Liver-derived extracellular vesicles: A cell by cell overview to isolation and characterization practices

https://doi.org/10.1016/j.bbagen.2020.129559Get rights and content

Highlights

  • Overview of methods for extracellular vesicles (EV) from liver resident cells.

  • EVs from hepatocytes and hepatic stellate cells are more frequently analysed.

  • EVs from cholangiocytes, sinusoidal endothelial cells, Kupffer cells, stem cells too.

  • Discussion of transferability between human, mouse, rat cells in vitro and ex vivo.

  • Focus on method and reporting standardization (EV yield, storage, cell viability).

Abstract

Background

Extracellular vesicles (EVs) are a diverse group of membrane-bound nanovesicles potentially released by every cell. With the liver's unique ensemble of cells and its fundamental physiological tasks, elucidating the role of EV-mediated hepatic cellular crosstalk and their role in different pathologies has been gaining the attention of many scientists.

Scope of review

The present review shifts the perspective into practice: we aim to critically discuss the methods used to purify and to biochemically analyse EVs from specific liver resident cells, including hepatocytes, hepatic stellate cells, cholangiocytes, liver sinusoidal endothelial cells, Kupffer cells, liver stem cells. The review offers a reference guide to current approaches.

Major conclusions

Strategies for EV isolation and characterization are as varied as the research groups performing them. We present main advantages and disadvantages for the methods, highlighting common causes for concern, such as FBS handling, reporting of cell viability, EV yield and storage, differences in differential centrifugations, suboptimal method descriptions, and method transferability. We both looked at how adaptable the research between human and rodent cells in vitro is, and also assessed how well either of them translates to ex vivo settings.

General significance

We reviewed methodological practices for the isolation and analysis of liver-derived EVs, making a cell type specific user guide that shows where to start, what has worked so far and to what extent. We critically discussed room for improvement, placing a particular focus on working towards a potential standardization of methods.

Introduction

Extracellular vesicles (EVs) is a collective term referring to a diverse group of small membrane vesicles virtually released by all cell types, and which are generally being categorized according to their biogenesis: [1,2] apoptotic bodies are blebs of the dying cell membrane and have the broadest size range; microvesicles, sometimes referred to as microparticles or ectosomes, stem from the outward budding of the cellular membrane. Exosomes, which tend to be the smallest subpopulation, are released into the extracellular space after multivesicular bodies (MVB) fuse with the cell membrane (Fig. 1). It is still not really possible to isolate one subpopulation from the others, and while it is believed that they may display biomolecules that are enriched to different extents, their overlapping composition, density and size, as well as the absence of subtype-specific markers still make for a considerable challenge. [3,4] The recent discovery of their role in intercellular communication captivated the attention of a growing number of scientists anticipating the enormous potential of EVs in the fields of diagnostics and drug delivery. [5,6] For some pathological dispositions EVs can be applied as liquid biopsies, and that has sparked a lot of interest from a diagnostic perspective. EVs are enriched in selected biomolecules, they are intrinsically equipped to protect their cargo from degradation, and while their complexity offers many characterization opportunities (see Fig. 1), they are still simpler to analyse than total blood or serum samples. The interest in EVs as drug delivery systems stems from their potential advantages over synthetic carriers: they are bioavailable, biocompatible, resistant to RNAases and proteases (high physicochemical stability), capable of long-distance communication and they are intrinsically able to interact with cells even across species. [[7], [8], [9]].

The increasing scientific interest in EVs [10] has led to the establishment of dedicated, open access databases such as Vesiclepedia, [11] EVpedia [12] and EV-TRACK, [13] which are being regularly updated. The majority of the information is concerning proteins, whereas EV-TRACK sets itself apart by focusing on method transparency.

As reviewed elsewhere, [14,15] there are significant challenges (small yields, co-purification of contaminants, etc.) in finding the most efficient protocols for the isolation and sufficient characterization of EVs. We will discuss them in an effort to encourage sharing the current knowledge of the more practical scientific trends in those areas, starting with the present review on liver-derived EVs.

The liver is a large and complex organ responsible for a variety of essential physiological tasks including protein synthesis, lipid storage regulation, xenobiotic detoxification, and offering support to both immunological activity and food digestion. [16] It is difficult to understate its importance. When organ function is compromised, hepatic diseases are directly responsible for as many as 2 million deaths per year: liver cirrhosis alone kills 1.16 million people every year, and hepatocellular carcinoma accounts for the death of 788,000 more, meaning that combined they cause 3.5% of all yearly deaths in the world. [17,18] The global health burden of liver associated conditions is not sufficiently addressed as of yet. [19] With its unique ensemble of diverse cells (see Fig. 2), the liver offers the opportunity to study intra- and inter-cellular communication. Elucidating the role of EV-mediated hepatic cellular crosstalk has gained the attention of many researchers, who have been able to review its critical role in both health and disease, pointing to differences in the set of EVs that are released, especially in the case of tumors. [[20], [21], [22], [23], [24], [25]].

