1
|
Kalsi P, Gupta N, Goyal G, Sharma H. Decoding the role of extracellular vesicles in pathogenesis of cystic fibrosis. Mol Cell Pediatr 2025; 12:5. [PMID: 40257719 PMCID: PMC12011702 DOI: 10.1186/s40348-025-00190-4] [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: 10/25/2024] [Accepted: 03/05/2025] [Indexed: 04/22/2025] Open
Abstract
BACKGROUND Intercellular communication is a critical process that ensures cooperation between distinct cell types and maintains homeostasis. In the past decades, extracellular vesicles (EVs) have been recognized as key components in cell-to-cell communication. These EVs carry multiple factors such as active enzymes, metabolites, nucleic acids and surface molecules that can alter the behavior of recipient cells. Thus, the role of EVs in exacerbating disease pathology by transporting inflammatory mediators, and other molecular signals that contribute to chronic inflammation and immune dysregulation in various diseases including cystic fibrosis (CF) is well documented. MAIN BODY CF is a genetic disorder characterized by chronic inflammation and persistent infections, primarily affecting the respiratory system. This review explores the multifaceted roles of EVs in CF lung disease, focusing on their biogenesis, cargo, and contributions to disease progression. It is well known that CF results from mutations in the CFTR (cystic fibrosis transmembrane conductance regulator) gene, leading to defective ion transport, thick mucus secretion, and a propensity for bacterial infections. However, it has been observed that EVs derived from CF patients carry altered molecular cargo, including proteins, lipids, RNA, and DNA, which can exacerbate these conditions by promoting inflammation, and modulating immune responses. Beyond their pathogenic roles, EVs also hold significant therapeutic potential. Their natural ability to transfer bioactive molecules positions them as promising vectors for delivering therapeutic agents, such as gene therapy constructs and anti-inflammatory compounds. Accordingly, a study has shown that these EVs can act as a carrier molecule for transport of functional CFTR mRNA, helping to restore proper chloride ion channel function by correcting defective CFTR proteins in affected cells. CONCLUSION This review aims to summarize the role of EVs and their molecular cargo in pathogenesis of CF lung disease via modulation of intracellular signaling leading to persistent inflammation and increased disease severity. We also explored the mechanisms of EV biogenesis, cargo selection, and their effects on recipient cells which may provide novel insights into CF pathogenesis and open new avenues for EV-based therapies aimed at improving disease management.
Collapse
Affiliation(s)
- Priya Kalsi
- Department of Biochemistry, All India Institute of Medical Sciences, Bathinda, 151001, Punjab, India
| | - Nikhil Gupta
- Department of Biochemistry, All India Institute of Medical Sciences, Bathinda, 151001, Punjab, India
| | - Gitanjali Goyal
- Department of Biochemistry, All India Institute of Medical Sciences, Bathinda, 151001, Punjab, India
| | - Himanshu Sharma
- Department of Biochemistry, All India Institute of Medical Sciences, Bathinda, 151001, Punjab, India.
| |
Collapse
|
2
|
R S A, R M, Sastri KT, G S M, A R A, V B. Precision medicine advances in cystic fibrosis: Exploring genetic pathways for targeted therapies. Life Sci 2024; 358:123186. [PMID: 39471902 DOI: 10.1016/j.lfs.2024.123186] [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: 08/12/2024] [Revised: 10/14/2024] [Accepted: 10/24/2024] [Indexed: 11/01/2024]
Abstract
Personalized medicine has transformed the treatment of cystic fibrosis (CF), providing customized therapeutic approaches based on individual genetic profiles. This review explores the genetic foundations of CF, focusing on mutations in the cystic fibrosis transmembrane conductance regulator (CFTR) gene and their implications for the development of the disease. The advent of genetic testing has enabled the association of specific mutations to disease severity, leading to the development of CFTR modulators like Ivacaftor, Lumacaftor, and Tezacaftor. Beyond CFTR mutations, genetic modifiers, including gene replacement therapy, genetic manipulation, lentivirus, and non-viral gene therapy formulations, along with environmental factors, play critical roles in influencing disease expression and outcomes. The identification of these modifiers is essential for optimizing therapeutic strategies. Emerging biomarkers, including inflammatory markers and pulmonary function indicators, aid in early disease detection and monitoring progression. Omics technologies are uncovering novel biomarkers, enabling more precise disease management. Pharmacogenomics has become integral to CF care, allowing for personalized approaches that consider genetic variations influencing drug metabolism, especially in antibiotics and anti-inflammatory therapies. The future of CF treatment lies in precision therapies, including CFTR modulators and cutting-edge techniques like gene therapy and CRISPR-Cas9 for mutation correction. As research evolves, these advances can improve patient outcomes while minimizing adverse effects. Ethical considerations and regulatory challenges remain critical as personalized medicine advances, ensuring equitable access and the long-term effectiveness of these innovative therapies.
Collapse
Affiliation(s)
- Abinesh R S
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Shivarathreeshwara Nagara, Mysuru, India
| | - Madhav R
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Shivarathreeshwara Nagara, Mysuru, India
| | - K Trideva Sastri
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Shivarathreeshwara Nagara, Mysuru, India.
| | - Meghana G S
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Shivarathreeshwara Nagara, Mysuru, India
| | - Akhila A R
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Shivarathreeshwara Nagara, Mysuru, India
| | - Balamuralidhara V
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Shivarathreeshwara Nagara, Mysuru, India
| |
Collapse
|
3
|
Luo L, Zhang W, You S, Cui X, Tu H, Yi Q, Wu J, Liu O. The role of epithelial cells in fibrosis: Mechanisms and treatment. Pharmacol Res 2024; 202:107144. [PMID: 38484858 DOI: 10.1016/j.phrs.2024.107144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 02/19/2024] [Accepted: 03/12/2024] [Indexed: 03/19/2024]
Abstract
Fibrosis is a pathological process that affects multiple organs and is considered one of the major causes of morbidity and mortality in multiple diseases, resulting in an enormous disease burden. Current studies have focused on fibroblasts and myofibroblasts, which directly lead to imbalance in generation and degradation of extracellular matrix (ECM). In recent years, an increasing number of studies have focused on the role of epithelial cells in fibrosis. In some cases, epithelial cells are first exposed to external physicochemical stimuli that may directly drive collagen accumulation in the mesenchyme. In other cases, the source of stimulation is mainly immune cells and some cytokines, and epithelial cells are similarly altered in the process. In this review, we will focus on the multiple dynamic alterations involved in epithelial cells after injury and during fibrogenesis, discuss the association among them, and summarize some therapies targeting changed epithelial cells. Especially, epithelial mesenchymal transition (EMT) is the key central step, which is closely linked to other biological behaviors. Meanwhile, we think studies on disruption of epithelial barrier, epithelial cell death and altered basal stem cell populations and stemness in fibrosis are not appreciated. We believe that therapies targeted epithelial cells can prevent the progress of fibrosis, but not reverse it. The epithelial cell targeting therapies will provide a wonderful preventive and delaying action.
Collapse
Affiliation(s)
- Liuyi Luo
- Xiangya Stomatological Hospital & Xiangya School of Stomatology, Central South University, Changsha, Hunan, China; Academician Workstation for Oral-maxilofacial and Regenerative Medicine, Central South University, Changsha, Hunan, China
| | - Wei Zhang
- Department of Oral Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Siyao You
- Xiangya Stomatological Hospital & Xiangya School of Stomatology, Central South University, Changsha, Hunan, China; Academician Workstation for Oral-maxilofacial and Regenerative Medicine, Central South University, Changsha, Hunan, China
| | - Xinyan Cui
- Xiangya Stomatological Hospital & Xiangya School of Stomatology, Central South University, Changsha, Hunan, China; Academician Workstation for Oral-maxilofacial and Regenerative Medicine, Central South University, Changsha, Hunan, China
| | - Hua Tu
- Xiangya Stomatological Hospital & Xiangya School of Stomatology, Central South University, Changsha, Hunan, China; Academician Workstation for Oral-maxilofacial and Regenerative Medicine, Central South University, Changsha, Hunan, China
| | - Qiao Yi
- Xiangya Stomatological Hospital & Xiangya School of Stomatology, Central South University, Changsha, Hunan, China; Academician Workstation for Oral-maxilofacial and Regenerative Medicine, Central South University, Changsha, Hunan, China
| | - Jianjun Wu
- Xiangya Stomatological Hospital & Xiangya School of Stomatology, Central South University, Changsha, Hunan, China; Academician Workstation for Oral-maxilofacial and Regenerative Medicine, Central South University, Changsha, Hunan, China.
