Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 -- 2455 2023-01-09 09:19:37 |
2 format correct + 1 word(s) 2456 2023-01-10 05:22:20 | |
3 format correct Meta information modification 2456 2023-01-10 05:23:45 | |
4 format correct + 1 word(s) 2457 2023-01-10 05:25:36 | |
5 format correct + 4 word(s) 2461 2023-01-10 05:27:17 | |
6 format correct Meta information modification 2461 2023-01-11 05:18:11 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Muñoz-Hernández, R.;  Rojas, �.;  Gato, S.;  Gallego, J.;  Gil-Gómez, A.;  Castro, M.J.;  Ampuero, J.;  Romero-Gómez, M. Extracellular Vesicles as a Biomarker in Liver Disease. Encyclopedia. Available online: https://encyclopedia.pub/entry/39890 (accessed on 01 July 2024).
Muñoz-Hernández R,  Rojas �,  Gato S,  Gallego J,  Gil-Gómez A,  Castro MJ, et al. Extracellular Vesicles as a Biomarker in Liver Disease. Encyclopedia. Available at: https://encyclopedia.pub/entry/39890. Accessed July 01, 2024.
Muñoz-Hernández, Rocío, Ángela Rojas, Sheila Gato, Javier Gallego, Antonio Gil-Gómez, María José Castro, Javier Ampuero, Manuel Romero-Gómez. "Extracellular Vesicles as a Biomarker in Liver Disease" Encyclopedia, https://encyclopedia.pub/entry/39890 (accessed July 01, 2024).
Muñoz-Hernández, R.,  Rojas, �.,  Gato, S.,  Gallego, J.,  Gil-Gómez, A.,  Castro, M.J.,  Ampuero, J., & Romero-Gómez, M. (2023, January 09). Extracellular Vesicles as a Biomarker in Liver Disease. In Encyclopedia. https://encyclopedia.pub/entry/39890
Muñoz-Hernández, Rocío, et al. "Extracellular Vesicles as a Biomarker in Liver Disease." Encyclopedia. Web. 09 January, 2023.
Extracellular Vesicles as a Biomarker in Liver Disease
Edit

Extracellular vesicles (EVs) are membrane-derived vesicles released by a variety of cell types, including hepatocytes, hepatic stellate cells, and immune cells in normal and pathological conditions. Depending on their biogenesis, there is a complex repertoire of EVs that differ in size and origin. EVs can carry lipids, proteins, coding and non-coding RNAs, and mitochondrial DNA causing alterations to the recipient cells, functioning as intercellular mediators of cell–cell communication (auto-, para-, juxta-, or even endocrine). The use of EVs as biomarkers in liver disease development are described. 

liver disease biomarkers extracellular vesicles

1. Introduction

More than eight hundred million people suffer from chronic liver disease, which accounts for approximately two million deaths per year worldwide [1], with cirrhosis, viral hepatitis, and hepatocellular carcinoma (HCC) being the leading causes of liver-related deaths. Liver biopsy remains the gold standard diagnostic tool to assess the stage of liver diseases despite its significant disadvantages (low acceptance, highly invasive, and heterogeneous). The lack of non-invasive tools constitutes a significant barrier to the clinical management of liver diseases.
Circulating extracellular vesicles (EVs), a heterogeneous population of small membrane-encapsulated particles identified in several body fluids such as blood, saliva, and urine, have been proposed as markers for liquid biopsies in several diseases [2][3]. In the past decade, several shreds of evidence have suggested that EVs have a key role in liver disease, since they have been recognized as potent vehicles of intercellular communication due to their capacity to transfer proteins, lipids, and nucleic acids, thereby influencing various physiological functions of the recipient cells [4][5]. In this way, they may contribute to the pathogenesis, initiation, and progression of different liver diseases [6], emerging as a key player in cell–cell communication during acute and chronic liver disease [7]. Previous studies report changes in the number, surface markers, and cargos in the circulating EVs after liver injury, thus representing a potential biomarker for liver disorders [8]. Interestingly, changes in EVs have been demonstrated before histological signs appear, providing strong evidence of their utility as non-invasive tools even for early diagnosis [9].

2. Non-Alcoholic Fatty Liver Disease (NAFLD)

Non-alcoholic fatty liver disease (NAFLD) is the most common chronic liver disease worldwide, affecting about a quarter of the world’s adult population [10]. The diagnosis of steatohepatitis (NASH), a more severe form of the disease, requires a histological examination to confirm the presence of ballooning and inflammation; thus, the development of non-invasive biomarkers in NAFLD has generated considerable attention in the scientific community. Previous studies reported the role of EVs in NAFLD development, such as hepatocyte-derived sphingosine 1-phosphate (S1P)-containing EVs with pro-inflammatory activity in NASH [11]. Indeed, hepatocyte-derived EVs released from damaged hepatocytes in experimental models of NASH activate non-parenchymal cells, such as endothelial, stellate, and hepatic macrophages, contributing to the progression of liver disease [12]. A few studies support the use of several types of EVs for diagnostic purposes in patients with NASH. Circulating CD14+ microvesicles (MVs) in NAFLD correlated with alanine aminotransferase (ALT) levels and NAScore [6]. Recently, Povero et al. [13] analyzed hepatocyte-derived EVs from pre-cirrhotic and cirrhotic NASH patients, using asialoglycoprotein receptor 1 (ASGPR1) and long-chain fatty acid transport protein 5 (FATP-5) as hepatocyte markers. Levels of ASGPR1 EVs were found to be increased twofold in pre-cirrhotic NASH and threefold in cirrhotic NASH, compared to healthy controls. Furthermore, ASGPR1+ EVs were also found to correlate with the hepatic vein portal gradient (HVPG), being a potential non-invasive biomarker of portal hypertension in patients with NASH and cirrhosis. Finally, a proteomic analysis of circulating EV cargo revealed two feature pairs (IL13Ra1 > TNR4; WISP-1 > BMP-14) with strong predictive power for liver fibrosis and cirrhosis (80% sensitivity and 80% specificity). Platelet-derived MVs have also been proposed for NASH diagnosis, with levels of CD61+ MVs correlating with fat fraction, ballooning, and fibrosis stage in NAFLD patients [14]. Another study used ASGR2 (asialoglycoprotein receptor 2) and CYP2E1 (cytochrome P450 2E1) antibodies as markers of hepatocyte-derived EVs and observed a decrease in ASGR2+ and CYP2E1+ MVs levels after bariatric surgery in 28 biopsy-proven NAFLD patients [8].

