (-/-) mice. In contrast, target cell interactions of EVs collected from Tspan15 (-/-) cortical donor neurons (KO-EVs) were significantly impaired, as compared to WT-EVs. Our data suggest that Tspan15 is dispensable at target neuron plasma membranes, but is required at the EV surface to promote EV docking at target neurons.
Collapse
Affiliation(s)
- Daniele Stajano
- Institute of Molecular Neurogenetics, Center for Molecular Neurobiology, ZMNHUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| | - Franco L. Lombino
- Institute of Molecular Neurogenetics, Center for Molecular Neurobiology, ZMNHUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| | - Michaela Schweizer
- Core Facility Morphology, Center for Molecular Neurobiology, ZMNHUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| | - Markus Glatzel
- Institute of NeuropathologyUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| | - Paul Saftig
- Biochemical InstituteChristian‐Albrechts‐University KielKielGermany
| | - Kira V. Gromova
- Institute of Molecular Neurogenetics, Center for Molecular Neurobiology, ZMNHUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| | - Matthias Kneussel
- Institute of Molecular Neurogenetics, Center for Molecular Neurobiology, ZMNHUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| |
Collapse
|
14
|
Jiang C, Zhang J, Wang W, Shan Z, Sun F, Tan Y, Tong Y, Qiu Y. Extracellular vesicles in gastric cancer: role of exosomal lncRNA and microRNA as diagnostic and therapeutic targets. Front Physiol 2023; 14:1158839. [PMID: 37664422 PMCID: PMC10469264 DOI: 10.3389/fphys.2023.1158839] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Accepted: 07/31/2023] [Indexed: 09/05/2023] Open
Abstract
Extracellular vesicles (EVs), including exosomes, play a crucial role in intercellular communication and have emerged as important mediators in the development and progression of gastric cancer. This review discusses the current understanding of the role of EVs, particularly exosomal lncRNA and microRNA, in gastric cancer and their potential as diagnostic and therapeutic targets. Exosomes are small membrane-bound particles secreted by both cancer cells and stromal cells within the tumor microenvironment. They contain various ncRNA and biomolecules, which can be transferred to recipient cells to promote tumor growth and metastasis. In this review, we highlighted the importance of exosomal lncRNA and microRNA in gastric cancer. Exosomal lncRNAs have been shown to regulate gene expression by interacting with transcription factors or chromatin-modifying enzymes, which regulate gene expression by binding to target mRNAs. We also discuss the potential use of exosomal lncRNAs and microRNAs as diagnostic biomarkers for gastric cancer. Exosomes can be isolated from various bodily fluids, including blood, urine, and saliva. They contain specific molecules that reflect the molecular characteristics of the tumor, making them promising candidates for non-invasive diagnostic tests. Finally, the potential of targeting exosomal lncRNAs and microRNAs as a therapeutic strategy for gastric cancer were reviewed as wee. Inhibition of specific molecules within exosomes has been shown to suppress tumor growth and metastasis in preclinical models. In conclusion, this review article provides an overview of the current understanding of the role of exosomal lncRNA and microRNA in gastric cancer. We suggest that further research into these molecules could lead to new diagnostic tools and therapeutic strategies for this deadly disease.
