Next Article in Journal
Golgi’s Role in the Development of Possible New Therapies in Cancer
Previous Article in Journal
Extracellular Vesicles Released by Genetically Modified Macrophages Activate Autophagy and Produce Potent Neuroprotection in Mouse Model of Lysosomal Storage Disorder, Batten Disease
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Air Pollution: Role of Extracellular Vesicles-Derived Non-Coding RNAs in Environmental Stress Response

by
Giuseppa D’Amico
1,†,
Radha Santonocito
1,†,
Alessandra Maria Vitale
1,2,†,
Federica Scalia
1,2,
Antonella Marino Gammazza
1,
Claudia Campanella
1,
Fabio Bucchieri
1,
Francesco Cappello
1,2 and
Celeste Caruso Bavisotto
1,2,*
1
Section of Human Anatomy and Histology, Department of Biomedicine, Neuroscience and Advanced Diagnostics (BIND), University of Palermo, 90133 Palermo, Italy
2
Euro-Mediterranean Institute of Science and Technology (IEMEST), 90139 Palermo, Italy
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Cells 2023, 12(11), 1498; https://doi.org/10.3390/cells12111498
Submission received: 31 March 2023 / Revised: 17 May 2023 / Accepted: 27 May 2023 / Published: 29 May 2023

Abstract

:
Air pollution has increased over the years, causing a negative impact on society due to the many health-related problems it can contribute to. Although the type and extent of air pollutants are known, the molecular mechanisms underlying the induction of negative effects on the human body remain unclear. Emerging evidence suggests the crucial involvement of different molecular mediators in inflammation and oxidative stress in air pollution-induced disorders. Among these, non-coding RNAs (ncRNAs) carried by extracellular vesicles (EVs) may play an essential role in gene regulation of the cell stress response in pollutant-induced multiorgan disorders. This review highlights EV-transported ncRNAs’ roles in physiological and pathological conditions, such as the development of cancer and respiratory, neurodegenerative, and cardiovascular diseases following exposure to various environmental stressors.

1. Discovery and Classification of Non-Coding RNAs

Between the late 1950s and early 1960s, Crick, Jacob, and Monod devised the “central dogma” of molecular biology, stating that genetic information flows only in one direction, from DNA to RNA and RNA to protein [1,2,3]. However, over the past decades, many exceptions to this dogma have been reported demonstrating that, even if almost all the human genome is actively transcribed, only ~1.5% of it is made up of protein-coding genes, which are first transcribed into messenger RNAs (mRNAs) [4,5,6,7]. The rest of the transcribed elements were collectively called non-coding RNAs (ncRNAs) [8], which were initially considered “junk” RNA with unknown functionality. However, this concept was rapidly withdrawn since increasing evidence demonstrated the involvement of ncRNAs in multiple critical biological events, such as development, differentiation, cellular homeostasis, and the onset of disease [9,10,11]. ncRNAs are mainly distinguished into two main classes: housekeeping and regulatory ncRNAs (Table 1). The first ones, constitutively expressed in all cell types and necessary for cell viability, are ribosomal RNAs (rRNAs), transfer RNAs (tRNAs), small nuclear RNAs (snRNAs), small nucleolar RNAs (snoRNAs), and telomerase RNAs (TER) [12]. In addition to housekeeping ncRNAs, an increasing number of regulatory ncRNAs have been identified and characterized thanks to the rapid development of in-depth and high-throughput transcriptome sequencing techniques. Regulatory ncRNAs function as regulators of gene expression at different levels: epigenetic, transcriptional, and post-transcriptional; and are classified according to their size into small ncRNAs (sncRNAs) that have less than 200 nucleotides (nts), such as microRNAs (miRNAs), small interfering RNAs (siRNAs), piwi-interacting RNAs (piRNAs), and long ncRNAs (lncRNAs) that have more than 200 nts [11,12] (Table 1). The first evidence that RNA may function as a regulator of gene expression came from experiments with prokaryotes (bacteria) over forty years ago, in the 1980s, when the Escherichia coli regulatory micF ncRNA was first identified, and its abilities to hybridize with a complementary target mRNA (ompF mRNA) and to inhibit its translation were described. It was the first RNA-regulating gene expression through sense-antisense base pairing to be reported [13,14,15]. However, ncRNAs represent only a tiny fraction of prokaryotes’ genomes, most containing protein-coding sequences [8]. Instead, the genome of higher multicellular eukaryotic organisms has fewer protein-coding sequences that diminish with increasing complexity. On the contrary, there is a large amount of non-coding intergenic and intronic sequences, which are also transcribed [16,17].

2. Biogenesis and Role of Regulatory ncRNAs under Physiological Conditions

Regulatory ncRNAs control various physiological pathways, such as development, differentiation, and cellular homeostasis.
siRNAs form a class of double-stranded (ds) RNA molecules (~20–25 nts) able to silence target genes post-transcriptionally. Based on their origin, they can be divided into two subcategories: exogenous siRNA (exo-siRNA), originating from exogenous ds-nucleic acid resulting from an artificial insertion or virus infections, and endogenous siRNAs (endo-siRNA), originating from the endogenous genomic locus, and transcribed from transposon elements [18,19]. In both cases, the precursor dsRNA is enzymatically processed to produce a smaller ds-siRNA, which is loaded in a RISC complex and then unwound, leading to the formation of a functional RISC, mediating the target mRNA cleavage and degradation, ultimately resulting in gene silencing [20].
miRNAs, the most studied and abundant class of small ncRNAs, are endogenous short non-coding single-stranded (ss)-RNAs (~22 nts), which negatively regulate the expression of hundreds of target genes post-transcriptionally by affecting either the stability or the translation of their mRNAs [21]. Similarly to endo-siRNAs, they originate as longer primary transcripts (pri-miRNAs), which undergo two highly regulated cleavage events, resulting in the formation of a miRNA-induced silencing complex (miRISC), targeting a specific mRNA and causing mRNA cleavage or translation inhibition [22,23,24,25].
To date, thousands of miRNAs from different organisms have been identified. In humans, over 2000 miRNAs have been annotated and validated, which regulate many protein-coding genes, and, thus, many biological events, both in normal cellular homeostasis and disease [26,27,28,29,30]. The critical role of miRNAs in cell differentiation and organisms’ development appeared clear ever since the discovery of the first miRNA, controlling the timing of larval development in C. elegans [31,32,33]. One of the earliest developmental events is the switch from pluripotent to lineage-specified cells, achieved through the downregulation of pluripotency markers, decreased self-renewal potential, and the subsequent activation of a lineage-specific gene expression pattern. All these changes are orchestrated and regulated by various miRNAs. DGCR8 depletion in embryonic stem cells hinders the downregulation of pluripotency markers and allows the expression of lineage-specific genes [34]. In this regard, in humans, the miR-302 family members are exclusively expressed during gastrulation and play a key role in promoting the mesendoderm formation and suppressing the neuroectoderm lineage by acting on LEFTY1 and LEFTY2 regulatory factors of the Nodal signaling pathway [35]. Many identified miRNAs have been found expressed at specific differentiation and developmental stages of certain cells and tissues, suggesting their involvement in these pathways. For instance, several miRNAs are exclusively or highly expressed in the brain tissue. The simultaneous overexpression of four of them (miR-124a, miR-9, miR-125b, and miR-22) in neuronal progenitor cells reduced the number of astroglia-like cells and increased that of neuronal cells. It was suggested that this event might result from the miRNAs-driven downregulation of non-neuronal transcripts [36]. Particularly, miR-9 has been found to indirectly modulate the phosphorylation status of STAT3, an important intracellular signaling molecule mediating the inhibition of neuronal terminal differentiation [36,37]. Among the first miRNAs to be identified as major regulators of differentiation and lineage determination were those regulating the formation of skeletal muscles [38,39,40]. Among these are miR-1 and miR-133, which were defined as MyomiRs since they are both uniquely expressed in skeletal and cardiac muscle cells, under the control of myogenic transcriptional factors, and cooperate to finely regulate their proliferation and differentiation from myoblast precursors [41,42,43].
piRNAs form a novel class of small silencing RNAs (26–31 nts) with distinct features from known miRNAs and siRNAs. They were first reported in 2001 in Drosophila melanogaster testes and subsequently in mammals’ germline and be involved in germline stem cell maintenance and self-renewal [44,45,46,47]. However, emerging evidence has demonstrated that they are also expressed in a tissue-specific manner in multiple human somatic tissues other than the germline [48,49]. A large fraction of piRNA precursors is produced from discrete genetic loci named piRNA clusters, which can be transcribed in one direction or convergently from two ends (unidirectional or bidirectional clusters). Instead, a smaller fraction of piRNA precursors are generated from the 3′ UTR of protein-coding genes or individual transposons [50,51,52]. piRNA precursors are then processed by enzymatic machinery different from that involved in the processing of miRNA and siRNA through two different mechanisms: the primary synthesis mechanism and the ‘ping-pong’ amplification mechanism [50,53]. Once formed, mature piRNAs can bind the piwi proteins of the Argonaute family to form a piRNA/piwi complex, which is involved in germ stem cell maintenance and self-renewal by mediating chromatin rearrangement, silencing of transposon transcription, and suppression of translation [53,54,55,56,57].
lncRNAs are a class of regulatory ncRNAs that has attracted particular interest recently, first discovered in the early 1990s [58,59,60]. They can be classified according to various criteria, like their genomic origins (sense, antisense, bidirectional, intronic, and intergenic lncRNAs), their function, their subcellular localization, and so on [61]. lncRNAs are generated through pathways like that of protein-coding genes and undergo similar post-transcriptional processing (possess a 5′ capping and a 3′ poly-A tail and may be alternatively spliced) [62]. In humans, recent annotations have reported between 30,000 and 60,000 lncRNAs [63,64,65], involved in a broad spectrum of biological processes, including cell proliferation, apoptosis, and cell differentiation [65,66,67,68,69,70], through the regulation of gene expression at both a transcriptional and post-transcriptional level. In addition, many lncRNAs can induce epigenetic modifications by recruiting chromatin-remodeling complexes, such as the polycomb repressive complex 2 or other chromatin-modifying proteins [71,72,73]. Some lncRNAs, such as Evf2, act as co-factors, regulating the activity of transcriptional factors and, thus, the expression of target genes [74]. At a post-transcriptional level, lncRNAs have been demonstrated to regulate target mRNA processing, such as capping, splicing, editing, transport, translation, degradation, and stability [75].

3. Cellular Response to Stress and Air Pollutants

Cells are continuously exposed to multiple intrinsic or extrinsic stimuli that evoke a specific biological response known as stress. Stress is an environmental stimulus that differs from optimal conditions and influences cell growth and development. Cells respond to stress with adaptive responses to maintain homeostasis [76]. Several molecular mediators participate in the stress response, such as molecular chaperones, whose activity can be regulated by ncRNAs. Chaperones are ubiquitous proteins highly conserved throughout evolution, from bacteria to humans [77]. Many of them are also known as Heat Shock Proteins (Hsps) and can be classified according to their molecular weight: small heat shock proteins (sHsps) family from 10 to 30 kDa; Hsp40 family (40 kDa); Hsp60 (or chaperonins) with a molecular weight close to 60 kDa; Hsp70 family (70 kDa); Hsp90 family (83–90 kDa); and Hsp100/110 family with a molecular weight equal to or higher than 100 kDa [78]. They are essential for controlling protein homeostasis and maintaining the correct and functional conformation of proteins and nascent polypeptides, preventing their aggregation and misfolding [79]. Other functions of Hsps are regulating gene expression and DNA replication, cell differentiation, signal transduction, inflammatory response, carcinogenesis, senescence, and cell apoptosis [80,81,82]. The main factor involved in the heat shock response is heat shock factor 1 (HSF1), which regulates the expression of Hsps [83]. Under stress conditions, besides changing gene expression levels, the HSF1 induces several pre-translational, co-translational, and post-translational processes, likely through the involvement of some ncRNAs [84,85]. Therefore, ncRNAs participate in stress response [86,87]. Moreover, exposure to various stressors may alter their expression, with critical physio-pathological implications [88,89].
Air pollution is a main external stimulus that evokes a stress response. Oxidative stress is at the root of the toxic effect of air pollutants; it triggers several signaling pathways, such as the inflammatory response and the production of proinflammatory cytokines [90]. In addition, oxidative stress leads to the generation of reactive oxygen species (ROS), alteration of mitochondrial function and oxidative DNA damage [91,92]. The damage caused by air pollution affects almost every organ in the body, causing or contributing to many diseases [93].
Environmental pollution has several sources, both anthropogenic and natural, such as the use of fossil fuels in electricity generation, transport, industry, and housing; industrial processes and solvent use; agriculture; waste treatment; volcanic eruptions, airborne dust, and emissions of volatile organic compounds (VOCs). Air pollution can be distinguished into outdoor and indoor pollution. The latter is especially important because people in developed countries now spend most of their time (>80%) in enclosed spaces (e.g., home, school, office, public buildings or means of transport). Common indoor pollutants include environmental tobacco smoke, particulate matter (PM), nitrogen dioxide, carbon monoxide, VOCs, and biological allergens. Indoor air pollution can increase the risk of irritative phenomena, allergic sensitization, and acute and chronic respiratory disorders [94].
Air pollutants modify the normal chemical composition of the air and can be divided into primary and secondary pollutants. The former is emitted into the air as they are, such as carbon monoxide (CO), nitrogen oxide (NOx), sulfur dioxide (SO2), VOCs and particulate matter (PM). PM, a mixture of solid particles and liquid droplets, can, in turn, be divided into fractions based on the particle size, known as the equivalent aerodynamic diameter (AED), e.g., PM 10, PM 2.5 and Ultra-Fine Particulate Matter (UFPM). Particles larger than 10 μm are unlikely to enter the lower airways as most of them will be filtered by the nose and upper airways, whereas PM 10, PM 2.5 and UFPM usually reach the lower airways [95]. SO2 is produced by volcanoes and various industrial processes. Coal and oil usually contain sulfur compounds, so their combustion generates sulfur dioxide and, thus, acid precipitation. This is one of the reasons for worrying about the environmental impact of using these fuels as energy sources. CO is a gas formed from the incomplete combustion of fuels such as propane, natural gas, petrol, oil, coal or wood. Sources of carbon monoxide are devices such as furnaces, gas water heaters, wood stoves and other fuel-burning appliances that do not work efficiently, clogged chimneys, car exhaust fumes and tobacco smoke, and various industrial processes. In addition, CO is released from erupting volcanoes, forest fires, swamp gas and seaweed [96,97]. Natural sources of NOx include the activity of nitrogen-consuming microorganisms in the soil and the increased worldwide use of fertilizers in recent decades. The anthropogenic sources of NOx are motor vehicles (49%), electric utilities (27%), other industrial, commercial and residential sources (19%) and all other activities (5%) that burn fuel [98]. Natural VOC emissions arise from vegetation and the degradation of organic material; anthropogenic emissions, on the other hand, are mainly from the incomplete combustion of hydrocarbons and the evaporation of solvents and fuels [99].
Secondary pollutants are subsequently formed in the atmosphere because of chemical reactions involving the primary pollutants and sometimes the natural components of the atmosphere. Among the processes involved in the formation of secondary pollutants, there is photochemical smog, which consists of a series of reactions between nitrogen oxides and hydrocarbons in the presence of UV light. This series of reactions results in the oxidation of nitrogen monoxide (NO) to nitrogen dioxide (NO2), the production of ozone (O3) and the oxidation of hydrocarbons, with the formation of peroxyacetyl nitrate (PAN), formaldehyde, nitric acid, nitrates, and nitro-derivatives in the particle phase [100]. Other vital mechanisms leading to secondary pollutants are oxidation reactions and secondary organic aerosols (SOA) formation. The former occurs when primary pollutants react with oxygen in the atmosphere. For example, sulfur dioxide can be oxidized to form sulfuric acid, which can contribute to the formation of acid rain. Secondary organic aerosol formation occurs through the condensation of gases in the atmosphere [101,102]. When primary pollutants, such as VOCs, react with other atmospheric chemicals, they can form small particles known as aerosols. These aerosols can then condense to form larger particles, contributing to haze formation and other forms of air pollution.
Climate change could alter the dispersion of primary pollutants, especially PM, and intensify the formation of secondary pollutants, such as ozone. In addition, meteorological variables like temperature, humidity, and wind characteristics influence the emission, transport, dispersion, chemical transformation, and deposition of environmental pollutants [103].