| | - Ousheng Liu
- Xiangya Stomatological Hospital & Xiangya School of Stomatology, Central South University, Changsha, Hunan, China; Academician Workstation for Oral-maxilofacial and Regenerative Medicine, Central South University, Changsha, Hunan, China.
| |
Collapse
|
4
|
Al-Humiari MA, Yu L, Liu LP, Nouri MZ, Tuna KM, Denslow ND, Alli AA. Extracellular vesicles from BALF of pediatric cystic fibrosis and asthma patients increase epithelial sodium channel activity in small airway epithelial cells. BIOCHIMICA ET BIOPHYSICA ACTA. BIOMEMBRANES 2024; 1866:184219. [PMID: 37634857 PMCID: PMC11632644 DOI: 10.1016/j.bbamem.2023.184219] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 08/20/2023] [Accepted: 08/23/2023] [Indexed: 08/29/2023]
Abstract
Extracellular Vesicles (EVs) are nanosized vesicles derived from all cell types. EV cargo allows for intercellular communication, intracellular signaling, and regulation of proteins in recipient cells. We tested the hypothesis that EVs isolated from the bronchoalveolar-lavage fluid (BALF) of pediatric cystic fibrosis (CF) or pediatric asthma patients increase epithelial sodium channel (ENaC) activity in normal human small airway epithelial cells (SAECs) and the mechanism involves specific EV lipids. We characterized EVs from BALF of pediatric CF and pediatric asthma patients by nanoparticle tracking analysis, transmission electron microscopy, and Western blotting. The CF and asthma pediatric groups were similar in BALF electrolytes concentration and cell count, except for neutrophils, which were higher in the CF group. Lipidomic analyses for each group of EVs were performed using targeted mass spectrometry. Phosphatidylethanolamine, sphingomyelins, and triacylglycerol were enriched in both groups, but phosphatidylcholine and phosphatidylinositol concentrations were greater in the CF group compared to the asthma group, and the opposite trend was found for phosphatidylserine. Endogenous ENaC activity, measured by the single-channel patch-clamp technique, increased in normal human SAECs after challenging SAEC with EVs from either the CF or asthma groups compared to control EVs. In conclusion, EVs isolated from BALF of pediatric patients with CF or asthma have unique lipid profiles. Despite the differences, both types of EVs increase ENaC activity in normal human SAECs compared to control EVs isolated from the conditioned media of these cells.
Collapse
Affiliation(s)
- Mohammed A Al-Humiari
- Department of Pediatrics, Pediatric Pulmonology, University of Florida, Gainesville, FL, United States of America
| | - Ling Yu
- Department of Physiology and Aging, University of Florida, Gainesville, FL, United States of America
| | - Lauren P Liu
- Department of Physiology and Aging, University of Florida, Gainesville, FL, United States of America
| | - Mohammad-Zaman Nouri
- Department of Physiological Sciences and Center for Environmental and Human Toxicology, University of Florida, Gainesville, FL, United States of America
| | - Kubra M Tuna
- Department of Endocrinology, University of Florida, Gainesville, FL, United States of America
| | - Nancy D Denslow
- Department of Physiological Sciences and Center for Environmental and Human Toxicology, University of Florida, Gainesville, FL, United States of America
| | - Abdel A Alli
- Department of Pediatrics, Pediatric Pulmonology, University of Florida, Gainesville, FL, United States of America; Department of Physiology and Aging, University of Florida, Gainesville, FL, United States of America; Department of Medicine, Division of Nephrology, Hypertension, and Renal Transplantation, University of Florida, Gainesville, FL, United States of America.
| |
Collapse
|
5
|
Roerig J, Schulz-Siegmund M. Standardization Approaches for Extracellular Vesicle Loading with Oligonucleotides and Biologics. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2301763. [PMID: 37287374 DOI: 10.1002/smll.202301763] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 05/13/2023] [Indexed: 06/09/2023]
Abstract
Extracellular vesicles (EVs) are widely recognized for their potential as drug delivery systems. EVs are membranous nanoparticles shed from cells. Among their natural features are their ability to shield cargo molecules against degradation and enable their functional internalization into target cells. Especially biological or bio-inspired large molecules (LMs), like nucleic acids, proteins, peptides, and others, may profit from encapsulation in EVs for drug delivery purposes. In the last years, a variety of loading protocols are explored for different LMs. The lack of standardization in the EV drug delivery field has impeded their comparability so far. Currently, the first reporting frameworks and workflows for EV drug loading are proposed. The aim of this review is to summarize these evolving standardization approaches and set recently developed methods into context. This will allow for enhanced comparability of future work on EV drug loading with LMs.
Collapse
Affiliation(s)
- Josepha Roerig
- Pharmaceutical Technology, Institute of Pharmacy, Medical Faculty, Leipzig University, 04317, Leipzig, Germany
| | - Michaela Schulz-Siegmund
- Pharmaceutical Technology, Institute of Pharmacy, Medical Faculty, Leipzig University, 04317, Leipzig, Germany
| |
Collapse
|
6
|
Di Mambro T, Pellielo G, Agyapong ED, Carinci M, Chianese D, Giorgi C, Morciano G, Patergnani S, Pinton P, Rimessi A. The Tricky Connection between Extracellular Vesicles and Mitochondria in Inflammatory-Related Diseases. Int J Mol Sci 2023; 24:8181. [PMID: 37175888 PMCID: PMC10179665 DOI: 10.3390/ijms24098181] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 04/21/2023] [Accepted: 05/01/2023] [Indexed: 05/15/2023] Open
Abstract
Mitochondria are organelles present in almost all eukaryotic cells, where they represent the main site of energy production. Mitochondria are involved in several important cell processes, such as calcium homeostasis, OXPHOS, autophagy, and apoptosis. Moreover, they play a pivotal role also in inflammation through the inter-organelle and inter-cellular communications, mediated by the release of mitochondrial damage-associated molecular patterns (mtDAMPs). It is currently well-documented that in addition to traditional endocrine and paracrine communication, the cells converse via extracellular vesicles (EVs). These small membrane-bound particles are released from cells in the extracellular milieu under physio-pathological conditions. Importantly, EVs have gained much attention for their crucial role in inter-cellular communication, translating inflammatory signals into recipient cells. EVs cargo includes plasma membrane and endosomal proteins, but EVs also contain material from other cellular compartments, including mitochondria. Studies have shown that EVs may transport mitochondrial portions, proteins, and/or mtDAMPs to modulate the metabolic and inflammatory responses of recipient cells. Overall, the relationship between EVs and mitochondria in inflammation is an active area of research, although further studies are needed to fully understand the mechanisms involved and how they may be targeted for therapeutic purposes. Here, we have reported and discussed the latest studies focused on this fascinating and recent area of research, discussing of tricky connection between mitochondria and EVs in inflammatory-related diseases.
Collapse
Affiliation(s)
- Tommaso Di Mambro
- Department of Medical Sciences, Section of Experimental Medicine, Laboratory for Technologies of Advanced Therapies, University of Ferrara, 44121 Ferrara, Italy; (T.D.M.); (G.P.); (E.D.A.); (M.C.); (D.C.); (C.G.); (G.M.); (S.P.); (P.P.)
| | - Giulia Pellielo
- Department of Medical Sciences, Section of Experimental Medicine, Laboratory for Technologies of Advanced Therapies, University of Ferrara, 44121 Ferrara, Italy; (T.D.M.); (G.P.); (E.D.A.); (M.C.); (D.C.); (C.G.); (G.M.); (S.P.); (P.P.)
| | - Esther Densu Agyapong
- Department of Medical Sciences, Section of Experimental Medicine, Laboratory for Technologies of Advanced Therapies, University of Ferrara, 44121 Ferrara, Italy; (T.D.M.); (G.P.); (E.D.A.); (M.C.); (D.C.); (C.G.); (G.M.); (S.P.); (P.P.)
| | - Marianna Carinci
- Department of Medical Sciences, Section of Experimental Medicine, Laboratory for Technologies of Advanced Therapies, University of Ferrara, 44121 Ferrara, Italy; (T.D.M.); (G.P.); (E.D.A.); (M.C.); (D.C.); (C.G.); (G.M.); (S.P.); (P.P.)
| | - Diego Chianese
- Department of Medical Sciences, Section of Experimental Medicine, Laboratory for Technologies of Advanced Therapies, University of Ferrara, 44121 Ferrara, Italy; (T.D.M.); (G.P.); (E.D.A.); (M.C.); (D.C.); (C.G.); (G.M.); (S.P.); (P.P.)
| | - Carlotta Giorgi
- Department of Medical Sciences, Section of Experimental Medicine, Laboratory for Technologies of Advanced Therapies, University of Ferrara, 44121 Ferrara, Italy; (T.D.M.); (G.P.); (E.D.A.); (M.C.); (D.C.); (C.G.); (G.M.); (S.P.); (P.P.)
| | - Giampaolo Morciano
- Department of Medical Sciences, Section of Experimental Medicine, Laboratory for Technologies of Advanced Therapies, University of Ferrara, 44121 Ferrara, Italy; (T.D.M.); (G.P.); (E.D.A.); (M.C.); (D.C.); (C.G.); (G.M.); (S.P.); (P.P.)
| | - Simone Patergnani
- Department of Medical Sciences, Section of Experimental Medicine, Laboratory for Technologies of Advanced Therapies, University of Ferrara, 44121 Ferrara, Italy; (T.D.M.); (G.P.); (E.D.A.); (M.C.); (D.C.); (C.G.); (G.M.); (S.P.); (P.P.)
| | - Paolo Pinton
- Department of Medical Sciences, Section of Experimental Medicine, Laboratory for Technologies of Advanced Therapies, University of Ferrara, 44121 Ferrara, Italy; (T.D.M.); (G.P.); (E.D.A.); (M.C.); (D.C.); (C.G.); (G.M.); (S.P.); (P.P.)
