Next Article in Journal
Automatic Classification for Sagittal Craniofacial Patterns Based on Different Convolutional Neural Networks
Next Article in Special Issue
The Expression Levels and Concentrations of PD-1 and PD-L1 Proteins in Septic Patients: A Systematic Review
Previous Article in Journal
Diagnostic Performance of 99mTc-Methoxy-Isobuty-Isonitrile (MIBI) for Risk Stratification of Hypofunctioning Thyroid Nodules: A European Multicenter Study
Previous Article in Special Issue
Weaning Failure in Critically Ill Patients Is Related to the Persistence of Sepsis Inflammation
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Non-Coding RNA Networks as Potential Novel Biomarker and Therapeutic Target for Sepsis and Sepsis-Related Multi-Organ Failure

Department of Promoting Health, Maternal-Infant, Excellence and Internal and Specialized Medicine (Promise) “G. D’Alessandro”, University of Palermo, Piazza delle Cliniche, 90100 Palermo, Italy
*
Author to whom correspondence should be addressed.
These authors contributed equally to this paper.
Diagnostics 2022, 12(6), 1355; https://doi.org/10.3390/diagnostics12061355
Submission received: 5 April 2022 / Revised: 27 May 2022 / Accepted: 28 May 2022 / Published: 31 May 2022
(This article belongs to the Special Issue Biomarkers of Sepsis)

Abstract

:
According to “Sepsis-3” consensus, sepsis is a life-threatening clinical syndrome caused by a dysregulated inflammatory host response to infection. A rapid identification of sepsis is mandatory, as the extent of the organ damage triggered by both the pathogen itself and the host’s immune response could abruptly evolve to multiple organ failure and ultimately lead to the death of the patient. The most commonly used therapeutic strategy is to provide hemodynamic and global support to the patient and to rapidly initiate broad-spectrum empiric antibiotic therapy. To date, there is no gold standard diagnostic test that can ascertain the diagnosis of sepsis. Therefore, once sepsis is suspected, the presence of organ dysfunction can be assessed using the Sepsis-related Organ Failure Assessment (SOFA) score, although the diagnosis continues to depend primarily on clinical judgment. Clinicians can now rely on several serum biomarkers for the diagnosis of sepsis (e.g., procalcitonin), and promising new biomarkers have been evaluated, e.g., presepsin and adrenomedullin, although their clinical relevance in the hospital setting is still under discussion. Non-codingRNA, including long non-codingRNAs (lncRNAs), circularRNAs (circRNAs) and microRNAs (miRNAs), take part in a complex chain of events playing a pivotal role in several important regulatory processes in humans. In this narrative review we summarize and then analyze the function of circRNAs-miRNA-mRNA networks as putative novel biomarkers and therapeutic targets for sepsis, focusing only on data collected in clinical settings in humans.

