Hostname: page-component-7f64f4797f-42qgm Total loading time: 0 Render date: 2025-11-10T23:28:46.532Z Has data issue: false hasContentIssue false

Extracellular Vesicles: Multimodal Tools for Diagnosis, Prognosis, and Therapy in Respiratory Diseases

Published online by Cambridge University Press:  21 October 2025

Miquéias Lopes-Pacheco*
Affiliation:
Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA Center for Cystic Fibrosis and Airways Disease Research, Emory University and Children’s Healthcare of Atlanta, Atlanta, GA, USA
*
Corresponding author: Miquéias Lopes-Pacheco; Email: mlopesp@emory.edu
Rights & Permissions [Opens in a new window]

Abstract

Background

Respiratory diseases are increasing global health burden with persistently high morbidity and mortality. Extracellular vesicles (EVs), which are virtually released by all cell types and carry a variety of molecules like miRNAs, have emerged as crucial mediators of intercellular communication. They play a key role in maintaining lung homeostasis and are involved in the pathogenesis of various respiratory conditions. Furthermore, mesenchymal stromal cell-derived EVs (MSC-EVs) have shown significant therapeutic potential due to their anti-inflammatory, antimicrobial, and reparative properties.

Methods

This narrative review critically assesses the current body of literature on the roles of EVs in respiratory diseases. We examine evidence from pre-clinical and clinical studies that investigate EVs as biomarkers and therapeutics for conditions including asthma, bronchopulmonary dysplasia (BPD), chronic obstructive pulmonary disease (COPD), cystic fibrosis (CF), idiopathic pulmonary fibrosis (IPF), lung cancer, and pulmonary arterial hypertension (PAH).

Results

EVs reflect the physiological or pathological state of their parental cells, making them promising multimodal biomarkers for the early diagnosis and monitoring of disease progression. Additionally, MSC-EVs function as effective, cell-free therapeutic agents. In a variety of disease models, they demonstrate efficacy by modulating immune responses, enhancing alveolar fluid clearance, and restoring epithelial and endothelial barrier integrity, leading to improved survival and outcomes.

Conclusions

EVs hold a dual and transformative potential in respiratory medicine. They may serve as valuable diagnostic and prognostic tools, and their application as cell-free therapeutics represents a novel and promising strategy for treating a wide spectrum of debilitating respiratory diseases.

Information

Type
Review
Creative Commons
Creative Common License - CCCreative Common License - BY
This is an Open Access article, distributed under the terms of the Creative Commons Attribution licence (http://creativecommons.org/licenses/by/4.0), which permits unrestricted re-use, distribution and reproduction, provided the original article is properly cited.
Copyright
© The Author(s), 2025. Published by Cambridge University Press

Introduction

Respiratory illnesses represent an increasing societal burden worldwide and encompass various pathological conditions, including obstructive and restrictive ventilatory disorders, pulmonary infections, and lung cancer. Among these, chronic respiratory diseases have a prevalence of more than 450 million cases and claim over four million lives annually, being therefore among the global leading causes of death (Refs. Reference Momtazmanesh, Moghaddam, Ghamari, Rad, Rezaei, Shobeiri, Aali, Abbasi-Kangevari, Abbasi-Kangevari, Abdelmasseh, Abdoun, Abdulah, Md Abdullah, Abedi, Abolhassani, Abrehdari-Tafreshi, Achappa, Adane Adane, Adane, Addo, Adnan, Sakilah Adnani, Ahmad, Ahmadi, Ahmadi, Ahmed, Ahmed, Rashid, al Hamad, Alahdab, Alemayehu, Alif, Aljunid, Almustanyir, Altirkawi, Alvis-Guzman, Dehkordi, Amir-Behghadami, Ancuceanu, Andrei, Andrei, Antony, Anyasodor, Arabloo, Arulappan, Ashraf, Athari, Attia, Ayele, Azadnajafabad, Babu, Bagherieh, Baltatu, Banach, Bardhan, Barone-Adesi, Barrow, Basu, Bayileyegn, Bensenor, Bhardwaj, Bhardwaj, Bhat, Bhattacharyya, Bouaoud, Braithwaite, Brauer, Butt, Butt, Calina, Cámera, Chanie, Charalampous, Chattu, Chimed-Ochir, Chu, Cohen, Cruz-Martins, Dadras, Darwesh, das, Debela, Delgado-Ortiz, Dereje, Dianatinasab, Diao, Diaz, Digesa, Dirirsa, Doku, Dongarwar, Douiri, Dsouza, Eini, Ekholuenetale, Ekundayo, Mustafa Elagali, Elhadi, Enyew, Erkhembayar, Etaee, Fagbamigbe, Faro, Fatehizadeh, Fekadu, Filip, Fischer, Foroutan, Franklin, Gaal, Gaihre, Gaipov, Gebrehiwot, Gerema, Getachew, Getachew, Ghafourifard, Ghanbari, Ghashghaee, Gholami, Gil, Golechha, Goleij, Golinelli, Guadie, Gupta, Gupta, Gupta, Gupta, Hadei, Halwani, Hanif, Hargono, Harorani, Hartono, Hasani, Hashi, Hay, Heidari, Hellemons, Herteliu, Holla, Horita, Hoseini, Hosseinzadeh, Huang, Hussain, Hwang, Iavicoli, Ibitoye, Ibrahim, Ilesanmi, Ilic, Ilic, Immurana, Ismail, Merin J, Jakovljevic, Jamshidi, Janodia, Javaheri, Jayapal, Jayaram, Jha, Johnson, Joo, Joseph, Jozwiak, K, Kaambwa, Kabir, Kalankesh, Kalhor, Kandel, Karanth, Karaye, Kassa, Kassie, Keikavoosi-Arani, Keykhaei, Khajuria, Khan, Khan, Khan, Khreis, Kim, Kisa, Kisa, Knibbs, Kolkhir, Komaki, Kompani, Koohestani, Koolivand, Korzh, Koyanagi, Krishan, Krohn, Kumar, Kumar, Kurmi, Kuttikkattu, la Vecchia, Lám, Lan, Lasrado, Latief, Lauriola, Lee, Lee, Legesse, Lenzi, Li, Lin, Liu, Liu, Lo, Lorenzovici, Lu, Mahalingam, Mahmoudi, Mahotra, Malekpour, Malik, Mallhi, Malta, Mansouri, Mathews, Maulud, Mechili, Nasab, Menezes, Mengistu, Mentis, Meshkat, Mestrovic, Micheletti Gomide Nogueira de Sá, Mirrakhimov, Misganaw, Mithra, Moghadasi, Mohammadi, Mohammadi, Mohammadshahi, Mohammed, Mohan, Moka, Monasta, Moni, Moniruzzaman, Montazeri, Moradi, Mostafavi, Mpundu-Kaambwa, Murillo-Zamora, Murray, Nair, Nangia, Swamy, Narayana, Natto, Nayak, Negash, Nena, Kandel, Niazi, Nogueira de Sá, Nowroozi, Nzoputam, Nzoputam, Oancea, Obaidur, Odukoya, Okati-Aliabad, Okekunle, Okonji, Olagunju, Bali, Ostojic, A, Padron-Monedero, Padubidri, Pahlevan Fallahy, Palicz, Pana, Park, Patel, Paudel, Paudel, Pedersini, Pereira, Pereira, Petcu, Pirestani, Postma, Prashant, Rabiee, Radfar, Rafiei, Rahim, Ur Rahman, Rahman, Rahman, Rahmani, Rahmani, Rahmanian, Rajput, Rana, Rao, Rao, Rashedi, Rashidi, Ratan, Rawaf, Rawaf, Rawal, Rawassizadeh, Razeghinia, Mohamed Redwan, Rezaei, Rezaei, Rezaei, Rezaeian, Rodrigues, Buendia Rodriguez, Roever, Rojas-Rueda, Rudd, Saad, Sabour, Saddik, Sadeghi, Sadeghi, Saeed, Sahebazzamani, Sahebkar, Sahoo, Sajid, Sakhamuri, Salehi, Samy, Santric-Milicevic, Sao Jose, Sathian, Satpathy, Saya, Senthilkumaran, Seylani, Shahabi, Shaikh, Shanawaz, Shannawaz, Sheikhi, Shekhar, Sibhat, Simpson, Singh, Singh, Singh, Skryabin, Skryabina, Soltani-Zangbar, Song, Soyiri, Steiropoulos, Stockfelt, Sun, Takahashi, Talaat, Tan, Tat, Tat, Taye, Thangaraju, Thapar, Thienemann, Tiyuri, Ngoc Tran, Tripathy, Car, Tusa, Ullah, Ullah, Vacante, Valdez, Valizadeh, van Boven, Vasankari, Vaziri, Violante, Vo, Wang, Wei, Westerman, Wickramasinghe, Xu, Xu, Yadav, Yismaw, Yon, Yonemoto, Yu, Yu, Yunusa, Zahir, Zangiabadian, Zareshahrabadi, Zarrintan, Zastrozhin, Zegeye, Zhang, Naghavi, Larijani and Farzadfar1, 2). These diseases are characterized by chronic airway/alveolar inflammation and progressive deterioration of lung function, with common clinical symptoms including chronic cough, chest tightness, shortness of breath, and mucus production. Furthermore, pulmonary exacerbations can frequently occur and contribute to accelerating disease progression and increasing the mortality rate (Refs. Reference Bhatt3Reference Ritchie and Wedzicha5). Despite considerable advances in understanding the underlying mechanisms in the pathogenesis and progression of severe respiratory diseases, there is an urgent need for more precise biomarkers and effective therapies to reduce morbidity and mortality associated with these devastating conditions.

Extracellular vesicles (EVs) are a multifaceted group of non-replicating lipid bilayer membrane-delimited particles (Figure 1), which are ubiquitously released or shed by all cell types (Refs. Reference Lötvall, Hill, Hochberg, Buzás, di Vizio, Gardiner, Gho, Kurochkin, Mathivanan, Quesenberry, Sahoo, Tahara, Wauben, Witwer and Théry6Reference Welsh, Goberdhan and O’Driscoll8). These particles can range from nano- to micrometres and once released into the extracellular milieu, they are extensively distributed and transported through biofluids (bloodstream, saliva, breast milk, urine, bronchoalveolar lavage fluid (BALF), and others), being thus able to travel long distances within the human body (Ref. Reference Alberro, Iparraguirre, Fernandes and Otaegui9). Accordingly, EVs play a key role in intercellular communication and participate in an intricate interplay among cells and tissues via autocrine, paracrine, or endocrine signalling mechanisms. Indeed, numerous studies have demonstrated that the internal content of EVs (nucleic acids, proteins, glycan structures, metabolites, and lipids) exhibits high stability due to protection from external enzymatic degradation (Refs. Reference Turchinovich, Weiz and Burwinkel10,Reference dos Santos, Lopes-Pacheco, English, Rolandsson Enes, Krasnodembskaya and Rocco11) and reflects the state of their cells of origin (Refs. Reference Zhang, Liu, Liu and Tang12Reference Pinheiro, Torrecilhas and de Freitas Souza14). After binding to specific plasma membrane receptors (e.g., integrins, tetraspanin, proteoglycans, and immunoglobulin) of target cells, EVs activate ligand-receptor downstream signals or release their content into the cytosol (or deliver it to specific organelles) to promote alterations in gene and protein expression, signal transduction, cell proliferation, differentiation, and survival (Refs. Reference Holtzman and Lee15, Reference Mulcahy, Pink and Carter16).

Figure 1. Extracellular vesicle (EV) composition. EVs are small, non-replicating membrane-bound particles released by cells into the extracellular environment. These particles are delimited by a lipid bilayer with membrane proteins (e.g., tetraspanins, receptors, immunoglobulins, and adhesion molecules), and their internal content is composed mainly of cytosolic proteins, lipids, and nucleic acids (e.g., DNA and RNA), which reflects the state of their parental cells. Some EVs can also contain metabolites and cellular organelles (e.g., mitochondria).

The first report potentially documenting the existence of EVs came in 1946 when Chargaff and West indicated the presence of ‘lipoproteins of very high particle weight’ in serum (Ref. Reference Chargaff and West17). A subsequent study from Wolf in 1967 reported the acceleration of coagulation by lipid-rich particles originating from the granules of platelets (Ref. Reference Wolf18). In the 1980s, studies from Harding and Pan reported the fusion of multivesicular endosomes with the plasma membrane of rat and sheep reticulocytes and the subsequent release of their cargo into the extracellular space (Refs. Reference Harding, Heuser and Stahl19, Reference Pan, Teng, Wu, Adam and Johnstone20). Since then, a growing body of research has been performed to provide further understanding of EV biology from different cell types and their potential implications for diagnosis, prognosis, and therapeutics.

Due to large heterogeneity in the literature and aiming to guide EV studies, the International Society for Extracellular Vesicles (ISEV) has established minimal criteria for their definition and characterization (Refs. Reference Lötvall, Hill, Hochberg, Buzás, di Vizio, Gardiner, Gho, Kurochkin, Mathivanan, Quesenberry, Sahoo, Tahara, Wauben, Witwer and Théry6Reference Welsh, Goberdhan and O’Driscoll8). Accordingly, EVs can be categorized based on their known cellular origin/biosynthesis, secretion pathway, and size (Table 1). Although significant progress has been attained in developing several methods for EV isolation and characterization (Tables 2 and 3), these are still unable to obtain pure EV populations since different EV types can overlap in size, density, and molecular composition. Furthermore, the identification of the cellular origin of EVs is particularly relevant in the clinical context, and certain markers are cell/tissue-specific; however, EV purification should not rely solely on EV-surface protein, and a limited consensus persists on appropriate markers to define biogenesis and origin for all EV subtypes.

Abbreviations: Alix: ALIG-2-interacting protein X; ARF: ADP-ribosylation factor; Cav: caveolin; CK: cytokeratin; ESCRT: endosomal sorting complexes for transport; Hsp: heat shock protein; Tsg: tumor susceptibility gene; Tspan: tetraspanin.

a Recommended operational term based on the diameter of the separated particles.

b There is no strict consensus on size cut-offs since it may be affected by the separation method.

c Related to formation or specialized cell processes and recommended to be used only when subcellular origin can be demonstrated.

Table 2. Methods commonly used for EV separation and concentration

Table 3. Methods commonly used for EV characterization

In the lungs, EVs have been demonstrated to be essential contributors to maintaining cellular homeostasis, regulating immune responses, and facilitating tissue healing/remodelling in both health and disease conditions (Refs. Reference Abreu, Lopes-Pacheco, Weiss and Rocco13, Reference Kadota, Fujita, Araya, Ochiya and Kuwano21). EVs promote these actions by operating as intercellular messengers and transferring their cargo to different lung resident and immune cells, which influence their behaviour and responses to environmental stimuli. In this review, we discuss the utility of EVs as biomarkers for early diagnosis and monitoring disease progression and their emerging role in therapeutic strategies for various respiratory diseases, including asthma, bronchopulmonary dysplasia (BPD), chronic obstructive pulmonary disease (COPD), cystic fibrosis (CF), idiopathic pulmonary fibrosis (IPF), lung cancer and pulmonary arterial hypertension (PAH).

Influence of EVs in lung homeostasis and intercellular communication

The respiratory tract is composed of a variety of cell types with distinct functions that maintain their structural and functional integrity based on complex intercellular communication (Figure 2). Defence against invaders also requires well-coordinated communication between structural and immune cells to eliminate pathogens and prevent excessive inflammation that may lead to tissue damage and remodelling (Ref. Reference Schneider, Rowe, Garcia-de-Alba, Kim, Sharpe and Haigis22). In this context, the airway epithelium acts as a physical barrier to prevent the entrance of allergens, pollutants, and pathogens and efficiently activates innate and adaptive immune responses for clearance and restoration of lung homeostasis (Ref. Reference Bartel, Deshane, Wilkinson and Gabrielsson23). EVs participate in such processes by supporting epithelial and endothelial cell function, enhancing barrier integrity, controlling oxidative stress, recruiting immune cells, and modulating their responses.

Figure 2. Extracellular vesicles (EVs) in lung physiological or pathological processes. The respiratory system is composed of diverse cell types, each with specialized roles, relying on intricate communication to preserve the structural and functional integrity of airways and alveoli. Under normal physiological (or pathological) conditions, EVs participate in lung homeostasis by promoting intercellular communication, thus regulating resident and immune cell responses, supporting barrier integrity, and facilitating tissue repair (or remodelling). EVs have been considered suitable biomarkers for diagnosis due to differences in their size, quantity, cellular origin, and content (both on the surface and within their lumen) in diverse diseases, which can facilitate the distinction between pathological conditions and severity with high accuracy and enable early and personalized therapeutic interventions. Mesenchymal stromal cell (MSC)-derived EVs have been demonstrated to be attractive therapeutic agents for respiratory diseases due to their ability to reduce inflammation and fibrosis, improve fluid clearance, and repair epithelial and endothelial barrier permeability.

As bronchial epithelial cells are frequently exposed to pollutants and pathogens, their EVs contain factors that are essential for an innate mucosal defence, such as surface-associated mucin (MUC)-1, MUC-4, and MUC-16 (Refs. Reference Kesimer, Scull, Brighton, DeMaria, Burns, O’Neal, Pickles and Sheehan24, Reference Gupta, Radicioni, Abdelwahab, Dang, Carpenter, Chua, Mieczkowski, Sheridan, Randell and Kesimer25). Bronchial epithelial cell-derived EVs are also a primary source of reactive pro-inflammatory mediators (Ref. Reference Wahlund, Eklund, Grunewald and Gabrielsson26) and demonstrated to activate receptor for advanced glycation end-products and mitogen-activated protein kinase pathways in neutrophils (Ref. Reference Useckaite, Ward, Trappe, Reilly, Lennon, Davage, Matallanas, Cassidy, Dillon, Brennan, Doyle, Carter, Donnelly, Linnane, McKone, McNally and Coppinger27). Meanwhile, pathogens defend themselves by releasing EVs with a cargo that prevents host immune responses and creates an optimal environment for their proliferation (Refs. Reference Pinheiro, Torrecilhas and de Freitas Souza14, Reference Liu, Defourny, Smid and Abee28).

Alveolar epithelial cell-derived EVs exhibit abundant expression of mediators involved in the modulation of inflammatory signals (e.g., miR-223 and suppressor of cytokine signalling) (Refs. Reference Ismail, Wang, Dakhlallah, Moldovan, Agarwal, Batte, Shah, Wisler, Eubank, Tridandapani, Paulaitis, Piper and Marsh29, Reference Bourdonnay, Zasłona, Penke, Speth, Schneider, Przybranowski, Swanson, Mancuso, Freeman, Curtis and Peters-Golden30). Endothelial cell-derived EVs assist in protecting lung tissue from inflammatory injury by delivering miR-10a and preventing monocyte activation (Ref. Reference Njock, Cheng, Dang, Nazari-Jahantigh, Lau, Boudreau, Roufaiel, Cybulsky, Schober and Fish31). Upon pulmonary artery damage, the abundance of EVs released by endothelial cells is significantly enhanced (Ref. Reference Zhao, Luo, Li, Li, He, Qi, Liu and Yu32). Endothelial cell-derived EVs can also participate in various biological processes, including angiogenesis, coagulation, and inflammation (Ref. Reference Kadota, Fujita, Yoshioka, Araya, Kuwano and Ochiya33).

Immune cell-secreted EVs play an important role in lung defence and homeostasis. Mast cell-derived EVs contribute to the production of antibody IgE (Ref. Reference Dhar, Mukherjee, Mukerjee, Mukherjee, Devi, Ashraf, Alserihi, Tayeb, Hashem, Alexiou and Thorate34) and can stimulate epithelial-mesenchymal transition (EMT) of airway cells due to transforming growth factor (TGF)-β expression (Ref. Reference Yin, Shelke, Lässer, Brismar and Lötvall35). Immune cell-derived EVs can stimulate or inhibit fibroblast differentiation into myofibroblasts, contributing to tissue healing or remodelling (Ref. Reference Lacy, Woeller, Thatcher, Pollock, Small, Sime and Phipps36). Dendritic cell-derived EVs can affect T cell maturation and regulation (Ref. Reference Dhar, Mukherjee, Mukerjee, Mukherjee, Devi, Ashraf, Alserihi, Tayeb, Hashem, Alexiou and Thorate34), while macrophage-derived EVs can promote the differentiation of monocytes by transferring miR-223 (Ref. Reference Ismail, Wang, Dakhlallah, Moldovan, Agarwal, Batte, Shah, Wisler, Eubank, Tridandapani, Paulaitis, Piper and Marsh29). Altogether, these examples support the relevance of different cell type-derived EVs in protecting the respiratory tract against invaders and maintaining or restoring tissue homeostasis.

EVs as biomarkers for respiratory diseases

Given the ability of EVs to carry molecular signatures that reflect the physiological or pathological state of their parental cells (Figure 2), they have been investigated as potential multimodal tools for disease diagnosis and prognosis as well as therapy monitoring (Refs. Reference Zhang, Liu, Liu and Tang12Reference Pinheiro, Torrecilhas and de Freitas Souza14). Since EVs are abundantly present in all body fluids and have a relatively stable cargo, they represent a valuable source for biomarker assessment (Refs. Reference Zhou, Xu, Zheng, Chen, Wang, Song, Shao and Zheng37, Reference Grueso-Navarro, Navarro, Laserna-Mendieta, Lucendo and Arias-González38).

EVs have been frequently obtained from BALF and blood for respiratory research studies, with the latest representing a minimally invasive source that could be utilized for routine testing (Refs. Reference Carnino, Lee and Jin39, Reference Paes Leme, Yokoo, Normando, Ormonde, Domingues, Cruz, Silva, Souza, dos Santos, Castro-Faria-Neto, Martins, Lopes-Pacheco and Rocco40). EV profiling may also assist in identifying disease and monitoring progression, or even ensuring the differentiation of similar respiratory conditions, thus facilitating early and personalized interventions (Refs. Reference Abreu, Lopes-Pacheco, Weiss and Rocco13, Reference Bacalhau, Camargo and Lopes-Pacheco41). For instance, alterations in the quantity and content of EVs have been documented in patients with asthma (Refs. Reference Duarte, Taveira-Gomes, Sokhatska, Palmares, Costa, Negrão, Guimarães, Delgado, Soares and Moreira42, Reference Rollet-Cohen, Bourderioux, Lipecka, Chhuon, Jung, Mesbahi, Nguyen-Khoa, Guérin-Pfyffer, Schmitt, Edelman, Sermet-Gaudelus and Guerrera43), COPD (Ref. Reference Takahashi, Kobayashi, Fujino, Suzuki, Ota, He, Yamada, Suzuki, Yanai, Kurosawa, Yamaya and Kubo44), CF (Refs. Reference Useckaite, Ward, Trappe, Reilly, Lennon, Davage, Matallanas, Cassidy, Dillon, Brennan, Doyle, Carter, Donnelly, Linnane, McKone, McNally and Coppinger27, Reference Rollet-Cohen, Bourderioux, Lipecka, Chhuon, Jung, Mesbahi, Nguyen-Khoa, Guérin-Pfyffer, Schmitt, Edelman, Sermet-Gaudelus and Guerrera43), and primary ciliary dyskinesia (Ref. Reference Rollet-Cohen, Bourderioux, Lipecka, Chhuon, Jung, Mesbahi, Nguyen-Khoa, Guérin-Pfyffer, Schmitt, Edelman, Sermet-Gaudelus and Guerrera43). Analysis of EV-transported cargo was able to differentiate children with allergic airway disease from healthy controls (Ref. Reference Samra, Lim, Han, Jee, Kim and Kim45) as well as disease severity in COVID-19 (Refs. Reference Paes Leme, Yokoo, Normando, Ormonde, Domingues, Cruz, Silva, Souza, dos Santos, Castro-Faria-Neto, Martins, Lopes-Pacheco and Rocco40, Reference Ciccosanti, Antonioli, Sacchi, Notari, Farina, Beccacece, Fusto, Vergori, D’Offizi, Taglietti, Antinori, Nicastri, Marchioni, Palmieri, Ippolito, Piacentini, Agrati and Fimia46).