4. ncRNAs as Part of EVs Cargo

RNA is part of the EVs’ cargo and is at the center of many functions attributed to EVs since it can alter gene expression and function in recipient cells. EVs are a heterogeneous group of vesicles released from cells under physiological and pathological conditions [104,105] that contain a cargo of bioactive molecules reflecting their parental cells and can modulate recipient cells’ behavior [106,107]. EVs are now considered an additional mechanism for intercellular communication, enabling cells to exchange information through proteins, lipids, and genetic material [108]. EVs are released from cells into the extracellular space and can be found in all biological fluids: blood [109], urine [110], saliva [111], cerebrospinal fluid [112], breast milk [113], bronchoalveolar lavage fluid (BALF) [114] etc. Furthermore, EV’s biogenesis has been a significant evolutionary advancement because the cargo encapsulated inside the vesicular structures is protected from the degradation of ribonucleases, deoxyribonucleases, and proteases that are abundant in the extracellular space. These enzymes cannot traverse the EVs lipid bilayer [115]. The uptake of EVs by recipient cells can be either selective (via ligand-receptor interaction) or non-selective [116]. EVs are generally classified according to certain intrinsic properties, including density, size, and biogenesis processes. They are generally divided into three subtypes according to their biogenesis mechanism: exosomes, microvesicles and apoptotic bodies [117]. The International Society for Extracellular Vesicles (ISEV) currently encourages the use of the term “extracellular vesicles” as a generic term for all secreted vesicles, considering the lack of consensus for the identification of specific markers to distinguish between the different subtypes of EVs [118]. Based on their size, we can distinguish: ‘small EVs’ (sEVs) and ‘medium/large EVs’ (m/lEVs), which have defined ranges of <100 nm or <200 nm for small EVs and >200 nm for large and/or medium EVs, respectively [118].
EV-associated RNAs (EV-RNA) consist of RNA fragments of different sizes and include mRNA, pre-miRNA and mature miRNA precursors, snoRNA, rRNA, tRNA, lncRNA, piRNA, and mitochondrial RNA [119]. Initially, the focus on EVs cargo was on proteins, but the discovery 2007 of biologically active RNA particles in small EVs amplified the potential role of EVs in biology [120]. Currently, the Vesiclepedia database includes more than 27,000 entries for mRNAs and more than 10,000 entries for non-coding RNAs [121]. The role of non-coding encapsulated RNAs in EVs is emerging, especially in liquid biopsy diagnostics, a promising non-invasive alternative to traditional methods of diagnosis and prognosis [122]. The liquid biopsy method is based on identifying clinically relevant biomarkers. It allows for early diagnosis and monitoring of diseases from their earliest stages, which would bring enormous benefits to human health.
It has been shown that the ncRNA content inside EVs is higher than in the cells from which they originate, in contrast to the protein and lipid content that reflect the parent cells [123,124]. This has led researchers to hypothesize the existence of highly selective RNA-loading mechanisms inside EVs. The interactions with RNA-binding proteins (RBPs), such as argonaute 2 (AGO2), ALIX and annexin A2, appear to be crucial to explain the highly controlled and specific process of selective ncRNA packaging in EVs [125,126,127,128,129]. It must be noted that RBPs constitute about 25% of the protein content of EVs [130]. Furthermore, it was shown that post-transcriptional modifications at the 3′ end of miRNAs could also influence their selective loading into EVs [129]. RNA-EVs are divided into three types: RNAs with known functions, such as intact mRNA and miRNA; RNAs with probable but unproven intercellular mediator functions, such as piRNA; and, thirdly, RNA fragments (e.g., tRNA and mRNA and rRNA fragments) with unknown functions, which may be non-functional degradation products [119]. EVs could also play a potential role in drug therapy due to their cargo-protecting characteristic, as they could be used to deliver drugs in a targeted manner to cells (e.g., cancer cells). This opens the door to potential RNA-based therapies, including siRNAs, miRNAs, antisense oligonucleotides, mRNAs, guide RNAs and self-amplifying RNAs. The advantages of the therapeutic use of EVs include their high biocompatibility and stability and their limited immunogenicity [130].

5. The Effects of Pollutants on the Production, Release, and Cargo of Circulating EVs

Environmental pollutants contribute to the pathogenesis of numerous human diseases, with chronic inflammation and oxidative stress as common hallmarks. Emerging evidence suggests that EVs may play a role in the relationship between environmental pollutants and the pathogenesis of chronic systemic diseases [131]. Physiologically, EVs from the pulmonary region exhibit protective effects against stress signals since they participate in maintaining pulmonary homeostasis. However, air pollution alters the composition of pulmonary EVs, leading to a dysregulation of the EVs cargo and increasing their production and release [132]. One of the functions of the respiratory epithelium is to defend the body against pathogens or environmental pollutants. In addition, it is involved in the regulation of immune responses, as well as in tissue repair and remodeling after injury [133]. However, repeated exposure of the respiratory epithelium to air pollution induces chronic airway inflammation that can lead to the release of inflammatory molecular signals in the circulatory system [134]. For instance, exposure to PM has been shown to lead to an increased release of EVs, especially in overweight subjects [135].
The role of alveolar macrophages and the respiratory epithelium is to rapidly eliminate inhaled harmful substances and thus maintain an anti-inflammatory state. EVs from alveolar macrophages contribute to this anti-inflammatory state by providing a suppressor of cytokine signaling (SOCS) 1 and 3 to epithelial cells. Studies have been conducted on mice exposed to cigarette smoke extract (CSE) and human smokers. Both showed reduced SOCS concentrations in BALF compared to non-smoking controls, confirming a loss in smokers of the anti-inflammatory status derived from EVs [136]. Exposure to CSE, PM and other air pollutants has been shown to alter transcriptomic (miRNAs) composition and increase the release of circulating EVs derived from endothelial cells and immune cells, as well as pro-inflammatory molecules [137,138,139]. Bronchial epithelial cells and lung fibroblasts have been shown to release CD63 + CD81 + and TF + EVs in response to respiratory toxicants [140]. Thus, exposure to environmental pollutants appears to shift the functionality of EVs secreted by mononuclear, epithelial, and endothelial cells from an anti-inflammatory to a pro-inflammatory phenotype. Exposure to environmental agents may ultimately trigger epigenetic changes because abnormal ncRNA expression within EVs may induce developmental changes or lead to disease progression.

6. ncRNAs and Links to Respiratory Diseases

Pollution is known to cause and aggravate several chronic respiratory diseases. The World Health Organization has ranked air pollution as the most common environmental cause of premature death [141]. Several studies have suggested that ncRNAs inside EVs are involved in respiratory diseases caused by air pollution. For example, one study found that exposure to PM increased the expression of miR-222-3p in EVs isolated from bronchial epithelial cells [142]. This miRNA was shown to target genes involved in oxidative stress and inflammation, suggesting that it may contribute to the development of airway inflammation and damage caused by PM exposure. In another study, miR-146a and miR-146b were significantly elevated in bronchoalveolar lavage fluid (BALF) and lung tissue homogenate from mice exposed to PM 2.5 [143]. Furthermore, it was seen that in bronchial epithelial cells treated with PM 2.5, there was the secretion of EVs that increased bronchial smooth muscle cell contractility and contributed to airway hyperresponsiveness [144].
Other studies have also reported that EVs, carrying ncRNAs in their cargo, are functionally involved in cancer initiation, progression and the development of metastases [145]. Exposure to PM 2.5 induced differential expression of vesicular miRNAs, leading to tumour development [146]. ncRNAs as cargo of EVs can be used as biomarkers of lung cancer (LC) because it was discovered that certain miRNAs are dysregulated in LC patients, including miR-21, miR-23b-3p, miR-10b-5p, miR-139-5p, miR-200b-5p, miR-378a, miR-379 and miR-4257 [147,148].
lncRNAs regulate certain biochemical and cellular processes, such as gene expression, RNA splicing and ligand-receptor engagement, which in turn mediate the pathogenesis of respiratory disorders [149]. They have also emerged as novel master regulators of initiation, progression, and response to therapy in various cancers. lncRNA HOX transcribed antisense RNA (HOTAIR) represses gene expression, and its high expression in LC correlates with metastasis and poor prognosis [150]. MALAT1 is one of the most highly valued lncRNAs in lung cancer. It modulates miR-124/STAT3 to promote carcinogenesis [151] and the miR-204/SLUG axis to enhance epithelial-mesenchymal transition (EMT) and lymph node metastasis [152]. Another widely evaluated oncogenic lncRNA in lung cancer is XIST because its downregulation has been shown to inhibit tumor growth via the upregulation of E-cadherin and downregulation of Bcl-2 [153].
In COPD, there is severe airway inflammation with consequent damage to the lung parenchyma. This leads to the destruction of the alveolar wall and rarefaction of the alveolar sacs, resulting in breathing difficulties and reduced lung function (irreversible airway obstruction) [154]. Air pollution is a significant risk factor for COPD, and exposure to particulate matter (PM) has been shown to induce inflammation and oxidative stress in the lungs, which can contribute to the development and progression of COPD. Prevention is the key to minimizing risk factors, such as air pollution and cigarette smoke. It has been shown that miR-210 and miR-218 expression is significantly higher in EVs released from primary human bronchial epithelial cells (HBEC) after exposure to cigarette smoke. This promotes airway fibrosis in the pathogenesis of COPD [155]. It was subsequently observed that following exposure to cigarette smoke, EVs showed a reduced expression of specific miRNAs, including let-7d, miR-191, miR-126 and miR125a, promoting apoptotic cell clearance [156]. Most studies report a global downregulation of miRNAs in response to cigarette smoke, especially from human alveolar macrophages, probably by modification of DICER, an RNA endonuclease involved in miRNA maturation [157,158]. The expression of some miRNAs, such as miR-638 and miR-101, appear to correlate with the severity of regional emphysema [159,160]. Among lncRNAs, increased expression of SCAL1 was seen in airway epithelial cells as part of the oxidative stress response to cigarette smoke exposure [161].
Asthma is a chronic inflammatory disease characterized by airway remodeling and bronchial hyperresponsiveness caused by both genetic factors and environmental stress factors. Air pollution is a major environmental factor that can exacerbate asthma symptoms. Exposure to air pollutants, such as particulate matter, ozone, nitrogen dioxide, and sulfur dioxide, can trigger inflammation in the airways, leading to asthma attacks. EVs derived from bronchoalveolar lavage fluid of asthmatic patients contained higher levels of certain miRNAs than healthy controls, suggesting that these EVs may play a role in the development of asthma [162,163]. ncRNAs play a key role in the onset and progression of asthma by regulating gene transcription [164]. miRNAs influence asthma pathogenesis by regulating immune cells, bronchial epithelial cells and airway smooth muscle cells (ASMCs). It has been shown that miR-1, miR-18a, miR-21, miR-146a, miR-155, miR-210 and miR-1248 can regulate T-cell function and the production of Th2 cytokines involved in the pathogenesis of asthma [165,166,167]. A significantly reduced expression of let-7a, miR-21, miR-133a, miR-155, miR-328 and miR-1248 was found in exhaled breath condensates from asthmatic subjects compared to healthy subjects [163]. Among lncRNAs, it was shown that lncRNA-MEG3 could competitively adsorb miR-17 in CD4+ T cells of asthmatic patients [168]. lncRNA MALAT1, on the other hand, adsorbs miR-155 by negatively regulating it and altering the Th1/Th2 balance among CD4+ T cells [169]. Another lncRNA under investigation is lncRNA PVT1, which, if over-regulated, could inhibit miR-149 expression in bronchial epithelial cells, promoting airway inflammation and cell barrier destruction, thus accelerating the development of asthma [170] (Table 2).

