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 -- 3158 2023-02-08 20:36:40 |
2 Reference format revised. + 6 word(s) 3164 2023-02-09 06:31:42 | |
3 Figure format revised. Meta information modification 3164 2023-02-09 06:32:26 |

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.
Allegra, A.;  Murdaca, G.;  Gammeri, L.;  Ettari, R.;  Gangemi, S. Alarmins and MicroRNAs in Genesis of Respiratory Diseases. Encyclopedia. Available online: https://encyclopedia.pub/entry/41000 (accessed on 18 May 2024).
Allegra A,  Murdaca G,  Gammeri L,  Ettari R,  Gangemi S. Alarmins and MicroRNAs in Genesis of Respiratory Diseases. Encyclopedia. Available at: https://encyclopedia.pub/entry/41000. Accessed May 18, 2024.
Allegra, Alessandro, Giuseppe Murdaca, Luca Gammeri, Roberta Ettari, Sebastiano Gangemi. "Alarmins and MicroRNAs in Genesis of Respiratory Diseases" Encyclopedia, https://encyclopedia.pub/entry/41000 (accessed May 18, 2024).
Allegra, A.,  Murdaca, G.,  Gammeri, L.,  Ettari, R., & Gangemi, S. (2023, February 08). Alarmins and MicroRNAs in Genesis of Respiratory Diseases. In Encyclopedia. https://encyclopedia.pub/entry/41000
Allegra, Alessandro, et al. "Alarmins and MicroRNAs in Genesis of Respiratory Diseases." Encyclopedia. Web. 08 February, 2023.
Alarmins and MicroRNAs in Genesis of Respiratory Diseases
Edit

Alarmins support the pathogenesis of chronic respiratory diseases as asthma through ferroptosis and the axis composed of high mobility group box 1 (HMGB1) and interleukin (IL)-33. The mechanisms supporting the development and progression of chronic respiratory diseases and the role of HMBG1 and IL-33 are described.

respiratory diseases alarmins high mobility group box 1 interleukin-33 microRNAs

1. Introduction

1.1. Chronic Respiratory Diseases

Chronic respiratory disorders are the third most important cause of mortality worldwide after cardiovascular pathologies and cancers [1]. Numerous reports show that the occurrence of asthma, chronic obstructive pulmonary disease (COPD), pulmonary tumours, infection and hypertension, acute lung injury (ALI), and acute respiratory distress syndrome (ARDS) is progressively rising [2][3][4].
These diseases are highly heterogeneous, with varying pathogenesis, clinical manifestations or histopathology ranging from slight inflammation to the generation of disseminated nodular lesions or lung fibrosis [5]. However, changes in the humoral or cellular immune responses are described in all subjects suffering from respiratory diseases. These changes can influence the onset, progression, and prognosis of these diseases, and may be therapeutic targets.

1.2. General Considerations Regarding High Mobility Group Box 1, Interleukin-33 and microRNAs

1.2.1. High Mobility Group Box 1

HMGB1 is a chromatin-binding protein that weakly binds DNA. It is formed by 215 amino acid residues divided into three different parts: two tandem high mobility group box domains separated by a short linker and an acidic C-terminal tail. It binds DNA to modify the chromatin configuration while preserving genome stability [6].
HMGB1 produces numerous effects inside the nucleus. It helps nucleosome development, operates as a DNA chaperone to support DNA duplication and repair [7][8], and is doubtlessly implicated in the pathogenesis of tumours [9][10]. It is also involved in V(D)J transcription and stimulates autophagy and cell survival when it occurs. It is produced and released by macrophages, natural killer (NK) cells, and dendritic cells (DCs) during inflammation, or by cells after necrosis or cell death.
HMGB1 is an alarmin and, thus, causes reactions of both adaptive and innate immune effectors. It operates via the receptor for advanced glycation end products (RAGE) and other receptors, such as Toll-like receptors (TLRs) 2 and 4 [11]. HMGB1 works as a damage-associated molecular pattern (DAMP) when discharged from injured cells or released from stimulated macrophages, triggering numerous ligands [12][13][14][15][16][17]. This alarmin may be involved in other processes, such as coagulation. HMGB1 intensifies the sterile inflammation correlated with infection and thrombosis [18]. Platelets accumulate HMGB1, transfer it to the cell surface when stimulated and then discharge it into microparticles. Moreover, thrombocytes can connect to HMGB1 via the RAGE, coherent with possible autocrine effects [19][20][21].

1.2.2. Interleukin-33

IL-33 is a 30-KDa protein belonging to the IL-1 cytokine family. Its structure presents two different domains: the C-terminal and the N-terminal domain. The C-terminal domain is responsible for extracellular ST2-dependent activity. The N-terminal domain matches IL-33 to the nucleus and has a transcriptional repressor effect [22]. The IL-33 chromatin-binding motif present in this domain facilitates attachment to histone dimers and changes the chromatin configuration. This mechanism is implicated in the transcription of genes, altering gene repression [8]. The active form of IL-33 does not hold a signal peptide, so it is not discharged via a usual secretory pathway; rather, this cytokine is discharged when cells are injured and works like an alarmin [23]. IL-33 is released by epithelial and endothelial cells but can be released into the airways by other cells, such as DCs and macrophages [24][25].
IL-33 is an essential controller of immune responses operating on DCs, mast cells, group 2 innate lymphoid cells (ILC2s), macrophages, and CD4 + T2 cells, provoking a type 2 immune response [5]. Moreover, this cytokine can stimulate the ST2 of Tregs, demonstrating its capability to reduce inflammation. However, IL-33 can stimulate the synthesis of IL-8 via the stimulation of the JNK/c-Jun/AP-1 pathway, provoking inflammatory conditions. The control of gene expression is also due to the silencing of RNAs. IL-33 has been shown to silence Rip2 by sequentially reducing the expression of mRNAs associated with factors such as IL-17, iNOS, E-selectin, ICAM-1, VCAM-1, and TSLP. Furthermore, NOD-like receptor protein 3 (NLRP3) gene silencing reduces IL-33 synthesis [26].

1.2.3. microRNAs

Modifications in gene expression can be caused by numerous forms of non-coding RNA, including long non-coding RNA (lncRNA), circular RNA (circRNA), and microRNA (miRNA). MicroRNAs are RNA sequences of 17–24 nucleotides that modify gene expression by connecting to messenger RNA (mRNA) transcripts in the 3′-untranslated region or coding sequence, causing a reduction in mRNA translation and protein production, and stimulating mRNA elimination [27][28]. However, connecting miRNAs to mRNA transcripts may be an inaccurate process, involving chains of only 6–8 nucleotides; this implies that each miRNA can have several different mRNA targets and, hypothetically, can affect numerous genes and different functional systems [29][30].
Furthermore, miRNAs can be categorized into isomiRs, which contain modifications in the sequence dimension or nucleotides at the 3′ or 5′ ends, occasionally with alterations in activity and targets [31]. It has been shown that although miRNAs are present and operate within the cytoplasm, they can also be discharged into vesicles called exosomes. These vesicles arrive at the circulatory system, where they may facilitate cellular interactions [32]. MiRNA profile changes and altered expression compared to healthy subjects can be observed in different diseases. These variations in miRNA profiles promote some genes and reduce the expression of others [29].