Asthma

Asthma is a complex respiratory disease characterized by chronic inflammatory responses that contribute to airflow obstruction and airway hyperresponsiveness, and remodelling (Ref. Reference Holgate, Wenzel, Postma, Weiss, Renz and Sly47). Over 260 million people worldwide are affected by asthma with many individuals remaining undiagnosed and untreated (Ref. Reference Momtazmanesh, Moghaddam, Ghamari, Rad, Rezaei, Shobeiri, Aali, Abbasi-Kangevari, Abbasi-Kangevari, Abdelmasseh, Abdoun, Abdulah, Md Abdullah, Abedi, Abolhassani, Abrehdari-Tafreshi, Achappa, Adane Adane, Adane, Addo, Adnan, Sakilah Adnani, Ahmad, Ahmadi, Ahmadi, Ahmed, Ahmed, Rashid, al Hamad, Alahdab, Alemayehu, Alif, Aljunid, Almustanyir, Altirkawi, Alvis-Guzman, Dehkordi, Amir-Behghadami, Ancuceanu, Andrei, Andrei, Antony, Anyasodor, Arabloo, Arulappan, Ashraf, Athari, Attia, Ayele, Azadnajafabad, Babu, Bagherieh, Baltatu, Banach, Bardhan, Barone-Adesi, Barrow, Basu, Bayileyegn, Bensenor, Bhardwaj, Bhardwaj, Bhat, Bhattacharyya, Bouaoud, Braithwaite, Brauer, Butt, Butt, Calina, Cámera, Chanie, Charalampous, Chattu, Chimed-Ochir, Chu, Cohen, Cruz-Martins, Dadras, Darwesh, das, Debela, Delgado-Ortiz, Dereje, Dianatinasab, Diao, Diaz, Digesa, Dirirsa, Doku, Dongarwar, Douiri, Dsouza, Eini, Ekholuenetale, Ekundayo, Mustafa Elagali, Elhadi, Enyew, Erkhembayar, Etaee, Fagbamigbe, Faro, Fatehizadeh, Fekadu, Filip, Fischer, Foroutan, Franklin, Gaal, Gaihre, Gaipov, Gebrehiwot, Gerema, Getachew, Getachew, Ghafourifard, Ghanbari, Ghashghaee, Gholami, Gil, Golechha, Goleij, Golinelli, Guadie, Gupta, Gupta, Gupta, Gupta, Hadei, Halwani, Hanif, Hargono, Harorani, Hartono, Hasani, Hashi, Hay, Heidari, Hellemons, Herteliu, Holla, Horita, Hoseini, Hosseinzadeh, Huang, Hussain, Hwang, Iavicoli, Ibitoye, Ibrahim, Ilesanmi, Ilic, Ilic, Immurana, Ismail, Merin J, Jakovljevic, Jamshidi, Janodia, Javaheri, Jayapal, Jayaram, Jha, Johnson, Joo, Joseph, Jozwiak, K, Kaambwa, Kabir, Kalankesh, Kalhor, Kandel, Karanth, Karaye, Kassa, Kassie, Keikavoosi-Arani, Keykhaei, Khajuria, Khan, Khan, Khan, Khreis, Kim, Kisa, Kisa, Knibbs, Kolkhir, Komaki, Kompani, Koohestani, Koolivand, Korzh, Koyanagi, Krishan, Krohn, Kumar, Kumar, Kurmi, Kuttikkattu, la Vecchia, Lám, Lan, Lasrado, Latief, Lauriola, Lee, Lee, Legesse, Lenzi, Li, Lin, Liu, Liu, Lo, Lorenzovici, Lu, Mahalingam, Mahmoudi, Mahotra, Malekpour, Malik, Mallhi, Malta, Mansouri, Mathews, Maulud, Mechili, Nasab, Menezes, Mengistu, Mentis, Meshkat, Mestrovic, Micheletti Gomide Nogueira de Sá, Mirrakhimov, Misganaw, Mithra, Moghadasi, Mohammadi, Mohammadi, Mohammadshahi, Mohammed, Mohan, Moka, Monasta, Moni, Moniruzzaman, Montazeri, Moradi, Mostafavi, Mpundu-Kaambwa, Murillo-Zamora, Murray, Nair, Nangia, Swamy, Narayana, Natto, Nayak, Negash, Nena, Kandel, Niazi, Nogueira de Sá, Nowroozi, Nzoputam, Nzoputam, Oancea, Obaidur, Odukoya, Okati-Aliabad, Okekunle, Okonji, Olagunju, Bali, Ostojic, A, Padron-Monedero, Padubidri, Pahlevan Fallahy, Palicz, Pana, Park, Patel, Paudel, Paudel, Pedersini, Pereira, Pereira, Petcu, Pirestani, Postma, Prashant, Rabiee, Radfar, Rafiei, Rahim, Ur Rahman, Rahman, Rahman, Rahmani, Rahmani, Rahmanian, Rajput, Rana, Rao, Rao, Rashedi, Rashidi, Ratan, Rawaf, Rawaf, Rawal, Rawassizadeh, Razeghinia, Mohamed Redwan, Rezaei, Rezaei, Rezaei, Rezaeian, Rodrigues, Buendia Rodriguez, Roever, Rojas-Rueda, Rudd, Saad, Sabour, Saddik, Sadeghi, Sadeghi, Saeed, Sahebazzamani, Sahebkar, Sahoo, Sajid, Sakhamuri, Salehi, Samy, Santric-Milicevic, Sao Jose, Sathian, Satpathy, Saya, Senthilkumaran, Seylani, Shahabi, Shaikh, Shanawaz, Shannawaz, Sheikhi, Shekhar, Sibhat, Simpson, Singh, Singh, Singh, Skryabin, Skryabina, Soltani-Zangbar, Song, Soyiri, Steiropoulos, Stockfelt, Sun, Takahashi, Talaat, Tan, Tat, Tat, Taye, Thangaraju, Thapar, Thienemann, Tiyuri, Ngoc Tran, Tripathy, Car, Tusa, Ullah, Ullah, Vacante, Valdez, Valizadeh, van Boven, Vasankari, Vaziri, Violante, Vo, Wang, Wei, Westerman, Wickramasinghe, Xu, Xu, Yadav, Yismaw, Yon, Yonemoto, Yu, Yu, Yunusa, Zahir, Zangiabadian, Zareshahrabadi, Zarrintan, Zastrozhin, Zegeye, Zhang, Naghavi, Larijani and Farzadfar1). Asthma has the highest prevalence among chronic respiratory diseases with variable incidence across different regions depending on several factors, including socioeconomic status and genetic predisposition (Ref. Reference Song, Adeloye, Salim, dos Santos, Campbell, Sheikh and Rudan48). EVs can be released by epithelial cells on both apical and basolateral sides (Refs. Reference Bartel, La Grutta and Cilluffo49, Reference Mwase, Phung, O’Sullivan, Mitchel, de Marzio, Kılıç, Weiss, Fredberg and Park50), and these contribute to the pathogenesis and progression of asthma by influencing pro-inflammatory responses (Refs. Reference Abreu, Lopes-Pacheco, Weiss and Rocco13, Reference Kanannejad, Arab, Soleimanian, Mazare and Kheshtchin51). For instance, house dust mite (HDM) extract significantly altered the content of BALF-derived EVs in experimental asthma (Ref. Reference Gon, Maruoka, Inoue, Kuroda, Yamagishi, Kozu, Shikano, Soda, Lötvall and Hashimoto52), corroborating findings of EVs from asthmatic patients (Ref. Reference Francisco-Garcia, Garrido-Martín, Rupani, Lau, Martinez-Nunez, Howarth and Sanchez-Elsner53). BALF- and nasal lavage fluid-derived EVs from asthmatic patients exhibited increased concentrations of interleukin (IL)-4, leukotriene C4, and other chemoattractant factors (Refs. Reference Torregrosa Paredes, Esser, Admyre, Nord, Rahman, Lukic, Rådmark, Grönneberg, Grunewald, Eklund, Scheynius and Gabrielsson54, Reference Lässer, O’Neil, Shelke, Sihlbom, Hansson, Gho, Lundbäck and Lötvall55). Likewise, an increase in EV release was observed in human bronchial epithelial cells treated with Th2 (IL-4 and IL-13) and Th17 cytokines (IL-17A and tumour necrosis factor (TNF)-α) (Ref. Reference Ax, Jevnikar, Cvjetkovic, Malmhäll, Olsson, Rådinger and Lässer56). Eosinophil-derived EVs from asthmatic patients were able to delay wound repair, promote in vitro apoptosis of small airway epithelial cells, and stimulate proliferation of smooth muscle cells (Refs. Reference Cañas, Sastre, Mazzeo, Fernández-Nieto, Rodrigo-Muñoz, González-Guerra, Izquierdo, Barranco, Quirce, Sastre and del Pozo57, Reference Cañas, Sastre, Rodrigo-Muñoz, Fernández-Nieto, Barranco, Quirce, Sastre and del Pozo58). Treatment with EV-production inhibitor GW4869 ameliorated asthma symptoms in an animal model by reducing the release of lung cell-derived EVs (Ref. Reference Gon, Maruoka, Inoue, Kuroda, Yamagishi, Kozu, Shikano, Soda, Lötvall and Hashimoto52). Such reduction was also accompanied by a reduction in the number of inflammatory cells, bronchial hyperresponsiveness, and IgE levels (Ref. Reference Kulshreshtha, Ahmad, Agrawal and Ghosh59). In BALF samples, positive correlations were established for EV counts versus eosinophilia and EV counts versus IgE titer (Ref. Reference Hough, Wilson, Trevor, Strenkowski, Maina, Kim, Spell, Wang, Chanda, Dager, Sharma, Curtiss, Antony, Dransfield, Chaplin, Steele, Barnes, Duncan, Prasain, Thannickal and Deshane60). Profiling studies have been performed to further understand EV involvement in asthma-mediated inflammatory responses. In BALF-derived EVs, proteomic and lipidomic analyses revealed increased levels of toll-like receptor (TLR) signalling-related proteins (Ref. Reference Rollet-Cohen, Bourderioux, Lipecka, Chhuon, Jung, Mesbahi, Nguyen-Khoa, Guérin-Pfyffer, Schmitt, Edelman, Sermet-Gaudelus and Guerrera43) and altered lipid mediator composition of EVs from asthmatic patients compared to healthy controls (Ref. Reference Hough, Wilson, Trevor, Strenkowski, Maina, Kim, Spell, Wang, Chanda, Dager, Sharma, Curtiss, Antony, Dransfield, Chaplin, Steele, Barnes, Duncan, Prasain, Thannickal and Deshane60).

Several studies have investigated EV-transported miRNA for potential biomarkers of asthma. Along these lines, EV miRNA content was significantly distinct in samples from asthmatic patients compared to healthy controls (Refs. Reference Duarte, Taveira-Gomes, Sokhatska, Palmares, Costa, Negrão, Guimarães, Delgado, Soares and Moreira42, Reference Levänen, Bhakta, Torregrosa Paredes, Barbeau, Hiltbrunner, Pollack, Sköld, Svartengren, Grunewald, Gabrielsson, Eklund, Larsson, Woodruff, Erle and Wheelock61). In mice, HDM exposure altered the expression of >100 miRNAs in EV content, including miR-346 and miR-574-5p (Ref. Reference Gon, Maruoka, Inoue, Kuroda, Yamagishi, Kozu, Shikano, Soda, Lötvall and Hashimoto52). Most BALF-derived EVs are released from epithelial cells in experimental asthma; however, a significant increase in EV number from immune cells was evidenced following allergen exposure. Increased levels of miR-142a and miR-223 were found in immune cell-derived EVs from this animal model (Ref. Reference Pua, Happ, Gray, Mar, Chiou, Hesse and Ansel62). In plasma-derived EVs, miR-21 and miR-223 levels were increased in patients with moderate asthma compared to healthy controls (Refs. Reference Rostami Hir, Alizadeh, Mazinani, Mahlooji Rad, Fazlollahi, Kazemnejad, Hosseini and Moin63, Reference Soccio, Moriondo, Lacedonia, Tondo, Pescatore, Quarato, Carone, Foschino Barbaro and Scioscia64). A promising miRNA to identify mildly asymptomatic asthma is let-7, which was demonstrated to be downregulated in BALF-derived EVs from patients with asthma (Ref. Reference Levänen, Bhakta, Torregrosa Paredes, Barbeau, Hiltbrunner, Pollack, Sköld, Svartengren, Grunewald, Gabrielsson, Eklund, Larsson, Woodruff, Erle and Wheelock61). On the contrary, PM2.5 led to asthma exacerbation by increasing levels of EV-packaged let-7i-5p in human bronchial epithelial cells (Ref. Reference Zheng, Du and Tian65). PM2.5 also increased levels of EV-packaged miR-129-2-30 that led to increased secretion of inflammatory mediators (IL-6, IL-8, and TNF-α) by epithelial cells (Ref. Reference Zheng, Du and Tian65). Levels of miR-126 were elevated in serum-derived EVs from patients with asthma and lung tissue of asthmatic mice (Ref. Reference Zhao, Li, Geng, Zhao, Ma, Yang, Deng, Luo and Pan66). Blood eosinophil counts positively correlated with miR-122-5a expression in plasma- and sputum-derived EVs with the potential to differentiate endotypes of asthma (Ref. Reference Bahmer, Krauss-Etschmann, Buschmann, Behrends, Watz, Kirsten, Pedersen, Waschki, Fuchs, Pfaffl, von Mutius, Rabe, Hansen, Kopp, König and Bartel67). Compared to healthy controls, EVs from asthmatic epithelial cells demonstrated increased levels of miR-9, which has been associated with steroid-resistant neutrophilic asthma (Ref. Reference Li, Tay, Maltby, Xiang, Eyers, Hatchwell, Zhou, Toop, Morris, Nair, Mattes, Foster and Yang68).

Bronchopulmonary dysplasia

BPD is a chronic lung disorder characterized by inflammation and disruption in airways and lung parenchymal vasculature, primarily affecting preterm infants or neonates requiring oxygen therapy. Although progress has been made in improving the survival of preterm infants, BPD remains a major cause of morbidity among survivors (Ref. Reference Stoll, Hansen, Bell, Walsh, Carlo, Shankaran, Laptook, Sánchez, van Meurs, Wyckoff, das, Hale, Ball, Newman, Schibler, Poindexter, Kennedy, Cotten, Watterberg, D’Angio, DeMauro, Truog, Devaskar and Higgins69). EVs have been isolated from various biofluids to investigate their role in BPD; however, studies have demonstrated that tracheal aspirate fluid is a feasible source for EV isolation and may represent the lung development stage in neonates or preterm infants (Refs. Reference Lal, Olave, Travers, Rezonzew, Dolma, Simpson, Halloran, Aghai, das, Sharma, Xu, Genschmer, Russell, Szul, Yi, Blalock, Gaggar, Bhandari and Ambalavanan70, Reference Ransom, Bunn, Negretti, Jetter, Bressman, Sucre and Pua71). Indeed, gestational age was demonstrated to be directly and negatively correlated to EV concentration and size, respectively (Refs. Reference Ransom, Bunn, Negretti, Jetter, Bressman, Sucre and Pua71, Reference Barnes, Pantazi and Holder72). Likewise, experimental hyperoxia-induced bronchial epithelial cells release more EVs of smaller size compared to cells under normoxia (Ref. Reference Lal, Olave, Travers, Rezonzew, Dolma, Simpson, Halloran, Aghai, das, Sharma, Xu, Genschmer, Russell, Szul, Yi, Blalock, Gaggar, Bhandari and Ambalavanan70). Umbilical cord-derived EVs from neonates who developed BPD reduced cell proliferation and capillary tube formation in cultured endothelial cells (Ref. Reference Zhong, Yan, Chen, Jia, Li, Liang, Gu, Wei, Lian, Zheng and Cui73). Differences in EV-surface markers have also been documented in neonates with BPD compared with those without it (Refs. Reference Lal, Olave, Travers, Rezonzew, Dolma, Simpson, Halloran, Aghai, das, Sharma, Xu, Genschmer, Russell, Szul, Yi, Blalock, Gaggar, Bhandari and Ambalavanan70, Reference Ransom, Bunn, Negretti, Jetter, Bressman, Sucre and Pua71). Tracheal aspirate-derived EVs exhibited increased levels of CD14 and CD24 (usually expressed by immune and epithelial cells, respectively) when obtained from infants with BPD (Ref. Reference Ransom, Bunn, Negretti, Jetter, Bressman, Sucre and Pua71). In experimental hyperoxia-induced BPD, rats demonstrated an increase in EV-packaged surfactant protein C in plasma. When these EVs were administered in control animals, lung and brain damage occurred, as well as morphological alterations consistent with BPD (Ref. Reference Ali, Zambrano, Duncan, Chen, Luo, Yuan, Chen, Benny, Schmidt, Young, Kerr, de Rivero Vaccari, Keane, Dietrich and Wu74).

Analysis of EV miRNAs for BPD has gained traction in recent years. Over 400 EV-derived miRNAs were differentially expressed in the umbilical cord of neonates with BPD compared to those without it (Ref. Reference Zhong, Yan, Chen, Jia, Li, Liang, Gu, Wei, Lian, Zheng and Cui73). Among these, the most significant reduction was observed for miRNA-103a-3p and miRNA-185-5p, while miRNA-200a-3p had increased expression (Ref. Reference Zhong, Yan, Chen, Jia, Li, Liang, Gu, Wei, Lian, Zheng and Cui73). An increase in endothelial cell proliferation and migration, and tube formation was observed by miR-103a-3p and miR-185-5p overexpression. On the contrary, miR-200a-3p overexpression prevented cellular angiogenic responses (Ref. Reference Zhong, Yan, Chen, Jia, Li, Liang, Gu, Wei, Lian, Zheng and Cui73). In another study, miRNA profiling revealed 40 miRNAs differentially expressed in EVs from infants with severe BPD compared to age-matched neonates without BPD (Ref. Reference Lal, Olave, Travers, Rezonzew, Dolma, Simpson, Halloran, Aghai, das, Sharma, Xu, Genschmer, Russell, Szul, Yi, Blalock, Gaggar, Bhandari and Ambalavanan70). The most sensitive alteration to predict BPD severity was the reduction of miR-876-3p. When exogenous miR-876-3p was administered in experimental hyperoxia-induced BPD, animals exhibited reduced inflammation and improved alveologenesis (Ref. Reference Lal, Olave, Travers, Rezonzew, Dolma, Simpson, Halloran, Aghai, das, Sharma, Xu, Genschmer, Russell, Szul, Yi, Blalock, Gaggar, Bhandari and Ambalavanan70). EV miR-21 has been implicated in adults with lung diseases and was upregulated in serum-derived EVs of preterm infants (≤32 weeks gestation) with versus without lung disorder (Ref. Reference Go, Maeda, Miyazaki, Maeda, Kume, Namba, Momoi, Hashimoto, Otsuru, Kawasaki, Hosoya and Dennery75).

Chronic obstructive pulmonary disease

COPD is a heterogeneous group of respiratory diseases characterized by progressive and partially irreversible restriction of airflow due to small airway damage (chronic bronchitis) or extensive alveolar wall disruption with airspace enlargement (emphysema) (Ref. Reference Antunes, Lopes-Pacheco and Rocco76). COPD has the highest mortality rate among chronic respiratory diseases (Ref. Reference Momtazmanesh, Moghaddam, Ghamari, Rad, Rezaei, Shobeiri, Aali, Abbasi-Kangevari, Abbasi-Kangevari, Abdelmasseh, Abdoun, Abdulah, Md Abdullah, Abedi, Abolhassani, Abrehdari-Tafreshi, Achappa, Adane Adane, Adane, Addo, Adnan, Sakilah Adnani, Ahmad, Ahmadi, Ahmadi, Ahmed, Ahmed, Rashid, al Hamad, Alahdab, Alemayehu, Alif, Aljunid, Almustanyir, Altirkawi, Alvis-Guzman, Dehkordi, Amir-Behghadami, Ancuceanu, Andrei, Andrei, Antony, Anyasodor, Arabloo, Arulappan, Ashraf, Athari, Attia, Ayele, Azadnajafabad, Babu, Bagherieh, Baltatu, Banach, Bardhan, Barone-Adesi, Barrow, Basu, Bayileyegn, Bensenor, Bhardwaj, Bhardwaj, Bhat, Bhattacharyya, Bouaoud, Braithwaite, Brauer, Butt, Butt, Calina, Cámera, Chanie, Charalampous, Chattu, Chimed-Ochir, Chu, Cohen, Cruz-Martins, Dadras, Darwesh, das, Debela, Delgado-Ortiz, Dereje, Dianatinasab, Diao, Diaz, Digesa, Dirirsa, Doku, Dongarwar, Douiri, Dsouza, Eini, Ekholuenetale, Ekundayo, Mustafa Elagali, Elhadi, Enyew, Erkhembayar, Etaee, Fagbamigbe, Faro, Fatehizadeh, Fekadu, Filip, Fischer, Foroutan, Franklin, Gaal, Gaihre, Gaipov, Gebrehiwot, Gerema, Getachew, Getachew, Ghafourifard, Ghanbari, Ghashghaee, Gholami, Gil, Golechha, Goleij, Golinelli, Guadie, Gupta, Gupta, Gupta, Gupta, Hadei, Halwani, Hanif, Hargono, Harorani, Hartono, Hasani, Hashi, Hay, Heidari, Hellemons, Herteliu, Holla, Horita, Hoseini, Hosseinzadeh, Huang, Hussain, Hwang, Iavicoli, Ibitoye, Ibrahim, Ilesanmi, Ilic, Ilic, Immurana, Ismail, Merin J, Jakovljevic, Jamshidi, Janodia, Javaheri, Jayapal, Jayaram, Jha, Johnson, Joo, Joseph, Jozwiak, K, Kaambwa, Kabir, Kalankesh, Kalhor, Kandel, Karanth, Karaye, Kassa, Kassie, Keikavoosi-Arani, Keykhaei, Khajuria, Khan, Khan, Khan, Khreis, Kim, Kisa, Kisa, Knibbs, Kolkhir, Komaki, Kompani, Koohestani, Koolivand, Korzh, Koyanagi, Krishan, Krohn, Kumar, Kumar, Kurmi, Kuttikkattu, la Vecchia, Lám, Lan, Lasrado, Latief, Lauriola, Lee, Lee, Legesse, Lenzi, Li, Lin, Liu, Liu, Lo, Lorenzovici, Lu, Mahalingam, Mahmoudi, Mahotra, Malekpour, Malik, Mallhi, Malta, Mansouri, Mathews, Maulud, Mechili, Nasab, Menezes, Mengistu, Mentis, Meshkat, Mestrovic, Micheletti Gomide Nogueira de Sá, Mirrakhimov, Misganaw, Mithra, Moghadasi, Mohammadi, Mohammadi, Mohammadshahi, Mohammed, Mohan, Moka, Monasta, Moni, Moniruzzaman, Montazeri, Moradi, Mostafavi, Mpundu-Kaambwa, Murillo-Zamora, Murray, Nair, Nangia, Swamy, Narayana, Natto, Nayak, Negash, Nena, Kandel, Niazi, Nogueira de Sá, Nowroozi, Nzoputam, Nzoputam, Oancea, Obaidur, Odukoya, Okati-Aliabad, Okekunle, Okonji, Olagunju, Bali, Ostojic, A, Padron-Monedero, Padubidri, Pahlevan Fallahy, Palicz, Pana, Park, Patel, Paudel, Paudel, Pedersini, Pereira, Pereira, Petcu, Pirestani, Postma, Prashant, Rabiee, Radfar, Rafiei, Rahim, Ur Rahman, Rahman, Rahman, Rahmani, Rahmani, Rahmanian, Rajput, Rana, Rao, Rao, Rashedi, Rashidi, Ratan, Rawaf, Rawaf, Rawal, Rawassizadeh, Razeghinia, Mohamed Redwan, Rezaei, Rezaei, Rezaei, Rezaeian, Rodrigues, Buendia Rodriguez, Roever, Rojas-Rueda, Rudd, Saad, Sabour, Saddik, Sadeghi, Sadeghi, Saeed, Sahebazzamani, Sahebkar, Sahoo, Sajid, Sakhamuri, Salehi, Samy, Santric-Milicevic, Sao Jose, Sathian, Satpathy, Saya, Senthilkumaran, Seylani, Shahabi, Shaikh, Shanawaz, Shannawaz, Sheikhi, Shekhar, Sibhat, Simpson, Singh, Singh, Singh, Skryabin, Skryabina, Soltani-Zangbar, Song, Soyiri, Steiropoulos, Stockfelt, Sun, Takahashi, Talaat, Tan, Tat, Tat, Taye, Thangaraju, Thapar, Thienemann, Tiyuri, Ngoc Tran, Tripathy, Car, Tusa, Ullah, Ullah, Vacante, Valdez, Valizadeh, van Boven, Vasankari, Vaziri, Violante, Vo, Wang, Wei, Westerman, Wickramasinghe, Xu, Xu, Yadav, Yismaw, Yon, Yonemoto, Yu, Yu, Yunusa, Zahir, Zangiabadian, Zareshahrabadi, Zarrintan, Zastrozhin, Zegeye, Zhang, Naghavi, Larijani and Farzadfar1), with cigarette smoke (CS) representing the major risk factor for COPD development (Ref. Reference Osei, Mirbolouk, Orimoloye, Dzaye, Uddin, Benjamin, Hall, DeFilippis, Bhatnagar, Biswal and Blaha77). Alterations in lung structural and immune cell-derived EVs have been documented in COPD. Harmful gases or particles were demonstrated to stimulate the release of stressed lung cell-derived EVs that contribute to COPD-associated tissue damage (Ref. Reference Tan, Armitage, Teo, Ong, Shin and Moodley78). Activated neutrophils participate in alveolar deterioration by enhancing the release of EV-transported elastase, which causes significant tissue damage by degrading the lung extracellular matrix (Ref. Reference Genschmer, Russell, Lal, Szul, Bratcher, Noerager, Abdul Roda, Xu, Rezonzew, Viera, Dobosh, Margaroli, Abdalla, King, McNicholas, Wells, Dransfield, Tirouvanziam, Gaggar and Blalock79). An increase in the production of endothelial cell-derived EVs was also found in patients with exacerbated COPD (Ref. Reference Takahashi, Kobayashi, Fujino, Suzuki, Ota, He, Yamada, Suzuki, Yanai, Kurosawa, Yamaya and Kubo44). Compared to patients with stable disease, exacerbated ones exhibited a greater number of serum-derived EVs, which correlated with IL-6, C-reactive protein, and soluble TNF-R1 values (Ref. Reference Tan, Armitage, Teo, Ong, Shin and Moodley78). In an early study, a reduction in lung function (FEV1/FVC ratio) was correlated with increased release of endothelial cell-derived EVs in COPD (Ref. Reference Takahashi, Kobayashi, Fujino, Suzuki, Ota, He, Yamada, Suzuki, Yanai, Kurosawa, Yamaya and Kubo44), while a subsequent study evidenced a negative correlation between FEV1 and the number of circulating EVs (Ref. Reference Takahashi, Kobayashi, Fujino, Suzuki, Ota, Tando, Yamada, Yanai, Yamaya, Kurosawa, Yamauchi and Kubo80). Because pathogens also release EVs, infected patients with COPD may present worse symptoms. For instance, E. coli-derived EVs could accelerate alveolar disruption by intensifying neutrophilic inflammation via IL-17A-dependent signalling (Ref. Reference Kim, Lee, Choi, Choi, Heo, Gho, Jee, Oh and Kim81).