- Center of Research for Innovative Therapies in Cystic Fibrosis, University of Ferrara, 44121 Ferrara, Italy
| | - Alessandro Rimessi
- Department of Medical Sciences, Section of Experimental Medicine, Laboratory for Technologies of Advanced Therapies, University of Ferrara, 44121 Ferrara, Italy; (T.D.M.); (G.P.); (E.D.A.); (M.C.); (D.C.); (C.G.); (G.M.); (S.P.); (P.P.)
- Center of Research for Innovative Therapies in Cystic Fibrosis, University of Ferrara, 44121 Ferrara, Italy
| |
Collapse
|
7
|
Ulpiano C, da Silva CL, Monteiro GA. Bioengineered Mesenchymal-Stromal-Cell-Derived Extracellular Vesicles as an Improved Drug Delivery System: Methods and Applications. Biomedicines 2023; 11:biomedicines11041231. [PMID: 37189850 DOI: 10.3390/biomedicines11041231] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 03/30/2023] [Accepted: 04/12/2023] [Indexed: 05/17/2023] Open
Abstract
Extracellular vesicles (EVs) are cell-derived nano-sized lipid membranous structures that modulate cell-cell communication by transporting a variety of biologically active cellular components. The potential of EVs in delivering functional cargos to targeted cells, their capacity to cross biological barriers, as well as their high modification flexibility, make them promising drug delivery vehicles for cell-free therapies. Mesenchymal stromal cells (MSCs) are known for their great paracrine trophic activity, which is largely sustained by the secretion of EVs. MSC-derived EVs (MSC-EVs) retain important features of the parental cells and can be bioengineered to improve their therapeutic payload and target specificity, demonstrating increased therapeutic potential in numerous pre-clinical animal models, including in the treatment of cancer and several degenerative diseases. Here, we review the fundamentals of EV biology and the bioengineering strategies currently available to maximize the therapeutic value of EVs, focusing on their cargo and surface manipulation. Then, a comprehensive overview of the methods and applications of bioengineered MSC-EVs is presented, while discussing the technical hurdles yet to be addressed before their clinical translation as therapeutic agents.
Collapse
Affiliation(s)
- Cristiana Ulpiano
- Department of Bioengineering and iBB-Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal
- Associate Laboratory i4HB-Institute for Health and Bioeconomy at Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal
| | - Cláudia L da Silva
- Department of Bioengineering and iBB-Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal
- Associate Laboratory i4HB-Institute for Health and Bioeconomy at Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal
| | - Gabriel A Monteiro
- Department of Bioengineering and iBB-Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal
- Associate Laboratory i4HB-Institute for Health and Bioeconomy at Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal
| |
Collapse
|
8
|
Soto-Vázquez YM, Genschmer KR. Impact of extracellular vesicles on the pathogenesis, diagnosis, and potential therapy in cardiopulmonary disease. Front Pharmacol 2023; 14:1081015. [PMID: 36891265 PMCID: PMC9986338 DOI: 10.3389/fphar.2023.1081015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 02/08/2023] [Indexed: 02/22/2023] Open
Abstract
Cardiopulmonary diseases span a wide breadth of conditions affecting both heart and lung, the burden of which is globally significant. Chronic pulmonary disease and cardiovascular disease are two of the leading causes of morbidity and mortality worldwide. This makes it critical to understand disease pathogenesis, thereby providing new diagnostic and therapeutic avenues to improve clinical outcomes. Extracellular vesicles provide insight into all three of these features of the disease. Extracellular vesicles are membrane-bound vesicles released by a multitude, if not all, cell types and are involved in multiple physiological and pathological processes that play an important role in intercellular communication. They can be isolated from bodily fluids, such as blood, urine, and saliva, and their contents include a variety of proteins, proteases, and microRNA. These vesicles have shown to act as effective transmitters of biological signals within the heart and lung and have roles in the pathogenesis and diagnosis of multiple cardiopulmonary diseases as well as demonstrate potential as therapeutic agents to treat said conditions. In this review article, we will discuss the role these extracellular vesicles play in the diagnosis, pathogenesis, and therapeutic possibilities of cardiovascular, pulmonary, and infection-related cardiopulmonary diseases.
Collapse
Affiliation(s)
- Yixel M Soto-Vázquez
- Department of Medicine, Division of Pulmonary, Allergy & Critical Care Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Kristopher R Genschmer
- Department of Medicine, Division of Pulmonary, Allergy & Critical Care Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| |
Collapse
|
9
|
Abstract
Chronic obstructive pulmonary disease (COPD) is a complex, heterogeneous, smoking-related disease of significant global impact. The complex biology of COPD is ultimately driven by a few interrelated processes, including proteolytic tissue remodeling, innate immune inflammation, derangements of the host-pathogen response, aberrant cellular phenotype switching, and cellular senescence, among others. Each of these processes are engendered and perpetuated by cells modulating their environment or each other. Extracellular vesicles (EVs) are powerful effectors that allow cells to perform a diverse array of functions on both adjacent and distant tissues, and their pleiotropic nature is only beginning to be appreciated. As such, EVs are candidates to play major roles in these fundamental mechanisms of disease behind COPD. Furthermore, some such roles for EVs are already established, and EVs are implicated in significant aspects of COPD pathogenesis. Here, we discuss known and potential ways that EVs modulate the environment of their originating cells to contribute to the processes that underlie COPD.
Collapse
Affiliation(s)
- Derek W Russell
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA;
- Birmingham VA Medical Center, Birmingham, Alabama, USA
| | - Kristopher R Genschmer
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA;
| | - J Edwin Blalock
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA;
| |
Collapse
|
10
|
Le Saux S, Aubert-Pouëssel A, Mohamed KE, Martineau P, Guglielmi L, Devoisselle JM, Legrand P, Chopineau J, Morille M. Interest of extracellular vesicles in regards to lipid nanoparticle based systems for intracellular protein delivery. Adv Drug Deliv Rev 2021; 176:113837. [PMID: 34144089 DOI: 10.1016/j.addr.2021.113837] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 06/04/2021] [Accepted: 06/12/2021] [Indexed: 12/14/2022]
Abstract
Compared to chemicals that continue to dominate the overall pharmaceutical market, protein therapeutics offer the advantages of higher specificity, greater activity, and reduced toxicity. While nearly all existing therapeutic proteins were developed against soluble or extracellular targets, the ability for proteins to enter cells and target intracellular compartments can significantly broaden their utility for a myriad of exiting targets. Given their physical, chemical, biological instability that could induce adverse effects, and their limited ability to cross cell membranes, delivery systems are required to fully reveal their biological potential. In this context, as natural protein nanocarriers, extracellular vesicles (EVs) hold great promise. Nevertheless, if not present naturally, bringing an interest protein into EV is not an easy task. In this review, we will explore methods used to load extrinsic protein into EVs and compare these natural vectors to their close synthetic counterparts, liposomes/lipid nanoparticles, to induce intracellular protein delivery.