Collapse
Affiliation(s)
- Chengyao Jiang
- Department of Gastric Surgery, Liaoning Cancer Hospital and Institute, Cancer Hospital of China Medical University, Shenyang, China
| | - Jianjun Zhang
- Department of Gastric Surgery, Liaoning Cancer Hospital and Institute, Cancer Hospital of China Medical University, Shenyang, China
| | - Wentao Wang
- Department of Gastric Surgery, Liaoning Cancer Hospital and Institute, Cancer Hospital of China Medical University, Shenyang, China
| | - Zexing Shan
- Department of Gastric Surgery, Liaoning Cancer Hospital and Institute, Cancer Hospital of China Medical University, Shenyang, China
| | - Fan Sun
- Department of Gastric Surgery, Liaoning Cancer Hospital and Institute, Cancer Hospital of China Medical University, Shenyang, China
| | - Yuen Tan
- Department of Gastric Surgery, Liaoning Cancer Hospital and Institute, Cancer Hospital of China Medical University, Shenyang, China
| | - Yilin Tong
- Department of Gastric Surgery, Liaoning Cancer Hospital and Institute, Cancer Hospital of China Medical University, Shenyang, China
| | - Yue Qiu
- Medical Oncology Department of Gastrointestinal Cancer, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, Liaoning, China
| |
Collapse
|
15
|
Nicolazzo JA, Abasszade JH. From the gut to the blood-brain barrier: novel approaches for the treatment and targeted delivery of therapeutics in Alzheimer's disease. Adv Drug Deliv Rev 2023; 196:114818. [PMID: 37003489 DOI: 10.1016/j.addr.2023.114818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/03/2023]
Affiliation(s)
- Joseph A Nicolazzo
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia.
| | | |
Collapse
|
16
|
Yin T, Liu Y, Ji W, Zhuang J, Chen X, Gong B, Chu J, Liang W, Gao J, Yin Y. Engineered mesenchymal stem cell-derived extracellular vesicles: A state-of-the-art multifunctional weapon against Alzheimer's disease. Theranostics 2023; 13:1264-1285. [PMID: 36923533 PMCID: PMC10008732 DOI: 10.7150/thno.81860] [Citation(s) in RCA: 22] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 01/21/2023] [Indexed: 02/15/2023] Open
Abstract
With the increase of population aging, the number of Alzheimer's disease (AD) patients is also increasing. According to current estimates, approximately 11% of people over 65 suffer from AD, and that percentage rises to 42% among people over 85. However, no effective treatment capable of decelerating or stopping AD progression is available. Furthermore, AD-targeted drugs composed of synthetic molecules pose concerns regarding biodegradation, clearance, immune response, and neurotoxicity. Mesenchymal stem cell-derived extracellular vesicles (MSC-EVs) are essential intercellular communication mediators holding great promise as AD therapeutics owing to their biocompatibility, versatility, effortless storage, superior safety, and the ability to transport messenger and noncoding RNAs, proteins, lipids, DNAs, and other bioactive compounds derived from cells. The functionalisation and engineering strategies of MSC-EVs are highlighted (e.g. preconditioning, drug loading, surface modification, and artificial EV fabrication), which could improve AD treatment by multiple therapeutic effects, including clearing abnormal protein accumulation and achieving neuroprotection and immunomodulatory effects. Herein, this review summarises state-of-the-art strategies to engineer MSC-EVs, discusses progress in their use as AD therapeutics, presents the perspectives and challenges associated with the related clinical applications, and concludes that engineered MSC-EVs show immense potential in AD therapy.
Collapse
Affiliation(s)
- Tong Yin
- Department of Neurology, Second Affiliated Hospital of Naval Medical University (Shanghai Changzheng Hospital), Shanghai 200003, China
| | - Yan Liu
- Department of Clinical Pharmacy, Xinhua Hospital; Clinical pharmacy innovation institute, Shanghai Jiao Tong University of Medicine, Shanghai 200000, China
| | - Wenbo Ji
- Department of Neurology, Second Affiliated Hospital of Naval Medical University (Shanghai Changzheng Hospital), Shanghai 200003, China
- Changhai Clinical Research Unit, Shanghai Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - Jianhua Zhuang
- Department of Neurology, Second Affiliated Hospital of Naval Medical University (Shanghai Changzheng Hospital), Shanghai 200003, China
| | - Xiaohan Chen
- Department of Neurology, Second Affiliated Hospital of Naval Medical University (Shanghai Changzheng Hospital), Shanghai 200003, China
| | - Baofeng Gong