1. Introduction

Sepsis is a life-threatening clinical syndrome caused by a dysregulated host inflammatory response to infection, often associated with multiple organ dysfunction syndrome and death [1]. It is recognized as a leading cause of death worldwide, as highlighted by the Global Burden of Disease, which estimated 48.9 million incident cases in 2017, accounting for 19.7% of all global deaths [2]. The economic and health burden of sepsis worldwide is alarming; mortality in sepsis patients has been estimated to be ≥10%, rising above 40% when evolving to septic shock [1]; in 2011 the total sepsis-related costs for US hospitals accounted for more than US $20 billion [1].
The definition of sepsis has undergone several revisions over the years because of the highly variable clinical spectrum: the 2001 American College of Chest Physicians/Society of Critical Care Medicine (ACCP/SCCM) Consensus Conference Committee criteria for the host systemic inflammatory response syndrome (SIRS) are currently outdated because of their demonstrated poor ability to discriminate between different degrees of clinical severity [3]. Reassessment of these criteria in a clinical setting has shown that they are often found in many inpatients, including those who are noninfectious and who do not proceed to adverse outcomes [3]. The latest definition of sepsis, named “Sepsis-3”, was proposed in 2016 by the SCCM and the European Society of Intensive Care Medicine (ESICM) [1]. According to the SCCM/ESICM, sepsis is defined as a life-threatening organ dysfunction (ascertained as acute change in Sepsis-related Organ Failure Assessment (SOFA score) total score ≥2) “due to a dysregulated host response to infection” [1]. Septic shock is defined as a “subset of sepsis in which particularly profound circulatory, cellular, and metabolic abnormalities substantially increase mortality” [1].
Rapid detection of sepsis is mandatory since the patient’s overall clinical impairment and degree of organ damage are triggered by an extremely complex chain of events involving the recognition of pathogen-associated molecular patterns (PAMPs) of microorganisms by the host immune system [4]. As happens in other acute pathologic conditions such as acute encephalitis [5], but also in several diseases not directly provoked by an infection such as ischemic stroke [6,7,8], atrial fibrillation [9], and many others [10], the characteristics of the interaction between host and pathogen fundamentally affect the degree and severity of the systemic involvement of the patient. The damage-associated molecular patterns (DAMPs) released by the spillover from the injured cells [11] can result in an escalating state of inflammation that can abruptly lead to multiple organ failure (MOF) and can result in death. The prompt initiation of a broad spectrum empirical antibiotic therapy and patient-driven supportive strategies such as fluid resuscitation optimize outcomes. The so-called early goal-directed therapy in the first hour of documented hypotension leads to a 79.9% survival rate, each hour of delay being associated with an average decrease in survival of 7.6% [12]. Despite the development of bedside screening tools to facilitate the early detection of septic patients, a tool to which a definitive diagnostic value can be attributed is still missing, thus the diagnosis is today still challenging, and it continues to depend on the clinical judgment based on nonspecific clinical and laboratory variables. In addition, rapid discrimination between infectious and noninfectious causes presents a daunting challenge. The diagnosis of systemic infection is mainly based on direct microbiological tests such as cultures or polymerase chain reaction-based methods or indirectly using specific immunoglobulin dosage. Unfortunately, microbiology results often take several days to became positive and are not diagnostic in patients with ongoing infection in up to one-third of cases, especially if cultures were collected when antibiotic treatment had already been started [13].
Since the combined sensitivity and specificity of actual biomarkers (e.g., C-reactive protein (CPR), Procalcitonin (PCT) and Interleukin-6 (IL-6)) do not allow for the rapid ascertainment of the diagnosis [14,15] and sepsis-related adverse outcomes rise with every hour of delay of proper intervention, new early biomarkers are urgently needed.
There is a growing amount of data about non-codingRNA, a group of transcripts that do not code proteins at first deemed as redundant RNAs but lately described as highly conserved transcripts involved in gene expression regulation through the modulation of chromatin rearrangement, histone modification, alternative splicing regulation and many other biological processes [16]. Recent findings speculate that circularRNAs (circRNAs), a particular type of long non-codingRNAs (lncRNAs) distinguished by a covalently closed-loop structure with neither 5′ to 3′ polarity nor polyadenosine tail, participates in gene regulation in a different way, regulating the microRNA (miRNAs) concentration in body fluids by competing with several miRNAs and regulating the downstream of messenger RNAs (mRNAs) [17].
Further demonstrating the increasing biological value that non-coding RNAs are proving to have, they seem to play a role in the pathogenesis of different diseases [18], and, given the complex interweaving between circRNAs, miRNAs, lncRNAs and mRNAs, various studies have addressed the issue of their role as novel diagnostic markers and therapeutic targets in many pathologic conditions including sepsis [19,20,21,22,23,24,25].
The objective of this review is to summarize the current findings of circRNA-miRNA-lncRNA networks in the context of sepsis as a biomarker and therapeutic target with a focus on their clinical use in a hospital setting and their effectiveness in providing reliable data for the improvement of clinical practice in the adequacy of early diagnosis and treatment of sepsis and septic shock.

2. Methodology of Literature Search

A comprehensive literature search was carried out in the MEDLINE database (search terms: “sepsis” + “noncodingRNA”, “miRNA”, “circRNA”, “mRNA”, “lncRNA”, “RNA”, “network”, “biomarker”, “therapy”, “prognosis”, “organ failure”). The search has been restricted to papers published in English without time limit. The authors sought literature by examining reference lists in original articles and reviews. We have included in this review only systematic reviews, metanalyses, randomized trials and randomized controlled trials, selecting studies in which the main objective of the study was the identification and function of circRNA, miRNA, or mRNA networks in the context of novel biomarkers with remarkable prognostic value and/or therapeutic target for sepsis and sepsis-related organ failure.
Each author involved independently evaluated the results of the literature research extracting the most pertinent knowledge, while others verified the accuracy and completeness of the extracted data. Each author made a judgement as to whether the search results were different or confounding, trying to provide as complete an overview of the field as possible to date.

