Minireviews Open Access
Copyright ©The Author(s) 2016. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Immunol. Mar 27, 2016; 6(1): 67-74
Published online Mar 27, 2016. doi: 10.5411/wji.v6.i1.67
Noncanonical intercellular communication in immune response
Malgorzata Kloc, Department of Surgery, Methodist Hospital, Houston, TX 77030, United States
Malgorzata Kloc, Xian C Li, Rafik M Ghobrial, Houston Methodist Research Institute, Houston, TX 77030, United States
Jacek Z Kubiak, Centre National de la Recherche Scientifique Unités Mixtes de Recherche 6290, Institute of Genetics and Development of Rennes, Cell Cycle Group, Faculty of Medicine, University of Rennes, Rennes, 35043 Rennes Cedex, France
Jacek Z Kubiak, Faculty of Medicine, University of Rennes 1, 35043 Rennes Cedex, France
Rafik M Ghobrial, Sherrie and Alan Conover Center for Liver Disease and Transplantation, Houston, TX 77030, United States
Author contributions: Kloc M contributed to concept, manuscript writing, figures drawing; Kubiak JZ, Li XC and Ghobrial RM contributed to manuscript co-writing.
Supported by William Stamps Farish Fund and Donald D. Hammill Foundation.
Conflict-of-interest statement: There is no conflict of interest to disclose.
Open-Access: This article is an open-access article which was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution Non Commercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: http://creativecommons.org/licenses/by-nc/4.0/
Correspondence to: Dr. Malgorzata Kloc, Department of Surgery, Methodist Hospital, 6550 Fannin St., Houston, TX 77030, United States. mkloc@houstonmethodist.org
Telephone: +1-713-4416875 Fax: +1-713-7903755
Received: July 22, 2015
Peer-review started: August 5, 2015
First decision: October 13, 2015
Revised: November 24, 2015
Accepted: December 17, 2015
Article in press: December 18, 2015
Published online: March 27, 2016

Abstract

The classical view of signaling between cells of immune system includes two major routes of intercellular communication: Through the release of extracellular molecules or a direct interaction between membrane bound receptor and its membrane bound ligand, which initiate a cascade of signaling in target cell. However, recent studies indicate that besides these canonical modes of signaling there are also noncanonical routs of intercellular communications through membrane stripping/membrane exchange/trogocytosis, extracellular traps, exosomes and ectososmes/microparticles. In this review we discuss what are the components of noncanonical pathways of signaling and what role they play in immune cells interactions.

Key Words: Trogocytosis, Membrane stripping, Extracellular traps, Exosomes, Ectososmes, Microparticles

Core tip: Noncanonical routes of intercellular communications through membrane stripping, trogocytosis, extracellular traps, microparticles and exosomes and their function in immune response are highlighted.



INTRODUCTION

For many decades general belief has been that during an immune response the immune cells communicate either by a direct contact between molecules anchored at the plasma membrane of adjacent cells (juxtracrine signaling) or via short (autocrine and paracrine) or long (endocrine) distance signaling using various cytokines or hormones and their cognate receptors systems (Figure 1). However, recent years studies, amounting to hundreds of publications, indicate that besides these well-known (canonical) signaling pathways there is a cornucopia of nonclassical (noncanonical) signaling mechanisms, which modify behavior of immune cells and shape the immune response. Below we give a brief overview of features and functions of these noncanonical signaling pathways.

Figure 1
Figure 1 Types of canonical signaling. In autocrine signaling, the cell regulates itself (autoregulates) through internally produced signaling molecules, which after release from the cell bind to cell’s own receptors. The examples are: Interleukin-1 produced by monocyte in response to external stimuli binds to its own receptor on the same monocyte; IL-2 released from activated T cell binds to its own receptor leading to self-stimulation. The juxtacrine signaling occurs between closely apposing cells when signaling molecule attached to one cell interacts with its receptor on adjacent cells or when signaling molecule excreted to the intercellular matrix of one cell binds to the receptor on neighboring cell. In juxtacrine signaling the signaling molecules do not diffuse freely between cells. The examples include cytokine signaling in immune system and Notch pathway signaling. In paracrine signaling, released signaling molecules such as, for example, cytokines or retinoic acid diffuse at short distances and act on the cells located in vicinity. In endocrine signaling, signaling molecules such as hormones or cytokines are transported through the circulation to the target cells.
MEMBRANE STRIPPING/MEMBRANE EXCHANGE/TROGOCYTOSIS