3. Alcoholic Hepatitis

Uncontrolled alcohol consumption results in a liver injury characterized by steatosis, inflammation, hepatitis, and cirrhosis [15]. Alcohol exposure increases the number of circulating EVs of hepatic origin (mostly hepatocytes and hepatic stellate cells (HSCs)). EVs act on target cells (macrophages, endothelial cells, and HSCs) promoting inflammation and fibrosis [3]. Its release is related to a partial inhibition of autophagy promoted by a decreased level of lysosomal-associated membrane proteins LAMP1 and LAMP2 through miR-155 expression [16]. In that sense, it has been demonstrated that alcohol-treated hepatocytes cross-talk with immune cells via microRNAs (miRNAs) contained in exosomes. In alcoholic liver disease (ALD), serum/plasma miR-122 and miR-155 levels are increased and predominantly associated with exosomes [17]. Thus, hepatocyte-derived exosomes with miR-122 increase the secretion of pro-inflammatory cytokines in monocytes [18]. Also, in hepatocytes, alcohol increases the delivery and formation of exosomes containing mitochondrial double-stranded RNA (mtdsRNA) that participate in the production of pro-inflammatory cytokines (IL1B) in Kupffer cells [19]. Related to this, hepatic EVs derived from alcoholic hepatitis mice are able to activate primary HSCs, inducing a-SMA and collagen through upregulating miRNAs and increasing IL1B and IL17 production in a TLR9-dependent manner in macrophages [20]. In this regard, CD40 ligand (CD40L) in EVs, in a caspase-dependent manner in response to alcohol exposure, has a critical role as a mediator of macrophage activation [21]. Furthermore, alcohol-exposed monocytes can communicate with naive monocytes via miR-27a-loaded EVs that program naive monocytes into M2 macrophages [22]. Also, protein cargo in EVs can be important, since, in animal models of alcoholic liver disease, macrophage activation was induced by hepatocyte-derived EVs harboring heat shock HSP90 protein [23].
Alcohol has been implicated in fibrosis through the release of a major fibrogenic cytokine, transforming growth factor-beta-1 (TGF-β1), and HSCs activation [24]. A study associated alcohol with the increase of profibrogenic factors through the levels of miR-19b in HSCs and derived exosomes. Interestingly, decreased miR-19b levels in activated HSCs resulted in a change in the expression of other miRNAs (miR-17–92 cluster). However, miR-19b was induced at the plasma and exosomal levels in this alcohol-induced hepatic fibrogenesis model [25]. In addition, elevated levels of EVs have been observed containing CYP2E1 derived from the liver in patients with alcoholism and in alcohol-exposed animals. CYP2E1 activity is associated with oxidative and endoplasmic reticulum stress after alcohol consumption, leading to the activation of apoptotic pathways and toxicity to monocytes and hepatocytes. Thus, these EVs with CYP2E1 cargo could act as a biomarker for liver damage from long-term alcohol exposure [26].
Moreover, EVs secreted from other organs such as intestinal epithelial cells have effects on hepatocytes during acute alcohol injury, highlighting the importance of the gut-liver axis in ALD progression [27].
Lastly, some studies have tried to find soluble markers to diagnose alcoholic hepatitis in a non-invasive manner. The plasma levels and EVs of cytokeratin-18 fragments (M30 and M65) are reliable non-invasive markers of alcoholic hepatitis [28] High levels of CD34+ and ASGPR1+ EVs can be used as markers of non-response to corticosteroid therapy in severe alcoholic hepatitis [29]. Recently, plasma EV concentration and sphingolipid cargo were found to correlate with the severity and mortality of alcoholic steatohepatitis [30].

4. Viral Hepatitis

EVs are potent modulators of the immune response. In vitro studies showed that hepatocytes infected with replicating HBV release EVs that induced a programmed cell death 1 ligand 1 (PD-L1) expression in monocytes, possibly suppressing host antiviral activity [31]. Notably, Montaldo et al. analyzed EVs in the plasma of HCV patients after direct-acting antiviral therapy, finding that miR204-5p, miR181a-5p, miR143-3p, and miR-122-p were decreased in the EVs from HCV patients compared to healthy donors. After that, EV cargo was determined after 6 months of therapy, and miR204-p and miR143-3p were still different between healthy and HCV-treated patients, indicating that EV-mediated signals could play a causal role in fibrosis progression despite viral eradication [32]. Another study showed that patients with active hepatitis C (ALT > 100 IU/mL) had an elevated number of T cell-derived MPs compared to patients with mild hepatitis C (ALT< 40U/mL) and healthy controls [33]. Researcher's group recently reported a decrease in endothelial and platelet apoptotic MV levels after a sustained virological response in HCV patients, concluding that this may be directly involved in the improvement of inflammation and endothelial dysfunction observed in these patients after HCV eradication [34].