- Department of Neurology, Second Affiliated Hospital of Naval Medical University (Shanghai Changzheng Hospital), Shanghai 200003, China
| | - Jianjian Chu
- Department of Neurology, Second Affiliated Hospital of Naval Medical University (Shanghai Changzheng Hospital), Shanghai 200003, China
| | - Wendanqi Liang
- Department of Neurology, Second Affiliated Hospital of Naval Medical University (Shanghai Changzheng Hospital), Shanghai 200003, China
| | - Jie Gao
- Changhai Clinical Research Unit, Shanghai Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - You Yin
- Department of Neurology, Second Affiliated Hospital of Naval Medical University (Shanghai Changzheng Hospital), Shanghai 200003, China
| |
Collapse
|
17
|
Burgelman M, Dujardin P, Vandendriessche C, Vandenbroucke RE. Free complement and complement containing extracellular vesicles as potential biomarkers for neuroinflammatory and neurodegenerative disorders. Front Immunol 2023; 13:1055050. [PMID: 36741417 PMCID: PMC9896008 DOI: 10.3389/fimmu.2022.1055050] [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: 09/27/2022] [Accepted: 12/07/2022] [Indexed: 01/21/2023] Open
Abstract
The complement system is implicated in a broad range of neuroinflammatory disorders such as Alzheimer's disease (AD) and multiple sclerosis (MS). Consequently, measuring complement levels in biofluids could serve as a potential biomarker for these diseases. Indeed, complement levels are shown to be altered in patients compared to controls, and some studies reported a correlation between the level of free complement in biofluids and disease progression, severity or the response to therapeutics. Overall, they are not (yet) suitable as a diagnostic tool due to heterogeneity of reported results. Moreover, measurement of free complement proteins has the disadvantage that information on their origin is lost, which might be of value in a multi-parameter approach for disease prediction and stratification. In light of this, extracellular vesicles (EVs) could provide a platform to improve the diagnostic power of complement proteins. EVs are nanosized double membrane particles that are secreted by essentially every cell type and resemble the (status of the) cell of origin. Interestingly, EVs can contain complement proteins, while the cellular origin can still be determined by the presence of EV surface markers. In this review, we summarize the current knowledge and future opportunities on the use of free and EV-associated complement proteins as biomarkers for neuroinflammatory and neurodegenerative disorders.
Collapse
Affiliation(s)
- Marlies Burgelman
- VIB Center for Inflammation Research, VIB, Ghent, Belgium,Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Pieter Dujardin
- VIB Center for Inflammation Research, VIB, Ghent, Belgium,Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Charysse Vandendriessche
- VIB Center for Inflammation Research, VIB, Ghent, Belgium,Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Roosmarijn E. Vandenbroucke
- VIB Center for Inflammation Research, VIB, Ghent, Belgium,Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium,*Correspondence: Roosmarijn E. Vandenbroucke,
| |
Collapse
|
18
|
Schiavello M, Vizio B, Bosco O, Pivetta E, Mariano F, Montrucchio G, Lupia E. Extracellular Vesicles: New Players in the Mechanisms of Sepsis- and COVID-19-Related Thromboinflammation. Int J Mol Sci 2023; 24:ijms24031920. [PMID: 36768242 PMCID: PMC9916541 DOI: 10.3390/ijms24031920] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 01/11/2023] [Accepted: 01/13/2023] [Indexed: 01/21/2023] Open
Abstract
Sepsis and COVID-19 patients often manifest an imbalance in inflammation and coagulation, a complex pathological mechanism also named thromboinflammation, which strongly affects patient prognosis. Extracellular vesicles (EVs) are nanoparticles released by cells into extracellular space that have a relevant role in cell-to-cell communication. Recently, EVs have been shown to act as important players in a variety of pathologies, including cancer and cardiovascular disease. The biological properties of EVs in the mechanisms of thromboinflammation during sepsis and COVID-19 are still only partially known. Herein, we summarize the current experimental evidence on the role of EVs in thromboinflammation, both in bacterial sepsis and in COVID-19. A better understanding of EV involvement in these processes could be useful in describing novel diagnostic and therapeutic applications of EVs in these diseases.