The trogocytosis - the nibbling (gnawing) off the fragments of live cells is probably an ancient mechanism applied by feeding amoebas[1,2]. However, in contrast to amoebic trogocytosis, which ultimate goal is devouring, and death of cellular pray, the immune cells developed mechanisms of vital trogocytosis or membrane stripping or exchange (Figure 2). During such a process, upon close contact between cells, the recipient cells acquire (borrow) foreign molecules, which they normally do not produce, and the donor cells either completely lose given molecules or the level of these molecules become, at least temporarily, reduced[3,4]. Recent studies indicate that the membrane exchange between various cell types, including immune cells, is a much more common and frequent phenomenon than previously thought. The in vitro and in vivo studies in different model systems showed that upon disengagement from immunological synapse with antigen presenting cells (APCs) or from a direct contact with other target cells, the activated CD4+, CD8+ T cells, regulatory T cells, helper T cells, macrophages, B cells, monocytes, granulocytes and natural killer (NK) cells, are able to internalize fragments of APCs/target cell membrane together with monoclonal antibodies, antigens, ligands, major histocompatibility complex (MHC), adhesion or co-stimulatory molecules[5-18]. For example, studies of Baba et al[19] showed that OX40 ligand (OX40L) expressed by COS-1 cells is transferred to CD4+ T (OX40L-, OX40+) cells, and that the acquired OX40L is functionally active. Other studies showed that the trogocytic acquisition of m157 (the murine cytomegalovirus-encoded ligand for the Ly49H-activating receptor) from target cells regulates NK cells function making them hypo-responsive both in vivo and in vitro[8]. In contrast, acquisition of anti-CD19 chimeric antigen receptors by NK cells enhances their cytotoxicity against the B-cell acute lymphoblastic leukemia cells[7]. Trogocytosis can also lead to acquisition of the MHC complexes by the non-APCs, which in turn may reinforce and/or propagate immune response, and activate or regulate T cells[4]. There are indications that trogocytosis/membrane internalization depends on GTPase TC21 and RhoG-dependent phagocytosis pathway (Figure 2)[16,20]. Membrane internalization not only leads to the acquisition of novel qualities by recipient cells but may also down-regulates the MHC/antigen/co-stimulatory molecules level in bestower APCs[11,16]. There are also instances of multicellular exchange and serial trogocytosis when immune cells acquire novel molecules from multiple sources and then transfer them to other recipient cells. For example the membrane bound molecules from multiple cancer cells can be acquired by CD4+ and CD8+ T cells and monocytes through multiple trogocytosis[21]. It has been shown that monocytes are able to transfer these molecules to other T cells[21]. Thus, trogocytosis/membrane exchange/stripping leads to acquisition/depletion of molecules and their cognate functions in recipient/donor cell, and ultimately modify or modulate an immune response (Figure 2)[3,4,6,18,22,23]. Trogocytosis and its outcomes can be either beneficial or harmful for the organism. Depending on circumstances and cell partners involved the trogocytosis may either promote or prevent development of various pathological conditions or diseases. For example trogocytosis is involved in the ablation of red blood cells in autoimmune hemolytic anemia[24] but when it removes antibodies binding to self-antigens it can prevent autoimmune diseases[25]. Another example of harmful trogocytosis is “oncologic trogocytosis” occurring between ovarian epithelial cancer cells and stromal cells allows cancer cells to acquire multiple drug resistance protein and thus chemoresitance[26].

Figure 2
Figure 2 Trogocytosis/ membrane exchange/stripping. A: During trogocytosis the recipient cell acquires membrane bound molecules from donor cell. The process of internalization of donor cell membrane is similar to phagocytosis and depends on actin ring contraction and small GTPases; B: During APC/T cell interaction the T cell may acquire MHC/peptide complexes and co-stimulatory molecules. Subsequently, such T cells can prime/activate naïve T cells in the absence of APCs, and/or by interacting with activated T cells lead to propagation of immune response[16,20]. APC: Antigen presenting cell; MHC: Major histocompatibility complex.
EXTRACELLULAR TRAPS

Extracellular traps (ETs) were discovered in 2004 in neutrophils and thus have been named Neutrophil Extracellular Traps (NETs)[27]. The process of ETs and NETs formation is called ETosis and NETosis, respectively. ETs consist of filamentous network of chromosomal and/or mitochondrial DNA, which is released from the cell after the break of nuclear/mitochondrial membrane (Figure 3). Because the process of ETosis involves nuclear/mitochondrial/plasma membrane breakage it usually leads to a suicidal, distinct from apoptosis or necrosis, death of ETs’ producing cells[28]. However, there are instances of non-suicidal (vital) NETosis, when nucleus-deprived neutrophils retain motility and chemotactic and phagocytic functions[29,30]. Another example is the vital mitochondrial NETosis when neutrophils primed, for example, with granulocyte/macrophage colony-stimulating factorand stimulated with short-term toll-like receptor 4 or complement factor 5a receptor retain intact nucleus and produce NETs containing exclusively mitochondrial DNA[31]. Besides DNA, the ETs contain various histones (which by themselves have antimicrobial properties) and a plethora of antimicrobial enzymes (Figure 3)[32-35]. The main role of ETs is the immobilization of microbes, which prevents dissemination, and exposing them to a high concentration of antimicrobial agents. Interestingly, many microbes developed defense mechanisms allowing them to escape from or neutralize ETs or ETs producing cells. For example Staphylococcus and Vibrio cholera produce endonucleases, which digest NETs, or/and convert NETs’ DNA into toxic metabolite (deoxyadenosine), which induces apoptosis and promotes death of immune cells[35-37]. The fact that ETosis occurs in many different cell types, and not only in vertebrates but also in invertebrates and plants, suggests that ETs are one of the primordial and evolutionary ancient mechanism of host defense. Studies of molecular pathways involved in initiation and execution of NETosis indicate that stimulation with microbes, inflammatory molecules or endogenous inducers leads, via protein kinase C and NADPH oxidase, to the production of reactive oxygen species and nitrix oxide[35]. These, in turn, induce nuclear/mitochondrial/granule membrane rapture, followed by proteolytic cleavage, deamination (citrullination) of histones, chromatin decondensation and eventual release of NETs[38]. It has been shown that besides consistent presence of DNA the other components of ETs vary, as they are stimulatory signal-specific and cell type-specific[32]. Although ETs play beneficial role in host defense, the presence of DNA and various enzymes makes ETs harmful, especially if they persist for long period of time; they become a very potent inducer of autoimmune response and various pathological conditions, such as lupus, psoriasis, vasculitis, rheumatoid arthritis, type I diabetes, allergic asthma and deep-vein thrombosis (Figure 3)[33,35,39]. It has been also shown that, at least in vitro, ETs influence the behavior of immune cells. NETs are able to down regulate lipopolysaccharide-induced activation of monocyte-derived dendritic cells, inhibit their capacity to activate proliferation of CD4+ T lymphocytes and to polarize naïve CD4+ T cells toward Th1/Th17 phenotypes, promoting Th2 response instead[40]. In addition, prolonged exposure to NETs can induce macrophage and dendritic cells death, which may limit ongoing inflammation[41]. However, it is still unknown, which components of NETs are responsible for these effects. The fact that persisting ETs can modify molecular and cellular components of immune system indicates that fast clearing of ETs is extremely important for proper functioning of immune response[42]. Recent studies indicate that macrophages serve as such clearing agents. Thus, macrophages seem to have a dual role; they can produce ETs and also remove them through phagocytosis[35]. Macrophage ETs, named METs, were discovered in 2010 in murine RAW 264. 7 (Abelson murine leukemia virus transformed macrophages) cell line and since then have been described in many different macrophage types[35,43]. In contrast to the neutrophils where the NETs formation is their main strategy (neutrophils are short-lived “by design”) the formations of METs in macrophages, which are long-lived cells, is an auxiliary strategy and is (regardless of stimulation) self-limited to less than 25% of total macrophage population[35]. Recently, the eosinophil extracellular traps (EETs) and their role in allergic diseases such as human eosinophilic chronic rhinosinusitis and eosinophilic otitis and eosinophilic esophagitis have been described[44,45]. Eosinophil traps released during local cytolysis contain DNA/histone H1 complex, which form globular fibers ticker than those present in neutrophil-derived traps. The EETs can trap fungi and bacteria and at least in eosinophilic esophagitis (characterized by esophageal epithelial barrier defects) can guard against pathogens infiltration through the impaired esophageal wall[45].

