1
|
Németh K, Kestecher BM, Ghosal S, Bodnár BR, Kittel Á, Hambalkó S, Kovácsházi C, Giricz Z, Ferdinandy P, Osteikoetxea X, Burkhardt R, Buzas EI, Orsó E. Therapeutic and pharmacological applications of extracellular vesicles and lipoproteins. Br J Pharmacol 2024; 181:4733-4749. [PMID: 39491825 DOI: 10.1111/bph.17336] [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: 04/12/2024] [Revised: 06/15/2024] [Accepted: 07/13/2024] [Indexed: 11/05/2024] Open
Abstract
In recent years, various approaches have been undertaken to eliminate lipoproteins co-isolated with extracellular vesicles, as they were initially regarded as contaminating entities. However, novel discoveries are reshaping our perspective. In body fluids, these distinct particles not only co-exist, but also interactions between them are likely to occur. Extracellular vesicles and lipoproteins can associate with each other, share cargo, influence each other's functions, and jointly have a role in the pathomechanisms of diseases. Additionally, their association carries important implications for therapeutic and pharmacological aspects of lipid-lowering strategies. Extracellular vesicles and lipoprotein particles may have roles in the elimination of each other from the circulation. The objective of this minireview is to delve into these aspects. Here, we show that under certain physiological and pathological conditions, extracellular vesicles and lipoproteins are 'partners' rather than 'strangers' or 'rivals'.
Collapse
Affiliation(s)
- Krisztina Németh
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, Budapest, Hungary
- HUN-REN-SU Translational Extracellular Vesicle Research Group, Budapest, Hungary
| | - Brachyahu M Kestecher
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, Budapest, Hungary
- HUN-REN-SU Translational Extracellular Vesicle Research Group, Budapest, Hungary
- HCEMM-SU Extracellular Vesicle Research Group, Budapest, Hungary
| | - Sayam Ghosal
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, Budapest, Hungary
- HCEMM-SU Extracellular Vesicle Research Group, Budapest, Hungary
| | - Bernadett R Bodnár
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, Budapest, Hungary
- HCEMM-SU Extracellular Vesicle Research Group, Budapest, Hungary
| | - Ágnes Kittel
- HUN-REN-SU Translational Extracellular Vesicle Research Group, Budapest, Hungary
- HUN-REN, Institute of Experimental Medicine, Budapest, Hungary
| | - Szabolcs Hambalkó
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
| | - Csenger Kovácsházi
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
| | - Zoltán Giricz
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
- Pharmahungary Group, Szeged, Hungary
| | - Péter Ferdinandy
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
- Pharmahungary Group, Szeged, Hungary
| | - Xabier Osteikoetxea
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, Budapest, Hungary
- HCEMM-SU Extracellular Vesicle Research Group, Budapest, Hungary
| | - Ralph Burkhardt
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital Regensburg, Regensburg, Germany
| | - Edit I Buzas
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, Budapest, Hungary
- HUN-REN-SU Translational Extracellular Vesicle Research Group, Budapest, Hungary
- HCEMM-SU Extracellular Vesicle Research Group, Budapest, Hungary
| | - Evelyn Orsó
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, Budapest, Hungary
| |
Collapse
|
2
|
Wardhani K, Levina A, Grau GER, Lay PA. Fluorescent, phosphorescent, magnetic resonance contrast and radioactive tracer labelling of extracellular vesicles. Chem Soc Rev 2024; 53:6779-6829. [PMID: 38828885 DOI: 10.1039/d2cs00238h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/05/2024]
Abstract
This review focusses on the significance of fluorescent, phosphorescent labelling and tracking of extracellular vesicles (EVs) for unravelling their biology, pathophysiology, and potential diagnostic and therapeutic uses. Various labeling strategies, such as lipid membrane, surface protein, luminal, nucleic acid, radionuclide, quantum dot labels, and metal complex-based stains, are evaluated for visualizing and characterizing EVs. Direct labelling with fluorescent lipophilic dyes is simple but generally lacks specificity, while surface protein labelling offers selectivity but may affect EV-cell interactions. Luminal and nucleic acid labelling strategies have their own advantages and challenges. Each labelling approach has strengths and weaknesses, which require a suitable probe and technique based on research goals, but new tetranuclear polypyridylruthenium(II) complexes as phosphorescent probes have strong phosphorescence, selective staining, and stability. Future research should prioritize the design of novel fluorescent probes and labelling platforms that can significantly enhance the efficiency, accuracy, and specificity of EV labeling, while preserving their composition and functionality. It is crucial to reduce false positive signals and explore the potential of multimodal imaging techniques to gain comprehensive insights into EVs.
Collapse
Affiliation(s)
- Kartika Wardhani
- School of Chemistry, The University of Sydney, Sydney, New South Wales, 2006, Australia.
- Biochemistry and Biotechnology (B-TEK) Group, Bioscience Division, Los Alamos National Laboratory, Los Alamos, New Mexico, 87545, USA
| | - Aviva Levina
- School of Chemistry, The University of Sydney, Sydney, New South Wales, 2006, Australia.
| | - Georges E R Grau
- Sydney Nano, The University of Sydney, Sydney, New South Wales, 2006, Australia
- Sydney Cancer Network, The University of Sydney, Sydney, New South Wales, 2006, Australia
- Marie Bashir Institute, The University of Sydney, Sydney, New South Wales, 2006, Australia
- Vascular Immunology Unit, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, 2006, Australia
| | - Peter A Lay
- School of Chemistry, The University of Sydney, Sydney, New South Wales, 2006, Australia.