2. Alarmins and Chronic Respiratory Diseases

2.1. Asthma and Alarmins

Asthma is a chronic pathology characterized by airway inflammation, hyperresponsiveness, submucosal fibrosis and increased mucus production [33]. It affects more than 270 million people, affecting both children and adults. Approximately 3–10% of all patients present with a severe form of the disease [34].
Asthma is distinguished by several immunological alterations provoked by the relationship between the environment and the patient’s genetic substrate. Based on immunological alterations, the disease can be classified into T2 asthma and non-T2 asthma [35]. This classification depends on the concentrations of type 2 cytokines [36].
The pathogenesis of asthma is due to type 2-mediated allergic inflammation that induces barrier defences at the mucosal surfaces and stimulates the generation of type 2 cytokines, including IL-4, IL-5, IL-9, and IL-13, and the shift of antibodies to IgE [37][38][39]. An innate type 2 immune response requires CD4+ T cells but depends on ILC2s [40][41][42]. ILC2s were initially recognized as cells not derived from the bone marrow and spleen. These are IL-25-dependent non-T non-B cells that were identified in the lung, spleen, liver, mesenteric lymph node, and peritoneum [43][44]. ILC2s induce T-helper cell (Th)2-related lung inflammation by controlling the delivery of Th2 cytokines. IL-33 plus IL-2, IL-7, IL-25, and TSLP stimulate lung ILC2s by reacting to allergen-induced tissue injury [45][46]. Lung ILC2s induce the production of a significant amount of IL-5 and IL-13 but not IL-4. In any case, ILC2 stimulation is mainly due to IL-33, which is induced in type 2 pneumocytes after contact with allergens [47][48][49][50][51]. It is noteworthy that different asthma types may respond to various treatment regimens [52][53].
A different approach to show the correlation between IL-33 and HMGB1 evaluated whether the P2Y13 receptor (P2Y13-R), a purinergic G protein-coupled receptor (GPCR), controlled the discharge of IL-33 and HMGB1 [54]. Human and mouse airway epithelial cells and C57Bl/6 mice were treated with different aeroallergens or respiratory viruses. The discharge and transfer of alarmins from the nucleus to the cytoplasm were evaluated. The effect of the P2Y13-R on the activity of airway epithelial cells was evaluated during and after experimental asthma by employing antagonists or animals with P2Y13-R gene deletion. Allergen contact provoked the discharge of ADP and ATP, molecules that stimulate P2Y13-R. Allergens, ATP, ADP, or contact with viruses caused the transfer of IL-33 and HMGB1 from the nucleus to the cytoplasm and their subsequent release; this effect was annulled by genetic deletion or the use of P2Y13 antagonists [54]
The function of alarmins in some pathological processes, such as ferroptosis, supports the existence of an axis composed of HMGB1 and IL-33. A link between IL-33 and HMGB1 in neutrophilic asthma has been evidenced and could be represented by ferroptosis. Ferroptosis is a type of regulated cell death (RCD) system that is dependent on reactive oxygen species (ROS) [55]
HMGB1 is discharged into the extracellular area in numerous forms of RCD and is correlated to the genesis of inflammation, both due to infections and aseptic. As an alarmin, HMGB1 could provoke the discharge of inflammatory substances, such as cytokines, after ferroptosis initiation. It has been reported that ferroptosis could significantly stimulate inflammation through IL-33 delivery [56]. The reduction in IL-33 decreases the inflammation caused by house dust mites (HDM), including eosinophil and neutrophil infiltration [57].

2.2. Alarmins and Respiratory Syncytial Virus Infection

Interesting data on the role of HMGB1 and IL-33 have also emerged in the context of different respiratory diseases, such as disorders derived from respiratory syncytial virus (RSV) infection and cystic fibrosis (CF).
RSV is a rapidly transmissible virus that provokes acute lung infections in children. The signs of RSV infection are slight; nevertheless, a subset of patients presents with severe RSV-correlated bronchiolitis. Inflammation provoked by RSV infection can include both the superior zone with nasal inflammation and rhinorrhoea and the inferior respiratory area, resulting in wheezing, bronchiolitis, and coughing [58][59]. Severe RSV-induced bronchiolitis causes necrosis, oedema, the shedding of airway epithelial cells, mucus overproduction and peribronchiolar inflammation. These elements cause airway obstruction [60].
Generally, RSV induces a type 1 immune response. Nevertheless, type 2 cytokines may also be released after RSV infection. There are increasing suggestions that children with critical RSV-correlated bronchiolitis are at a higher risk of presenting with asthma throughout their lives [61][62][63][64].
The epithelial-originated cytokines IL-33 and IL-25 and the alarmin HMGB1 stimulate and control ILC2 activity, driving the development of type 2-provoked pulmonary pathologies [65][66][67].
The management of RSV-infected airway epithelial cells in a culture with a xanthine oxidase inhibitor reduced the generation of both IL-33 and TSLP [68]. In vivo, the dispensation of a xanthine oxidase inhibitor reduced the generation of bronchoalveolar lavage fluid (BALF) IL-33 and decreased the number of ILC2s in the lungs of mice [68].
The role played by the IL-33/HMGB1 axis in respiratory infections has been confirmed in other experimental studies. IFN-beta promoter stimulator I (IPS-1) deficit influences susceptibility to viral bronchiolitis and provokes the onset of type 2 inflammation and airway alterations. The main reason for this condition is a deficiency in antiviral cytokine generation that can cause a significantly higher viral burden in epithelial cells, which die due to necrosis, and the production of IL-33 and HMGB1. This condition is associated with the increased generation of ILC2 type 2 cytokines, such as IL-5 and IL-13, and airway smooth muscle modification. Significantly, this type 2 inflammatory condition does not occur in adult animals with an IPS-1 deficit affected by infection, indicating that this condition is dependent on age and reflecting the human distribution of sensitivity to RSV bronchiolitis. The appearance of a type 2 immune response after an acute and severe alteration of epithelial function is coherent with its fundamental function in regulating lesion repair [69][70] as ILC2s, which are the principal ILC component in the lungs, are the main controllers of tissue damage repair [71][72].