Mechanistic studies have provided novel insights into the onset and progression of COPD by investigating EV miRNA content. In human bronchial epithelial cells, CS extract exposure upregulated miR-21 and miR-210, increasing the production of α-smooth muscle actin and collagen type I and inducing lung fibroblast differentiation into myofibroblasts (Refs. Reference Fujita, Araya, Ito, Kobayashi, Kosaka, Yoshioka, Kadota, Hara, Kuwano and Ochiya82, Reference Xu, Ling, Xue, Dai, Sun, Chen, Liu, Zhou, Liu, Luo, Bian and Liu83). These alterations facilitated tissue remodelling that was prevented by administering a miR-21 inhibitor (Ref. Reference Xu, Ling, Xue, Dai, Sun, Chen, Liu, Zhou, Liu, Luo, Bian and Liu83). CS-exposed bronchial epithelial cells also demonstrated upregulation of miR-500a-5p, miR-574-5p, miR-656-5p, miR-3180-5p, and miR-3913-5p, and downregulation of miR-130b-5p, miR-222-5p, and miR-618 (Ref. Reference Corsello, Kudlicki, Garofalo and Casola84). Furthermore, EVs from CS-exposed endothelial cells were enriched with specific miRNAs (let-7d, miR-125a, miR-126, and miR-191) that, once engulfed by macrophages, inhibited their clearance abilities (Ref. Reference Serban, Rezania, Petrusca, Poirier, Cao, Justice, Patel, Tsvetkova, Kamocki, Mikosz, Schweitzer, Jacobson, Cardoso, Carlesso, Hubbard, Kechris, Dragnea, Berdyshev, McClintock and Petrache85). CS-exposed airway epithelial cells released EVs containing miR-7, miR-21-3p, miR-27b-3p, miR-125a-5p, and miR-221-3p that induced macrophage polarization toward pro-inflammatory M1 phenotype (Refs. Reference Chen, Wu, Shi, Fan, Zhang, Su, Wang and Li86Reference Jiang, Wang, Zhang, Min and Gu89). In another study using CS-induced emphysema, airway epithelial cells exhibited increased levels of EV miR-93, which increased levels of macrophage-produced metalloproteinases 9 and 12 and promoted lung remodelling by excessively degrading elastin (Ref. Reference Xia, Wu, Zhao, Li, Lu, Ma, Cheng, Sun, Xiang, Bian and Liu90). CS-exposed airway epithelial cells can also contribute to lung remodelling by reducing EV-packaged miR-422a, leading to greater secretion of phosphoprotein-1 and production of collagen type I and smooth muscle actin by fibroblasts (Refs. Reference Xu, Ling, Xue, Dai, Sun, Chen, Liu, Zhou, Liu, Luo, Bian and Liu83, Reference Dai, Lin, Xu, Hu, Gou and Xu91).

Several studies have investigated alterations in EV miRNA expression to distinguish patients with distinct severity of COPD or from other diseases. Levels of miR-199a-5p were upregulated in plasma-derived EVs from patients with COPD compared to non-smokers (Ref. Reference Sundar, Li and Rahman92). A negative correlation between this miRNA and FEV1 was observed (Ref. Reference Mizuno, Bogaard, Gomez-Arroyo, Alhussaini, Kraskauskas, Cool and Voelkel93). In plasma-derived EVs, a set of four miRNAs (miR-92b-3p, miR-106b-3p, miR-223-3p, and miR-374a-5p) demonstrated high diagnostic accuracy in segregating stable and exacerbated patients (Ref. Reference O’Farrell, Bowman and Fong94), while neutrophil count combined with serum-derived EV miR-1258 also demonstrated high accuracy for such (Ref. Reference Wang, Yang, Qiao, Bai, Li, Sun, Liu, Yang and Cui95). Muscle dysfunction in COPD patients was detected by upregulation of three plasma-derived EV miRNAs (miR-133a-3p, miR-133a-5p, miR-206) (Ref. Reference Carpi, Polini, Nieri, Dubbini, Celi, Nieri and Neri96). Patients with lung cancer or stable COPD differed in the expression of 14 plasma-derived EV miRNAs, of which the highest diagnostic accuracy was found by the expression of miR-27a-3p combined with miR-106b-3p and miR-361-5p (Ref. Reference O’Farrell, Bowman, Fong and Yang97). Inflammatory endotypes of COPD (neutrophilic versus eosinophilic) were also segregated by differentially expressed EV miRNAs in the BALF (Ref. Reference Burke, Cellura, Freeman, Hicks, Ostridge, Watson, Williams, Spalluto, Staples and Wilkinson98).

Cystic fibrosis

CF is a life-limiting genetic disease caused by mutations in the CF transmembrane conductance regulator (CFTR) gene and affects over 100,000 individuals worldwide. Despite the multiorgan involvement, CF morbidity and mortality primarily result from end-stage lung disease due to impaired mucociliary clearance, chronic inflammation, and recurrent infection (Ref. Reference Lopes-Pacheco99). Emerging evidence suggests that EVs participate in inflammation and immune responses in CF (Ref. Reference Bacalhau, Camargo and Lopes-Pacheco41). In this context, higher EV counts were observed in CF cell lines (CFBE41o and CuFi-5) compared to control cells (16HBE14o and Nuli-1) (Ref. Reference Useckaite, Ward, Trappe, Reilly, Lennon, Davage, Matallanas, Cassidy, Dillon, Brennan, Doyle, Carter, Donnelly, Linnane, McKone, McNally and Coppinger27). Differences in EV cargo from CF and non-CF cell lines were also evidenced in response to Pseudomonas aeruginosa (Ref. Reference Lozano-Iturbe, Blanco-Agudín and Vázquez-Espinosa100), which is the most prevalent pathogen found in CF lungs. Interestingly, primary bronchial epithelial cell-derived EVs inhibited P. aeruginosa biofilm formation by mitigating levels of biofilm-related proteins in a CF mouse model (Ref. Reference Sarkar, Barnaby, Nymon, Taatjes, Kelley and Stanton101). On the contrary, both WT and CF airway epithelial cell-derived EVs exposed to P. aeruginosa exhibited greater content of pro-inflammatory mediators; however, responses from CF macrophages were mitigated in comparison to WT macrophages (Ref. Reference Koeppen, Nymon, Barnaby, Li, Hampton, Ashare and Stanton102). BALF-derived EVs revealed an enrichment of protein signatures related to macrophage activation and inflammation in a murine model of CF-like muco-inflammatory lung disease (Ref. Reference Mao, Suryawanshi, Patial and Saini103).

In sputum from CF patients, a high number of EVs – predominantly from granulocytes – was evidenced, and when these EVs were intratracheally administered in mice, there was strong neutrophilia in their lungs (Ref. Reference Porro, Di Gioia and Trotta104). Likewise, CF BALF-derived EVs contained higher levels of leukocyte chemotaxis-related proteins and drove neutrophil recruitment (Refs. Reference Useckaite, Ward, Trappe, Reilly, Lennon, Davage, Matallanas, Cassidy, Dillon, Brennan, Doyle, Carter, Donnelly, Linnane, McKone, McNally and Coppinger27, Reference Rollet-Cohen, Bourderioux, Lipecka, Chhuon, Jung, Mesbahi, Nguyen-Khoa, Guérin-Pfyffer, Schmitt, Edelman, Sermet-Gaudelus and Guerrera43). By analysing BALF-derived EVs, unique protein signatures were identified to differentiate CF stable and exacerbated versus controls (Ref. Reference Useckaite, Ward, Trappe, Reilly, Lennon, Davage, Matallanas, Cassidy, Dillon, Brennan, Doyle, Carter, Donnelly, Linnane, McKone, McNally and Coppinger27). BALF-derived EVs from paediatric CF patients (similar to those from asthmatic patients) were found to enhance epithelial sodium channel activity in small airway epithelia (Ref. Reference Al-Humiari, Yu and Liu105). Compared to controls, CF EVs exhibited alterations in lipid profile with increased ceramide production, which might accelerate the release of EV-packaged pro-inflammatory ceramides and perpetuate the inflammatory state in CF (Ref. Reference Zulueta, Peli, Dei Cas, Colombo, Paroni, Falleni, Baisi, Bollati, Chiaramonte, del Favero, Ghidoni and Caretti106). CF airway fluid-derived EVs demonstrated an enrichment with IL-1α, IL-1β, IL-18, and active caspase-1, which stimulate a hyperinflammatory state by inducing inflammasome activation of surrounding cells (namely, epithelial cells and newly recruited neutrophils) (Ref. Reference Forrest, Dobosh, Ingersoll, Rao, Rojas, Laval, Alvarez, Brown, Tangpricha and Tirouvanziam107). In sputum-derived EVs from CF patients treated for pulmonary exacerbation, levels of EV-transported neutrophil elastase and myeloperoxidase were decreased and demonstrated good correlation with improvement in lung function (Ref. Reference Ben-Meir, Antounians, Eisha, Ratjen, Zani and Grasemann108).

While BALF and sputum have been utilized as the main sources of EVs in CF studies, recent evidence demonstrated that urine-derived EVs can be a suitable source to distinguish CF patients and healthy controls (Ref. Reference Gauthier, Pranke, Jung, Martignetti, Stoven, Nguyen-Khoa, Semeraro, Hinzpeter, Edelman, Guerrera and Sermet-Gaudelus109). Indeed, CF EVs exhibited increased expression of epidermal growth factor receptor (EGFR) and decreased expression of klotho and matrisome, providing insights into CFTR-related kidney dysfunction (Ref. Reference Gauthier, Pranke, Jung, Martignetti, Stoven, Nguyen-Khoa, Semeraro, Hinzpeter, Edelman, Guerrera and Sermet-Gaudelus109).

Recent studies have assessed the impact of clinically approved CFTR modulators on CF EVs (Refs. Reference Useckaite, Ward, Trappe, Reilly, Lennon, Davage, Matallanas, Cassidy, Dillon, Brennan, Doyle, Carter, Donnelly, Linnane, McKone, McNally and Coppinger27, Reference Trappe, Lakkappa, Carter, Dillon, Wynne, McKone, McNally and Coppinger110) since these drugs can correct the basic defects of the mutant CFTR protein (Refs. Reference Lopes-Pacheco99, Reference Oliver, Carlon, Pedemonte and Lopes-Pacheco111). In CFBE41o cells, treatment with lumacaftor/ivacaftor or tezacaftor/ivacaftor led to a significant reduction of EV release (Ref. Reference Useckaite, Ward, Trappe, Reilly, Lennon, Davage, Matallanas, Cassidy, Dillon, Brennan, Doyle, Carter, Donnelly, Linnane, McKone, McNally and Coppinger27). Although children with CF and healthy controls demonstrated equivalent EV counts, treatment with elexacaftor/tezacaftor/ivacaftor altered EV protein content in serum-derived EVs of patients with CF from different age groups (Ref. Reference Trappe, Lakkappa, Carter, Dillon, Wynne, McKone, McNally and Coppinger110).

Idiopathic pulmonary fibrosis

IPF is a progressive disorder characterized by abnormal deposition of extracellular matrix in diverse regions of the lung parenchyma in response to chronic epithelial cell damage. The disease affects over three million people worldwide and presents a median survival rate of 2–5 years with only 20–25% of patients living beyond 10 years (Ref. Reference Nathan, Shlobin, Weir, Ahmad, Kaldjob, Battle, Sheridan and du Bois112). Under normal conditions, lung fibroblasts communicate with surrounding cells by EVs enriched with prostaglandins to inhibit myofibroblast differentiation; however, a reduction in EV-transported prostaglandins could trigger pulmonary fibrosis (Refs. Reference Lacy, Woeller, Thatcher, Pollock, Small, Sime and Phipps36, Reference Kadota, Yoshioka, Fujita, Araya, Minagawa, Hara, Miyamoto, Suzuki, Fujimori, Kohno, Fujii, Kishi, Kuwano and Ochiya113). Several studies have demonstrated the detrimental influence of EV-transported cargo on cell senescence and EMT during IPF development and progression. For instance, decreased reparative properties were observed for senescent cell-derived EVs (Refs. Reference Kadota, Fujita, Yoshioka, Araya, Kuwano and Ochiya114, Reference Antunes, Braga, Oliveira, Kitoko, Castro, Xisto, Coelho, Rocha, Silva-Aguiar, Caruso-Neves, Martins, Carvalho, Galina, Weiss, Lapa e Silva, Lopes-Pacheco, Cruz and Rocco115). Administration of syndecan-1-positive-EVs into mice lungs led to epithelial reprogramming by upregulating TGF-β and WNT signalling (Ref. Reference Parimon, Yao, Habiel, Ge, Bora, Brauer, Evans, Xie, Alonso-Valenteen, Medina-Kauwe, Jiang, Noble, Hogaboam, Deng, Burgy, Antes, Königshoff, Stripp, Gharib and Chen116). WNT signalling plays a role in IPF pathogenesis by promoting fibroblast proliferation, and an elevated number of WNT5A-positive EVs was found in BALF from IPF patients compared to non-IPF controls (Ref. Reference Martin-Medina, Lehmann, Burgy, Hermann, Baarsma, Wagner, de Santis, Ciolek, Hofer, Frankenberger, Aichler, Lindner, Gesierich, Guenther, Walch, Coughlan, Wolters, Lee, Behr and Königshoff117). Fibroblast-derived EVs contain high levels of fibronectin on their surface that, once bound to other fibroblasts, stimulate invasion by focal adhesion kinase- and steroid receptor coactivator kinase-mediated mechanisms (Ref. Reference Chanda, Otoupalova, Hough, Locy, Bernard, Deshane, Sanderson, Mobley and Thannickal118).

Various studies have investigated the influence of EV-transferred miRNAs on aberrant EMT and fibrosis development. For instance, bronchial epithelial cells from healthy subjects release much less EVs than those cells from IPF patients, which also contain increased levels of miR-7, miR-137, miR-195, and miR-411. These EVs can also induce senescence in naïve epithelial cells (Ref. Reference Asghar, Monkley, Smith, Hewitt, Grime, Murray, Overed-Sayer and Molyneaux119). In IPF sputum-derived EVs, seven miRNAs were upregulated, and there was a negative correlation between miR-142-3p levels and lung diffusing capacity (Ref. Reference Njock, Guiot, Henket, Nivelles, Thiry, Dequiedt, Corhay, Louis and Struman120). Interestingly, increased expression of miR-142-3p was suggested to come from macrophages and was able to reduce TGFβ-R1 expression in lung fibroblasts and alveolar epithelial cells (Ref. Reference Guiot, Cambier, Boeckx, Henket, Nivelles, Gester, Louis, Malaise, Dequiedt, Louis, Struman and Njock121). Compared to non-IPF cells, lung fibroblasts from IPF patients release more EVs that can induce increased senescence and mitochondrial dysfunction in airway epithelial cells (Refs. Reference Kadota, Yoshioka, Fujita, Araya, Minagawa, Hara, Miyamoto, Suzuki, Fujimori, Kohno, Fujii, Kishi, Kuwano and Ochiya113, Reference Chanda, Otoupalova, Hough, Locy, Bernard, Deshane, Sanderson, Mobley and Thannickal118). IPF fibroblast-derived EVs carry high levels of miR-23b-3p and miR-494-3p, which lead to epithelial phenotypic alterations by inhibiting sirtuin-3. Such mechanisms may be involved in mitochondrial dysfunction and increased levels of reactive oxygen species that result in DNA damage and epithelial cell senescence (Ref. Reference Kadota, Yoshioka, Fujita, Araya, Minagawa, Hara, Miyamoto, Suzuki, Fujimori, Kohno, Fujii, Kishi, Kuwano and Ochiya113). Upregulation of miR-21-5 was found in serum-derived EVs of IPF patients with a good prediction of mortality in the 30-month follow-up (Ref. Reference Makiguchi, Yamada, Yoshioka, Sugiura, Koarai, Chiba, Fujino, Tojo, Ota, Kubo, Kobayashi, Yanai, Shimura, Ochiya and Ichinose122). Levels of EV-transferred miR-21-5p and the rate of decline of spirometry FVC also demonstrated good correlation (Ref. Reference Makiguchi, Yamada, Yoshioka, Sugiura, Koarai, Chiba, Fujino, Tojo, Ota, Kubo, Kobayashi, Yanai, Shimura, Ochiya and Ichinose122).

Lung cancer

Lung cancer is the leading cause of malignant neoplasms and accounts for ~25% of all global cancer deaths (Ref. Reference Siegel, Miller, Wagle and Jemal123). Extensive research has been performed to investigate EVs and their cargo for early cancer detection. For instance, increased expression of EGFR has been detected in lung cancer cells with an even higher level of EGFR in EVs from patients with lung cancer compared to healthy controls (Ref. Reference Yamashita, Kamada, Kanasaki, Maeda, Nagano, Abe, Inoue, Yoshioka, Tsutsumi, Katayama, Inoue and Tsunoda124). Lung cancer cell-derived EVs contain miR-21 and miR-29a that bind to TLR8 on immune cells within the tumour niche, leading to a pro-inflammatory state via NF-κB activation that results in tumour progression and metastasis (Ref. Reference Fabbri, Paone, Calore, Galli, Gaudio, Santhanam, Lovat, Fadda, Mao, Nuovo, Zanesi, Crawford, Ozer, Wernicke, Alder, Caligiuri, Nana-Sinkam, Perrotti and Croce125). Early tumour formation is usually marked by hypoxia with increased levels of EV hypoxic signature proteins associated with EMT (Ref. Reference Lobb, Visan, Wu, Norris, Hastie, Everitt, Yang, Bowman, Siva, Larsen, Gorman, MacManus, Leimgruber, Fong and Möller126). Under hypoxic conditions, lung cancer cell-derived EVs express high levels of EGFR, TGF-β, miR-23a, and miR-619-5p that create an immunosuppressive state by polarizing macrophages (Ref. Reference Kim, Park, Kim, Choi, Kim, Sung, Sung, Choi, Yun, Yi, Lee, Kim, Lee and Rho127) and eliminating NK cells (Ref. Reference Cohnen, Chiang, Stojanovic, Schmidt, Claus, Saftig, Janßen, Cerwenka, Bryceson and Watzl128), while inducing angiogenesis (Ref. Reference Fabbri, Paone, Calore, Galli, Gaudio, Santhanam, Lovat, Fadda, Mao, Nuovo, Zanesi, Crawford, Ozer, Wernicke, Alder, Caligiuri, Nana-Sinkam, Perrotti and Croce125), thus facilitating the dissemination of abnormal cells.

The cargo of lung cancer-derived EVs contributes to drug resistance (Ref. Reference Liu, Gao, Ma, Huang, Chen, Zeng, Ashby, Zou and Chen129). Analysis of lung cancer-derived EVs indicated cisplatin response linked to high expression of miR-146a-5p and cisplatin resistance associated with miR-96 and miR-425-3p (Refs. Reference Yamashita, Kamada, Kanasaki, Maeda, Nagano, Abe, Inoue, Yoshioka, Tsutsumi, Katayama, Inoue and Tsunoda124, Reference Zhang and Xu130). Furthermore, poor prognosis was observed by miR-378 upregulation of serum-derived EVs from patients with non-small cell lung cancer (NSCLC) (Ref. Reference Zhang and Xu130). In patients with lung adenocarcinoma, plasma EVs exhibited elevated expression of Ras homolog family member V. By EV array, CD151/tetraspanin-24, CD171/L1CAM, and tetraspanin-8 were found to be abnormally expressed in histological slices of lung tumours (Ref. Reference Kato, Vykoukal, Fahrmann and Hanash131). Prospective miRNAs for early diagnosis and prognosis have been identified for lung cancer. In transcriptomic analysis, miR-30a-3p, miR-30e-3p, miR-181-5p, and miR-361-5p were identified for the diagnosis of lung adenocarcinoma, while miR-10b-5p, miR-15b-5p, and miR-320b were suggested for lung squamous cell carcinoma (Ref. Reference Jin, Chen, Chen, Fei, Chen, Cai, Liu, Lin, Su, Zhao, Su, Pan, Shen, Xie and Xie132). Prediction of survival rate by lung cancer was also suggested by analysis of let-7, miR-137, miR-182, miR-221, and miR-372 expressed in lung cancer cell-derived EVs (Ref. Reference Jin, Chen, Chen, Fei, Chen, Cai, Liu, Lin, Su, Zhao, Su, Pan, Shen, Xie and Xie132).

Pulmonary arterial hypertension

PAH is a rare, progressive disease characterized by pulmonary vascular inflammation and remodelling that leads to right ventricular failure. In PAH patients, the number of circulating EVs was demonstrated to be significantly increased (Refs. Reference Bakouboula, Morel, Faure, Zobairi, Jesel, Trinh, Zupan, Canuet, Grunebaum, Brunette, Desprez, Chabot, Weitzenblum, Freyssinet, Chaouat and Toti133Reference Khandagale, Åberg, Wikström, Bergström Lind, Shevchenko, Björklund, Siegbahn and Christersson135) and directly correlated with functional impairment (Ref. Reference Bakouboula, Morel, Faure, Zobairi, Jesel, Trinh, Zupan, Canuet, Grunebaum, Brunette, Desprez, Chabot, Weitzenblum, Freyssinet, Chaouat and Toti133) and mortality rate (Ref. Reference Amabile, Heiss, Chang, Angeli, Damon, Rame, McGlothlin, Grossman, de Marco and Yeghiazarians136). Proteomic analysis revealed 13 proteins associated with coagulation and oxidative stress that were differentially expressed in blood-derived EVs from PAH patients compared to healthy controls (Ref. Reference Khandagale, Åberg, Wikström, Bergström Lind, Shevchenko, Björklund, Siegbahn and Christersson135). Endothelial cell-derived EV counts and levels of tricuspid annular plane systolic excursion (prognostic predictor for hypertension) were also higher in urine samples from PAH patients compared to controls (Ref. Reference Rose, Wanner, Cheong, Queisser, Barrett, Park, Hite, Naga Prasad, Erzurum and Asosingh137). Treatment with prostacyclin analogues reduced platelet- and leukocyte-derived EV counts and inhibited platelet reactivity and thrombus formation in PAH patients (Ref. Reference Gąsecka, Banaszkiewicz, Nieuwland, van der Pol, Hajji, Mutwil, Rogula, Rutkowska, Pluta, Eyileten, Postuła, Darocha, Huczek, Opolski, Filipiak, Torbicki and Kurzyna138).

EV content has been profiled to identify miRNAs and other factors that may be useful for PAH diagnosis and prognosis. For instance, miR-26a-5p was downregulated and miR-486-5p was upregulated in plasma-derived EVs of PAH patients (Ref. Reference Khandagale, Corcoran, Nikpour, Isaksson, Wikström, Siegbahn and Christersson139). A high level of miR-596 in plasma-derived EVs was associated with severe PAH and poor prognosis (Ref. Reference Huang, Wang, Tang, Gong, Sun, Wang, Jiang, Wu, Luo, Zhang, Yang, Li, Yuan, Zhao and Yuan140). Translationally controlled tumour protein has increased expression in lung tissue sections of PAH patients, and its transfer by EVs led to proliferation and apoptosis resistance of pulmonary arterial smooth muscle cells that resulted in vascular remodelling (Ref. Reference Ferrer, Dunmore, Hassan, Ormiston, Moore, Deighton, Long, Yang, Stewart and Morrell141). In experimental monocrotaline-induced PAH, EV-packaged miR-211 induced proliferation of pulmonary arterial smooth muscle cells by inhibiting CaMK1/PPAR-γ signalling. Downregulation of miR-211 mitigated PAH in rats (Ref. Reference Zhang, Liu, Zheng, Chen, Sun, Yao, Sun, Lin, Lin and Yuan142).