Collapse
|
11
|
Allan KM, Farrow N, Donnelley M, Jaffe A, Waters SA. Treatment of Cystic Fibrosis: From Gene- to Cell-Based Therapies. Front Pharmacol 2021; 12:639475. [PMID: 33796025 PMCID: PMC8007963 DOI: 10.3389/fphar.2021.639475] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Accepted: 01/27/2021] [Indexed: 12/11/2022] Open
Abstract
Prognosis of patients with cystic fibrosis (CF) varies extensively despite recent advances in targeted therapies that improve CF transmembrane conductance regulator (CFTR) function. Despite being a multi-organ disease, extensive lung tissue destruction remains the major cause of morbidity and mortality. Progress towards a curative treatment strategy that implements a CFTR gene addition-technology to the patients’ lungs has been slow and not yet developed beyond clinical trials. Improved delivery vectors are needed to overcome the body’s defense system and ensure an efficient and consistent clinical response before gene therapy is suitable for clinical care. Cell-based therapy–which relies on functional modification of allogenic or autologous cells ex vivo, prior to transplantation into the patient–is now a therapeutic reality for various diseases. For CF, pioneering research has demonstrated proof-of-principle for allogenic transplantation of cultured human airway stem cells into mouse airways. However, applying a cell-based therapy to the human airways has distinct challenges. We review CF gene therapies using viral and non-viral delivery strategies and discuss current advances towards autologous cell-based therapies. Progress towards identification, correction, and expansion of a suitable regenerative cell, as well as refinement of pre-cell transplant lung conditioning protocols is discussed.
Collapse
Affiliation(s)
- Katelin M Allan
- School of Women's and Children's Health, Faculty of Medicine, University of New South Wales, Sydney, Australia.,Molecular and Integrative Cystic Fibrosis Research Centre (miCF_RC), University of New South Wales and Sydney Children's Hospital, Sydney, Australia
| | - Nigel Farrow
- Respiratory and Sleep Medicine, Women's and Children's Health Network, Adelaide, Australia.,Robinson Research Institute, The University of Adelaide, Adelaide, Australia.,Adelaide Medical School, The University of Adelaide, Adelaide, Australia
| | - Martin Donnelley
- Respiratory and Sleep Medicine, Women's and Children's Health Network, Adelaide, Australia.,Robinson Research Institute, The University of Adelaide, Adelaide, Australia.,Adelaide Medical School, The University of Adelaide, Adelaide, Australia
| | - Adam Jaffe
- School of Women's and Children's Health, Faculty of Medicine, University of New South Wales, Sydney, Australia.,Molecular and Integrative Cystic Fibrosis Research Centre (miCF_RC), University of New South Wales and Sydney Children's Hospital, Sydney, Australia.,Department of Respiratory Medicine, Sydney Children's Hospital, Sydney, Australia
| | - Shafagh A Waters
- School of Women's and Children's Health, Faculty of Medicine, University of New South Wales, Sydney, Australia.,Molecular and Integrative Cystic Fibrosis Research Centre (miCF_RC), University of New South Wales and Sydney Children's Hospital, Sydney, Australia.,Department of Respiratory Medicine, Sydney Children's Hospital, Sydney, Australia
| |
Collapse
|
12
|
Abreu SC, Lopes-Pacheco M, Weiss DJ, Rocco PRM. Mesenchymal Stromal Cell-Derived Extracellular Vesicles in Lung Diseases: Current Status and Perspectives. Front Cell Dev Biol 2021; 9:600711. [PMID: 33659247 PMCID: PMC7917181 DOI: 10.3389/fcell.2021.600711] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2020] [Accepted: 01/11/2021] [Indexed: 12/11/2022] Open
Abstract
Extracellular vesicles (EVs) have emerged as a potential therapy for several diseases. These plasma membrane-derived fragments are released constitutively by virtually all cell types-including mesenchymal stromal cells (MSCs)-under stimulation or following cell-to-cell interaction, which leads to activation or inhibition of distinct signaling pathways. Based on their size, intracellular origin, and secretion pathway, EVs have been grouped into three main populations: exosomes, microvesicles (or microparticles), and apoptotic bodies. Several molecules can be found inside MSC-derived EVs, including proteins, lipids, mRNA, microRNAs, DNAs, as well as organelles that can be transferred to damaged recipient cells, thus contributing to the reparative process and promoting relevant anti-inflammatory/resolutive actions. Indeed, the paracrine/endocrine actions induced by MSC-derived EVs have demonstrated therapeutic potential to mitigate or even reverse tissue damage, thus raising interest in the regenerative medicine field, particularly for lung diseases. In this review, we summarize the main features of EVs and the current understanding of the mechanisms of action of MSC-derived EVs in several lung diseases, such as chronic obstructive pulmonary disease (COPD), pulmonary infections [including coronavirus disease 2019 (COVID-19)], asthma, acute respiratory distress syndrome (ARDS), idiopathic pulmonary fibrosis (IPF), and cystic fibrosis (CF), among others. Finally, we list a number of limitations associated with this therapeutic strategy that must be overcome in order to translate effective EV-based therapies into clinical practice.
Collapse
Affiliation(s)
- Soraia C. Abreu
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- National Institute of Science and Technology for Regenerative Medicine, Rio de Janeiro, Brazil
| | - Miquéias Lopes-Pacheco
- Biosystems & Integrative Sciences Institute, Faculty of Sciences, University of Lisbon, Lisbon, Portugal
| | - Daniel J. Weiss
- Department of Medicine, College of Medicine, University of Vermont Larner, Burlington, VT, United States
| | - Patricia R. M. Rocco
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- National Institute of Science and Technology for Regenerative Medicine, Rio de Janeiro, Brazil
| |
Collapse
|
13
|
Han Y, Jones TW, Dutta S, Zhu Y, Wang X, Narayanan SP, Fagan SC, Zhang D. Overview and Update on Methods for Cargo Loading into Extracellular Vesicles. Processes (Basel) 2021; 9. [PMID: 33954091 PMCID: PMC8096148 DOI: 10.3390/pr9020356] [Citation(s) in RCA: 74] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The enormous library of pharmaceutical compounds presents endless research avenues. However, several factors limit the therapeutic potential of these drugs, such as drug resistance, stability, off-target toxicity, and inadequate delivery to the site of action. Extracellular vesicles (EVs) are lipid bilayer-delimited particles and are naturally released from cells. Growing evidence shows that EVs have great potential to serve as effective drug carriers. Since EVs can not only transfer biological information, but also effectively deliver hydrophobic drugs into cells, the application of EVs as a novel drug delivery system has attracted considerable scientific interest. Recently, EVs loaded with siRNA, miRNA, mRNA, CRISPR/Cas9, proteins, or therapeutic drugs show improved delivery efficiency and drug effect. In this review, we summarize the methods used for the cargo loading into EVs, including siRNA, miRNA, mRNA, CRISPR/Cas9, proteins, and therapeutic drugs. Furthermore, we also include the recent advance in engineered EVs for drug delivery. Finally, both advantages and challenges of EVs as a new drug delivery system are discussed. Here, we encourage researchers to further develop convenient and reliable loading methods for the potential clinical applications of EVs as drug carriers in the future.
Collapse
Affiliation(s)
- Yohan Han
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia and Charlie Norwood VA Medical Center, Augusta, GA 30912, USA
| | - Timothy W. Jones
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia and Charlie Norwood VA Medical Center, Augusta, GA 30912, USA
| | - Saugata Dutta
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia and Charlie Norwood VA Medical Center, Augusta, GA 30912, USA
| | - Yin Zhu
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia and Charlie Norwood VA Medical Center, Augusta, GA 30912, USA
| | - Xiaoyun Wang
- Center for Vaccines and Immunology, University of Georgia, Athens, GA 30602, USA
| | - S. Priya Narayanan
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia and Charlie Norwood VA Medical Center, Augusta, GA 30912, USA
- Vascular Biology Center, Augusta University, Augusta, GA 30912, USA
- James and Jean Culver Vision Discovery Institute, Augusta University, Augusta, GA 30912, USA
| | - Susan C. Fagan
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia and Charlie Norwood VA Medical Center, Augusta, GA 30912, USA
| | - Duo Zhang
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia and Charlie Norwood VA Medical Center, Augusta, GA 30912, USA
- Vascular Biology Center, Augusta University, Augusta, GA 30912, USA
- Correspondence: ; Tel.: +1-706-721-6491; Fax: +1-706-721-3994
| |
Collapse
|
14
|
Shrivastava S, Morris KV. The Multifunctionality of Exosomes; from the Garbage Bin of the Cell to a Next Generation Gene and Cellular Therapy. Genes (Basel) 2021; 12:genes12020173. [PMID: 33513776 PMCID: PMC7912150 DOI: 10.3390/genes12020173] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2020] [Revised: 01/23/2021] [Accepted: 01/24/2021] [Indexed: 02/07/2023] Open
Abstract
Exosomes are packaged with a variety of cellular cargo including RNA, DNA, lipids and proteins. For several decades now there has been ongoing debate as to what extent exosomes are the garbage bin of the cell or if these entities function as a distributer of cellular cargo which acts in a meaningful mechanistic way on target cells. Are the contents of exosomes unwanted excess cellular produce or are they selective nucleic acid packaged nanoparticles used to communicate in a paracrine fashion? Overexpressed RNAs and fragments of DNA have been shown to collect into exosomes which are jettisoned from cells in response to particular stimuli to maintain homeostasis suggesting exosomes are functional trash bins of the cell. Other studies however have deciphered selective packaging of particular nucleic acids into exosomes. Nucleic acids packaged into exosomes are increasingly reported to exert transcriptional control on recipient cells, supporting the notion that exosomes may provide a role in signaling and intracellular communication. We survey the literature and conclude that exosomes are multifunctional entities, with a plethora of roles that can each be taken advantage to functionally modulate cells. We also note that the potential utility of developing exosomes as a next generation genetic therapy may in future transform cellular therapies. We also depict three models of methodologies which can be adopted by researchers intending to package nucleic acid in exosomes for developing gene and cell therapy.