2.3. Alarmins and Cystic Fibrosis

In CF, recurrent lung inflammation provokes increasing tissue injury. After cell damage and the onset of inflammation, necrotic cells discharge proteins as danger signals, which participate in the eradication of pathogens and stimulate cell repair [73]. However, the excessive liberation of such substances may increase inflammation, accelerating tissue alterations.
Considering the Th17/Th2 form of pulmonary phlogosis in CF patients and its correlation to CF-related pathogens, IL-33 and HMGB1 may participate in inflammation. Unlike the diffuse production of HMGB1, IL-33 is basically produced in epithelial cells [73] and has a role in tissue inflammation correlated to pathogen infection [74][75][76][77].
To evaluate the influence of IL-33 and HMGB1 in CF inflammation, researchers considered the IL-33 concentrations in the BALF of CF patients and evaluated subjects with frequent pulmonary infections. The IL-33 concentrations were significantly higher in CF subjects than in non-CF subjects, while the HMGB1 concentrations were higher in non-CF controls with repeated infections [78].
Furthermore, the Th2 cytokine pattern in CF patients has been correlated to a deficit of IFN-c [79], possibly due to an effect of IL-33. Moreover, IL-33 regulates neutrophil inflow, a specific aspect of CF. In addition, contact with Pseudomonas aeruginosa has been reported to stimulate IL-33 but not HMGB1 expression, and this finding might correlate this infection with an increased Th2 reaction [80]. Finally, other relationships between Il-33 and CF have been observed, such as an association between IL-33 expression and the lack of a CF transmembrane conductance regulator [80].

3. MicroRNAs and Chronic Respiratory Diseases

3.1. MicroRNAs and Asthma

The role played by miRNAs in respiratory diseases is closely linked to the specific miRNA, as the non-coding genetic material can play a triggering or protective role.
MiRNAs are involved in the genesis of asthma, as a relevant modification in the miRNA generation of airway cells has been described by numerous studies [81][82][83][84][85][86]. An analysis stated that epithelial miRNA-141 controlled the presence of airway mucus in asthma [86], while different studies implicated epithelial miRNAs in airway eosinophilia; moreover, an evaluation of miRNAs in asthma stated that a group of epithelial miRNAs was reduced in these subjects [86]. For instance, miRNA-30a-3pa, an miRNA also implicated in the growth and apoptosis of tumour cells [87][88], was considerably reduced in the blood of asthmatic subjects and is probably involved in airway eosinophilic inflammation.
Moreover, it was reported that miRNA-3934 concentrations might be employed to differentiate asthma subjects from non-asthmatic subjects. Regarding the mechanism, miRNA-3934 suppressed the increase in AGEs, which provoked an increase in apoptosis in basophils [89]. These findings suggest that miRNA-3934 can reduce the onset of asthma by targeting the RAGE and probably inhibiting other pathways, such as the TGF-β/Smad signalling pathway.
Even more interesting are the data relating to a possible correlation between miRNAs and HMGB1 in the pathogenesis of asthmatic disease. There are three runt-related transcriptional factor (RUNX) genes, namely RUNX1, RUNX2, and RUNX3, as well as maternal smoking, which have been reported to potentially support the onset of asthma in children by increasing the expression of RUNX1 [90]. Moreover, RUNX2 is described as stimulating the gene transcription of a sterile alpha motif (SAM) pointed domain comprising an ETS transcription factor (SPDEF) and may join the promoter of HMGB1 [91][92]. Wu et al. evaluated miRNA-30a-3p production in asthma patients and non-asthmatic subjects and considered the relationship between miRNA-30a-3p and airway eosinophilia [93]. They found that miRNA-30a-3p production was significantly reduced in the bronchial brushings of asthma patients compared to normal controls. Epithelial miRNA-30a-3p production was negatively related to several factors indicating airway eosinophilia, such as eosinophils in sputum or bronchial biopsies and exhaled nitric oxide in patients. The scholars demonstrated that RUNX2 is a target of miRNA-30a-3p and augments HMGB1 expression. HMGB1 and RUNX2 production are both increased in the airway epithelium and are related to each other in asthmatic subjects. A reduction in miRNA-30a-3p increased RUNX2 and HMGB1 production, while increasing RUNX2 stimulated HMGB1 in BEAS-2B cells. Interestingly, an airway increases in mmu-miRNA-30a-3p inhibited HMGB1 and RUNX2 expression and reduced airway eosinophilia in an experimental animal model [93]. Therefore, epithelial miRNA-30a-3p could target the RUNX2/HMGB1 axis to reduce airway eosinophilia in asthma (Figure 1).
Figure 1. Protective and negative effects of miRNAs on asthma. MiRNA-23a downregulates CXCL12 expression in fibroblasts [94]. In bronchial epithelial cells, it negatively regulates BLC2 expression, while miRNA-145-5p suppresses KIF3A [94][95]. These miRNAs promote bronchial inflammation and disease exacerbation. In contrast, protective effects are related to the action of miRNA-126-3p on the endothelial barrier through PIK3R2 [96]. Additionally, miRNA-3934 acts to mitigate IL-33 levels [89]. Magenta indicates downregulation, green indicates upregulation.

3.2. MicroRNAs and Acute Respiratory Distress Syndrome

ALI or its more critical manifestation, ARDS, is a severe acute pulmonary pathology with a significant mortality rate. ALI/ARDS is caused by different pulmonary conditions, such as pneumonia, or indirect extra-pulmonary damage, such as sepsis [97]. Its characteristics include alteration of the alveolar-capillary membrane, increased inflammation and reduced alveolar fluid clearance, with secondary hypoxemia, pulmonary oedema, and abnormal gas exchange [98][99][100].

MiRNAs and HMGB1 could also play a role in the genesis of ARDS. A report stated that the increased expression of miRNA-181b reduced the gene expression of importin-a3 and was able to decrease lung damage and mortality rates in ARDS animals [101]. Furthermore, Rao et al. showed that modification of the cytokine signalling suppressor 1 (SOCS1) in an animal model of ARDS reduced inflammatory cytokine production and inflammatory cell accumulation in miRNA-155 (−/−) animals compared to wild-type mice [102].

Finally, in different experiments, researchers assessed which miRNA could improve ARDS by targeting HMGB1 [103]. In experimental in vitro and in vivo models of LPS-induced ARDS models, they studied the effect of miRNA-574-5p on the production of HMGB1, pro-inflammatory cytokines and inflammasomes. MiRNA-574-5p seemed able to reduce the inflammatory reaction by operating on HMGB1. Stimulating the production of miRNA-574-5p or HMGB1 siRNA silencing reduced the stimulation of the NLRP3 inflammasome. Additionally, increased expression of HMGB1 overturned the anti-inflammatory action of miRNA-574-5p. In vivo, the increased production of miRNA-574-5p reduced interstitial oedema and alveolar leucocyte infiltration in ARDS animals [103].