7. ncRNAs as Mediators of the Crosstalk between Air Pollution and Cardiovascular Diseases

Cardiovascular diseases (CVDs) are a group of disorders affecting the heart and the blood vessels, such as heart failure, coronary heart disease and acute myocardial infarction (AMI) and represent one of the leading causes of morbidity and mortality in the world [171].
Besides genetic causes, increasing compelling evidence has demonstrated that exposure to environmental air pollution, especially particulate matter (PM), significantly contributes to the development of CVDs and acute cardiac events like stroke, likely by triggering pulmonary inflammation, systemic inflammation, autonomic nervous system imbalance, oxidative stress, endothelial dysfunction and prothrombotic changes [172,173,174,175,176]. Accordingly, it has been shown that short-term exposures to PM and nitrogen oxides are consistently associated with increased risks of hypertension and triggering of myocardial infarction (MI) and stroke. In contrast, long-term exposures are largely associated with increased risk of atherosclerosis, incident MI, hypertension, and incident stroke and stroke mortality [177].
In recent years, researchers have started to investigate the underlying epigenetic component of CVDs. NcRNA-mediated gene regulation has been proposed to play a key role in the possible correlation between air pollutant exposure and the risk of CVDs [178,179,180]. It has been well established that short and long ncRNAs are involved in cardiac development and physiological functions. Moreover, they significantly contribute to the development and progression of various CVDs [181,182,183].
A cohort study by Bollati et al. analyzed male workers’ miRNA profile in peripheral blood leukocytes after a three-day exposure to ambient PM rich in metals. The examined subjects were not affected by CVDs, or other conditions in which systemic inflammation occurs, such as cancer and respiratory diseases. A qRT-PCR analysis revealed a significant increase in miR-21 and miR-222. Moreover, a positive correlation was found between the levels of miR-21 and 8-hydroxy-guanine (8-OH-dG), suggesting a relationship with oxidative stress [184]. Similarly, mir-222 has been found in higher concentrations in the extracellular fraction of saliva in schoolchildren following recent exposure to ultrafine particles (UFP) [185]. Both miR-21 and miR-222 are highly expressed in the cardiovascular system, regulating critical biological pathways. For instance, miR-21 has been reported to exert a cardioprotective role [186].
On the contrary, they are dysregulated in numerous CVDs, suggesting they could be used as promising cardiovascular biomarkers and novel therapeutic targets/agents in CVDs, including air pollution-correlated ones [187,188]. However, in a cohort of 153 elderly males, an inverse relationship was found between eight leukocyte miRNAs, including miR-21 and miR-222, and PM 2.5 exposure. According to the authors, considering the activity of the predicted target mRNAs, this negative association should result in increased inflammation, endothelial dysfunction, and atherosclerosis [189]. The contradictory results could be due to differences in the methodologies used by the two research teams, the particle composition, the time of exposure and/or the characteristics of the participants.
Chen and colleagues observed that in healthy young subjects, short-term exposure to PM was positively associated with the expression (mRNAs, proteins, or both) of cytokines involved in inflammation, coagulation, and endothelial dysfunction, including IL1, IL6, TNF, toll-like receptor 2, coagulation factor 3, and endothelin 1. On the contrary, it was negatively associated with the expression of miRNAs predicted to target these cytokines’ mRNAs, such as miR-21-5p, miR-187-3p, miR-146a-5p, miR-1-3p, and miR-199a-5p. Thus, the authors suggested that a possible correlation between PM exposure and CVD development might be mediated by miRNAs controlling specific cytokine expression [190].
Another interesting line of research regarding the correlation between exposure to environmental contaminants and increased CVD risk focused on investigating EVs’ miRNA content. Numerous studies have shown that exposure to ambient air pollutants, including fine particulate matter (PM 2.5), coarse particulate matter (PM 10), ozone (O3), and nitrogen oxides (NOx), was significantly associated with changes in the expression of some circulating miRNAs, including miR-26a-5p, miR-146a-5p, miR-150–5p and miR-21-5p, some of which may mediate their effects on the downstream inflammation, coagulation, and blood lipid biomarkers [134,139,190,191,192,193,194,195,196].
In this context, Bollati and coworkers investigated whether PM and metal-rich PM may alter microvesicles (MVs) content, finding that miR-128 and miR-302 were significantly overexpressed after three days of PM exposure, compared with the beginning of the working week [134]. Both miRNAs regulate gene expression linked with CVDs, including coronary artery disease, cardiac hypertrophy, and heart failure pathways, suggesting MV-associated miRNAs as mediators of air pollution toxicity [134]. Furthermore, the effects of metal-rich PM have been evaluated in a subset of the same study population, identifying four PM-sensitive miRNAs (miR-29a-3p, miR-146a-5p, miR-421, and let-7 g-5p) that were differentially expressed compared to control samples and seem to favor a pro-inflammatory response, potentially contributing to several diseases, including CVDs and respiratory system disorders [139,191]. Similarly, Rodosthenous and colleagues investigated the relationship between short-, intermediate-, and long-term exposures to PM and the EVs-miRNome in a cohort of healthy adults. They observed long-term exposure to PM 2.5 increased some EVs’ miRNAs in serum. The predicted target genes, including interleukin 6 (IL-6), C-X-C motif chemokine ligand 12 (CXCL12), vascular cell adhesion molecule 1 (VCAM-1), a cluster of differentiation 40 (CD40), and platelet-derived growth factor subunit beta (PDGFB), are linked to CVD-related signaling pathways, such as oxidative stress, inflammation, atherosclerosis, and cardiac hypertrophy [193]. These results were confirmed by a subsequent study by the same authors, where it was demonstrated that an increase of multiple EVs’ mRNAs in serum from elderly males could modify the association between PM 2.5 and systolic blood pressure by targeting proteins involved in essential cardiovascular functions [194]. These results aligned with previous data from Motta et al., which demonstrated that miRNAs are responsible for the association between exposure to particulate air pollution and increased blood pressure, a well-established risk factor for cardiovascular disease [197].
Pergoli and colleagues observed that short-term exposure to PM 10 was associated with an increased release of EVs, especially from monocytes/macrophages (CD14+) and platelets (CD61+), as well as high fibrinogen levels in overweight/obese people [192]. The analysis of these EVs’ content revealed that nine miRNAs were downregulated in response to PM exposure. As highlighted by integrated network analysis, five of them, i.e., let-7c, miR-106a, miR-185, miR-331, and miR-652, modulate cardiovascular-linked processes, including fibrinogen levels, suggesting an association between extracellular miRNA released after short-term exposure to PM and increased coagulation [192]. Therefore, air pollutant exposure may affect the EVs’ content and downstream effects on target cells. Most recently, a change of small EV (sEVs) subpopulations released from the respiratory system has also been observed in response to PM 2.5 exposure, increasing CD63/CD81/CD9-positive particles [196]. In particular, the authors observed that these sEVs contained miR-421, which contributed to cardiac dysfunction by targeting the cardiac angiotensin-converting enzyme 2 (ACE2), suggesting the sEVs’ miRNome as responsible for the crosstalk between lung and heart upon PM 2.5 exposure [196] (Table 2).

8. ncRNAs and Neurodegenerative Diseases

Neurodegenerative diseases (NDDs), including Parkinson’s disease (PD), Alzheimer’s disease (AD), and other dementia disorders, affect millions of people worldwide and are caused by the progressive degeneration and death of select populations of neurons. This process leads to debilitating conditions such as cognitive, motor, and behavioral impairments [198]. Recent studies indicate that air pollution may also adversely affect the brain, contributing to the etiopathogenesis of neurodegenerative diseases. There is growing evidence that traffic-related air pollution (TRAP) exposure might lead to the development and progression of neurodegenerative disorders [199]. A study conducted in the U.S. reported that the risk of developing dementia for residents in areas with high PM 2.5 was increased by 92% compared to those who lived in areas with lower levels of particulate matter [200]. In addition, another study conducted on mice demonstrated the induced glutaminase-containing EVs release from microglial cells. The increment of glutamate is associated with neurodegeneration [201]. Several studies suggest that exposure to air pollution might be associated with PD, a long-term degenerative disorder of the central nervous system that mainly affects the motor system. Different expression levels of several miRNAs can be used as potential diagnostic biomarkers for PD. Particularly, EV-derived miRNAs are increasingly used to diagnose the disease [202]. A study on blood-derived EVs in PD reported higher miR-34a-5p levels in PD patients compared to controls, whereas other similar studies on serum EV-derived miRNAs found upregulated miRNAs, such as miR-22, miR-23a, miR-24, andmiR-222, and a downregulated miRNA, miR-505, in PD patients [203,204]. Also, EVs isolated from the cerebrospinal fluid (CSF) are a promising source for PD diagnostic markers. The qRT-PCR analysis showed differences in the miR-181a-5p, miR-181b-5p, miR-9-5p and let-7b in PD patients versus control comparisons [203].
Various studies have demonstrated a strong association between air pollution and AD. In this neurodegenerative disease, the amyloid-beta protein (Aβ) dysregulation leads to aggregates of amyloid fibrils and neurotoxicity. A German study showed that exposure to PM 2.5 and NOx was associated with an increased risk for AD [205]. Moreover, a study conducted in London concluded that residents with the most exposure to nitrogen dioxide and PM 2.5 were the most likely to be diagnosed with dementia, with associations more consistent for AD [206]. Compared to healthy controls, a study on plasma-derived EVs of AD patients reported downregulation of miR-342-3p, miR-451 and miR-21-5p. On the contrary, other studies revealed several EV-derived miRNAs, such as miR-29a, -451a, -125b, -582-5p, -106a-5p, and -106b-5p, were upregulated in AD patients, compared to healthy controls [207,208,209].
Table 2. ncRNAs role in environmental stress response and their involvement in respiratory, cardiovascular, and nervous system pathogenesis. The table also summarizes their possible contribution as biomarkers.
Table 2. ncRNAs role in environmental stress response and their involvement in respiratory, cardiovascular, and nervous system pathogenesis. The table also summarizes their possible contribution as biomarkers.
DiseaseType of DiseaseEnvironmental StressorsncRNADescriptionRefs.
Respiratory diseases Exposure to PMIncreased expression of miR-222-3p in EVs isolated from bronchial epithelial cellsmiRNA involved in oxidative stress and inflammation; contribution to the development of airway inflammation[142]
Exposure to PM 2.5miR-146a and miR-146bSignificantly elevated BALF and lung tissue homogenate from mice [143]
Bronchial epithelial cells treated with PM 2.5EVs carry ncRNAs in their cargoSecretion of EVs that increased bronchial smooth muscle cell contractility and contributed to airway hyperresponsiveness[144]
Lung cancer (LC)Exposure to PM 2.5miR-21, miR-23b-3p, miR-10b-5p, miR-139-5p, miR-200b-5p, miR-378a, miR-379 and miR-4257Dysregulated ncRNAs as EVs cargo can be used as biomarkers of LC[147,148]
Lung cancer (LC) lncRNA HOTAIR HOTAIR high expression in LC correlates with metastasis and poor prognosis[150]
COPDExposure to cigarette smokemiR-210 and miR-218Their expression is highest in EVs released from HBECs. This promotes airway fibrosis in the pathogenesis of COPD[155]
Exposure to cigarette smokelet-7d, miR-191, miR-126 and miR125aEVs showed a reduced expression of these miRNAs, promoting apoptotic cell clearance[156]
Exposure to cigarette smokelncRNA SCAL1 Increased expression of SCAL1 in airway epithelial cells as part of the oxidative stress response[161]
AsthmaAir pollution can exacerbate asthma symptomsmiR-1, miR-18a, miR-21, miR-146a, miR-155, miR-210 and miR-1248They can regulate T-cell function and the production of Th2 cytokines involved in the pathogenesis of asthma[164,165,166]
AsthmaAir pollution can exacerbate asthma symptomslet-7a, miR-21, miR-133a, miR-155, miR-328 and miR-1248Reduced expression in exhaled breath condensates from asthmatic subjects[167]
AsthmaAir pollution can exacerbate asthma symptomslncRNA-MEG3
MALAT1
PVT1
Altered expression of these lncRNAs compared to controls[168,169,170]
Cardiovascular diseases Three-day exposure to ambient PM rich in metals miRNA profile of peripheral blood leukocytesIncreased levels of miR-21 and miR-222 and positive correlation with 8-OH-dG level and oxidative stress[182]
Exposure to ultrafine particlesmiRNAs from the extracellular fraction of saliva in schoolchildrenIncreased concentration of miR-222 and possible positive correlation with CVDs[185]
Exposure to PM 2.5 Eight leukocyte miRNAsReduced levels of miRNAs, including miR-21 and miR-222. Increase in inflammation, endothelial dysfunction, and atherosclerosis processes[189]
Short-term exposure to PMmiR-21-5p, miR-187-3p, miR-146a-5p, miR-1-3p, and miR-199a-5pReduced levels of miRNAs and increase of the targeted cytokines, which are involved in inflammation, coagulation, and endothelial dysfunction[189]
Coronary artery disease, cardiac hypertrophy, and heart failureThree-day exposure to PMmiR-128 and miR-302 from circulating microvesiclesmiRNAs overexpression and possible positive correlation with CVD development[190]
Exposure to metal-rich PMmiR-29a-3p, miR-146a-5p, miR-421, and let-7 g-5pAltered expression of miRNAs and positive correlation with a pro-inflammatory response contributing to CVD development[191]
Short-term exposure to PM 10miRNAs from monocytes/macrophages (CD14+) and platelets (CD61+)-derived EVs Downregulation of miRNAs expression and increase of coagulation[192]
Long-term exposure to PM 2.5miRNAs from serum EVs Increase of EVs’ miRNAs, whose targets are involved in some CVD-related signaling pathways, such as oxidative stress, inflammation, atherosclerosis, and cardiac hypertrophy[193]
Long-term exposure to PM 2.5miRNAs from serum EVs Increase of EVs’ miRNAs, whose targets are involved in essential cardiovascular functions[194]
Exposure to PM 2.5miR-421 from small EVs released from the respiratory systemPositive correlation with cardiac dysfunction[196]
Neurodegenerative diseasesPDExposure to air pollutionmiR-34a-5p, miR-181a-5p, miR-181b-5p, miR-9-5p and let-7bAltered expression of these miRNAs compared to controls[203]
PDExposure to air pollutionmiR-22, miR-23a, miR-24, miR-222, mir-505Altered expression of these EVs’ miRNAs[204]
ADExposure to air pollutionmiR-342-3p, miR-451 and miR-21-5pDownregulation of EVs’ miRNAs expression[207]
ADExposure to air pollutionmiR-29a, -451a, -125b, -582-5p, -106a-5p, and -106b-5pIncreased expression of EVs’ miRNAs[208,209]
VDExposure to air pollutionmiR-135a, miR-193b, -23a, -29a, -130bAltered expression of these EVs’ miRNAs[207]
FTDExposure to air pollutionmiR-204-5p and mirR-632Downregulation of miRNAs expression[207]
Furthermore, exposure to air pollution is also associated with other types of dementia disorders, including vascular dementia (VD) and frontotemporal dementia (FTD) [210,211]. VD is dementia caused by reduced blood flow in various brain regions. In contrast, FTD is a rare type of dementia characterized by progressive nerve cell loss in the frontal and temporal lobes of the brain. Recent studies have shown that EV-derived miRNAs are involved in the pathogenesis of dementia disorders, making them valuable biomarkers for diagnosing dementia. Serum levels of EV-derived miR-135a increased, whereas levels of EV-derived miR-193b, -23a, -29a, -130b decreased in VD patients versus healthy controls. EVs isolated from FTD patients’ cerebrospinal fluid (CSF) showed downregulated expression of miR-204-5p and mirR-632 compared to healthy controls [207] (Table 2).

9. Conclusions and Future Perspectives

Poor air quality has become a significant threat to public health globally, and it is estimated to cause approximately 7 million deaths each year, as it increases the risk of a wide range of diseases. Considering its health impacts, conducting biological impact assessments of air pollution has thus become an urgent task for public health practitioners [212].
Growing evidence highlights the clear connection between environmental pollutants and human pathologies. Nevertheless, the mechanisms linking air pollutant exposure with mediating biological effects in cells and disease onset have not yet been fully elucidated.
In response to environmental stresses, cells produce multiple mediators secreted into the extracellular environment via EVs [213,214,215]. Among these mediators, ncRNAs can determine multiorgan and systemic effects due to their ability to affect numerous genetic targets and manipulate the gene expression of target cells. Moreover, circulating EVs can be detected in the plasma of subjects exposed to particulate air pollutants, to identify a possible predictive role of peripheral blood ncRNA signatures in human diseases. Evaluating different EV-carried ncRNA signatures could represent a good opportunity to find new biomarkers for the diagnostics and subsequent stratification, prognostics and evaluating treatment efficacy in patients with air pollutant exposure-associated diseases. In addition, ncRNAs could be used to optimize EV-based therapeutic approaches, such as personalized medicine (Figure 1).