Collapse
Affiliation(s)
- Surya Shrivastava
- Center for Gene Therapy, City of Hope-Beckman Research Institute, Duarte, CA 91010, USA;
- Hematological Malignancy and Stem Cell Transplantation Institute at the City of Hope, Duarte, CA 91010, USA
| | - Kevin V. Morris
- Center for Gene Therapy, City of Hope-Beckman Research Institute, Duarte, CA 91010, USA;
- Hematological Malignancy and Stem Cell Transplantation Institute at the City of Hope, Duarte, CA 91010, USA
- School of Medical Science, Gold Coast Campus, Griffith University, Southport 4222, Australia
- Correspondence:
| |
Collapse
|
15
|
Delivery of genome-editing biomacromolecules for treatment of lung genetic disorders. Adv Drug Deliv Rev 2021; 168:196-216. [PMID: 32416111 DOI: 10.1016/j.addr.2020.05.002] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 04/28/2020] [Accepted: 05/08/2020] [Indexed: 02/06/2023]
Abstract
Genome-editing systems based on clustered, regularly interspaced, short palindromic repeat (CRISPR)/associated protein (CRISPR/Cas), are emerging as a revolutionary technology for the treatment of various genetic diseases. To date, the delivery of genome-editing biomacromolecules by viral or non-viral vectors have been proposed as new therapeutic options for lung genetic disorders, such as cystic fibrosis (CF) and α-1 antitrypsin deficiency (AATD), and it has been accepted that these delivery vectors can introduce CRISPR/Cas9 machineries into target cells or tissues in vitro, ex vivo and in vivo. However, the efficient local or systemic delivery of CRISPR/Cas9 elements to the lung, enabled by either viral or by non-viral carriers, still remains elusive. Herein, we first introduce lung genetic disorders and their current treatment options, and then summarize CRISPR/Cas9-based strategies for the therapeutic genome editing of these disorders. We further summarize the pros and cons of different routes of administration for lung genetic disorders. In particular, the potentials of aerosol delivery for therapeutic CRISPR/Cas9 biomacromolecules for lung genome editing are discussed and highlighted. Finally, current challenges and future outlooks in this emerging area are briefly discussed.
Collapse
|
16
|
Da Silva Sanchez A, Paunovska K, Cristian A, Dahlman JE. Treating Cystic Fibrosis with mRNA and CRISPR. Hum Gene Ther 2020; 31:940-955. [PMID: 32799680 PMCID: PMC7495921 DOI: 10.1089/hum.2020.137] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Accepted: 08/13/2020] [Indexed: 12/16/2022] Open
Abstract
Less than 20% of the protein coding genome is thought to be targetable using small molecules. mRNA therapies are not limited in the same way since in theory, they can silence or edit any gene by encoding CRISPR nucleases, or alternatively, produce any missing protein. Yet not all mRNA therapies are equally likely to succeed. Over the past several years, an increasing number of clinical trials with siRNA- and antisense oligonucleotide-based drugs have revealed three key concepts that will likely extend to mRNA therapies delivered by nonviral systems. First, scientists have come to understand that some genes make better targets for RNA therapies than others. Second, scientists have learned that the type and position of chemical modifications made to an RNA drug can alter its therapeutic window, toxicity, and bioavailability. Third, scientists have found that safe and targeted drug delivery vehicles are required to ferry mRNA therapies into diseased cells. In this study, we apply these learnings to cystic fibrosis (CF). We also describe lessons learned from a subset of CF gene therapies that have already been tested in patients. Finally, we highlight the scientific advances that are still required for nonviral mRNA- or CRISPR-based drugs to treat CF successfully in patients.
Collapse
Affiliation(s)
- Alejandro Da Silva Sanchez
- Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, Georgia, USA
- School of Chemical & Biomolecular Engineering, Georgia Institute of Technology, Atlanta, Georgia, USA
| | - Kalina Paunovska
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, Georgia, USA
| | - Ana Cristian
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, Georgia, USA
| | - James E. Dahlman
- Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, Georgia, USA
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, Georgia, USA
| |
Collapse
|
17
|
Shi X, Cheng Q, Zhang Y. Reprogramming extracellular vesicles with engineered proteins. Methods 2020; 177:95-102. [PMID: 31568822 DOI: 10.1016/j.ymeth.2019.09.017] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Revised: 09/13/2019] [Accepted: 09/25/2019] [Indexed: 12/15/2022] Open
Abstract
Extracellular vesicles (EVs) have been emerging as a new class of cell-free therapy for the treatment of a variety of diseases, including cancer, tissue injuries, and inflammatory diseases. Reprograming native EVs by genetic engineering and other approaches offers an attractive prospect of extending therapeutic capabilities of EVs beyond their natural functions and properties. In this review article, we survey the state-of-the-art methods of EVs engineering and summarize major therapeutic applications of the reprogrammed EVs.
Collapse
Affiliation(s)
- Xiaojing Shi
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA 90089, USA
| | - Qinqin Cheng
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA 90089, USA
| | - Yong Zhang
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA 90089, USA; Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA 90089, USA; Department of Chemistry, Dornsife College of Letters, Arts and Sciences, University of Southern California, Los Angeles, CA 90089, USA; Research Center for Liver Diseases, University of Southern California, Los Angeles, CA 90089, USA.
| |
Collapse
|
18
|
Useckaite Z, Ward MP, Trappe A, Reilly R, Lennon J, Davage H, Matallanas D, Cassidy H, Dillon ET, Brennan K, Doyle SL, Carter S, Donnelly S, Linnane B, McKone EF, McNally P, Coppinger JA. Increased extracellular vesicles mediate inflammatory signalling in cystic fibrosis. Thorax 2020; 75:449-458. [PMID: 32265339 PMCID: PMC7279202 DOI: 10.1136/thoraxjnl-2019-214027] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 03/09/2020] [Accepted: 03/20/2020] [Indexed: 02/06/2023]
Abstract
Rationale Mutations in the cystic fibrosis transmembrane regulator (CFTR) gene form the basis of cystic fibrosis (CF). There remains an important knowledge gap in CF as to how diminished CFTR activity leads to the dominant inflammatory response within CF airways. Objectives To investigate if extracellular vesicles (EVs) contribute to inflammatory signalling in CF. Methods EVs released from CFBE41o-, CuFi-5, 16HBE14o- and NuLi-1 cells were characterised by nanoparticle tracking analysis (NTA). EVs isolated from bronchoalveolar lavage fluid (BALF) from 30 people with CF (PWCF) were analysed by NTA and mass spectrometry and compared with controls. Neutrophils were isolated from the blood of 8 PWCF to examine neutrophil migration in the presence of CFBE41o- EVs. Results A significantly higher level of EVs were released from CFBE41o- (p<0.0001) and CuFi-5 (p=0.0209) relative to control cell lines. A significantly higher level of EVs were detected in BALF of PWCF, in three different age groups relative to controls (p=0.01, 0.001, 0.002). A significantly lower level of EVs were released from CFBE41o- (p<0.001) and CuFi-5 (p=0.0002) cell lines treated with CFTR modulators. Significant changes in the protein expression of 126 unique proteins was determined in EVs obtained from the BALF of PWCF of different age groups (p<0.001–0.05). A significant increase in chemotaxis of neutrophils derived from PWCF was observed in the presence of CFBE41o EVs (p=0.0024) compared with controls. Conclusion This study demonstrates that EVs are produced in CF airway cells, have differential protein expression at different ages and drive neutrophil recruitment in CF.