Figure 3
Figure 3 Extracellular traps formation and side effects. Various external or internal inducers may lead to the breakage of nuclear or mitochondrial (or both) membranes and release of extracellular traps (ETs). The ETs contain network of nuclear/mitochondrial DNA (blue/green), antimicrobial compounds (yellow) such as LL37, myeoloperoxidase (lysosomal protein) and elastase (chymotrypsin-like protease), and deiminated (citrullinatied) histones (red H). Under normal circumstances the ETs are promptly removed by macrophages, however if the ETs persist they can lead to inflammatory and autoimmune response[31,32].
EXOSOMAL AND ECTOSOMAL/MICROPARTICLE SIGNALING

Exosomes are small (30-150 nm) endocytic membranous vesicles, which are produced by various cell types including immune cells[46]. They were discovered over 30 years ago and for many years they were believed to be the non-functional cell debris or debris disposal vehicles. However, over the last several years numerous studies have proven that exosomes are an important component of intercellular communication circuit and as such play a crucial role in initiation and/or modulation of immune response[46-54]. Exosomes form inside the cell through endosome/mulitvesicular body (MVB) pathway in which they acquire various cytoplasmic, membrane bound and/or nuclear components (Figure 4). Fully matured exosomes released (via exocytosis) from a founder cells deliver, sometimes over long distances, various molecules to their targets. Exososmes may contain a variety of molecules such as: (1) genetic material: retrotransposomal DNA, mitochondrial DNA, mRNAs, miRNAs, rRNA, tRNA; (2) lipids; and (3) proteins: Cytoskeletal proteins, heat shock proteins, channels and transporters, adhesion proteins, tetraspanis and various receptors (Table 1), listed in Exocarta database http://www.exocarta.org[49,55-61]. There are many studies showing how exososmal signaling can influence and modify immune cells and immune response. For example the exosomes released by B cells and dendritic cells contain functional MHC - antigenic peptide complexes, which induce adaptive immune responses in vitro and in vivo[62,63]. Andreola et al[64], showed that death-inducing Fas ligand - bearing exosomes secreted by tumor cells induce lymphocyte apoptosis, which in turn suppress anti-tumor response. In addition, other studies showed that exosomes derived from B lymphocytes expressing FasL can kill T helper (TH) lymphocytes[65], and that antigen-specific suppression of immune response is exerted by microRNA-150 (miRNA-150)-containing exosomes derived from T CD8+ suppressor (Ts) cells[66]. There are also studies showing that exosomes participate in the signaling between pathogens and immune cells. For example exosomes derived from Schistosoma japonicum worm induce macrophage differentiation into M1 subtype[67] and Leishmania-derived exosomes deliver Leishmania specific molecules into the host macrophages and induce secretion of IL-8[68]. Bhatnagar et al[69] showed that pathogen-associated molecular patterns (PAMPs)-rich exosomes secreted from macrophages infected with various mycobacteria are able to stimulate proinflammatory response in naïve macrophages, and when transferred into mice they stimulate synthesis of IL-12 and TNF-α and promote infiltration of lungs with neutrophils and macrophages. Authors suggest that PAMPs-containing exosomes play a major role in immune surveillance[69]. Because exosomes’ content is cell/pathogen specific and they are able to carry and deliver biologically active molecules to the target cells there is also tremendous interest in application of exosomes as biomarkers and the custom engineered exosomes as an immune and anti-cancer therapeutics[70-75].