5. Fibrosis

Besides amplifying inflammation and modulating injury, EVs have also been demonstrated to promote liver fibrosis in NAFLD and ALD [35]. HSCs regulate the establishment and sustaining of liver fibrosis [36] partly due to their ability to chronically secrete EVs. Previous studies indicate that lipotoxic hepatocyte-derived pro-inflammatory miRNA-rich EVs could activate TLR-3 in HSCs [37], inducing their activation and migration [38]. These activated HSCs (aHSCs) in turn release EVs that contain various profibrotic proteins, lipids, and nucleic acids [39]. Moreover, the release of these EVs increases in response to liver injury [40]. Unlike aHSCs, EVs secreted by quiescent HSCs display antifibrotic properties since their cargo is shown to suppress HSC activation. Furthermore, they reduce inflammation, promote cell viability, inhibit hepatocyte apoptosis, and decrease liver transaminase levels, indicating their therapeutic potential [41][42]. Also, in the progression of NAFLD, exosomes from visceral adipose tissue (VAT) were related to fibrosis through TGF-B dysregulation in the hepatocytes and HSCs [43].
Although HCV does not replicate in HSCs, EVs from HCV-infected hepatocytes induce the expression of profibrogenic genes. miR-19a in these hepatocyte-derived HCV-EVs was able to promote fibrosis by targeting SOCS3 which caused the activation of the STAT3–TGF-B signaling pathway [44]. Previous studies aimed at determining the role of platelet and monocyte-derived MVs as biomarkers of fibrosis in biopsy-proven NAFLD patients [45]. CD14+ CD16+ EVs improved the ability of liver fibrosis scores to identify patients with F3/F4 fibrosis in a small preliminary cohort. Weil et al. reported 2.5-fold higher levels of platelet-derived MVs in 10 healthy subjects compared with 90 cirrhotic patients [46]. Finally, another study found that hepatocyte-derived MVs were 4.0-fold and 2.2-fold higher in patients with Child–Pugh C compare with those with Child–Pugh A or B respectively. Indeed, hepatocyte-derived MVs correlated with HVPG and were able to predict 6-month mortality independently of the Child–Pugh score or Model for End-Stage Liver Disease (MELD) [47].

6. Hepatobiliary Tumors: Hepatocellular Carcinoma and Cholangiocarcinoma

HCC is the most common primary liver cancer, being the fourth cause of cancer-related deaths worldwide [48]. HCC has a poor prognosis due to the lack of early symptoms and the low sensitivity and specificity of available diagnostic tools. Early detection is essential to improving surveillance and the adoption of curative surgical therapies. In the same scenario, the earlier the cholangiocarcinoma (CCA) detection, the more opportunities there are for curative treatments. In the context of cancer, the role of EVs has emerged as another promising strategy for liver cancer surveillance. Besides cell-to-cell contact, intercellular communication also happens through EVs to set up and modify tumor microenvironments. EVs are released by cancer cells in order to promote tumor growth and improve the tumor microenvironment for the spreading of these cells [49]. EVs are present in circulation at the early and advanced stages of the disease. The stability and integrity of EVs and their molecular cargos may serve as useful early-stage cancer diagnostic biomarkers and therapeutic approaches [50][51][52].
Current guidelines suggest the need for non-invasive tools for the diagnosis of HCC and CCA [53]. Circulating MV levels were found to increase in HCC patients in comparison to cirrhotic patients. Furthermore, they correlated with HCC tumor size, pathological type, and TNM stages, tending to a decrease after surgical intervention [54]. Several HCC-associated surface markers have been used to isolate and quantify liver tumor EVs. Julich-Haertel and colleagues showed that the combination of annexin V+ EpCAM+ ASGPR1+ CD133+ taMPs allowed one to distinguish liver malignancies and cirrhosis. Furthermore, EpCAM+ ASGPR1+ and annexin V+ were increased in liver cancer (HCC and CCA) compared to cirrhotic patients. In addition, 7 days after tumor resection, EpCAM+ ASGPR1+ annexin V+ levels significantly decreased, showing a strong association with tumors [55]. A high expression of MMP-7-EVs could be a marker for the differential diagnosis of CCA [56]. Regarding early diagnosis, three EV subpopulations, EpCAM+ CD63+, CD147+ CD63+, and GPC3+ CD63+ were highly associated with the early diagnosis of HCC (AUROC of 0.95 (95% CI = 0.90–0.99) with a sensitivity of 91% and a specificity of 90%) [57]. Another pan-cancer marker was proposed for the diagnosis of HCC and CCA, such as EpCAM+ CD147+ EVs which were increased in HCC, CCA, and other cancers [55]. A complex technique integrating covalent chemistry-mediated EV capture/release, multimarker antibody cocktails, nanostructured substrates, and microfluidic chaotic mixers showed that purified EpCAM, ASGPR1, and CD147 EVs have a 10-gene HCC-specific signature that allows one to distinguish HCC patients from at-risk cirrhotic patients (AUROC: 0.93 (95% CI, 0.86–1.00; S: 94.4% and S: 88.5%)) [58]. Early recurrence after liver resection was related to higher levels of Hepar-1+ microparticles before surgery, suggesting its potential role as a prognostic biomarker [59]. An emerging body of evidence supports the idea that platelets have an important role in carcinogenesis, mainly in HCC development [60][61]. In fact, platelet-derived EVs have also been related to colon cancer [62], but further studies are needed in liver tumors.
The content of cancer-derived EVs significantly differs from that of healthy cells, including different types of RNA such as miRNA, lncRNA, and cancer-specific proteins [53]. miR-122 EVs allowed the differentiation of HCC from liver cirrhosis (AUC:0.990, 95% CI, 0.945–1.00). In addition, the combination of miRNA-122, miRNA-148a, and Alpha-fetoprotein (AFP) increases diagnostic accuracy (AUC:0.931, 95% CI, 0.857–0.973), suggesting that the serum vesicle microRNA signature alone or in combination with available markers could be used as a screening tool for HCC [63]. Higher miR-21 in the circulating EVs of HCC patients are better markers than serum miR-21 in differentiating HCC from cirrhotic and healthy patients [64][65]. Another cluster of miRNAs, miR-18a, miR-221, miR-222, miR-224, miR-101, miR-106b, miR-122, and miR-195, were found to have an increase in exosomes from HCC [53]. Inside purified ASGPR1+ EVs, four miRNAs, miR-10b-5p, miR-21-5p, miR-221-3p, and miR-223-3p were found to be increased in those patients with lower AFP levels [66]. The lncRNA LINC00853 in EVs showed a good diagnostic capacity for HCC (AUC:0.934, 95% IC 0.887–0.966) [67]. Another study found that lnc85 was higher in the exosomes of HCC patients with high and low levels of AFP compared to the healthy control and liver cirrhosis (AUC:0.869) [68]. Recently, a study showed that cancer-associated fibroblast (CAF)-derived MVs can be implicated in HCC progression. The survival rate in patients with low antitumoral miR-150-3p levels in plasma CAF-derived exosomes was significantly poor compared to patients with high miR-150-3p levels. [69]
Regarding CCA, it is well known that bile EVs were significantly higher in CCA patients [70]. A proteomic study showed that EVs from CCA patients expressed a specific protein profile showing potential usefulness as a diagnostic tool [71]. Many studies have shown that EVs are involved in the development and progression of liver cancer. Further pieces of evidence to better understand the role of EVs in diagnosis and prognosis are needed.