Author Contributions

Conceptualization, F.C. and C.C.B.; writing—original draft preparation, G.D., R.S., A.M.V. and F.S.; writing—review and editing, A.M.G., C.C., F.B., F.C. and C.C.B.; supervision, C.C.B. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Data Availability Statement

No new data were created.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Crick, F.H. On Protein Synthesis. Symp. Soc. Exp. Biol. 1958, 12, 138–163. [Google Scholar] [PubMed]
  2. Crick, F. Central Dogma of Molecular Biology. Nature 1970, 227, 5258. [Google Scholar] [CrossRef] [PubMed]
  3. Jacob, F.; Monod, J. Genetic Regulatory Mechanisms in the Synthesis of Proteins. J. Mol. Biol. 1961, 3, 318–356. [Google Scholar] [CrossRef] [PubMed]
  4. Lander, E.S.; Linton, L.M.; Birren, B.; Nusbaum, C.; Zody, M.C.; Baldwin, J.; Devon, K.; Dewar, K.; Doyle, M.; Fitzhugh, W.; et al. Initial Sequencing and Analysis of the Human Genome. Nature 2001, 409, 860–921. [Google Scholar] [CrossRef]
  5. Craig Venter, J.; Adams, M.D.; Myers, E.W.; Li, P.W.; Mural, R.J.; Sutton, G.G.; Smith, H.O.; Yandell, M.; Evans, C.A.; Holt, R.A.; et al. The Sequence of the Human Genome. Science 2001, 291, 1304–1351. [Google Scholar] [CrossRef]
  6. Birney, E.; Stamatoyannopoulos, J.A.; Dutta, A.; Guigó, R.; Gingeras, T.R.; Margulies, E.H.; Weng, Z.; Snyder, M.; Dermitzakis, E.T.; Thurman, R.E.; et al. Identification and Analysis of Functional Elements in 1% of the Human Genome by the ENCODE Pilot Project. Nature 2007, 447, 799–816. [Google Scholar] [CrossRef]
  7. Nurk, S.; Koren, S.; Rhie, A.; Rautiainen, M.; Bzikadze, A.V.; Mikheenko, A.; Vollger, M.R.; Altemose, N.; Uralsky, L.; Gershman, A.; et al. The Complete Sequence of a Human Genome. Science 2022, 376, 44–53. [Google Scholar] [CrossRef]
  8. Mattick, J.S. Challenging the Dogma: The Hidden Layer of Non-Protein-Coding RNAs in Complex Organisms. Bioessays 2003, 25, 930–939. [Google Scholar] [CrossRef]
  9. Pertea, M. The Human Transcriptome: An Unfinished Story. Genes 2012, 3, 344–360. [Google Scholar] [CrossRef]
  10. Richard Boland, C. Non-Coding RNA: It’s Not Junk. Dig. Dis. Sci. 2017, 62, 1107–1109. [Google Scholar] [CrossRef]
  11. Beermann, J.; Piccoli, M.T.; Viereck, J.; Thum, T. Non-Coding RNAs in Development and Disease: Background, Mechanisms, and Therapeutic Approaches. Physiol. Rev. 2016, 96, 1297–1325. [Google Scholar] [CrossRef] [PubMed]
  12. Zhang, P.; Wu, W.; Chen, Q.; Chen, M. Non-Coding RNAs and Their Integrated Networks. J. Integr. Bioinform. 2019, 16, 20190027. [Google Scholar] [CrossRef]
  13. Mizuno, T.; Chou, M.Y.; Inouye, M. A Unique Mechanism Regulating Gene Expression: Translational Inhibition by a Complementary RNA Transcript (MicRNA). Proc. Natl. Acad. Sci. USA 1984, 81, 1966–1970. [Google Scholar] [CrossRef]
  14. Andersen, J.; Delihas, N.; Ikenaka, K.; Green, P.J.; Pines, O.; Ilercil, O.; Inouye, M. The Isolation and Characterization of RNA Coded by the MicF Gene in Escherichia Coli. Nucleic Acids Res. 1987, 15, 2089. [Google Scholar] [CrossRef]
  15. Andersen, J.; Forst, S.A.; Zhao, K.; Inouye, M.; Delihas, N. The Function of MicF RNA. MicF RNA Is a Major Factor in the Thermal Regulation of OmpF Protein in Escherichia Coli. J. Biol. Chem. 1989, 264, 17961–17970. [Google Scholar] [CrossRef] [PubMed]
  16. Mattick, J.S. Non-Coding RNAs: The Architects of Eukaryotic Complexity. EMBO Rep. 2001, 2, 986–991. [Google Scholar] [CrossRef] [PubMed]
  17. Frith, M.C.; Pheasant, M.; Mattick, J.S. The Amazing Complexity of the Human Transcriptome. Eur. J. Hum. Genet. 2005, 13, 894–897. [Google Scholar] [CrossRef]
  18. Hamilton, A.J.; Baulcombe, D.C. A Species of Small Antisense RNA in Posttranscriptional Gene Silencing in Plants. Science 1999, 286, 950–952. [Google Scholar] [CrossRef]
  19. Waterhouse, P.M.; Graham, M.W.; Wang, M.B. Virus Resistance and Gene Silencing in Plants Can Be Induced by Simultaneous Expression of Sense and Antisense RNA. Proc. Natl. Acad. Sci. USA 1998, 95, 13959–13964. [Google Scholar] [CrossRef]
  20. MacRae, I.J.; Ma, E.; Zhou, M.; Robinson, C.V.; Doudna, J.A. In Vitro Reconstitution of the Human RISC-Loading Complex. Proc. Natl. Acad. Sci. USA 2008, 105, 512–517. [Google Scholar] [CrossRef]
  21. Bartel, D.P. MicroRNAs: Genomics, Biogenesis, Mechanism, and Function. Cell 2004, 116, 281–297. [Google Scholar] [CrossRef] [PubMed]
  22. Morlando, M.; Ballarino, M.; Gromak, N.; Pagano, F.; Bozzoni, I.; Proudfoot, N.J. Primary MicroRNA Transcripts Are Processed Co-Transcriptionally. Nat. Struct. Mol. Biol. 2008, 15, 902–909. [Google Scholar] [CrossRef] [PubMed]
  23. Park, J.E.; Heo, I.; Tian, Y.; Simanshu, D.K.; Chang, H.; Jee, D.; Patel, D.J.; Kim, V.N. Dicer Recognizes the 5′ End of RNA for Efficient and Accurate Processing. Nature 2011, 475, 201–205. [Google Scholar] [CrossRef]
  24. Gregory, R.I.; Chendrimada, T.P.; Cooch, N.; Shiekhattar, R. Human RISC Couples MicroRNA Biogenesis and Posttranscriptional Gene Silencing. Cell 2005, 123, 631–640. [Google Scholar] [CrossRef]
  25. Lytle, J.R.; Yario, T.A.; Steitz, J.A. Target MRNAs Are Repressed as Efficiently by MicroRNA-Binding Sites in the 5′ UTR as in the 3′ UTR. Proc. Natl. Acad. Sci. USA 2007, 104, 9667–9672. [Google Scholar] [CrossRef] [PubMed]
  26. Lewis, B.P.; Burge, C.B.; Bartel, D.P. Conserved Seed Pairing, Often Flanked by Adenosines, Indicates That Thousands of Human Genes Are MicroRNA Targets. Cell 2005, 120, 15–20. [Google Scholar] [CrossRef]
  27. Shi, Y.; Jin, Y.X. MicroRNA in Cell Differentiation and Development. Sci. China C Life Sci. 2009, 52, 205–211. [Google Scholar] [CrossRef]
  28. Sayed, D.; Abdellatif, M. MicroRNAs in Development and Disease. Physiol. Rev. 2011, 91, 827–887. [Google Scholar] [CrossRef]
  29. Alles, J.; Fehlmann, T.; Fischer, U.; Backes, C.; Galata, V.; Minet, M.; Hart, M.; Abu-Halima, M.; Grässer, F.A.; Lenhof, H.P.; et al. An Estimate of the Total Number of True Human MiRNAs. Nucleic Acids Res. 2019, 47, 3353–3364. [Google Scholar] [CrossRef]
  30. Kozomara, A.; Birgaoanu, M.; Griffiths-Jones, S. MiRBase: From MicroRNA Sequences to Function. Nucleic Acids Res. 2019, 47, D155–D162. [Google Scholar] [CrossRef]
  31. Lee, R.C.; Feinbaum, R.L.; Ambros, V. The C. Elegans Heterochronic Gene Lin-4 Encodes Small RNAs with Antisense Complementarity to Lin-14. Cell 1993, 75, 843–854. [Google Scholar] [CrossRef] [PubMed]
  32. Wightman, B.; Ha, I.; Ruvkun, G. Posttranscriptional Regulation of the Heterochronic Gene Lin-14 by Lin-4 Mediates Temporal Pattern Formation in C. Elegans. Cell 1993, 75, 855–862. [Google Scholar] [CrossRef] [PubMed]
  33. Reinhart, B.J.; Slack, F.J.; Basson, M.; Pasquienelll, A.E.; Bettlnger, J.C.; Rougvle, A.E.; Horvitz, H.R.; Ruvkun, G. The 21-Nucleotide Let-7 RNA Regulates Developmental Timing in Caenorhabditis Elegans. Nature 2000, 403, 901–906. [Google Scholar] [CrossRef] [PubMed]
  34. Wang, Y.; Medvid, R.; Melton, C.; Jaenisch, R.; Blelloch, R. DGCR8 Is Essential for MicroRNA Biogenesis and Silencing of Embryonic Stem Cell Self-Renewal. Nat. Genet. 2007, 39, 380–385. [Google Scholar] [CrossRef]
  35. Rosa, A.; Spagnoli, F.M.; Brivanlou, A.H. The MiR-430/427/302 Family Controls Mesendodermal Fate Specification via Species-Specific Target Selection. Dev. Cell 2009, 16, 517–527. [Google Scholar] [CrossRef] [PubMed]
  36. Krichevsky, A.M.; Sonntag, K.-C.; Isacson, O.; Kosik, K.S. Specific MicroRNAs Modulate Embryonic Stem Cell-Derived Neurogenesis. Stem Cells 2006, 24, 857–864. [Google Scholar] [CrossRef]
  37. Delaloy, C.; Liu, L.; Lee, J.A.; Su, H.; Shen, F.; Yang, G.Y.; Young, W.L.; Ivey, K.N.; Gao, F.B. MicroRNA-9 Coordinates Proliferation and Migration of Human Embryonic Stem Cell–Derived Neural Progenitors. Cell Stem Cell 2010, 6, 323. [Google Scholar] [CrossRef]
  38. Luo, W.; Nie, Q.; Zhang, X. MicroRNAs Involved in Skeletal Muscle Differentiation. J. Genet. Genom. 2013, 40, 107–116. [Google Scholar] [CrossRef]
  39. Horak, M.; Novak, J.; Bienertova-Vasku, J. Muscle-Specific MicroRNAs in Skeletal Muscle Development. Dev. Biol. 2016, 410, 1–13. [Google Scholar] [CrossRef]
  40. Wang, J.; Yang, L.Z.; Zhang, J.S.; Gong, J.X.; Wang, Y.H.; Zhang, C.L.; Chen, H.; Fang, X.T. Effects of MicroRNAs on Skeletal Muscle Development. Gene 2018, 668, 107–113. [Google Scholar] [CrossRef]
  41. Zhao, Y.; Samal, E.; Srivastava, D. Serum Response Factor Regulates a Muscle-Specific MicroRNA That Targets Hand2 during Cardiogenesis. Nature 2005, 436, 214–220. [Google Scholar] [CrossRef] [PubMed]
  42. Chen, J.F.; Mandel, E.M.; Thomson, J.M.; Wu, Q.; Callis, T.E.; Hammond, S.M.; Conlon, F.L.; Wang, D.Z. The Role of MicroRNA-1 and MicroRNA-133 in Skeletal Muscle Proliferation and Differentiation. Nat. Genet. 2006, 38, 228–233. [Google Scholar] [CrossRef] [PubMed]
  43. Liu, N.; Williams, A.H.; Kim, Y.; McAnally, J.; Bezprozvannaya, S.; Sutherland, L.B.; Richardson, J.A.; Bassel-Duby, R.; Olson, E.N. An Intragenic MEF2-Dependent Enhancer Directs Muscle-Specific Expression of MicroRNAs 1 and 133. Proc. Natl. Acad. Sci. USA 2007, 104, 20844–20849. [Google Scholar] [CrossRef]
  44. Aravin, A.A.; Naumova, N.M.; Tulin, A.V.; Vagin, V.V.; Rozovsky, Y.M.; Gvozdev, V.A. Double-Stranded RNA-Mediated Silencing of Genomic Tandem Repeats and Transposable Elements in the D. Melanogaster Germline. Curr. Biol. 2001, 11, 1017–1027. [Google Scholar] [CrossRef]
  45. Siomi, M.C.; Sato, K.; Pezic, D.; Aravin, A.A. PIWI-Interacting Small RNAs: The Vanguard of Genome Defence. Nat. Rev. Mol. Cell Biol. 2011, 12, 246–258. [Google Scholar] [CrossRef] [PubMed]
  46. Vagin, V.V.; Sigova, A.; Li, C.; Seitz, H.; Gvozdev, V.; Zamore, P.D. A Distinct Small RNA Pathway Silences Selfish Genetic Elements in the Germline. Science 2006, 313, 320–324. [Google Scholar] [CrossRef] [PubMed]
  47. Grivna, S.T.; Beyret, E.; Wang, Z.; Lin, H. A Novel Class of Small RNAs in Mouse Spermatogenic Cells. Genes Dev. 2006, 20, 1709–1714. [Google Scholar] [CrossRef]
  48. Yan, Z.; Hu, H.Y.; Jiang, X.; Maierhofer, V.; Neb, E.; He, L.; Hu, Y.; Hu, H.; Li, N.; Chen, W.; et al. Widespread Expression of PiRNA-like Molecules in Somatic Tissues. Nucleic Acids Res. 2011, 39, 6596–6607. [Google Scholar] [CrossRef]
  49. Perera, B.P.U.; Tsai, Z.T.Y.; Colwell, M.L.; Jones, T.R.; Goodrich, J.M.; Wang, K.; Sartor, M.A.; Faulk, C.; Dolinoy, D.C. Somatic Expression of PiRNA and Associated Machinery in the Mouse Identifies Short, Tissue-Specific PiRNA. Epigenetics 2019, 14, 504–521. [Google Scholar] [CrossRef]
  50. Brennecke, J.; Aravin, A.A.; Stark, A.; Dus, M.; Kellis, M.; Sachidanandam, R.; Hannon, G.J. Discrete Small RNA-Generating Loci as Master Regulators of Transposon Activity in Drosophila. Cell 2007, 128, 1089–1103. [Google Scholar] [CrossRef]
  51. Robine, N.; Lau, N.C.; Balla, S.; Jin, Z.; Okamura, K.; Kuramochi-Miyagawa, S.; Blower, M.D.; Lai, E.C. A Broadly Conserved Pathway Generates 3’UTR-Directed Primary PiRNAs. Curr. Biol. 2009, 19, 2066–2076. [Google Scholar] [CrossRef] [PubMed]
  52. Saito, K.; Inagaki, S.; Mituyama, T.; Kawamura, Y.; Ono, Y.; Sakota, E.; Kotani, H.; Asai, K.; Siomi, H.; Siomi, M.C. A Regulatory Circuit for Piwi by the Large Maf Gene Traffic Jam in Drosophila. Nature 2009, 461, 1296–1299. [Google Scholar] [CrossRef] [PubMed]
  53. Weick, E.M.; Miska, E.A. PiRNAs: From Biogenesis to Function. Development 2014, 141, 3458–3471. [Google Scholar] [CrossRef]
  54. Cox, D.N.; Chao, A.; Baker, J.; Chang, L.; Qiao, D.; Lin, H. A Novel Class of Evolutionarily Conserved Genes Defined by Piwi Are Essential for Stem Cell Self-Renewal. Genes Dev. 1998, 12, 3715–3727. [Google Scholar] [CrossRef] [PubMed]
  55. Deng, W.; Lin, H. Miwi, a Murine Homolog of Piwi, Encodes a Cytoplasmic Protein Essential for Spermatogenesis. Dev. Cell 2002, 2, 819–830. [Google Scholar] [CrossRef]
  56. Aravin, A.; Gaidatzis, D.; Pfeffer, S.; Lagos-Quintana, M.; Landgraf, P.; Iovino, N.; Morris, P.; Brownstein, M.J.; Kuramochi-Miyagawa, S.; Nakano, T.; et al. A Novel Class of Small RNAs Bind to MILI Protein in Mouse Testes. Nature 2006, 442, 203–207. [Google Scholar] [CrossRef] [PubMed]
  57. Girard, A.; Sachidanandam, R.; Hannon, G.J.; Carmell, M.A. A Germline-Specific Class of Small RNAs Binds Mammalian Piwi Proteins. Nature 2006, 442, 199–202. [Google Scholar] [CrossRef]
  58. Brannan, C.I.; Dees, E.C.; Ingram, R.S.; Tilghman, S.M. The Product of the H19 Gene May Function as an RNA. Mol. Cell Biol. 1990, 10, 28–36. [Google Scholar] [CrossRef]
  59. Brockdorff, N.; Ashworth, A.; Kay, G.F.; McCabe, V.M.; Norris, D.P.; Cooper, P.J.; Swift, S.; Rastan, S. The Product of the Mouse Xist Gene Is a 15 Kb Inactive X-Specific Transcript Containing No Conserved ORF and Located in the Nucleus. Cell 1992, 71, 515–526. [Google Scholar] [CrossRef]
  60. Brown, C.J.; Hendrich, B.D.; Rupert, J.L.; Lafrenière, R.G.; Xing, Y.; Lawrence, J.; Willard, H.F. The Human XIST Gene: Analysis of a 17 Kb Inactive X-Specific RNA That Contains Conserved Repeats and Is Highly Localized within the Nucleus. Cell 1992, 71, 527–542. [Google Scholar] [CrossRef]
  61. Alessio, E.; Bonadio, R.S.; Buson, L.; Chemello, F.; Cagnin, S. A Single Cell but Many Different Transcripts: A Journey into the World of Long Non-Coding RNAs. Int. J. Mol. Sci. 2020, 21, 302. [Google Scholar] [CrossRef] [PubMed]
  62. Yoon, J.H.; Abdelmohsen, K.; Gorospe, M. Posttranscriptional Gene Regulation by Long Noncoding RNA. J. Mol. Biol. 2013, 425, 3723–3730. [Google Scholar] [CrossRef] [PubMed]
