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 -- 2621 2022-12-30 15:35:02 |
2 format Meta information modification 2621 2023-01-03 03:25:03 |

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.
Yang, Q.;  Luo, Y.;  Lan, B.;  Dong, X.;  Wang, Z.;  Ge, P.;  Zhang, G.;  Chen, H. Exosomes and Acute Pancreatitis-Associated Acute Lung Injury. Encyclopedia. Available online: https://encyclopedia.pub/entry/39632 (accessed on 01 July 2024).
Yang Q,  Luo Y,  Lan B,  Dong X,  Wang Z,  Ge P, et al. Exosomes and Acute Pancreatitis-Associated Acute Lung Injury. Encyclopedia. Available at: https://encyclopedia.pub/entry/39632. Accessed July 01, 2024.
Yang, Qi, Yalan Luo, Bowen Lan, Xuanchi Dong, Zhengjian Wang, Peng Ge, Guixin Zhang, Hailong Chen. "Exosomes and Acute Pancreatitis-Associated Acute Lung Injury" Encyclopedia, https://encyclopedia.pub/entry/39632 (accessed July 01, 2024).
Yang, Q.,  Luo, Y.,  Lan, B.,  Dong, X.,  Wang, Z.,  Ge, P.,  Zhang, G., & Chen, H. (2022, December 30). Exosomes and Acute Pancreatitis-Associated Acute Lung Injury. In Encyclopedia. https://encyclopedia.pub/entry/39632
Yang, Qi, et al. "Exosomes and Acute Pancreatitis-Associated Acute Lung Injury." Encyclopedia. Web. 30 December, 2022.
Exosomes and Acute Pancreatitis-Associated Acute Lung Injury
Edit

Acute pancreatitis (AP) is a prevalent clinical condition of the digestive system, with a growing frequency each year. Approximately 20% of patients suffer from severe acute pancreatitis (SAP) with local consequences and multi-organ failure, putting a significant strain on patients’ health insurance. According to reports, the lungs are particularly susceptible to SAP. Acute respiratory distress syndrome, a severe type of acute lung injury (ALI), is the primary cause of mortality among AP patients. Controlling the mortality associated with SAP requires an understanding of the etiology of AP-associated ALI, the discovery of biomarkers for the early detection of ALI, and the identification of potentially effective drug treatments. Exosomes are a class of extracellular vesicles with a diameter of 30–150 nm that are actively released into tissue fluids to mediate biological functions. Exosomes are laden with bioactive cargo, such as lipids, proteins, DNA, and RNA. During the initial stages of AP, acinar cell-derived exosomes suppress forkhead box protein O1 expression, resulting in M1 macrophage polarization. Similarly, macrophage-derived exosomes activate inflammatory pathways within endothelium or epithelial cells, promoting an inflammatory cascade response. On the other hand, a part of exosome cargo performs tissue repair and anti-inflammatory actions and inhibits the cytokine storm during AP. 

exosome acute pancreatitis acute lung injury

1. Introduction

Acute pancreatitis (AP) is a frequent occurring, acute abdominal illness. The majority of patients present with mild AP, which may spontaneously resolve. Nonetheless, approximately 20% of patients report severe AP (SAP) that is fast progressive and aggressive [1][2]. Acute lung injury (ALI) is a life-threatening condition characterized by diffuse interstitial and alveolar edema resulting from the damage of pulmonary microvascular endothelial cells (PMVECs) and alveolar epithelial cells (AECs) [3][4]. ALI and its severe form, acute respiratory distress syndrome (ARDS), are among the most prevalent consequences of SAP and are leading causes of mortality in SAP patients [5].
New insights into the pathogenesis of AP and associated ALI have emerged in recent years. The systemic inflammatory response (SIRS) caused by the abnormal activation of pancreatic enzymes, mitochondrial dysfunction, impaired autophagy, endoplasmic reticulum stress, programmed cell death, intestinal mucosal barrier damage, and bacterial translocation are the initiating factors of multiple organ dysfunction syndromes (MODS) in AP [6]. Key molecules causing pulmonary air–blood barrier disruption and alveolar edema include pancreatic, intestinal, and liver-derived non-coding RNAs (ncRNAs), damage-associated molecular patterns (DAMPs), and pathogen-associated molecular patterns (PAMPs). Cross-signaling between immune cells (such as neutrophils, macrophages, and T cells) and parenchymal cells (such as acinar cells, intestinal epithelial cells, PMVECs, and AECs) is a crucial mechanism for maintaining the AP cytokine storm [7]. However, the molecular network of intercellular communication is intricate and requires immediate clarification. Exosome-related research has expanded quickly in recent years. The basics of exosomes, including the biogenesis, processes of secretion, and cargo they carry, have been steadily uncovered [8][9].

2. Exosomes and AP-Associated ALI

The molecular mechanisms involved in AP-associated ALI that lead to SIRS and diffuse alveolar damage have been studied in detail. Multiple signaling pathways are engaged during AP. To summarize, the activation of the cytokine storm is caused by the upregulation of extracellular mediators such as DAMPs, histones, and ncRNAs during AP. Intriguingly, emerging research has shown that the pancreas–lung axis [6] and the gut–lung axis [10] may mediate the cytokine storm in AP-associated ALI, and that exosomes may be major carriers of extracellular mediators transported along the signaling axis.
Zhu et al. discovered that plasma exosomal miR-216a was considerably elevated in AP patients with ALI compared to AP patients without ALI [11]. Exosomal miR-216a seems to be a particular modulator of inflammation in AP-induced ALI. As shown in animal investigations, miR-216a expression was undetectable in all organs save the pancreas, including the lung, gut, heart, and kidney. It is possible that exosomal miR-216a is pancreas-specific. Moreover, exosomal miR-216a enhanced the permeability of pulmonary microvascular endothelial cells, which was linked with the degree of ALI during AP. Xu et al. discovered that cold-inducible RNA-binding protein (CIRP) may play a crucial role in alveolar macrophage (AM) pyroptosis as well as neutrophil recruitment during AP-associated ALI [12]. The level of CIRP was found to be enhanced in the pancreatic tissue, serum, and lung tissue of AP rats by Xu and his colleagues. Interestingly, immunohistochemical staining revealed that pancreatic islet cells may be the predominant cell type that secretes CIRP, which may be an additional inflammatory mediator secreted by injured pancreatic tissue that induces ALI. In addition, Murao et al. discovered that CIRP may persist extracellularly as exosomes and mediate inflammation during sepsis.