3. Role of Biomarkers in Sepsis

According to the Biomarkers Definitions Working Group, a biological marker or biomarker is “a characteristic that is objectively measured and evaluated as an indicator of normal biological processes, pathogenic processes or pharmacologic responses to a therapeutic intervention” [26].
A biomarker finds application across four domains or functional classes:
  • As a diagnostic tool, i.e., a biomarker able to confirm a disease;
  • As a tool able to stage or to stratify disease severity;
  • As a prognostic tool;
  • An effective tool for prediction and monitoring of clinical response of an intervention [26].
Biomarkers of sepsis hold the promise of closing the gap in obtaining mycobacterial cultures by providing clinicians with clinically useful data. There is a strong demand for new and accurate sepsis biomarkers, especially in the era of personalized medicine in which physicians must increasingly tailor clinical and therapeutic management to each patient. In the following part of our manuscript, without aiming to address this topic comprehensively, the role of some of the major biomarkers currently used in sepsis will be analyzed by discussing the merits and demerits of their use during the management and treatment of such a serious disease that has its cornerstone in the timeliness of identification and early and appropriate intervention.

3.1. C-Reactive Protein (CRP)

C-Reactive Protein (CRP) is a plasma protein belonging to the group of the so-called acute phase reactants which may increase rapidly during inflammatory conditions or secondary to non-specific acute inflammatory stimuli [26]. The acute-phase proteins are produced in the liver during inflammatory states under the control of cytokines: CRP is mainly synthesized through Interleukin-6 (IL-6) and Interleukin-1β (IL-1β) stimulation via the transcription factors STAT3 and NF-κB [27]. CRP, as a component of the innate immune system, during infection may recognize various pathogens associated molecular patterns (PAMPs) such as phospholipid fragments released from damaged cells consequently activating the complement system and finally inducing the death of the targeted cells [28]. CRP is released into the bloodstream after 4–6 h after an inflammatory stimulus and a plasma peak is reached in 36 to 50 h [14,15,16,17,18,19,20,21,22,23,24,25,26,27,28,29]. Several conditions besides infection can result in the elevation of the CRP-serum level [30]; a meta-analysis of Simon et al. [31] demonstrated a sensitivity of 75% [95% CI: 62–84%] and a specificity of 67% [95% CI: 56–77%] for CRP in differentiating bacterial infection from the noninfective cause of inflammation.
Liu et al. [32], in a systematic review and meta-analysis including 45 studies and 5654 patients, showed an acceptable level of sensitivity of 75% (95% CI: 69–79%) but a weak level of specificity of 67% (95% CI: 58–74%) for the ability of CRP to differentiate patients with sepsis vs. non-infectious inflammatory state/disorders. Tan et al. [33], comparing the ability of CRP and PCT to serve as biomarkers for sepsis diagnosis show similar sensitivity (CRP: 80%, 95% CI: 63–90%, procalcitonin: 80%, 95% CI: 69–87%) but significantly lower specificity for CRP (61%; 95% CI: 50–72%) than procalcitonin (77%; 95% CI: 60–88%) [33]. A possible explanation for the lower diagnostic accuracy of CRP as a sepsis biomarker (low specificity and moderate sensibility) could account for the slow-release kinetics as a consequence of the inflammatory stimulus and its increase also due to other pathological conditions besides infections (e.g., trauma, burns, surgery or various immune-inflammatory conditions [34,35].
Finally, the limits showed that CRP remains a widely used diagnostic and therapeutic biomarker in sepsis to date, mainly because a decrease in its values correlates with the success of antimicrobial treatment [36].

3.2. Procalcitonin (PCT)

Procalcitonin is the precursor of calcitonin, released by the C-cells of parathyroid glands. Assicot et al. [37] in 1993 for the first time described the association between PCT serum levels and severe bacterial infection. Compared to CRP, the PCT has a better kinetic profile, increasing within 3–6 h after the onset of infection reaching its serum peak after 6–8 h [29]. Several studies investigated the diagnostic performance of PCT. A meta-analysis of Uzzan et al. [35], including studies from 1996 to 2004, showed a higher accuracy of PCT levels than CRP levels for the diagnosis of sepsis (Global diagnostic accuracy odds ratios: CRP 5.43 [95% CI: 3.19–9.23] vs. PCT 14.69 [95% CI 7.12–30.27] [35]. However, the authors included a restricted cohort study based only on surgery or trauma patients, and thus the conclusion cannot be extended to patients other than surgical conditions [35].
Tang et al. [38], in a meta-analysis of 18 studies, pointed out that PCT was not adequate in discriminating between sepsis and SIRS (both sensitivity and specificity were 71% [95% CI: 67–76] and the Area Under the Summary Receiver Operator Characteristic Curve was 0.78 [95% CI: 0.73–0.83] [38]. Another meta-analysis of 30 observational studies evaluating 3244 mixed subjects (pediatric and adult patients admitted in the Intensive Care Unit or Emergency Room), has given the PCT a sensitivity of 77% [95% CI: 72–81%] and a specificity of 79% [95% CI: 74–84%], with AUC 0.85 [95% CI 0.81–0.88] for accuracy in discriminating sepsis from a non-infectious state [39].
Several studies have also confirmed the clinical utility of PCT in driving antimicrobial therapy surveillance and the eventual de-escalation of antibiotic treatment [14,15,16,17,18,19,20,21,22,23,24,25,26,27,28,29].
To date, there are no established cut-off values of serum PCT concentrations that are able to discriminate sepsis versus septic shock [29].