Collapse
Affiliation(s)
- Zivile Useckaite
- School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin 2, Ireland.,National Children's Research Centre, Children's Health Ireland (CHI) at Crumlin, Dublin 12, Ireland
| | - Mark P Ward
- School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin 2, Ireland.,National Children's Research Centre, Children's Health Ireland (CHI) at Crumlin, Dublin 12, Ireland
| | - Anne Trappe
- School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin 2, Ireland.,National Children's Research Centre, Children's Health Ireland (CHI) at Crumlin, Dublin 12, Ireland
| | - Rebecca Reilly
- National Children's Research Centre, Children's Health Ireland (CHI) at Crumlin, Dublin 12, Ireland
| | - Jenny Lennon
- National Children's Research Centre, Children's Health Ireland (CHI) at Crumlin, Dublin 12, Ireland
| | - Holly Davage
- National Children's Research Centre, Children's Health Ireland (CHI) at Crumlin, Dublin 12, Ireland
| | - David Matallanas
- Systems Biology Ireland, UCD School of Medicine and Medical Sciences, University College Dublin, Dublin 4, Ireland
| | - Hilary Cassidy
- Systems Biology Ireland, UCD School of Medicine and Medical Sciences, University College Dublin, Dublin 4, Ireland.,School of Biomolecular and Biomedical Sciences, University College Dublin, Dublin 4, Ireland
| | - Eugene T Dillon
- UCD Conway Institute, University College Dublin, Dublin 4, Ireland
| | - Kiva Brennan
- National Children's Research Centre, Children's Health Ireland (CHI) at Crumlin, Dublin 12, Ireland.,Clinical Medicine, School of Medicine, Trinity College Dublin, Dublin 2, Ireland
| | - Sarah L Doyle
- National Children's Research Centre, Children's Health Ireland (CHI) at Crumlin, Dublin 12, Ireland.,Clinical Medicine, School of Medicine, Trinity College Dublin, Dublin 2, Ireland
| | | | - Seamas Donnelly
- Clinical Medicine, School of Medicine, Trinity College Dublin, Dublin 2, Ireland
| | - Barry Linnane
- National Children's Research Centre, Children's Health Ireland (CHI) at Crumlin, Dublin 12, Ireland.,Paediatrics, University Hospital Limerick, Limerick, Ireland
| | - Edward F McKone
- UCD Conway Institute, University College Dublin, Dublin 4, Ireland.,St. Vincent's University Hospital, Dublin 4, Ireland
| | - Paul McNally
- National Children's Research Centre, Children's Health Ireland (CHI) at Crumlin, Dublin 12, Ireland.,Paediatrics, Royal College of Surgeons in Ireland, Dublin 2, Ireland
| | - Judith A Coppinger
- School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin 2, Ireland .,National Children's Research Centre, Children's Health Ireland (CHI) at Crumlin, Dublin 12, Ireland
| |
Collapse
|
19
|
De Santi C, Fernández Fernández E, Gaul R, Vencken S, Glasgow A, Oglesby IK, Hurley K, Hawkins F, Mitash N, Mu F, Raoof R, Henshall DC, Cutrona MB, Simpson JC, Harvey BJ, Linnane B, McNally P, Cryan SA, MacLoughlin R, Swiatecka-Urban A, Greene CM. Precise Targeting of miRNA Sites Restores CFTR Activity in CF Bronchial Epithelial Cells. Mol Ther 2020; 28:1190-1199. [PMID: 32059764 DOI: 10.1016/j.ymthe.2020.02.001] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2019] [Accepted: 03/02/2020] [Indexed: 01/19/2023] Open
Abstract
MicroRNAs that are overexpressed in cystic fibrosis (CF) bronchial epithelial cells (BEC) negatively regulate CFTR and nullify the beneficial effects of CFTR modulators. We hypothesized that it is possible to reverse microRNA-mediated inhibition of CFTR using CFTR-specific target site blockers (TSBs) and to develop a drug-device combination inhalation therapy for CF. Lead microRNA expression was quantified in a series of human CF and non-CF samples and in vitro models. A panel of CFTR 3' untranslated region (UTR)-specific locked nucleic acid antisense oligonucleotide TSBs was assessed for their ability to increase CFTR expression. Their effects on CFTR activity alone or in combination with CFTR modulators were measured in CF BEC models. TSB encapsulation in poly-lactic-co-glycolic acid (PLGA) nanoparticles was assessed as a proof of principle of delivery into CF BECs. TSBs targeting the CFTR 3' UTR 298-305:miR-145-5p or 166-173:miR-223-3p sites increased CFTR expression and anion channel activity and enhanced the effects of ivacaftor/lumacaftor or ivacaftor/tezacaftor in CF BECs. Biocompatible PLGA-TSB nanoparticles promoted CFTR expression in primary BECs and retained desirable biophysical characteristics following nebulization. Alone or in combination with CFTR modulators, aerosolized CFTR-targeting TSBs encapsulated in PLGA nanoparticles could represent a promising drug-device combination therapy for the treatment for CFTR dysfunction in the lung.
Collapse
Affiliation(s)
- Chiara De Santi
- Department of Clinical Microbiology, Royal College of Surgeons in Ireland, Dublin 9, Ireland.
| | | | - Rachel Gaul
- School of Pharmacy and Tissue Engineering Research Group, Royal College of Surgeons in Ireland, Dublin 2, Ireland
| | - Sebastian Vencken
- Department of Clinical Microbiology, Royal College of Surgeons in Ireland, Dublin 9, Ireland
| | - Arlene Glasgow
- Department of Clinical Microbiology, Royal College of Surgeons in Ireland, Dublin 9, Ireland
| | - Irene K Oglesby
- Department of Medicine, Royal College of Surgeons in Ireland, Dublin 9, Ireland
| | - Killian Hurley
- Department of Medicine, Royal College of Surgeons in Ireland, Dublin 9, Ireland
| | - Finn Hawkins
- Center for Regenerative Medicine, Boston University and Boston Medical Center, Boston, MA 02118, USA; The Pulmonary Center and Department of Medicine, Boston University School of Medicine, Boston, MA 02118, USA
| | - Nilay Mitash
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Fangping Mu
- Center for Research Computing, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Rana Raoof
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin 2, Ireland
| | - David C Henshall
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin 2, Ireland
| | - Meritxell B Cutrona
- School of Biology and Environmental Science, Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin 4, Ireland
| | - Jeremy C Simpson
- School of Biology and Environmental Science, Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin 4, Ireland
| | - Brian J Harvey
- Department of Molecular Medicine, Royal College of Surgeons in Ireland, Dublin 9, Ireland
| | - Barry Linnane
- University Hospital Limerick, Dooradoyle, Limerick, Ireland
| | - Paul McNally
- Department of Pediatrics, Royal College of Surgeons in Ireland, Dublin 2, Ireland; National Children's Research Centre, Children's Health Ireland at Crumlin, Dublin 12, Ireland
| | - Sally Ann Cryan
- School of Pharmacy and Tissue Engineering Research Group, Royal College of Surgeons in Ireland, Dublin 2, Ireland
| | | | | | - Catherine M Greene
- Department of Clinical Microbiology, Royal College of Surgeons in Ireland, Dublin 9, Ireland
| |
Collapse
|
20
|
Villamizar O, Waters SA, Scott T, Saayman S, Grepo N, Urak R, Davis A, Jaffe A, Morris KV. Targeted Activation of Cystic Fibrosis Transmembrane Conductance Regulator. Mol Ther 2019; 27:1737-1748. [PMID: 31383454 PMCID: PMC6822231 DOI: 10.1016/j.ymthe.2019.07.002] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2019] [Revised: 06/24/2019] [Accepted: 07/03/2019] [Indexed: 01/05/2023] Open
Abstract
Cystic fibrosis (CF) is caused by mutations in the CF transmembrane conductance regulator (CFTR) gene. The majority of CFTR mutations result in impaired chloride channel function as only a fraction of the mutated CFTR reaches the plasma membrane. The development of a therapeutic approach that facilitates increased cell-surface expression of CFTR could prove clinically relevant. Here, we evaluate and contrast two molecular approaches to activate CFTR expression. We find that an RNA-guided nuclease null Cas9 (dCas9) fused with a tripartite activator, VP64-p65-Rta can activate endogenous CFTR in cultured human nasal epithelial cells from CF patients. We also find that targeting BGas, a long non-coding RNA involved in transcriptionally modulating CFTR expression with a gapmer, induced both strong knockdown of BGas and concordant activation of CFTR. Notably, the gapmer can be delivered to target cells when generated as electrostatic particles with recombinant HIV-Tat cell penetrating peptide (CPP), when packaged into exosomes, or when loaded into lipid nanoparticles (LNPs). Treatment of patient-derived human nasal epithelial cells containing F508del with gapmer-CPP, gapmer-exosomes, or LNPs resulted in increased expression and function of CFTR. Collectively, these observations suggest that CRISPR/dCas-VPR (CRISPR) and BGas-gapmer approaches can target and specifically activate CFTR.