The gut–lung axis is a commonly recognized pathophysiological signal of crosstalk between intestinal and pulmonary diseases. In the case of AP-associated ALI, intestinal damage and its subsequent response has an “amplifier” effect [13]. Firstly, intestinal barrier damage and increased intestinal permeability are prevalent in AP patients and models generated by a variety of causes [14][15]. Secondly, the intestinal barrier is a factor that exacerbates the inflammatory response to SIRS [16]. After intestinal barrier damage, the most direct consequence may be the “second strike” of intestine-derived endotoxins entering the circulation and lungs through the portal vein or mesenteric lymphatic system [17][18]. On the other hand, SIRS may be promoted by exosomes released from the injured gut during AP. Under physiological conditions, intestinal epithelial cells (IECs) or DC-derived exosomes carrying transforming growth factor-β, MHC class I and II complexes and co-stimulatory molecules coordinate the regulation of intestinal immunity and maintain immune homeostasis. However, miRNAs such as miR-122a and miR-29a released from damaged IECs can exacerbate intestinal barrier damage and increase intestinal permeability [19].

3. The Therapeutic Potential of Exosomes in AP and Associated ALI

Aa a double-edged sword, exosomes release both anti-inflammatory and pro-inflammatory substances into the cells to which they bind. They regulate the inflammatory cascade response during AP by selectively binding downstream molecules and modulating receptor cell activity. As a result, researchers are considering exosomes as possible therapeutic targets. The first step in the chain of AP is pancreatic acinar cell damage. Recently, in vitro experiments confirmed that exosomes produced by acinar cells were shown to drastically lower intracellular ROS and the inflammatory factor level and ameliorate pathological pancreatic injury [20]. These findings show that injured cells may be able to heal tissue damage but that certain triggering elements are required. Several in vivo and in vitro studies demonstrated emodin’s protective properties against AP-induced pancreatic injury, intestinal barrier dysfunction, and ALI [12][21]. Emodin was discovered to boost the differentiation and anti-inflammatory activity of regulatory T cells by stimulating the release of exosome-specific lncRNA taurine upregulated 1 (TUG1) from pancreatic acinar cells, hence limiting the development of AP [22]. Recent research suggests that the inflamed pancreas may release exosomes into the circulation during SAP. These exosomes have been shown to have a pro-inflammatory action. However, emodin may prevent “bad” exosomes from being secreting by acinar cells. Proteomics was employed to characterize the impact of rhodopsin on the plasma-derived exosome proteome in SAP rats. According to the results of the enrichment study, peroxisome proliferator-activated receptors (PPAR) signaling is the primary mechanism by which emodin influences the exosomal proteome. Mechanistic investigations showed that emodin protected the lungs by preventing exosome-mediated M1 polarization of alveolar macrophages via regulating PPAR signaling [23]. During AP-induced ALI, AECs are critical target cells in the lung, and their destruction directly leads to pulmonary edema and widespread alveolar injury. Evidence suggests that exosomes derived from AECs in ALI help to regulate the immune balance and the inflammatory cascade response [24]. As a natural antioxidant and anti-inflammatory agent, salidroside has been shown to protect against AP and ALI/ARDS caused by AP in mice. Activation of NF-κB, interleukin receptor-associated kinase, and tumor necrosis factor receptor-associated molecule 6 in AMs was inhibited by the salidroside-stimulated release of exosomal miR-146a from AECs and improved ALI in rats [25]. DAMPs and PAMPs activate AMs, an essential innate immune cell population during the outset of AP. They produce significant doses of inflammatory mediators that contribute to lung damage [26], unlike alveolar and alveolar epithelial cells. Similarly, pyroptotic AM-derived pyroptotic bodies increased AEC damage and vascular leakage [27]. Furthermore, the Hippo signaling pathway was activated in AECs by AMs-derived exosomal transfer RNA-derived fragments, which caused AECs ferroptosis and aided in the establishment of ALI [28]. Modulation of AMs-derived exosomes may therefore be a promising method for combating AP and associated ALI.
The therapeutic potential of mesenchymal stem cell (MSC)-derived exosomes cannot be overlooked in exosome-related treatment techniques [29][30][31]. Human umbilical cord mesenchymal stem cells (hucMSC) have been frequently recognized for their capacity for self-renewal and multilineage differentiation, particularly for the bioactive chemicals they contain for tissue repair and the management of inflammation [32][33]. Han et al. discovered that hucMSC-derived exosomes showed a remarkable tissue regeneration potential in rats suffering from traumatic pancreatitis (trauma-induced non-infectious AP) [34]. In particular, hucMSC-Evs injected intravenously could colonize injured pancreatic tissue and inhibit inflammatory response and apoptosis of acinar cells, promoting damaged tissue repair [34]. A less frequent consequence of AP is myocardial injury, which is challenging to treat and has a high death rate [35]. MSC-derived exosomes were found to upregulate vascular hemophilia factor and vascular endothelial growth factor through the activation of the Akt/nuclear factor E2 related factors 2/heme oxygenase 1 signaling pathway, which led to the amelioration of SAP-induced myocardial injury [36]. Unfortunately, the previous research did not characterize the components transported by stem-cell-derived exosomes, i.e., the molecules that exert the protective effects. Xia et al., on the other hand, discovered that adipose-derived MSC-derived exosome increased AM mitochondrial function by transferring mitochondrial components to them, which led to a reduction in pulmonary inflammation in mice [37].