- Sydney Nano, The University of Sydney, Sydney, New South Wales, 2006, Australia
- Sydney Cancer Network, The University of Sydney, Sydney, New South Wales, 2006, Australia
- Marie Bashir Institute, The University of Sydney, Sydney, New South Wales, 2006, Australia
- Sydney Analytical, The University of Sydney, Sydney, New South Wales, 2006, Australia
| |
Collapse
|
3
|
Jorfi S, Ansa-Addo EA, Mariniello K, Warde P, Bin Senian AA, Stratton D, Bax BE, Levene M, Lange S, Inal JM. A Coxsackievirus B1-mediated nonlytic Extracellular Vesicle-to-cell mechanism of virus transmission and its possible control through modulation of EV release. J Gen Virol 2023; 104. [PMID: 37665326 DOI: 10.1099/jgv.0.001884] [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] [Indexed: 09/05/2023] Open
Abstract
Like most non-enveloped viruses, CVB1 mainly uses cell lysis to spread. Details of a nonlytic virus transmission remain unclear. Extracellular Vesicles (EVs) transfer biomolecules between cells. We show that CVB1 entry into HeLa cells results in apoptosis and release of CVB1-induced 'medium-sized' EVs (CVB1i-mEVs). These mEVs (100-300 nm) harbour CVB1 as shown by immunoblotting with anti-CVB1-antibody; viral capsids were detected by transmission electron microscopy and RT-PCR revealed CVB1 RNA. The percentage of mEVs released from CVB1-infected HeLa cells harbouring virus was estimated from TEM at 34 %. Inhibition of CVB1i-mEV production, with calpeptin or siRNA knockdown of CAPNS1 in HeLa cells limited spread of CVB1 suggesting these vesicles disseminate CVB1 virions to new host cells by a nonlytic EV-to-cell mechanism. This was confirmed by detecting CVB1 virions inside HeLa cells after co-culture with CVB1i-mEVs; EV release may also prevent apoptosis of infected cells whilst spreading apoptosis to secondary sites of infection.
Collapse
Affiliation(s)
- Samireh Jorfi
- Cell Communication in Disease Pathology, School of Human Sciences, London Metropolitan University, London N7 8DB, UK
| | - Ephraim Abrokwa Ansa-Addo
- Cell Communication in Disease Pathology, School of Human Sciences, London Metropolitan University, London N7 8DB, UK
- Present address: Pelotonia Institute for Immuno-Oncology, The James, Ohio State University, Columbus, OH 43210, USA
| | - Katia Mariniello
- Cell Communication in Disease Pathology, School of Human Sciences, London Metropolitan University, London N7 8DB, UK
- Present address: William Harvey Research Institute, Queen Mary, University of London, London, UK
| | - Purva Warde
- Biosciences Research Group, School of Life and Medical Sciences, University of Hertfordshire, Hatfield AL10 9EU, UK
| | - Ahmad Asyraf Bin Senian
- Biosciences Research Group, School of Life and Medical Sciences, University of Hertfordshire, Hatfield AL10 9EU, UK
- Present address: Clinical Research Centre, Sarawak General Hospital, Kuching, Malaysia
| | - Dan Stratton
- School of Life, Health & Chemical Sciences, The Open University, Milton Keynes MK7 6AE, UK
| | - Bridget E Bax
- Molecular and Clinical Sciences Research Institute, St. George's, University of London, London SW17 0RE, UK
| | - Michelle Levene
- Molecular and Clinical Sciences Research Institute, St. George's, University of London, London SW17 0RE, UK
| | - Sigrun Lange
- Tissue Architecture and Regeneration Research Group, School of Life Sciences, University of Westminster, 116, New Cavendish St., London, UK
- University College London School of Pharmacy, Brunswick Sq., London, UK
| | - Jameel Malhador Inal
- Cell Communication in Disease Pathology, School of Human Sciences, London Metropolitan University, London N7 8DB, UK
- Biosciences Research Group, School of Life and Medical Sciences, University of Hertfordshire, Hatfield AL10 9EU, UK
| |
Collapse
|
4
|
Kumar P, Mehta D, Bissler JJ. Physiologically Based Pharmacokinetic Modeling of Extracellular Vesicles. BIOLOGY 2023; 12:1178. [PMID: 37759578 PMCID: PMC10525702 DOI: 10.3390/biology12091178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 08/13/2023] [Accepted: 08/22/2023] [Indexed: 09/29/2023]
Abstract
Extracellular vesicles (EVs) are lipid membrane bound-cell-derived structures that are a key player in intercellular communication and facilitate numerous cellular functions such as tumor growth, metastasis, immunosuppression, and angiogenesis. They can be used as a drug delivery platform because they can protect drugs from degradation and target specific cells or tissues. With the advancement in the technologies and methods in EV research, EV-therapeutics are one of the fast-growing domains in the human health sector. Therapeutic translation of EVs in clinics requires assessing the quality, safety, and efficacy of the EVs, in which pharmacokinetics is very crucial. We report here the application of physiologically based pharmacokinetic (PBPK) modeling as a principal tool for the prediction of absorption, distribution, metabolism, and excretion of EVs. To create a PBPK model of EVs, researchers would need to gather data on the size, shape, and composition of the EVs, as well as the physiological processes that affect their behavior in the body. The PBPK model would then be used to predict the pharmacokinetics of drugs delivered via EVs, such as the rate at which the drug is absorbed and distributed throughout the body, the rate at which it is metabolized and eliminated, and the maximum concentration of the drug in the body. This information can be used to optimize the design of EV-based drug delivery systems, including the size and composition of the EVs, the route of administration, and the dose of the drug. There has not been any dedicated review article that describes the PBPK modeling of EV. This review provides an overview of the absorption, distribution, metabolism, and excretion (ADME) phenomena of EVs. In addition, we will briefly describe the different computer-based modeling approaches that may help in the future of EV-based therapeutic research.
Collapse
Affiliation(s)
- Prashant Kumar
- Division of Biochemical Toxicology, National Center for Toxicological Research, United States Food and Drug Administration, Jefferson, AR 72079, USA;
| | - Darshan Mehta
- Division of Biochemical Toxicology, National Center for Toxicological Research, United States Food and Drug Administration, Jefferson, AR 72079, USA;
| | - John J. Bissler
- Department of Pediatrics, Division of Pediatrics Nephrology, University of Tennessee Health Science Center, Memphis, TN 38103, USA;
| |
Collapse
|
5
|
Garcia NA, Mellergaard M, Gonzalez-King H, Salomon C, Handberg A. Comprehensive Strategy for Identifying Extracellular Vesicle Surface Proteins as Biomarkers for Non-Alcoholic Fatty Liver Disease. Int J Mol Sci 2023; 24:13326. [PMID: 37686134 PMCID: PMC10487973 DOI: 10.3390/ijms241713326] [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: 07/18/2023] [Revised: 08/21/2023] [Accepted: 08/25/2023] [Indexed: 09/10/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a liver disorder that has become a global health concern due to its increasing prevalence. There is a need for reliable biomarkers to aid in the diagnosis and prognosis of NAFLD. Extracellular vesicles (EVs) are promising candidates in biomarker discovery, as they carry proteins that reflect the pathophysiological state of the liver. In this review, we developed a list of EV proteins that could be used as diagnostic biomarkers for NAFLD. We employed a multi-step strategy that involved reviewing and comparing various sources of information. Firstly, we reviewed papers that have studied EVs proteins as biomarkers in NAFLD and papers that have studied circulating proteins as biomarkers in NAFLD. To further identify potential candidates, we utilized the EV database Vesiclepedia.org to qualify each protein. Finally, we consulted the Human Protein Atlas to search for candidates' localization, focusing on membrane proteins. By integrating these sources of information, we developed a comprehensive list of potential EVs membrane protein biomarkers that could aid in diagnosing and monitoring NAFLD. In conclusion, our multi-step strategy for identifying EV-based protein biomarkers for NAFLD provides a comprehensive approach that can also be applied to other diseases. The protein candidates identified through this approach could have significant implications for the development of non-invasive diagnostic tests for NAFLD and improve the management and treatment of this prevalent liver disorder.