References

  1. Asrani, S.K.; Devarbhavi, H.; Eaton, J.; Kamath, P.S. Burden of liver diseases in the world. J. Hepatol. 2019, 70, 151–171.
  2. György, B.; Szabó, T.G.; Pásztói, M.; Pál, Z.; Misják, P.; Aradi, B.; László, V.; Pállinger, É.; Pap, E.; Kittel, Á.; et al. Membrane vesicles, current state-of-the-art: Emerging role of extracellular vesicles. Cell. Mol. Life Sci. 2011, 68, 2667–2688.
  3. Wu, D.; Zhu, H.; Wang, H. Extracellular Vesicles in Non-alcoholic Fatty Liver Disease and Alcoholic Liver Disease. Front. Physiol. 2021, 12, 707429.
  4. Yáñez-Mó, M.; Siljander, P.R.-M.; Andreu, Z.; Bedina Zavec, A.; Borràs, F.E.; Buzas, E.I.; Buzas, K.; Casal, E.; Cappello, F.; Carvalho, J.; et al. Biological properties of extracellular vesicles and their physiological functions. J. Extracell. Vesicles 2015, 4, 27066.
  5. Cazanave, S.C.; Mott, J.; Bronk, S.F.; Werneburg, N.W.; Fingas, C.D.; Meng, X.W.; Finnberg, N.; El-Deiry, W.; Kaufmann, S.; Gores, G.J. Death Receptor 5 Signaling Promotes Hepatocyte Lipoapoptosis. J. Biol. Chem. 2011, 286, 39336.
  6. Kornek, M.; Lynch, M.; Mehta, S.H.; Lai, M.; Exley, M.; Afdhal, N.H.; Schuppan, D. Circulating microparticles as disease-specific biomarkers of severity of inflammation in patients with hepatitis C or nonalcoholic steatohepatitis. Gastroenterology 2012, 143, 448–458.
  7. Sugimachi, K.; Matsumura, T.; Hirata, H.; Uchi, R.; Ueda, M.; Ueo, H.; Shinden, Y.; Iguchi, T.; Eguchi, H.; Shirabe, K.; et al. Identification of a bona fide microRNA biomarker in serum exosomes that predicts hepatocellular carcinoma recurrence after liver transplantation. Br. J. Cancer 2015, 112, 532–538.
  8. Nakao, Y.; Amrollahi, P.; Parthasarathy, G.; Mauer, A.S.; Sehrawat, T.S.; Vanderboom, P.; Nair, K.S.; Nakao, K.; Allen, A.M.; Hu, T.Y.; et al. Circulating extracellular vesicles are a biomarker for NAFLD resolution and response to weight loss surgery. Nanomedicine 2021, 36, 102430.
  9. Sarin, S.K.; APASL ACLF Research Consortium (AARC) for APASL ACLF Working Party; Choudhury, A.; Sharma, M.K.; Maiwall, R.; Al Mahtab, M.; Rahman, S.; Saigal, S.; Saraf, N.; Soin, A.S.; et al. Acute-on-chronic liver failure: Consensus recommendations of the Asian Pacific association for the study of the liver (APASL): An update. Hepatol. Int. 2019, 13, 353–390.
  10. Younossi, Z.M.; Koenig, A.B.; Abdelatif, D.; Fazel, Y.; Henry, L.; Wymer, M. Global epidemiology of nonalcoholic fatty liver disease-Meta-analytic assessment of prevalence, incidence, and outcomes. Hepatology 2016, 64, 73–84.
  11. Dasgupta, D.; Nakao, Y.; Mauer, A.S.; Thompson, J.M.; Sehrawat, T.S.; Liao, C.Y.; Krishnan, A.; Lucien, F.; Guo, Q.; Liu, M.; et al. IRE1A Stimulates Hepatocyte-Derived Extracellular Vesicles That Promote Inflammation in Mice with Steatohepatitis. Gastroenterology 2020, 159, 1487–1503.e17.
  12. Povero, D.; Eguchi, A.; Niesman, I.R.; Andronikou, N.; de Mollerat du Jeu, X.; Mulya, A.; Berk, M.; Lazic, M.; Thapaliya, S.; Parola, M.; et al. Lipid-induced toxicity stimulates hepatocytes to release angiogenic microparticles that require Vanin-1 for uptake by endothelial cells. Sci. Signal. 2013, 6, ra88.
  13. Povero, D.; Yamashita, H.; Ren, W.; Subramanian, M.G.; Myers, R.P.; Eguchi, A.; Simonetto, D.A.; Goodman, Z.D.; Harrison, S.A.; Sanyal, A.J.; et al. Characterization and Proteome of Circulating Extracellular Vesicles as Potential Biomarkers for NASH. Hepatol. Commun. 2020, 4, 1263–1278.
  14. Li, J.; Liu, H.; Mauer, A.S.; Lucien, F.; Raiter, A.; Bandla, H.; Mounajjed, T.; Yin, Z.; Glaser, K.J.; Yin, M.; et al. Characterization of Cellular Sources and Circulating Levels of Extracellular Vesicles in a Dietary Murine Model of Nonalcoholic Steatohepatitis. Hepatol. Commun. 2019, 3, 1235–1249.