Researchers have discovered that edible plants may generate nanoscale EVs with shapes and components comparable to animal exosomes [38]. Furthermore, plant-derived exosomes are biocompatible and safe to consume, with no side effects or possible toxicity [39][40]. Plant-derived exosomes may penetrate biological barriers to transport lipid-soluble and hydrophilic target molecules to tissues in vivo, increasing the target molecules’ bioavailability or effectiveness of the target molecules [41]. Ginger exosome-like nanoparticles (GELN) have been found to be taken up by lung macrophages and epithelial cells, with a preference for ACE2-positive cells. Furthermore, ginger GELN miRNA has the potential to bind to many locations in the SARS-CoV-2 virus genome and be transported to lung epithelial cells to decrease Nsp12 production and, consequently, lung inflammation [42]. As a result, plant-derived exosomes are potential AP therapeutic agents.
Exosomes maybe a therapeutic target for AP and related organ failures. Using novel medications to control the levels of lipids, proteins, and nucleic acids carried by exosomes, as well as the exosome-mediated drug delivery, offers fantastic potential to decrease the AP-induced cytokine storm. On the other hand, stem-cell-based exosome therapies have been in existence for some time and should be tested in AP clinical trials as soon as is feasible.

4. Exosome-Based Diagnostic Strategy

Compared with cell-free nucleic acids and proteins, exosome-specific nucleic acids and proteins are protected by a lipid bilayer and have better stability in the extracellular environment by avoiding degradation by RNA hydrolases [43][44]. In addition, exosomes are more accessible than solid biopsy samples and are present in almost all biological fluids, such as plasma, urine, saliva, ascites, breast milk, and amniotic fluid. Therefore, exosomes have been established as ideal biomarkers and have been widely used in disease diagnosis, prognosis evaluation, and treatment monitoring. The diagnostic and prognostic value of exosomes in pancreatic diseases such as pancreatic cancer and chronic pancreatitis has been widely confirmed [45][46][47][48].
Peripheral blood-derived exosomal miR-155, miR-216a, miR-21, (miR-603, miR-548ad-5p, miR-122-5p, miR-4477a, miR-192-5p, miR-215-5p, and miR-583), lncRNA PVT1, and MALAT1 [49] were linked with the severity of SAP and may provide new insight into the etiology of SAP and act as biomarkers of SAP. In addition, some AP serum/mesenteric lymph/plasma markers such as FBXL19-AS1 [50] and lnc-ITSN1-2 [51], miR-214-3p [52], miR-27a-5p [53], miR-217-5p [54], miR-193a-5p [55], miR-375 [56], miR-148a [57], miR-138-5p [58], miR-92b [59], miR-10a [59], miR-7 [60], miR-9 [60], miR-141 [61], miR-551b-5p [62], miR-126-5p [63], miR-24 [64], (miR-22-3p, miR-1260b, miR-762, miR-23b, miR-23a, miR-550a-5p, miR-324-5p, miR-484, miR-331-3p, miR-140-3p, and miR-342-3p [65]), miR-127 [66], miR-372 [67], miR-126-5p [68], miR-146 [69], miR-153 [70], miR-320-5p [55], Circ_0000284 [71], and Circ_0073748 [72], also exist in the exosome. These potential exosomal biomarkers also provide an important direction for the diagnosis and prognosis of AP in clinical applications.
Exosome-specific S100A8 correlates with the inflammatory response and predicts severity in individuals with SAP. Similarly, several free proteins and DNA are elevated in the plasma of patients with SAP, and this has implications for the diagnosis and prognostic assessment of the disease [73][74][75]. Moreover, the above substances such as HMGB1 [76], heat shock protein 70 [77], histones [49], CIRP [73], S100A12 [74], gamma-enolase [78], and mtDNA [75] have also been proven to be essential cargoes loaded by exosomes. Therefore, in the future, two areas of interest will be exploring whether the above exosome-specific cargoes can recognize AP and whether exosome-specific proteins or DNAs have better diagnostic performance than free proteins or DNAs.
In addition to blood samples, many exosomes exist in biological fluids, including urine and pancreatic juice. Urine samples are easy to obtain and non-invasive, which is desirable for both clinicians and patients. Several studies have found that nucleic acids and proteins carried by urine-derived exosomes have potential diagnostic value in pancreatic diseases [79][80][81]. In the case of AP, a 2014 study confirmed that urinary ketone bodies, glucose, plasma choline, and lipid levels were increased in patients’ urine, while levels of urinary hippurate, creatine, and plasma-branched chain amino acids decreased. A biomarker panel of guanine, hippurate, and creatine reliably identified AP with high sensitivity and specificity [82]. Later, a proteomic study confirmed that the peak intensity ratio of urinary β-2 microglobulin to saponin B has a better diagnostic performance in patients with SAP, especially with renal injury and inflammation [83]. In short, urine is also a promising biospecimen for mining AP biomarkers. Exploring the changes in exosomal cargo in urine during AP is urgent.
Pancreatic juice is secreted by pancreatic acinar cells and duct wall cells, an alkaline liquid with a strong digestibility. In terms of accuracy and the characterization that best reflects the pathological mechanisms of AP, pancreatic fluid is second to pancreatic tissue and is a biofluid superior to blood and urine [84]. In 2018, Osteikoetxea et al. found that the detection and characterization of EVs in pancreatic juice are feasible and confirmed that mucin, CFTR, and MDR1 proteins carried by pancreatic juice-derived EVs are potential biomarkers of pancreatic cancer [85]. Later, Nakamura et al. found that miR-21 and miR-15 carried by pancreatic juice-derived exosomes have the potential to diagnose patients with PDAC and CP [86]. The diagnostic value of pancreatic juice examination in pancreas-related diseases is constantly updated. The changes in the exosomal cargo of pancreatic juice in AP patients should be explored as soon as possible.
Bronchoalveolar lavage fluid (BALF), which is in direct contact with lung tissue, is an ideal biologic fluid for the diagnosis of lung diseases [87]. Previous studies have found that exosomes derived from BALF have potential diagnostic value in patients with ALI/ARDS [88], chronic obstructive pulmonary disease [89], nodular pulmonary disease [90], lung cancer [91], lung infections [92], and asthma [93].
Because of their stability, exosome-specific ncRNAs and proteins have been employed as biomarkers in AP and associated ALI research. Exosome isolation from blood, pleural fluid, urine, ascitic fluid, alveolar lavage fluid, and pancreatic fluid is an emerging method of fluid biopsy with broad potential clinical applications, especially for patients with AP who are experiencing multi-organ failure. Exosome-specific ncRNA and protein detection are complicated by several variables, as has been described. Different clinical investigations on the expression of a specific exosomal cargo in the bodily fluids of AP patients may find contradictory results. To further establish the sensitivity and specificity of exosomal cargoes, substantial cohort studies are still required before their use can be advocated for in clinical applications.