Collapse
Affiliation(s)
| | - Maiken Mellergaard
- Department of Clinical Biochemistry, Aalborg University Hospital, Aalborg Hobrovej 18-22, 9000 Aalborg, Denmark
- Department of Clinical Medicine, The Faculty of Medicine, Aalborg University, 9000 Aalborg, Denmark
| | - Hernan Gonzalez-King
- Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, 431 50 Gothenburg, Sweden
| | - Carlos Salomon
- Translational Extracellular Vesicles in Obstetrics and Gynae-Oncology Group, University of Queensland, Brisbane, QLD 4029, Australia
| | - Aase Handberg
- Department of Clinical Biochemistry, Aalborg University Hospital, Aalborg Hobrovej 18-22, 9000 Aalborg, Denmark
- Department of Clinical Medicine, The Faculty of Medicine, Aalborg University, 9000 Aalborg, Denmark
| |
Collapse
|
6
|
Pham MT, Lee JY, Ritter C, Thielemann R, Meyer J, Haselmann U, Funaya C, Laketa V, Rohr K, Bartenschlager R. Endosomal egress and intercellular transmission of hepatic ApoE-containing lipoproteins and its exploitation by the hepatitis C virus. PLoS Pathog 2023; 19:e1011052. [PMID: 37506130 PMCID: PMC10411793 DOI: 10.1371/journal.ppat.1011052] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 08/09/2023] [Accepted: 07/07/2023] [Indexed: 07/30/2023] Open
Abstract
Liver-generated plasma Apolipoprotein E (ApoE)-containing lipoproteins (LPs) (ApoE-LPs) play central roles in lipid transport and metabolism. Perturbations of ApoE can result in several metabolic disorders and ApoE genotypes have been associated with multiple diseases. ApoE is synthesized at the endoplasmic reticulum and transported to the Golgi apparatus for LP assembly; however, the ApoE-LPs transport pathway from there to the plasma membrane is largely unknown. Here, we established an integrative imaging approach based on a fully functional fluorescently tagged ApoE. We found that newly synthesized ApoE-LPs accumulate in CD63-positive endosomes of hepatocytes. In addition, we observed the co-egress of ApoE-LPs and CD63-positive intraluminal vesicles (ILVs), which are precursors of extracellular vesicles (EVs), along the late endosomal trafficking route in a microtubule-dependent manner. A fraction of ApoE-LPs associated with CD63-positive EVs appears to be co-transmitted from cell to cell. Given the important role of ApoE in viral infections, we employed as well-studied model the hepatitis C virus (HCV) and found that the viral replicase component nonstructural protein 5A (NS5A) is enriched in ApoE-containing ILVs. Interaction between NS5A and ApoE is required for the efficient release of ILVs containing HCV RNA. These vesicles are transported along the endosomal ApoE egress pathway. Taken together, our data argue for endosomal egress and transmission of hepatic ApoE-LPs, a pathway that is hijacked by HCV. Given the more general role of EV-mediated cell-to-cell communication, these insights provide new starting points for research into the pathophysiology of ApoE-related metabolic and infection-related disorders.
Collapse
Affiliation(s)
- Minh-Tu Pham
- Department of Infectious Diseases, Molecular Virology, Center for Integrative Infectious Diseases Research, Heidelberg University, Heidelberg, Germany
- German Center for Infection Research (DZIF), Partner Site Heidelberg, Heidelberg, Germany
| | - Ji-Young Lee
- Department of Infectious Diseases, Molecular Virology, Center for Integrative Infectious Diseases Research, Heidelberg University, Heidelberg, Germany
- German Center for Infection Research (DZIF), Partner Site Heidelberg, Heidelberg, Germany
| | - Christian Ritter
- BioQuant Center, IPMB, Biomedical Computer Vision Group, Heidelberg University, Heidelberg, Germany
| | - Roman Thielemann
- BioQuant Center, IPMB, Biomedical Computer Vision Group, Heidelberg University, Heidelberg, Germany
| | - Janis Meyer
- BioQuant Center, IPMB, Biomedical Computer Vision Group, Heidelberg University, Heidelberg, Germany
| | - Uta Haselmann
- Department of Infectious Diseases, Molecular Virology, Center for Integrative Infectious Diseases Research, Heidelberg University, Heidelberg, Germany
| | - Charlotta Funaya
- Electron Microscopy Core Facility (EMCF), Heidelberg University, Heidelberg, Germany
| | - Vibor Laketa
- German Center for Infection Research (DZIF), Partner Site Heidelberg, Heidelberg, Germany
- Department of Infectious Diseases, Virology, Center for Integrative Infectious Diseases Research, Heidelberg University, Heidelberg, Germany
| | - Karl Rohr
- BioQuant Center, IPMB, Biomedical Computer Vision Group, Heidelberg University, Heidelberg, Germany
| | - Ralf Bartenschlager
- Department of Infectious Diseases, Molecular Virology, Center for Integrative Infectious Diseases Research, Heidelberg University, Heidelberg, Germany
- German Center for Infection Research (DZIF), Partner Site Heidelberg, Heidelberg, Germany
- Division Virus-Associated Carcinogenesis, German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
7
|
Extracellular Vesicles from Interferon-γ-primed Human Umbilical Cord Mesenchymal Stromal Cells Reduce Escherichia coli-induced Acute Lung Injury in Rats. Anesthesiology 2020; 130:778-790. [PMID: 30870158 DOI: 10.1097/aln.0000000000002655] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