Table 1 Molecular content of exosomes[49,55-61,74,75].
Nucleic acidsExosome lumen proteinsExosome membrane proteinsLipids
mRNA miRNA tRNA rRNA mitochondrial DNA retrotransposonsActin Cofillin GAPDH Hsp70 Rab TubulinAnnexins Channels EGFR FasL I-CAM1 Integrins LBPA/CD63 LAMP1/2 MHC PD-1L Tetraspanins Tsg 101Cholesterol Diglycerides Eicosanoids fatty acids Gangliosides Lyso-phosphatidylcholine Lyso-bis phosphatidic acid Phosphatidylcholine phosp hatidylethanolamine Phosphatidylserine phosphatidylinositol Sphingomyelin
Figure 4
Figure 4 Exosome pathway. Exosome forms through endocytosis, which starts from the invagination of clathrin coated domain of plasma membrane (coated pit) bearing the receptors and other membrane-bound molecules. After entering cell interior, the coated vesicle loses clathrin coat and becomes the endosome. Subsequently, after acquiring variety of other molecules from Golgi apparatus and cytoplasm, the endosome membrane undergoes inward budding resulting in the formation of multivesiular multivesicular body (MVB) containing exososmes. Ultimately, the MVB fuses with plasma membrane and in the process of exocytosis releases exosomes outside the cell. The exososmes either fuse with the membrane of neighboring target cell or enter blood stream to be transported to distant targets. In the alternative outcome (not shown here), which serves as the pathway degradation pathway, the MVB fuses with lysosomes, which degrade its content[67,69].

Besides exosomes, various cells are able to release another type of membranous vesicles called the ectosomes/microparticles (MPs). The MPs are 0.2-2 mm in diameter and unlike exosomes they bud off the plasma membrane without the involvement of endosome/MVB pathway. The MPs contain a variety of bioactive molecules such as procoagulation compounds (for example P-selectin glycoprotein ligand-1 and tissue factor TF) and/or oncogenic proteins, mRNAs and micro RNAs[76-78]. Similar to exososmes, the circulating MPs may promote/inhibit inflammation, immune response, resistance to chemotherapeutics or activate oncogenic pathways[76-78].

CONCLUSION

One of the most fascinating aspects of noncanonical signaling is the fact that its cellular processes such as trogocytosis/membrane exchange, ETs, exosomes and microparticles are evolutionary ancient (amoebic trogocytsosis, ETs in acoelomate) and conserved in plants, invertebrates and vertebrates[79]. This indicates that noncanonical processes served as the primordial defense mechanisms and canonical signaling had developed later in evolution adding a new and more sophisticated quality to the ancient safeguards. Ironically, the existence of these ancient noncanonical pathways has been discovered much latter than canonical pathways, and only in recent decade they have been recognized as an extremely important regulators of innate and adaptive immunity and inflammatory responses.

Footnotes

P- Reviewer: Li W, Ramirez GA, Sakkas L, Vermi W S- Editor: Ji FF L- Editor: A E- Editor: Wu HL