3.3. Presepsin

Presepsin, the N-terminal fragment of 13 kDa of the sCD14 (the soluble form of the receptor of lipopolysaccharide-lipopolysaccharide binding protein), is an emerging biomarker and early indicator of bacterial infections [40]. Presepsin, as part of the Toll-like receptor group, takes part of the innate immune system, binding several pathogen-associated molecular patterns (PAMPs) such as lipopolysaccharide (LPS) of Gram- or peptidogligans [40]. In recent years, sCD14 has become one of the most widely sepsis biomarkers studied: the level of sCD14 increased significantly in patients with sepsis and septic shock compared with healthy people, and the change was significantly related to the severity and prognosis of the disease [29,41,42,43,44]. The diagnostic power of presepsin in detecting sepsis showed with a pooled sensitivity of 77–86% and a specificity of 73–78% [41,42,43,44]. Nevertheless, presepsin still needs wider investigation and further validation and comparison with standard sepsis biomarkers prior to being recommended for the hospital-setting.

4. Non-CodingRNA

Non-codingRNA, including long non-codingRNAs (lncRNAs), circularRNAs (circRNAs) and microRNAs (miRNAs), take part in a complex chain of events playing a pivotal role in several important regulatory processes in humans. Non-codingRNAs are traditionally classified based on the length of nucleotides (nt) in small non-codingRNAs (sncRNAs) and long non-codingRNAs. The ncRNAs with 200 nt or lower are referred to as sncRNAs including microRNAs (miRNAs), small interfering RNAs (siRNAs) and piwi-interactingRNAs (piRNAs) whereas those with 200 nt or higher are referred as lncRNAs, such as promoter-associated transcripts (PATs), enhancerRNAs (eRNAs) and circularRNAs (circRNAs).
Numerous studies have demonstrated that exosomes, phospholipid bilayer vesicles that originate from the membrane vesicles of the endosomes and are secreted by almost all cells, contain a variety of functional molecules that are crucial mediators of intercellular communication including lncRNAs, circRNAs and miRNAs. After exosomes are released into the tissue fluid, they arrive at the target cells and begin to deliver the different molecules contained (such as circRNAs), thus initiating functional responses and inducing subsequent phenotypic changes [45,46,47]. Recent studies have shown altered expressions of several non-coding RNAs such as lncRNAs, circRNAs and miRNAs during sepsis. Interestingly, noncoding RNAs have also been found to participate in the pathogenesis of multiple organ system failure through different mechanisms. In the following section of the review, the role of these three classes of noncoding RNAs in the pathophysiology of sepsis and sepsis-related multi-organ failure (MOF) will be examined.

4.1. CircRNAs

CircRNAs are an endogenous non-codingRNA which have as their main characteristic a closed loop structure with a covalent bond linking the 3′ to 5′ ends [48,49].
CircRNAs are highly conserved molecules expressed in a broad range of human cells, both in physiologic and pathologic conditions [50]. Memczak et al. detected 1950 circRNAs in HEK293 cells, 1903 circRNAs in the brain and fetal cells of mouse and through 724 circRNAs from different developing stages of Caenorhabditis Elegans (a nematode worm about 1 mm in length) [48].
Enuka et al. [51] estimate that circRNAs have a half-life of 18.8–23.7 h, roughly 2.5-fold higher that their linear form [52]. Also, circRNAs are insensitive to the common degradation pathways (i.e., RNAase or RNAexonucleases) [53]. Mature circRNAs are usually found in cytoplasms, whereas immature circRNAs that are still susceptible to intronic splicing remain in the nucleus [54].

4.1.1. CircRNAs Biogenesis

CircRNAs are primarily generated by the transcription of exonic and/or intronic sequences of a protein-coding gene in a complex reaction catalyzed by the RNA polymerase II (RNApol II). Both circRNAs and mRNAs, in their linear forms, are produced by the same precursor or pre-mRNAs undergoing a totally different splicing mechanism [55]. CircRNAs are produced by spliceosome machinery through a back-splicing process, resulting in a covalently closed loop structure between the 3′ upstream splice site and the 5′ downstream splice site [56,57,58,59,60,61].