BACKGROUND Human umbilical cord mesenchymal stromal cells possess considerable therapeutic promise for acute respiratory distress syndrome. Umbilical cord mesenchymal stromal cells may exert therapeutic effects via extracellular vesicles, while priming umbilical cord mesenchymal stromal cells may further enhance their effect. The authors investigated whether interferon-γ-primed umbilical cord mesenchymal stromal cells would generate mesenchymal stromal cell-derived extracellular vesicles with enhanced effects in Escherichia coli (E. coli) pneumonia. METHODS In a university laboratory, anesthetized adult male Sprague-Dawley rats (n = 8 to 18 per group) underwent intrapulmonary E. coli instillation (5 × 10 colony forming units per kilogram), and were randomized to receive (a) primed mesenchymal stromal cell-derived extracellular vesicles, (b) naïve mesenchymal stromal cell-derived extracellular vesicles (both 100 million mesenchymal stromal cell-derived extracellular vesicles per kilogram), or (c) vehicle. Injury severity and bacterial load were assessed at 48 h. In vitro studies assessed the potential for primed and naïve mesenchymal stromal cell-derived extracellular vesicles to enhance macrophage bacterial phagocytosis and killing. RESULTS Survival increased with primed (10 of 11 [91%]) and naïve (8 of 8 [100%]) mesenchymal stromal cell-derived extracellular vesicles compared with vehicle (12 of 18 [66.7%], P = 0.038). Primed-but not naïve-mesenchymal stromal cell-derived extracellular vesicles reduced alveolar-arterial oxygen gradient (422 ± 104, 536 ± 58, 523 ± 68 mm Hg, respectively; P = 0.008), reduced alveolar protein leak (0.7 ± 0.3, 1.4 ± 0.4, 1.5 ± 0.7 mg/ml, respectively; P = 0.003), increased lung mononuclear phagocytes (23.2 ± 6.3, 21.7 ± 5, 16.7 ± 5 respectively; P = 0.025), and reduced alveolar tumor necrosis factor alpha concentrations (29 ± 14.5, 35 ± 12.3, 47.2 ± 6.3 pg/ml, respectively; P = 0.026) compared with vehicle. Primed-but not naïve-mesenchymal stromal cell-derived extracellular vesicles enhanced endothelial nitric oxide synthase production in the injured lung (endothelial nitric oxide synthase/β-actin = 0.77 ± 0.34, 0.25 ± 0.29, 0.21 ± 0.33, respectively; P = 0.005). Both primed and naïve mesenchymal stromal cell-derived extracellular vesicles enhanced E. coli phagocytosis and bacterial killing in human acute monocytic leukemia cell line (THP-1) in vitro (36.9 ± 4, 13.3 ± 8, 0.1 ± 0.01%, respectively; P = 0.0004) compared with vehicle. CONCLUSIONS Extracellular vesicles from interferon-γ-primed human umbilical cord mesenchymal stromal cells more effectively attenuated E. coli-induced lung injury compared with extracellular vesicles from naïve mesenchymal stromal cells, potentially via enhanced macrophage phagocytosis and killing of E. coli.
Collapse
|
8
|
Henrich SE, Thaxton CS. An update on synthetic high-density lipoprotein-like nanoparticles for cancer therapy. Expert Rev Anticancer Ther 2019; 19:515-528. [DOI: 10.1080/14737140.2019.1624529] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Stephen E. Henrich
- Department of Urology, Simpson Querrey Institute for BioNanotechnology, Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Feinberg School of Medicine, Chicago, IL, USA
| | - C. Shad Thaxton
- Department of Urology, Simpson Querrey Institute for BioNanotechnology, Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Feinberg School of Medicine, Chicago, IL, USA
| |
Collapse
|
9
|
Reza-Zaldivar EE, Hernández-Sapiéns MA, Minjarez B, Gutiérrez-Mercado YK, Márquez-Aguirre AL, Canales-Aguirre AA. Potential Effects of MSC-Derived Exosomes in Neuroplasticity in Alzheimer's Disease. Front Cell Neurosci 2018; 12:317. [PMID: 30319358 PMCID: PMC6165870 DOI: 10.3389/fncel.2018.00317] [Citation(s) in RCA: 109] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Accepted: 08/30/2018] [Indexed: 12/23/2022] Open
Abstract
Alzheimer’s disease (AD) is the most common type of dementia affecting regions of the central nervous system that exhibit synaptic plasticity and are involved in higher brain functions such as learning and memory. AD is characterized by progressive cognitive dysfunction, memory loss and behavioral disturbances of synaptic plasticity and energy metabolism. Cell therapy has emerged as an alternative treatment of AD. The use of adult stem cells, such as neural stem cells and Mesenchymal Stem Cells (MSCs) from bone marrow and adipose tissue, have the potential to decrease cognitive deficits, possibly by reducing neuronal loss through blocking apoptosis, increasing neurogenesis, synaptogenesis and angiogenesis. These processes are mediated primarily by the secretion of many growth factors, anti-inflammatory proteins, membrane receptors, microRNAs (miRNA) and exosomes. Exosomes encapsulate and transfer several functional molecules like proteins, lipids and regulatory RNA which can modify cell metabolism. In the proteomic characterization of the content of MSC-derived exosomes, more than 730 proteins have been identified, some of which are specific cell type markers and others are involved in the regulation of binding and fusion of exosomes with adjacent cells. Furthermore, some factors were found that promote the recruitment, proliferation and differentiation of other cells like neural stem cells. Moreover, within exosomal cargo, a wide range of miRNAs were found, which can control functions related to neural remodeling as well as angiogenic and neurogenic processes. Taking this into consideration, the use of exosomes could be part of a strategy to promote neuroplasticity, improve cognitive impairment and neural replacement in AD. In this review, we describe how exosomes are involved in AD pathology and discuss the therapeutic potential of MSC-derived exosomes mediated by miRNA and protein cargo.