References
1.  Ralston KS. Chew on this: amoebic trogocytosis and host cell killing by Entamoeba histolytica. Trends Parasitol. 2015;31:442-452.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 25]  [Cited by in F6Publishing: 25]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
2.  Ralston KS, Solga MD, Mackey-Lawrence NM, Somlata A, Petri WA. Trogocytosis by Entamoeba histolytica contributes to cell killing and tissue invasion. Nature. 2014;508:526-530.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 151]  [Cited by in F6Publishing: 149]  [Article Influence: 14.9]  [Reference Citation Analysis (0)]
3.  Joly E, Hudrisier D. What is trogocytosis and what is its purpose? Nat Immunol. 2003;4:815.  [PubMed]  [DOI]  [Cited in This Article: ]
4.  Nakayama M. Antigen Presentation by MHC-Dressed Cells. Front Immunol. 2014;5:672.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 50]  [Cited by in F6Publishing: 83]  [Article Influence: 9.2]  [Reference Citation Analysis (0)]
5.  Pham T, Mero P, Booth JW. Dynamics of macrophage trogocytosis of rituximab-coated B cells. PLoS One. 2011;6:e14498.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 39]  [Cited by in F6Publishing: 41]  [Article Influence: 3.2]  [Reference Citation Analysis (0)]
6.  Taylor RP, Lindorfer MA. Fcγ-receptor-mediated trogocytosis impacts mAb-based therapies: historical precedence and recent developments. Blood. 2015;125:762-766.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 99]  [Cited by in F6Publishing: 103]  [Article Influence: 10.3]  [Reference Citation Analysis (0)]
7.  Cho FN, Chang TH, Shu CW, Ko MC, Liao SK, Wu KH, Yu MS, Lin SJ, Hong YC, Chen CH. Enhanced cytotoxicity of natural killer cells following the acquisition of chimeric antigen receptors through trogocytosis. PLoS One. 2014;9:e109352.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 26]  [Cited by in F6Publishing: 27]  [Article Influence: 2.7]  [Reference Citation Analysis (0)]
8.  Miner CA, Giri TK, Meyer CE, Shabsovich M, Tripathy SK. Acquisition of activation receptor ligand by trogocytosis renders NK cells hyporesponsive. J Immunol. 2015;194:1945-1953.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 28]  [Cited by in F6Publishing: 27]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
9.  Huang JF, Yang Y, Sepulveda H, Shi W, Hwang I, Peterson PA, Jackson MR, Sprent J, Cai Z. TCR-Mediated internalization of peptide-MHC complexes acquired by T cells. Science. 1999;286:952-954.  [PubMed]  [DOI]  [Cited in This Article: ]
10.  Hwang I, Huang JF, Kishimoto H, Brunmark A, Peterson PA, Jackson MR, Surh CD, Cai Z, Sprent J. T cells can use either T cell receptor or CD28 receptors to absorb and internalize cell surface molecules derived from antigen-presenting cells. J Exp Med. 2000;191:1137-1148.  [PubMed]  [DOI]  [Cited in This Article: ]
11.  Walker MR, Mannie MD. Acquisition of functional MHC class II/peptide complexes by T cells during thymic development and CNS-directed pathogenesis. Cell Immunol. 2002;218:13-25.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 15]  [Cited by in F6Publishing: 16]  [Article Influence: 0.7]  [Reference Citation Analysis (0)]
12.  Kedl RM, Schaefer BC, Kappler JW, Marrack P. T cells down-modulate peptide-MHC complexes on APCs in vivo. Nat Immunol. 2002;3:27-32.  [PubMed]  [DOI]  [Cited in This Article: ]
13.  Wetzel SA, McKeithan TW, Parker DC. Peptide-specific intercellular transfer of MHC class II to CD4+ T cells directly from the immunological synapse upon cellular dissociation. J Immunol. 2005;174:80-89.  [PubMed]  [DOI]  [Cited in This Article: ]
14.  Zhou G, Ding ZC, Fu J, Levitsky HI. Presentation of acquired peptide-MHC class II ligands by CD4+ regulatory T cells or helper cells differentially regulates antigen-specific CD4+ T cell response. J Immunol. 2011;186:2148-2155.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 35]  [Cited by in F6Publishing: 38]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
15.  Romagnoli PA, Premenko-Lanier MF, Loria GD, Altman JD. CD8 T cell memory recall is enhanced by novel direct interactions with CD4 T cells enabled by MHC class II transferred from APCs. PLoS One. 2013;8:e56999.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 23]  [Cited by in F6Publishing: 23]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
16.  Dhainaut M, Moser M. Regulation of immune reactivity by intercellular transfer. Front Immunol. 2014;5:112.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 30]  [Cited by in F6Publishing: 34]  [Article Influence: 3.4]  [Reference Citation Analysis (0)]
17.  Rossi EA, Goldenberg DM, Michel R, Rossi DL, Wallace DJ, Chang CH. Trogocytosis of multiple B-cell surface markers by CD22 targeting with epratuzumab. Blood. 2013;122:3020-3029.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 77]  [Cited by in F6Publishing: 83]  [Article Influence: 7.5]  [Reference Citation Analysis (0)]
18.  Zhu X, Niedermann G. Rapid and efficient transfer of the T cell aging marker CD57 from glioblastoma stem cells to CAR T cells. Oncoscience. 2015;2:476-482.  [PubMed]  [DOI]  [Cited in This Article: ]
19.  Baba E, Takahashi Y, Lichtenfeld J, Tanaka R, Yoshida A, Sugamura K, Yamamoto N, Tanaka Y. Functional CD4 T cells after intercellular molecular transfer of 0X40 ligand. J Immunol. 2001;167:875-883.  [PubMed]  [DOI]  [Cited in This Article: ]
20.  Martínez-Martín N, Fernández-Arenas E, Cemerski S, Delgado P, Turner M, Heuser J, Irvine DJ, Huang B, Bustelo XR, Shaw A. T cell receptor internalization from the immunological synapse is mediated by TC21 and RhoG GTPase-dependent phagocytosis. Immunity. 2011;35:208-222.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 123]  [Cited by in F6Publishing: 143]  [Article Influence: 11.0]  [Reference Citation Analysis (0)]
21.  Alegre E, Howangyin KY, Favier B, Baudhuin J, Lesport E, Daouya M, Gonzalez A, Carosella ED, Lemaoult J. Membrane redistributions through multi-intercellular exchanges and serial trogocytosis. Cell Res. 2010;20:1239-1251.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in F6Publishing: 18]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