  15. Rahman, M.A.; Patters, B.J.; Kodidela, S.; Kumar, S. Extracellular Vesicles: Intercellular Mediators in Alcohol-Induced Pathologies. J. Neuroimmune Pharmacol. 2020, 15, 409–421.
  16. Babuta, M.; Furi, I.; Bala, S.; Bukong, T.N.; Lowe, P.; Catalano, D.; Calenda, C.; Kodys, K.; Szabo, G. Dysregulated Autophagy and Lysosome Function Are Linked to Exosome Production by Micro-RNA 155 in Alcoholic Liver Disease. Hepatology 2019, 70, 2123–2141.
  17. Bala, S.; Petrasek, J.; Mundkur, S.; Catalano, D.; Levin, I.; Ward, J.; Alao, H.; Kodys, K.; Szabo, G. Circulating microRNAs in exosomes indicate hepatocyte injury and inflammation in alcoholic, drug-induced, and inflammatory liver diseases. Hepatology 2012, 56, 1946–1957.
  18. Momen-Heravi, F.; Bala, S.; Kodys, K.; Szabo, G. Exosomes derived from alcohol-treated hepatocytes horizontally transfer liver specific miRNA-122 and sensitize monocytes to LPS. Sci. Rep. 2015, 5, 9991.
  19. Lee, J.H.; Shim, Y.R.; Seo, W.; Kim, M.H.; Choi, W.M.; Kim, H.H.; Kim, Y.E.; Yang, K.; Ryu, T.; Jeong, J.M.; et al. Mitochondrial Double-Stranded RNA in Exosome Promotes Interleukin-17 Production Through Toll-Like Receptor 3 in Alcohol-associated Liver Injury. Hepatology 2020, 72, 609–625.
  20. Eguchi, A.; Yan, R.; Pan, S.Q.; Wu, R.; Kim, J.; Chen, Y.; Ansong, C.; Smith, R.D.; Tempaku, M.; Ohno-Machado, L.; et al. Comprehensive characterization of hepatocyte-derived extracellular vesicles identifies direct miRNA-based regulation of hepatic stellate cells and DAMP-based hepatic macrophage IL-1β and IL-17 upregulation in alcoholic hepatitis mice. J. Mol. Med. 2020, 98, 1021–1034.
  21. Verma, V.K.; Li, H.; Wang, R.; Hirsova, P.; Mushref, M.; Liu, Y.; Cao, S.; Contreras, P.C.; Malhi, H.; Kamath, P.S.; et al. Alcohol stimulates macrophage activation through caspase-dependent hepatocyte derived release of CD40L containing extracellular vesicles. J. Hepatol. 2016, 64, 651–660.
  22. Saha, B.; Momen-Heravi, F.; Kodys, K.; Szabo, G. MicroRNA Cargo of Extracellular Vesicles from Alcohol-exposed Monocytes Signals Naive Monocytes to Differentiate into M2 Macrophages. J. Biol. Chem. 2016, 291, 149–159.
  23. Saha, B.; Momen-Heravi, F.; Furi, I.; Kodys, K.; Catalano, D.; Gangopadhyay, A.; Haraszti, R.; Satishchandran, A.; Iracheta-Vellve, A.; Adejumo, A.; et al. Extracellular vesicles from mice with alcoholic liver disease carry a distinct protein cargo and induce macrophage activation through heat shock protein 90. Hepatology 2018, 67, 1986–2000.
  24. Breitkopf, K.; Haas, S.; Wiercinska, E.; Singer, M.V.; Dooley, S. Anti-TGF-beta strategies for the treatment of chronic liver disease. Alcohol. Clin. Exp. Res. 2005, 29, 121S–131S.
  25. Brandon-Warner, E.; Feilen, N.A.; Culberson, C.R.; Field, C.O.; Delemos, A.S.; Russo, M.W.; Schrum, L.W. Processing of miR17-92 Cluster in Hepatic Stellate Cells Promotes Hepatic Fibrogenesis During Alcohol-Induced Injury. Alcohol. Clin. Exp. Res. 2016, 40, 1430–1442.
  26. Cho, Y.E.; Mezey, E.; Hardwick, J.P.; Salem, N.; Clemens, D.L.; Song, B.J. Increased ethanol-inducible cytochrome P450-2E1 and cytochrome P450 isoforms in exosomes of alcohol-exposed rodents and patients with alcoholism through oxidative and endoplasmic reticulum stress. Hepatol. Commun. 2017, 1, 675–690.
  27. Lamas-Paz, A.; Morán, L.; Peng, J.; Salinas, B.; López-Alcántara, N.; Sydor, S.; Vilchez-Vargas, R.; Asensio, I.; Hao, F.; Zheng, K.; et al. Intestinal Epithelial Cell-Derived Extracellular Vesicles Modulate Hepatic Injury via the Gut-Liver Axis During Acute Alcohol Injury. Front. Pharmacol. 2020, 11, 603771.