4.1.2. CircRNAs Functions

CircRNAs act by separating miRNAs from their target mRNAs, thereby influencing miRNA-mediated gene suppression or expression. The interaction between circRNA-miRNA appears to be critical to the optimal functionality of our organism [62,63]. CircRNAs may act as a dynamic scaffold influencing protein interactions having the potential to regulate protein function by binding, storing, sequencing and isolating proteins to specific subcellular locations [64]. Finally, nuclear circRNAs may act as regulators of the transcription by promoting the extended activity of RNA polymerase II [46,65] or through other alternative pathways which are still being studied.
Considering all of these features, the tissue development specific expression patterns and the putative crucial regulatory functions in various diseases, circRNAs thus have all of the qualities to be used as novel biomarkers in several pathologic conditions [19,21,66,67].

4.2. Long Non-CodingRNAs (lncRNAs)

The lncRNAs (200 nt or more) belong to the large class of non-codingRNAs that perform housekeeping functions in numerous biological processes through the regulation of gene expression at the post-transcriptional and transcriptional level [68,69,70]. Changes in the expression levels of lncRNAs affect the malignancy phenotype in various types of cancer such as colorectal, lung, liver, breast, ovarian cancers and leukemia [71,72], and are linked to development state [73] and may be observed during various phases of T-cell differentiation [74].

4.2.1. lncRNAs Biogenesis

To date, the lncRNAs biogenesis is not completely clarified. Transcriptome-wide studies show that lncRNAs biogenesis owns a peculiar expression pattern that is cell type-specific and stage-specific [75,76], which is also regulated by cell type and stage-specific stimuli [77] Briefly, lncRNAs are widely interspersed in the genome. Enhancers, promoters, and intragenic regions are the main DNA elements from which lncRNAs are transcribed [78].

4.2.2. lncRNAs Functions

The molecular function of lncRNAs may be summarized as four archetypes: (1) signal: lncRNAs can serve as molecular signal able to activate/silence specific genome sequences by interacting with chromatin and recruiting the chromatin modifying systems [76]; (2) Decoy: lncRNAs play a central role in the regulation of genome transcription, mainly by inhibiting it but occasionally also by activating it [76,77]; (3) Guide: lncRNAs can modify the chromatin structure guiding specific proteins to specific targets that ultimately cause gene silencing [76,77]; (4) Scaffold: lncRNAs can serve as a scaffold for assembling two or more proteins by inducing changes in chromatin as a consequence. They may therefore play a role in the activation or transcriptional silencing of specific genome sequences [76,77].

4.3. MicroRNAs (miRNAs)

miRNAs are endogenous non-coding transcripts of 19–22 nucleotides that modulate the translation of target mRNAs at a post-transcriptional level [79].

4.3.1. miRNAs Biogenesis

The RNA polymerase II transcribes a primary miRNA called pri-miRNA (~500–3000 nt); starting with this precursor a premature miRNA (pre-miRNA) is formed [80,81]. Through exportin 5, the pre-miRNA is exported in the cytoplasm and an miRNA duplex is processed. The “miRNA duplex” is bound into the RISC (RNA-induced silencing complex) with the final release of the mature miRNA [80]. As previously mentioned, circRNAs sequester mature miRNAs, acting as an miRNA sponge, through the interaction between the RNA binding proteins, diminishing miRNA functions.

4.3.2. miRNAs Functions

miRNAs are believed to be among the most important regulators of cellular communication, playing a pivotal role in maneuvering the linear RNA’s expression at various levels. The most notably and predominant function of miRNAs is the binding of complementary sequences of the target linear RNAs that results in the inhibition of mRNA translation [82].