Other respiratory diseases

Recent evidence indicates that cilia-derived EVs possess distinct mechanisms of release and molecular features compared to cytosolic-derived EVs (Ref. Reference Mohieldin, Pala, Beuttler, Moresco, Yates and Nauli143). In primary ciliary dyskinesia, a rare genetic disease caused by mutations in genes related to cilia development and function, proteomic analysis demonstrated high levels of proteins involved in leukocyte chemotaxis and antioxidant activity (Ref. Reference Rollet-Cohen, Bourderioux, Lipecka, Chhuon, Jung, Mesbahi, Nguyen-Khoa, Guérin-Pfyffer, Schmitt, Edelman, Sermet-Gaudelus and Guerrera43).

Both silicosis and pulmonary sarcoidosis exhibit granuloma formation and extensive lung parenchyma fibrosis resulting from different aetiologies. While silicosis is caused by silica inhalation mainly in work environments, systemic inflammation with unknown causes is responsible for sarcoidosis. In experimental silicosis, macrophage-derived EVs contributed to silica-induced lung fibrosis by activating fibroblasts in a mechanism dependent on endoplasmic reticulum stress. A significant decrease in lung fibrosis and levels of pro-inflammatory mediators (TNF-α, IL-1β, and IL-6) in BALF was observed when animals were pre-treated with the EV-production inhibitor GW4869 (Ref. Reference Qin, Lin, Liu, Li, Li, Deng, Chen, Chen, Niu, Li and Hu144). Serum-derived EVs from an animal model of silicosis demonstrated increased levels of miR-107 and miR-125-5p, which can promote fibroblast differentiation and contribute to lung fibrosis (Refs. Reference Xia, Wang, Guo, Pei, Zhang, Bao, Li, Qu, Zhao, Hao and Yao145,Reference Ding, Pei, Zhang, Qi, Xia, Hao and Yao146). In addition, levels of miR-223-3p were significantly altered in EVs and tissue from silicosis patients, thus being considered a promising biomarker (Ref. Reference Hou, Zhu, Jiang, Zhao, Jia, Jiang, Wang, Xue, Wang and Tian147). On the contrary, sarcoidosis patients demonstrated altered levels of CD14 and lipopolysaccharide-binding protein on serum-derived EVs by proteomics analysis (Ref. Reference Futami, Takeda, Koba, Narumi, Nojima, Ito, Nakayama, Ishida, Yoshimura, Naito, Fukushima, Takimoto, Edahiro, Matsuki, Nojima, Hirata, Koyama, Iwahori, Nagatomo, Shirai, Suga, Satoh, Futami, Miyake, Shiroyama, Inoue, Adachi, Tomonaga, Ueda and Kumanogoh148). Treatment with methotrexate also reduced levels of EV-transported serpin C1 in sarcoidosis patients, suggesting that it can be a good predictor for therapy monitoring (Ref. Reference Kraaijvanger, Janssen Bonás, Grutters, Paspali, Veltkamp, de Kleijn and van Moorsel149).

Emerging therapeutic properties of EVs for respiratory diseases

One of the most attractive applications of EVs is in cell-free therapeutics, as these particles carry several molecules that can modulate lung resident and immune responses and assist in tissue repair (Figure 2). In this context, mesenchymal stromal cell (MSC)-derived EVs are particularly promising for such purposes due to their multimodal ability to promote anti-inflammatory and antimicrobial actions, fluid clearance, and recovery of epithelial and endothelial permeability in a variety of experimental models of respiratory diseases (Figure 3). As EVs are non-replicating, they are also considered safer and easier to store and distribute for therapeutic purposes than living cells. In addition, recent evidence indicates that EV therapeutic properties may be potentiated by stimulating MSCs with biological, chemical, or physical methods before EV isolation (Ref. Reference Lopes-Pacheco and Rocco150).

Figure 3. Mesenchymal stromal cell (MSC)-derived extracellular vesicles (EVs) in respiratory diseases. Major therapeutic effects of MSC-derived EVs were found in experimental models of asthma, bronchopulmonary dysplasia (BPD), chronic obstructive pulmonary disease (COPD), lung cancer, lung fibrosis, and pulmonary arterial hypertension (PAH).

Asthma

Therapeutic effects of MSC-derived EVs have been investigated in several experimental models of asthma, including Aspergillus hyphal extract, HDM, and ovalbumin. In an early study, MSC-derived conditioned medium prevented peribronchial inflammation and airway smooth muscle thickening in experimental asthma by an adiponectin-promoted mechanism (Ref. Reference Ionescu, Alphonse, Arizmendi, Morgan, Abel, Eaton, Duszyk, Vliagoftis, Aprahamian, Walsh and Thébaud151). Subsequent studies demonstrated that EVs from both murine and human MSCs reduce airway hyperresponsiveness and lung inflammation by mitigating BALF neutrophil and eosinophil counts and levels of IL-4, IL-5, and IL-17 in Aspergillus hyphal extract-induced asthma (Ref. Reference Cruz, Borg, Goodwin, Sokocevic, Wagner, Coffey, Antunes, Robinson, Mitsialis, Kourembanas, Thane, Hoffman, McKenna, Rocco and Weiss152). MSC-derived EVs also stimulated T regulatory cell proliferation and immunosuppressive properties by enhancing IL-10 and TGF-β levels in peripheral blood mononuclear cells of asthmatic patients (Ref. Reference Du, Zhuansun and Chen153).

In experimental ovalbumin-induced asthma, MSC-derived EVs were efficient at reducing lung eosinophilia, fibrosis, and IgE and TGF-β levels (Refs. Reference de Castro, Xisto, Kitoko, Cruz, Olsen, Redondo, Ferreira, Weiss, Martins, Morales and Rocco154, Reference Mun, Kang, Park, Yu, Cho and Roh155), decreasing the number of mucus-producing goblet cells (Ref. Reference Song, Zhu and Wei156) and modulating macrophage polarization (Ref. Reference Fang, Zhang, Meng, Wang, He, Peng, Xu, Fan, Wu, Wu, Zheng and Fu157). MSC-derived EVs mitigated airway remodelling by promoting M2-like macrophage activation via FoxO1 signalling (Ref. Reference Shang, Sun, Xu, Ge, Hu, Xiao, Ning, Dong and Bai158). TNF receptor-associated factor 1 was also involved in macrophage polarization by MSC-derived EVs by regulating NF-κB and AKT signalling pathways (Ref. Reference Dong, Wang, Zhang, Zhang, Gu, Guo, Zuo, Pan, Hsu, Wang and Wang159). Furthermore, EMT and airway remodelling in asthma are influenced by WNT/β-catenin signalling activation, and MSC-derived EVs prevented tissue remodelling by inhibiting WNT/β-catenin signalling pathway-related factors. Such effects were reversed by the administration of BML-284, a WNT agonist (Ref. Reference Song, Zhu and Wei156). Likewise, the downregulation of miR-188, an EV-enriched miRNA, mitigated the protective effects of MSC-derived EVs in experimental ovalbumin-induced asthma (Ref. Reference Shan, Liu, Zhang, Zhou and Shang160). EV-transferred miR-221-3p inhibited FGF2 expression and ERK1/2 signalling, leading to mitigated lung inflammation and remodelling in experimental asthma (Ref. Reference Liu, Lin, Nie, Wan, Jiang and Zhang161). In samples from asthmatic patients, STAT3 and miR-301a-3p levels were demonstrated to be negatively correlated. MSC-derived EV miR-301a-3p was efficiently internalized by airway smooth muscle cells, which inhibits their proliferation and migration by targeting STAT3 (Ref. Reference Feng, Bai, Li, Zhao, Sun, Bao, Ren and Su162). miR-223-3p is highly expressed in MSC-derived EVs and has a major protective action on asthma-induced airway remodelling by regulating NLRP3/caspase-1/gasdermin D-mediated inflammasome activation and pyroptosis (Ref. Reference Li and Yang163).

Under hypoxic conditions, a higher number of EVs was released by MSCs (Refs. Reference Dong, Wang, Zheng, Pu, Ma, Qi, Zhang, Xue, Shan, Liu, Wang and Mao164, Reference Sholihah and Barlian165). When these EVs were administered in ovalbumin-induced asthma, there was greater improvement in lung inflammation (BALF total cell and eosinophil counts and levels of IL-4 and IL-13) and remodelling (levels of α-smooth muscle actin, collagen-1, and TGF-β1) compared to EVs from normoxia-cultured MSCs (Ref. Reference Dong, Wang, Zheng, Pu, Ma, Qi, Zhang, Xue, Shan, Liu, Wang and Mao164). Nebulization of hypoxic MSC-derived EVs was also effective at reducing lung inflammation and remodelling (Ref. Reference Xu, Wang, Luo, Gao, Gu, Ma, Xu, Yu, Liu, Liu, Wang, Zheng, Mao and Dong166), levels of IgE and pro-inflammatory cytokines (Ref. Reference Luo, Wang, Mao, Xu, Gu, Li, Mao, Zheng and Dong167). Upregulation of miR-146a-5p was evidenced on these EVs, and reduction of this EV-transferred miRNA impaired lung protective effects in experimental ovalbumin-induced asthma (Refs. Reference Dong, Wang, Zheng, Pu, Ma, Qi, Zhang, Xue, Shan, Liu, Wang and Mao164, Reference Xu, Wang, Luo, Gao, Gu, Ma, Xu, Yu, Liu, Liu, Wang, Zheng, Mao and Dong166).

Bronchopulmonary dysplasia

Several studies have investigated the potential of MSC-derived EVs for BPD therapy. In an initial study, MSC-derived EVs were demonstrated to improve pulmonary hypertension and lung function by reducing vascular remodelling, alveolar simplification, and fibrosis in hyperoxia-induced BPD in newborn mice (Ref. Reference Willis, Fernandez-Gonzalez, Anastas, Vitali, Liu, Ericsson, Kwong, Mitsialis and Kourembanas168). These effects were associated with macrophage polarization to M2 anti-inflammatory phenotype instead of M1 pro-inflammatory one (Ref. Reference Willis, Fernandez-Gonzalez, Anastas, Vitali, Liu, Ericsson, Kwong, Mitsialis and Kourembanas168). Both early and late administration of MSC-derived EVs were effective at improving core features of BPD on this model, thus preventing or reversing cardiorespiratory abnormalities (Ref. Reference Willis, Fernandez-Gonzalez, Reis, Yeung, Liu, Ericsson, Andrews, Mitsialis and Kourembanas169). Biodistribution analysis revealed MSC-derived EVs in lung tissue of hyperoxia-exposed newborn mice and epigenetic and phenotypic reprogramming of myeloid cells to a non-inflammatory phenotype (Ref. Reference Willis, Reis, Gheinani, Fernandez-Gonzalez, Taglauer, Yeung, Liu, Ericsson, Haas, Mitsialis and Kourembanas170). MSC-derived EVs also restored thymocyte development/maturation profile and thymic medullary structure by enhancing the expression of antioxidant-stress-related genes (Ref. Reference Reis, Willis, Fernandez-Gonzalez, Yeung, Taglauer, Magaletta, Parsons, Derr, Liu, Maehr, Kourembanas and Mitsialis171).

In a rat model of hyperoxia-induced BPD, MSC-derived EVs reduced alveolar simplification, lung damage, and fibrosis in a dose-dependent manner (Ref. Reference Ai, Shen, Sun, Zhu, Gao, du, Yuan, Chen and Zhou172). In vitro analysis revealed that EVs delay the transdifferentiation of alveolar type II cells into type I cells by downregulating WNT5a (Ref. Reference Ai, Shen, Sun, Zhu, Gao, du, Yuan, Chen and Zhou172). Antenatal administration of MSC-derived EVs was also able to reduce NLRP3- and IL-1β-mediated inflammation and preserve distal lung growth and mechanics in chorioamnionitis-induced rat BPD (Ref. Reference Abele, Taglauer, Almeda, Wilson, Abikoye, Seedorf, Mitsialis, Kourembanas and Abman173). These therapeutic benefits were associated with increased expression of vascular endothelial growth factor (VEGF) (Ref. Reference Abele, Taglauer, Almeda, Wilson, Abikoye, Seedorf, Mitsialis, Kourembanas and Abman173), which is decreased in preterm infants (Ref. Reference Lassus, Ristimäki and Ylikorkala174). Along these lines, EV-transferred VEGF mitigated lung injury by decreasing inflammation and improving alveolarization and angiogenesis in hyperoxia-induced rat BPD. Such effects were not found by EVs from VEGF-knockdown MSCs (Ref. Reference Ahn, Park, Kim, Sung, Sung, Ahn and Chang175). Neurologic alterations are also observed in infants with BPD. MSC-derived EVs not only reduced lung damage but also improved neurodevelopmental features in an animal model (Ref. Reference Lithopoulos, Strueby, O’Reilly, Zhong, Möbius, Eaton, Fung, Hurskainen, Cyr-Depauw, Suen, Xu, Collins, Vadivel, Stewart, Burger and Thébaud176).

Chronic obstructive pulmonary disease

Initial studies assessed the effects of MSC-derived conditioned media in COPD models and demonstrated a variety of therapeutic benefits. In CS-induced emphysema, conditioned media promoted tissue repair and increased the number of small pulmonary vessels (Ref. Reference Huh, Kim, Lee, Lee, van Ta, Kim, Oh, Lee and Lee177). Hepatocyte growth factor-mediated protection was observed in lung tissue when MSC-derived conditioned media were administered at the onset of elastase-induced emphysema (Ref. Reference Kennelly, Mahon and English178).

EVs released by damaged alveolar type II cells promoted MSC migration and upregulated genes related to mitochondrial synthesis and transfer (Ref. Reference Song, Peng and Guo179). In co-culture experiments, MSCs released EVs to protect the BEAS2B lung epithelial cell line against CS-induced mitochondrial abnormalities (Ref. Reference Maremanda, Sundar and Rahman180). MSC-derived EVs also reduced CS-induced mitochondrial dysfunction, peribronchial and perivascular inflammation, alveolar septa thickening, and mucus-producing goblet cell counts in rodent lungs (Refs. Reference Maremanda, Sundar and Rahman180Reference Zhu, Lian, Su, Wu, Zeng and Chen182). CS exposure elevated pyroptosis and inhibited phagocytic abilities in alveolar macrophages, which was reversed by MSC-derived EVs (Ref. Reference Zhu, Lian, Su, Wu, Zeng and Chen182). In experimental papain-induced emphysema, MSC-derived EVs prevented endothelial cell apoptosis by activating VEGF/VEGF receptor-2-mediated AKT and MEK/ERK signalling pathways (Ref. Reference Chen, Lin, Deng and Qian183). MSC-derived EVs were also able to improve lung function and mitigate airway inflammation and levels of pro-inflammatory cytokines in CS-exposed mice (Ref. Reference Harrell, Miloradovic, Sadikot, Fellabaum, Markovic, Miloradovic, Acovic, Djonov, Arsenijevic and Volarevic184). Interestingly, cardiorespiratory abnormalities in emphysematous mice were mitigated by MSC-derived EVs from healthy donors but not from elastase-induced emphysema (Ref. Reference Antunes, Braga, Oliveira, Kitoko, Castro, Xisto, Coelho, Rocha, Silva-Aguiar, Caruso-Neves, Martins, Carvalho, Galina, Weiss, Lapa e Silva, Lopes-Pacheco, Cruz and Rocco115). These EVs exhibited downregulation of various anti-inflammatory and anti-oxidant mediators (Ref. Reference Antunes, Braga, Oliveira, Kitoko, Castro, Xisto, Coelho, Rocha, Silva-Aguiar, Caruso-Neves, Martins, Carvalho, Galina, Weiss, Lapa e Silva, Lopes-Pacheco, Cruz and Rocco115). Artificial EVs from MSCs were generated by sequential penetration through polycarbonate membranes and demonstrated similar size, shape, and surface markers as natural MSC-derived EVs. When these EVs were administered in a model of elastase-induced emphysema, lower doses of artificial EVs promoted similar reparative actions as higher doses of natural EVs (Ref. Reference Kim, Kim, Cho, Shin, Lee and Oh185).

Lung cancer

Current research suggests MSCs as a double-edged sword in cancer as they demonstrate pro- and anti-tumorigenic actions (Refs. Reference Liang, Chen, Zhang, Fang, Chen, Xu and Chen186, Reference Slama, Ah-Pine, Khettab, Arcambal, Begue, Dutheil and Gasque187). On the contrary, most studies revealed tumour-suppression effects by MSC-derived EVs, although further research is still needed. MSC EV-overexpressing miR-30b-5p prevented tumorigenesis and promoted NSCLC cell apoptosis by regulating the EZH2/PI3K/AKT pathway (Ref. Reference Wu, Tian, Liu, Xu, Shi, Zhang, Gao, Yin, Xu and Wang188). Both miR-204 and miR-598 have reduced expression in NSCLC tissues, which was associated with poor prognosis (Refs. Reference Li and Wu189, Reference Liu, Zhang, Lin, Tang, Zhou, Wen and Li190). MSC-derived EVs transferred miR-598 into NSCLC cells and prevented proliferation and migration by inhibiting THBS2 production (Ref. Reference Li and Wu189). Inhibition of EMT and NSCLC cell migration and invasion was also evidenced by EV-transferred miR-204 by targeting the KLF7/AKT/HIF-1α pathway (Ref. Reference Liu, Zhang, Lin, Tang, Zhou, Wen and Li190). NSCLC tissues exhibited high expression of CCNE1 and CCNE2 and low expression of miR-144. MSC-derived EVs reduced NSCLC proliferation and the number of S-phase-blocked cells by transferring miR-144 in both in vitro and in vivo models (Ref. Reference Liang, Zhang, Li, Xin, Zhao, Ma and du191). Another therapeutically relevant miRNA in lung cancer is miR-320a which, once transferred by MSC-derived EVs, reduced cancer cell migration and invasion by binding to sex-determining region Y-box 4 (Ref. Reference Xie and Wang192). Let-7i has a reduced expression in lung cells, and its transfer by MSC-derived EVs inhibited lung cell proliferation by downregulating the KDM3A/DCLK1/FXYD3 signalling pathway (Ref. Reference Liu, Feng, Zeng, He, Gong and Liu193).

Despite promising therapeutic actions, some studies demonstrated an opposite effect in which MSC-derived EVs facilitate lung cancer development. In lung adenocarcinoma cells, EV-transferred miR-410 increased their proliferation and reduced apoptosis (Ref. Reference Dong, Pu, Zhang, Qi, Xu, Li, Wei, Wang, Zhou, Zhu, Wang, Liu, Chen and Su194). Under hypoxic conditions, MSC-derived EVs express certain miRNAs (miR-21-5p, miR-193a-3p, miR-210-3p, and miR-5100) that activate STAT3-mediated EMT and accelerate cancer cell invasion (Refs. Reference Zhang, Sai, Wang, Wang, Wang, Zheng, Li, Tang and Xiang195, Reference Ren, Hou, Yang, Wang, Wu, Wu, Zhao and Lu196).

Lung fibrosis

MSC-derived EVs have demonstrated a variety of therapeutic benefits in experimental lung fibrosis. In lung fibroblasts, MSC-derived EVs inhibited TGF-β1-mediated myofibroblast differentiation by a Thy-1-dependent mechanism (Ref. Reference Shentu, Huang, Cernelc-Kohan, Chan, Wong, Espinoza, Tan, Gramaglia, van der Heyde, Chien and Hagood197). Blockage of Thy-1 reduced EV uptake and preserved myofibroblast differentiation (Ref. Reference Shentu, Huang, Cernelc-Kohan, Chan, Wong, Espinoza, Tan, Gramaglia, van der Heyde, Chien and Hagood197). MSC-derived EVs mitigated bleomycin-induced lung fibrosis by stimulating the proliferation of bronchoalveolar stem cells (Ref. Reference Tan, Lau, Leaw, Nguyen, Salamonsen, Saad, Chan, Zhu, Krause, Kim, Sievert, Wallace and Lim198). Proteomic analysis demonstrated that human MSC-derived EVs modulate macrophages to an anti-inflammatory phenotype, thus mitigating lung inflammation and fibrosis in a model of bleomycin-induced lung fibrosis (Ref. Reference Mansouri, Willis, Fernandez-Gonzalez, Reis, Nassiri, Mitsialis and Kourembanas199). In PM2.5-induced lung fibrosis, MSC-derived EVs reduced levels of reactive oxygen species and apoptosis of alveolar epithelial cells (Ref. Reference Gao, Sun, Dong, Zhao, Hu and Jin200). MSC-derived EVs were effective at mitigating macrophage counts, collagen fibre content, and granuloma size in lung parenchyma, thus improving lung function in silica-induced lung fibrosis (Ref. Reference Bandeira, Oliveira, Silva, Menna-Barreto, Takyia, Suk, Witwer, Paulaitis, Hanes, Rocco and Morales201). EVs from three-dimensional cultured MSCs also reduced collagen deposition in silica-exposed fibroblasts (Ref. Reference Xu, Zhao, Li, Hou, Wang, Li, Jiang, Zhu and Tian202).

The influence of MSC EV-transferred miRNAs in preventing or reversing lung fibrosis has been documented in several studies. For instance, miR-29b-3p demonstrated low expression in lung tissue, and its overexpression in MSC-derived EVs prevented interstitial fibroblast proliferation, differentiation, migration, and invasion by targeting frizzled 6 (Ref. Reference Wan, Chen, Fang, Zuo, Cui and Xie203). EV-transferred miR-186 also prevented fibroblast activation by downregulating SRY-related HMG box transcription factor 4 and its downstream gene Dickkopf-1, thus alleviating lung fibrosis in mice (Ref. Reference Zhou, Lin, Kang, Liu, Zhang and Xu204). TGF-β signalling pathway was inhibited by EV-transferred miR-21 and miR-23, preventing myofibroblast differentiation and lung damage in experimental bleomycin-induced lung fibrosis (Ref. Reference Shi, Ren, Li, Wang, Wang, Qin, Li, Zhang, Li and Wang205). EV-transferred let-7 mitigated lung epithelial fibrosis and remodelling in mice by reducing NLRP3-mediated inflammation activation, mitochondrial damage, and levels of reactive oxygen species in alveolar epithelial cells (Ref. Reference Sun, Zhu, Feng, Lin, Yin, Jin and Wang206). Levels of miR-30b were downregulated in the serum of IPF patients, and EV-transferred miR-30b reduced inflammation and apoptosis of TGF-β1-stimulated lung epithelial cell lines by targeting Runx1 and Spred2 (Ref. Reference Zhu, Xu, Wang, Zhang, Zhou and Wu207). EV-transferred miR-29c and miR-129 reduced lung damage and myofibroblast differentiation in experimental bleomycin-induced lung fibrosis (Ref. Reference Nataliya, Mikhail, Vladimir, Olga, Maksim, Ivan, Ekaterina, Georgy, Natalia, Pavel, Andrey, Maria, Maxim, Anastasiya, Uliana, Zhanna, Vsevolod, Natalia and Anastasiya208). In radiation-induced pulmonary fibrosis, EV-transferred miR-214-3p reduced endothelial cell damage, inflammation, and fibrosis by downregulating ataxia telangiectasia mutated/p53/p21-mediated signalling pathway (Ref. Reference Lei, He, Zhu, Zhou, Zhang, Wang, Huang, Chen, Li, Liu, Han, Guo, Han and Li209). EV-transferred miR-218 inhibited endothelial-mesenchymal transition and restored endothelial properties by downregulating the MeCP2/BMP2 pathway (Ref. Reference Zhao, Du and Sun210). Knockdown of miR-218 reduced the therapeutic effects of MSC-derived EVs on endothelial-mesenchymal transition (Ref. Reference Zhao, Du and Sun210).

In silica-induced lung fibrosis, MSCs release EV-containing miRNAs that inhibit macrophage activation by suppressing TLR signalling (Ref. Reference Phinney, Di Giuseppe and Njah211). EV-transferred miR-223-3p reduced silica-induced inflammation (IL-1β, IL-18, and cleaved caspase-1) and remodelling (collagen I and III, fibronectin, and α-smooth muscle actin) by suppressing NLRP3-mediated signalling pathway (Ref. Reference Hou, Zhu, Jiang, Zhao, Jia, Jiang, Wang, Xue, Wang and Tian147). In experimental silicosis, EV-containing let-7i-5p prevented fibroblast activation by targeting the TGF-β receptor/Smad3 pathway (Ref. Reference Xu, Hou, Zhao, Wang, Jiang, Jiang, Zhu and Tian212), while EV-transferred let-7d-5p reduced PM2.5-induced lung fibrosis (Ref. Reference Gao, Sun, Dong, Zhao, Hu and Jin200). Likewise, miR-26a-5p is downregulated in silicosis, and its transfer by MSC-derived EVs prevented EMT by targeting Adam17/Notch signalling, thus mitigating lung fibrosis in mice (Ref. Reference Zhao, Jiang, Xu, Jia, Wang, Xue, Wang, Zhu and Tian213).