  28. Bissonnette, J.; Altamirano, J.; Devue, C.; Roux, O.; Payancé, A.; Lebrec, D.; Bedossa, P.; Valla, D.; Durand, F.; Ait-Oufella, H.; et al. A prospective study of the utility of plasma biomarkers to diagnose alcoholic hepatitis. Hepatology 2017, 66, 555–563.
  29. Sukriti, S.; Maras, J.S.; Bihari, C.; Das, S.; Vyas, A.K.; Sharma, S.; Hussain, S.; Shasthry, S.; Choudhary, A.; Premkumar, M.; et al. Microvesicles in hepatic and peripheral vein can predict nonresponse to corticosteroid therapy in severe alcoholic hepatitis. Aliment. Pharmacol. Ther. 2018, 47, 1151–1161.
  30. Sehrawat, T.S.; Arab, J.P.; Liu, M.; Amrollahi, P.; Wan, M.; Fan, J.; Nakao, Y.; Pose, E.; Navarro-Corcuera, A.; Dasgupta, D.; et al. Circulating Extracellular Vesicles Carrying Sphingolipid Cargo for the Diagnosis and Dynamic Risk Profiling of Alcoholic Hepatitis. Hepatology 2021, 73, 571–585.
  31. Kakizaki, M.; Yamamoto, Y.; Yabuta, S.; Kurosaki, N.; Kagawa, T.; Kotani, A. The immunological function of extracellular vesicles in hepatitis B virus-infected hepatocytes. PLoS ONE 2018, 13, e0205886.
  32. Montaldo, C.; Terri, M.; Riccioni, V.; Battistelli, C.; Bordoni, V.; D’Offizi, G.; Prado, M.G.; Trionfetti, F.; Vescovo, T.; Tartaglia, E.; et al. Fibrogenic signals persist in DAA-treated HCV patients after sustained virological response. J. Hepatol. 2021, 75, 1301–1311.
  33. Kornek, M.; Popov, Y.; Libermann, T.A.; Afdhal, N.H.; Schuppan, D. Human T cell microparticles circulate in blood of hepatitis patients and induce fibrolytic activation of hepatic stellate cells. Hepatology 2011, 53, 230–242.
  34. Muñoz-Hernández, R.; Ampuero, J.; Millán, R.; Gil-Gómez, A.; Rojas, Á.; Macher, H.C.; Gallego-Durán, R.; Gato, S.; Montero-Vallejo, R.; Rico, M.C.; et al. Hepatitis C Virus Clearance by Direct-Acting Antivirals Agents Improves Endothelial Dysfunction and Subclinical Atherosclerosis: HEPCAR Study. Clin. Transl. Gastroenterol. 2020, 11, e00203.
  35. Gabriel, K.; Ingram, A.; Austin, R.; Kapoor, A.; Tang, D.; Majeed, F.; Qureshi, T.; Al-Nedawi, K. Regulation of the tumor suppressor PTEN through exosomes: A diagnostic potential for prostate cancer. PLoS ONE 2013, 8, e70047.
  36. Chen, Z.; Jain, A.; Liu, H.; Zhao, Z.; Cheng, K. Targeted Drug Delivery to Hepatic Stellate Cells for the Treatment of Liver Fibrosis. J. Pharmacol. Exp. Ther. 2019, 370, 695–702.
  37. Seo, W.; Eun, H.S.; Kim, S.Y.; Yi, H.S.; Lee, Y.S.; Park, S.H.; Jang, M.J.; Jo, E.; Kim, S.C.; Han, Y.M.; et al. Exosome-mediated activation of toll-like receptor 3 in stellate cells stimulates interleukin-17 production by γδ T cells in liver fibrosis. Hepatology 2016, 64, 616–631.
  38. Hirsova, P.; Ibrahim, S.H.; Verma, V.K.; Morton, L.A.; Shah, V.H.; LaRusso, N.F.; Gores, G.J.; Malhi, H. Extracellular vesicles in liver pathobiology: Small particles with big impact. Hepatology 2016, 64, 2219–2233.
  39. Devaraj, E.; Perumal, E.; Subramaniyan, R.; Mustapha, N. Liver fibrosis: Extracellular vesicles mediated intercellular communication in perisinusoidal space. Hepatology 2022, 76, 275–285.
  40. Hirsova, P.; Ibrahim, S.H.; Verma, V.K.; Morton, L.A.; Shah, V.H.; LaRusso, N.F.; Gores, G.J.; Malhi, H. Lipid-Induced Signaling Causes Release of Inflammatory Extracellular Vesicles From Hepatocytes. Gastroenterology 2016, 150, 956–967.
  41. Bruno, S.; Chiabotto, G.; Camussi, G. Extracellular Vesicles: A Therapeutic Option for Liver Fibrosis. Int. J. Mol. Sci. 2020, 21, 4255.
  42. Huang, R.; Pan, Q.; Ma, X.; Wang, Y.; Liang, Y.; Dai, B.; Liao, X.; Li, M.; Miao, H. Hepatic Stellate Cell-Derived Microvesicles Prevent Hepatocytes from Injury Induced by APAP/H2O2. Stem Cells Int. 2016, 2016, 8357567.
  43. Hirsova, P.; Ibrahim, S.H.; Verma, V.K.; Morton, L.A.; Shah, V.H.; LaRusso, N.F.; Gores, G.J.; Malhi, H. Adipocyte exosomes induce transforming growth factor beta pathway dysregulation in hepatocytes: A novel paradigm for obesity-related liver disease. J. Surg. Res. 2014, 192, 268–275.
  44. Devhare, P.B.; Sasaki, R.; Shrivastava, S.; Di Bisceglie, A.M.; Ray, R.; Ray, R.B. Exosome-Mediated Intercellular Communication between Hepatitis C Virus-Infected Hepatocytes and Hepatic Stellate Cells. J. Virol. 2017, 91, e02225-16.
  45. Welsh, J.A.; Scorletti, E.; Clough, G.F.; Englyst, N.A.; Byrne, C.D. Leukocyte extracellular vesicle concentration is inversely associated with liver fibrosis severity in NAFLD. J. Leukoc. Biol. 2018, 104, 631–639.
  46. Weil, D.; Di Martino, V.; Mourey, G.; Biichle, S.; Renaudin, A.; Laheurte, C.; Cypriani, B.; Delabrousse, E.; Grandclément, E.; Thévenot, T.; et al. Small Annexin V-Positive Platelet-Derived Microvesicles Affect Prognosis in Cirrhosis: A Longitudinal Study. Clin. Transl. Gastroenterol. 2021, 12, e00333.
  47. Payancé, A.; Silva-Junior, G.; Bissonnette, J.; Tanguy, M.; Pasquet, B.; Levi, C.; Roux, O.; Nekachtali, O.; Baiges, A.; Hernández-Gea, V.; et al. Hepatocyte microvesicle levels improve prediction of mortality in patients with cirrhosis. Hepatology 2018, 68, 1508–1518.
  48. Sung, H.; Ferlay, J.; Siegel, R.L.; Laversanne, M.; Soerjomataram, I.; Jemal, A.; Bray, F. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J. Clin. 2021, 71, 209–249.
  49. Bebelman, M.P.; Smit, M.J.; Pegtel, D.M.; Baglio, S.R. Biogenesis and function of extracellular vesicles in cancer. Pharmacol. Ther. 2018, 188, 1–11.
  50. Xie, F.; Feng, S.; Yang, H.; Mao, Y. Extracellular vesicles in hepatocellular cancer and cholangiocarcinoma. Ann. Transl. Med. 2019, 7, 86.
  51. Gandhi, C.R. Oxidative Stress and Hepatic Stellate Cells: A Paradoxical Relationship. Trends Cell Mol. Biol. 2012, 7, 1–10.
  52. Lee, Y.T.; Tran, B.V.; Wang, J.J.; Liang, I.Y.; You, S.; Zhu, Y.; Agopian, V.G.; Tseng, H.R.; Yang, J.D. The Role of Extracellular Vesicles in Disease Progression and Detection of Hepatocellular Carcinoma. Cancers 2021, 13, 3076.
  53. Sberna, A.L.; Bouillet, B.; Rouland, A.; Brindisi, M.C.; Nguyen, A.; Mouillot, T.; Duvillard, L.; Denimal, D.; Loffroy, R.; Vergès, B.; et al. European Association for the Study of the Liver (EASL), European Association for the Study of Diabetes (EASD) and European Association for the Study of Obesity (EASO) clinical practice recommendations for the management of non-alcoholic fatty liver diseas. Diabet. Med. 2018, 35, 368–375.
  54. Wang, W.; Li, H.; Zhou, Y.; Jie, S. Peripheral blood microvesicles are potential biomarkers for hepatocellular carcinoma. Cancer Biomark. 2013, 13, 351–357.
  55. Julich-Haertel, H.; Urban, S.K.; Krawczyk, M.; Willms, A.; Jankowski, K.; Patkowski, W.; Kruk, B.; Krasnodębski, M.; Ligocka, J.; Schwab, R.; et al. Cancer-associated circulating large extracellular vesicles in cholangiocarcinoma and hepatocellular carcinoma. J. Hepatol. 2017, 67, 282–292.
  56. Lang, S.A.; Bednarsch, J.; Joechle, K.; Amygdalos, I.; Czigany, Z.; Heij, L.; Ulmer, T.F.; Neumann, U.P. Prognostic biomarkers for cholangiocarcinoma (CCA): State of the art. Expert Rev. Gastroenterol. Hepatol. 2021, 15, 497–510.
  57. Sun, N.; Zhang, C.; Lee, Y.; Tran, B.V.; Wang, J.; Kim, H.; Lee, J.; Zhang, R.Y.; Wang, J.J.; Hu, J.; et al. HCC EV ECG Score: An Extracellular Vesicle-based Protein Assay for Detection of Early-Stage Hepatocellular Carcinoma. Hepatology 2022.
  58. Sun, N.; Lee, Y.-T.; Zhang, R.Y.; Kao, R.; Teng, P.-C.; Yang, Y.; Yang, P.; Wang, J.J.; Smalley, M.; Chen, P.-J.; et al. Purification of HCC-specific extracellular vesicles on nanosubstrates for early HCC detection by digital scoring. Nat. Commun. 2020, 11, 4489.