4. Exosomal MicroRNA, Circular RNA, and Long Non-Coding RNA in Respiratory Diseases

Not only circulating miRNAs but also those contained in exosomes could play a role in respiratory diseases. Endothelial progenitor cells (EPCs) are an encouraging possibility for ALI treatment [104][105]. Some studies have demonstrated that EPCs can reduce inflammation and vascular leakage and increase bacterial elimination in ALI and infection-induced pulmonary damage [106][107][108][109]. Lately, exosomes have appeared as a relevant paracrine system that allows cell-to-cell communication by easing the transport of miRNAs from one cell to another [110].
The significant alteration of circRNAs has been demonstrated in respiratory diseases. For instance, circ_0007385 presence is greater in non-small cell lung cancer (NSCLC) cells and is correlated with a poor prognosis. Silencing circ_0007385 is possible to reduce cell growth and invasion in A549 and H1975 cells and decrease cisplatin resistance [111]. Furthermore, circ_0007385 silencing reduced the tumour proliferation of A549 cells in vivo. There was a direct interface between HMGB1, miRNA-519d-3p and circ_0007385. The production of miRNA-519d-3p was reduced in NSCLC in a circ_0007385-related manner, and circ_0007385 could secondarily control HMGB1 through miRNA-519d-3p. Thus, by both reducing miRNA-519d-3p and re-establishing HMGB1, it is possible to reverse the inhibitory action of circ_0007385 reduction in cell growth and invasion [111].
The significant alteration of circRNAs has been demonstrated in respiratory diseases. For instance, circ_0007385 presence is greater in non-small cell lung cancer (NSCLC) cells and is correlated with a poor prognosis. Silencing circ_0007385 is possible to reduce cell growth and invasion in A549 and H1975 cells and decrease cisplatin resistance [111]. Furthermore, circ_0007385 silencing reduced the tumour proliferation of A549 cells in vivo. There was a direct interface between HMGB1, miRNA-519d-3p and circ_0007385. The production of miRNA-519d-3p was reduced in NSCLC in a circ_0007385-related manner, and circ_0007385 could secondarily control HMGB1 through miRNA-519d-3p. Thus, by both reducing miRNA-519d-3p and re-establishing HMGB1, it is possible to reverse the inhibitory action of circ_0007385 reduction in cell growth and invasion [111].