  63. Iyer, M.K.; Niknafs, Y.S.; Malik, R.; Singhal, U.; Sahu, A.; Hosono, Y.; Barrette, T.R.; Prensner, J.R.; Evans, J.R.; Zhao, S.; et al. The Landscape of Long Noncoding RNAs in the Human Transcriptome. Nat. Genet. 2015, 47, 199–208. [Google Scholar] [CrossRef] [PubMed]
  64. Hon, C.C.; Ramilowski, J.A.; Harshbarger, J.; Bertin, N.; Rackham, O.J.L.; Gough, J.; Denisenko, E.; Schmeier, S.; Poulsen, T.M.; Severin, J.; et al. An Atlas of Human Long Non-Coding RNAs with Accurate 5’ Ends. Nature 2017, 543, 199–204. [Google Scholar] [CrossRef]
  65. Guttman, M.; Amit, I.; Garber, M.; French, C.; Lin, M.F.; Feldser, D.; Huarte, M.; Zuk, O.; Carey, B.W.; Cassady, J.P.; et al. Chromatin Signature Reveals over a Thousand Highly Conserved Large Non-Coding RNAs in Mammals. Nature 2009, 458, 223–227. [Google Scholar] [CrossRef]
  66. Clark, M.B.; Mattick, J.S. Long Noncoding RNAs in Cell Biology. Semin Cell Dev. Biol. 2011, 22, 366–376. [Google Scholar] [CrossRef]
  67. Batista, P.J.; Chang, H.Y. Long Noncoding RNAs: Cellular Address Codes in Development and Disease. Cell 2013, 152, 1298–1307. [Google Scholar] [CrossRef]
  68. Sauvageau, M.; Goff, L.A.; Lodato, S.; Bonev, B.; Groff, A.F.; Gerhardinger, C.; Sanchez-Gomez, D.B.; Hacisuleyman, E.; Li, E.; Spence, M.; et al. Multiple Knockout Mouse Models Reveal LincRNAs Are Required for Life and Brain Development. eLife 2013, 2, e01749. [Google Scholar] [CrossRef]
  69. Guttman, M.; Donaghey, J.; Carey, B.W.; Garber, M.; Grenier, J.K.; Munson, G.; Young, G.; Lucas, A.B.; Ach, R.; Bruhn, L.; et al. LincRNAs Act in the Circuitry Controlling Pluripotency and Differentiation. Nature 2011, 477, 7364. [Google Scholar] [CrossRef]
  70. Klattenhoff, C.A.; Scheuermann, J.C.; Surface, L.E.; Bradley, R.K.; Fields, P.A.; Steinhauser, M.L.; Ding, H.; Butty, V.L.; Torrey, L.; Haas, S.; et al. Braveheart, a Long Noncoding RNA Required for Cardiovascular Lineage Commitment. Cell 2013, 152, 570–583. [Google Scholar] [CrossRef]
  71. Plath, K.; Mlynarczyk-Evans, S.; Nusinow, D.A.; Panning, B. Xist RNA and the Mechanism of X Chromosome Inactivation. Annu. Rev. Genet. 2002, 36, 233–278. [Google Scholar] [CrossRef] [PubMed]
  72. Davidovich, C.; Cech, T.R. The Recruitment of Chromatin Modifiers by Long Noncoding RNAs: Lessons from PRC2. RNA 2015, 21, 2007–2022. [Google Scholar] [CrossRef]
  73. Jin, J.J.; Lv, W.; Xia, P.; Xu, Z.Y.; Zheng, A.D.; Wang, X.J.; Wang, S.S.; Zeng, R.; Luo, H.M.; Li, G.L.; et al. Long Noncoding RNA SYISL Regulates Myogenesis by Interacting with Polycomb Repressive Complex 2. Proc. Natl. Acad. Sci. USA 2018, 115, E9802–E9811. [Google Scholar] [CrossRef] [PubMed]
  74. Cajigas, I.; Chakraborty, A.; Swyter, K.R.; Luo, H.; Bastidas, M.; Nigro, M.; Morris, E.R.; Chen, S.; VanGompel, M.J.W.; Leib, D.; et al. The Evf2 Ultraconserved Enhancer LncRNA Functionally and Spatially Organizes Megabase Distant Genes in the Developing Forebrain. Mol. Cell 2018, 71, 956–972.e9. [Google Scholar] [CrossRef] [PubMed]
  75. Tripathi, V.; Ellis, J.D.; Shen, Z.; Song, D.Y.; Pan, Q.; Watt, A.T.; Freier, S.M.; Bennett, C.F.; Sharma, A.; Bubulya, P.A.; et al. The Nuclear-Retained Noncoding RNA MALAT1 Regulates Alternative Splicing by Modulating SR Splicing Factor Phosphorylation. Mol. Cell 2010, 39, 925–938. [Google Scholar] [CrossRef]
  76. Vígh, L.; Török, Z.; Balogh, G.; Glatz, A.; Piotto, S.; Horváth, I. Membrane-Regulated Stress Response: A Theoretical and Practical Approach. Adv. Exp. Med. Biol. 2007, 594, 114–131. [Google Scholar] [CrossRef]
  77. Lindquist, S. The Heat-Shock Response. Annu. Rev. Biochem. 1986, 55, 1151–1191. [Google Scholar] [CrossRef]
  78. Kampinga, H.H.; Hageman, J.; Vos, M.J.; Kubota, H.; Tanguay, R.M.; Bruford, E.A.; Cheetham, M.E.; Chen, B.; Hightower, L.E. Guidelines for the Nomenclature of the Human Heat Shock Proteins. Cell Stress Chaperones 2009, 14, 105–111. [Google Scholar] [CrossRef]
  79. Macario, A.J.L.; Conway de Macario, E. Sick Chaperones, Cellular Stress, and Disease. N. Engl. J. Med. 2005, 353, 1489–1501. [Google Scholar] [CrossRef]
  80. Marino Gammazza, A.; Caruso Bavisotto, C.; David, S.; Barone, R.; Rappa, F.; Campanella, C.; Conway de Macario, E.; Cappello, F.; Macario, A.J.L. HSP60 Is a Ubiquitous Player in the Physiological and Pathogenic Interactions between the Chaperoning and the Immune Systems. Curr. Immunol. Rev. 2017, 13, 44–55. [Google Scholar] [CrossRef]
  81. D’Anneo, A.; Caruso Bavisotto, C.; Marino Gammazza, A.; Paladino, L.; Carlisi, D.; Cappello, F.; Conway de Macario, E.; Macario, A.J.L.; Lauricella, M. Lipid Chaperones and Associated Diseases: A Group of Chaperonopathies Defining a New Nosological Entity with Implications for Medical Research and Practice. Cell Stress Chaperones 2020, 25, 805–820. [Google Scholar] [CrossRef] [PubMed]
  82. Caruso Bavisotto, C.; Marino Gammazza, A.; Lo Cascio, F.; Mocciaro, E.; Vitale, A.M.; Vergilio, G.; Pace, A.; Cappello, F.; Campanella, C.; Palumbo Piccionello, A. Curcumin Affects HSP60 Folding Activity and Levels in Neuroblastoma Cells. Int. J. Mol. Sci. 2020, 21, 661. [Google Scholar] [CrossRef] [PubMed]
  83. Dai, C.; Whitesell, L.; Rogers, A.B.; Lindquist, S. Heat Shock Factor 1 Is a Powerful Multifaceted Modifier of Carcinogenesis. Cell 2007, 130, 1005–1018. [Google Scholar] [CrossRef] [PubMed]
  84. Biamonti, G.; Caceres, J.F. Cellular Stress and RNA Splicing. Trends Biochem. Sci. 2009, 34, 146–153. [Google Scholar] [CrossRef] [PubMed]
  85. Caruso Bavisotto, C.; Alberti, G.; Vitale, A.M.; Paladino, L.; Campanella, C.; Rappa, F.; Gorska, M.; Conway de Macario, E.; Cappello, F.; Macario, A.J.L.; et al. Hsp60 Post-Translational Modifications: Functional and Pathological Consequences. Front. Mol. Biosci. 2020, 7, 95. [Google Scholar] [CrossRef]
  86. Xu, C.; Lu, Y.; Pan, Z.; Chu, W.; Luo, X.; Lin, H.; Xiao, J.; Shan, H.; Wang, Z.; Yang, B. The Muscle-Specific MicroRNAs MiR-1 and MiR-133 Produce Opposing Effects on Apoptosis by Targeting HSP60, HSP70 and Caspase-9 in Cardiomyocytes. J. Cell Sci. 2007, 120, 3045–3052. [Google Scholar] [CrossRef]
  87. Shan, Z.X.; Lin, Q.X.; Deng, C.Y.; Zhu, J.N.; Mai, L.P.; Liu, J.L.; Fu, Y.H.; Liu, X.Y.; Li, Y.X.; Zhang, Y.Y.; et al. MiR-1/MiR-206 Regulate Hsp60 Expression Contributing to Glucose-Mediated Apoptosis in Cardiomyocytes. FEBS Lett. 2010, 584, 3592–3600. [Google Scholar] [CrossRef]
  88. Yu, J.; Liu, F.; Yin, P.; Zhu, X.; Cheng, G.; Wang, N.; Lu, A.; Luan, W.; Zhang, N.; Li, J.; et al. Integrating MiRNA and MRNA Expression Profiles in Response to Heat Stress-Induced Injury in Rat Small Intestine. Funct. Integr. Genom. 2011, 11, 203–213. [Google Scholar] [CrossRef]
  89. Mahmoudi, Z.; Karamali, N.; Roghani, S.A.; Assar, S.; Pournazari, M.; Soufivand, P.; Salari, F.; Rezaiemanesh, A. Efficacy of DMARDs and Methylprednisolone Treatment on the Gene Expression Levels of HSPA5, MMD, and Non-Coding RNAs MALAT1, H19, MiR-199a-5p, and MiR-1-3p, in Patients with Rheumatoid Arthritis. Int. Immunopharmacol. 2022, 108, 108878. [Google Scholar] [CrossRef]
  90. Kelly, F.J. Oxidative Stress: Its Role in Air Pollution and Adverse Health Effects. Occup. Environ. Med. 2003, 60, 612–616. [Google Scholar] [CrossRef]
  91. González-Flecha, B. Oxidant Mechanisms in Response to Ambient Air Particles. Mol. Asp. Med. 2004, 25, 169–182. [Google Scholar] [CrossRef] [PubMed]
  92. Risom, L.; Møller, P.; Loft, S. Oxidative Stress-Induced DNA Damage by Particulate Air Pollution. Mutat. Res. Fundam. Mol. Mech. Mutagen. 2005, 592, 119–137. [Google Scholar] [CrossRef] [PubMed]
  93. Miguel, V.; Lamas, S.; Espinosa-Diez, C. Role of Non-Coding-RNAs in Response to Environmental Stressors and Consequences on Human Health. Redox Biol. 2020, 37, 101580. [Google Scholar] [CrossRef] [PubMed]
  94. González-Martín, J.; Kraakman, N.J.R.; Pérez, C.; Lebrero, R.; Muñoz, R. A State-of-the-Art Review on Indoor Air Pollution and Strategies for Indoor Air Pollution Control. Chemosphere 2021, 262, 128376. [Google Scholar] [CrossRef]
  95. Anderson, J.O.; Thundiyil, J.G.; Stolbach, A. Clearing the Air: A Review of the Effects of Particulate Matter Air Pollution on Human Health. J. Med. Toxicol. 2012, 8, 166–175. [Google Scholar] [CrossRef]
  96. Carbon Monoxide Poisoning—Canada.Ca. Available online: https://www.canada.ca/en/health-canada/services/air-quality/indoor-air-contaminants/keep-carbon-monoxide-out-your-home.html (accessed on 18 February 2023).
  97. National Pollutant Inventory—DCCEEW. Available online: https://www.dcceew.gov.au/environment/protection/npi (accessed on 18 February 2023).
  98. Jin, Y.; Veiga, M.C.; Kennes, C. Bioprocesses for the Removal of Nitrogen Oxides from Polluted Air. J. Chem. Technol. Biotechnol. 2005, 80, 483–494. [Google Scholar] [CrossRef]
  99. Gli Inquinanti|Ministero Dell’Ambiente e Della Sicurezza Energetica. Available online: https://www.mase.gov.it/pagina/gli-inquinanti (accessed on 19 February 2023).
  100. Heicklen, J. The Formation and Inhibition of Photochemical Smog. Ann. N. Y. Acad. Sci. 1987, 502, 145–159. [Google Scholar] [CrossRef]
  101. Zhang, Y.; Cheng, M.; Gao, J.; Li, J. Review of the Influencing Factors of Secondary Organic Aerosol Formation and Aging Mechanism Based on Photochemical Smog Chamber Simulation Methods. J. Environ. Sci. 2023, 123, 545–559. [Google Scholar] [CrossRef]
  102. Déméautis, T.; Delles, M.; Tomaz, S.; Monneret, G.; Glehen, O.; Devouassoux, G.; George, C.; Bentaher, A. Pathogenic Mechanisms of Secondary Organic Aerosols. Chem. Res. Toxicol. 2022, 35, 1146–1161. [Google Scholar] [CrossRef]
  103. Kinney, P.L. Climate Change, Air Quality, and Human Health. Am. J. Prev. Med. 2008, 35, 459–467. [Google Scholar] [CrossRef]
  104. van der Pol, E.; Böing, A.N.; Harrison, P.; Sturk, A.; Nieuwland, R. Classification, Functions, and Clinical Relevance of Extracellular Vesicles. Pharmacol. Rev. 2012, 64, 676–705. [Google Scholar] [CrossRef] [PubMed]
  105. Yuana, Y.; Sturk, A.; Nieuwland, R. Extracellular Vesicles in Physiological and Pathological Conditions. Blood Rev. 2013, 27, 31–39. [Google Scholar] [CrossRef]
  106. Yáñez-Mó, M.; Siljander, P.R.M.; Andreu, Z.; Zavec, A.B.; 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. [Google Scholar] [CrossRef]
  107. Zaborowski, M.P.; Balaj, L.; Breakefield, X.O.; Lai, C.P. Extracellular Vesicles: Composition, Biological Relevance, and Methods of Study. Bioscience 2015, 65, 783–797. [Google Scholar] [CrossRef] [PubMed]
  108. van Niel, G.; D’Angelo, G.; Raposo, G. Shedding Light on the Cell Biology of Extracellular Vesicles. Nat. Rev. Mol. Cell Biol. 2018, 19, 213–228. [Google Scholar] [CrossRef] [PubMed]
  109. Caby, M.P.; Lankar, D.; Vincendeau-Scherrer, C.; Raposo, G.; Bonnerot, C. Exosomal-like Vesicles Are Present in Human Blood Plasma. Int. Immunol. 2005, 17, 879–887. [Google Scholar] [CrossRef] [PubMed]
  110. Pisitkun, T.; Shen, R.F.; Knepper, M.A. Identification and Proteomic Profiling of Exosomes in Human Urine. Proc. Natl. Acad. Sci. USA 2004, 101, 13368–13373. [Google Scholar] [CrossRef]
  111. Michael, A.; Bajracharya, S.D.; Yuen, P.S.T.; Zhou, H.; Star, R.A.; Illei, G.G.; Alevizos, I. Exosomes from Human Saliva as a Source of MicroRNA Biomarkers. Oral Dis. 2010, 16, 34–38. [Google Scholar] [CrossRef]
  112. Manek, R.; Moghieb, A.; Yang, Z.; Kumar, D.; Kobessiy, F.; Sarkis, G.A.; Raghavan, V.; Wang, K.K.W. Protein Biomarkers and Neuroproteomics Characterization of Microvesicles/Exosomes from Human Cerebrospinal Fluid Following Traumatic Brain Injury. Mol. Neurobiol. 2018, 55, 6112–6128. [Google Scholar] [CrossRef]
  113. Admyre, C.; Johansson, S.M.; Qazi, K.R.; Filén, J.-J.; Lahesmaa, R.; Norman, M.; Neve, E.P.A.; Scheynius, A.; Gabrielsson, S. Exosomes with Immune Modulatory Features Are Present in Human Breast Milk. J. Immunol. 2007, 179, 1969–1978. [Google Scholar] [CrossRef]
  114. Admyre, C.; Grunewald, J.; Thyberg, J.; Bripenäck, S.; Tornling, G.; Eklund, A.; Scheynius, A.; Gabrielsson, S. Exosomes with Major Histocompatibility Complex Class II and Co-Stimulatory Molecules Are Present in Human BAL Fluid. Eur. Respir. J. 2003, 22, 578–583. [Google Scholar] [CrossRef] [PubMed]
  115. Cheng, L.; Sharples, R.A.; Scicluna, B.J.; Hill, A.F. Exosomes Provide a Protective and Enriched Source of MiRNA for Biomarker Profiling Compared to Intracellular and Cell-Free Blood. J. Extracell. Vesicles 2014, 3, 23743. [Google Scholar] [CrossRef] [PubMed]
  116. Mulcahy, L.A.; Pink, R.C.; Carter, D.R.F. Routes and Mechanisms of Extracellular Vesicle Uptake. J. Extracell. Vesicles 2014, 3, 24641. [Google Scholar] [CrossRef] [PubMed]
  117. Colombo, M.; Raposo, G.; Théry, C. Biogenesis, Secretion, and Intercellular Interactions of Exosomes and Other Extracellular Vesicles. Annu. Rev. Cell Dev. Biol. 2014, 30, 255–289. [Google Scholar] [CrossRef]
  118. Théry, C.; Witwer, K.W.; Aikawa, E.; Alcaraz, M.J.; Anderson, J.D.; Andriantsitohaina, R.; Antoniou, A.; Arab, T.; Archer, F.; Atkin-Smith, G.K.; et al. Minimal Information for Studies of Extracellular Vesicles 2018 (MISEV2018): A Position Statement of the International Society for Extracellular Vesicles and Update of the MISEV2014 Guidelines. J. Extracell. Vesicles 2018, 7, 1535750. [Google Scholar] [CrossRef]