  59. Abbate, V.; Marcantoni, M.; Giuliante, F.; Vecchio, F.M.; Gatto, I.; Mele, C.; Saviano, A.; Arciuolo, D.; Gaetani, E.; Ferrari, M.C.; et al. HepPar1-Positive Circulating Microparticles Are Increased in Subjects with Hepatocellular Carcinoma and Predict Early Recurrence after Liver Resection. Int. J. Mol. Sci. 2017, 18, 1043.
  60. Lazar, S.; Goldfinger, L.E. Platelets and extracellular vesicles and their cross talk with cancer. Blood 2021, 137, 3192.
  61. Malehmir, M.; Pfister, D.; Gallage, S.; Szydlowska, M.; Inverso, D.; Kotsiliti, E.; Leone, V.; Peiseler, M.; Surewaard, B.G.J.; Rath, D.; et al. Platelet GPIbα is a mediator and potential interventional target for NASH and subsequent liver cancer. Nat. Med. 2019, 25, 641–655.
  62. Zhao, L.; Bi, Y.; Kou, J.; Shi, J.; Piao, D. Phosphatidylserine exposing-platelets and microparticles promote procoagulant activity in colon cancer patients. J. Exp. Clin. Cancer Res. 2016, 35, 1–12.
  63. Wang, Y.; Zhang, C.; Zhang, P.; Guo, G.; Jiang, T.; Zhao, X.; Jiang, J.; Huang, X.; Tong, H.; Tian, Y. Serum exosomal microRNAs combined with alpha-fetoprotein as diagnostic markers of hepatocellular carcinoma. Cancer Med. 2018, 7, 1670–1679.
  64. Wang, H.; Hou, L.; Li, A.; Duan, Y.; Gao, H.; Song, X. Expression of Serum Exosomal MicroRNA-21 in Human Hepatocellular Carcinoma. Biomed. Res. Int. 2014, 2014, 864894.
  65. Tian, X.-P.; Wang, C.-Y.; Jin, X.-H.; Li, M.; Wang, F.-W.; Huang, W.-J.; Yun, J.-P.; Xu, R.-H.; Cai, Q.-Q.; Xie, D. Acidic microenvironment up-regulates exosomal mir-21 and mir-10b in early-stage hepatocellular carcinoma to promote cancer cell proliferation and metastasis. Theranostics 2019, 9, 1965–1979.
  66. Ghosh, S.; Bhowmik, S.; Majumdar, S.; Goswami, A.; Chakraborty, J.; Gupta, S.; Aggarwal, S.; Ray, S.; Chatterjee, R.; Bhattacharyya, S.; et al. The exosome encapsulated microRNAs as circulating diagnostic marker for hepatocellular carcinoma with low alpha-fetoprotein. Int. J. Cancer 2020, 147, 2934–2947.
  67. Kim, S.S.; Baek, G.O.; Ahn, H.R.; Sung, S.; Seo, C.W.; Cho, H.J.; Nam, S.W.; Cheong, J.Y.; Eun, J.W. Serum small extracellular vesicle-derived LINC00853 as a novel diagnostic marker for early hepatocellular carcinoma. Mol. Oncol. 2020, 14, 2646–2659.
  68. Huang, X.; Sun, L.; Wen, S.; Deng, D.; Wan, F.; He, X.; Tian, L.; Liang, L.; Wei, C.; Gao, K.; et al. RNA sequencing of plasma exosomes revealed novel functional long noncoding RNAs in hepatocellular carcinoma. Cancer Sci. 2020, 111, 3338–3349.
  69. Yugawa, K.; Yoshizumi, T.; Mano, Y.; Itoh, S.; Harada, N.; Ikegami, T.; Kohashi, K.; Oda, Y.; Mori, M. Cancer-associated fibroblasts promote hepatocellular carcinoma progression through downregulation of exosomal miR-150-3p. Eur. J. Surg. Oncol. 2021, 47, 384–393.
  70. Severino, V.; Dumonceau, J.-M.; Delhaye, M.; Moll, S.; Annessi-Ramseyer, I.; Robin, X.; Frossard, J.-L.; Farina, A. Extracellular Vesicles in Bile as Markers of Malignant Biliary Stenoses. Gastroenterology 2017, 153, 495–504.e8.
  71. Arbelaiz, A.; Azkargorta, M.; Krawczyk, M.; Santos-Laso, A.; Lapitz, A.; Perugorria, M.J.; Erice, O.; Gonzalez, E.; Jimenez-Agüero, R.; Lacasta, A.; et al. Serum extracellular vesicles contain protein biomarkers for primary sclerosing cholangitis and cholangiocarcinoma. Hepatology 2017, 66, 1125–1143.
More
Information
Subjects: Pathology
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : , , , , , , ,
View Times: 493
Entry Collection: Gastrointestinal Disease
Revisions: 6 times (View History)
Update Date: 11 Jan 2023
1000/1000
Video Production Service