References

  1. GBD Chronic Respiratory Disease Collaborators. Prevalence and attributable health burden of chronic respiratory diseases, 1990-2017: A systematic analysis for the Global Burden of Disease Study 2017. Lancet Respir. Med. 2020, 8, 585–596.
  2. Asher, M.I.; García-Marcos, L.; Pearce, N.E.; Strachan, D.P. Trends in worldwide asthma prevalence. Eur. Respir. J. 2020, 56, 2002094.
  3. Wang, C.; Xu, J.; Yang, L.; Xu, Y.; Zhang, X.; Bai, C.; Kang, J.; Ran, P.; Shen, H.; Wen, F.; et al. China Pulmonary Health Study Group. Prevalence and risk factors of chronic obstructive pulmonary disease in China (the China Pulmonary Health study): A national cross-sectional study. Lancet 2018, 391, 1706–1717.
  4. 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 A Cancer J. Clin. 2021, 71, 209–249.
  5. Lynch, D.A.; Sverzellati, N.; Travis, W.D.; Brown, K.K.; Colby, T.V.; Galvin, J.R.; Goldin, J.G.; Hansell, D.M.; Inoue, Y.; Johkoh, T.; et al. Diagnostic criteria for idiopathic pulmonary fibrosis: A Fleischner Society White Paper. Lancet Respir. Med. 2018, 6, 138–153.
  6. Wei, L.; Zhang, W.; Li, Y.; Zhai, J. The SIRT1-HMGB1 axis: Therapeutic potential to ameliorate inflammatory responses and tumor occurrence. Front. Cell Dev. Biol. 2022, 10, 986511.
  7. Lotze, M.T.; Tracey, K.J. High-mobility group box 1 protein (HMGB1): Nuclear weapon in the immune arsenal. Nat. Rev. Immunol. 2005, 5, 331–342.
  8. Lee, S.A.; Kwak, M.S.; Kim, S.; Shin, J.S. The role of high mobility group box 1 in innate immunity. Yonsei Med. J. 2014, 55, 1165–1176.
  9. Murdaca, G.; Allegra, A.; Paladin, F.; Calapai, F.; Musolino, C.; Gangemi, S. Involvement of Alarmins in the Pathogenesis and Progression of Multiple Myeloma. Int. J. Mol. Sci. 2021, 22, 9039.
  10. Li Pomi, F.; Borgia, F.; Custurone, P.; Vaccaro, M.; Pioggia, G.; Gangemi, S. Role of HMGB1 in Cutaneous Melanoma: State of the Art. Int. J. Mol. Sci. 2022, 23, 9327.
  11. Tang, D.; Kang, R.; Livesey, K.M.; Cheh, C.W.; Farkas, A.; Loughran, P.; Hoppe, G.; Bianchi, M.E.; Tracey, K.J.; Zeh, H.J.; et al. Endogenous HMGB1 regulates autophagy. J. Cell Biol. 2010, 190, 881–892.
  12. Ullah, M.A.; Loh, Z.; Gan, W.J.; Zhang, V.; Yang, H.; Li, J.H.; Yamamoto, Y.; Schmidt, A.M.; Armour, C.L.; Hughes, J.M.; et al. Receptor for advanced glycation end products and its ligand high-mobility group box-1 mediate allergic airway sensitization and airway inflammation. J. Allergy Clin. Immunol. 2014, 134, 440–450.
  13. Gardella, S.; Andrei, C.; Ferrera, D.; Lotti, L.V.; Torrisi, M.R.; Bianchi, M.E.; Rubartelli, A. The nuclear protein HMGB1 is secreted by monocytes via a non-classical, vesicle-mediated secretory pathway. EMBO Rep. 2002, 3, 995–1001.
  14. Park, J.S.; Gamboni-Robertson, F.; He, Q.; Svetkauskaite, D.; Kim, J.Y.; Strassheim, D.; Sohn, J.W.; Yamada, S.; Maruyama, I.; Banerjee, A.; et al. High mobility group box 1 protein interacts with multiple Toll-like receptors. Am. J. Physiol. Cell Physiol. 2006, 290, C917–C924.
  15. Treutiger, C.J.; Mullins, G.E.; Johansson, A.S.; Rouhiainen, A.; Rauvala, H.M.; Erlandsson-Harris, H.; Andersson, U.; Yang, H.; Tracey, K.J.; Andersson, J.; et al. High mobility group 1 B-box mediates activation of human endothelium. J. Intern. Med. 2003, 254, 375–385.
  16. Huebener, P.; Pradere, J.P.; Hernandez, C.; Gwak, G.Y.; Caviglia, J.M.; Mu, X.; Loike, J.D.; Jenkins, R.E.; Antoine, D.J.; Schwabe, R.F. The HMGB1/RAGE axis triggers neutrophil-mediated injury amplification following necrosis. J. Clin. Investig. 2019, 130, 1802.
  17. Taverna, S.; Tonacci, A.; Ferraro, M.; Cammarata, G.; Cuttitta, G.; Bucchieri, S.; Pace, E.; Gangemi, S. High Mobility Group Box 1: Biological Functions and Relevance in Oxidative Stress Related Chronic Diseases. Cells 2022, 11, 849.
  18. Stark, K.; Philippi, V.; Stockhausen, S.; Busse, J.; Antonelli, A.; Miller, M.; Schubert, I.; Hoseinpour, P.; Chandraratne, S.; von Brühl, M.L.; et al. Disulfide HMGB1 derived from platelets coordinates venous thrombosis in mice. Blood 2016, 128, 2435–2449.
  19. Yu, Y.; Tang, D.; Kang, R. Oxidative stress-mediated HMGB1 biology. Front. Physiol. 2015, 6, 93.
  20. Maugeri, N.; Franchini, S.; Campana, L.; Baldini, M.; Ramirez, G.A.; Sabbadini, M.G.; Rovere-Querini, P.; Manfredi, A.A. Circulating platelets as a source of the damage-associated molecular pattern HMGB1 in patients with systemic sclerosis. Autoimmunity 2012, 45, 584–587.
  21. Ahrens, I.; Chen, Y.C.; Topcic, D.; Bode, M.; Haenel, D.; Hagemeyer, C.E.; Seeba, H.; Duerschmied, D.; Bassler, N.; Jandeleit-Dahm, K.A.; et al. HMGB1 binds to activated platelets via the receptor for advanced glycation end products and is present in platelet rich human coronary artery thrombi. Thromb. Haemost. 2015, 114, 994–1003.
  22. Carriere, V.; Roussel, L.; Ortega, N.; Lacorre, D.A.; Americh, L.; Aguilar, L.; Bouche, G.; Girard, J.P. IL-33, the IL-1-like cytokine ligand for ST2 receptor, is a chromatin-associated nuclear factor in vivo. Proc. Natl. Acad. Sci. USA 2007, 104, 282–287.
  23. Liew, F.Y.; Girard, J.P.; Turnquist, H.R. Interleukin-33 in health and disease. Nat. Rev. Immunol. 2016, 16, 676–689.
  24. Qi, F.; Bai, S.; Wang, D.; Xu, L.; Hu, H.; Zeng, S.; Chai, R.; Liu, B. Macrophages produce IL-33 by activating MAPK signaling pathway during RSV infection. Mol. Immunol. 2017, 87, 284–292.
  25. Murdaca, G.; Greco, M.; Tonacci, A.; Negrini, S.; Borro, M.; Puppo, F.; Gangemi, S. IL-33/IL-31 Axis in Immune-Mediated and Allergic Diseases. Int. J. Mol. Sci. 2019, 20, 5856.
  26. Di Salvo, E.; Casciaro, M.; Gangemi, S. IL-33 genetics and epigenetics in immune-related diseases. Clin. Mol. Allergy 2021, 19, 18.
  27. Allegra, A.; Alonci, A.; Campo, S.; Penna, G.; Petrungaro, A.; Gerace, D.; Musolino, C. Circulating microRNAs: New biomarkers in diagnosis, prognosis and treatment of cancer (review). Int. J. Oncol. 2012, 41, 1897–1912.
  28. Musolino, C.; Oteri, G.; Allegra, A.; Mania, M.; D’Ascola, A.; Avenoso, A.; Innao, V.; Allegra, A.G.; Campo, S. Altered microRNA expression profile in the peripheral lymphoid compartment of multiple myeloma patients with bisphosphonate-induced osteonecrosis of the jaw. Ann. Hematol. 2018, 97, 1259–1269.