References

  1. Leppäniemi, A.; Tolonen, M.; Tarasconi, A.; Lohse, H.A.S.; Gamberini, E.; Kirkpatrick, A.W.; Ball, C.G.; Parry, N.; Sartelli, M.; Wolbrink, D.R.J.; et al. 2019 WSES guidelines for the management of severe acute pancreatitis. World J. Emerg. Surg. 2019, 14, 1–20.
  2. Iannuzzi, J.P.; King, J.A.; Leong, J.H.; Quan, J.; Windsor, J.W.; Tanyingoh, D.; Stephanie, C.; Nauzer, F.; Steven, J.H.; Abdel-Aziz, S.; et al. Global Incidence of Acute Pancreatitis Is Increasing Over Time: A Systematic Review and Meta-Analysis. Gastroenterology 2022, 162, 122–134.
  3. Liu, G.; Zhang, J.; Chen, H.; Wang, C.; Qiu, Y.; Liu, Y.; Wan, J.; Guo, H. Effects and mechanisms of alveolar type II epithelial cell apoptosis in severe pancreati-tis-induced acute lung injury. Exp. Ther. Med. 2014, 7, 565–572.
  4. Vrolyk, V.; Singh, B. Animal models to study the role of pulmonary intravascular macrophages in spontaneous and induced acute pancreatitis. Cell Tissue Res. 2020, 380, 207–222.
  5. Luiken, I.; Eisenmann, S.; Garbe, J.; Sternby, H.; Verdonk, R.C.; Dimova, A.; Ignatavicius, P.; Ilzarbe, L.; Koiva, P.; Penttilä, A.K.; et al. Pleuropulmonary pathologies in the early phase of acute pancreatitis correlate with disease severity. PLoS ONE 2022, 17, e0263739.
  6. Ge, P.; Luo, Y.; Okoye, C.S.; Chen, H.; Liu, J.; Zhang, G.; Xu, C.; Chen, H. Intestinal barrier damage, systemic inflammatory response syndrome, and acute lung injury: A troublesome trio for acute pancreatitis. Biomed. Pharmacother. 2020, 132, 110770.
  7. Liu, D.; Wen, L.; Wang, Z.; Hai, Y.; Yang, D.; Zhang, Y.; Bai, M.; Song, B.; Wang, Y. The Mechanism of Lung and Intestinal Injury in Acute Pancreatitis: A Review. Front. Med. 2022, 9, 904078.
  8. Suire, C.N.; Hade, M.D. Extracellular Vesicles in Type 1 Diabetes: A Versatile Tool. Bioengineering 2022, 9, 105.
  9. Hade, M.; Suire, C.; Suo, Z. Mesenchymal Stem Cell-Derived Exosomes: Applications in Regenerative Medicine. Cells 2021, 10, 1959.
  10. Wang, Z.; Liu, J.; Li, F.; Luo, Y.; Ge, P.; Zhang, Y.; Wen, H.; Yang, Q.; Ma, S.; Chen, H. The gut-lung axis in severe acute Pancreatitis-associated lung injury: The protection by the gut microbiota through short-chain fatty acids. Pharmacol. Res. 2022, 182, 106321.
  11. Zhu, H.; Song, Y.; Kong, X.; Du, Y. Expression of microrNA-216A in patients with acute pancreatitis complicated with lung injury and its effect on endothelial cell permeability. J. Chin. Pancreas Dis. 2020, 20, 4.
  12. Xu, Q.; Wang, M.; Guo, H.; Liu, H.; Zhang, G.; Xu, C.; Chen, H. Emodin Alleviates Severe Acute Pancreatitis-Associated Acute Lung Injury by Inhibiting the Cold-Inducible RNA-Binding Protein (CIRP)-Mediated Activation of the NLRP3/IL-1β/CXCL1 Signaling. Front. Pharmacol. 2021, 12, 655372.
  13. Liang, X.Y.; Jia, T.X.; Zhang, M. Intestinal bacterial overgrowth in the early stage of severe acute pancreatitis is associated with acute respiratory distress syndrome. World J. Gastroenterol. 2021, 27, 1643–1654.
  14. Jabłońska, B.; Mrowiec, S. Nutritional Support in Patients with Severe Acute Pancreatitis-Current Standards. Nutrients 2021, 13, 1498.
  15. Kanthasamy, K.A.; Akshintala, V.S.; Singh, V.K. Nutritional Management of Acute Pancreatitis. Gastroenterol. Clin. N. Am. 2021, 50, 141–150.
  16. Singh, N.; Sonika, U.; Moka, P.; Sharma, B.; Sachdev, V.; Mishra, S.K.; Upadhyay, A.D.; Saraya, A. Association of endotoxaemia & gut permeability with complications of acute pancre-atitis: Secondary analysis of data. Indian J. Med. Res. 2019, 149, 763–770.
  17. Hu, X.; Gong, L.; Zhou, R.; Han, Z.; Ji, L.; Zhang, Y.; Zhang, S.; Wu, D. Variations in Gut Microbiome are Associated with Prognosis of Hypertriglyceridem-ia-Associated Acute Pancreatitis. Biomolecules 2021, 11, 695.
  18. Tang, Y.; Kong, J.; Zhou, B.; Wang, X.; Liu, X.; Wang, Y.; Zhu, S. Mesenteric Lymph Duct Ligation Alleviates Acute Lung Injury Caused by Severe Acute Pancreatitis Through Inhibition of High Mobility Group Box 1-Induced Inflammation in Rats. Am. J. Dig. Dis. 2021, 66, 4344–4353.
  19. Park, E.J.; Shimaoka, M.; Kiyono, H. Functional Flexibility of Exosomes and MicroRNAs of Intestinal Epithelial Cells in Affecting Inflammation. Front. Mol. Biosci. 2022, 9, 854487.