The present review shifts the perspective into practice: we aim to critically consider the methods used to purify and to biochemically analyse EVs from specific cells, offering a user/reference guide to current practices (see Table 1). The most common and (when applicable) the most original/promising protocols will be examined in order to provide a discussion frame. Diving into the different methodological approaches, we will also highlight limitations and possible inconsistencies. Finally, the (interspecies) method transferability and the translational applicability of these practices will be examined. We will look within the individual cell specific sections if the same or similar methods could be transferred to different in vitro systems (rodent cells, co-cultures), but we will also explore the practical strategies for EV-isolation and characterization that were applied ex vivo and in the clinic from patients with liver-related conditions (Section 8). It is worth noting, that we are only covering method employment and it is not our role to comment on the quality of the results gained from them. As to the nomenclature, we strived to keep it as it was applied in the referenced manuscripts, only changing it to “EVs” if we needed to paraphrase, since it is the preferred generic term. [1,2].

Section snippets

EVs from hepatocytes

Hepatocytes are the most abundant cells in the liver, comprising about 70–80% of its mass, they are dedicated to protein synthesis (serum albumin, transferrin and other glycoproteins), while also being the main site for glycolysis and only site for bile salts production in the body. [[222], [223], [224]] Another essential task relying on hepatocytes is detoxification: they metabolize both exogenous (e.g., drugs, toxins) and endogenous (e.g., steroids) compounds. [225,226] More notable still is

EVs from hepatic stellate cells

Hepatic stellate cells reside in the space of Disse (Fig. 2), between hepatocytes and endothelial cells, and are mainly responsible for storing vitamin A in cytoplasmic lipid droplets. [238] Upon liver injury, however, they undergo transdifferentiation into a myofibroblast-like state, i.e., they become activated, progressively lose their lipid droplets and start promoting fibrogenesis, most notably by deposition of excessive and collagen rich extracellular matrix. [239,240] When the injuries

EVs from cholangiocytes

Cholangiocytes are epithelial cells lining the bile ducts (Fig. 2), which can differ in size and morphology just as the bile duct tree itself does. Their main physiological role is the modification of the bile coming from the liver while it is being transported along the biliary ducts into the intestine. [[247], [248], [249], [250]] Pathologies directly associated with cholangiocytes include primary biliary cholangitis, and primary sclerosing cholangitis, for the latter of which liver

EVs from liver sinusoidal endothelial cells

Liver sinusoidal endothelial cells (LSECs) form the fenestrated endothelial layer at the interface between cells in the blood and the HSCs in the space of Disse. LSECs act as efficient pinocytotic scavengers for particles that are smaller than 0.2 μm, making them the central players in clearing the body of blood-borne viruses. [253,254] When working with EVs from LSECs, researchers were interested in infections caused by hepatitis B and C viruses (HBV and HCV).

EVs from Kupffer cells

The resident macrophages in the liver, found in the hepatic sinusoid, are known as Kupffer cells. [258] Since the liver is frequently in contact with exogenous material, suppressing “unwanted” immune responses is essential. Kupffer cells have been shown to provide anti-inflammatory signals that allow homeostatic immunological tolerance under healthy conditions. As part of the innate immune system, these macrophages phagocyte invading pathogens and play a critical role for the initiation of

EVs from liver stem cells

Several studies have suggested the presence of liver-resident stem cells, which along with hepatocytes contribute to liver regeneration, but it remains a controversial topic, as reviewed elsewhere. [264] The identification of oval cells and their role in liver regeneration contributes to our current understanding of the process, [265] more confidently so after establishing their precursor role to hepatocytes, [266] and their localization in the canals of Hering [267] between bile capillaries

EVs from clinical settings

There has been extensive research delving into the diagnostic potential of EVs in the context of liver-associated conditions, albeit mostly looking into circulating vesicles, both in human patients [[100], [101], [102], [103], [104], [105], [106], [107], [108], [109], [110], [111], [112], [113], [114], [115], [116], [117], [118], [119], [120], [121], [122], [123], [124], [125], [126], [127], [128], [129], [130], [131], [132], [133], [134], [135], [136], [137], [138], [139], [140], [141], [142],

Final general remarks

Strategies for liver-derived EV isolation and characterization are as varied as the research groups tackling the challenge. While the main advantages and disadvantages for each approach have been presented, we would like to highlight some of the key aspects that emerged.