  29. Mott, J.L.; Mohr, A.M. Overview of MicroRNA biology. Semin. Liver Dis. 2015, 35, 3–11.
  30. Chevillet, J.R.; Lee, I.; Briggs, H.A.; He, Y.; Wang, K. Issues and prospects of microRNA-based biomarkers in blood and other body fluids. Molecules 2014, 19, 6080–6105.
  31. Tomasello, L.; Distefano, R.; Nigita, G.; Croce, C.M. The MicroRNA family gets wider: The IsomiRs classifcation and role. Front. Cell Dev. Biol. 2021, 9, 668648.
  32. O’Brien, J.; Hayder, H.; Zayed, Y.; Peng, C. Overview of MicroRNA biogenesis, mechanisms of actions, and circulation. Front. Endocrinol. 2018, 9, 402.
  33. Papi, A.; Brightling, C.; Pedersen, S.E.; Reddel, H.K. Asthma. Lancet 2018, 391, 783–800.
  34. Bloom, C.; de Preux, L.; Sheikh, A.; Quint, J. Health and cost impact of stepping down asthma medication for UK patients, 2001–2017: A population-based observational study. PLoS Med. 2020, 17, e1003145.
  35. Hoshi, M.; Matsunaga, M.; Nogami, K.; Hamada, K.; Kobori, T.; Kainuma, K.; Nagao, M.; Fujisawa, T. Three cases of severe adolescent asthma treated with mepolizumab: Lung function trajectories. Asia Pac. Allergy 2020, 10, e13.
  36. Fahy, J.V. Type 2 inflammation in asthma–present in most, absent in many. Nat. Rev. Immunol. 2015, 15, 57–65.
  37. Kubo, M. Innate and adaptive type 2 immunity in lung allergic inflammation. Immunol. Rev. 2017, 278, 162–172.
  38. Kim, H.Y.; De Kruyff, R.H.; Umetsu, D.T. The many paths to asthma: Phenotype shaped by innate and adaptive immunity. Nat. Immunol. 2010, 11, 577–584.
  39. Pulendran, B.; Artis, D. New paradigms in type 2 immunity. Science 2012, 337, 431–435.
  40. Halim, T.Y.F.; McKenzie, A.N.J. New kids on the block: Group 2 innate lymphoid cells and type 2 inflammation in the lung. Chest 2013, 144, 1681–1686.
  41. Neill, D.R.; Wong, S.H.; Bellosi, A.; Flynn, R.J.; Daly, M.; Langford, T.K.; Bucks, C.; Kane, C.M.; Fallon, P.G.; Pannell, R.; et al. Nuocytes represent a new innate effector leukocyte that mediates type-2 immunity. Nature 2010, 464, 1367–1370.
  42. Price, A.E.; Liang, H.E.; Sullivan, B.M.; Reinhardt, R.L.; Eisley, C.J.; Erle, D.J.; Locksley, R.M. Systemically dispersed innate IL-13-expressing cells in type 2 immunity. Proc. Natl. Acad. Sci. USA 2010, 107, 11489–11494.
  43. Fallon, P.G.; Ballantyne, S.J.; Mangan, N.E.; Barlow, J.L.; Dasvarma, A.; Hewett, D.R.; McIlgorm, A.; Jolin, H.E.; McKenzie, A.N. Identification of an interleukin (IL)-25-dependent cell population that provides IL-4, IL-5, and IL-13 at the onset of helminth expulsion. J. Exp. Med. 2006, 203, 1105–1116.
  44. Herbert, D.R.; Douglas, B.; Zullo, K. Group 2 Innate Lymphoid Cells (ILC2): Type 2 immunity and Helminth immunity. Int. J. Mol. Sci. 2019, 20, 2276.
  45. Halim, T.Y.; Krauss, R.H.; Sun, A.C.; Takei, F. Lung natural helper cells are a critical source of Th2 cell-type cytokines in protease allergen-induced airway inflammation. Immunity 2012, 36, 451–463.
  46. Mjösberg, J.M.; Trifari, S.; Crellin, N.K.; Peters, C.P.; van Drunen, C.M.; Piet, B.; Fokkens, W.J.; Cupedo, T.; Spits, H. Human IL-25- and IL-33-responsive type 2 innate lymphoid cells are defined by expression of CRTH2 and CD161. Nat. Immunol. 2011, 12, 1055–1062.
  47. Cayrol, C.; Girard, J.P. The IL-1-like cytokine IL-33 is inactivated after maturation by caspase-1. Proc. Natl. Acad. Sci. USA 2009, 106, 9021902–9021906.
  48. Kulkarni, N.S.; Hollins, F.; Sutcliffe, A.; Saunders, R.; Shah, S.; Siddiqui, S.; Gupta, S.; Haldar, P.; Green, R.; Pavord, I.; et al. Eosinophil protein in airway macrophages: A novel biomarker of eosinophilic inflammation in patients with asthma. J. Allergy Clin. Immunol. 2010, 126, 61–69.e3.
  49. Suzukawa, M.; Morita, H.; Nambu, A.; Arae, K.; Shimura, E.; Shibui, A.; Yamaguchi, S.; Suzukawa, K.; Nakanishi, W.; Oboki, K.; et al. Epithelial cell-derived IL-25, but not Th17 cell-derived IL-17 or IL-17F, is crucial for murine asthma. J. Immunol. 2012, 189, 3641–3652.
  50. Soumelis, V.; Reche, P.A.; Kanzler, H.; Yuan, W.; Edward, G.; Homey, B.; Gilliet, M.; Ho, S.; Antonenko, S.; Lauerma, A.; et al. Human epithelial cells trigger dendritic cell mediated allergic inflammation by producing TSLP. Nat. Immunol. 2002, 3, 673–680.
  51. Lai, Y.; Altemeier, W.A.; Vandree, J.; Piliponsky, A.M.; Johnson, B.; Appel, C.L.; Frevert, C.W.; Hyde, D.M.; Ziegler, S.F.; Smith, D.E.; et al. Increased density of intraepithelial mast cells in patients with exercise-induced bronchoconstriction regulated through epithelially derived thymic stromal lymphopoietin and IL-33. J. Allergy Clin. Immunol. 2014, 133, 1448–1455.
  52. Wenzel, S.E. Asthma Phenotypes: The Evolution from Clinical to Molecular Approaches. Nat. Med. 2012, 18, 716–725.
  53. Lee, H.W.; Baek, M.G.; Choi, S.; Ahn, Y.H.; Bang, J.Y.; Sohn, K.H.; Kang, M.G.; Jung, J.W.; Choi, J.H.; Cho, S.H.; et al. Peripheral blood transcriptomic clusters uncovered immune phenotypes of asthma. Respir. Res. 2022, 23, 237.
  54. Werder, R.B.; Ullah, M.A.; Rahman, M.M.; Simpson, J.; Lynch, J.P.; Collinson, N.; Rittchen, S.; Rashid, R.B.; Sikder, M.A.A.; Handoko, H.Y.; et al. Targeting the P2Y13 Receptor Suppresses IL-33 and HMGB1 Release and Ameliorates Experimental Asthma. Am. J. Respir. Crit. Care Med. 2022, 205, 300–312.
  55. Yao, X.; Xie, R.; Cao, Y.; Tang, J.; Men, Y.; Peng, H.; Yang, W. Simvastatin induced ferroptosis for triple-negative breast cancer therapy. J. Nanobiotechnol. 2021, 19, 311.
  56. Stockwell, B.R.; Friedmann Angeli, J.P.; Bayir, H.; Bush, A.I.; Conrad, M.; Dixon, S.J.; Fulda, S.; Gascón, S.; Hatzios, S.K.; Kagan, V.E.; et al. Ferroptosis: A Regulated Cell Death Nexus Linking Metabolism, Redox Biology, and Disease. Cell 2017, 171, 273–285.
  57. Allinne, J.; Scott, G.; Lim, W.K.; Birchard, D.; Erjefält, J.S.; Sandén, C.; Ben, L.H.; Agrawal, A.; Kaur, N.; Kim, J.H.; et al. IL-33 blockade affects mediators of persistence and exacerbation in a model of chronic airway inflammation. J. Allergy Clin. Immunol. 2019, 144, 1624–1637.e10.