  20. Guo, Y.; Cao, F.; Ding, Y.; Lu, J.; Liu, S.; Li, F. Acinar Cells Derived Exosomes Alleviate the Severity of Acute Pancreatitis. Discov. Med. 2021, 31, 95–105.
  21. Zhou, Q.; Xiang, H.; Liu, H.; Qi, B.; Shi, X.; Guo, W.; Zou, J.; Wan, X.; Wu, W.; Wang, Z.; et al. Emodin Alleviates Intestinal Barrier Dysfunction by Inhibiting Apoptosis and Regulating the Immune Response in Severe Acute Pancreatitis. Pancreas 2021, 50, 1202–1211.
  22. Wen, X.; He, B.; Tang, X.; Wang, B.; Chen, Z. Emodin inhibits the progression of acute pancreatitis via regulation of lncRNA TUG1 and exosomal lncRNA TUGMol. Med. Rep. 2021, 24, 785.
  23. Hu, Q.; Yao, J.; Wu, X.; Li, J.; Li, G.; Tang, W.; Liu, J.; Wan, M. Emodin attenuates severe acute pancreatitis-associated acute lung injury by suppressing pancreatic exosome-mediated alveolar macrophage activation. Acta Pharm. Sin. B 2021, 12, 3986–4003.
  24. Feng, Z.; Zhou, J.; Liu, Y.; Xia, R.; Li, Q.; Yan, L.; Chen, Q.; Chen, X.; Jiang, Y.; Chao, G.; et al. Epithelium- and endothelium-derived exosomes regulate the alveolar macrophages by targeting RGS1 mediated calcium signaling-dependent immune response. Cell Death Differ. 2021, 28, 2238–2256.
  25. Zheng, L.; Su, J.; Zhang, Z.; Jiang, L.; Wei, J.; Xu, X.; Lv, S. Salidroside regulates inflammatory pathway of alveolar macrophages by influencing the se-cretion of miRNA-146a exosomes by lung epithelial cells. Sci. Rep. 2020, 10, 20750.
  26. Ye, C.; Li, H.; Bao, M.; Zhuo, R.; Jiang, G.; Wang, W. Alveolar macrophage—Derived exosomes modulate severity and outcome of acute lung injury. Aging 2020, 12, 6120–6128.
  27. Qin, X.; Zhou, Y.; Jia, C.; Chao, Z.; Qin, H.; Liang, J.; Liu, X.; Liu, Z.; Sun, T.; Yuan, Y.; et al. Caspase-1-mediated extracellular vesicles derived from pyroptotic alveolar macrophages promote inflammation in acute lung injury. Int. J. Biol. Sci. 2022, 18, 1521–1538.
  28. Wang, W.; Zhu, L.; Li, H.; Ren, W.; Zhuo, R.; Feng, C.; He, Y.; Hu, Y.; Ye, C. Alveolar macrophage-derived exosomal tRF-22-8BWS7K092 activates Hippo signaling pathway to induce ferroptosis in acute lung injury. Int. Immunopharmacol. 2022, 107, 108690.
  29. Cheng, Y.; Cao, X.; Qin, L. Mesenchymal Stem Cell-Derived Extracellular Vesicles: A Novel Cell-Free Therapy for Sepsis. Front. Immunol. 2020, 11, 647.
  30. Goodman, R.R.; Jong, M.K.; Davies, J.E. Concise review: The challenges and opportunities of employing mesenchymal stromal cells in the treatment of acute pancreatitis. Biotechnol. Adv. 2019, 42, 107338.
  31. Pu, Q.; Xiu, G.; Sun, J.; Liu, P.; Ling, B. Progress on the effect of mesenchymal stem cell derived exosomes on multiple organ dysfunction in sepsis. Zhonghua Wei Zhong Bing Ji Jiu Yi Xue 2021, 33, 757–760.
  32. Wang, Y.; Li, H.; Li, X.; Su, X.; Xiao, H.; Yang, J. Hypoxic Preconditioning of Human Umbilical Cord Mesenchymal Stem Cells Is an Effective Strategy for Treating Acute Lung Injury. Stem Cells Dev. 2021, 30, 128–134.
  33. Shaikh, M.S.; Shahzad, Z.; Tash, E.A.; Janjua, O.S.; Khan, M.I.; Zafar, M.S. Human Umbilical Cord Mesenchymal Stem Cells: Current Literature and Role in Periodontal Regeneration. Cells 2022, 11, 1168.
  34. Han, L.; Zhao, Z.; Chen, X.; Yang, K.; Tan, Z.; Huang, Z.; Zhou, L.; Dai, R. Human umbilical cord mesenchymal stem cells-derived exosomes for treating traumatic pan-creatitis in rats. Stem Cell Res. Ther. 2022, 13, 221.
  35. Luo, Y.; Li, Z.; Ge, P.; Guo, H.; Li, L.; Zhang, G.; Xu, C.; Chen, H. Comprehensive Mechanism, Novel Markers and Multidisciplinary Treatment of Severe Acute Pan-creatitis-Associated Cardiac Injury—A Narrative Review. J. Inflamm. Res. 2021, 14, 3145–3169.
  36. Chen, M.; Chen, J.; Huang, W.; Li, C.; Luo, H.; Xue, Z.; Xiao, Y.; Wu, Q.; Chen, C. Exosomes from human induced pluripotent stem cells derived mesenchymal stem cells improved myocardial injury caused by severe acute pancreatitis through activating Akt/Nrf2/HO-1 axis. Cell Cycle 2022, 21, 1578–1589.
  37. Xia, L.; Zhang, C.; Lv, N.; Liang, Z.; Ma, T.; Cheng, H.; Xia, Y.; Shi, L. AdMSC-derived exosomes alleviate acute lung injury via transferring mitochondrial component to improve homeostasis of alveolar macrophages. Theranostics 2022, 12, 2928–2947.
  38. Kim, S.Q.; Kim, K.-H. Emergence of Edible Plant-Derived Nanovesicles as Functional Food Components and Nanocarriers for Therapeutics Delivery: Potentials in Human Health and Disease. Cells 2022, 11, 2232.