5. Non-Coding RNA and Sepsis

During sepsis the immune system is widely activated; the level and extent of the immune response triggered by the pathogen is different from subject to subject and depends largely on his/her state of immunocompetence [83,84].
Studies performed mostly in experimental animal models investigating the role of non-codingRNAs in the modulation of inflammation have identified some interesting networks that have been shown to be actively involved. Accumulating evidence shows that lncRNAs and miRNAs are involved in the sepsis inflammatory response, but the role played by different non-coding RNA networks in different biological contexts is extremely complex, variable, and involves the intervention of several mediators and effectors. An example will make clear the complexity of the field of research we are discussing: lncRNA taurine upregulated gene 1 (lncRNA TUG1) seems to participate in several pathophysiological processes. The overexpression of TUG1 has demonstrated the ability to alleviate the inflammatory response (including apoptosis and autophagy) in acute lung injury [85]. These relevant clinical effects seem to be mediated via targeting miR-34b-5p [86] and miR-27a-3p [87]. On the other hand, TUG1 silencing reduces the inflammation and apoptosis of renal tubular cells in an ischemia-reperfusion model via targeting of miR-449B-5p [88], and regulates the expression of various genes such as matrix metalloproteinase [89], protects against myocardial ischemia upregulating miR 142-3p [90] and downregulating miR-29a-3p [91] through the modulation of the miR-532-5p/Sox8 axis [92] and the miR-145-5p-Binp3 axis [93,94]. These are just a few of the many and complex interactions highlighted to date.
Despite the mentioned challenge of accurately defining the roles played by specific non-coding RNAs, some research has helped us identify some molecules of likely clinical interest. LncRNA metastasis-associated lung adenocarcinoma transcript 1 (MALAT1) has been linked to sepsis. MALAT1 downregulation inhibits the LPS-induced inflammatory response by preventing the release of IL-6 and TNF-α and the NF-κB signaling pathway by upregulating miR-150-5p [95]. The MALAT1/miR-214/Toll-like receptor (TLR)5 signaling pathway dysregulation enhances the risk of post-burn sepsis by promoting greater production of proinflammatory cytokines [96]. LncRNA nuclear-enriched abundant transcript 1 (NEAT1) seems to also be involved in sepsis progression. In rats, NEAT1 knockdown could significantly improve the sepsis-induced myocardial injury preventing cardiac insufficiency and consensually increasing the ejection fraction (p < 0.05) [97]. Furthermore, another study indicates that NEAT1 inhibits the LPS-induced progression of sepsis in RAW264.7 cells by modulating the miR-31-5p/POU2F1 axis [98], suggesting that NEAT1, which also positively correlates with Th1 and Th17 levels [99], will be a potential target for clinical treatment of sepsis-induced organ damage.
Several miRNAs have been found to influence the course of sepsis, providing another substantial amount of evidence [100]. Over a hundred of different miRNAs have been investigated [101], identifying more or less prominent roles in the pathophysiology of sepsis-related injury by influencing the level of inflammasome induced.
What emerges is therefore an extremely interconnected network in which it is difficult to disentangle, especially if the objective is, as in our case, to try to determine whether there are the premises to attribute the role of biomarker of sepsis to one or more of the non-coding RNAs analyzed so far. It is a picture in which for each different pathological condition studied, different networks of non-coding RNA play different roles, making it impossible to assign a unique label to a specific lncRNA or miRNA.

6. Assessment of the Clinical and Prognostic Value of Non-Coding RNAs as Biomarkers in Sepsis

The role actually played in sepsis by the many non-coding RNAs that have been studied so far is only partially clarified. Many studies using cell and animal models have tried to demonstrate that non-coding RNAs may be used to control the multi-organ damage due to the septic process, but considerable challenges must be overcome in order to successfully translate these approaches into clinical practice. The concrete risk is to discuss notions that remain theoretical, not having the clinical relevance to propose themselves as concrete novelties in the future day-to-day clinical practice. Attempting to provide indications that are as translatable as possible into clinical practice, the following will be listed only the evidence available to date in which it was assessed the usefulness of miRNAs in sepsis in human clinical settings.

6.1. circRNAs

Using the research criteria previously described, only three studies to date have evaluated circ-RNAs by addressing the issue of their reliability as biomarker and prognostic value in clinical settings of sepsis in humans.
Wei et al. [102] explored the clinical values of circular RNA protein kinase C iota (circ-PRKCI) and its target microRNA-545 (miR-545) in sepsis patients. Plasma samples of 121 sepsis patients and 60 healthy controls were collected. Decreased circ-PRKCI expression and increased miR-545 expression were observed in sepsis patients compared to healthy controls, both of which had close correlations with sepsis risk. Decreased circ-PRKCI and increased miR-545 expressions were associated to 28-day mortality risk in sepsis patients, which were slightly lower than the predictive values of APACHE II score and SOFA score for predicting 28-day mortality risk.
Tian et al. [103] studied changes in circRNA expression in exosomes by circRNA microarray analysis in sepsis patients. ROC analysis showed that hsa_circRNA_104484 and hsa_circRNA_104670 have the potential to be used as novel diagnostic biomarkers and molecular therapeutic targets for sepsis.
Hong et al. [104] evaluated circFADS2 expression, a circRNA with putative protective roles in LPS-induced inflammation. Their results suggest that CircFADS2 is upregulated in sepsis to suppress LPS-induced lung cell apoptosis.

6.2. lncRNAs

Using the research criteria previously described, twelve studies addressed the issue of reliability as a biomarker and prognostic tool of lncRNAs in clinical settings of sepsis in humans. Table 1 shows the main findings as well as the diagnostic power of the different lncRNAs evaluated.