Collapse
Affiliation(s)
- Olga Villamizar
- Center for Gene Therapy, City of Hope-Beckman Research Institute at the City of Hope, Duarte, CA 91010, USA
| | - Shafagh A Waters
- Faculty of Medicine, School of Women's & Children's Health, University of New South Wales (UNSW), Sydney, NSW, Australia; Molecular and Integrative Cystic Fibrosis Research Centre (miCF_RC), School of Women's & Children's Health, Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia
| | - Tristan Scott
- Center for Gene Therapy, City of Hope-Beckman Research Institute at the City of Hope, Duarte, CA 91010, USA
| | - Sheena Saayman
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, CA, USA
| | - Nicole Grepo
- Center for Gene Therapy, City of Hope-Beckman Research Institute at the City of Hope, Duarte, CA 91010, USA
| | - Ryan Urak
- Center for Gene Therapy, City of Hope-Beckman Research Institute at the City of Hope, Duarte, CA 91010, USA
| | - Alicia Davis
- Center for Gene Therapy, City of Hope-Beckman Research Institute at the City of Hope, Duarte, CA 91010, USA
| | - Adam Jaffe
- Faculty of Medicine, School of Women's & Children's Health, University of New South Wales (UNSW), Sydney, NSW, Australia; Molecular and Integrative Cystic Fibrosis Research Centre (miCF_RC), School of Women's & Children's Health, Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia; Department of Respiratory Medicine, Sydney Children's Hospital, Sydney, NSW, Australia
| | - Kevin V Morris
- Center for Gene Therapy, City of Hope-Beckman Research Institute at the City of Hope, Duarte, CA 91010, USA.
| |
Collapse
|
21
|
Wiklander OPB, Brennan MÁ, Lötvall J, Breakefield XO, El Andaloussi S. Advances in therapeutic applications of extracellular vesicles. Sci Transl Med 2019; 11:eaav8521. [PMID: 31092696 PMCID: PMC7104415 DOI: 10.1126/scitranslmed.aav8521] [Citation(s) in RCA: 675] [Impact Index Per Article: 112.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Accepted: 04/26/2019] [Indexed: 12/18/2022]
Abstract
Extracellular vesicles (EVs) are nanometer-sized, lipid membrane-enclosed vesicles secreted by most, if not all, cells and contain lipids, proteins, and various nucleic acid species of the source cell. EVs act as important mediators of intercellular communication that influence both physiological and pathological conditions. Given their ability to transfer bioactive components and surmount biological barriers, EVs are increasingly being explored as potential therapeutic agents. EVs can potentiate tissue regeneration, participate in immune modulation, and function as potential alternatives to stem cell therapy, and bioengineered EVs can act as delivery vehicles for therapeutic agents. Here, we cover recent approaches and advances of EV-based therapies.
Collapse
Affiliation(s)
- Oscar P B Wiklander
- Department of Laboratory Medicine, Karolinska Institutet, 141 86 Stockholm, Sweden.
- Evox Therapeutics Limited, Medawar Centre, Robert Robinson Avenue, Oxford OX4 4HG, UK
| | - Meadhbh Á Brennan
- Harvard School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA
- Departments of Neurology and Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02129, USA
- INSERM UMR 1238, PhyOS, Faculty of Medicine, Université de Nantes, 44034 Nantes cedex 1, France
| | - Jan Lötvall
- Krefting Research Centre, Institute of Medicine at Sahlgrenska Academy, University of Gothenburg, 405 30 Gothenburg, Sweden
| | - Xandra O Breakefield
- Departments of Neurology and Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02129, USA
| | - Samir El Andaloussi
- Department of Laboratory Medicine, Karolinska Institutet, 141 86 Stockholm, Sweden.
- Evox Therapeutics Limited, Medawar Centre, Robert Robinson Avenue, Oxford OX4 4HG, UK
| |
Collapse
|
22
|
Letsiou E, Bauer N. Endothelial Extracellular Vesicles in Pulmonary Function and Disease. CURRENT TOPICS IN MEMBRANES 2018; 82:197-256. [PMID: 30360780 DOI: 10.1016/bs.ctm.2018.09.002] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The pulmonary vascular endothelium is involved in the pathogenesis of acute and chronic lung diseases. Endothelial cell (EC)-derived products such as extracellular vesicles (EVs) serve as EC messengers that mediate inflammatory as well as cytoprotective effects. EC-EVs are a broad term, which encompasses exosomes and microvesicles of endothelial origin. EVs are comprised of lipids, nucleic acids, and proteins that reflect not only the cellular origin but also the stimulus that triggered their biogenesis and secretion. This chapter presents an overview of the biology of EC-EVs and summarizes key findings regarding their characteristics, components, and functions. The role of EC-EVs is specifically delineated in pulmonary diseases characterized by endothelial dysfunction, including pulmonary hypertension, acute respiratory distress syndrome and associated conditions, chronic obstructive pulmonary disease, and obstructive sleep apnea.
Collapse
Affiliation(s)
- Eleftheria Letsiou
- Division of Pulmonary Inflammation, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Natalie Bauer
- Department of Pharmacology & Center for Lung Biology, College of Medicine, University of South Alabama, Mobile, AL, United States.
| |
Collapse
|
23
|
Osteikoetxea X, Benke M, Rodriguez M, Pálóczi K, Sódar BW, Szvicsek Z, Szabó-Taylor K, Vukman KV, Kittel Á, Wiener Z, Vékey K, Harsányi L, Szűcs Á, Turiák L, Buzás EI. Detection and proteomic characterization of extracellular vesicles in human pancreatic juice. Biochem Biophys Res Commun 2018; 499:37-43. [PMID: 29550476 DOI: 10.1016/j.bbrc.2018.03.107] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Accepted: 03/13/2018] [Indexed: 02/07/2023]
Abstract
AIMS The prognosis of patients with pancreatic cancer has remained virtually unchanged with a high mortality rate compared to other types of cancers. An earlier detection would provide a time window of opportunity for treatment and prevention of deaths. In the present study we investigated extracellular vesicle (EV)-associated potential biomarkers for pancreatic cancer by directly assessing EV size-based subpopulations in pancreatic juice samples of patients with chronic pancreatitis or pancreatic cancer. In addition, we also studied blood plasma and pancreatic cancer cell line-derived EVs. METHODS Comparative proteomic analysis was performed of 102 EV preparations from human pancreatic juices, blood, and pancreatic cancer cell lines Capan-1 and MIA PaCa-2. EV preparations were also characterized by electron microscopy, tunable resistive pulse sensing, and flow cytometry. RESULTS Here we describe the presence of EVs in human pancreatic juice samples. Pancreatic juice EV-associated proteins that we identified as possible candidate markers for pancreatic cancer included mucins, such as MUC1, MUC4, MUC5AC, MUC6 and MUC16, CFTR, and MDR1 proteins. These candidate biomarkers could also be detected by flow cytometry in EVs found in pancreatic juice and those secreted by pancreatic cancer cell lines. CONCLUSIONS Together our data show that detection and characterization of EVs directly in pancreatic juice is feasible and may prove to be a valuable source of potential biomarkers of pancreatic cancer.
Collapse
Affiliation(s)
- Xabier Osteikoetxea
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, Budapest, Hungary.
| | - Márton Benke
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, Budapest, Hungary; 1st Department of Surgery, Semmelweis University, Budapest, Hungary
| | - Marta Rodriguez
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital-The Norwegian Radium Hospital, Oslo, Norway
| | - Krisztina Pálóczi
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, Budapest, Hungary
| | - Barbara W Sódar
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, Budapest, Hungary
| | - Zsuzsanna Szvicsek
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, Budapest, Hungary
| | - Katalin Szabó-Taylor
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, Budapest, Hungary
| | - Krisztina V Vukman
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, Budapest, Hungary
| | - Ágnes Kittel
- Institute of Experimental Medicine, Hungarian Academy of Sciences, Budapest, Hungary
| | - Zoltán Wiener
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, Budapest, Hungary
| | - Károly Vékey
- Research Centre for Natural Sciences, Hungarian Academy of Sciences, Budapest Hungary
| | - László Harsányi
- 1st Department of Surgery, Semmelweis University, Budapest, Hungary
| | - Ákos Szűcs
- 1st Department of Surgery, Semmelweis University, Budapest, Hungary
| | - Lilla Turiák
- Research Centre for Natural Sciences, Hungarian Academy of Sciences, Budapest Hungary
| | - Edit I Buzás
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, Budapest, Hungary; MTA-SE Immune-Proteogenomics Extracellular Vesicles Research Group, Budapest, Hungary.