  39. Cai, Y.; Zhang, L.; Zhang, Y.; Lu, R. Plant-Derived Exosomes as a Drug-Delivery Approach for the Treatment of Inflammatory Bowel Disease and Colitis-Associated Cancer. Pharmaceutics 2022, 14, 822.
  40. Zhang, Z.; Yu, Y.; Zhu, G.; Zeng, L.; Xu, S.; Cheng, H.; Ouyang, Z.; Chen, J.; Pathak, J.L.; Wu, L.; et al. The Emerging Role of Plant-Derived Exosomes-Like Nanoparticles in Immune Regulation and Periodontitis Treatment. Front. Immunol. 2022, 13, 896745.
  41. Nemati, M.; Singh, B.; Mir, R.A.; Nemati, M.; Babaei, A.; Ahmadi, M.; Rasmi, Y.; Golezani, A.G.; Rezaie, J. Plant-derived extracellular vesicles: A novel nanomedicine approach with advantages and challenges. Cell Commun. Signal. 2022, 20, 69.
  42. Teng, Y.; Xu, F.; Zhang, X.; Mu, J.; Sayed, M.; Hu, X.; Lei, C.; Sriwastva, M.; Kumar, A.; Sundaram, K.; et al. Plant-derived exosomal microRNAs inhibit lung inflammation induced by exosomes SARS-CoV-2 Nsp12. Mol. Ther. 2021, 29, 2424–2440.
  43. Yang, D.; Zhang, W.; Zhang, H.; Zhang, F.; Chen, L.; Ma, L.; Larcher, L.M.; Chen, S.; Liu, N.; Zhao, Q.; et al. Progress, opportunity, and perspective on exosome isolation—Efforts for efficient exosome-based theranostics. Theranostics 2020, 10, 3684–3707.
  44. Liu, W.-Z.; Ma, Z.-J.; Kang, X.-W. Current status and outlook of advances in exosome isolation. Anal. Bioanal. Chem. 2022, 414, 7123–7141.
  45. Desai, C.S.; Khan, A.; Bellio, M.A.; Willis, M.L.; Mahung, C.; Ma, X.; Baldwin, X.; Williams, B.M.; Baron, T.H.; Coleman, L.G.; et al. Characterization of extracellular vesicle miRNA identified in peripheral blood of chronic pancreatitis patients. Mol. Cell. Biochem. 2021, 476, 4331–4341.
  46. Wang, L.; Wu, J.; Ye, N.; Li, F.; Zhan, H.; Chen, S.; Xu, J. Plasma-Derived Exosome MiR-19b Acts as a Diagnostic Marker for Pancreatic Cancer. Front. Oncol. 2021, 11, 739111.
  47. Wu, Y.; Zeng, H.; Yu, Q.; Huang, H.; Fervers, B.; Chen, Z.-S.; Lu, L. A Circulating Exosome RNA Signature Is a Potential Diagnostic Marker for Pancreatic Cancer, a Systematic Study. Cancers 2021, 13, 2565.
  48. Yang, J.; Zhang, Y.; Gao, X.; Yuan, Y.; Zhao, J.; Zhou, S.; Wang, H.; Wang, L.; Xu, G.; Li, X.; et al. Plasma-Derived Exosomal ALIX as a Novel Biomarker for Diagnosis and Classification of Pancreatic Cancer. Front. Oncol. 2021, 11, 628346.
  49. Liu, T.; Huang, W.; Szatmary, P.; Abrams, S.T.; Alhamdi, Y.; Lin, Z.; Greenhalf, W.; Wang, G.; Sutton, R.; Toh, C.H. Accuracy of circulating histones in predicting persistent organ failure and mortality in patients with acute pancreatitis. Br. J. Surg. 2017, 104, 1215–1225.
  50. Ma, Q.; Gan, G.-F.; Niu, Y.; Tong, S.-J. Analysis of associations of FBXL19-AS1 with occurrence, development and prognosis of acute pancreatitis. Eur. Rev. Med. Pharmacol. Sci. 2020, 24, 12763–12769.
  51. Li, J.; Bu, X.; Chen, X.; Xiong, P.; Chen, Z.; Yu, L. Predictive value of long non-coding RNA intersectin 1-2 for occurrence and in-hospital mortality of severe acute pancreatitis. J. Clin. Lab. Anal. 2019, 34, e23170.
  52. Yan, Z.; Zang, B.; Gong, X.; Ren, J.; Wang, R. MiR-214-3p exacerbates kidney damages and inflammation induced by hyperlipidemic pancreatitis complicated with acute renal injury. Life Sci. 2019, 241, 117118.
  53. Zhu, Y.; Liu, S.; Wang, F. MicroRNA MiR-27a-5p Alleviates the Cerulein-Induced Cell Apoptosis and Inflammatory Injury of AR42J Cells by Targeting Traf3 in Acute Pancreatitis. Inflammation 2020, 43, 1988–1998.
  54. Erdos, Z.; Barnum, J.E.; Wang, E.; DeMaula, C.; Dey, P.M.; Forest, T.; Bailey, W.J.; Glaab, W.E. Evaluation of the Relative Performance of Pancreas-Specific MicroRNAs in Rat Plasma as Biomarkers of Pancreas Injury. Toxicol. Sci. 2019, 173, 5–18.
  55. Yu, W.; Zhang, M.; Li, X.; Pan, N.; Bian, X.; Wu, W. Protective Effect of miR-193a-5p and miR-320-5p on Caerulein-Induced Injury in AR42J Cells. Am. J. Dig. Dis. 2021, 66, 4333–4343.
  56. Usborne, A.L.; Smith, A.T.; Engle, S.K.; Watson, D.E.; Sullivan, J.M.; Walgren, J.L. Biomarkers of exocrine pancreatic injury in 2 rat acute pancreatitis models. Toxicol. Pathol. 2014, 42, 195–203.
  57. Miao, B.; Qi, W.; Zhang, S.; Wang, H.; Wang, C.; Hu, L.; Huang, G.; Li, S.; Wang, H. miR-148a suppresses autophagy by down-regulation of IL-6/STAT3 signaling in ceru-lein-induced acute pancreatitis. Pancreatology 2019, 19, 557–565.