6.3. miRNAs

The research that has evaluated miRNAs with this aim in clinical settings of sepsis are of significant number, attracting the interest of researchers. We have tried to summarize in Table 2 the main elements of all available studies and in Figure 1 the main regulatory mechanisms of the pro-inflammatory state and interaction with host defense mechanisms that may substantiate their role as biomarkers in sepsis.

7. Conclusions

When facing sepsis or septic shock, time management is crucial to ensure the maximum chance of survival for the patient. Since the combined sensitivity and specificity of actual biomarkers (e.g., CPR, PCT, IL-6, etc.) do not allow for the rapid ascertainment of the diagnosis [14,15], they cannot always discriminate early enough between infectious and non-infectious patients and rapidly evolving patients from the more stable ones, whilst sepsis-related adverse outcomes rise with every hour of delay of proper intervention; new early biomarkers are urgently needed.
Non-codingRNA, including lncRNAs, circRNAs and miRNAs, cooperate in a comprehensive network deeply involved in gene function modulation, playing a pivotal role in several important regulatory mechanisms in humans, including the pathophysiology of sepsis and the sepsis-associated organ dysfunction.
The numerous studies carried out in the field of non-coding RNAs, their biogenesis, and their biological and clinical significance in recent years have identified the finding of the selective enrichment of exosomes with various networks of non-codingRNAs, different in health and disease according to options and mechanisms almost unknown today. This issue is certainly one of those most deserving of attention and investigation in the future.
Among the many studies conducted in clinical settings to ascertain the effective role of specific non-coding RNAs in supporting the early diagnosis of sepsis and in guiding therapeutic management, some have provided data that are worthy of highlighting and may provide a concrete starting point for future investigations. Some lncRNa, such as NEAT1, MALAT1, and HULC have shown interesting and promising potentials, as well as many dozens of miRNAs that seem to have the potential to flank, if not replace, the role of biomarkers in the molecules that we currently use today.
Much remains to be investigated and written about non-coding RNAs as a prognostic biomarker for sepsis. Targeted clinical studies aimed at identifying their role in everyday practice more accurately are therefore needed in the coming years.

Author Contributions

All authors have read and agreed to the published version of the manuscript.

Funding

The APC was partly funded by University of Palermo, fund assigned to Irene Simonetta as contribution for the PhD program “Clinic and Molecular Medicine”.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Conflicts of Interest

The authors declare that they have no conflicts of interest.