Collapse
Affiliation(s)
- Edwin E Reza-Zaldivar
- Unidad de Evaluación Preclínica, Biotecnología Médica y Farmacéutica, CONACYT Centro de Investigación y Asistencia en Tecnología y Diseño del Estado de Jalisco (CIATEJ), Guadalajara, Mexico
| | - Mercedes A Hernández-Sapiéns
- Unidad de Evaluación Preclínica, Biotecnología Médica y Farmacéutica, CONACYT Centro de Investigación y Asistencia en Tecnología y Diseño del Estado de Jalisco (CIATEJ), Guadalajara, Mexico
| | - Benito Minjarez
- Centro Universitario de Ciencias Biológicas y Agropecuarias (CUCBA), Universidad de Guadalajara, Guadalajara, Mexico
| | - Yanet K Gutiérrez-Mercado
- Unidad de Evaluación Preclínica, Biotecnología Médica y Farmacéutica, CONACYT Centro de Investigación y Asistencia en Tecnología y Diseño del Estado de Jalisco (CIATEJ), Guadalajara, Mexico
| | - Ana L Márquez-Aguirre
- Unidad de Evaluación Preclínica, Biotecnología Médica y Farmacéutica, CONACYT Centro de Investigación y Asistencia en Tecnología y Diseño del Estado de Jalisco (CIATEJ), Guadalajara, Mexico
| | - Alejandro A Canales-Aguirre
- Unidad de Evaluación Preclínica, Biotecnología Médica y Farmacéutica, CONACYT Centro de Investigación y Asistencia en Tecnología y Diseño del Estado de Jalisco (CIATEJ), Guadalajara, Mexico.,Profesor del programa de Maestría en Ciencias de la Salud Ambiental, Centro Universitario de Ciencias Biológicas y Agropecuarias (CUCBA), Universidad de Guadalajara, Guadalajara, Mexico
| |
Collapse
|
10
|
Muresan XM, Sticozzi C, Belmonte G, Cervellati F, Ferrara F, Lila MA, Valacchi G. SR-B1 involvement in keratinocytes in vitro wound closure. Arch Biochem Biophys 2018; 658:1-6. [PMID: 30240595 DOI: 10.1016/j.abb.2018.09.014] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Revised: 09/16/2018] [Accepted: 09/17/2018] [Indexed: 12/17/2022]
Abstract
Skin represents the most extended organ of human body, having as main function the protection of our body from outdoor stressors. Its protective ability is compromised when the skin is disrupted as a consequence of mechanical insults. For this purpose, cutaneous tissue is equipped with an efficient and fine mechanism involved in repairing the wounded area. Among the numerous players that take part in the wound healing process, SR-B1 has been recently shown to have a role in keratinocyte re-epithelialization. SR-B1 is a mediator of cholesterol uptake from HDLs, whereas it is implicated in other cellular processes such as vitamins absorption, vesicle trafficking or pathogen identification. The aim of this study was to investigate the mechanisms involved in SR-B1 role in skin wound closure. Our in vitro data demonstrated that SR-B1 influenced keratinocyte proliferation and migration through a downregulation of nuclear cyclin D1 levels and active MMP9 expression respectively possibly in an NF-kB-dependent mechanism. In addition, SR-B1 was also able to modulate keratinocyte morphology into a pro-migratory cytoskeleton rearrangement. The present in vitro study suggests a new role of SRB1 as a possible new key player in cutaneous wound healing mechanism.
Collapse
Affiliation(s)
- Ximena M Muresan
- Dept. Life Sciences and Biotechnology, University of Ferrara, Ferrara, Italy
| | - Claudia Sticozzi
- Dept. Life Sciences and Biotechnology, University of Ferrara, Ferrara, Italy
| | - Giuseppe Belmonte
- Dept. Life Sciences and Biotechnology, University of Ferrara, Ferrara, Italy
| | - Franco Cervellati
- Dept. Life Sciences and Biotechnology, University of Ferrara, Ferrara, Italy
| | - Francesca Ferrara
- Dept. Life Sciences and Biotechnology, University of Ferrara, Ferrara, Italy
| | - Mary Ann Lila
- Plants for Human Health Institute, Animal Sciences Dept., NC Research Campus, NC State University, NC, USA
| | - Giuseppe Valacchi
- Dept. Life Sciences and Biotechnology, University of Ferrara, Ferrara, Italy; Plants for Human Health Institute, Animal Sciences Dept., NC Research Campus, NC State University, NC, USA.
| |
Collapse
|
11
|
Buzás EI, Tóth EÁ, Sódar BW, Szabó-Taylor KÉ. Molecular interactions at the surface of extracellular vesicles. Semin Immunopathol 2018; 40:453-464. [PMID: 29663027 PMCID: PMC6208672 DOI: 10.1007/s00281-018-0682-0] [Citation(s) in RCA: 206] [Impact Index Per Article: 34.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Accepted: 03/26/2018] [Indexed: 12/21/2022]
Abstract
Extracellular vesicles such as exosomes, microvesicles, apoptotic bodies, and large oncosomes have been shown to participate in a wide variety of biological processes and are currently under intense investigation in many different fields of biomedicine. One of the key features of extracellular vesicles is that they have relatively large surface compared to their volume. Some extracellular vesicle surface molecules are shared with those of the plasma membrane of the releasing cell, while other molecules are characteristic for extracellular vesicular surfaces. Besides proteins, lipids, glycans, and nucleic acids are also players of extracellular vesicle surface interactions. Being secreted and present in high number in biological samples, collectively extracellular vesicles represent a uniquely large interactive surface area which can establish contacts both with cells and with molecules in the extracellular microenvironment. Here, we provide a brief overview of known components of the extracellular vesicle surface interactome and highlight some already established roles of the extracellular vesicle surface interactions in different biological processes in health and disease.
Collapse
Affiliation(s)
- Edit I Buzás
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, Budapest, Hungary.
- MTA-SE Immune-Proteogenomics Research Group, Budapest, Hungary.
| | - Eszter Á Tóth
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, Budapest, Hungary
| | - Barbara W Sódar
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, Budapest, Hungary
| | - Katalin É Szabó-Taylor
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, Budapest, Hungary
| |
Collapse
|
12
|
Natural low- and high-density lipoproteins as mighty bio-nanocarriers for anticancer drug delivery. Cancer Chemother Pharmacol 2018; 82:371-382. [DOI: 10.1007/s00280-018-3626-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Accepted: 06/12/2018] [Indexed: 02/07/2023]
|
13
|
Menard JA, Cerezo-Magaña M, Belting M. Functional role of extracellular vesicles and lipoproteins in the tumour microenvironment. Philos Trans R Soc Lond B Biol Sci 2018; 373:rstb.2016.0480. [PMID: 29158310 DOI: 10.1098/rstb.2016.0480] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/31/2017] [Indexed: 12/11/2022] Open
Abstract
Cancer can be regarded as an invasive organ that exhibits unique plasticity provided by coordinated, cancer cell-stromal cell communication in the tumour microenvironment. Typical stress factors in the tumour niche, such as hypoxia and acidosis, are major drivers and modulators of these events. Recent findings reveal an important role of extracellular vesicles and lipoproteins in cancer cell adaption to exogenous stress. Adaptive mechanisms include stimulation of angiogenesis and increased metastasis. Here, we will discuss the similarities and distinct features of these endogenous nanoparticles and their roles as signalosomes and nutrient sources in cancer. We will focus on the accumulating evidence for a central role of cell-surface heparan sulphate proteoglycans in the uptake of extracellular vesicles and lipoproteins.This article is part of the discussion meeting issue 'Extracellular vesicles and the tumour microenvironment'.