  58. Song, G.; Zhou, J.; Song, R.; Liu, D.; Yu, W.; Xie, W.; Ma, Z.; Gong, J.; Meng, H.; Yang, T.; et al. Long noncoding RNA H19 regulates the therapeutic efficacy of mesenchymal stem cells in rats with severe acute pancreatitis by sponging miR-138-5p and miR-141-3p. Stem Cell Res. Ther. 2020, 11, 420.
  59. Liu, P.; Xia, L.; Zhang, W.-L.; Ke, H.-J.; Su, T.; Deng, L.-B.; Chen, Y.-X.; Lv, N.-H. Identification of serum microRNAs as diagnostic and prognostic biomarkers for acute pancreatitis. Pancreatology 2014, 14, 159–166.
  60. Lu, P.; Wang, F.; Wu, J.; Wang, C.; Yan, J.; Li, Z.-L.; Song, J.-X.; Wang, J.-J. Elevated Serum miR-7, miR-9, miR-122, and miR-141 Are Noninvasive Biomarkers of Acute Pancreatitis. Dis. Markers 2017, 2017, 7293459.
  61. Zhu, H.; Huang, L.; Zhu, S.; Li, X.; Li, Z.; Yu, C.; Yu, X. Regulation of autophagy by systemic admission of microRNA-141 to target HMGB1 in l-arginine-induced acute pancreatitis in vivo. Pancreatology 2016, 16, 337–346.
  62. Zhang, Y.; Yan, L.; Han, W. Elevated Level of miR-551b-5p is Associated With Inflammation and Disease Progression in Patients With Severe Acute Pancreatitis. Ther. Apher. Dial. 2018, 22, 649–655.
  63. Kuśnierz-Cabala, B.; Nowak, E.; Sporek, M.; Kowalik, A.; Kuźniewski, M.; Enguita, F.J.; Stępień, E. Serum levels of unique miR-551-5p and endothelial-specific miR-126a-5p allow discrimination of patients in the early phase of acute pancreatitis. Pancreatology 2015, 15, 344–351.
  64. Meng, S.; Wang, H.; Xue, D.; Zhang, W. Screening and validation of differentially expressed extracellular miRNAs in acute pan-creatitis. Mol. Med. Rep. 2017, 16, 6412–6418.
  65. Lu, X.G.; Kang, X.; Zhan, L.B.; Kang, L.M.; Fan, Z.W.; Bai, L.Z. Circulating miRNAs as biomarkers for severe acute pancreatitis associated with acute lung injury. World J. Gastroenterol. 2017, 23, 7440–7449.
  66. Shi, N.; Deng, L.; Chen, W.; Zhang, X.; Luo, R.; Jin, T.; Ma, Y.; Du, C.; Lin, Z.; Jiang, K.; et al. Is MicroRNA-127 a Novel Biomarker for Acute Pancreatitis with Lung Injury? Dis. Markers 2017, 2017, 1204295.
  67. Shan, Y.; Kong, W.; Zhu, A.; Zhang, J.; Ying, R.; Zhu, W. Increased levels of miR-372 correlate with disease progression in patients with hyper-lipidemic acute pancreatitis. Exp. Ther. Med. 2020, 19, 3845–3850.
  68. Chen, Y.J.; Lin, T.L.; Cai, Z.; Yan, C.H.; Gou, S.R.; Zhuang, Y.D. Assessment of acute pancreatitis severity via determination of serum levels of hsa-miR-126-5p and IL-6. Exp. Ther. Med. 2021, 21, 26.
  69. Li, X.Y.; Wang, Y.F.; Li, N. Circulating microRNA-146a and microRNA-146b exhibit potential to serve as markers for acute pancreatitis management and prognosis. Eur. Rev. Med. Pharmacol. Sci. 2020, 24, 12770–12780.
  70. Dai, J.; Jiang, M.; Hu, Y.; Xiao, J.; Hu, B.; Xu, J.; Han, X.; Shen, S.; Li, B.; Wu, Z.; et al. Dysregulated SREBP1c/miR-153 signaling induced by hypertriglyceridemia worsens acute pancreatitis and delays tissue repair. JCI Insight 2021, 6, e138584.
  71. Huang, H.; Chen, W.; Lu, J.; Zhang, S.; Xiang, X.; Wang, X.; Tang, G. Circ_0000284 Promoted Acute Pancreatitis Progression through the Regulation of miR-10a-5p/Wnt/β-Catenin Pathway. Chem. Biodivers. 2022, 19, e202101006.
  72. Ren, S.; Pan, L.; Yang, L.; Niu, Z.; Wang, L.; Gao, Y.; Liu, J.; Liu, Z.; Pei, H. Interfering hsa_circ_0073748 alleviates caerulein-induced ductal cell injury in acute pancreatitis by inhibiting miR-132-3p/TRAF3/NF-κB pathway. Cell Cycle 2022, 21, 172–186.
  73. Gong, J.-D.; Qi, X.-F.; Zhang, Y.; Li, H.-L. Increased admission serum cold-inducible RNA-binding protein concentration is associated with prognosis of severe acute pancreatitis. Clin. Chim. Acta 2017, 471, 135–142.
  74. Zhao, B.; Chen, Y.; Sun, W.W.; Chen, W.W.; Ma, L.; Yang, Z.T.; Huang, J.; Chen, E.Z.; Fei, J.; Mao, E.Q. Effect of S100A12 and soluble receptor for advanced glycation end products on the occurrence of severe acute pancreatitis. J. Dig. Dis. 2016, 17, 475–482.
  75. Wu, L.; Xu, W.; Wang, F.; Lv, T.; Yin, Z.; Song, Y. Plasma mtDNA Analysis Aids in Predicting Pancreatic Necrosis in Acute Pancreatitis Patients: A Pilot Study. Am. J. Dig. Dis. 2018, 63, 2975–2982.
  76. Li, N.; Wang, B.; Cai, S.; Liu, P. The Role of Serum High Mobility Group Box 1 and Interleukin-6 Levels in Acute Pancreatitis: A Meta-Analysis. J. Cell. Biochem. 2017, 119, 616–624.