22.  LeMaoult J, Caumartin J, Daouya M, Switala M, Rebmann V, Arnulf B, Carosella ED. Trogocytic intercellular membrane exchanges among hematological tumors. J Hematol Oncol. 2015;8:24.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in F6Publishing: 18]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
23.  Uzana R, Eisenberg G, Merims S, Frankenburg S, Pato A, Yefenof E, Engelstein R, Peretz T, Machlenkin A, Lotem M. Human T cell crosstalk is induced by tumor membrane transfer. PLoS One. 2015;10:e0118244.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 8]  [Article Influence: 0.9]  [Reference Citation Analysis (0)]
24.  Chadebech P, Michel M, Janvier D, Yamada K, Copie-Bergman C, Bodivit G, Bensussan A, Fournie JJ, Godeau B, Bierling P. IgA-mediated human autoimmune hemolytic anemia as a result of hemagglutination in the spleen, but independent of complement activation and FcαRI. Blood. 2010;116:4141-4147.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 28]  [Cited by in F6Publishing: 31]  [Article Influence: 2.2]  [Reference Citation Analysis (0)]
25.  Masuda S, Iwasaki S, Tomaru U, Baba T, Katsumata K, Ishizu A. Possible implication of Fc γ receptor-mediated trogocytosis in susceptibility to systemic autoimmune disease. Clin Dev Immunol. 2013;2013:345745.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 7]  [Article Influence: 0.6]  [Reference Citation Analysis (0)]
26.  Rafii A, Mirshahi P, Poupot M, Faussat AM, Simon A, Ducros E, Mery E, Couderc B, Lis R, Capdet J. Oncologic trogocytosis of an original stromal cells induces chemoresistance of ovarian tumours. PLoS One. 2008;3:e3894.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 71]  [Cited by in F6Publishing: 73]  [Article Influence: 4.6]  [Reference Citation Analysis (0)]
27.  Brinkmann V, Reichard U, Goosmann C, Fauler B, Uhlemann Y, Weiss DS, Weinrauch Y, Zychlinsky A. Neutrophil extracellular traps kill bacteria. Science. 2004;303:1532-1535.  [PubMed]  [DOI]  [Cited in This Article: ]
28.  Wartha F, Henriques-Normark B. ETosis: a novel cell death pathway. Sci Signal. 2008;1:pe25.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 105]  [Cited by in F6Publishing: 111]  [Article Influence: 6.9]  [Reference Citation Analysis (0)]
29.  Yipp BG, Kubes P. NETosis: how vital is it? Blood. 2013;122:2784-2794.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 534]  [Cited by in F6Publishing: 623]  [Article Influence: 56.6]  [Reference Citation Analysis (0)]
30.  Zhao W, Fogg DK, Kaplan MJ. A novel image-based quantitative method for the characterization of NETosis. J Immunol Methods. 2015;423:104-110.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 65]  [Cited by in F6Publishing: 69]  [Article Influence: 7.7]  [Reference Citation Analysis (0)]
31.  Yousefi S, Mihalache C, Kozlowski E, Schmid I, Simon HU. Viable neutrophils release mitochondrial DNA to form neutrophil extracellular traps. Cell Death Differ. 2009;16:1438-1444.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 568]  [Cited by in F6Publishing: 664]  [Article Influence: 44.3]  [Reference Citation Analysis (0)]
32.  Xu J, Zhang X, Pelayo R, Monestier M, Ammollo CT, Semeraro F, Taylor FB, Esmon NL, Lupu F, Esmon CT. Extracellular histones are major mediators of death in sepsis. Nat Med. 2009;15:1318-1321.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 987]  [Cited by in F6Publishing: 1086]  [Article Influence: 72.4]  [Reference Citation Analysis (0)]
33.  Kaplan MJ, Radic M. Neutrophil extracellular traps: double-edged swords of innate immunity. J Immunol. 2012;189:2689-2695.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 801]  [Cited by in F6Publishing: 783]  [Article Influence: 65.3]  [Reference Citation Analysis (0)]
34.  Badimon L, Vilahur G. Neutrophil extracellular traps: a new source of tissue factor in atherothrombosis. Eur Heart J. 2015;36:1364-1366.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 20]  [Cited by in F6Publishing: 19]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
35.  Boe DM, Curtis BJ, Chen MM, Ippolito JA, Kovacs EJ. Extracellular traps and macrophages: new roles for the versatile phagocyte. J Leukoc Biol. 2015;97:1023-1035.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 79]  [Cited by in F6Publishing: 80]  [Article Influence: 8.9]  [Reference Citation Analysis (0)]
36.  Seper A, Hosseinzadeh A, Gorkiewicz G, Lichtenegger S, Roier S, Leitner DR, Röhm M, Grutsch A, Reidl J, Urban CF. Vibrio cholerae evades neutrophil extracellular traps by the activity of two extracellular nucleases. PLoS Pathog. 2013;9:e1003614.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 105]  [Cited by in F6Publishing: 97]  [Article Influence: 8.8]  [Reference Citation Analysis (0)]
37.  Thammavongsa V, Missiakas DM, Schneewind O. Staphylococcus aureus degrades neutrophil extracellular traps to promote immune cell death. Science. 2013;342:863-866.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 252]  [Cited by in F6Publishing: 272]  [Article Influence: 24.7]  [Reference Citation Analysis (0)]
38.  Stoiber W, Obermayer A, Steinbacher P, Krautgartner WD. The Role of Reactive Oxygen Species (ROS) in the Formation of Extracellular Traps (ETs) in Humans. Biomolecules. 2015;5:702-723.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 133]  [Cited by in F6Publishing: 163]  [Article Influence: 18.1]  [Reference Citation Analysis (0)]
39.  Pinegin B, Vorobjeva N, Pinegin V. Neutrophil extracellular traps and their role in the development of chronic inflammation and autoimmunity. Autoimmun Rev. 2015;14:633-640.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 96]  [Cited by in F6Publishing: 107]  [Article Influence: 11.9]  [Reference Citation Analysis (0)]