  119. O’Brien, K.; Breyne, K.; Ughetto, S.; Laurent, L.C.; Breakefield, X.O. RNA Delivery by Extracellular Vesicles in Mammalian Cells and Its Applications. Nat. Rev. Mol. Cell Biol. 2020, 21, 585–606. [Google Scholar] [CrossRef]
  120. Valadi, H.; Ekström, K.; Bossios, A.; Sjöstrand, M.; Lee, J.J.; Lötvall, 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. [Google Scholar] [CrossRef]
  121. Fabbiano, F.; Corsi, J.; Gurrieri, E.; Trevisan, C.; Notarangelo, M.; D’Agostino, V.G. RNA Packaging into Extracellular Vesicles: An Orchestra of RNA-binding Proteins? J. Extracell. Vesicles 2020, 10. [Google Scholar] [CrossRef]
  122. Heitzer, E.; Haque, I.S.; Roberts, C.E.S.; Speicher, M.R. Current and Future Perspectives of Liquid Biopsies in Genomics-Driven Oncology. Nat. Rev. Genet. 2019, 20, 71–88. [Google Scholar] [CrossRef]
  123. Mittelbrunn, M.; Gutiérrez-Vázquez, C.; Villarroya-Beltri, C.; González, S.; Sánchez-Cabo, F.; González, M.Á.; Bernad, A.; Sánchez-Madrid, F. Unidirectional Transfer of MicroRNA-Loaded Exosomes from T Cells to Antigen-Presenting Cells. Nat. Commun. 2011, 2, 282. [Google Scholar] [CrossRef]
  124. Baglio, S.R.; Rooijers, K.; Koppers-Lalic, D.; Verweij, F.J.; Pérez Lanzón, M.; Zini, N.; Naaijkens, B.; Perut, F.; Niessen, H.W.M.; Baldini, N.; et al. Human Bone Marrow- and Adipose-Mesenchymal Stem Cells Secrete Exosomes Enriched in Distinctive MiRNA and TRNA Species. Stem Cell Res. Ther. 2015, 6, 127. [Google Scholar] [CrossRef] [PubMed]
  125. Villarroya-Beltri, C.; Gutiérrez-Vázquez, C.; Sánchez-Cabo, F.; Pérez-Hernández, D.; Vázquez, J.; Martin-Cofreces, N.; Martinez-Herrera, D.J.; Pascual-Montano, A.; Mittelbrunn, M.; Sánchez-Madrid, F. Sumoylated HnRNPA2B1 Controls the Sorting of MiRNAs into Exosomes through Binding to Specific Motifs. Nat. Commun. 2013, 4, 2980. [Google Scholar] [CrossRef]
  126. Iavello, A.; Frech, V.S.L.; Gai, C.; Deregibus, M.C.; Quesenberry, P.J.; Camussi, G. Role of Alix in MiRNA Packaging during Extracellular Vesicle Biogenesis. Int. J. Mol. Med. 2016, 37, 958–966. [Google Scholar] [CrossRef] [PubMed]
  127. Hagiwara, K.; Katsuda, T.; Gailhouste, L.; Kosaka, N.; Ochiya, T. Commitment of Annexin A2 in Recruitment of MicroRNAs into Extracellular Vesicles. FEBS Lett. 2015, 589, 4071–4078. [Google Scholar] [CrossRef] [PubMed]
  128. Sork, H.; Corso, G.; Krjutskov, K.; Johansson, H.J.; Nordin, J.Z.; Wiklander, O.P.B.; Lee, Y.X.F.; Westholm, J.O.; Lehtiö, J.; Wood, M.J.A.; et al. Heterogeneity and Interplay of the Extracellular Vesicle Small RNA Transcriptome and Proteome. Sci. Rep. 2018, 8, 10813. [Google Scholar] [CrossRef]
  129. Wani, S.; Kaul, D. Cancer Cells Govern MiR-2909 Exosomal Recruitment through Its 3’-End Post-Transcriptional Modification. Cell Biochem. Funct. 2018, 36, 106–111. [Google Scholar] [CrossRef]
  130. Meng, W.; He, C.; Hao, Y.; Wang, L.; Li, L.; Zhu, G. Prospects and Challenges of Extracellular Vesicle-Based Drug Delivery System: Considering Cell Source. Drug Deliv. 2020, 27, 585–598. [Google Scholar] [CrossRef]
  131. Eckhardt, C.M.; Baccarelli, A.A.; Wu, H. Environmental Exposures and Extracellular Vesicles: Indicators of Systemic Effects and Human Disease. Curr. Environ. Health Rep. 2022, 9, 465–476. [Google Scholar] [CrossRef]
  132. Guiot, J.; Struman, I.; Louis, E.; Louis, R.; Malaise, M.; Njock, M.S. Exosomal MiRNAs in Lung Diseases: From Biologic Function to Therapeutic Targets. J. Clin. Med. 2019, 8, 1345. [Google Scholar] [CrossRef]
  133. Rao Tata, P.; Rajagopal, J. Plasticity in the Lung: Making and Breaking Cell Identity. Development 2017, 144, 755–766. [Google Scholar] [CrossRef]
  134. Bollati, V.; Angelici, L.; Rizzo, G.; Pergoli, L.; Rota, F.; Hoxha, M.; Nordio, F.; Bonzini, M.; Tarantini, L.; Cantone, L.; et al. Microvesicle-Associated MicroRNA Expression Is Altered upon Particulate Matter Exposure in Healthy Workers and in A549 Cells. J. Appl. Toxicol. 2015, 35, 59–67. [Google Scholar] [CrossRef] [PubMed]
  135. Bonzini, M.; Pergoli, L.; Cantone, L.; Hoxha, M.; Spinazzè, A.; del Buono, L.; Favero, C.; Carugno, M.; Angelici, L.; Broggi, L.; et al. Short-Term Particulate Matter Exposure Induces Extracellular Vesicle Release in Overweight Subjects. Environ. Res. 2017, 155, 228. [Google Scholar] [CrossRef] [PubMed]
  136. Bourdonnay, E.; Zasłona, Z.; Penke, L.R.K.; Speth, J.M.; Schneider, D.J.; Przybranowski, S.; Swanson, J.A.; Mancuso, P.; Freeman, C.M.; Curtis, J.L.; et al. Transcellular Delivery of Vesicular SOCS Proteins from Macrophages to Epithelial Cells Blunts Inflammatory Signaling. J. Exp. Med. 2015, 212, 729–742. [Google Scholar] [CrossRef]
  137. Pope, C.A.; Bhatnagar, A.; McCracken, J.P.; Abplanalp, W.; Conklin, D.J.; O’Toole, T. Exposure to Fine Particulate Air Pollution Is Associated with Endothelial Injury and Systemic Inflammation. Circ. Res. 2016, 119, 1204–1214. [Google Scholar] [CrossRef] [PubMed]
  138. Lim, J.h.; Song, M.K.; Cho, Y.; Kim, W.; Han, S.O.; Ryu, J.C. Expression of Exosomal and Cellular MicroRNAs: As Biomarkers for Toluene, Ethylbenzene, Xylene (TEX) Exposure. Mol. Cell. Toxicol. 2016, 12, 359–369. [Google Scholar] [CrossRef]
  139. Pavanello, S.; Bonzini, M.; Angelici, L.; Motta, V.; Pergoli, L.; Hoxha, M.; Cantone, L.; Pesatori, A.C.; Apostoli, P.; Tripodi, A.; et al. Extracellular Vesicle-Driven Information Mediates the Long-Term Effects of Particulate Matter Exposure on Coagulation and Inflammation Pathways. Toxicol. Lett. 2016, 259, 143–150. [Google Scholar] [CrossRef]
  140. Stassen, F.R.M.; van Eijck, P.H.; Savelkoul, P.H.M.; Wouters, E.F.M.; Rohde, G.G.U.; Briedé, J.J.; Reynaert, N.L.; de Kok, T.M.; Benedikter, B.J. Cell Type- and Exposure-Specific Modulation of CD63/CD81-Positive and Tissue Factor-Positive Extracellular Vesicle Release in Response to Respiratory Toxicants. Oxid. Med. Cell. Longev. 2019, 2019, 5204218. [Google Scholar] [CrossRef]
  141. Schraufnagel, D.E.; Balmes, J.R.; Cowl, C.T.; de Matteis, S.; Jung, S.H.; Mortimer, K.; Perez-Padilla, R.; Rice, M.B.; Riojas-Rodriguez, H.; Sood, A.; et al. Air Pollution and Noncommunicable Diseases: A Review by the Forum of International Respiratory Societies’ Environmental Committee, Part 2: Air Pollution and Organ Systems. Chest 2019, 155, 417–426. [Google Scholar] [CrossRef]
  142. Cheng, M.; Wang, B.; Yang, M.; Ma, J.; Ye, Z.; Xie, L.; Zhou, M.; Chen, W. MicroRNAs Expression in Relation to Particulate Matter Exposure: A Systematic Review. Environ. Pollut. 2020, 260, 113961. [Google Scholar] [CrossRef]
  143. Hou, T.; Chen, Q.; Ma, Y. Elevated Expression of MiR-146 Involved in Regulating Mice Pulmonary Dysfunction after Exposure to PM2.5. J. Toxicol. Sci. 2021, 46, 437–443. [Google Scholar] [CrossRef]
  144. Zheng, R.; Du, M.; Tian, M.; Zhu, Z.; Wei, C.; Chu, H.; Gan, C.; Liang, J.; Xue, R.; Gao, F.; et al. Fine Particulate Matter Induces Childhood Asthma Attacks via Extracellular Vesicle-Packaged Let-7i-5p-Mediated Modulation of the MAPK Signaling Pathway. Adv. Sci. 2022, 9, 2102460. [Google Scholar] [CrossRef]
  145. Paramanantham, A.; Asfiya, R.; Das, S.; McCully, G.; Srivastava, A. Extracellular Vesicle (EVs) Associated Non-Coding RNAs in Lung Cancer and Therapeutics. Int. J. Mol. Sci. 2022, 23, 13637. [Google Scholar] [CrossRef] [PubMed]
  146. Wang, Y.; Zhong, Y.; Sun, K.; Fan, Y.; Liao, J.; Wang, G. Identification of Exosome MiRNAs in Bronchial Epithelial Cells after PM2.5 Chronic Exposure. Ecotoxicol. Environ. Saf. 2021, 215, 112127. [Google Scholar] [CrossRef] [PubMed]
  147. He, X.; Park, S.; Chen, Y.; Lee, H. Extracellular Vesicle-Associated MiRNAs as a Biomarker for Lung Cancer in Liquid Biopsy. Front. Mol. Biosci. 2021, 8, 630718. [Google Scholar] [CrossRef] [PubMed]
  148. Thandra, K.C.; Barsouk, A.; Saginala, K.; Aluru, J.S.; Barsouk, A. Epidemiology of Lung Cancer. Contemp. Oncol. 2021, 25, 45–52. [Google Scholar] [CrossRef]
  149. Djebali, S.; Davis, C.A.; Merkel, A.; Dobin, A.; Lassmann, T.; Mortazavi, A.; Tanzer, A.; Lagarde, J.; Lin, W.; Schlesinger, F.; et al. Landscape of Transcription in Human Cells. Nature 2012, 489, 101–108. [Google Scholar] [CrossRef]
  150. Loewen, G.; Jayawickramarajah, J.; Zhuo, Y.; Shan, B. Functions of LncRNA HOTAIR in Lung Cancer. J. Hematol. Oncol. 2014, 7, 90. [Google Scholar] [CrossRef]
  151. Li, S.; Mei, Z.; Hu, H.B.; Zhang, X. The LncRNA MALAT1 Contributes to Non-Small Cell Lung Cancer Development via Modulating MiR-124/STAT3 Axis. J. Cell Physiol. 2018, 233, 6679–6688. [Google Scholar] [CrossRef]
  152. Li, J.; Wang, J.; Chen, Y.; Li, S.; Jin, M.; Wang, H.; Chen, Z.; Yu, W. LncRNA MALAT1 Exerts Oncogenic Functions in Lung Adenocarcinoma by Targeting MiR-204. Am. J. Cancer Res. 2016, 6, 1099. [Google Scholar]
  153. Zhang, Y.L.; Li, X.B.; Hou, Y.X.; Fang, N.Z.; You, J.C.; Zhou, Q.H. The LncRNA XIST Exhibits Oncogenic Properties via Regulation of MiR-449a and Bcl-2 in Human Non-Small Cell Lung Cancer Article Has Been Corrected since Advanced Online Publication, and an Erratum Is Also Printed in This Issue. Acta Pharmacol. Sin. 2017, 38, 371. [Google Scholar] [CrossRef]
  154. Hogg, J.C.; Chu, F.; Utokaparch, S.; Woods, R.; Elliott, W.M.; Buzatu, L.; Cherniack, R.M.; Rogers, R.M.; Sciurba, F.C.; Coxson, H.O.; et al. The Nature of Small-Airway Obstruction in Chronic Obstructive Pulmonary Disease. N. Engl. J. Med. 2004, 350, 2645–2653. [Google Scholar] [CrossRef] [PubMed]
  155. Fujita, Y.; Araya, J.; Ito, S.; Kobayashi, K.; Kosaka, N.; Yoshioka, Y.; Kadota, T.; Hara, H.; Kuwano, K.; Ochiya, T. Suppression of Autophagy by Extracellular Vesicles Promotes Myofibroblast Differentiation in COPD Pathogenesis. J. Extracell. Vesicles 2015, 4, 28388. [Google Scholar] [CrossRef] [PubMed]
  156. Serban, K.A.; Rezania, S.; Petrusca, D.N.; Poirier, C.; Cao, D.; Justice, M.J.; Patel, M.; Tsvetkova, I.; Kamocki, K.; Mikosz, A.; et al. Structural and Functional Characterization of Endothelial Microparticles Released by Cigarette Smoke. Sci. Rep. 2016, 6, 31596. [Google Scholar] [CrossRef] [PubMed]
  157. Graff, J.W.; Powers, L.S.; Dickson, A.M.; Kim, J.; Reisetter, A.C.; Hassan, I.H.; Kremens, K.; Gross, T.J.; Wilson, M.E.; Monick, M.M. Cigarette Smoking Decreases Global MicroRNA Expression in Human Alveolar Macrophages. PLoS ONE 2012, 7, e44066. [Google Scholar] [CrossRef]
  158. Gross, T.J.; Powers, L.S.; Boudreau, R.L.; Brink, B.; Reisetter, A.; Goel, K.; Gerke, A.K.; Hassan, I.H.; Monick, M.M. A Microrna Processing Defect in Smokers’ Macrophages Is Linked to Sumoylation of the Endonuclease DICER. J. Biol. Chem. 2014, 289, 12823–12834. [Google Scholar] [CrossRef]
  159. Christenson, S.A.; Brandsma, C.A.; Campbell, J.D.; Knight, D.A.; Pechkovsky, D.v.; Hogg, J.C.; Timens, W.; Postma, D.S.; Lenburg, M.; Spira, A. MiR-638 Regulates Gene Expression Networks Associated with Emphysematous Lung Destruction. Genome Med. 2013, 5, 114. [Google Scholar] [CrossRef]
  160. Zhu, Q.-Y.; Liu, Q.; Chen, J.-X.; Lan, K.; Ge, B.-X. MicroRNA-101 Targets MAPK Phosphatase-1 To Regulate the Activation of MAPKs in Macrophages. J. Immunol. 2010, 185, 7435–7442. [Google Scholar] [CrossRef]
  161. Mestdagh, P.; Vandesompele, J.; Brusselle, G.; Vermaelen, K. Non-Coding RNAs and Respiratory Disease. Thorax 2015, 70, 388–390. [Google Scholar] [CrossRef]
  162. Bartel, S.; la Grutta, S.; Cilluffo, G.; Perconti, G.; Bongiovanni, A.; Giallongo, A.; Behrends, J.; Kruppa, J.; Hermann, S.; Chiang, D.; et al. Human Airway Epithelial Extracellular Vesicle MiRNA Signature Is Altered upon Asthma Development. Allergy 2020, 75, 346–356. [Google Scholar] [CrossRef]
  163. Weidner, J.; Bartel, S.; Kılıç, A.; Zissler, U.M.; Renz, H.; Schwarze, J.; Schmidt-Weber, C.B.; Maes, T.; Rebane, A.; Krauss-Etschmann, S.; et al. Spotlight on MicroRNAs in Allergy and Asthma. Allergy 2021, 76, 1661–1678. [Google Scholar] [CrossRef]
  164. Wang, X.; Chen, H.; Liu, J.; Gai, L.; Yan, X.; Guo, Z.; Liu, F. Emerging Advances of Non-Coding RNAs and Competitive Endogenous RNA Regulatory Networks in Asthma. Bioengineered 2021, 12, 7820–7836. [Google Scholar] [CrossRef] [PubMed]
  165. Lu, T.X.; Munitz, A.; Rothenberg, M.E. MicroRNA-21 Is up-Regulated in Allergic Airway Inflammation and Regulates IL-12p35 Expression. J. Immunol. 2009, 182, 4994–5002. [Google Scholar] [CrossRef] [PubMed]
  166. Tian, M.; Zhou, Y.; Jia, H.; Zhu, X.; Cui, Y. The Clinical Significance of Changes in the Expression Levels of MicroRNA-1 and Inflammatory Factors in the Peripheral Blood of Children with Acute-Stage Asthma. Biomed. Res. Int. 2018, 2018, 7632487. [Google Scholar] [CrossRef]
  167. Tsai, M.J.; Tsai, Y.C.; Chang, W.A.; Lin, Y.S.; Tsai, P.H.; Sheu, C.C.; Kuo, P.L.; Hsu, Y.L. Deducting MicroRNA-Mediated Changes Common in Bronchial Epithelial Cells of Asthma and Chronic Obstructive Pulmonary Disease-A Next-Generation Sequencing-Guided Bioinformatic Approach. Int. J. Mol. Sci. 2019, 20, 553. [Google Scholar] [CrossRef]
  168. Qiu, Y.y.; Wu, Y.; Lin, M.j.; Bian, T.; Xiao, Y.l.; Qin, C. LncRNA-MEG3 Functions as a Competing Endogenous RNA to Regulate Treg/Th17 Balance in Patients with Asthma by Targeting MicroRNA-17/ RORγt. Biomed. Pharmacother. 2019, 111, 386–394. [Google Scholar] [CrossRef]
  169. Liang, Z.; Tang, F. The Potency of LncRNA MALAT1/MiR-155/CTLA4 Axis in Altering Th1/Th2 Balance of Asthma. Biosci. Rep. 2020, 40, BSR20190397. [Google Scholar] [CrossRef]
  170. Ma, L.; Zhang, Q.; Hao, J.; Wang, J.; Wang, C. LncRNA PVT1 Exacerbates the Inflammation and Cell-Barrier Injury during Asthma by Regulating MiR-149. J. Biochem. Mol. Toxicol. 2020, 34, e22563. [Google Scholar] [CrossRef] [PubMed]