| |
Collapse
|
24
|
Horibe T, Okushima N, Torisawa A, Akiyoshi R, Hatta‐Ohashi Y, Suzuki H, Kawakami K. Evaluation of chemical chaperones based on the monitoring of Bip promoter activity and visualization of extracellular vesicles by real-time bioluminescence imaging. LUMINESCENCE 2018; 33:249-255. [PMID: 28929569 PMCID: PMC6084373 DOI: 10.1002/bio.3388] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2017] [Revised: 06/28/2017] [Accepted: 07/25/2017] [Indexed: 01/20/2023]
Abstract
It is known that endoplasmic reticulum (ER) stress in cells and extracellular vesicles (EVs) plays a significant role in cancer cells, therefore the evaluation of compounds that can regulate ER stress and EV secretion would be a suitable system for further screening and development of new drugs. In this study, we evaluated chemical chaperones derived from natural products based on monitoring Bip/GRP78 promoter activity during cancer cell growth, at the level of the single cell, by a bioluminescence microscopy system that had several advantages compared with fluorescence imaging. It was found that several chemical chaperones, such as ferulic acid (FA), silybin, and rutin, affected the activity. We visualized EVs from cancer cells using bioluminescence imaging and showed that several EVs could be observed when using CD63 fused with NanoLuc luciferase, which has a much smaller molecular weight and higher intensity than conventional firefly luciferase. We then examined the effects of the chemical chaperones on EVs from cancer cells by bioluminescence imaging and quantified the expression of CD63 in these EVs. It was found that the chemical chaperones examined in this study affected CD63 levels in EVs. These results showed that imaging at the level of the single cell using bioluminescence is a powerful tool and could be used to evaluate chemical chaperones and EVs from cancer cells. This approach may produce new information in this field when taken together with conventional and classical methods.
Collapse
Affiliation(s)
- Tomohisa Horibe
- Department of Pharmacoepidemiology, Graduate School of Medicine and Public HealthKyoto UniversityKyotoJapan
| | - Nanako Okushima
- Department of Pharmacoepidemiology, Graduate School of Medicine and Public HealthKyoto UniversityKyotoJapan
| | - Aya Torisawa
- Department of Pharmacoepidemiology, Graduate School of Medicine and Public HealthKyoto UniversityKyotoJapan
| | - Ryutaro Akiyoshi
- Evaluation Technology Department 1Olympus CorporationHachioji‐shiTokyoJapan
| | - Yoko Hatta‐Ohashi
- Evaluation Technology Department 1Olympus CorporationHachioji‐shiTokyoJapan
| | - Hirobumi Suzuki
- Evaluation Technology Department 1Olympus CorporationHachioji‐shiTokyoJapan
| | - Koji Kawakami
- Department of Pharmacoepidemiology, Graduate School of Medicine and Public HealthKyoto UniversityKyotoJapan
| |
Collapse
|
25
|
Hart SL, Harrison PT. Genetic therapies for cystic fibrosis lung disease. Curr Opin Pharmacol 2017; 34:119-124. [PMID: 29107808 DOI: 10.1016/j.coph.2017.10.006] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Revised: 10/02/2017] [Accepted: 10/16/2017] [Indexed: 12/26/2022]
Abstract
Gene therapy for cystic fibrosis (CF) has been the subject of intense research over the last twenty-five years or more, using both viral and liposomal delivery methods, but so far without the emergence of a clinical therapy. New approaches to CF gene therapy involving recent improvements to vector systems, both viral and non-viral, as well as new nucleic acid technologies have led to renewed interest in the field. The field of therapeutic gene editing is rapidly developing with the emergence of CRISPR/Cas9 as well as chemically modified mRNA therapeutics. These new types of nucleic acid therapies are also a good fit with delivery by non-viral delivery approaches which has led to a renewed interest in lipid-based and other nanoformulations.
Collapse
Affiliation(s)
- Stephen L Hart
- Experimental and Personalised Medicines Section, Genetics and Genomic Medicine Programme, UCL Great Ormond Street Institute of Child Health, 30 Guilford St, London WC1N 1EH, UK.
| | - Patrick T Harrison
- Department of Physiology, BioSciences Institute, University College Cork, Cork, Ireland
| |
Collapse
|
26
|
Stranford DM, Hung ME, Gargus ES, Shah RN, Leonard JN. A Systematic Evaluation of Factors Affecting Extracellular Vesicle Uptake by Breast Cancer Cells. Tissue Eng Part A 2017; 23:1274-1282. [PMID: 28586292 DOI: 10.1089/ten.tea.2017.0158] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Extracellular vesicles (EVs) are nanometer-scale particles that are secreted by cells and mediate intercellular communication by transferring biomolecules between cells. Harnessing this mechanism for therapeutic biomolecule delivery represents a promising frontier for regenerative medicine and other clinical applications. One challenge to realizing this goal is that to date, our understanding of which factors affect EV uptake by recipient cells remains incomplete. In this study, we systematically investigated such delivery questions in the context of breast cancer cells, which are one of the most well-studied cell types with respect to EV delivery and therefore comprise a facile model system for this investigation. By displaying various targeting peptides on the EV surface, we observed that although displaying GE11 on EVs modestly increased uptake by MCF-7 cells, neuropeptide Y (NPY) display had no effect on uptake by the same cells. In contrast, neurotensin (NTS) and urokinase plasminogen activator (uPA) display reduced EV uptake by MDA-MB-231 cells. Interestingly, EV uptake rate did not depend on the source of the EVs; breast cancer cells demonstrated no increase in uptake on administration of breast cancer-derived EVs in comparison to HEK293FT-derived EVs. Moreover, EV uptake was greatly enhanced by delivery in the presence of polybrene and spinoculation, suggesting that maximal EV uptake rates are much greater than those observed under basal conditions in cell culture. By investigating how the cell's environment might provide cues that impact EV uptake, we also observed that culturing cells on soft matrices significantly enhanced EV uptake, compared to culturing on stiff tissue culture polystyrene. Each of these observations provides insights into the factors impacting EV uptake by breast cancer cells, while also providing a basis of comparison for systematically evaluating and perhaps enhancing EV uptake by various cell types.
Collapse
Affiliation(s)
- Devin M Stranford
- 1 Department of Chemical and Biological Engineering, Northwestern University , Evanston, Illinois.,2 Center for Synthetic Biology, Northwestern University , Evanston, Illinois
| | - Michelle E Hung
- 3 Interdisciplinary Biological Sciences Program, Northwestern University , Evanston, Illinois
| | - Emma S Gargus
- 4 Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University , Chicago, Illinois.,5 Simpson Querrey Institute for BioNanotechnology, Northwestern University , Chicago, Illinois
| | - Ramille N Shah
- 5 Simpson Querrey Institute for BioNanotechnology, Northwestern University , Chicago, Illinois.,6 Department of Materials Science and Engineering, Northwestern University , Evanston, Illinois.,7 Department of Surgery, Feinberg School of Medicine, Northwestern University , Chicago, Illinois.,8 Department of Biomedical Engineering, Northwestern University , Evanston, Illinois
| | - Joshua N Leonard
- 1 Department of Chemical and Biological Engineering, Northwestern University , Evanston, Illinois.,2 Center for Synthetic Biology, Northwestern University , Evanston, Illinois.,9 Chemistry of Life Processes Institute, Northwestern University , Evanston, Illinois.,10 Member, Robert H. Lurie Comprehensive Cancer Center, Northwestern University , Evanston, Illinois
| |
Collapse
|
27
|
Stranford DM, Leonard JN. Delivery of Biomolecules via Extracellular Vesicles. ADVANCES IN GENETICS 2017; 98:155-175. [DOI: 10.1016/bs.adgen.2017.08.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
|
28
|
Exosomes and Exosomal miRNA in Respiratory Diseases. Mediators Inflamm 2016; 2016:5628404. [PMID: 27738390 PMCID: PMC5055958 DOI: 10.1155/2016/5628404] [Citation(s) in RCA: 97] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Accepted: 08/28/2016] [Indexed: 12/21/2022] Open
Abstract
Exosomes are nanosized vesicles released from every cell in the body including those in the respiratory tract and lungs. They are found in most body fluids and contain a number of different biomolecules including proteins, lipids, and both mRNA and noncoding RNAs. Since they can release their contents, particularly miRNAs, to both neighboring and distal cells, they are considered important in cell-cell communication. Recent evidence has shown their possible importance in the pathogenesis of several pulmonary diseases. The differential expression of exosomes and of exosomal miRNAs in disease has driven their promise as biomarkers of disease enabling noninvasive clinical diagnosis in addition to their use as therapeutic tools. In this review, we summarize recent advances in this area as applicable to pulmonary diseases.
Collapse
|