Pulmonary arterial hypertension

In early studies, MSC-derived conditioned media demonstrated protective effects against PAH by preventing cardiac remodelling and improving pulmonary blood flow (Ref. Reference Rathinasabapathy, Bruce, Espejo, Horowitz, Sudhan, Nair, Guzzo, Francis, Raizada, Shenoy and Katovich214) and by downregulating calcineurin and nuclear factor of activated T-cells, thus inhibiting the inflammation-mediated over-proliferation of pulmonary artery smooth muscle cells (Ref. Reference Liu, Han and Han215). Subsequently, MSC-derived EVs reduced right ventricular hypertrophy and small pulmonary artery area index in experimental sugen/hypoxia- (Ref. Reference Klinger, Pereira and Del Tatto216) and monocrotaline-induced PAH (Ref. Reference Chen, An, Liu, Wang, Chen, Hong, Liu, Xiao and Chen217). MSC-derived EVs promoted therapeutic effects on experimental monocrotaline-induced PAH by transferring miR-34a, miR-122, miR-124, and miR-127 (Ref. Reference Aliotta, Pereira, Wen, Dooner, del Tatto, Papa, Goldberg, Baird, Ventetuolo, Quesenberry and Klinger218) and by shifting the balance from the ACE/AngII/AT1R axis toward ACE2/Ang(1–7)/Mas axis (Ref. Reference Liu, Liu, Xiao, Wang, Yao, Zeng, Yu, Guan, Wei, Peng, Zhu, Wang, Yang, Zhong and Chen219). MSC-derived EVs also reduced monocrotaline-induced lung fibrosis, right ventricular hypertrophy, and pulmonary vascular remodelling by regulating the WNT5A/BMP signalling pathway (Ref. Reference Zhang, Ge, Zhang, Wang, Jiang, Xin and Luan220). In experimental hypoxia-induced PAH, macrophages were polarized to M2 phenotype by MSC-derived EVs, leading to reduced levels of inflammatory mediators and increased levels of IL-10, inhibiting over-proliferation of pulmonary artery smooth muscle cells (Ref. Reference Liu, Zhang, Liu, Zhang, Wang, Huang, Ma and Ge221).

Concluding remarks

Significant progress has been made in translational medicine for respiratory diseases; however, more precise biomarkers and effective therapies remain an urgent need. EVs have emerged as essential mediators of intercellular communication by interacting and transferring their content to recipient cells, thus regulating various biological pathways. Their ability to modulate resident and immune cell responses, inflammation, and tissue repair – particularly in respiratory diseases – highlights their multimodal potential. By enabling the identification of unique signatures that may distinguish pathological conditions and severity with high accuracy, EVs may hold an optimal role for diagnosis. In addition, MSC-derived EVs carry anti-inflammatory and regenerative factors that can influence cellular signalling processing, offering therapeutic avenues for lung inflammatory and fibrotic conditions. Lastly, EVs possess intrinsic properties that make them optimal candidates for gene/drug delivery by combining natural biocompatibility with the ability to target specific cells and tissues.

Despite encouraging pre-clinical findings, several challenges should be addressed before EVs become a tool in the clinical scenario. A major barrier is the need for high-quality, standardized methods for EV isolation and characterization. Current techniques frequently yield heterogeneous populations of EVs, which can hamper reproducibility and clinical development. Improved protocols for EV purification associated with rigorous characterization will be fundamental to ensuring consistency in therapeutic development. Moreover, the EV research field needs to adopt more transparent and stringent reporting practices in scientific publications to facilitate cross-study comparisons.

Although current evidence strongly supports the role of EVs in lung tissue repair and inflammation modulation, large-scale clinical studies are needed to establish standardized dosing, delivery methods, and long-term safety and efficacy. In parallel, interdisciplinary collaboration will be crucial in overcoming existing technical and regulatory barriers and in establishing guidelines for EV-based products to streamline their approval process.

In conclusion, EVs represent a transformative tool in biomedicine, with vast potential for both diagnostics and therapeutics. However, their successful translation into patient care will depend on overcoming current limitations in isolation techniques, biomarker specificity, and clinical validation. By addressing these challenges through rigorous research and collaborative efforts, the EV field can pave the way for novel, effective treatments for respiratory diseases and beyond. The future of EV-based medicine is promising, but its realization hinges on continued innovation, standardization, and evidence-based development.

Author contribution

Miquéias Lopes-Pacheco confirms being the sole contributor to this study and has approved it for publication.

Competing interests

The author declare none.