  171. Cardiovascular Diseases. Available online: https://www.who.int/health-topics/cardiovascular-diseases#tab=tab_1 (accessed on 19 February 2023).
  172. Brook, R.D. Cardiovascular Effects of Air Pollution. Clin. Sci. 2008, 115, 175–187. [Google Scholar] [CrossRef]
  173. Mills, N.L.; Donaldson, K.; Hadoke, P.W.; Boon, N.A.; MacNee, W.; Cassee, F.R.; Sandström, T.; Blomberg, A.; Newby, D.E. Adverse Cardiovascular Effects of Air Pollution. Nat. Clin. Pract. Cardiovasc. Med. 2009, 61, 2008. [Google Scholar] [CrossRef]
  174. Martinelli, N.; Olivieri, O.; Girelli, D. Air Particulate Matter and Cardiovascular Disease: A Narrative Review. Eur. J. Intern. Med. 2013, 24, 295–302. [Google Scholar] [CrossRef]
  175. Meo, S.A.; Suraya, F. Effect of Environmental Air Pollution on Cardiovascular Diseases. Eur. Rev. Med. Pharmacol. Sci. 2015, 19, 4890–4897. [Google Scholar]
  176. Lederer, A.M.; Fredriksen, P.M.; Nkeh-Chungag, B.N.; Everson, F.; Strijdom, H.; de Boever, P.; Goswami, N. Cardiovascular Effects of Air Pollution: Current Evidence from Animal and Human Studies. Am. J. Physiol. Hearth Circ. Physiol. 2021, 320, H1417–H1439. [Google Scholar] [CrossRef]
  177. de Bont, J.; Jaganathan, S.; Dahlquist, M.; Persson, Å.; Stafoggia, M.; Ljungman, P. Ambient Air Pollution and Cardiovascular Diseases: An Umbrella Review of Systematic Reviews and Meta-Analyses. J. Intern. Med. 2022, 291, 779–800. [Google Scholar] [CrossRef] [PubMed]
  178. Perera, B.P.U.; Faulk, C.; Svoboda, L.K.; Goodrich, J.M.; Dolinoy, D.C. The Role of Environmental Exposures and the Epigenome in Health and Disease. Environ. Mol. Mutagen. 2020, 61, 176–192. [Google Scholar] [CrossRef]
  179. Micheu, M.M.; Birsan, M.V.; Szép, R.; Keresztesi, Á.; Nita, I.A. From Air Pollution to Cardiovascular Diseases: The Emerging Role of Epigenetics. Mol. Biol. Rep. 2020, 47, 5559–5567. [Google Scholar] [CrossRef]
  180. Tumolo, M.R.; Panico, A.; De Donno, A.; Mincarone, P.; Leo, C.G.; Guarino, R.; Bagordo, F.; Serio, F.; Idolo, A.; Grassi, T.; et al. The Expression of MicroRNAs and Exposure to Environmental Contaminants Related to Human Health: A Review. Int. J. Environ. Health Res. 2022, 32, 332–354. [Google Scholar] [CrossRef]
  181. Gangwar, R.S.; Rajagopalan, S.; Natarajan, R.; Deiuliis, J.A. Noncoding RNAs in Cardiovascular Disease: Pathological Relevance and Emerging Role as Biomarkers and Therapeutics. Am. J. Hypertens. 2018, 31, 150–165. [Google Scholar] [CrossRef]
  182. Sweta, S.; Dudnakova, T.; Sudheer, S.; Baker, A.H.; Bhushan, R. Importance of Long Non-Coding RNAs in the Development and Disease of Skeletal Muscle and Cardiovascular Lineages. Front. Cell Dev. Biol. 2019, 7, 228. [Google Scholar] [CrossRef]
  183. Toni, L.; Hailu, F.; Sucharov, C.C. Dysregulated Micro-RNAs and Long Noncoding RNAs in Cardiac Development and Pediatric Heart Failure. Am. J. Physiol. Hearth Circ. Physiol. 2020, 318, H1308–H1315. [Google Scholar] [CrossRef]
  184. Bollati, V.; Marinelli, B.; Apostoli, P.; Bonzini, M.; Nordio, F.; Hoxha, M.; Pegoraro, V.; Motta, V.; Tarantini, L.; Cantone, L.; et al. Exposure to Metal-Rich Particulate Matter Modifies the Expression of Candidate MicroRNAs in Peripheral Blood Leukocytes. Environ. Health Perspect. 2010, 118, 763–768. [Google Scholar] [CrossRef]
  185. Vriens, A.; Nawrot, T.S.; Saenen, N.D.; Provost, E.B.; Kicinski, M.; Lefebvre, W.; Vanpoucke, C.; Van Deun, J.; De Wever, O.; Vrijens, K.; et al. Recent Exposure to Ultrafine Particles in School Children Alters MIR-222 Expression in the Extracellular Fraction of Saliva. Environ. Health A Glob. Access Sci. Source 2016, 15, 80. [Google Scholar] [CrossRef]
  186. Sothivelr, V.; Hasan, M.Y.; Saffian, S.M.; Zainalabidin, S.; Ugusman, A.; Mahadi, M.K. Revisiting MiRNA-21 as a Therapeutic Strategy for Myocardial Infarction: A Systematic Review. J. Cardiovasc. Pharmacol. 2022, 80, 393–406. [Google Scholar] [CrossRef]
  187. Cheng, Y.; Zhang, C. MicroRNA-21 in Cardiovascular Disease. J. Cardiovasc. Transl. Res. 2010, 3, 251–255. [Google Scholar] [CrossRef]
  188. Ding, S.; Huang, H.; Xu, Y.; Zhu, H.; Zhong, C. MiR-222 in Cardiovascular Diseases: Physiology and Pathology. Biomed Res. Int. 2017, 2017, 4962426. [Google Scholar] [CrossRef]
  189. Fossati, S.; Baccarelli, A.; Zanobetti, A.; Hoxha, M.; Vokonas, P.S.; Wright, R.O.; Schwartz, J. Ambient Particulate Air Pollution and MicroRNAs in Elderly Men. Epidemiology 2014, 25, 68–78. [Google Scholar] [CrossRef]
  190. Chen, R.; Li, H.; Cai, J.; Wang, C.; Lin, Z.; Liu, C.; Niu, Y.; Zhao, Z.; Li, W.; Kan, H. Fine Particulate Air Pollution and the Expression of MicroRNAs and Circulating Cytokines Relevant to Inflammation, Coagulation, and Vasoconstriction. Environ. Health Perspect. 2018, 126, 017007. [Google Scholar] [CrossRef]
  191. Motta, V.; Angelici, L.; Nordio, F.; Bollati, V.; Fossati, S.; Frascati, F.; Tinaglia, V.; Bertazzi, P.A.; Battaglia, C.; Baccarelli, A.A. Integrative Analysis of Mirna and Inflammatory Gene Expression after Acute Particulate Matter Exposure. Toxicol. Sci. 2013, 132, 307–316. [Google Scholar] [CrossRef]
  192. Pergoli, L.; Cantone, L.; Favero, C.; Angelici, L.; Iodice, S.; Pinatel, E.; Hoxha, M.; Dioni, L.; Letizia, M.; Albetti, B.; et al. Extracellular Vesicle-Packaged MiRNA Release after Short-Term Exposure to Particulate Matter Is Associated with Increased Coagulation. Part. Fibre Toxicol. 2017, 14, 32. [Google Scholar] [CrossRef]
  193. Rodosthenous, R.S.; Coull, B.A.; Lu, Q.; Vokonas, P.S.; Schwartz, J.D.; Baccarelli, A.A. Ambient Particulate Matter and MicroRNAs in Extracellular Vesicles: A Pilot Study of Older Individuals. Part. Fibre Toxicol. 2016, 13, 13. [Google Scholar] [CrossRef]
  194. Rodosthenous, R.S.; Kloog, I.; Colicino, E.; Zhong, J.; Herrera, L.A.; Vokonas, P.; Schwartz, J.; Baccarelli, A.A.; Prada, D. Extracellular Vesicle-Enriched MicroRNAs Interact in the Association between Long-Term Particulate Matter and Blood Pressure in Elderly Men. Environ. Res. 2018, 167, 640–649. [Google Scholar] [CrossRef]
  195. Chen, H.; Zhang, S.; Yu, B.; Xu, Y.; Rappold, A.G.; Diaz-Sanchez, D.; Samet, J.M.; Tong, H. Circulating MicroRNAs as Putative Mediators in the Association between Short-Term Exposure to Ambient Air Pollution and Cardiovascular Biomarkers. Ecotoxicol. Environ. Saf. 2022, 239, 113604. [Google Scholar] [CrossRef]
  196. Wang, H.; Wang, T.; Rui, W.; Xie, J.; Xie, Y.; Zhang, X.; Guan, L.; Li, G.; Lei, Z.; Schiffelers, R.M.; et al. Extracellular Vesicles Enclosed-MiR-421 Suppresses Air Pollution (PM2.5)-Induced Cardiac Dysfunction via ACE2 Signalling. J. Extracell. Vesicles 2022, 11, e12222. [Google Scholar] [CrossRef]
  197. Motta, V.; Favero, C.; Dioni, L.; Iodice, S.; Battaglia, C.; Angelici, L.; Vigna, L.; Pesatori, A.C.; Bollati, V. MicroRNAs Are Associated with Blood-Pressure Effects of Exposure to Particulate Matter: Results from a Mediated Moderation Analysis. Environ. Res. 2016, 146, 274–281. [Google Scholar] [CrossRef]
  198. Dugger, B.N.; Dickson, D.W. Pathology of Neurodegenerative Diseases. Cold Spring Harb. Perspect. Biol. 2017, 9, a028035. [Google Scholar] [CrossRef] [PubMed]
  199. Costa, L.G.; Cole, T.B.; Dao, K.; Chang, Y.C.; Coburn, J.; Garrick, J.M. Effects of Air Pollution on the Nervous System and Its Possible Role in Neurodevelopmental and Neurodegenerative Disorders. Pharmacol. Ther. 2020, 210, 107523. [Google Scholar] [CrossRef] [PubMed]
  200. Cacciottolo, M.; Wang, X.; Driscoll, I.; Woodward, N.; Saffari, A.; Reyes, J.; Serre, M.L.; Vizuete, W.; Sioutas, C.; Morgan, T.E.; et al. Particulate Air Pollutants, APOE Alleles and Their Contributions to Cognitive Impairment in Older Women and to Amyloidogenesis in Experimental Models. Transl. Psychiatry 2017, 7, e1022. [Google Scholar] [CrossRef]
  201. Chen, X.; Guo, J.; Huang, Y.; Liu, S.; Huang, Y.; Zhang, Z.; Zhang, F.; Lu, Z.; Li, F.; Zheng, J.C.; et al. Urban Airborne PM2.5-Activated Microglia Mediate Neurotoxicity through Glutaminase-Containing Extracellular Vesicles in Olfactory Bulb. Environ. Pollut. 2020, 264, 114716. [Google Scholar] [CrossRef]
  202. He, S.; Huang, L.; Shao, C.; Nie, T.; Xia, L.; Cui, B.; Lu, F.; Zhu, L.; Chen, B.; Yang, Q. Several MiRNAs Derived from Serum Extracellular Vesicles Are Potential Biomarkers for Early Diagnosis and Progression of Parkinson’s Disease. Transl. Neurodegener 2021, 10, 25. [Google Scholar] [CrossRef]
  203. Upadhya, R.; Shetty, A.K. Extracellular Vesicles for the Diagnosis and Treatment of Parkinson’s Disease. Aging Dis. 2021, 12, 1438–1450. [Google Scholar] [CrossRef]
  204. Caldi Gomes, L.; Roser, A.E.; Jain, G.; Pena Centeno, T.; Maass, F.; Schilde, L.; May, C.; Schneider, A.; Bähr, M.; Marcus, K.; et al. MicroRNAs from Extracellular Vesicles as a Signature for Parkinson’s Disease. Clin. Transl. Med. 2021, 11, e357. [Google Scholar] [CrossRef]
  205. Schikowski, T.; Vossoughi, M.; Vierkötter, A.; Schulte, T.; Teichert, T.; Sugiri, D.; Fehsel, K.; Tzivian, L.; Bae, I.S.; Ranft, U.; et al. Association of Air Pollution with Cognitive Functions and Its Modification by APOE Gene Variants in Elderly Women. Environ. Res. 2015, 142, 10–16. [Google Scholar] [CrossRef]
  206. Carey, I.M.; Anderson, H.R.; Atkinson, R.W.; Beevers, S.D.; Cook, D.G.; Strachan, D.P.; Dajnak, D.; Gulliver, J.; Kelly, F.J. Are Noise and Air Pollution Related to the Incidence of Dementia? A Cohort Study in London, England. BMJ Open 2018, 8, e022404. [Google Scholar] [CrossRef] [PubMed]
  207. Dong, X.; Zheng, D.; Nao, J. Circulating Exosome MicroRNAs as Diagnostic Biomarkers of Dementia. Front. Aging Neurosci. 2020, 12, 290. [Google Scholar] [CrossRef]
  208. Gámez-Valero, A.; Campdelacreu, J.; Vilas, D.; Ispierto, L.; Reñé, R.; Álvarez, R.; Armengol, M.P.; Borràs, F.E.; Beyer, K. Exploratory Study on MicroRNA Profiles from Plasma-Derived Extracellular Vesicles in Alzheimer’s Disease and Dementia with Lewy Bodies. Transl. Neurodegener. 2019, 8, 31. [Google Scholar] [CrossRef]
  209. Wang, L.; Zhang, L. Circulating Exosomal MiRNA as Diagnostic Biomarkers of Neurodegenerative Diseases. Front. Mol. Neurosci. 2020, 13, 53. [Google Scholar] [CrossRef]
  210. Li, C.Y.; Li, C.H.; Martini, S.; Hou, W.H. Association between Air Pollution and Risk of Vascular Dementia: A Multipollutant Analysis in Taiwan. Environ. Int. 2019, 133, 105233. [Google Scholar] [CrossRef]
  211. Calderón-Garcidueñas, L.; Hernández-Luna, J.; Mukherjee, P.S.; Styner, M.; Chávez-Franco, D.A.; Luévano-Castro, S.C.; Crespo-Cortés, C.N.; Stommel, E.W.; Torres-Jardón, R. Hemispheric Cortical, Cerebellar and Caudate Atrophy Associated to Cognitive Impairment in Metropolitan Mexico City Young Adults Exposed to Fine Particulate Matter Air Pollution. Toxics 2022, 10, 156. [Google Scholar] [CrossRef]
  212. Fuller, R.; Landrigan, P.J.; Balakrishnan, K.; Bathan, G.; Bose-O’Reilly, S.; Brauer, M.; Caravanos, J.; Chiles, T.; Cohen, A.; Corra, L.; et al. Pollution and health: A progress update. Lancet Planet Health 2022, 6, e535–e547. [Google Scholar] [CrossRef]
  213. Caruso Bavisotto, C.; Marino Gammazza, A.; Rappa, F.; Fucarino, A.; Pitruzzella, A.; David, S.; Campanella, C. Exosomes: Can doctors still ignore their existence? EuroMediterr. Biomed. J. 2013, 8, 7–9. [Google Scholar] [CrossRef]
  214. Campanella, C.; D’Anneo, A.; Marino Gammazza, A.; Caruso Bavisotto, C.; Barone, R.; Emanuele, S.; Lo Cascio, F.; Mocciaro, E.; Fais, S.; Conway de Macario, E.; et al. The histone deacetylase inhibitor SAHA induces HSP60 nitration and its extracellular release by exosomal vesicles in human lung-derived carcinoma cells. Oncotarget 2016, 7, 28849–28867. [Google Scholar] [CrossRef] [PubMed]
  215. Cappello, F.; Logozzi, M.; Campanella, C.; Caruso Bavisotto, C.; Marcilla, A.; Properzi, F.; Fais, S. Exosome levels in human body fluids: A tumor marker by themselves? Eur. J. Pharm. Sci. 2017, 96, 93–98. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Schematic representation of the benefits of ncRNA in liquid biopsy in the study of diseases associated with air pollutant exposure. ncRNAs can be found in blood-derived EVs and represent reliable prognostic and predictive markers, useful in screening, diagnosis, and EV-based personalized therapy.
Figure 1. Schematic representation of the benefits of ncRNA in liquid biopsy in the study of diseases associated with air pollutant exposure. ncRNAs can be found in blood-derived EVs and represent reliable prognostic and predictive markers, useful in screening, diagnosis, and EV-based personalized therapy.
Cells 12 01498 g001
Table 1. Classification of ncRNAs.
Table 1. Classification of ncRNAs.
TypesSizeFunction
Housekeeping RNAsrRNAs120–5000 ntsProteins synthesis
tRNAs70–100 ntsProteins synthesis
snRNAs~150 ntsSplicing of mRNAs
snoRNAs60–300 ntsModification of rRNAs
TERC400–600 ntsTemplate for telomere replication
Regulatory RNAssiRNAs20–25 ntsRegulation of gene expression at the post-transcriptional level
miRNAs21–23 ntsRegulation of gene expression at the post-transcriptional level
piRNAs26–31 ntsRegulation of gene expression at epigenetic, transcriptional, and post-transcriptional level
lncRNAs>200 ntsRegulation of gene expression at epigenetic, transcriptional, and transcriptional level
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