Collapse
Affiliation(s)
- Julien A Menard
- Department of Clinical Sciences, Section of Oncology and Pathology, Lund University, Lund, Sweden.,Department of Oncology, Skåne University Hospital, Lund, Sweden
| | - Myriam Cerezo-Magaña
- Department of Clinical Sciences, Section of Oncology and Pathology, Lund University, Lund, Sweden.,Department of Oncology, Skåne University Hospital, Lund, Sweden
| | - Mattias Belting
- Department of Clinical Sciences, Section of Oncology and Pathology, Lund University, Lund, Sweden .,Department of Oncology, Skåne University Hospital, Lund, Sweden
| |
Collapse
|
14
|
Rink JS, Sun W, Misener S, Wang JJ, Zhang ZJ, Kibbe MR, Dravid VP, Venkatraman S, Thaxton CS. Nitric Oxide-Delivering High-Density Lipoprotein-like Nanoparticles as a Biomimetic Nanotherapy for Vascular Diseases. ACS APPLIED MATERIALS & INTERFACES 2018; 10:6904-6916. [PMID: 29385802 PMCID: PMC8495904 DOI: 10.1021/acsami.7b18525] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
Disorders of blood vessels cause a range of severe health problems. As a powerful vasodilator and cellular second messenger, nitric oxide (NO) is known to have beneficial vascular functions. However, NO typically has a short half-life and is not specifically targeted. On the other hand, high-density lipoproteins (HDLs) are targeted natural nanoparticles (NPs) that transport cholesterol in the systemic circulation and whose protective effects in vascular homeostasis overlap with those of NO. Evolving the AuNP-templated HDL-like nanoparticles (HDL NPs), a platform of bioinspired HDL, we set up a targeted biomimetic nanotherapy for vascular disease that combines the functions of NO and HDL. A synthetic S-nitrosylated (SNO) phospholipid (1,2-dipalmitoyl-sn-glycero-3-phosphonitrosothioethanol) was synthesized and assembled with S-containing phospholipids and the principal protein of HDL, apolipoprotein A-I, to construct NO-delivering HDL-like particles (SNO HDL NPs). SNO HDL NPs self-assemble under mild conditions similar to natural processes, avoiding the complex postassembly modification needed for most synthetic NO-release nanoparticles. In vitro data demonstrate that the SNO HDL NPs merge the functional properties of NO and HDL into a targeted nanocarrier. Also, SNO HDL NPs were demonstrated to reduce ischemia/reperfusion injury in vivo in a mouse kidney transplant model and atherosclerotic plaque burden in a mouse model of atherosclerosis. Thus, the synthesis of SNO HDL NPs provides not only a bioinspired nanotherapy for vascular disease but also a foundation to construct diversified multifunctional platforms based on HDL NPs in the future.
Collapse
Affiliation(s)
- Jonathan S. Rink
- Northwestern University, Feinberg School of Medicine, Department of Urology, 303 East Chicago Avenue, Chicago, IL 60611, United States
- Northwestern University, Simpson Querrey Institute for BioNanotechnology, 303 East Superior, Chicago, IL 60611, United States
| | - Wangqiang Sun
- Northwestern University, Feinberg School of Medicine, Department of Urology, 303 East Chicago Avenue, Chicago, IL 60611, United States
- Northwestern University, Simpson Querrey Institute for BioNanotechnology, 303 East Superior, Chicago, IL 60611, United States
| | - Sol Misener
- Northwestern University, Feinberg School of Medicine, Department of Urology, 303 East Chicago Avenue, Chicago, IL 60611, United States
| | - Jiao-Jing Wang
- Northwestern University, Feinberg School of Medicine, Department of Surgery, Division of Transplantation, 303 East Chicago Ave, Chicago, IL 60611, United States
| | - Zheng Jenny Zhang
- Northwestern University, Feinberg School of Medicine, Department of Surgery, Division of Transplantation, 303 East Chicago Ave, Chicago, IL 60611, United States
| | - Melina R. Kibbe
- University of North Carolina at Chapel Hill, Department of Surgery, 101 Manning Dr., Chapel Hill, NC, 27599, United States
| | - Vinayak P. Dravid
- Northwestern University, Department of Materials Science and Engineering, 2220 Campus Drive, Evanston, IL 60208, United States
- Northwestern University, Applied Physics Program, Evanston, IL 60208, United States
- Northwestern University, International Institute for Nanotechnology, Evanston, IL60208, United States
| | - Subbu Venkatraman
- Nanyang Technological University, School of Materials Science and Engineering, 50 Nanyang Avenue, Singapore, 639798
| | - C. Shad Thaxton
- Northwestern University, Feinberg School of Medicine, Department of Urology, 303 East Chicago Avenue, Chicago, IL 60611, United States
- Northwestern University, Simpson Querrey Institute for BioNanotechnology, 303 East Superior, Chicago, IL 60611, United States
- Northwestern University, International Institute for Nanotechnology, Evanston, IL60208, United States
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL, 60611, United States
- Corresponding Author
| |
Collapse
|
15
|
Wu Z, Zeng Q, Cao K, Sun Y. Exosomes: small vesicles with big roles in hepatocellular carcinoma. Oncotarget 2018; 7:60687-60697. [PMID: 27463001 PMCID: PMC5312412 DOI: 10.18632/oncotarget.10807] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Accepted: 07/13/2016] [Indexed: 02/06/2023] Open
Abstract
Despite improvements in the diagnosis and treatment of hepatocellular carcinoma (HCC), the prognosis is still poor. Pioneering work has demonstrated a potential role for tumour cell-derived exosomes (TEXs) in HCC. TEXs can mediate immune responses, antigen presentation and intracellular communication by serving as vehicles for the transfer of proteins, viruses, lipids and RNA between cells. An improved understanding of the roles played by exosomes could lead to a powerful new strategy for preventing and treating HCC. In this review, we summarise current understanding on the topic. The literature points to two faces of TEXs in HCC: 1) They can promote invasion, metastasis, immune evasion and modulation and 2) they can act as diagnostic and prognostic biomarkers, and can be used in anti-cancer drug resistance and immunotherapy in the future.