  77. Arriaga-Pizano, L.; Boscó-Gárate, I.; Martínez-Ordaz, J.L.; Wong-Baeza, I.; Gutiérrez-Mendoza, M.; Sánchez-Fernandez, P.; Macías, C.I.R.L.; Isibasi, A.; Pelaez-Luna, M.; Cérbulo-Vázquez, A.; et al. High Serum Levels of High-Mobility Group Box 1 (HMGB1) and Low Levels of Heat Shock Protein 70 (Hsp70) are Associated with Poor Prognosis in Patients with Acute Pancreatitis. Arch. Med. Res. 2018, 49, 504–511.
  78. Owusu, L.; Xu, C.; Chen, H.; Liu, G.; Zhang, G.; Zhang, J.; Tang, Z.; Sun, Z.; Yi, X. Gamma-enolase predicts lung damage in severe acute pancreatitis-induced acute lung injury. Histochem. J. 2018, 49, 347–356.
  79. Moutinho-Ribeiro, P.; Macedo, G.; Melo, S.A. Pancreatic Cancer Diagnosis and Management: Has the Time Come to Prick the Bubble? Front. Endocrinol. 2019, 9, 779.
  80. Yoshizawa, N.; Sugimoto, K.; Tameda, M.; Inagaki, Y.; Ikejiri, M.; Inoue, H.; Usui, M.; Ito, M.; Takei, Y. miR-3940-5p/miR-8069 ratio in urine exosomes is a novel diagnostic biomarker for pancreatic ductal adenocarcinoma. Oncol. Lett. 2020, 19, 2677–2684.
  81. Yang, J.; Xu, R.; Wang, C.; Qiu, J.; Ren, B.; You, L. Early screening and diagnosis strategies of pancreatic cancer: A comprehensive review. Cancer Commun. 2021, 41, 1257–1274.
  82. Villaseñor, A.; Kinross, J.M.; Li, J.V.; Penney, N.; Barton, R.H.; Nicholson, J.K.; Darzi, A.; Barbas, C.; Holmes, E. 1H NMR Global Metabolic Phenotyping of Acute Pancreatitis in the Emergency Unit. J. Proteome Res. 2014, 13, 5362–5375.
  83. Chang, C.-T.; Liao, H.-Y.; Huang, W.-H.; Lin, S.-Y.; Tsai, T.-Y.; Yang, C.-Y.; Tsai, F.-J.; Chen, C.-J. Early prediction of severe acute pancreatitis by urinary β-2 microglobulin/saposin B peak ratios on MALDI-TOF. Clin. Chim. Acta 2015, 440, 115–122.
  84. Pallagi, P.; Hegyi, P.; Rakonczay, Z., Jr. The Physiology and Pathophysiology of Pancreatic Ductal Secretion: The Back-ground for Clinicians. Pancreas 2015, 44, 1211–1233.
  85. Osteikoetxea, X.; Benke, M.; Rodriguez, M.; Pálóczi, K.; Sódar, B.W.; Szvicsek, Z.; Szabó-Taylor, K.; Vukman, K.V.; Kittel, Á.; Wiener, Z.; et al. Detection and proteomic characterization of extracellular vesicles in human pancreatic juice. Biochem. Biophys. Res. Commun. 2018, 499, 37–43.
  86. Nakamura, S.; Sadakari, Y.; Ohtsuka, T.; Okayama, T.; Nakashima, Y.; Gotoh, Y.; Saeki, K.; Mori, Y.; Nakata, K.; Miyasaka, Y.; et al. Pancreatic Juice Exosomal MicroRNAs as Biomarkers for Detection of Pancreatic Ductal Adenocarcinoma. Ann. Surg. Oncol. 2019, 26, 2104–2111.
  87. Ge, P.; Zhang, J.; Zhang, G.; Chen, H. Research progress on application of metabolomics in acute lung injury. Zhonghua Wei Zhong Bing Ji Jiu Yi Xue 2021, 33, 1266–1271.
  88. Papadopoulos, S.; Kazepidou, E.; Antonelou, M.H.; Leondaritis, G.; Tsapinou, A.; Koulouras, V.P.; Avgeropoulos, A.; Nakos, G.; Lekka, M.E. Secretory Phospholipase A(2)-IIA Protein and mRNA Pools in Extra-cellular Vesicles of Bronchoalveolar Lavage Fluid from Patients with Early Acute Respiratory Distress Syndrome: A New Perception in the Dissemination of Inflammation. Pharmaceuticals 2020, 13, 415.
  89. Kaur, G.; Maremanda, K.P.; Campos, M.; Chand, H.S.; Li, F.; Hirani, N.; Haseeb, M.A.; Li, D.; Rahman, I. Distinct Exosomal miRNA Profiles from BALF and Lung Tissue of COPD and IPF Patients. Int. J. Mol. Sci. 2021, 22, 11830.
  90. Kishore, A.; Navratilova, Z.; Kolek, V.; Novosadova, E.; Čépe, K.; Du Bois, R.M.; Petrek, M. Expression analysis of extracellular microRNA in bronchoalveolar lavage fluid from patients with pulmonary sarcoidosis. Respirology 2018, 23, 1166–1172.
  91. Domagala-Kulawik, J. The relevance of bronchoalveolar lavage fluid analysis for lung cancer patients. Expert Rev. Respir. Med. 2019, 14, 329–337.
  92. Zhou, B.; Guo, M.; Hao, X.; Lou, B.; Liu, J.; She, J. Altered exosomal microRNA profiles in bronchoalveolar lavage fluid can mediate metabolism in patients with Acinetobacter baumannii ventilator-associated pneumonia. Ann. Transl. Med. 2020, 8, 1561.
  93. Zhao, M.; Li, Y.-P.; Geng, X.-R.; Zhao, M.; Ma, S.-B.; Yang, Y.-H.; Deng, Z.-H.; Luo, L.-M.; Pan, X.-Q. Expression Level of MiRNA-126 in Serum Exosomes of Allergic Asthma Patients and Lung Tissues of Asthmatic Mice. Curr. Drug Metab. 2019, 20, 799–803.
More
Information
Subjects: Biology
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: 318
Entry Collection: Gastrointestinal Disease
Revisions: 2 times (View History)
Update Date: 03 Jan 2023
1000/1000
Video Production Service