  58. Smith, D.K.; Seales, S.; Budzik, C. Respiratory syncytial virus bronchiolitis in children. Am. Fam. Physician 2017, 95, 94–99.
  59. Walsh, E.E. Respiratory Syncytial Virus Infection: An Illness for All Ages. Clin. Chest Med. 2017, 38, 29–36.
  60. Johnson, J.E.; Gonzales, R.A.; Olson, S.J.; Wright, P.F.; Graham, B.S. The histopathology of fatal untreated human respiratory syncytial virus infection. Mod. Pathol. 2007, 20, 108–119.
  61. Piedimonte, G.; Perez, M.K. Respiratory syncytial virus infection and bronchiolitis. Pediatr. Rev. 2014, 35, 519–530.
  62. Plesca, D.A.; Cora, F.; Buzoianu, E.; Moiceanu, M.; Hurduc, V. Risk factors for severe bronchiolitis—A retrospective study. Eur. Respir. J. 2012, 40, 4660.
  63. Borchers, A.T.; Chang, C.; Gershwin, M.E.; Gershwin, L.J. Respiratory syncytial virus—A comprehensive review. Clin. Rev. Allergy Immunol. 2013, 45, 331–379.
  64. Hashem, M.; Hall, C.B. Respiratory syncytial virus in healthy adults: The cost of a cold. J. Clin. Virol. 2003, 27, 14–21.
  65. Camelo, A.; Rosignoli, G.; Ohne, Y.; Stewart, R.A.; Overed-Sayer, C.; Sleeman, M.A.; May, R.D. IL-33, IL-25, and TSLP induce a distinct phenotypic and activation profile in human type 2 innate lymphoid cells. Blood Adv. 2017, 1, 577–589.
  66. Hinde, H.L.; Wu, Q.; Bentley, J.K.; Han, M.B.M.; Rajput, C.; Hong, J.Y.; Lei, J. The Innate Cytokines IL-25, IL-33, and TSLP Cooperate in the Induction of Type 2 Innate Lymphoid Cell Expansion and Mucous Metaplasia in Rhinovirus-Infected Immature Mice. J. Immunol. 2017, 199, 1308–1318.
  67. Zhang, K.; Jin, Y.; Lai, D.; Wang, J.; Wang, Y.; Wu, X.; Scott, M.; Li, Y.; Hou, J.; Billiar, T.; et al. RAGE-induced ilc2 expansion in acute lung injury due to haemorrhagic shock. Thorax 2020, 75, 209–219.
  68. Fonseca, W.; Malinczak, C.-A.; Schuler, C.F.; Best, S.K.K.; Rasky, A.J.; Morris, S.B.; Cui, T.X.; Popova, A.P.; Lukacs, N.W. Uric acid pathway activation during respiratory virus infection promotes Th2 immune response via innate cytokine production and ILC2 accumulation. Mucosal Immunol. 2020, 13, 691–701.
  69. Gause, W.C.; Wynn, T.A.; Allen, J.E. Type 2 immunity and wound healing: Evolutionary refinement of adaptive immunity by helminths. Nat. Rev. Immunol. 2013, 13, 607–614.
  70. Phipps, S.; Ying, S.; Wangoo, A.; Ong, Y.E.; Levi-Schaffer, F.; Kay, A.B. The Relationship Between Allergen-Induced Tissue Eosinophilia and Markers of Repair and Remodeling in Human Atopic Skin. J. Immunol. 2002, 169, 4604–4612.
  71. Monticelli, L.A.; Sonnenberg, G.F.; Abt, M.C.; Alenghat, T.; Ziegler, C.G.; Doering, T.A.; Angelosanto, J.M.; Laidlaw, B.J.; Yang, C.Y.; Sathaliyawala, T.; et al. Innate lymphoid cells promote lung-tissue homeostasis after infection with influenza virus. Nat. Immunol. 2011, 12, 1045–1054.
  72. Sonnenberg, G.F.; Artis, D. Innate lymphoid cells in the initiation, regulation and resolution of inflammation. Nat. Med. 2015, 21, 698–708.
  73. Hirsiger, S.; Simmen, H.P.; Werner, C.M.; Wanner, G.A.; Rittirsch, D. Danger signals activating the immune response after trauma. Mediat. Inflamm. 2012, 2012, 315941.
  74. Akdis, M.; Burgler, S.; Crameri, R.; Eiwegger, T.; Fujita, H.; Gomez, E.; Klunker, S.; Meyer, N.; O’Mahony, L.; Palomares, O.; et al. Interleukins, from 1 to 37, and interferon-γ: Receptors, functions, and roles in diseases. J. Allergy Clin. Immunol. 2011, 127, e1–e70.
  75. Roussel, L.; Farias, R.; Rousseau, S. IL-33 is expressed in epithelia from patients with cystic fibrosis and potentiates neutrophil recruitment. J. Allergy Clin. Immunol. 2013, 131, 913–916.
  76. Schmitz, J.; Owyang, A.; Oldham, E.; Song, Y.; Murphy, E.; McClanahan, T.K.; Zurawski, G.; Moshrefi, M.; Qin, J.; Li, X.; et al. IL-33, an interleukin-1-like cytokine that signals via the IL-1 receptor-related protein ST2 and induces T helper type 2-associated cytokines. Immunity 2005, 23, 479–490.
  77. Yanai, H.; Ban, T.; Taniguchi, T. High-mobility group box family of proteins: Ligand and sensor for innate immunity. Trends Immunol. 2012, 33, 633–640.
  78. Rowe, S.M.; Jackson, P.L.; Liu, G.; Hardison, M.; Livraghi, A.; Solomon, G.M.; McQuaid, D.B.; Noerager, B.D.; Gaggar, A.; Clancy, J.P.; et al. Potential role of high-mobility group box 1 in cystic fibrosis airway disease. Am. J. Respir. Crit. Care Med. 2008, 178, 822–831.
  79. Brazova, J.; Sediva, A.; Pospisilova, D.; Vavrova, V.; Pohunek, P.; Macek, M., Jr.; Bartunkova, J.; Lauschmann, H. Differential cytokine profile in children with cystic fibrosis. Clin. Immunol. 2005, 115, 210–215.
  80. Hartl, D.; Griese, M.; Kappler, M.; Zissel, G.; Reinhardt, D.; Rebhan, C.; Schendel, D.J.; Krauss-Etschmann, S. Pulmonary T(H)2 response in Pseudomonas aeruginosa-infected patients with cystic fibrosis. J. Allergy Clin. Immunol. 2006, 117, 204–211.
  81. Heffler, E.; Allegra, A.; Pioggia, G.; Picardi, G.; Musolino, C.; Gangemi, S. MicroRNA Profiling in Asthma: Potential Biomarkers and Therapeutic Targets. Am. J. Respir. Cell Mol. Biol. 2017, 57, 642–650.
  82. Bartel, S.; Carraro, G.; Alessandrini, F.; Krauss-Etschmann, S.; Ricciardolo, F.L.M.; Bellusci, S. miR-142-3p is associated with aberrant WNT signaling during airway remodeling in asthma. Am. J. Physiol. Lung Cell Mol. Physiol. 2018, 315, 328–333.
  83. Pua, H.H.; Happ, H.C.; Gray, C.J.; Mar, D.J.; Chiou, N.T.; Hesse, L.E.; Ansel, K.M. Increased hematopoietic extracellular RNAs and vesicles in the lung during allergic airway responses. Cell Rep. 2019, 26, 933–944.e934.
  84. Zhang, K.; Liang, Y.; Feng, Y.; Wu, W.; Zhang, H.; He, J.; Hu, Q.; Zhao, J.; Xu, Y.; Liu, Z.; et al. Decreased epithelial and sputum miR-221-3p associates with airway eosinophilic inflammation and CXCL17 expression in asthma. Am. J. Physiol. Lung Cell Mol. Physiol. 2018, 315, 253–264.
  85. Liang, Y.; Feng, Y.; Wu, W.; Chang, C.; Chen, D.; Chen, S.; Zhen, G. microRNA-218-5p plays a protective role in eosinophilic airway inflammation via targeting δ-catenin, a novel catenin in asthma. Clin. Exp. Allergy 2020, 50, 29–40.