References

Momtazmanesh, S, Moghaddam, SS, Ghamari, S-H, Rad, EM, Rezaei, N, Shobeiri, P, Aali, A, Abbasi-Kangevari, M, Abbasi-Kangevari, Z, Abdelmasseh, M, Abdoun, M, Abdulah, DM, Md Abdullah, AY, Abedi, A, Abolhassani, H, Abrehdari-Tafreshi, Z, Achappa, B, Adane Adane, DE, Adane, TD, Addo, IY, Adnan, M, Sakilah Adnani, QE, Ahmad, S, Ahmadi, A, Ahmadi, K, Ahmed, A, Ahmed, A, Rashid, TA, al Hamad, H, Alahdab, F, Alemayehu, A, Alif, SM, Aljunid, SM, Almustanyir, S, Altirkawi, KA, Alvis-Guzman, N, Dehkordi, JA, Amir-Behghadami, M, Ancuceanu, R, Andrei, CL, Andrei, T, Antony, CM, Anyasodor, AE, Arabloo, J, Arulappan, J, Ashraf, T, Athari, SS, Attia, EF, Ayele, MT, Azadnajafabad, S, Babu, AS, Bagherieh, S, Baltatu, OC, Banach, M, Bardhan, M, Barone-Adesi, F, Barrow, A, Basu, S, Bayileyegn, NS, Bensenor, IM, Bhardwaj, N, Bhardwaj, P, Bhat, AN, Bhattacharyya, K, Bouaoud, S, Braithwaite, D, Brauer, M, Butt, MH, Butt, ZA, Calina, D, Cámera, LA, Chanie, GS, Charalampous, P, Chattu, VK, Chimed-Ochir, O, Chu, DT, Cohen, AJ, Cruz-Martins, N, Dadras, O, Darwesh, AM, das, S, Debela, SA, Delgado-Ortiz, L, Dereje, D, Dianatinasab, M, Diao, N, Diaz, D, Digesa, LE, Dirirsa, G, Doku, PN, Dongarwar, D, Douiri, A, Dsouza, HL, Eini, E, Ekholuenetale, M, Ekundayo, TC, Mustafa Elagali, AE, Elhadi, M, Enyew, DB, Erkhembayar, R, Etaee, F, Fagbamigbe, AF, Faro, A, Fatehizadeh, A, Fekadu, G, Filip, I, Fischer, F, Foroutan, M, Franklin, RC, Gaal, PA, Gaihre, S, Gaipov, A, Gebrehiwot, M, Gerema, U, Getachew, ME, Getachew, T, Ghafourifard, M, Ghanbari, R, Ghashghaee, A, Gholami, A, Gil, AU, Golechha, M, Goleij, P, Golinelli, D, Guadie, HA, Gupta, B, Gupta, S, Gupta, VB, Gupta, VK, Hadei, M, Halwani, R, Hanif, A, Hargono, A, Harorani, M, Hartono, RK, Hasani, H, Hashi, A, Hay, SI, Heidari, M, Hellemons, ME, Herteliu, C, Holla, R, Horita, N, Hoseini, M, Hosseinzadeh, M, Huang, J, Hussain, S, Hwang, BF, Iavicoli, I, Ibitoye, SE, Ibrahim, S, Ilesanmi, OS, Ilic, IM, Ilic, MD, Immurana, M, Ismail, NE, Merin J, L, Jakovljevic, M, Jamshidi, E, Janodia, MD, Javaheri, T, Jayapal, SK, Jayaram, S, Jha, RP, Johnson, O, Joo, T, Joseph, N, Jozwiak, JJ, K, V, Kaambwa, B, Kabir, Z, Kalankesh, LR, Kalhor, R, Kandel, H, Karanth, SD, Karaye, IM, Kassa, BG, Kassie, GM, Keikavoosi-Arani, L, Keykhaei, M, Khajuria, H, Khan, IA, Khan, MAB, Khan, YH, Khreis, H, Kim, MS, Kisa, A, Kisa, S, Knibbs, LD, Kolkhir, P, Komaki, S, Kompani, F, Koohestani, HR, Koolivand, A, Korzh, O, Koyanagi, A, Krishan, K, Krohn, KJ, Kumar, N, Kumar, N, Kurmi, OP, Kuttikkattu, A, la Vecchia, C, Lám, J, Lan, Q, Lasrado, S, Latief, K, Lauriola, P, Lee, SW, Lee, YH, Legesse, SM, Lenzi, J, Li, MC, Lin, RT, Liu, G, Liu, W, Lo, CH, Lorenzovici, L, Lu, Y, Mahalingam, S, Mahmoudi, E, Mahotra, NB, Malekpour, MR, Malik, AA, Mallhi, TH, Malta, DC, Mansouri, B, Mathews, E, Maulud, SQ, Mechili, EA, Nasab, EM, Menezes, RG, Mengistu, DA, Mentis, AF, Meshkat, M, Mestrovic, T, Micheletti Gomide Nogueira de Sá, AC, Mirrakhimov, EM, Misganaw, A, Mithra, P, Moghadasi, J, Mohammadi, E, Mohammadi, M, Mohammadshahi, M, Mohammed, S, Mohan, S, Moka, N, Monasta, L, Moni, MA, Moniruzzaman, M, Montazeri, F, Moradi, M, Mostafavi, E, Mpundu-Kaambwa, C, Murillo-Zamora, E, Murray, CJL, Nair, TS, Nangia, V, Swamy, SN, Narayana, AI, Natto, ZS, Nayak, BP, Negash, WW, Nena, E, Kandel, SN, Niazi, RK, Nogueira de Sá, AT, Nowroozi, A, Nzoputam, CI, Nzoputam, OJ, Oancea, B, Obaidur, RM, Odukoya, OO, Okati-Aliabad, H, Okekunle, AP, Okonji, OC, Olagunju, AT, Bali, AO, Ostojic, SM, A, MP, Padron-Monedero, A, Padubidri, JR, Pahlevan Fallahy, MT, Palicz, T, Pana, A, Park, EK, Patel, J, Paudel, R, Paudel, U, Pedersini, P, Pereira, M, Pereira, RB, Petcu, IR, Pirestani, M, Postma, MJ, Prashant, A, Rabiee, M, Radfar, A, Rafiei, S, Rahim, F, Ur Rahman, MH, Rahman, M, Rahman, MA, Rahmani, AM, Rahmani, S, Rahmanian, V, Rajput, P, Rana, J, Rao, CR, Rao, SJ, Rashedi, S, Rashidi, MM, Ratan, ZA, Rawaf, DL, Rawaf, S, Rawal, L, Rawassizadeh, R, Razeghinia, MS, Mohamed Redwan, EM, Rezaei, M, Rezaei, N, Rezaei, N, Rezaeian, M, Rodrigues, M, Buendia Rodriguez, JA, Roever, L, Rojas-Rueda, D, Rudd, KE, Saad, AMA, Sabour, S, Saddik, B, Sadeghi, E, Sadeghi, M, Saeed, U, Sahebazzamani, M, Sahebkar, A, Sahoo, H, Sajid, MR, Sakhamuri, S, Salehi, S, Samy, AM, Santric-Milicevic, MM, Sao Jose, BP, Sathian, B, Satpathy, M, Saya, GK, Senthilkumaran, S, Seylani, A, Shahabi, S, Shaikh, MA, Shanawaz, M, Shannawaz, M, Sheikhi, RA, Shekhar, S, Sibhat, MM, Simpson, CR, Singh, JA, Singh, P, Singh, S, Skryabin, VY, Skryabina, AA, Soltani-Zangbar, MS, Song, S, Soyiri, IN, Steiropoulos, P, Stockfelt, L, Sun, J, Takahashi, K, Talaat, IM, Tan, KK, Tat, NY, Tat, VY, Taye, BT, Thangaraju, P, Thapar, R, Thienemann, F, Tiyuri, A, Ngoc Tran, MT, Tripathy, JP, Car, LT, Tusa, BS, Ullah, I, Ullah, S, Vacante, M, Valdez, PR, Valizadeh, R, van Boven, JFM, Vasankari, TJ, Vaziri, S, Violante, FS, Vo, B, Wang, N, Wei, MY, Westerman, R, Wickramasinghe, ND, Xu, S, Xu, X, Yadav, L, Yismaw, Y, Yon, DK, Yonemoto, N, Yu, C, Yu, Y, Yunusa, I, Zahir, M, Zangiabadian, M, Zareshahrabadi, Z, Zarrintan, A, Zastrozhin, MS, Zegeye, ZB, Zhang, Y, Naghavi, M, Larijani, B and Farzadfar, F (2023) Global burden of chronic respiratory diseases and risk factors, 1990–2019: An update from the global burden of disease study 2019. eClinicalMedicine. 59, 101936.Google Scholar
Bhatt, JM (2013) Treatment of pulmonary exacerbations in cystic fibrosis. European Respiratory Review 22, 205216.Google Scholar
Ryerson, CJ, Cottin, V, Brown, KK and Collard, HR (2015) Acute exacerbation of idiopathic pulmonary fibrosis: Shifting the paradigm. The European Respiratory Journal 46, 512520.Google Scholar
Ritchie, AI and Wedzicha, JA (2020) Definition, causes, pathogenesis, and consequences of chronic obstructive pulmonary disease exacerbations. Clinics in Chest Medicine 41, 421438.Google Scholar
Lötvall, J, Hill, AF, Hochberg, F, Buzás, EI, di Vizio, D, Gardiner, C, Gho, YS, Kurochkin, IV, Mathivanan, S, Quesenberry, P, Sahoo, S, Tahara, H, Wauben, MH, Witwer, KW and Théry, C (2014) Minimal experimental requirements for definition of extracellular vesicles and their functions: A position statement from the International Society for Extracellular Vesicles. Journal of Extracellular Vesicles 3, 26913.Google Scholar
Théry, C, Witwer, KW, Aikawa, E, Alcaraz, MJ, Anderson, JD, Andriantsitohaina, R, Antoniou, A, Arab, T, Archer, F, Atkin-Smith, GK, Ayre, DC, Bach, JM, Bachurski, D, Baharvand, H, Balaj, L, Baldacchino, S, Bauer, NN, Baxter, AA, Bebawy, M, Beckham, C, Bedina Zavec, A, Benmoussa, A, Berardi, AC, Bergese, P, Bielska, E, Blenkiron, C, Bobis-Wozowicz, S, Boilard, E, Boireau, W, Bongiovanni, A, Borràs, FE, Bosch, S, Boulanger, CM, Breakefield, X, Breglio, AM, Brennan, , Brigstock, DR, Brisson, A, Broekman, MLD, Bromberg, JF, Bryl-Górecka, P, Buch, S, Buck, AH, Burger, D, Busatto, S, Buschmann, D, Bussolati, B, Buzás, EI, Byrd, JB, Camussi, G, Carter, DRF, Caruso, S, Chamley, LW, Chang, YT, Chen, C, Chen, S, Cheng, L, Chin, AR, Clayton, A, Clerici, SP, Cocks, A, Cocucci, E, Coffey, RJ, Cordeiro-da-Silva, A, Couch, Y, Coumans, FAW, Coyle, B, Crescitelli, R, Criado, MF, D’Souza-Schorey, C, das, S, Datta Chaudhuri, A, de Candia, P, de Santana, EF Jr, de Wever, O, del Portillo, HA, Demaret, T, Deville, S, Devitt, A, Dhondt, B, di Vizio, D, Dieterich, LC, Dolo, V, Dominguez Rubio, AP, Dominici, M, Dourado, MR, Driedonks, TAP, Duarte, FV, Duncan, HM, Eichenberger, RM, Ekström, K, el Andaloussi, S, Elie-Caille, C, Erdbrügger, U, Falcón-Pérez, JM, Fatima, F, Fish, JE, Flores-Bellver, M, Försönits, A, Frelet-Barrand, A, Fricke, F, Fuhrmann, G, Gabrielsson, S, Gámez-Valero, A, Gardiner, C, Gärtner, K, Gaudin, R, Gho, YS, Giebel, B, Gilbert, C, Gimona, M, Giusti, I, Goberdhan, DCI, Görgens, A, Gorski, SM, Greening, DW, Gross, JC, Gualerzi, A, Gupta, GN, Gustafson, D, Handberg, A, Haraszti, RA, Harrison, P, Hegyesi, H, Hendrix, A, Hill, AF, Hochberg, FH, Hoffmann, KF, Holder, B, Holthofer, H, Hosseinkhani, B, Hu, G, Huang, Y, Huber, V, Hunt, S, Ibrahim, AGE, Ikezu, T, Inal, JM, Isin, M, Ivanova, A, Jackson, HK, Jacobsen, S, Jay, SM, Jayachandran, M, Jenster, G, Jiang, L, Johnson, SM, Jones, JC, Jong, A, Jovanovic-Talisman, T, Jung, S, Kalluri, R, Kano, SI, Kaur, S, Kawamura, Y, Keller, ET, Khamari, D, Khomyakova, E, Khvorova, A, Kierulf, P, Kim, KP, Kislinger, T, Klingeborn, M, Klinke, DJ II, Kornek, M, Kosanović, MM, Kovács, ÁF, Krämer-Albers, EM, Krasemann, S, Krause, M, Kurochkin, IV, Kusuma, GD, Kuypers, S, Laitinen, S, Langevin, SM, Languino, LR, Lannigan, J, Lässer, C, Laurent, LC, Lavieu, G, Lázaro-Ibáñez, E, le Lay, S, Lee, MS, Lee, YXF, Lemos, DS, Lenassi, M, Leszczynska, A, Li, ITS, Liao, K, Libregts, SF, Ligeti, E, Lim, R, Lim, SK, Linē, A, Linnemannstöns, K, Llorente, A, Lombard, CA, Lorenowicz, MJ, Lörincz, ÁM, Lötvall, J, Lovett, J, Lowry, MC, Loyer, X, Lu, Q, Lukomska, B, Lunavat, TR, Maas, SLN, Malhi, H, Marcilla, A, Mariani, J, Mariscal, J, Martens-Uzunova, ES, Martin-Jaular, L, Martinez, MC, Martins, VR, Mathieu, M, Mathivanan, S, Maugeri, M, McGinnis, LK, McVey, MJ, Meckes, DG Jr, Meehan, KL, Mertens, I, Minciacchi, VR, Möller, A, Møller Jørgensen, M, Morales-Kastresana, A, Morhayim, J, Mullier, F, Muraca, M, Musante, L, Mussack, V, Muth, DC, Myburgh, KH, Najrana, T, Nawaz, M, Nazarenko, I, Nejsum, P, Neri, C, Neri, T, Nieuwland, R, Nimrichter, L, Nolan, JP, Nolte-’t Hoen, ENM, Noren Hooten, N, O’Driscoll, L, O’Grady, T, O’Loghlen, A, Ochiya, T, Olivier, M, Ortiz, A, Ortiz, LA, Osteikoetxea, X, Østergaard, O, Ostrowski, M, Park, J, Pegtel, DM, Peinado, H, Perut, F, Pfaffl, MW, Phinney, DG, Pieters, BCH, Pink, RC, Pisetsky, DS, Pogge von Strandmann, E, Polakovicova, I, Poon, IKH, Powell, BH, Prada, I, Pulliam, L, Quesenberry, P, Radeghieri, A, Raffai, RL, Raimondo, S, Rak, J, Ramirez, MI, Raposo, G, Rayyan, MS, Regev-Rudzki, N, Ricklefs, FL, Robbins, PD, Roberts, DD, Rodrigues, SC, Rohde, E, Rome, S, Rouschop, KMA, Rughetti, A, Russell, AE, Saá, P, Sahoo, S, Salas-Huenuleo, E, Sánchez, C, Saugstad, JA, Saul, MJ, Schiffelers, RM, Schneider, R, Schøyen, TH, Scott, A, Shahaj, E, Sharma, S, Shatnyeva, O, Shekari, F, Shelke, GV, Shetty, AK, Shiba, K, Siljander, PRM, Silva, AM, Skowronek, A, Snyder, OL II, Soares, RP, Sódar, BW, Soekmadji, C, Sotillo, J, Stahl, PD, Stoorvogel, W, Stott, SL, Strasser, EF, Swift, S, Tahara, H, Tewari, M, Timms, K, Tiwari, S, Tixeira, R, Tkach, M, Toh, WS, Tomasini, R, Torrecilhas, AC, Tosar, JP, Toxavidis, V, Urbanelli, L, Vader, P, van Balkom, BWM, van der Grein, SG, van Deun, J, van Herwijnen, MJC, van Keuren-Jensen, K, van Niel, G, van Royen, ME, van Wijnen, AJ, Vasconcelos, MH, Vechetti, IJ Jr, Veit, TD, Vella, LJ, Velot, É, Verweij, FJ, Vestad, B, Viñas, JL, Visnovitz, T, Vukman, KV, Wahlgren, J, Watson, DC, Wauben, MHM, Weaver, A, Webber, JP, Weber, V, Wehman, AM, Weiss, DJ, Welsh, JA, Wendt, S, Wheelock, AM, Wiener, Z, Witte, L, Wolfram, J, Xagorari, A, Xander, P, Xu, J, Yan, X, Yáñez-Mó, M, Yin, H, Yuana, Y, Zappulli, V, Zarubova, J, Žėkas, V, Zhang, JY, Zhao, Z, Zheng, L, Zheutlin, AR, Zickler, AM, Zimmermann, P, Zivkovic, AM, Zocco, D and Zuba-Surma, EK (2018) Minimal information for studies of extracellular vesicles 2018 (MISEV2018): A position statement of the International Society for Extracellular Vesicles and update of the MISEV2014 guidelines. Journal of Extracellular Vesicles 7, 1535750.Google Scholar
Welsh, JA, Goberdhan, DCI, O’Driscoll, L, et al. (2024) Minimal information for studies of extracellular vesicles (MISEV2023): From basic to advanced approaches. Journal of Extracellular Vesicles 13, e12404.Google Scholar
Alberro, A, Iparraguirre, L, Fernandes, A and Otaegui, D (2021) Extracellular vesicles in blood: Sources, effects, and applications. International Journal of Molecular Sciences 22, 8163.Google Scholar
Turchinovich, A, Weiz, L and Burwinkel, B (2012) Extracellular miRNAs: The mystery of their origin and function. Trends in Biochemical Sciences 37, 460465.Google Scholar
dos Santos, CC, Lopes-Pacheco, M, English, K, Rolandsson Enes, S, Krasnodembskaya, A and Rocco, PRM (2024) The MSC-EV-microRNAome: A perspective on therapeutic mechanisms of action in sepsis and ARDS. Cells 13, 122.Google Scholar
Zhang, Y, Liu, Y, Liu, H and Tang, WH (2019) Exosomes: Biogenesis, biologic function and clinical potential. Cell & Bioscience 9, 19.Google Scholar
Abreu, SC, Lopes-Pacheco, M, Weiss, DJ and Rocco, PRM (2021) Mesenchymal stromal cell-derived extracellular vesicles in lung diseases: Current status and perspectives. Frontiers in Cell and Development Biology 9, 600711.Google Scholar
Pinheiro, AAS, Torrecilhas, AC, de Freitas Souza, BS, et al. (2024) Potential of extracellular vesicles in the pathogenesis, diagnosis and therapy for parasitic diseases. Journal of Extracellular Vesicles 13, e12496.Google Scholar
Holtzman, J and Lee, H (2020) Emerging role of extracellular vesicles in the respiratory system. Experimental & Molecular Medicine 52, 887895.Google Scholar
Mulcahy, LA, Pink, RC and Carter, DRF (2014 ) Routes and mechanisms of extracellular vesicle uptake. Journal of Extracellular Vesicles 4, 3.Google Scholar
Chargaff, E and West, R (1946) The biological significance of the thromboplastic protein of blood. The Journal of Biological Chemistry 166, 189197.Google Scholar
Wolf, P (1967) The nature and significance of platelet products in human plasma. British Journal of Haematology 13, 269288.Google Scholar
Harding, C, Heuser, J and Stahl, P (1983) Receptor-mediated endocytosis of transferrin and recycling of the transferrin receptor in rat reticulocytes. The Journal of Cell Biology 97, 329339.Google Scholar
Pan, BT, Teng, K, Wu, C, Adam, M and Johnstone, RM (1985) Electron microscopic evidence for externalization of the transferrin receptor in vesicular form in sheep reticulocytes. The Journal of Cell Biology 101, 942948.Google Scholar
Kadota, T, Fujita, Y, Araya, J, Ochiya, T and Kuwano, K (2022) Extracellular vesicle-mediated cellular crosstalk in lung repair, remodelling and regeneration. European Respiratory Review 31, 210106.Google Scholar
Schneider, JL, Rowe, JH, Garcia-de-Alba, C, Kim, CF, Sharpe, AH and Haigis, MC (2021) The aging lung: Physiology, disease, and immunity. Cell 184, 19902019.Google Scholar
Bartel, S, Deshane, J, Wilkinson, T and Gabrielsson, S (2020) Extracellular vesicles as mediators of cellular cross talk in the lung microenvironment. Frontiers in Medicine 7, 326.Google Scholar
Kesimer, M, Scull, M, Brighton, B, DeMaria, G, Burns, K, O’Neal, W, Pickles, RJ and Sheehan, JK (2009) Characterization of exosome-like vesicles released from human tracheobronchial ciliated epithelium: A possible role in innate defense. The FASEB Journal 23, 18581868.Google Scholar
Gupta, R, Radicioni, G, Abdelwahab, S, Dang, H, Carpenter, J, Chua, M, Mieczkowski, PA, Sheridan, JT, Randell, SH and Kesimer, M (2019) Intercellular communication between airway epithelial cells is mediated by exosome-like vesicles. American Journal of Respiratory Cell and Molecular Biology 60, 209220.Google Scholar
Wahlund, CJE, Eklund, A, Grunewald, J and Gabrielsson, S (2017) Pulmonary extracellular vesicles as mediators of local and systemic inflammation. Frontiers in Cell and Development Biology 5, 39.Google Scholar
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 and Coppinger, JA (2020) Increased extracellular vesicles mediate inflammatory signalling in cystic fibrosis. Thorax 75, 449458.Google Scholar
Liu, Y, Defourny, KAY, Smid, EJ and Abee, T (2018) Gram-positive bacterial extracellular vesicles and their impact on health and disease. Frontiers in Microbiology 9, 1502.Google Scholar
Ismail, N, Wang, Y, Dakhlallah, D, Moldovan, L, Agarwal, K, Batte, K, Shah, P, Wisler, J, Eubank, TD, Tridandapani, S, Paulaitis, ME, Piper, MG and Marsh, CB (2013) Macrophage microvesicles induce macrophage differentiation and miR-223 transfer. Blood 121, 984995.Google Scholar
Bourdonnay, E, Zasłona, Z, Penke, LRK, Speth, JM, Schneider, DJ, Przybranowski, S, Swanson, JA, Mancuso, P, Freeman, CM, Curtis, JL and Peters-Golden, M (2015) Transcellular delivery of vesicular SOCS proteins from macrophages to epithelial cells blunts inflammatory signaling. The Journal of Experimental Medicine 212, 729742.Google Scholar
Njock, M-S, Cheng, HS, Dang, LT, Nazari-Jahantigh, M, Lau, AC, Boudreau, E, Roufaiel, M, Cybulsky, MI, Schober, A and Fish, JE (2015) Endothelial cells suppress monocyte activation through secretion of extracellular vesicles containing antiinflammatory microRNAs. Blood 125, 32023212.Google Scholar
Zhao, L, Luo, H, Li, X, Li, T, He, J, Qi, Q, Liu, Y and Yu, Z (2017) Exosomes derived from human pulmonary artery endothelial cells shift the balance between proliferation and apoptosis of smooth muscle cells. Cardiology 137, 4353.Google Scholar
Kadota, T, Fujita, Y, Yoshioka, Y, Araya, J, Kuwano, K and Ochiya, T (2016) Extracellular vesicles in chronic obstructive pulmonary disease. International Journal of Molecular Sciences 17, 1801.Google Scholar
Dhar, R, Mukherjee, S, Mukerjee, N, Mukherjee, D, Devi, A, Ashraf, GM, Alserihi, RF, Tayeb, HH, Hashem, AM, Alexiou, A and Thorate, N (2022) Interrelation between extracellular vesicles miRNAs with chronic lung diseases. Journal of Cellular Physiology 237, 40214036.Google Scholar
Yin, Y, Shelke, GV, Lässer, C, Brismar, H and Lötvall, J (2020) Extracellular vesicles from mast cells induce mesenchymal transition in airway epithelial cells. Respiratory Research 21, 101.Google Scholar
Lacy, SH, Woeller, CF, Thatcher, TH, Pollock, SJ, Small, EM, Sime, PJ and Phipps, RP (2019) Activated human lung fibroblasts produce extracellular vesicles with Antifibrotic prostaglandins. American Journal of Respiratory Cell and Molecular Biology 60, 269278.Google Scholar
Zhou, B, Xu, K, Zheng, X, Chen, T, Wang, J, Song, Y, Shao, Y and Zheng, S (2020) Application of exosomes as liquid biopsy in clinical diagnosis. Signal Transduction and Targeted Therapy 5, 144.Google Scholar
Grueso-Navarro, E, Navarro, P, Laserna-Mendieta, EJ, Lucendo, AJ and Arias-González, L (2023) Blood-based biomarkers for Eosinophilic esophagitis and concomitant atopic diseases: A look into the potential of extracellular vesicles. International Journal of Molecular Sciences 24, 3669.Google Scholar
Carnino, JM, Lee, H and Jin, Y (2019) Isolation and characterization of extracellular vesicles from Broncho-alveolar lavage fluid: A review and comparison of different methods. Respiratory Research 20, 240.Google Scholar
Paes Leme, AF, Yokoo, S, Normando, AGC, Ormonde, JVS, Domingues, RR, Cruz, FF, Silva, PL, Souza, BSF, dos Santos, CC, Castro-Faria-Neto, H, Martins, CM, Lopes-Pacheco, M and Rocco, PRM (2024) Proteomics of serum-derived extracellular vesicles are associated with the severity and different clinical profiles of patients with COVID-19: An exploratory secondary analysis. Cytotherapy 26, 444455.Google Scholar
Bacalhau, M, Camargo, M and Lopes-Pacheco, M (2024) Laboratory tools to predict CFTR modulator therapy effectiveness and to monitor disease severity in cystic fibrosis. Journal of Personalized Medicine 14, 93.Google Scholar
Duarte, D, Taveira-Gomes, T, Sokhatska, O, Palmares, C, Costa, R, Negrão, R, Guimarães, JT, Delgado, L, Soares, R and Moreira, A (2013) Increased circulating platelet microparticles as a potential biomarker in asthma. Allergy 68, 10731075.Google Scholar
Rollet-Cohen, V, Bourderioux, M, Lipecka, J, Chhuon, C, Jung, VA, Mesbahi, M, Nguyen-Khoa, T, Guérin-Pfyffer, S, Schmitt, A, Edelman, A, Sermet-Gaudelus, I and Guerrera, IC (2018) Comparative proteomics of respiratory exosomes in cystic fibrosis, primary ciliary dyskinesia and asthma. Journal of Proteomics 185, 17.Google Scholar
Takahashi, T, Kobayashi, S, Fujino, N, Suzuki, T, Ota, C, He, M, Yamada, M, Suzuki, S, Yanai, M, Kurosawa, S, Yamaya, M and Kubo, H (2012) Increased circulating endothelial microparticles in COPD patients: A potential biomarker for COPD exacerbation susceptibility. Thorax 67, 10671074.Google Scholar
Samra, MS, Lim, DH, Han, MY, Jee, HM, Kim, YK and Kim, JH (2021) Bacterial microbiota-derived extracellular vesicles in children with allergic airway diseases: Compositional and functional features. Allergy, Asthma & Immunology Research 13, 5674.Google Scholar
Ciccosanti, F, Antonioli, M, Sacchi, A, Notari, S, Farina, A, Beccacece, A, Fusto, M, Vergori, A, D’Offizi, G, Taglietti, F, Antinori, A, Nicastri, E, Marchioni, L, Palmieri, F, Ippolito, G, Piacentini, M, Agrati, C and Fimia, GM (2022) Proteomic analysis identifies a signature of disease severity in the plasma of COVID-19 pneumonia patients associated to neutrophil, platelet and complement activation. Clinical Proteomics 19, 38.Google Scholar
Holgate, ST, Wenzel, S, Postma, DS, Weiss, ST, Renz, H and Sly, PD (2015) Asthma. Nature Reviews Disease Primers 1, 15025.Google Scholar
Song, P, Adeloye, D, Salim, H, dos Santos, JPR, Campbell, H, Sheikh, A and Rudan, I (2022) Global, regional, and national prevalence of asthma in 2019: A systematic analysis and modelling study. Journal of Global Health 12, 04052.Google Scholar
Bartel, S, La Grutta, S, Cilluffo, G, et al. (2020) Human airway epithelial extracellular vesicle miRNA signature is altered upon asthma development. Allergy 75, 346356.Google Scholar
Mwase, C, Phung, T-KN, O’Sullivan, MJ, Mitchel, JA, de Marzio, M, Kılıç, A, Weiss, ST, Fredberg, JJ and Park, JA (2022) Mechanical compression of human airway epithelial cells induces release of extracellular vesicles containing Tenascin C. Cells 11, 256.Google Scholar
Kanannejad, Z, Arab, S, Soleimanian, S, Mazare, A and Kheshtchin, N (2024) Exosomes in asthma: Underappreciated contributors to the pathogenesis and novel therapeutic tools. Immunity, Inflammation and Disease 12, e1325.Google Scholar
Gon, Y, Maruoka, S, Inoue, T, Kuroda, K, Yamagishi, K, Kozu, Y, Shikano, S, Soda, K, Lötvall, J and Hashimoto, S (2017) Selective release of miRNAs via extracellular vesicles is associated with house-dust mite allergen-induced airway inflammation. Clinical and Experimental Allergy 47, 15861598.Google Scholar
Francisco-Garcia, AS, Garrido-Martín, EM, Rupani, H, Lau, LCK, Martinez-Nunez, RT, Howarth, PH and Sanchez-Elsner, T (2019) Small RNA species and microRNA profiles are altered in severe asthma Nanovesicles from Broncho alveolar lavage and associate with impaired lung function and inflammation. Non-coding RNA. 5, 51.Google Scholar
Torregrosa Paredes, P, Esser, J, Admyre, C, Nord, M, Rahman, QK, Lukic, A, Rådmark, O, Grönneberg, R, Grunewald, J, Eklund, A, Scheynius, A and Gabrielsson, S (2012) Bronchoalveolar lavage fluid exosomes contribute to cytokine and leukotriene production in allergic asthma. Allergy 67, 911919.Google Scholar
Lässer, C, O’Neil, SE, Shelke, GV, Sihlbom, C, Hansson, SF, Gho, YS, Lundbäck, B and Lötvall, J (2016) Exosomes in the nose induce immune cell trafficking and harbour an altered protein cargo in chronic airway inflammation. Journal of Translational Medicine 14, 181.Google Scholar
Ax, E, Jevnikar, Z, Cvjetkovic, A, Malmhäll, C, Olsson, H, Rådinger, M and Lässer, C (2020) T2 and T17 cytokines alter the cargo and function of airway epithelium-derived extracellular vesicles. Respiratory Research 21, 155.Google Scholar
Cañas, JA, Sastre, B, Mazzeo, C, Fernández-Nieto, M, Rodrigo-Muñoz, JM, González-Guerra, A, Izquierdo, M, Barranco, P, Quirce, S, Sastre, J and del Pozo, V (2017) Exosomes from eosinophils autoregulate and promote eosinophil functions. Journal of Leukocyte Biology 101, 11911199.Google Scholar
Cañas, JA, Sastre, B, Rodrigo-Muñoz, JM, Fernández-Nieto, M, Barranco, P, Quirce, S, Sastre, J and del Pozo, V (2018) Eosinophil-derived exosomes contribute to asthma remodelling by activating structural lung cells. Clinical and Experimental Allergy 48, 11731185.Google Scholar
Kulshreshtha, A, Ahmad, T, Agrawal, A and Ghosh, B (2013) Proinflammatory role of epithelial cell-derived exosomes in allergic airway inflammation. The Journal of Allergy and Clinical Immunology 131, 11941203, e1–14.Google Scholar
Hough, KP, Wilson, LS, Trevor, JL, Strenkowski, JG, Maina, N, Kim, YI, Spell, ML, Wang, Y, Chanda, D, Dager, JR, Sharma, NS, Curtiss, M, Antony, VB, Dransfield, MT, Chaplin, DD, Steele, C, Barnes, S, Duncan, SR, Prasain, JK, Thannickal, VJ and Deshane, JS (2018) Unique lipid signatures of extracellular vesicles from the Airways of Asthmatics. Scientific Reports 8, 10340.Google Scholar
Levänen, B, Bhakta, NR, Torregrosa Paredes, P, Barbeau, R, Hiltbrunner, S, Pollack, JL, Sköld, CM, Svartengren, M, Grunewald, J, Gabrielsson, S, Eklund, A, Larsson, BM, Woodruff, PG, Erle, DJ and Wheelock, ÅM (2013) Altered microRNA profiles in bronchoalveolar lavage fluid exosomes in asthmatic patients. The Journal of Allergy and Clinical Immunology 131, 894903.Google Scholar
Pua, HH, Happ, HC, Gray, CJ, Mar, DJ, Chiou, NT, Hesse, LE, and Ansel, KM (2019 ) Increased hematopoietic extracellular RNAs and vesicles in the lung during allergic airway responses. Cell Reports 26, 933944.e4.Google Scholar
Rostami Hir, S, Alizadeh, Z, Mazinani, M, Mahlooji Rad, M, Fazlollahi, MR, Kazemnejad, A, Hosseini, AZ and Moin, M (2021) Exosomal MicroRNAs as biomarkers in allergic asthma. Iranian Journal of Allergy, Asthma, and Immunology 20, 160168.Google Scholar
Soccio, P, Moriondo, G, Lacedonia, D, Tondo, P, Pescatore, D, Quarato, CMI, Carone, M, Foschino Barbaro, MP and Scioscia, G (2023) MiRNA and Exosomal miRNA as new biomarkers useful to phenotyping severe asthma. Biomolecules 13, 1542.Google Scholar
Zheng, R, Du, M, Tian, M, et al. (2022) Fine particulate matter induces childhood asthma attacks via extracellular vesicle-packaged let-7i-5p-mediated modulation of the MAPK Signaling pathway. Advancement of Science 9, e2102460.Google Scholar
Zhao, M, Li, Y-P, Geng, X-R, Zhao, M, Ma, SB, Yang, YH, Deng, ZH, Luo, LM and Pan, XQ (2019) Expression level of MiRNA-126 in serum Exosomes of allergic asthma patients and lung tissues of asthmatic mice. Current Drug Metabolism 20, 799803.Google Scholar
Bahmer, T, Krauss-Etschmann, S, Buschmann, D, Behrends, J, Watz, H, Kirsten, AM, Pedersen, F, Waschki, B, Fuchs, O, Pfaffl, MW, von Mutius, E, Rabe, KF, Hansen, G, Kopp, MV, König, IR and Bartel, S (2021) RNA-seq-based profiling of extracellular vesicles in plasma reveals a potential role of miR-122-5p in asthma. Allergy 76, 366371.Google Scholar
Li, JJ, Tay, HL, Maltby, S, Xiang, Y, Eyers, F, Hatchwell, L, Zhou, H, Toop, HD, Morris, JC, Nair, P, Mattes, J, Foster, PS and Yang, M (2015) MicroRNA-9 regulates steroid-resistant airway hyperresponsiveness by reducing protein phosphatase 2A activity. The Journal of Allergy and Clinical Immunology 136, 462473.Google Scholar
Stoll, BJ, Hansen, NI, Bell, EF, Walsh, MC, Carlo, WA, Shankaran, S, Laptook, AR, Sánchez, PJ, van Meurs, K, Wyckoff, M, das, A, Hale, EC, Ball, MB, Newman, NS, Schibler, K, Poindexter, BB, Kennedy, KA, Cotten, CM, Watterberg, KL, D’Angio, CT, DeMauro, S, Truog, WE, Devaskar, U, Higgins, RD and Eunice Kennedy Shriver National Institute of Child Health and Human Development Neonatal Research Network (2015) Trends in care practices, morbidity, and mortality of extremely preterm neonates, 1993–2012. Journal of the American Medical Association 314, 10391051.Google Scholar
Lal, CV, Olave, N, Travers, C, Rezonzew, G, Dolma, K, Simpson, A, Halloran, B, Aghai, Z, das, P, Sharma, N, Xu, X, Genschmer, K, Russell, D, Szul, T, Yi, N, Blalock, JE, Gaggar, A, Bhandari, V and Ambalavanan, N (2018) Exosomal microRNA predicts and protects against severe bronchopulmonary dysplasia in extremely premature infants. JCI Insight 3, e93994.Google Scholar
Ransom, MA, Bunn, KE, Negretti, NM, Jetter, CS, Bressman, ZJ, Sucre, JMS and Pua, HH (2023) Developmental trajectory of extracellular vesicle characteristics from the lungs of preterm infants. American Journal of Physiology-Lung Cellular and Molecular Physiology 324, L385L392.Google Scholar
Barnes, MVC, Pantazi, P and Holder, B (2023) Circulating extracellular vesicles in healthy and pathological pregnancies: A scoping review of methodology, rigour and results. Journal of Extracellular Vesicles 12, e12377.Google Scholar
Zhong, X-Q, Yan, Q, Chen, Z-G, Jia, CH, Li, XH, Liang, ZY, Gu, J, Wei, HL, Lian, CY, Zheng, J and Cui, QL (2021) Umbilical cord blood-derived Exosomes from very preterm infants with Bronchopulmonary dysplasia impaired endothelial angiogenesis: Roles of Exosomal MicroRNAs. Frontiers in Cell and Development Biology 9, 637248.Google Scholar
Ali, A, Zambrano, R, Duncan, MR, Chen, S, Luo, S, Yuan, H, Chen, P, Benny, M, Schmidt, A, Young, K, Kerr, N, de Rivero Vaccari, JP, Keane, RW, Dietrich, WD and Wu, S (2021) Hyperoxia-activated circulating extracellular vesicles induce lung and brain injury in neonatal rats. Scientific Reports 11, 8791.Google Scholar
Go, H, Maeda, H, Miyazaki, K, Maeda, R, Kume, Y, Namba, F, Momoi, N, Hashimoto, K, Otsuru, S, Kawasaki, Y, Hosoya, M and Dennery, PA (2020) Extracellular vesicle miRNA-21 is a potential biomarker for predicting chronic lung disease in premature infants. American Journal of Physiology-Lung Cellular and Molecular Physiology 318, L845L851.Google Scholar
Antunes, MA, Lopes-Pacheco, M and Rocco, PRM (2021) Oxidative stress-derived mitochondrial dysfunction in chronic obstructive pulmonary disease: A concise review. Oxidative Medicine and Cellular Longevity 2021, 6644002.Google Scholar
Osei, AD, Mirbolouk, M, Orimoloye, OA, Dzaye, O, Uddin, SMI, Benjamin, EJ, Hall, ME, DeFilippis, AP, Bhatnagar, A, Biswal, SS and Blaha, MJ (2020) Association between E-cigarette use and chronic obstructive pulmonary disease by smoking status: Behavioral risk factor surveillance system 2016 and 2017. American Journal of Preventive Medicine 58, 336342.Google Scholar
Tan, DBA, Armitage, J, Teo, T-H, Ong, NE, Shin, H and Moodley, YP (2017) Elevated levels of circulating exosome in COPD patients are associated with systemic inflammation. Respiratory Medicine 132, 261264.Google Scholar
Genschmer, KR, Russell, DW, Lal, C, Szul, T, Bratcher, PE, Noerager, BD, Abdul Roda, M, Xu, X, Rezonzew, G, Viera, L, Dobosh, BS, Margaroli, C, Abdalla, TH, King, RW, McNicholas, CM, Wells, JM, Dransfield, MT, Tirouvanziam, R, Gaggar, A and Blalock, JE (2019); Activated PMN Exosomes: Pathogenic entities causing matrix destruction and disease in the lung. Cell 176, 113126.e15.Google Scholar
Takahashi, T, Kobayashi, S, Fujino, N, Suzuki, T, Ota, C, Tando, Y, Yamada, M, Yanai, M, Yamaya, M, Kurosawa, S, Yamauchi, M and Kubo, H (2014) Annual FEV1 changes and numbers of circulating endothelial microparticles in patients with COPD: A prospective study. BMJ Open 4, e004571.Google Scholar
Kim, Y-S, Lee, W-H, Choi, E-J, Choi, JP, Heo, YJ, Gho, YS, Jee, YK, Oh, YM and Kim, YK (2015) Extracellular vesicles derived from gram-negative bacteria, such as Escherichia coli, induce emphysema mainly via IL-17A-mediated neutrophilic inflammation. Journal of Immunology 194, 33613368.Google Scholar
Fujita, Y, Araya, J, Ito, S, Kobayashi, K, Kosaka, N, Yoshioka, Y, Kadota, T, Hara, H, Kuwano, K and Ochiya, T (2015) Suppression of autophagy by extracellular vesicles promotes myofibroblast differentiation in COPD pathogenesis. Journal of Extracellular Vesicles 4, 28388.Google Scholar
Xu, H, Ling, M, Xue, J, Dai, X, Sun, Q, Chen, C, Liu, Y, Zhou, L, Liu, J, Luo, F, Bian, Q and Liu, Q (2018) Exosomal microRNA-21 derived from bronchial epithelial cells is involved in aberrant epithelium-fibroblast cross-talk in COPD induced by cigarette smoking. Theranostics 8, 54195433.Google Scholar
Corsello, T, Kudlicki, AS, Garofalo, RP and Casola, A (2019) Cigarette smoke condensate exposure changes RNA content of extracellular vesicles released from Small airway epithelial cells. Cells 8, 1652.Google Scholar
Serban, KA, Rezania, S, Petrusca, DN, Poirier, C, Cao, D, Justice, MJ, Patel, M, Tsvetkova, I, Kamocki, K, Mikosz, A, Schweitzer, KS, Jacobson, S, Cardoso, A, Carlesso, N, Hubbard, WC, Kechris, K, Dragnea, B, Berdyshev, EV, McClintock, J and Petrache, I (2016) Structural and functional characterization of endothelial microparticles released by cigarette smoke. Scientific Reports 6, 31596.Google Scholar
Chen, Z, Wu, H, Shi, R, Fan, W, Zhang, J, Su, W, Wang, Y and Li, P (2021) miRNAomics analysis reveals the promoting effects of cigarette smoke extract-treated Beas-2B-derived exosomes on macrophage polarization. Biochemical and Biophysical Research Communications 572, 157163.Google Scholar
Jia, H, He, W, Wu, B, Zhong, Z, Chang, Y, Liu, Y, Wang, M and Xia, S (2024) Cigarette smoke-induced exosomal miR-221-3p facilitates M1 macrophage polarization via the STAT3 pathway in chronic obstructive pulmonary disease. Aging (Albany NY) 16, 1237912391.Google Scholar
Wang, R, Zhu, Z, Peng, S, Xu, J, chen, Y, Wei, S and Liu, X (2024) Exosome microRNA-125a-5p derived from epithelium promotes M1 macrophage polarization by targeting IL1RN in chronic obstructive pulmonary disease. International Immunopharmacology 137, 112466.Google Scholar
Jiang, Y, Wang, J, Zhang, H, Min, Y and Gu, T (2023) Serum exosome-derived MiR-7 exacerbates chronic obstructive pulmonary disease by regulating macrophage differentiation. Iranian Journal of Public Health 52, 563574.Google Scholar
Xia, H, Wu, Y, Zhao, J, Li, W, Lu, L, Ma, H, Cheng, C, Sun, J, Xiang, Q, Bian, T and Liu, Q (2022) The aberrant cross-talk of epithelium-macrophages via METTL3-regulated extracellular vesicle miR-93 in smoking-induced emphysema. Cell Biology and Toxicology 38, 167183.Google Scholar
Dai, Z, Lin, L, Xu, Y, Hu, L, Gou, S and Xu, X (2024) Extracellular vesicle dynamics in COPD: Understanding the role of miR-422a, SPP1 and IL-17 A in smoking-related pathology. BMC Pulmonary Medicine 24, 173.Google Scholar
Sundar, IK, Li, D and Rahman, I (2019) Small RNA-sequence analysis of plasma-derived extracellular vesicle miRNAs in smokers and patients with chronic obstructive pulmonary disease as circulating biomarkers. Journal of Extracellular Vesicles 8, 1684816.Google Scholar
Mizuno, S, Bogaard, HJ, Gomez-Arroyo, J, Alhussaini, A, Kraskauskas, D, Cool, CD and Voelkel, NF (2012) MicroRNA-199a-5p is associated with hypoxia-inducible factor-1α expression in lungs from patients with COPD. Chest 142, 663672.Google Scholar
O’Farrell, HE, Bowman, RV, Fong, KM, et al. (2022) Plasma extracellular vesicle miRNA profiles distinguish chronic obstructive pulmonary disease exacerbations and disease severity. International Journal of COPD 17, 28212833.Google Scholar
Wang, F, Yang, B, Qiao, J, Bai, L, Li, Z, Sun, W, Liu, Q, Yang, S and Cui, L (2023) Serum exosomal microRNA-1258 may as a novel biomarker for the diagnosis of acute exacerbations of chronic obstructive pulmonary disease. Scientific Reports 13, 18332.Google Scholar
Carpi, S, Polini, B, Nieri, D, Dubbini, N, Celi, A, Nieri, P and Neri, T (2020) Expression analysis of muscle-specific miRNAs in plasma-derived extracellular vesicles from patients with chronic obstructive pulmonary disease. Diagnostics. 10, 502.Google Scholar
O’Farrell, HE, Bowman, RV, Fong, KM and Yang, IA (2021) Plasma extracellular vesicle miRNAs can identify lung cancer, current smoking status, and stable COPD. International Journal of Molecular Sciences 22, 5803.Google Scholar
Burke, H, Cellura, D, Freeman, A, Hicks, A, Ostridge, K, Watson, A, Williams, NP, Spalluto, CM, Staples, KJ and Wilkinson, TMA (2022) Pulmonary EV miRNA profiles identify disease and distinct inflammatory endotypes in COPD. Frontiers in Medicine 9, 1039702.Google Scholar
Lopes-Pacheco, M (2020) CFTR modulators: The changing face of cystic fibrosis in the era of precision medicine. Frontiers in Pharmacology 10, 1662.Google Scholar
Lozano-Iturbe, V, Blanco-Agudín, N, Vázquez-Espinosa, E, et al. (2024) The binding of Pseudomonas aeruginosa to cystic fibrosis bronchial epithelial model cells alters the composition of the Exosomes they produce compared to healthy control cells. International Journal of Molecular Sciences [Internet] 25, 895. Available at http://www.ncbi.nlm.nih.gov/pubmed/38255969.Google Scholar
Sarkar, S, Barnaby, R, Nymon, AB, Taatjes, DJ, Kelley, TJ and Stanton, BA (2024) Extracellular vesicles secreted by primary human bronchial epithelial cells reduce Pseudomonas aeruginosa burden and inflammation in cystic fibrosis mouse lung. American Journal of Physiology-Lung Cellular and Molecular Physiology 326, L164L174.Google Scholar
Koeppen, K, Nymon, A, Barnaby, R, Li, Z, Hampton, TH, Ashare, A and Stanton, BA (2021) CF monocyte-derived macrophages have an attenuated response to extracellular vesicles secreted by airway epithelial cells. American Journal of Physiology-Lung Cellular and Molecular Physiology 320, L530L544.Google Scholar
Mao, Y, Suryawanshi, A, Patial, S and Saini, Y (2024) Airspaces-derived exosomes contain disease-relevant protein signatures in a mouse model of cystic fibrosis (CF)-like mucoinflammatory lung disease. Frontiers in Pharmacology 15, 1460692.Google Scholar
Porro, C, Di Gioia, S, Trotta, T, et al. (2013) Pro-inflammatory effect of cystic fibrosis sputum microparticles in the murine lung. Journal of Cystic Fibrosis 12, 721728.Google Scholar
Al-Humiari, MA, Yu, L, Liu, LP, et al. (2024) 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 1866, 184219.Google Scholar
Zulueta, A, Peli, V, Dei Cas, M, Colombo, M, Paroni, R, Falleni, M, Baisi, A, Bollati, V, Chiaramonte, R, del Favero, E, Ghidoni, R and Caretti, A (2019) Inflammatory role of extracellular sphingolipids in cystic fibrosis. The International Journal of Biochemistry & Cell Biology 116, 105622.Google Scholar
Forrest, OA, Dobosh, B, Ingersoll, SA, Rao, S, Rojas, A, Laval, J, Alvarez, JA, Brown, MR, Tangpricha, V and Tirouvanziam, R (2022) Neutrophil-derived extracellular vesicles promote feed-forward inflammasome signaling in cystic fibrosis airways. Journal of Leukocyte Biology 112, 707716.Google Scholar
Ben-Meir, E, Antounians, L, Eisha, S, Ratjen, F, Zani, A and Grasemann, H (2024) Extracellular vesicles in sputum of children with cystic fibrosis pulmonary exacerbations. ERJ Open Research 10, 005472024.Google Scholar
Gauthier, S, Pranke, I, Jung, V, Martignetti, L, Stoven, V, Nguyen-Khoa, T, Semeraro, M, Hinzpeter, A, Edelman, A, Guerrera, IC and Sermet-Gaudelus, I (2020) Urinary Exosomes of patients with cystic fibrosis unravel CFTR-related renal disease. International Journal of Molecular Sciences 21, 6625.Google Scholar
Trappe, A, Lakkappa, N, Carter, S, Dillon, E, Wynne, K, McKone, E, McNally, P and Coppinger, JA (2023) Investigating serum extracellular vesicles in cystic fibrosis. Journal of Cystic Fibrosis 22, 674679.Google Scholar
Oliver, KE, Carlon, MS, Pedemonte, N and Lopes-Pacheco, M (2023) The revolution of personalized pharmacotherapies for cystic fibrosis: What does the future hold? Expert Opinion on Pharmacotherapy 24, 15451565.Google Scholar
Nathan, SD, Shlobin, OA, Weir, N, Ahmad, S, Kaldjob, JM, Battle, E, Sheridan, MJ and du Bois, RM (2011) Long-term course and prognosis of idiopathic pulmonary fibrosis in the new millennium. Chest 140, 221229.Google Scholar
Kadota, T, Yoshioka, Y, Fujita, Y, Araya, J, Minagawa, S, Hara, H, Miyamoto, A, Suzuki, S, Fujimori, S, Kohno, T, Fujii, T, Kishi, K, Kuwano, K and Ochiya, T (2020) Extracellular vesicles from fibroblasts induce epithelial-cell senescence in pulmonary fibrosis. American Journal of Respiratory Cell and Molecular Biology 63, 623636.Google Scholar
Kadota, T, Fujita, Y, Yoshioka, Y, Araya, J, Kuwano, K and Ochiya, T (2018) Emerging role of extracellular vesicles as a senescence-associated secretory phenotype: Insights into the pathophysiology of lung diseases. Molecular Aspects of Medicine 60, 92103.Google Scholar
Antunes, MA, Braga, CL, Oliveira, TB, Kitoko, JZ, Castro, LL, Xisto, DG, Coelho, MS, Rocha, N, Silva-Aguiar, RP, Caruso-Neves, C, Martins, EG, Carvalho, CF, Galina, A, Weiss, DJ, Lapa e Silva, JR, Lopes-Pacheco, M, Cruz, FF and Rocco, PRM (2021) Mesenchymal stromal cells from emphysematous donors and their extracellular vesicles are unable to reverse cardiorespiratory dysfunction in experimental severe emphysema. Frontiers in Cell and Development Biology 9, 661385.Google Scholar
Parimon, T, Yao, C, Habiel, DM, Ge, L, Bora, SA, Brauer, R, Evans, CM, Xie, T, Alonso-Valenteen, F, Medina-Kauwe, LK, Jiang, D, Noble, PW, Hogaboam, CM, Deng, N, Burgy, O, Antes, TJ, Königshoff, M, Stripp, BR, Gharib, SA and Chen, P (2019) Syndecan-1 promotes lung fibrosis by regulating epithelial reprogramming through extracellular vesicles. JCI Insight 5, 129359.Google Scholar
Martin-Medina, A, Lehmann, M, Burgy, O, Hermann, S, Baarsma, HA, Wagner, DE, de Santis, MM, Ciolek, F, Hofer, TP, Frankenberger, M, Aichler, M, Lindner, M, Gesierich, W, Guenther, A, Walch, A, Coughlan, C, Wolters, P, Lee, JS, Behr, J and Königshoff, M (2018) Increased extracellular vesicles mediate WNT5A Signaling in idiopathic pulmonary fibrosis. American Journal of Respiratory and Critical Care Medicine 198, 15271538.Google Scholar
Chanda, D, Otoupalova, E, Hough, KP, Locy, ML, Bernard, K, Deshane, JS, Sanderson, RD, Mobley, JA and Thannickal, VJ (2019) Fibronectin on the surface of extracellular vesicles mediates fibroblast invasion. American Journal of Respiratory Cell and Molecular Biology 60, 279288.Google Scholar
Asghar, S, Monkley, S, Smith, DJF, Hewitt, RJ, Grime, K, Murray, LA, Overed-Sayer, CL and Molyneaux, PL (2023) Epithelial senescence in idiopathic pulmonary fibrosis is propagated by small extracellular vesicles. Respiratory Research 24, 51.Google Scholar
Njock, M-S, Guiot, J, Henket, MA, Nivelles, O, Thiry, M, Dequiedt, F, Corhay, JL, Louis, RE and Struman, I (2019) Sputum exosomes: Promising biomarkers for idiopathic pulmonary fibrosis. Thorax 74, 309312.Google Scholar
Guiot, J, Cambier, M, Boeckx, A, Henket, M, Nivelles, O, Gester, F, Louis, E, Malaise, M, Dequiedt, F, Louis, R, Struman, I and Njock, MS (2020) Macrophage-derived exosomes attenuate fibrosis in airway epithelial cells through delivery of antifibrotic miR-142-3p. Thorax 75, 870881.Google Scholar
Makiguchi, T, Yamada, M, Yoshioka, Y, Sugiura, H, Koarai, A, Chiba, S, Fujino, N, Tojo, Y, Ota, C, Kubo, H, Kobayashi, S, Yanai, M, Shimura, S, Ochiya, T and Ichinose, M (2016) Serum extracellular vesicular miR-21-5p is a predictor of the prognosis in idiopathic pulmonary fibrosis. Respiratory Research 17, 110.Google Scholar
Siegel, RL, Miller, KD, Wagle, NS and Jemal, A (2023) Cancer statistics, 2023. CA: A Cancer Journal for Clinicians 73, 1748.Google Scholar
Yamashita, T, Kamada, H, Kanasaki, S, Maeda, Y, Nagano, K, Abe, Y, Inoue, M, Yoshioka, Y, Tsutsumi, Y, Katayama, S, Inoue, M and Tsunoda, S (2013) Epidermal growth factor receptor localized to exosome membranes as a possible biomarker for lung cancer diagnosis. Die Pharmazie 68, 969973.Google Scholar
Fabbri, M, Paone, A, Calore, F, Galli, R, Gaudio, E, Santhanam, R, Lovat, F, Fadda, P, Mao, C, Nuovo, GJ, Zanesi, N, Crawford, M, Ozer, GH, Wernicke, D, Alder, H, Caligiuri, MA, Nana-Sinkam, P, Perrotti, D and Croce, CM (2012) MicroRNAs bind to toll-like receptors to induce prometastatic inflammatory response. Proceedings of the National Academy of Sciences of the United States of America 109, E2110E2116.Google Scholar
Lobb, RJ, Visan, KS, Wu, L-Y, Norris, EL, Hastie, ML, Everitt, S, Yang, IA, Bowman, RV, Siva, S, Larsen, JE, Gorman, JJ, MacManus, M, Leimgruber, A, Fong, KM and Möller, A (2023) An epithelial-to-mesenchymal transition induced extracellular vesicle prognostic signature in non-small cell lung cancer. Communications Biology 6, 68.Google Scholar
Kim, DH, Park, S, Kim, H, Choi, YJ, Kim, SY, Sung, KJ, Sung, YH, Choi, CM, Yun, M, Yi, YS, Lee, CW, Kim, SY, Lee, JC and Rho, JK (2020) Tumor-derived exosomal miR-619-5p promotes tumor angiogenesis and metastasis through the inhibition of RCAN1.4. Cancer Letters 475, 213.Google Scholar
Cohnen, A, Chiang, SC, Stojanovic, A, Schmidt, H, Claus, M, Saftig, P, Janßen, O, Cerwenka, A, Bryceson, YT and Watzl, C (2013) Surface CD107a/LAMP-1 protects natural killer cells from degranulation-associated damage. Blood 122, 14111418.Google Scholar
Liu, K, Gao, L, Ma, X, Huang, JJ, Chen, J, Zeng, L, Ashby, CR Jr, Zou, C and Chen, ZS (2020) Long non-coding RNAs regulate drug resistance in cancer. Molecular Cancer 19, 54.Google Scholar
Zhang, Y and Xu, H (2020) Serum exosomal miR-378 upregulation is associated with poor prognosis in non-small-cell lung cancer patients. Journal of Clinical Laboratory Analysis 34, e23237.Google Scholar
Kato, T, Vykoukal, JV, Fahrmann, JF and Hanash, S (2021) Extracellular vesicles in lung cancer: Prospects for diagnostic and therapeutic applications. Cancers (Basel) 13, 4604.Google Scholar
Jin, X, Chen, Y, Chen, H, Fei, S, Chen, D, Cai, X, Liu, L, Lin, B, Su, H, Zhao, L, Su, M, Pan, H, Shen, L, Xie, D and Xie, C (2017) Evaluation of tumor-derived Exosomal miRNA as potential diagnostic biomarkers for early-stage non-Small cell lung cancer using next-generation sequencing. Clinical Cancer Research 23, 53115319.Google Scholar
Bakouboula, B, Morel, O, Faure, A, Zobairi, F, Jesel, L, Trinh, A, Zupan, M, Canuet, M, Grunebaum, L, Brunette, A, Desprez, D, Chabot, F, Weitzenblum, E, Freyssinet, JM, Chaouat, A and Toti, F (2008) Procoagulant membrane microparticles correlate with the severity of pulmonary arterial hypertension. American Journal of Respiratory and Critical Care Medicine 177, 536543.Google Scholar
Diehl, P, Aleker, M, Helbing, T, Sossong, V, Germann, M, Sorichter, S, Bode, C and Moser, M (2011) Increased platelet, leukocyte and endothelial microparticles predict enhanced coagulation and vascular inflammation in pulmonary hypertension. Journal of Thrombosis and Thrombolysis 31, 173179.Google Scholar
Khandagale, A, Åberg, M, Wikström, G, Bergström Lind, S, Shevchenko, G, Björklund, E, Siegbahn, A and Christersson, C (2020) Role of extracellular vesicles in pulmonary arterial hypertension: Modulation of pulmonary endothelial function and angiogenesis. Arteriosclerosis, Thrombosis, and Vascular Biology 40, 22932309.Google Scholar
Amabile, N, Heiss, C, Chang, V, Angeli, FS, Damon, L, Rame, EJ, McGlothlin, D, Grossman, W, de Marco, T and Yeghiazarians, Y (2009) Increased CD62e(+) endothelial microparticle levels predict poor outcome in pulmonary hypertension patients. The Journal of Heart and Lung Transplantation 28, 10811086.Google Scholar
Rose, JA, Wanner, N, Cheong, HI, Queisser, K, Barrett, P, Park, M, Hite, C, Naga Prasad, SV, Erzurum, S and Asosingh, K (2016) Flow Cytometric quantification of peripheral blood cell β-adrenergic receptor density and urinary endothelial cell-derived microparticles in pulmonary arterial hypertension. PLoS One 11, e0156940.Google Scholar
Gąsecka, A, Banaszkiewicz, M, Nieuwland, R, van der Pol, E, Hajji, N, Mutwil, H, Rogula, S, Rutkowska, W, Pluta, K, Eyileten, C, Postuła, M, Darocha, S, Huczek, Z, Opolski, G, Filipiak, K, Torbicki, A and Kurzyna, M (2021) Prostacyclin analogues inhibit platelet reactivity, extracellular vesicle release and thrombus formation in patients with pulmonary arterial hypertension. Journal of Clinical Medicine 10, 1024.Google Scholar
Khandagale, A, Corcoran, P, Nikpour, M, Isaksson, A, Wikström, G, Siegbahn, A and Christersson, C (2022) MircoRNA in extracellular vesicles from patients with pulmonary arterial hypertension alters endothelial Angiogenic response. International Journal of Molecular Sciences 23, 11964.Google Scholar
Huang, Y, Wang, Z-G, Tang, L, Gong, SG, Sun, YY, Wang, L, Jiang, R, Wu, WH, Luo, CJ, Zhang, J, Yang, XJ, Li, JL, Yuan, XT, Zhao, QH and Yuan, P (2021) Plasma exosomal miR-596: A novel biomarker predicts survival in patients with idiopathic pulmonary artery hypertension. The Journal of International Medical Research 49, 3000605211002379.Google Scholar
Ferrer, E, Dunmore, BJ, Hassan, D, Ormiston, ML, Moore, S, Deighton, J, Long, L, Yang, XD, Stewart, DJ and Morrell, NW (2018) A potential role for Exosomal Translationally controlled tumor protein export in vascular Remodeling in pulmonary arterial hypertension. American Journal of Respiratory Cell and Molecular Biology 59, 467478.Google Scholar
Zhang, S, Liu, J, Zheng, K, Chen, L, Sun, Y, Yao, Z, Sun, Y, Lin, Y, Lin, K and Yuan, L (2021) Exosomal miR-211 contributes to pulmonary hypertension via attenuating CaMK1/PPAR-γaxis. Vascular Pharmacology 136, 106820.Google Scholar
Mohieldin, AM, Pala, R, Beuttler, R, Moresco, JJ, Yates, JR III and Nauli, SM (2021) Ciliary extracellular vesicles are distinct from the cytosolic extracellular vesicles. Journal of Extracellular Vesicles 10, e12086.Google Scholar
Qin, X, Lin, X, Liu, L, Li, Y, Li, X, Deng, Z, Chen, H, Chen, H, Niu, Z, Li, Z and Hu, Y (2021) Macrophage-derived exosomes mediate silica-induced pulmonary fibrosis by activating fibroblast in an endoplasmic reticulum stress-dependent manner. Journal of Cellular and Molecular Medicine 25, 44664477.Google Scholar
Xia, J, Wang, D, Guo, W, Pei, Y, Zhang, L, Bao, L, Li, Y, Qu, Y, Zhao, Y, Hao, C and Yao, W (2024) Exposure to micron-grade silica particles triggers pulmonary fibrosis through cell-to-cell delivery of exosomal miR-107. International Journal of Biological Macromolecules 266, 131058.Google Scholar
Ding, M, Pei, Y, Zhang, C, Qi, Y, Xia, J, Hao, C and Yao, W (2023) Exosomal miR-125a-5p regulates T lymphocyte subsets to promote silica-induced pulmonary fibrosis by targeting TRAF6. Ecotoxicology and Environmental Safety 249, 114401.Google Scholar
Hou, L, Zhu, Z, Jiang, F, Zhao, J, Jia, Q, Jiang, Q, Wang, H, Xue, W, Wang, Y and Tian, L (2023) Human umbilical cord mesenchymal stem cell-derived extracellular vesicles alleviated silica induced lung inflammation and fibrosis in mice via circPWWP2A/miR-223-3p/NLRP3 axis. Ecotoxicology and Environmental Safety 251, 114537.Google Scholar
Futami, Y, Takeda, Y, Koba, T, Narumi, R, Nojima, Y, Ito, M, Nakayama, M, Ishida, M, Yoshimura, H, Naito, Y, Fukushima, K, Takimoto, T, Edahiro, R, Matsuki, T, Nojima, S, Hirata, H, Koyama, S, Iwahori, K, Nagatomo, I, Shirai, Y, Suga, Y, Satoh, S, Futami, S, Miyake, K, Shiroyama, T, Inoue, Y, Adachi, J, Tomonaga, T, Ueda, K and Kumanogoh, A (2022) Identification of CD14 and lipopolysaccharide-binding protein as novel biomarkers for sarcoidosis using proteomics of serum extracellular vesicles. International Immunology 34, 327340.Google Scholar
Kraaijvanger, R, Janssen Bonás, M, Grutters, JC, Paspali, I, Veltkamp, M, de Kleijn, DPV and van Moorsel, CHM (2024) Decreased serpin C1 in extracellular vesicles predicts response to methotrexate treatment in patients with pulmonary sarcoidosis. Respiratory Research 25, 166.Google Scholar
Lopes-Pacheco, M and Rocco, PRM (2023) Functional enhancement strategies to potentiate the therapeutic properties of mesenchymal stromal cells for respiratory diseases. Frontiers in Pharmacology 14, 1067422.Google Scholar
Ionescu, LI, Alphonse, RS, Arizmendi, N, Morgan, B, Abel, M, Eaton, F, Duszyk, M, Vliagoftis, H, Aprahamian, TR, Walsh, K and Thébaud, B (2012) Airway delivery of soluble factors from plastic-adherent bone marrow cells prevents murine asthma. American Journal of Respiratory Cell and Molecular Biology 46, 207216.Google Scholar
Cruz, FF, Borg, ZD, Goodwin, M, Sokocevic, D, Wagner, DE, Coffey, A, Antunes, M, Robinson, KL, Mitsialis, SA, Kourembanas, S, Thane, K, Hoffman, AM, McKenna, DH, Rocco, PRM and Weiss, DJ (2015) Systemic Administration of Human Bone Marrow-Derived Mesenchymal Stromal Cell Extracellular Vesicles Ameliorates Aspergillus Hyphal Extract-Induced Allergic Airway Inflammation in Immunocompetent mice. Stem Cells Translational Medicine 4, 13021316.Google Scholar
Du, Y-M, Zhuansun, Y-X, Chen, R, et al. (2018) Mesenchymal stem cell exosomes promote immunosuppression of regulatory T cells in asthma. Experimental Cell Research 363, 114120.Google Scholar
de Castro, LL, Xisto, DG, Kitoko, JZ, Cruz, FF, Olsen, PC, Redondo, PAG, Ferreira, TPT, Weiss, DJ, Martins, MA, Morales, MM and Rocco, PRM (2017) Human adipose tissue mesenchymal stromal cells and their extracellular vesicles act differentially on lung mechanics and inflammation in experimental allergic asthma. Stem Cell Research & Therapy 8, 151.Google Scholar
Mun, SJ, Kang, SA, Park, H-K, Yu, HS, Cho, KS and Roh, HJ (2021) Intranasally administered extracellular vesicles from adipose stem cells have Immunomodulatory effects in a mouse model of asthma. Stem Cells International 2021, 6686625.Google Scholar
Song, J, Zhu, X-M and Wei, Q-Y (2020) MSCs reduce airway remodeling in the lungs of asthmatic rats through the Wnt/β-catenin signaling pathway. European Review for Medical and Pharmacological Sciences 24, 1119911211.Google Scholar
Fang, S-B, Zhang, H-Y, Meng, X-C, Wang, C, He, BX, Peng, YQ, Xu, ZB, Fan, XL, Wu, ZJ, Wu, ZC, Zheng, SG and Fu, QL (2020) Small extracellular vesicles derived from human MSCs prevent allergic airway inflammation via immunomodulation on pulmonary macrophages. Cell Death & Disease 11, 409.Google Scholar
Shang, Y, Sun, Y, Xu, J, Ge, X, Hu, Z, Xiao, J, Ning, Y, Dong, Y and Bai, C (2020) Exosomes from mmu_circ_0001359-modified ADSCs attenuate airway Remodeling by enhancing FoxO1 Signaling-mediated M2-like macrophage activation. Molecular Therapy Nucleic Acids 19, 951960.Google Scholar
Dong, B, Wang, C, Zhang, J, Zhang, J, Gu, Y, Guo, X, Zuo, X, Pan, H, Hsu, ACY, Wang, G and Wang, F (2021) Exosomes from human umbilical cord mesenchymal stem cells attenuate the inflammation of severe steroid-resistant asthma by reshaping macrophage polarization. Stem Cell Research & Therapy 12, 204.Google Scholar
Shan, L, Liu, S, Zhang, Q, Zhou, Q and Shang, Y (2022) Human bone marrow-mesenchymal stem cell-derived exosomal microRNA-188 reduces bronchial smooth muscle cell proliferation in asthma through suppressing the JARID2/Wnt/β-catenin axis. Cell Cycle 21, 352367.Google Scholar
Liu, W, Lin, H, Nie, W, Wan, J, Jiang, Q and Zhang, A (2022) Exosomal miR-221-3p derived from bone marrow mesenchymal stem cells alleviates asthma progression by targeting FGF2 and inhibiting the ERK1/2 Signaling pathway. Evidence-Based Complementary and Alternative Medicine 2022, 5910874.Google Scholar
Feng, C-Y, Bai, S-Y, Li, M-L, Zhao, JY, Sun, JM, Bao, HJ, Ren, Y and Su, XM (2022) Adipose-derived mesenchymal stem cell-derived Exosomal miR-301a-3p regulates airway smooth muscle cells during asthma by targeting STAT3. Journal of Asthma and Allergy 15, 99110.Google Scholar
Li, X and Yang, N (2023) Exosome miR-223-3p in the bone marrow-derived mesenchymal stem cells alleviates the inflammation and airway remodeling through NLRP3-induced ASC/Caspase-1/GSDMD signaling pathway. International Immunopharmacology 123, 110746.Google Scholar
Dong, L, Wang, Y, Zheng, T, Pu, Y, Ma, Y, Qi, X, Zhang, W, Xue, F, Shan, Z, Liu, J, Wang, X and Mao, C (2021) Hypoxic hUCMSC-derived extracellular vesicles attenuate allergic airway inflammation and airway remodeling in chronic asthma mice. Stem Cell Research & Therapy 12, 4.Google Scholar
Sholihah, IA and Barlian, A (2024) Anti-inflammatory potency of human Wharton’s jelly mesenchymal stem cell-derived Exosomes on L2 cell line induced by lipopolysaccharides. Advanced Pharmaceutical Bulletin 14, 434444.Google Scholar
Xu, X, Wang, Y, Luo, X, Gao, X, Gu, W, Ma, Y, Xu, L, Yu, M, Liu, X, Liu, J, Wang, X, Zheng, T, Mao, C and Dong, L (2023) A non-invasive strategy for suppressing asthmatic airway inflammation and remodeling: Inhalation of nebulized hypoxic hUCMSC-derived extracellular vesicles. Frontiers in Immunology 14, 1150971.Google Scholar
Luo, X, Wang, Y, Mao, Y, Xu, X, Gu, W, Li, W, Mao, C, Zheng, T and Dong, L (2024) Nebulization of hypoxic hUCMSC-EVs attenuates airway epithelial barrier defects in chronic asthma mice by transferring CAV-1. International Journal of Nanomedicine 19, 1094110959.Google Scholar
Willis, GR, Fernandez-Gonzalez, A, Anastas, J, Vitali, SH, Liu, X, Ericsson, M, Kwong, A, Mitsialis, SA and Kourembanas, S (2018) Mesenchymal stromal cell Exosomes ameliorate experimental Bronchopulmonary dysplasia and restore lung function through macrophage immunomodulation. American Journal of Respiratory and Critical Care Medicine 197, 104116.Google Scholar
Willis, GR, Fernandez-Gonzalez, A, Reis, M, Yeung, V, Liu, X, Ericsson, M, Andrews, NA, Mitsialis, SA and Kourembanas, S (2020) Mesenchymal stromal cell-derived small extracellular vesicles restore lung architecture and improve exercise capacity in a model of neonatal hyperoxia-induced lung injury. Journal of Extracellular Vesicles 9, 1790874.Google Scholar
Willis, GR, Reis, M, Gheinani, AH, Fernandez-Gonzalez, A, Taglauer, ES, Yeung, V, Liu, X, Ericsson, M, Haas, E, Mitsialis, SA and Kourembanas, S (2021) Extracellular vesicles protect the neonatal lung from Hyperoxic injury through the epigenetic and Transcriptomic reprogramming of myeloid cells. American Journal of Respiratory and Critical Care Medicine 204, 14181432.Google Scholar
Reis, M, Willis, GR, Fernandez-Gonzalez, A, Yeung, V, Taglauer, E, Magaletta, M, Parsons, T, Derr, A, Liu, X, Maehr, R, Kourembanas, S and Mitsialis, SA (2021) Mesenchymal stromal cell-derived extracellular vesicles restore Thymic architecture and T cell function disrupted by neonatal Hyperoxia. Frontiers in Immunology 12, 640595.Google Scholar
Ai, D, Shen, J, Sun, J, Zhu, Z, Gao, R, du, Y, Yuan, L, Chen, C and Zhou, J (2022) Mesenchymal stem cell-derived extracellular vesicles suppress Hyperoxia-induced Transdifferentiation of rat alveolar type 2 epithelial cells. Stem Cells and Development 31, 5366.Google Scholar
Abele, AN, Taglauer, ES, Almeda, M, Wilson, N, Abikoye, A, Seedorf, GJ, Mitsialis, SA, Kourembanas, S and Abman, SH (2022) Antenatal mesenchymal stromal cell extracellular vesicle treatment preserves lung development in a model of bronchopulmonary dysplasia due to chorioamnionitis. American Journal of Physiology-Lung Cellular and Molecular Physiology 322, L179L190.Google Scholar
Lassus, P, Ristimäki, A, Ylikorkala, O, et al. (1999) Vascular endothelial growth factor in human preterm lung. American Journal of Respiratory and Critical Care Medicine 159, 14291433.Google Scholar
Ahn, SY, Park, WS, Kim, YE, Sung, DK, Sung, SI, Ahn, JY and Chang, YS (2018) Vascular endothelial growth factor mediates the therapeutic efficacy of mesenchymal stem cell-derived extracellular vesicles against neonatal hyperoxic lung injury. Experimental & Molecular Medicine 50, 112.Google Scholar
Lithopoulos, MA, Strueby, L, O’Reilly, M, Zhong, S, Möbius, MA, Eaton, F, Fung, M, Hurskainen, M, Cyr-Depauw, C, Suen, C, Xu, L, Collins, JJP, Vadivel, A, Stewart, DJ, Burger, D and Thébaud, B (2022) Pulmonary and neurologic effects of mesenchymal stromal cell extracellular vesicles in a multifactorial lung injury model. American Journal of Respiratory and Critical Care Medicine 205, 11861201.Google Scholar
Huh, JW, Kim, S-Y, Lee, JH, Lee, JS, van Ta, Q, Kim, MJ, Oh, YM, Lee, YS and Lee, SD (2011) Bone marrow cells repair cigarette smoke-induced emphysema in rats. American Journal of Physiology-Lung Cellular and Molecular Physiology 301, L255L266.Google Scholar
Kennelly, H, Mahon, BP and English, K (2016) Human mesenchymal stromal cells exert HGF dependent cytoprotective effects in a human relevant pre-clinical model of COPD. Scientific Reports 6, 38207.Google Scholar
Song, L, Peng, J and Guo, X (2021) Exosomal lncRNA TCONS_00064356 derived from injured alveolar epithelial type II cells affects the biological characteristics of mesenchymal stem cells. Life Sciences 278, 119568.Google Scholar
Maremanda, KP, Sundar, IK and Rahman, I (2019) Protective role of mesenchymal stem cells and mesenchymal stem cell-derived exosomes in cigarette smoke-induced mitochondrial dysfunction in mice. Toxicology and Applied Pharmacology 385, 114788.Google Scholar
Ridzuan, N, Zakaria, N, Widera, D, Sheard, J, Morimoto, M, Kiyokawa, H, Mohd Isa, SA, Chatar Singh, GK, Then, KY, Ooi, GC and Yahaya, BH (2021) Human umbilical cord mesenchymal stem cell-derived extracellular vesicles ameliorate airway inflammation in a rat model of chronic obstructive pulmonary disease (COPD). Stem Cell Research & Therapy 12, 54.Google Scholar
Zhu, Z, Lian, X, Su, X, Wu, W, Zeng, Y and Chen, X (2022) Exosomes derived from adipose-derived stem cells alleviate cigarette smoke-induced lung inflammation and injury by inhibiting alveolar macrophages pyroptosis. Respiratory Research 23, 5.Google Scholar
Chen, Q, Lin, J, Deng, Z and Qian, W (2022) Exosomes derived from human umbilical cord mesenchymal stem cells protect against papain-induced emphysema by preventing apoptosis through activating VEGF-VEGFR2-mediated AKT and MEK/ERK pathways in rats. Regenerative Therapy 21, 216224.Google Scholar
Harrell, CR, Miloradovic, D, Sadikot, R, Fellabaum, C, Markovic, BS, Miloradovic, D, Acovic, A, Djonov, V, Arsenijevic, N and Volarevic, V (2020) Molecular and cellular mechanisms responsible for beneficial effects of mesenchymal stem cell-derived product ‘Exo-d-MAPPS’ in attenuation of chronic airway inflammation. Analytical Cellular Pathology 2020, 3153891.Google Scholar
Kim, Y-S, Kim, J-Y, Cho, R, Shin, DM, Lee, SW and Oh, YM (2017) Adipose stem cell-derived nanovesicles inhibit emphysema primarily via an FGF2-dependent pathway. Experimental & Molecular Medicine 49, e284.Google Scholar
Liang, W, Chen, X, Zhang, S, Fang, J, Chen, M, Xu, Y and Chen, X (2021) Mesenchymal stem cells as a double-edged sword in tumor growth: Focusing on MSC-derived cytokines. Cellular & Molecular Biology Letters 26, 3.Google Scholar
Slama, Y, Ah-Pine, F, Khettab, M, Arcambal, A, Begue, M, Dutheil, F and Gasque, P (2023) The dual role of mesenchymal stem cells in cancer pathophysiology: Pro-tumorigenic effects versus therapeutic potential. International Journal of Molecular Sciences 24, 13511.Google Scholar
Wu, T, Tian, Q, Liu, R, Xu, K, Shi, S, Zhang, X, Gao, L, Yin, X, Xu, S and Wang, P (2023) Inhibitory role of bone marrow mesenchymal stem cells-derived exosome in non-small-cell lung cancer: microRNA-30b-5p, EZH2 and PI3K/AKT pathway. Journal of Cellular and Molecular Medicine 27, 35263538.Google Scholar
Li, X and Wu, F (2023) Mesenchymal stem cell-derived extracellular vesicles transfer miR-598 to inhibit the growth and metastasis of non-small-cell lung cancer by targeting THBS2. Cell Death Dscovery 9, 3.Google Scholar
Liu, X-N, Zhang, C-B, Lin, H, Tang, XY, Zhou, R, Wen, HL and Li, J (2021) microRNA-204 shuttled by mesenchymal stem cell-derived exosomes inhibits the migration and invasion of non-small-cell lung cancer cells via the KLF7/AKT/HIF-1α axis. Neoplasma 68, 719731.Google Scholar
Liang, Y, Zhang, D, Li, L, Xin, T, Zhao, Y, Ma, R and du, J (2020) Exosomal microRNA-144 from bone marrow-derived mesenchymal stem cells inhibits the progression of non-small cell lung cancer by targeting CCNE1 and CCNE2. Stem Cell Research & Therapy 11, 87.Google Scholar
Xie, H and Wang, J (2022) MicroRNA-320a-containing exosomes from human umbilical cord mesenchymal stem cells curtail proliferation and metastasis in lung cancer by binding to SOX4. Journal of Receptor and Signal Transduction Research 42, 268278.Google Scholar
Liu, J, Feng, Y, Zeng, X, He, M, Gong, Y and Liu, Y (2021) Extracellular vesicles-encapsulated let-7i shed from bone mesenchymal stem cells suppress lung cancer via KDM3A/DCLK1/FXYD3 axis. Journal of Cellular and Molecular Medicine 25, 19111926.Google Scholar
Dong, L, Pu, Y, Zhang, L, Qi, Q, Xu, L, Li, W, Wei, C, Wang, X, Zhou, S, Zhu, J, Wang, X, Liu, F, Chen, X and Su, C (2018) Human umbilical cord mesenchymal stem cell-derived extracellular vesicles promote lung adenocarcinoma growth by transferring miR-410. Cell Death & Disease 9, 218.Google Scholar
Zhang, X, Sai, B, Wang, F, Wang, L, Wang, Y, Zheng, L, Li, G, Tang, J and Xiang, J (2019) Hypoxic BMSC-derived exosomal miRNAs promote metastasis of lung cancer cells via STAT3-induced EMT. Molecular Cancer 18, 40.Google Scholar
Ren, W, Hou, J, Yang, C, Wang, H, Wu, S, Wu, Y, Zhao, X and Lu, C (2019) Extracellular vesicles secreted by hypoxia pre-challenged mesenchymal stem cells promote non-small cell lung cancer cell growth and mobility as well as macrophage M2 polarization via miR-21-5p delivery. Journal of Experimental & Clinical Cancer Research 38, 62.Google Scholar
Shentu, T-P, Huang, T-S, Cernelc-Kohan, M, Chan, J, Wong, SS, Espinoza, CR, Tan, C, Gramaglia, I, van der Heyde, H, Chien, S and Hagood, JS (2017) Thy-1 dependent uptake of mesenchymal stem cell-derived extracellular vesicles blocks myofibroblastic differentiation. Scientific Reports 7, 18052.Google Scholar
Tan, JL, Lau, SN, Leaw, B, Nguyen, HPT, Salamonsen, LA, Saad, MI, Chan, ST, Zhu, D, Krause, M, Kim, C, Sievert, W, Wallace, EM and Lim, R (2018) Amnion epithelial cell-derived Exosomes restrict lung injury and enhance endogenous lung repair. Stem Cells Translational Medicine 7, 180196.Google Scholar
Mansouri, N, Willis, GR, Fernandez-Gonzalez, A, Reis, M, Nassiri, S, Mitsialis, SA and Kourembanas, S (2019) Mesenchymal stromal cell exosomes prevent and revert experimental pulmonary fibrosis through modulation of monocyte phenotypes. JCI Insight 4, e128060.Google Scholar
Gao, Y, Sun, J, Dong, C, Zhao, M, Hu, Y and Jin, F (2020) Extracellular vesicles derived from adipose mesenchymal stem cells alleviate PM2.5-induced lung injury and pulmonary fibrosis. Medical Science Monitor 26, e922782.Google Scholar
Bandeira, E, Oliveira, H, Silva, JD, Menna-Barreto, RFS, Takyia, CM, Suk, JS, Witwer, KW, Paulaitis, ME, Hanes, J, Rocco, PRM and Morales, MM (2018) Therapeutic effects of adipose-tissue-derived mesenchymal stromal cells and their extracellular vesicles in experimental silicosis. Respiratory Research 19, 104.Google Scholar
Xu, C, Zhao, J, Li, Q, Hou, L, Wang, Y, Li, S, Jiang, F, Zhu, Z and Tian, L (2020) Exosomes derived from three-dimensional cultured human umbilical cord mesenchymal stem cells ameliorate pulmonary fibrosis in a mouse silicosis model. Stem Cell Research & Therapy 11, 503.Google Scholar
Wan, X, Chen, S, Fang, Y, Zuo, W, Cui, J and Xie, S (2020) Mesenchymal stem cell-derived extracellular vesicles suppress the fibroblast proliferation by downregulating FZD6 expression in fibroblasts via micrRNA-29b-3p in idiopathic pulmonary fibrosis. Journal of Cellular Physiology 235, 86138625.Google Scholar
Zhou, J, Lin, Y, Kang, X, Liu, Z, Zhang, W and Xu, F (2021) microRNA-186 in extracellular vesicles from bone marrow mesenchymal stem cells alleviates idiopathic pulmonary fibrosis via interaction with SOX4 and DKK1. Stem Cell Research & Therapy 12, 96.Google Scholar
Shi, L, Ren, J, Li, J, Wang, D, Wang, Y, Qin, T, Li, X, Zhang, G, Li, C and Wang, Y (2021) Extracellular vesicles derived from umbilical cord mesenchymal stromal cells alleviate pulmonary fibrosis by means of transforming growth factor-β signaling inhibition. Stem Cell Research & Therapy 12, 230.Google Scholar
Sun, L, Zhu, M, Feng, W, Lin, Y, Yin, J, Jin, J and Wang, Y (2019) Exosomal miRNA Let-7 from menstrual blood-derived endometrial stem cells alleviates pulmonary fibrosis through regulating mitochondrial DNA damage. Oxidative Medicine and Cellular Longevity 2019, 4506303.Google Scholar
Zhu, L, Xu, Y, Wang, J, Zhang, Y, Zhou, J and Wu, H (2024) Mesenchymal stem cells-derived exosomes carrying microRNA-30b confer protection against pulmonary fibrosis by downregulating Runx1 via Spred2. Molecular Genetics and Genomics 299, 33.Google Scholar
Nataliya, B, Mikhail, A, Vladimir, P, Olga, G, Maksim, V, Ivan, Z, Ekaterina, N, Georgy, S, Natalia, D, Pavel, M, Andrey, C, Maria, S, Maxim, K, Anastasiya, T, Uliana, D, Zhanna, A, Vsevolod, T, Natalia, K and Anastasiya, E (2023) Mesenchymal stromal cells facilitate resolution of pulmonary fibrosis by miR-29c and miR-129 intercellular transfer. Experimental & Molecular Medicine 55, 13991412.Google Scholar
Lei, X, He, N, Zhu, L, Zhou, M, Zhang, K, Wang, C, Huang, H, Chen, S, Li, Y, Liu, Q, Han, Z, Guo, Z, Han, Z and Li, Z (2021) Mesenchymal stem cell-derived extracellular vesicles attenuate radiation-induced lung injury via miRNA-214-3p. Antioxidants & Redox Signaling 35, 849862.Google Scholar
Zhao, Y, Du, L, Sun, J, et al. (2023) Exosomal miR-218 derived from mesenchymal stem cells inhibits endothelial-to-mesenchymal transition by epigenetically modulating of BMP2 in pulmonary fibrosis. Cell Biology and Toxicology 39, 29192936.Google Scholar
Phinney, DG, Di Giuseppe, M, Njah, J, et al. (2015) Mesenchymal stem cells use extracellular vesicles to outsource mitophagy and shuttle microRNAs. Nature Communications 6, 8472.Google Scholar
Xu, C, Hou, L, Zhao, J, Wang, Y, Jiang, F, Jiang, Q, Zhu, Z and Tian, L (2022) Exosomal let-7i-5p from three-dimensional cultured human umbilical cord mesenchymal stem cells inhibits fibroblast activation in silicosis through targeting TGFBR1. Ecotoxicology and Environmental Safety 233, 113302.Google Scholar
Zhao, J, Jiang, Q, Xu, C, Jia, Q, Wang, H, Xue, W, Wang, Y, Zhu, Z and Tian, L (2023) MiR-26a-5p from HucMSC-derived extracellular vesicles inhibits epithelial mesenchymal transition by targeting Adam17 in silica-induced lung fibrosis. Ecotoxicology and Environmental Safety 257, 114950.Google Scholar
Rathinasabapathy, A, Bruce, E, Espejo, A, Horowitz, A, Sudhan, DR, Nair, A, Guzzo, D, Francis, J, Raizada, MK, Shenoy, V and Katovich, MJ (2016) Therapeutic potential of adipose stem cell-derived conditioned medium against pulmonary hypertension and lung fibrosis. British Journal of Pharmacology 173, 28592879.Google Scholar
Liu, J, Han, Z, Han, Z, et al. (2016) Mesenchymal stem cell-conditioned media suppresses inflammation-associated overproliferation of pulmonary artery smooth muscle cells in a rat model of pulmonary hypertension. Experimental and Therapeutic Medicine 11, 467475.Google Scholar
Klinger, JR, Pereira, M, Del Tatto, M, et al. (2020) Mesenchymal stem cell extracellular vesicles reverse Sugen/hypoxia pulmonary hypertension in rats. American Journal of Respiratory Cell and Molecular Biology 62, 577587.Google Scholar
Chen, J, An, R, Liu, Z, Wang, JJ, Chen, SZ, Hong, MM, Liu, JH, Xiao, MY and Chen, YF (2014) Therapeutic effects of mesenchymal stem cell-derived microvesicles on pulmonary arterial hypertension in rats. Acta Pharmacologica Sinica 35, 11211128.Google Scholar
Aliotta, JM, Pereira, M, Wen, S, Dooner, MS, del Tatto, M, Papa, E, Goldberg, LR, Baird, GL, Ventetuolo, CE, Quesenberry, PJ and Klinger, JR (2016) Exosomes induce and reverse monocrotaline-induced pulmonary hypertension in mice. Cardiovascular Research 110, 319330.Google Scholar
Liu, Z, Liu, J, Xiao, M, Wang, J, Yao, F, Zeng, W, Yu, L, Guan, Y, Wei, W, Peng, Z, Zhu, K, Wang, J, Yang, Z, Zhong, J and Chen, J (2018) Mesenchymal stem cell-derived microvesicles alleviate pulmonary arterial hypertension by regulating renin-angiotensin system. Journal of the American Society of Hypertension 12, 470478.Google Scholar
Zhang, Z, Ge, L, Zhang, S, Wang, J, Jiang, W, Xin, Q and Luan, Y (2020) The protective effects of MSC-EXO against pulmonary hypertension through regulating Wnt5a/BMP signalling pathway. Journal of Cellular and Molecular Medicine 24, 1393813948.Google Scholar
Liu, H, Zhang, Q, Liu, C, Zhang, Y, Wang, Y, Huang, P, Ma, L and Ge, R (2024) Human umbilical cord mesenchymal stromal cell-derived Exosomes alleviate hypoxia-induced pulmonary arterial hypertension in mice via macrophages. Stem Cells 42, 329345.Google Scholar
Jeppesen, DK, Hvam, ML, Primdahl-Bengtson, B, Boysen, AT, Whitehead, B, Dyrskjøt, L, Ørntoft, TF, Howard, KA and Ostenfeld, MS (2014) Comparative analysis of discrete exosome fractions obtained by differential centrifugation. Journal of Extracellular Vesicles 3, 25011.Google Scholar
Kowal, J, Arras, G, Colombo, M, Jouve, M, Morath, JP, Primdal-Bengtson, B, Dingli, F, Loew, D, Tkach, M and Théry, C (2016) Proteomic comparison defines novel markers to characterize heterogeneous populations of extracellular vesicle subtypes. Proceedings of the National Academy of Sciences of the United States of America 113, E968E977.Google Scholar
Yuana, Y, Levels, J, Grootemaat, A, Sturk, A and Nieuwland, R (2014) Co-isolation of extracellular vesicles and high-density lipoproteins using density gradient ultracentrifugation. Journal of Extracellular Vesicles 3.Google Scholar
Corso, G, Mäger, I, Lee, Y, Görgens, A, Bultema, J, Giebel, B, Wood, MJA, Nordin, JZ and Andaloussi, SEL (2017) Reproducible and scalable purification of extracellular vesicles using combined bind-elute and size exclusion chromatography. Scientific Reports 7, 11561.Google Scholar
Karimi, N, Cvjetkovic, A, Jang, SC, Crescitelli, R, Hosseinpour Feizi, MA, Nieuwland, R, Lötvall, J and Lässer, C (2018) Detailed analysis of the plasma extracellular vesicle proteome after separation from lipoproteins. Cellular and Molecular Life Sciences 75, 28732886.Google Scholar
Karttunen, J, Heiskanen, M, Navarro-Ferrandis, V, das Gupta, S, Lipponen, A, Puhakka, N, Rilla, K, Koistinen, A and Pitkänen, A (2019) Precipitation-based extracellular vesicle isolation from rat plasma co-precipitate vesicle-free microRNAs. Journal of Extracellular Vesicles 8, 1555410.Google Scholar
Contreras-Naranjo, JC, Wu, H-J and Ugaz, VM (2017) Microfluidics for exosome isolation and analysis: Enabling liquid biopsy for personalized medicine. Lab on a Chip 17, 35583577.Google Scholar
Preußer, C, Stelter, K, Tertel, T, Linder, M, Helmprobst, F, Szymanski, W, Graumann, J, Giebel, B, Reinartz, S, Müller, R, Weber, G and von Strandmann, EP (2022) Isolation of native EVs from primary biofluids-free-flow electrophoresis as a novel approach to purify ascites-derived EVs. Journal of Extracellular Biology 1, e71.Google Scholar
Ghosh, A, Davey, M, Chute, IC, Griffiths, SG, Lewis, S, Chacko, S, Barnett, D, Crapoulet, N, Fournier, S, Joy, A, Caissie, MC, Ferguson, AD, Daigle, M, Meli, MV, Lewis, SM and Ouellette, RJ (2014) Rapid isolation of extracellular vesicles from cell culture and biological fluids using a synthetic peptide with specific affinity for heat shock proteins. PLoS One 9, e110443.Google Scholar
Mathieu, M, Névo, N, Jouve, M, Valenzuela, JI, Maurin, M, Verweij, FJ, Palmulli, R, Lankar, D, Dingli, F, Loew, D, Rubinstein, E, Boncompain, G, Perez, F and Théry, C (2021) Specificities of exosome versus small ectosome secretion revealed by live intracellular tracking of CD63 and CD9. Nature Communications 12, 4389.Google Scholar
Figure 0