40.  Barrientos L, Bignon A, Gueguen C, de Chaisemartin L, Gorges R, Sandré C, Mascarell L, Balabanian K, Kerdine-Römer S, Pallardy M. Neutrophil extracellular traps downregulate lipopolysaccharide-induced activation of monocyte-derived dendritic cells. J Immunol. 2014;193:5689-5698.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 39]  [Cited by in F6Publishing: 47]  [Article Influence: 4.7]  [Reference Citation Analysis (0)]
41.  Donis-Maturano L, Sánchez-Torres LE, Cerbulo-Vázquez A, Chacón-Salinas R, García-Romo GS, Orozco-Uribe MC, Yam-Puc JC, González-Jiménez MA, Paredes-Vivas YL, Calderón-Amador J. Prolonged exposure to neutrophil extracellular traps can induce mitochondrial damage in macrophages and dendritic cells. Springerplus. 2015;4:161.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in F6Publishing: 19]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
42.  Anjos PM, Fagundes-Netto FS, Volpe CM, Nogueira-Machado JA. Impaired clearance of neutrophils extracellular trap (NET) may induce detrimental tissular effect. Recent Pat Endocr Metab Immune Drug Discov. 2014;8:186-190.  [PubMed]  [DOI]  [Cited in This Article: ]
43.  Chow OA, von Köckritz-Blickwede M, Bright AT, Hensler ME, Zinkernagel AS, Cogen AL, Gallo RL, Monestier M, Wang Y, Glass CK. Statins enhance formation of phagocyte extracellular traps. Cell Host Microbe. 2010;8:445-454.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 282]  [Cited by in F6Publishing: 309]  [Article Influence: 23.8]  [Reference Citation Analysis (0)]
44.  Ueki S, Konno Y, Takeda M, Moritoki Y, Hirokawa M, Matsuwaki Y, Honda K, Ohta N, Yamamoto S, Takagi Y. Eosinophil extracellular trap cell death-derived DNA traps: Their presence in secretions and functional attributes. J Allergy Clin Immunol. 2016;137:258-267.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 134]  [Cited by in F6Publishing: 153]  [Article Influence: 17.0]  [Reference Citation Analysis (0)]
45.  Simon D, Radonjic-Hösli S, Straumann A, Yousefi S, Simon HU. Active eosinophilic esophagitis is characterized by epithelial barrier defects and eosinophil extracellular trap formation. Allergy. 2015;70:443-452.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 95]  [Cited by in F6Publishing: 98]  [Article Influence: 10.9]  [Reference Citation Analysis (0)]
46.  Théry C. Exosomes: secreted vesicles and intercellular communications. F1000 Biol Rep. 2011;3:15.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 583]  [Cited by in F6Publishing: 666]  [Article Influence: 51.2]  [Reference Citation Analysis (0)]
47.  Hegmans JP, Gerber PJ, Lambrecht BN. Exosomes. Methods Mol Biol. 2008;484:97-109.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 20]  [Cited by in F6Publishing: 21]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
48.  Bu N, Wu H, Zhang G, Zhan S, Zhang R, Sun H, Du Y, Yao L, Wang H. Exosomes from Dendritic Cells Loaded with Chaperone-Rich Cell Lysates Elicit a Potent T Cell Immune Response Against Intracranial Glioma in Mice. J Mol Neurosci. 2015;56:631-643.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 41]  [Cited by in F6Publishing: 48]  [Article Influence: 5.3]  [Reference Citation Analysis (0)]
49.  Greening DW, Gopal SK, Xu R, Simpson RJ, Chen W. Exosomes and their roles in immune regulation and cancer. Semin Cell Dev Biol. 2015;40:72-81.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 365]  [Cited by in F6Publishing: 432]  [Article Influence: 48.0]  [Reference Citation Analysis (0)]
50.  Bobrie A, Colombo M, Raposo G, Théry C. Exosome secretion: molecular mechanisms and roles in immune responses. Traffic. 2011;12:1659-1668.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 686]  [Cited by in F6Publishing: 747]  [Article Influence: 57.5]  [Reference Citation Analysis (0)]
51.  Lai FW, Lichty BD, Bowdish DM. Microvesicles: ubiquitous contributors to infection and immunity. J Leukoc Biol. 2015;97:237-245.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 45]  [Cited by in F6Publishing: 47]  [Article Influence: 4.7]  [Reference Citation Analysis (0)]
52.  Agarwal A, Fanelli G, Letizia M, Tung SL, Boardman D, Lechler R, Lombardi G, Smyth LA. Regulatory T cell-derived exosomes: possible therapeutic and diagnostic tools in transplantation. Front Immunol. 2014;5:555.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 53]  [Cited by in F6Publishing: 69]  [Article Influence: 6.9]  [Reference Citation Analysis (0)]
53.  De Jong OG, Van Balkom BW, Schiffelers RM, Bouten CV, Verhaar MC. Extracellular vesicles: potential roles in regenerative medicine. Front Immunol. 2014;5:608.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 161]  [Cited by in F6Publishing: 210]  [Article Influence: 21.0]  [Reference Citation Analysis (0)]
54.  Schorey JS, Cheng Y, Singh PP, Smith VL. Exosomes and other extracellular vesicles in host-pathogen interactions. EMBO Rep. 2015;16:24-43.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 430]  [Cited by in F6Publishing: 489]  [Article Influence: 48.9]  [Reference Citation Analysis (0)]
55.  Mathivanan S, Fahner CJ, Reid GE, Simpson RJ. ExoCarta 2012: database of exosomal proteins, RNA and lipids. Nucleic Acids Res. 2012;40:D1241-D1244.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 697]  [Cited by in F6Publishing: 771]  [Article Influence: 59.3]  [Reference Citation Analysis (0)]
56.  Benito-Martin A, Di Giannatale A, Ceder S, Peinado H. The new deal: a potential role for secreted vesicles in innate immunity and tumor progression. Front Immunol. 2015;6:66.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 71]  [Cited by in F6Publishing: 73]  [Article Influence: 8.1]  [Reference Citation Analysis (0)]
57.  Yoon YJ, Kim OY, Gho YS. Extracellular vesicles as emerging intercellular communicasomes. BMB Rep. 2014;47:531-539.  [PubMed]  [DOI]  [Cited in This Article: ]
58.  Salido-Guadarrama I, Romero-Cordoba S, Peralta-Zaragoza O, Hidalgo-Miranda A, Rodríguez-Dorantes M. MicroRNAs transported by exosomes in body fluids as mediators of intercellular communication in cancer. Onco Targets Ther. 2014;7:1327-1338.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 35]  [Cited by in F6Publishing: 94]  [Article Influence: 9.4]  [Reference Citation Analysis (0)]