Conclusions and perspectives

We reviewed the current methodological practices for the isolation and analysis of liver derived EVs, making a cell type specific user guide. Liver-derived EVs are gaining attention as a research topic, and there are already a few research papers addressing EVs from every liver resident cell type. Much work has already been published, but a rigorous standardization is needed. We moreover highlighted common causes for concern and critically reviewed room for improvement: the bigger issue that

Declaration of Competing Interest

The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper.

Acknowledgements

G.F. is a holder of a NanoMatFutur grant from the Federal Ministry of Research and Education (13XP5029A). The Phospholipids Research Center is gratefully acknowledged for its support.

References (283)

  • N.R. Kapoor et al.

    The HBx gene of hepatitis B virus can influence hepatic microenvironment via exosomes by transferring its mRNA and protein

    Virus Res.

    (2017)
  • E. Rodríguez-Suárez et al.

    Quantitative proteomic analysis of hepatocyte-secreted extracellular vesicles reveals candidate markers for liver toxicity

    J. Proteome

    (2014)
  • L. Duan et al.

    Role of extracellular vesicles in release of protein adducts after acetaminophen-induced liver injury in mice and humans

    Toxicol. Lett.

    (2019)
  • P. Hirsova et al.

    Lipotoxic lethal and sublethal stress signaling in hepatocytes: relevance to NASH pathogenesis

    J. Lipid Res.

    (2016)
  • H. Nojima et al.

    Hepatocyte exosomes mediate liver repair and regeneration via sphingosine-1-phosphate

    J. Hepatol.

    (2016)
  • E. Kakazu et al.

    Hepatocytes release ceramide-enriched pro-inflammatory extracellular vesicles in an IRE1α-dependent manner

    J. Lipid Res.

    (2016)
  • A. Charrier et al.

    Exosomes mediate intercellular transfer of pro-fibrogenic connective tissue growth factor (CCN2) between hepatic stellate cells, the principal fibrotic cells in the liver

    Surg. (United States).

    (2014)
  • L. Chen et al.

    Fibrogenic signaling is suppressed in hepatic stellate cells through targeting of connective tissue growth factor (CCN2) by cellular or exosomal microRNA-199a-5p

    Am. J. Pathol.

    (2016)
  • R. Wang et al.

    Exosome adherence and internalization by hepatic stellate cells triggers sphingosine 1-phosphate-dependent migration

    J. Biol. Chem.

    (2015)
  • B. Saha et al.

    MicroRNA cargo of extracellular vesicles from alcohol-exposed monocytes signals naive monocytes to differentiate into M2 macrophages

    J. Biol. Chem.

    (2016)
  • C. Théry et al.

    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

    J. Extracell. Vesicles

    (2018)
  • K.W. Witwer et al.

    Extracellular vesicles or exosomes? On primacy, precision, and popularity influencing a choice of nomenclature

    J. Extracell. Vesicles

    (2019)
  • G. van Niel et al.

    Shedding light on the cell biology of extracellular vesicles

    Nat. Rev. Mol. Cell Biol.

    (2018)
  • P.D. Stahl et al.

    Exosomes and extracellular vesicles: the path forward

    Essays Biochem.

    (2018)
  • O.G. de Jong et al.

    Drug delivery with extracellular vesicles: from imagination to innovation

    Acc. Chem. Res.

    (2019)
  • A. Goes et al.

    Biogenic and biomimetic carriers as versatile transporters to treat infections

    ACS Infect. Dis.

    (2018)
  • Y. Kawamura et al.

    Extracellular vesicles as trans-genomic agents: emerging roles in disease and evolution

    Cancer Sci.

    (2017)
  • R. Ono et al.

    Exosome-mediated horizontal gene transfer occurs in double-strand break repair during genome editing

    Commun. Biol.

    (2019)
  • E. Woith et al.

    Extracellular vesicles—connecting kingdoms

    Int. J. Mol. Sci.

    (2019)
  • B. Wang et al.

    The state of exosomes research: a global visualized analysis

    Biomed. Res. Int.

    (2019)
  • H. Kalra et al.

    Vesiclepedia: a compendium for extracellular vesicles with continuous community annotation

    PLoS Biol.

    (2012)
  • D.-K. Kim et al.

    EVpedia: an integrated database of high-throughput data for systemic analyses of extracellular vesicles

    J. Extracell. Vesicles

    (2013)
  • J. Van Deun et al.

    EV-TRACK: transparent reporting and centralizing knowledge in extracellular vesicle research

    Nat. Methods

    (2017)
  • M.I. Ramirez et al.

    Technical challenges of working with extracellular vesicles

    Nanoscale.

    (2018)
  • J. Jablonska et al.

    Challenges in the isolation and proteomic analysis of cancer exosomes—implications for translational research

    Proteomes

    (2019)
  • N. Ndugga et al.