Collapse
Affiliation(s)
- Zhitong Wu
- Department of Clinical Laboratory, Eighth Affiliated Hospital of Guangxi Medical University, Guigang City People's Hospital, Guigang, Guangxi, China
| | - Qinghai Zeng
- Department of Dermatology, Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Ke Cao
- Department of Oncology, Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yifan Sun
- Department of Clinical Laboratory, Third Affiliated Hospital of Guangxi University of Chinese Medicine, Liuzhou, Guangxi, China
| |
Collapse
|
16
|
Plebanek MP, Bhaumik D, Bryce PJ, Thaxton CS. Scavenger Receptor Type B1 and Lipoprotein Nanoparticle Inhibit Myeloid-Derived Suppressor Cells. Mol Cancer Ther 2017; 17:686-697. [PMID: 29282300 DOI: 10.1158/1535-7163.mct-17-0981] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2017] [Revised: 10/30/2017] [Accepted: 12/11/2017] [Indexed: 12/22/2022]
Abstract
Myeloid-derived suppressor cells (MDSC) are innate immune cells that potently inhibit T cells. In cancer, novel therapies aimed to activate T cells can be rendered ineffective due to the activity of MDSCs. Thus, targeted inhibition of MDSCs may greatly enhance T-cell-mediated antitumor immunity, but mechanisms remain obscure. Here we show, for the first time, that scavenger receptor type B-1 (SCARB1), a high-affinity receptor for spherical high-density lipoprotein (HDL), is expressed by MDSCs. Furthermore, we demonstrate that SCARB1 is specifically targeted by synthetic high-density lipoprotein-like nanoparticles (HDL NP), which reduce MDSC activity. Using in vitro T-cell proliferation assays, data show that HDL NPs specifically bind SCARB1 to inhibit MDSC activity. In murine cancer models, HDL NP treatment significantly reduces tumor growth, metastatic tumor burden, and increases survival due to enhanced adaptive immunity. Flow cytometry and IHC demonstrate that HDL NP-mediated suppression of MDSCs increased CD8+ T cells and reduced Treg cells in the metastatic tumor microenvironment. Using transgenic mice lacking SCARB1, in vivo data clearly show that the HDL NPs specifically target this receptor for suppressing MDSCs. Ultimately, our data provide a new mechanism and targeted therapy, HDL NPs, to modulate a critical innate immune cell checkpoint to enhance the immune response to cancer. Mol Cancer Ther; 17(3); 686-97. ©2017 AACR.
Collapse
Affiliation(s)
- Michael P Plebanek
- Department of Urology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois.,Driskill Graduate Program in the Life Sciences, Northwestern University, Chicago, Illinois
| | - Debayan Bhaumik
- Department of Urology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Paul J Bryce
- Division of Allergy-Immunology, Department of Medicine, Northwestern University, Feinberg School of Medicine, Chicago, Illinois
| | - C Shad Thaxton
- Department of Urology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois. .,Simpson Querrey Institute for BioNanotechnology, Northwestern University, Chicago, Illinois.,International Institute for Nanotechnology, Northwestern University, Chicago, Illinois.,Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, Illinois
| |
Collapse
|
17
|
McMahon KM, Scielzo C, Angeloni NL, Deiss-Yehiely E, Scarfo L, Ranghetti P, Ma S, Kaplan J, Barbaglio F, Gordon LI, Giles FJ, Thaxton CS, Ghia P. Synthetic high-density lipoproteins as targeted monotherapy for chronic lymphocytic leukemia. Oncotarget 2017; 8:11219-11227. [PMID: 28061439 PMCID: PMC5355259 DOI: 10.18632/oncotarget.14494] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Accepted: 12/26/2016] [Indexed: 12/18/2022] Open
Abstract
Chronic lymphocytic leukemia (CLL) remains incurable despite the introduction of new drugs. Therapies targeting receptors and pathways active specifically in malignant B cells might provide better treatment options. For instance, in B cell lymphoma, our group has previously shown that scavenger receptor type B-1 (SR-B1), the high-affinity receptor for cholesterol-rich high-density lipoproteins (HDL), is a therapeutic target. As evidence suggests that targeting cholesterol metabolism in CLL cells may have therapeutic benefit, we examined SR-B1 expression in primary CLL cells from patients. Unlike normal B cells that do not express SR-B1, CLL cells express the receptor. As a result, we evaluated cholesterol-poor synthetic HDL nanoparticles (HDL NP), known for targeting SR-B1, as a therapy for CLL. HDL NPs potently and selectively induce apoptotic cell death in primary CLL cells. HDL NPs had no effect on normal peripheral blood mononuclear cells from healthy individuals or patients with CLL. These data implicate SR-B1 as a target in CLL and HDL NPs as targeted monotherapy for CLL.