Collapse
|
19
|
Li X, Hsu JC, Son MH, Ha LN, Cai W. Cancer photodynamic therapy with chlorin e6-loaded, goat milk-derived extracellular vesicles: [ 18F]FDG lights up the way. Eur J Nucl Med Mol Imaging 2023; 50:247-250. [PMID: 36357594 PMCID: PMC9822859 DOI: 10.1007/s00259-022-06031-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Affiliation(s)
- Xiaoyan Li
- Departments of Radiology and Medical Physics, University of WI - Madison, Madison, WI, USA
| | - Jessica C Hsu
- Departments of Radiology and Medical Physics, University of WI - Madison, Madison, WI, USA
| | - Mai Hong Son
- Department of Nuclear Medicine, Hospital 108, Hanoi, Vietnam
| | - Le Ngoc Ha
- Department of Nuclear Medicine, Hospital 108, Hanoi, Vietnam
| | - Weibo Cai
- Departments of Radiology and Medical Physics, University of WI - Madison, Madison, WI, USA.
| |
Collapse
|
20
|
Emerging Roles of Extracellular Vesicles in Alzheimer's Disease: Focus on Synaptic Dysfunction and Vesicle-Neuron Interaction. Cells 2022; 12:cells12010063. [PMID: 36611856 PMCID: PMC9818402 DOI: 10.3390/cells12010063] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 12/16/2022] [Accepted: 12/20/2022] [Indexed: 12/28/2022] Open
Abstract
Alzheimer's disease (AD) is considered by many to be a synaptic failure. Synaptic function is in fact deeply affected in the very early disease phases and recognized as the main cause of AD-related cognitive impairment. While the reciprocal involvement of amyloid beta (Aβ) and tau peptides in these processes is under intense investigation, the crucial role of extracellular vesicles (EVs) released by different brain cells as vehicles for these molecules and as mediators of early synaptic alterations is gaining more and more ground in the field. In this review, we will summarize the current literature on the contribution of EVs derived from distinct brain cells to neuronal alterations and build a working model for EV-mediated propagation of synaptic dysfunction in early AD. A deeper understanding of EV-neuron interaction will provide useful targets for the development of novel therapeutic approaches aimed at hampering AD progression.
Collapse
|
21
|
Wang B, Liu W, Sun F. Nucleosome assembly protein 1-like 5 alleviates Alzheimer's disease-like pathological characteristics in a cell model. Front Mol Neurosci 2022; 15:1034766. [PMID: 36568274 PMCID: PMC9773259 DOI: 10.3389/fnmol.2022.1034766] [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: 09/02/2022] [Accepted: 11/24/2022] [Indexed: 12/14/2022] Open
Abstract
Alzheimer's disease (AD) remains one of the most common dementias of neurodegenerative disease-related diseases. Nucleosome assembly protein 1-like 5 (NAP1L5) belongs to the NAP1L protein family, which acts as a histone chaperone. However, the function and mechanism of NAP1L5 in AD are still unclear. Bioinformatics analysis, RT-qPCR, and Western blotting results showed that NAP1L5 was downregulated in the brain tissues of AD patients and a mouse cell model of AD. NAP1L5 overexpression alleviated (Amyloid-β precursor protein) APP metabolism and Tau phosphorylation. We further demonstrated that NAP1L5 regulated the AD-like pathological characteristics through the GSK3B/Wnt/β-Catenin signaling pathway. Moreover, we showed that the Wnt/β-Catenin signaling pathway, regulated by NAP1L5, was mediated by AQP1-mediated mechanism in N2a-APP695sw cell. In sum, these results suggested that NAP1L5 overexpression has neuroprotective effects and might act as potential biomarker and target for the diagnosis and treatment of AD.
Collapse
Affiliation(s)
- Bingyan Wang
- Department of Anesthesiology, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Weiying Liu
- Department of Pathogen Biology, School of Basic Medicine, Tianjin Medical University, Tianjin, China,*Correspondence: Weiying Liu,
| | - Fengxian Sun
- Department of Physiology and Pathophysiology, School of Basic Medicine, Tianjin Medical University, Tianjin, China,Fengxian Sun,
| |
Collapse
|