D’Amico, G.; Santonocito, R.; Vitale, A.M.; Scalia, F.; Marino Gammazza, A.; Campanella, C.; Bucchieri, F.; Cappello, F.; Caruso Bavisotto, C. Air Pollution: Role of Extracellular Vesicles-Derived Non-Coding RNAs in Environmental Stress Response. Cells 2023, 12, 1498. https://doi.org/10.3390/cells12111498

AMA Style

D’Amico G, Santonocito R, Vitale AM, Scalia F, Marino Gammazza A, Campanella C, Bucchieri F, Cappello F, Caruso Bavisotto C. Air Pollution: Role of Extracellular Vesicles-Derived Non-Coding RNAs in Environmental Stress Response. Cells. 2023; 12(11):1498. https://doi.org/10.3390/cells12111498

Chicago/Turabian Style

D’Amico, Giuseppa, Radha Santonocito, Alessandra Maria Vitale, Federica Scalia, Antonella Marino Gammazza, Claudia Campanella, Fabio Bucchieri, Francesco Cappello, and Celeste Caruso Bavisotto. 2023. "Air Pollution: Role of Extracellular Vesicles-Derived Non-Coding RNAs in Environmental Stress Response" Cells 12, no. 11: 1498. https://doi.org/10.3390/cells12111498

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop