Cell & Developmental Biology

Cell & Developmental Biology
Open Access

ISSN: 2168-9296

+44 1478 350008

Mini Review - (2016) Volume 5, Issue 2

Extracellular Vesicles Facilitate the Intercellular Communications in the Pathogenesis of Lung Injury

Heedoo Lee, Duo Zhang, Jasleen Minhas and Yang Jin*
1Division of Pulmonary and Critical Care Medicine, Department of Medicine, Boston University, Boston, USA, E-mail: hlee@changwon.ac.kr
2Internal Medicine, North Shore Medical Center, Salem Hospital, 81 Highland Ave, Salem, USA, E-mail: hlee@changwon.ac.kr
*Corresponding Author: Yang Jin, Division of Pulmonary and Critical Care, Department of Medicine, Boston University, Medical Campus 72 East Concord St., Boston, MA 02118, USA, Tel: 6174143291 Email:

Abstract

Extracellular vesicles (EVs) are a group of heterogeneous, nano-sized structures surrounded by lipid bilayer membranes that are released by cells. Depending on their size and mechanisms of formation, EVs are often referred to as exosomes, microvesicles (MVs) and apoptotic bodies (AB). EVs are evolutionally conserved vesicles that mediate intercellular communications and cross-talk, via transferring proteins, lipids and nucleic acids. Accumulating evidence suggests that EVs exert essential physiological and pathological functions on both their mother and recipient cells. Therefore, growing interests focus on the potentials of EVs to serve as novel targets for the development of therapeutic and diagnostic strategies. Currently, extensive reports are yielded from cancer research. However, besides malignancy, EVs may also serve as crucial regulators in other devastating conditions, such as the acute respiratory distress syndrome (ARDS) and acute lung injury (ALI). The generation, regulation and function of EVs in ARDS/ALI are largely unexplored. In this mini review, we will briefly review the current understanding of EVs and their known physiological/pathological functions in the pathogenesis of ARDS/ALI. Previously, only scattered reports have been published in this field. We believe that further investigations focusing on EVs and their compositions will shed light on novel insights in the research of ARDS/ALI.

<

Keywords: Apoptotic body; Inflammation; Oxidative stress; Hyperoxia; Extracellular vesicle (EV); Exosome; Infections; Lung injury

Background

Accumulating evidence suggest that extracellular vesicles (EVs) mediate cell-cell cross talk [1], particularly in the fields of tumor genesis [2-6]. In recent years, extracellular vesicles have been isolated from most non-malignant cells and biological fluids including saliva [7], bronchial lavage fluid (BALF) [8,9], breast milk [10], amniotic fluid [11], blood [12,13], and urine [14]. Findings have also suggested that EV-shuttling molecules, including proteins, RNAs, microRNAs (miRNAs) and lipids, potentially exert essential roles in the pathogenesis of human diseases [15].

Acute respiratory distress syndrome (ARDS) is a devastating entity encountered in critical ill patients. Despite the recent advances on medical knowledge and strategies, the mortality and mobility of ARDS remain unacceptably high [16]. There is dire need of identification of novel targets to develop diagnostic and therapeutic strategies Human lungs have a large surface area only second to skin and are in contact with air constantly. The lung epithelium plays an essential role in innate immunity and host defense due to constant exposure to environmental stimuli including microorganisms and disease pathogens [17]. Lung injury frequently occurs in response to diverse noxious stimuli and pathogens numerous types of cells reside in the lungs and intercellular communication during lung injury is poorly understood. The discovery of EVs shines a light on our understanding of the development of human lung injury.

Classification of EVs

EVs refer to a group of heterogeneous vesicles in which the contents, size and mechanisms of formation are different [15]. The International Society of Extracellular Vesicles recently defined three main subgroups of EVs [15]. Exosomes are the smallest subgroup measuring approximately 30-100 nm in diameter [18-20]. Microvesicles (MVs) are the second largest subgroup in size, ranging from 100 nm to 500 nm [18-20]. Apoptotic bodies (ABs) have the largest size and are the most variable vesicles amongst the three subgroups. They range from 500-2000 nm in diameter and are comparable to platelets [18-20].

Mechanistically, exosomes are released from cells after multivesicular bodies (MVBs) fuse with the plasma membrane [5,20]. MV formation involves direct protruding from plasma membranes [19,20]. Similarly, ABs is formed by plasma membrane blebbing during the process of apoptosis [21]. However, our interests focus more on the exosomes and MVs rather than ABs, given that vesicles and their compositions derived from live cells potentially play more crucial functions in the development of lung injury. That being said, currently there is no single marker can uniquely identify each subgroup of EVs. The groups of proteins which have often been used as markers of EVs are not specific to either exosomes or MVs. These proteins include, but are not limited to: tetraspanins such as CD9, CD63, CD81 and CD82; 14-3-3 proteins, major histocompatibility complex (MHC) molecules and heat shock proteins; HSPs, Tsg101 and the Endosomal Sorting Complex Required for Transport (ESCRT-3) binding protein Alix [22-26].

EV Composition

Thus far, proteins, nucleic acids and lipids have all been identified in EVs [27]. These compositions are derived based on the parent cells [27]. For example, lung surfactant proteins

(SPs) have been detected in the EVs derived from lung epithelial cells [28]. These cell-specific proteins can serve as markers to reflect the origins of the EVs. Abundant cytoskeletal-, cytosolic-, heat shock-, plasma membrane proteins, and proteins involved in vesicle trafficking have been found in both exosomes and MVs [29].

EV-shuttling cytokines identified so far include, but are not limited to, interleukin 1β (IL-1β), IL1α, IL-18, macrophage migration inhibitory factor (MIF), IL-32, tumor necrosis factor (TNF), IL-6, vascular endothelial growth factor (VEGF), IL-8 (CXCL8), fractalkine (CX3CL1), CCL2-5 and CCL20 [30]. Whether the EV-containing cytokines and chemokines are functional in their recipient cells remains unclear and requires further investigation.

RNAs were first identified in EVs in 2006 [31]. The EV-containing RNAs appear much smaller (less than 700 nucleotides (nt)) in comparison to cellular RNAs. However, fragments of long RNAs such as the mRNAs, long non-coding RNAs (lncRNAs), ribosomal RNA (rRNA) have all been identified in EVs [18,32,33]. More interestingly, miRNAs, the 20-22 nt small non-coding RNA molecules, have also been found in a variety of EVs [34], suggesting that EVs serve as a cargo for circulating miRNAs. Oncogenic DNAs, mitochondrial DNA (mtDNA), single-stranded DNA, double-stranded DNA (dsDNA) have all been identified in EVs [35-39].

Lipids were first described in prostate-derived EVs (named prostasomes) in 1989 [40]. Emerging new lipid families have been described in EVs, including prostaglandin E2, F2, J2 and D2 [41]. Lysobisphosphatidic acid may participate in exosome biogenesis and contribute to vesicle budding from cell membranes [42]. The lipids from cellular plasma membranes are expectedly found in the lipid bilayers of exosomes. These includesphingomyelin, phosphatidylcholine, phosphatidyl-ethanolamine, phosphatidylserine, ganglioside GM3 and phosphatidylinositol [43-45].

EV Uptakes by Target Cells

Multiple theories have been proposed on how EVs reach their recipient cells and transmit carried information Postulated mechanisms include, initial internalization of the EVs into the recipient cells, with subsequent transport of EV-shuttling molecules, such as proteins, cytokines, RNA/DNA molecules or fragments, non-coding RNAs, miRNAs, etc. [46,47] into the recipient cells. Currently, EVmediated small non-coding RNA or small interfering RNAs (siRNAs) delivery has been confirmed in a number of cell types [48-50]. The proposed mechanisms for EV uptake by the recipient cells primarily include clathrin-mediated endocytosis (CME), phagocytosis, macropinocytosis and plasma membrane fusion [46]. A second mechanism may include interactions between EV proteins and plasma membrane receptors on recipient cells [3,51-53]. Additionally, fusion with the plasma membrane of the recipient cells provides another route to deliver EV compositions into the recipient cells [54].

EV Functions in the Development of Lung Injury

The most commonly proposed roles of EVs include the emission and transportation of signaling/regulatory molecules for intercellular communications, subsequently resulting in the modulation of the immune system and antigen presentation. There is increasing attention on the application of EVs in diagnostics and therapeutics for human diseases, particularly in the field of cancer diagnosis and cancer metastasis diagnosis. Despite the growing applications of EVs in the field of oncology, the functions of EVs in lung diseases remain unclear.

Role of EVs in acute lung injury (ALI)

Numerous observations have linked EVs with the development of lung injury/ARDS. For example, during the pathogenesis of a variety of type of lung injury, the generation of “microparticles” (MPs) has been observed in the platelets, neutrophils, monocytes, lymphocytes, red blood cells, and endothelial and epithelial cells [55]. Endothelial cell-derived “EVs” have been reported to contain S1PR3 and represent the inflammatory states of acute lung injury (ALI) [56]. Endothelial cell (EC)-derived EVs are also believed to be important markers of lung vascular injury in the development of ventilator induced lung injury (VILI) [57]. Endothelial EVs significantly increase after exposure of endothelial cells to physiological or pathological mechanical stress, such as cyclic stretch. Similar observations have been made in the infection-associated ALI. Robustly higher amounts of endothelial EVs are noted after exposure to LPS [57].

Stored, packed RBCs release RBC-originated MPs which contribute to neutrophil priming, activation and transfusion associated ALI (TRALI) [58]. In addition to RBCs, the platelet-derived MPs increase during the storage period, prime the fMLP-activated PMN respiration burst, which may induce TRALI [59]. Moreover, monocyte-derived MPs up regulate the level of pro-inflammatory factors in lung epithelial cells, primarily through activating NF-κB and PPAR-γ dependent pathways [60].

Mitochondria-mediated ROS play a crucial function in the pathogenesis of ALI [61]. Mitochondria in bone-marrow-derived stromal cells are released in a microvesicle-containing manner, and subsequently play a protective role in ALI [62]. On the other hand, alveolar epithelial cell-derived “EVs” are reported to serve as the main source of tissue factor (TF) pro-coagulant activity in ARDS [63].

Recently, using the hyperoxia induced ALI mouse mode (HALI), Moon et al. have demonstrated that lung epithelial cells release a robust amount of EVs [28]. These EVs are derived from live cells rather than apoptotic or dying cells. Their sizes fall mainly the range of exosomes or MVs (100-500 nm). Interestingly, stromal cells remove harmful mitochondria via EVs in a similar mechanism. Lung epithelial cells also release robust amount of EV-enwrapped caspase 3. This observation suggests that in additional to being a messenger, EVs also serve as a cargo for “trash” disposal. Moreover, Moon et al further observed that epithelial EVs trigger alveolar macrophage activation and proinflammatory cytokine releases, confirming their roles in mediating cell-cell cross-talk [28].

In summary, the potential roles of EVs in the pathogenesis of ARDS/ lung injury have been reported in a variety of settings, including but not limited to - infection, oxidative stress, transfusion and mechanical stretch-associated ALI. Almost all cell types release EVs, and these EVs may be protective or detrimental, depending on the type of stimuli, the type of mother cells and the type and compositions of the EVs. Despite the above mentioned observations, detailed characterization and mechanistic exploration on EVs involving in ALI/ARDS remain largely unclear. Further directions include extensive investigations on the EVfunctions under diverse stimuli, EV-compositions and classifications, as well as the mechanisms of EV formation.

Acknowledgement

This work is support by NIH R01HL102076 (Y.J.), NIH R21 AI121644 (Y.J.) and NIH R01 GM111313 (Y.J.)

References

  1. Lee Y, El Andaloussi S, Wood MJ (2012)Exosomes and microvesicles: extracellular vesicles for genetic information transfer and gene therapy. Hum Mol Genet 21: R125-134.
  2. De Broe M, Wieme R, Roels F (1975) Letter: Membrane fragments with koinozymic properties released from villous adenoma of the rectum. Lancet 2: 1214-1215.
  3. Raposo G, Nijman HW, Stoorvogel W, Liejendekker R, Harding CV, et al. (1996) B lymphocytes secrete antigen-presenting vesicles. J Exp Med 183: 1161-1172.
  4. Harding C, Heuser J, Stahl P (1984) Endocytosis and intracellular processing of transferrin and colloidal gold-transferrin in rat reticulocytes: Demonstration of a pathway for receptor shedding. Eur J Cell Biol 35: 256-263.
  5. Johnstone RM, Adam M, Hammond JR, Orr L, Turbide C (1987) Vesicle formation during reticulocyte maturation. Association of plasma membrane activities with released vesicles (exosomes). J BiolChem 262: 9412-9420.
  6. Pan BT, Johnstone RM (1983) Fate of the transferrin receptor during maturation of sheep reticulocytes invitro: Selective externalization of the receptor. Cell 33: 967-978.
  7. Kapsogeorgou EK, Abu-Helu RF, Moutsopoulos HM, Manoussakis MN (2005) Salivary gland epithelial cell exosomes: A source of autoantigenicribonucleoproteins. Arthritis Rheum 52: 1517-1521.
  8. TorregrosaParedes P, Esser J, Admyre C, Nord M, Rahman QK, et al. (2012) Bronchoalveolar lavage fluid exosomes contribute to cytokine and leukotriene production in allergic asthma.Allergy 67: 911-919.
  9. Admyre C, Grunewald J, Thyberg J, Gripenback S, Tornling G, et al. (2003)Exosomes with major histocompatibility complex class II and co-stimulatory molecules are present in human BAL fluid. EurRespir J 22: 578-583.
  10. Zhou Q, Li M, Wang X, Li Q, Wang T, et al. (2012) Immune-related microRNAs are abundant in breast milk exosomes.Int J BiolSci 8: 118-123.
  11. Jean-Pierre C, Perni SC, Bongiovanni AM, Kalish RB, Karasahan E, et al. (2006) Extracellular 70-kd heat shock protein in mid-trimester amniotic fluid and its effect on cytokine production by ex vivo-cultured amniotic fluid cells.Am J ObstetGynecol 194: 694-698.
  12. Yang J, Wei F, Schafer C, Wong DT (2014) Detection of tumor cell-specific mRNA and protein in exosome-like microvesicles from blood and saliva.PLoS One 9: e110641.
  13. CHARGAFF E, WEST R (1946) The biological significance of the thromboplastic protein of blood.J BiolChem 166: 189-197.
  14. Dear JW, Street JM, Bailey MA (2013) Urinary exosomes: A reservoir for biomarker discovery and potential mediators of intra-renal signalling. Proteomics 13: 1572-1580.
  15. Yanez-Mo M, Siljander PRM, Andreu Z, Zavec AB, Borras FE, et al.(2015) Biological properties of extracellular vesicles and their physiological functions. J Extracell Vesicles 4: 27066.
  16. Baron RM, Levy BD (2016) Recent advances in understanding and treating ARDS.F1000Res 5.
  17. Hiemstra PS, McCray PB Jr, Bals R (2015) The innate immune function of airway epithelial cells in inflammatory lung disease.EurRespir J 45: 1150-1162.
  18. Crescitelli R, Lässer C, Szabó TG, Kittel A, Eldh M, et al. (2013) Distinct RNA profiles in sub-populations of extracellular vesicles: Apoptotic bodies, microvesicles and exosomes.J Extracell Vesicles 2.
  19. Turturici G, Tinnirello R, Sconzo G, Geraci F (2014) Extracellular membrane vesicles as a mechanism of cell-to-cell communication: Advantages and disadvantages.Am J Physiol Cell Physiol 306: C621-633.
  20. Théry C, Ostrowski M, Segura E (2009) Membrane vesicles as conveyors of immune responses.Nat Rev Immunol 9: 581-593.
  21. Akers JC, Gonda D, Kim R, Carter BS, Chen CC (2013) Biogenesis of extracellular vesicles (EV): exosomes, microvesicles, retrovirus-like vesicles and apoptotic bodies.J Neurooncol 113: 1-11.
  22. Gross C, Schmidt-Wolf IGH, Nagaraj S, Gastpar R, Ellwart J, et al. (2003) Heat shock protein 70-reactivity is associated with increased cell surface density of CD94/CD56 on primary natural killer cells. Cell Stress Chaperon 8: 348-360.
  23. Campanella C, Bucchieri F, Merendino AM, Fucarino A, Burgio G, et al. (2012) The Odyssey of Hsp60 from tumor cells to other destinations includes plasma membrane-associated stages and golgi and exosomal protein-trafficking modalities. Plos One 7: e42008.
  24. Merendino AM1, Bucchieri F, Campanella C, Marcianò V, Ribbene A, et al. (2010) Hsp60 is actively secreted by human tumor cells.PLoS One 5: e9247.
  25. Subra C, Grand D, Laulagnier K, Stella A, Lambeau G, et al. (2010)Exosomes account for vesicle-mediated transcellular transport of activatable phospholipases and prostaglandins. J Lipid Res 51: 2105-2120.
  26. Buschow SI, van Balkom BWM, Aalberts M, Heck AJR, Wauben M, et al. (2010) MHC class II-associated proteins in B-cell exosomes and potential functional implications for exosome biogenesis. Immunol Cell Biol 88: 851-856.
  27. Xu R, Greening DW, Zhu HJ, Takahashi N, Simpson RJ (2016) Extracellular vesicle isolation and characterization: Toward clinical application. J Clin Invest 126: 1152-1162.
  28. Moon HG, Cao Y, Yang J, Lee JH, Choi HS, et al. (2015) Lung epithelial cell-derived extracellular vesicles activate macrophage-mediated inflammatory responses via ROCK1 pathway. Cell Death Dis 6: e2016.
  29. Zaborowski MP, Balaj L, Breakefield XO, Lai CP (2015) Extracellular vesicles: Composition, biological relevance and methods of study. Bioscience 65: 783-797.
  30. Campos JH, Soares RP, Ribeiro K, Andrade AC, Batista WL, et al. (2015) Extracellular vesicles: Role in inflammatory responses and potential uses in vaccination in cancer and infectious diseases. J Immunol Res 2015: 832057.
  31. Ratajczak J, Miekus K, Kucia M, Zhang J, Reca R, et al. (2006)Embryonic stem cell-derived microvesicles reprogram hematopoietic progenitors: evidence for horizontal transfer of mRNA and protein delivery. Leukemia 20: 847-856.
  32. Mittelbrunn M, Sánchez-Madrid F (2012) Intercellular communication: Diverse structures for exchange of genetic information.Nat Rev Mol Cell Biol 13: 328-335.
  33. Kogure T, Yan IK, Lin WL, Patel T (2013) Extracellular vesicle-mediated transfer of a novel long noncoding RNA TUC339: A mechanism of intercellular signaling in human hepatocellular cancer. Genes Cancer 4: 261-272.
  34. Ha M, Kim VN (2014) Regulation of microRNA biogenesis.Nat Rev Mol Cell Biol 15: 509-524.
  35. Lee TH, Chennakrishnaiah S, Audemard E, Montermini L, Meehan B, et al. (2014) Oncogenic ras-driven cancer cell vesiculation leads to emission of double-stranded DNA capable of interacting with target cells. BiochemBiophys Res Commun 451: 295-301.
  36. Guescini M, Genedani S, Stocchi V, Agnati LF (2010) Astrocytes andglioblastoma cells release exosomes carrying mtDNA.J Neural Transm (Vienna) 117: 1-4.
  37. Balaj L, Lessard R, Dai L, Cho YJ, Pomeroy SL, et al. (2011)Tumourmicrovesicles contain retrotransposon elements and amplified oncogene sequences. Nat Commun2: 180.
  38. Thakur BK, Zhang H, Becker A, Matei I, Huang Y, et al. (2014) Double-stranded DNA in exosomes: a novel biomarker in cancer detection. Cell Res 24: 766-769.
  39. Waldenström A, Gennebäck N, Hellman U, Ronquist G (2012) Cardiomyocytemicrovesicles contain DNA/RNA and convey biological messages to target cells.PLoS One 7: e34653.
  40. Arvidson G, Ronquist G, Wikander G, Ojteg AC (1989) Human prostasome membranes exhibit very high cholesterol/phospholipid ratios yielding high molecular ordering. BiochimBiophysActa 984: 167-173.
  41. Yoon YJ, Kim OY, Gho YS (2014) Extracellular vesicles as emerging intercellular communicasomes.BMB Rep 47: 531-539.
  42. Brouwers JF, Aalberts M, Jansen JW, van Niel G, Wauben MH, et al. (2013) Distinct lipid compositions of two types of human prostasomes.Proteomics 13: 1660-1666.
  43. Laulagnier K, Motta C, Hamdi S, Roy S, Fauvelle F, et al. (2004) Mast cell- and dendritic cell-derived exosomes display a specific lipid composition and an unusual membrane organization. Biochem J 380: 161-171.
  44. Choi DS, Kim DK, Kim YK, Gho YS (2013) Proteomics, transcriptomics and lipidomics of exosomes and ectosomes.Proteomics 13: 1554-1571.