  86. Huo, X.; Zhang, K.; Yi, L.; Mo, Y.; Liang, Y.; Zhao, J.; Zhang, Z.; Xu, Y.; Zhen, G. Decreased epithelial and plasma miR-181b-5p expression associates with airway eosinophilic inflammation in asthma. Clin. Exp. Allergy 2016, 46, 1281–1290.
  87. Niu, Z.-R.; Han, T.; Sun, X.-L.; Luan, L.-X.; Gou, W.-L.; Zhu, X.-M. MicroRNA-30a-3p is overexpressed in the placentas of patients with preeclampsia and affects trophoblast invasion and apoptosis by its effects on IGF-1. Am. J. Obstet. Gynecol. 2018, 218, 249.e1–249.e12.
  88. Wang, Y.; Wang, F.; He, J.; Du, J.; Zhang, H.; Shi, H.; Chen, Y.; Wei, Y.; Xue, W.; Yan, J.; et al. miR-30a-3p targets MAD2L1 and regulates proliferation of gastric cancer cells. OncoTargets Ther. 2019, 12, 11313–11324.
  89. Dou, L.; Wang, W.; Wang, J.; Zhang, X.; Hu, X.; Zheng, W.; Han, K.; Wang, G. miR-3934 regulates the apoptosis and secretion of inflammatory cytokines of basophils via targeting RAGE in asthma. Allergy Asthma Clin. Immunol. 2022, 18, 66.
  90. Haley, K.J.; Lasky-Su, J.; Manoli, S.E.; Smith, L.A.; Shahsafaei, A.; Weiss, S.T.; Tantisira, K. RUNX transcription factors: Association with pediatric asthma and modulated by maternal smoking. Am. J. Physiol. Lung Cell Mol. Physiol. 2011, 301, 693–701.
  91. Shi, N.; Zhang, J.; Chen, S.-Y. Runx2, a novel regulator for goblet cell differentiation and asthma development. FASEB J. 2017, 31, 412–420.
  92. Van der Deen, M.; Akech, J.; Lapointe, D.; Gupta, S.; Young, D.W.; Montecino, M.A.; Galindo, M.; Lian, J.B.; Stein, J.L.; Stein, G.S.; et al. Genomic promoter occupancy of runt-related transcription factor RUNX2 in Osteosarcoma cells identifies genes involved in cell adhesion and motility. J. Biol. Chem. 2012, 287, 4503–4517.
  93. Wu, W.; Gao, J.; Chen, D.; Chen, G.; Feng, Y.; Chang, C.; Chen, S.; Yi, L.; Zhen, G. Epithelial microRNA-30a-3p targets RUNX2/HMGB1 axis to suppress airway eosinophilic inflammation in asthma. Respir. Res. 2022, 23, 17.
  94. Jin, A.; Bao, R.; Roth, M.; Liu, L.; Yang, X.; Tang, X.; Yang, X.; Sun, Q.; Lu, S. microRNA-23a contributes to asthma by targeting BCL2 in airway epithelial cells and CXCL12 in fibroblasts. J. Cell Physiol. 2019, 234, 21153–21165.
  95. Xiong, T.; Du, Y.; Fu, Z.; Geng, G. MicroRNA-145-5p promotes asthma pathogenesis by inhibiting kinesin family member 3A expression in mouse airway epithelial cells. J. Int. Med. Res. 2019, 47, 3307–3319.
  96. Xi, T.; Jin, F.; Zhu, Y.; Wang, J.; Tang, L.; Wang, Y.; Liebeskind, D.S.; He, Z. MicroRNA-126-3p attenuates blood-brain barrier disruption, cerebral edema and neuronal injury following intracerebral hemorrhage by regulating PIK3R2 and Akt. Biochem Biophys. Res. Commun. 2017, 494, 144–151.
  97. Weinberger, S.E.; Cockrill, B.A.; Mandel, J. Acute respiratory distress syndrome. In Principles of Pulmonary Medicine; Elsevier: Amsterdam, The Netherlands, 2019; pp. 357–369.
  98. Xu, B.; Chen, S.S.; Liu, M.Z.; Gan, C.X.; Li, J.Q.; Guo, G.H. Stem cell derived exosomes-based therapy for acute lung injury and acute respiratory distress syndrome: A novel therapeutic strategy. Life Sci. 2020, 254, 117766.
  99. Thompson, B.T.; Chambers, R.C.; Liu, K.D. Acute respiratory distress syndrome. N. Engl. J. Med. 2017, 377, 562–572.
  100. Rawal, G.; Yadav, S.; Kumar, R. Acute respiratory distress syndrome: An update and review. J. Transl. Int. Med. 2018, 6, 74–77.
  101. Sun, X.; Icli, B.; Wara, A.K.; Belkin, N.; He, S.; Kobzik, L.; Hunninghake, G.M.; Vera, M.P.; MICU Registry. MicroRNA-181b regulates NF-kB-mediated vascular inflammation. J. Clin. Investig. 2012, 122, 1973–1990.
  102. Rao, R.; Rieder, S.A.; Nagarkatti, P.; Nagarkatti, M. Staphylococcal enterotoxin B-induced microRNA-155 targets SOCS1 to promote acute inflammatory lung injury. Infect. Immun. 2014, 82, 2971–2979.
  103. He, B.; Zhou, W.; Rui, Y.; Liu, L.; Chen, B.; Su, X. MicroRNA-574-5p Attenuates Acute Respiratory Distress Syndrome by Targeting HMGB1. Am. J. Respir. Cell Mol. Biol. 2021, 64, 196–207.
  104. Lee, J.W.; Zhu, Y.; Matthay, M.A. Cell-based therapy for acute lung injury: Are we there yet? Anesthesiology 2012, 116, 1189–1191.
  105. Kawasaki, T.; Nishiwaki, T.; Sekine, A.; Nishimura, R.; Suda, R.; Urushibara, T.; Suzuki, T.; Takayanagi, S.; Terada, J.; Sakao, S.; et al. Vascular repair by tissue resident endothelial progenitor cells in endotoxin-induced lung injury. Am. J. Respir. Cell Mol. Biol. 2015, 53, 500–512.
  106. Cao, J.P.; He, X.Y.; Xu, H.T.; Zou, Z.; Shi, X.Y. Autologous transplantation of peripheral blood-derived circulating endothelial progenitor cells attenuates endotoxin-induced acute lung injury in rabbits by direct endothelial repair and indirect immunomodulation. Anesthesiology 2012, 116, 1278–1287.
  107. Lam, C.F.; Liu, Y.C.; Hsu, J.K.; Yeh, P.A.; Su, T.Y.; Huang, C.C.; Lin, M.W.; Wu, P.C.; Chang, P.J.; Tsai, Y.C. Autologous transplantation of endothelial progenitor cells attenuates acute lung injury in rabbits. Anesthesiology 2008, 108, 392–401.
  108. Mao, M.; Wang, S.N.; Lv, X.J.; Wang, Y.; Xu, J.C. Intravenous delivery of bone marrow-derived endothelial progenitor cells improves survival and attenuates lipopolysaccharide-induced lung injury in rats. Shock 2010, 34, 196–204.
  109. Fan, H.; Goodwin, A.J.; Chang, E.; Zingarelli, B.; Borg, K.; Guan, S.; Halushka, P.V.; Cook, J.A. Endothelial progenitor cells and a SDF-1alpha analogue synergistically improve survival in sepsis. Am. J. Respir. Crit. Care Med. 2014, 189, 1509–1519.
  110. Valadi, H.; Ekstrom, K.; Bossios, A.; Sjostrand, M.; Lee, J.J.; Lotvall, J.O. Exosome mediated transfer of mRNAs and microRNAs is a novel mechanism of genetic exchange between cells. Nat. Cell Biol. 2007, 9, 654–659.
  111. Ye, Y.; Zhao, L.; Li, Q.; Xi, C.; Li, Y.; Li, Z. circ_0007385 served as competing endogenous RNA for miR-519d-3p to suppress malignant behaviors and cisplatin resistance of non-small cell lung cancer cells. Thorac. Cancer 2020, 11, 2196–2208.
More
Information
Subjects: Immunology
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: 388
Revisions: 3 times (View History)
Update Date: 09 Feb 2023
1000/1000