Collapse
Affiliation(s)
- Kaylin M McMahon
- Department of Urology, Feinberg School of Medicine, Northwestern University, Tarry, Chicago, IL, USA
| | - Cristina Scielzo
- Università Vita-Salute San Raffaele, Milan, Italy.,Strategic Research Program On CLL and Unit of B cell Neoplasia, Division of Experimental Oncology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Nicholas L Angeloni
- Department of Urology, Feinberg School of Medicine, Northwestern University, Tarry, Chicago, IL, USA
| | - Elad Deiss-Yehiely
- Department of Urology, Feinberg School of Medicine, Northwestern University, Tarry, Chicago, IL, USA
| | - Lydia Scarfo
- Università Vita-Salute San Raffaele, Milan, Italy.,Strategic Research Program On CLL and Unit of B cell Neoplasia, Division of Experimental Oncology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Pamela Ranghetti
- Università Vita-Salute San Raffaele, Milan, Italy.,Strategic Research Program On CLL and Unit of B cell Neoplasia, Division of Experimental Oncology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Shuo Ma
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL, USA
| | - Jason Kaplan
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL, USA.,Developmental Therapeutics Program of The Division of Hematology Oncology, Feinberg School of Medicine, Chicago, IL, USA
| | - Federica Barbaglio
- Strategic Research Program On CLL and Unit of B cell Neoplasia, Division of Experimental Oncology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Leo I Gordon
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL, USA
| | - Francis J Giles
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL, USA.,Developmental Therapeutics Program of The Division of Hematology Oncology, Feinberg School of Medicine, Chicago, IL, USA
| | - C Shad Thaxton
- Department of Urology, Feinberg School of Medicine, Northwestern University, Tarry, Chicago, IL, USA.,Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL, USA.,Simpson Querrey Institute (SQI) for BioNanotechnology, Chicago, IL, USA.,International Institute for Nanotechnology, Evanston, IL, USA
| | - Paolo Ghia
- Università Vita-Salute San Raffaele, Milan, Italy.,Strategic Research Program On CLL and Unit of B cell Neoplasia, Division of Experimental Oncology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| |
Collapse
|
18
|
Harden ME, Munger K. Human papillomavirus 16 E6 and E7 oncoprotein expression alters microRNA expression in extracellular vesicles. Virology 2017; 508:63-69. [PMID: 28500882 DOI: 10.1016/j.virol.2017.05.005] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Revised: 05/03/2017] [Accepted: 05/05/2017] [Indexed: 01/08/2023]
Abstract
Extracellular vesicles released by cancer cells are mediators of intercellular communication that have been reported to contribute to carcinogenesis. Since they are readily detected in bodily fluids, they may also be used as cancer biomarkers. The E6/E7 oncoproteins drive human papillomavirus (HPV)-associated cancers, which account for approximately 5% of all human cancers worldwide. Here, we investigate how HPV16 E6/E7 oncogene expression in primary human epithelial cells alters miR expression in extracellular vesicles and compare these to changes in intracellular miR expression. Examining a panel of 68 cancer related miRs revealed that many miRs had similar expression patterns in cells and in extracellular vesicles, whereas some other miRs had different expression patterns and may be selectively packaged into extracellular vesicles. Interestingly, the set of miRs that may be selectively packaged in HPV16 E6/E7 extracellular vesicles is predicted to inhibit necrosis and apoptosis.
Collapse
Affiliation(s)
- Mallory E Harden
- Program in Virology, Division of Medical Sciences, Harvard Medical School, Boston, MA 02115, USA; Department of Developmental, Molecular and Chemical Biology, Tufts University School of Medicine, Boston, MA 02111, USA
| | - Karl Munger
- Department of Developmental, Molecular and Chemical Biology, Tufts University School of Medicine, Boston, MA 02111, USA.
| |
Collapse
|
19
|
Lai CT, Sun W, Palekar RU, Thaxton CS, Schatz GC. Molecular Dynamics Simulation and Experimental Studies of Gold Nanoparticle Templated HDL-like Nanoparticles for Cholesterol Metabolism Therapeutics. ACS APPLIED MATERIALS & INTERFACES 2017; 9:1247-1254. [PMID: 28001031 DOI: 10.1021/acsami.6b12249] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
High-density lipoprotein (HDL) plays an important role in the transport and metabolism of cholesterol. Mimics of HDL are being explored as potentially powerful therapeutic agents for removing excess cholesterol from arterial plaques. Gold nanoparticles (AuNPs) functionalized with apolipoprotein A-I and with the lipids 1,2-dipalmitoyl-sn-glycero-3-phosphocholine and 1,2-dipalmitoyl-sn-glycero-3-phosphoethanolamine-N-[3-(2-pyridyldithio)propionate] have been demonstrated to be robust acceptors of cellular cholesterol. However, detailed structural information about this functionalized HDL AuNP is still lacking. In this study, we have used X-ray photoelectron spectroscopy and lecithin/cholesterol acyltransferase activation experiments together with coarse-grained and all-atom molecular dynamics simulations to model the structure and cholesterol uptake properties of the HDL AuNP construct. By simulating different apolipoprotein-loaded AuNPs, we find that lipids are oriented differently in regions with and without apoA-I. We also show that in this functionalized HDL AuNP, the distribution of cholesteryl ester maintains a reverse concentration gradient that is similar to the gradient found in native HDL.
Collapse
Affiliation(s)
- Cheng-Tsung Lai
- Department of Chemistry, Northwestern University , Evanston, Illinois 60208, United States
| | - Wangqiang Sun
- Department of Urology, Northwestern University , Chicago, Illinois 60611, United States
- Simpson Querrey Institute for Bionanotechnology , 303 East Superior, Chicago, Illinois 60611, United States
- International Institute for Nanotechnology, 2145 Sheridan Road, Evanston, Illinois 60208, United States
| | - Rohun U Palekar
- Department of Urology, Northwestern University , Chicago, Illinois 60611, United States
- Simpson Querrey Institute for Bionanotechnology , 303 East Superior, Chicago, Illinois 60611, United States
- International Institute for Nanotechnology, 2145 Sheridan Road, Evanston, Illinois 60208, United States
| | - C Shad Thaxton
- Department of Urology, Northwestern University , Chicago, Illinois 60611, United States
- Simpson Querrey Institute for Bionanotechnology , 303 East Superior, Chicago, Illinois 60611, United States
- International Institute for Nanotechnology, 2145 Sheridan Road, Evanston, Illinois 60208, United States
| | - George C Schatz
- Department of Chemistry, Northwestern University , Evanston, Illinois 60208, United States
| |
Collapse
|
20
|
Tumor Microenvironment Modulation via Gold Nanoparticles Targeting Malicious Exosomes: Implications for Cancer Diagnostics and Therapy. Int J Mol Sci 2017; 18:ijms18010162. [PMID: 28098821 PMCID: PMC5297795 DOI: 10.3390/ijms18010162] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2016] [Revised: 01/03/2017] [Accepted: 01/09/2017] [Indexed: 12/18/2022] Open
Abstract
Exosomes are nanovesicles formed in the endosomal pathway with an important role in paracrine and autocrine cell communication. Exosomes secreted by cancer cells, malicious exosomes, have important roles in tumor microenvironment maturation and cancer progression. The knowledge of the role of exosomes in tumorigenesis prompted a new era in cancer diagnostics and therapy, taking advantage of the use of circulating exosomes as tumor biomarkers due to their stability in body fluids and targeting malignant exosomes’ release and/or uptake to inhibit or delay tumor development. In recent years, nanotechnology has paved the way for the development of a plethora of new diagnostic and therapeutic platforms, fostering theranostics. The unique physical and chemical properties of gold nanoparticles (AuNPs) make them suitable vehicles to pursuit this goal. AuNPs’ properties such as ease of synthesis with the desired shape and size, high surface:volume ratio, and the possibility of engineering their surface as desired, potentiate AuNPs’ role in nanotheranostics, allowing the use of the same formulation for exosome detection and restraining the effect of malicious exosomes in cancer progression.
Collapse
|