  45. Arienti G, Carlini E, Polci A, Cosmi EV, Palmerini CA (1998) Fatty acid pattern of human prostasome lipid.Arch BiochemBiophys 358: 391-395.
  46. Mulcahy LA, Pink RC, Carter DR (2014) Routes and mechanisms of extracellular vesicle uptake.J Extracell Vesicles 3.
  47. Franzen CA, Simms PE, Van Huis AF, Foreman KE, Kuo PC, et al. (2014) Characterization of uptake and internalization of exosomes by bladder cancer cells.Biomed Res Int 2014: 619829.
  48. Alvarez-Erviti L, Seow Y, Yin H, Betts C, Lakhal S, et al. (2011) Delivery of siRNA to the mouse brain by systemic injection of targeted exosomes.Nat Biotechnol 29: 341-345.
  49. Luo SS, Ishibashi O, Ishikawa G, Ishikawa T, Katayama A, et al. (2009) Human villous trophoblasts express and secrete placenta-specific microRNAs into maternal circulation via exosomes. BiolReprod 81: 717-729.
  50. Aliotta JM, Pereira M, Johnson KW, de Paz N, Dooner MS, et al. (2010)Microvesicle entry into marrow cells mediates tissue-specific changes in mRNA by direct delivery of mRNA and induction of transcription. ExpHematol 38: 233-245.
  51. Svensson KJ, Christianson HC, Wittrup A, Bourseau-Guilmain E, Lindqvist E, et al. (2013)Exosome uptake depends on ERK1/2-heat shock protein 27 signaling and lipid Raft-mediated endocytosis negatively regulated by caveolin-1. J BiolChem 288: 17713-17724.
  52. Morelli AE, Larregina AT, Shufesky WJ, Sullivan ML, Stolz DB, et al. (2004) Endocytosis, intracellular sorting and processing of exosomesby dendritic cells. Blood 104: 3257-3266.
  53. Tumne A, Prasad VS, Chen Y, Stolz DB, Saha K, et al. (2009) Non-cytotoxic suppression of human immunodeficiency virus type 1 transcription by exosomes secreted from CD8+ T cells. J Virol 83: 4354-4364.
  54. Parolini I, Federici C, Raggi C, Lugini L, Palleschi S, et al. (2009) Microenvironmental pH is a key factor for exosome traffic in tumor cells.J BiolChem 284: 34211-34222.
  55. McVey M, Tabuchi A, Kuebler WM (2012) Microparticles and acute lung injury.Am J Physiol Lung Cell MolPhysiol 303: L364-381.
  56. Sun X, Singleton PA, Letsiou E, Zhao J, Belvitch P, et al. (2012) Sphingosine-1-phosphate receptor-3 is a novel biomarker in acute lung injury.Am J Respir Cell MolBiol 47: 628-636.
  57. Letsiou E, Sammani S, Zhang W, Zhou T, Quijada H, et al. (2015) Pathologic mechanical stress and endotoxin exposure increases lung endothelial microparticle shedding. Am J Respir Cell MolBiol 52: 193-204.
  58. Belizaire RM, Prakash PS, Richter JR, Robinson BR, Edwards MJ, et al. (2012)Microparticles from stored red blood cells activates neutrophils and cause lung injury after hemorrhage and resuscitation. J Am CollSurg 214: 648-655.
  59. Xie RF, Hu P, Li W, Ren YN, Yang J, et al. (2014)The effect of platelet-derived microparticles in stored apheresis platelet concentrates on polymorphonuclear leucocyte respiratory burst. Vox Sang 106: 234-241.
  60. Neri T, Armani C, Pegoli A, Cordazzo C, Carmazzi Y, et al. (2011) Role of NF-kappa B and PPAR-gamma in lung inflammation induced by monocyte-derived microparticles.EurRespir J 37: 1494-1502.
  61. Schumacker PT, Gillespie MN, Nakahira K, Choi AM, Crouser ED, et al. (2014) Mitochondria in lung biology and pathology: more than just a powerhouse.Am J Physiol Lung Cell MolPhysiol 306: L962-974.
  62. Islam MN, Das SR, Emin MT, Wei M, Sun L, et al. (2012)Mitochondrial transfer from bone-marrow-derived stromal cells to pulmonary alveoli protects against acute lung injury. Nat Med 18: 759-765.
  63. Bastarache JA, Fremont RD, Kropski JA, Bossert FR, Ware LB (2009)Procoagulant alveolar microparticles in the lungs of patients with acute respiratory distress syndrome. Am J Physiol Lung Cell MolPhysiol 297: L1035-1041.
Citation: Lee H, Zhang D, Minhas J, Jin Y (2016) Extracellular Vesicles Facilitate the Intercellular Communications in the Pathogenesis of Lung Injury. Cell Dev Biol 5:175.

Copyright: © 2016 Lee H, et al. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.
Top