Figure 1. Extracellular vesicle (EV) composition. EVs are small, non-replicating membrane-bound particles released by cells into the extracellular environment. These particles are delimited by a lipid bilayer with membrane proteins (e.g., tetraspanins, receptors, immunoglobulins, and adhesion molecules), and their internal content is composed mainly of cytosolic proteins, lipids, and nucleic acids (e.g., DNA and RNA), which reflects the state of their parental cells. Some EVs can also contain metabolites and cellular organelles (e.g., mitochondria).

Figure 1

Table 1. EV subtypes based on size, formation/origin, and related markers (Refs. 6–8)

Figure 2

Table 2. Methods commonly used for EV separation and concentration

Figure 3

Table 3. Methods commonly used for EV characterization

Figure 4

Figure 2. Extracellular vesicles (EVs) in lung physiological or pathological processes. The respiratory system is composed of diverse cell types, each with specialized roles, relying on intricate communication to preserve the structural and functional integrity of airways and alveoli. Under normal physiological (or pathological) conditions, EVs participate in lung homeostasis by promoting intercellular communication, thus regulating resident and immune cell responses, supporting barrier integrity, and facilitating tissue repair (or remodelling). EVs have been considered suitable biomarkers for diagnosis due to differences in their size, quantity, cellular origin, and content (both on the surface and within their lumen) in diverse diseases, which can facilitate the distinction between pathological conditions and severity with high accuracy and enable early and personalized therapeutic interventions. Mesenchymal stromal cell (MSC)-derived EVs have been demonstrated to be attractive therapeutic agents for respiratory diseases due to their ability to reduce inflammation and fibrosis, improve fluid clearance, and repair epithelial and endothelial barrier permeability.

Figure 5

Figure 3. Mesenchymal stromal cell (MSC)-derived extracellular vesicles (EVs) in respiratory diseases. Major therapeutic effects of MSC-derived EVs were found in experimental models of asthma, bronchopulmonary dysplasia (BPD), chronic obstructive pulmonary disease (COPD), lung cancer, lung fibrosis, and pulmonary arterial hypertension (PAH).