    Disparities between research attention and burden in liver diseases: implications on uneven advances in pharmacological therapies in Europe and the USA

    BMJ Open

    (2017)
  • D. Povero et al.

    Circulating extracellular vesicles with specific proteome and liver microRNAs are potential biomarkers for liver injury in experimental fatty liver disease

    PLoS One

    (2014)
  • S. Urban et al.

    Extracellular vesicles in liver diseases: diagnostic, prognostic, and therapeutic application

    Semin. Liver Dis.

    (2019)
  • S. Sung et al.

    Liver-derived exosomes and their implications in liver pathobiology

    Int. J. Mol. Sci.

    (2018)
  • K. Sato et al.

    Intercellular communication between hepatic cells in liver diseases

    Int. J. Mol. Sci.

    (2019)
  • M.A. Angulo et al.

    Metabolic nano-machines: extracellular vesicles containing active enzymes and their contribution to liver diseases

    Curr. Pathobiol. Rep.

    (2019)
  • A.L. Mescher et al.

    Junqueira’s Basic Histology: Text and Atlas

    (2010)
  • M. Berardocco et al.

    RNA-seq reveals distinctive RNA profiles of small extracellular vesicles from different human liver cancer cell lines

    Oncotarget

    (2017)
  • T. Kogure et al.

    Intercellular nanovesicle-mediated microRNA transfer: a mechanism of environmental modulation of hepatocellular cancer cell growth

    Hepatology

    (2011)
  • T. Kogure et al.

    Extracellular vesicle-mediated transfer of a novel long noncoding RNA TUC339: a mechanism of intercellular signaling in human hepatocellular cancer

    Genes Cancer

    (2013)
  • W.-H. Xiao et al.

    Effect of 5-aza-2′-deoxycytidine on immune-associated proteins in exosomes from hepatoma

    World J. Gastroenterol.

    (2010)
  • W. Xiao et al.

    Effects of the epigenetic drug MS-275 on the release and function of exosome-related immune molecules in hepatocellular carcinoma cells

    Eur. J. Med. Res.

    (2013)
  • Y.-E. Cho et al.

    Increased liver-specific proteins in circulating extracellular vesicles as potential biomarkers for drug- and alcohol-induced liver injury

    PLoS One

    (2017)
  • Q. Rao et al.

    Tumor-derived exosomes elicit tumor suppression in murine hepatocellular carcinoma models and humans in vitro

    Hepatology

    (2016)
  • J. Wei et al.

    Vps4A functions as a tumor suppressor by regulating the secretion and uptake of exosomal microRNAs in human hepatoma cells

    Hepatology

    (2015)
  • Cited by (7)

    • Hepatic stellate cell-derived exosomes modulate macrophage inflammatory response

      2021, Experimental Cell Research
      Citation Excerpt :

      Following EXO release, the effect of EXOs on target cells depends on the specific components contained within the EXO, factors that can comprise of lipids, miRs, mRNAs, and proteins [7,8]. Previous studies report significant roles for EXOs released from hepatocytes [31,32], macrophages [25], cholangiocytes [33], and hepatoma cells [34] during the initiation and acceleration of HSC activation [35]. To our knowledge, our data represent the first reports of a functional role for HSC-EXOs in regulating macrophage function, implying a dynamic interaction within hepatic cell populations during HSC activation/fibrosis.

    • Extracellular vesicles in hepatology: Physiological role, involvement in pathogenesis, and therapeutic opportunities

      2021, Pharmacology and Therapeutics
      Citation Excerpt :

      As a conclusion, the presented studies reassure the notion of EVs as a specific messenger involved in the cellular crosstalk that occurs in the liver. More comprehensive review of the cargo content for each cell type can be found in (Zivko, Fuhrmann, & Luciani, 2020). In the last decade, it has been observed that hepatocyte-derived EVs play an essential role in the diagnosis of different diseases (reviewed in (Sato, Meng, Glaser, & Alpini, 2016)).

    • Macrophages in pancreatitis: Mechanisms and therapeutic potential

      2020, Biomedicine and Pharmacotherapy
      Citation Excerpt :

      During AP, hepatocytes, Kupffer cells, and other specific hepatocytes can remove exosomes produced by the pancreas that enter the liver through the portal vein system, but secrete liver-derived exosomes into the blood circulation. These exosomes furthermore act like pro-inflammatory cytokines to activate alveolar macrophages into pro-inflammatory M1 macrophages, thereby exacerbating lung injury [74,75]. Collectively, during AP, digestive enzymes, cytokines, chemokines, exosomes, and other inflammatory substances released from the pancreas may enter the liver through the portal vein system before being diluted into the blood circulation.

    View all citing articles on Scopus
    View full text