59.  Valadi H, Ekström K, Bossios A, Sjöstrand M, Lee JJ, Lötvall JO. Exosome-mediated transfer of mRNAs and microRNAs is a novel mechanism of genetic exchange between cells. Nat Cell Biol. 2007;9:654-659.  [PubMed]  [DOI]  [Cited in This Article: ]
60.  Yao Y, Wei W, Sun J, Chen L, Deng X, Ma L, Hao S. Proteomic analysis of exosomes derived from human lymphoma cells. Eur J Med Res. 2015;20:8.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in F6Publishing: 20]  [Article Influence: 2.2]  [Reference Citation Analysis (0)]
61.  Zhang X, Yuan X, Shi H, Wu L, Qian H, Xu W. Exosomes in cancer: small particle, big player. J Hematol Oncol. 2015;8:83.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 461]  [Cited by in F6Publishing: 532]  [Article Influence: 59.1]  [Reference Citation Analysis (0)]
62.  Raposo G, Nijman HW, Stoorvogel W, Liejendekker R, Harding CV, Melief CJ, Geuze HJ. B lymphocytes secrete antigen-presenting vesicles. J Exp Med. 1996;183:1161-1172.  [PubMed]  [DOI]  [Cited in This Article: ]
63.  Zitvogel L, Regnault A, Lozier A, Wolfers J, Flament C, Tenza D, Ricciardi-Castagnoli P, Raposo G, Amigorena S. Eradication of established murine tumors using a novel cell-free vaccine: dendritic cell-derived exosomes. Nat Med. 1998;4:594-600.  [PubMed]  [DOI]  [Cited in This Article: ]
64.  Andreola G, Rivoltini L, Castelli C, Huber V, Perego P, Deho P, Squarcina P, Accornero P, Lozupone F, Lugini L. Induction of lymphocyte apoptosis by tumor cell secretion of FasL-bearing microvesicles. J Exp Med. 2002;195:1303-1316.  [PubMed]  [DOI]  [Cited in This Article: ]
65.  Lundy SK, Klinker MW, Fox DA. Killer B lymphocytes and their fas ligand positive exosomes as inducers of immune tolerance. Front Immunol. 2015;6:122.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 28]  [Cited by in F6Publishing: 28]  [Article Influence: 3.1]  [Reference Citation Analysis (0)]
66.  Nazimek K, Ptak W, Nowak B, Ptak M, Askenase PW, Bryniarski K. Macrophages play an essential role in antigen-specific immune suppression mediated by T CD8⁺ cell-derived exosomes. Immunology. 2015;146:23-32.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 41]  [Cited by in F6Publishing: 45]  [Article Influence: 5.0]  [Reference Citation Analysis (0)]
67.  Wang L, Li Z, Shen J, Liu Z, Liang J, Wu X, Sun X, Wu Z. Exosome-like vesicles derived by Schistosoma japonicum adult worms mediates M1 type immune- activity of macrophage. Parasitol Res. 2015;114:1865-1873.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 80]  [Cited by in F6Publishing: 88]  [Article Influence: 9.8]  [Reference Citation Analysis (0)]
68.  Silverman JM, Clos J, de’Oliveira CC, Shirvani O, Fang Y, Wang C, Foster LJ, Reiner NE. An exosome-based secretion pathway is responsible for protein export from Leishmania and communication with macrophages. J Cell Sci. 2010;123:842-852.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 319]  [Cited by in F6Publishing: 344]  [Article Influence: 24.6]  [Reference Citation Analysis (0)]
69.  Bhatnagar S, Shinagawa K, Castellino FJ, Schorey JS. Exosomes released from macrophages infected with intracellular pathogens stimulate a proinflammatory response in vitro and in vivo. Blood. 2007;110:3234-3244.  [PubMed]  [DOI]  [Cited in This Article: ]
70.  Kalani A, Tyagi A, Tyagi N. Exosomes: mediators of neurodegeneration, neuroprotection and therapeutics. Mol Neurobiol. 2014;49:590-600.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 216]  [Cited by in F6Publishing: 238]  [Article Influence: 21.6]  [Reference Citation Analysis (0)]
71.  De Toro J, Herschlik L, Waldner C, Mongini C. Emerging roles of exosomes in normal and pathological conditions: new insights for diagnosis and therapeutic applications. Front Immunol. 2015;6:203.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 349]  [Cited by in F6Publishing: 414]  [Article Influence: 46.0]  [Reference Citation Analysis (0)]
72.  Zhang B, Yin Y, Lai RC, Lim SK. Immunotherapeutic potential of extracellular vesicles. Front Immunol. 2014;5:518.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 103]  [Cited by in F6Publishing: 121]  [Article Influence: 12.1]  [Reference Citation Analysis (0)]
73.  Lässer C. Exosomes in diagnostic and therapeutic applications: biomarker, vaccine and RNA interference delivery vehicle. Expert Opin Biol Ther. 2015;15:103-117.  [PubMed]  [DOI]  [Cited in This Article: ]
74.  Waldenström A, Ronquist G. Role of exosomes in myocardial remodeling. Circ Res. 2014;114:315-324.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 95]  [Cited by in F6Publishing: 106]  [Article Influence: 10.6]  [Reference Citation Analysis (0)]
75.  Subra C, Laulagnier K, Perret B, Record M. Exosome lipidomics unravels lipid sorting at the level of multivesicular bodies. Biochimie. 2007;89:205-212.  [PubMed]  [DOI]  [Cited in This Article: ]
76.  Cocucci E, Meldolesi J. Ectosomes and exosomes: shedding the confusion between extracellular vesicles. Trends Cell Biol. 2015;25:364-372.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 786]  [Cited by in F6Publishing: 937]  [Article Influence: 104.1]  [Reference Citation Analysis (0)]
77.  de Souza PS, Faccion RS, Bernardo PS, Maia RC. Membrane microparticles: shedding new light into cancer cell communication. J Cancer Res Clin Oncol. 2015;Aug 19; Epub ahead of print.  [PubMed]  [DOI]  [Cited in This Article: ]
78.  Morel O, Morel N, Jesel L, Freyssinet JM, Toti F. Microparticles: a critical component in the nexus between inflammation, immunity, and thrombosis. Semin Immunopathol. 2011;33:469-486.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 109]  [Cited by in F6Publishing: 115]  [Article Influence: 8.8]  [Reference Citation Analysis (0)]
79.  Robb CT, Dyrynda EA, Gray RD, Rossi AG, Smith VJ. Invertebrate extracellular phagocyte traps show that chromatin is an ancient defence weapon. Nat Commun. 2014;5:4627.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 100]  [Cited by in F6Publishing: 99]  [Article Influence: 9.9]  [Reference Citation Analysis (0)]