References

  1. Singer, M.; Deutschman, C.S.; Seymour, C.W.; Shankar-Hari, M.; Annane, D.; Bauer, M.; Bellomo, R.; Bernard, G.R.; Chiche, J.D.; Coopersmith, C.M.; et al. The Third International Consensus Definitions for Sepsis and Septic Shock (Sepsis-3). JAMA 2016, 315, 801–810. [Google Scholar] [CrossRef]
  2. Rudd, K.E.; Johnson, S.C.; Agesa, K.M.; Shackelford, K.A.; Tsoi, D.; Kievlan, D.R.; Colombara, D.V.; Ikuta, K.S.; Kissoon, N.; Finfer, S.; et al. Global, regional, and national sepsis incidence and mortality, 1990–2017: Analysis for the Global Burden of Disease Study. Lancet 2020, 395, 200–211. [Google Scholar] [CrossRef] [Green Version]
  3. Taeb, A.M.; Hooper, M.H.; Marik, P.E. Sepsis: Current Definition, Pathophysiology, Diagnosis, and Management. Nutr. Clin. Pract. 2017, 32, 296–308. [Google Scholar] [CrossRef] [PubMed]
  4. Takeuchi, O.; Akira, S. Pattern recognition receptors and inflammation. Cell 2010, 140, 805–820. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Tuttolomondo, A.; Colomba, C.; Di Bona, D.; Casuccio, A.; Di Raimondo, D.; Clemente, G.; Arnao, V.; Pecoraro, R.; Ragonese, P.; Aiello, A.; et al. HLA and killer cell immunoglobulin-like receptor (KIRs) genotyping in patients with acute viral encephalitis. Oncotarget 2018, 9, 17523–17532. [Google Scholar] [CrossRef] [Green Version]
  6. Tuttolomondo, A.; Di Raimondo, D.; Pecoraro, R.; Casuccio, A.; Di Bona, D.; Aiello, A.; Accardi, G.; Arnao, V.; Clemente, G.; Della Corte, V.; et al. HLA and killer cell immunoglobulin-like receptor (KIRs) genotyping in patients with acute ischemic stroke. J. Neuroinflamm. 2019, 16, 88. [Google Scholar] [CrossRef] [Green Version]
  7. Tuttolomondo, A.; Di Raimondo, D.; Pecoraro, R.; Arnao, V.; Pinto, A.; Licata, G. Inflammation in ischemic stroke subtypes. Curr. Pharm. Des. 2012, 18, 4289–4310. [Google Scholar] [CrossRef]
  8. Tuttolomondo, A.; Di Raimondo, D.; di Sciacca, R.; Pinto, A.; Licata, G. Inflammatory cytokines in acute ischemic stroke. Curr. Pharm. Des. 2008, 14, 3574–3589. [Google Scholar] [CrossRef]
  9. Maida, C.D.; Vasto, S.; Di Raimondo, D.; Casuccio, A.; Vassallo, V.; Daidone, M.; Del Cuore, A.; Pacinella, G.; Cirrincione, A.; Simonetta, I.; et al. Inflammatory activation and endothelial dysfunction markers in patients with permanent atrial fibrillation: A cross-sectional study. Aging 2020, 12, 8423–8433. [Google Scholar] [CrossRef]
  10. Dounousi, E.; Duni, A.; Naka, K.K.; Vartholomatos, G.; Zoccali, C. The Innate Immune System and Cardiovascular Disease in ESKD: Monocytes and Natural Killer Cells. Curr. Vasc. Pharmacol. 2021, 19, 63–76. [Google Scholar] [CrossRef]
  11. Pisetsky, D.S.; Gauley, J.; Ullal, A.J. HMGB1 and microparticles as mediators of the immune response to cell death. Antioxid. Redox Signal. 2011, 15, 2209–2219. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Kumar, A.; Roberts, D.; Wood, K.E.; Light, B.; Parrillo, J.E.; Sharma, S.; Suppes, R.; Feinstein, D.; Zanotti, S.; Taiberg, L.; et al. Duration of hypotension before initiation of effective antimicrobial therapy is the critical determinant of survival in human septic shock. Crit. Care Med. 2006, 34, 1589–1596. [Google Scholar] [CrossRef] [PubMed]
  13. Marshall, J.C.; Reinhart, K.; International Sepsis Forum. Biomarkers of sepsis. Crit. Care Med. 2009, 37, 2290–2298. [Google Scholar] [CrossRef] [PubMed]
  14. Bloos, F.; Reinhart, K. Rapid diagnosis of sepsis. Virulence 2014, 5, 154–160. [Google Scholar] [CrossRef] [PubMed]
  15. Pierrakos, C.; Vincent, J.L. Sepsis biomarkers: A review. Crit. Care 2010, 14, R15. [Google Scholar] [CrossRef] [Green Version]
  16. Zhang, X.; Wang, W.; Zhu, W.; Dong, J.; Cheng, Y.; Yin, Z.; Shen, F. Mechanisms and Functions of Long Non-Coding RNAs at Multiple Regulatory Levels. Int. J. Mol. Sci. 2019, 20, 5573. [Google Scholar] [CrossRef] [Green Version]
  17. Hansen, T.B.; Jensen, T.I.; Clausen, B.H.; Bramsen, J.B.; Finsen, B.; Damgaard, C.K.; Kjems, J. Natural RNA circles function as efficient microRNA sponges. Nature 2013, 495, 384–388. [Google Scholar] [CrossRef]
  18. Flores-Concha, M.; Oñate, Á.A. Long Non-coding RNAs in the Regulation of the Immune Response and Trained Immunity. Front. Genet. 2020, 11, 718. [Google Scholar] [CrossRef]
  19. Liang, Z.Z.; Guo, C.; Zou, M.M.; Meng, P.; Zhang, T.T. circRNA-miRNA-mRNA regulatory network in human lung cancer: An update. Cancer Cell Int. 2020, 20, 173. [Google Scholar] [CrossRef]
  20. Tuttolomondo, A.; Simonetta, I.; Pinto, A. MicroRNA and receptor mediated signaling pathways as potential therapeutic targets in heart failure. Expert Opin. Ther. Targets 2016, 20, 1287–1300. [Google Scholar] [CrossRef]
  21. Sakshi, S.; Jayasuriya, R.; Ganesan, K.; Xu, B.; Ramkumar, K.M. Role of circRNA-miRNA-mRNA interaction network in diabetes and its associated complications. Mol. Ther. Nucleic Acids 2021, 26, 1291–1302. [Google Scholar] [CrossRef] [PubMed]
  22. Zhang, M.; Bai, X.; Zeng, X.; Liu, J.; Liu, F.; Zhang, Z. circRNA-miRNA-mRNA in breast cancer. Clin. Chim. Acta Int. J. Clin. Chem. 2021, 523, 120–130. [Google Scholar] [CrossRef] [PubMed]
  23. Kong, H.; Sun, M.L.; Zhang, X.A.; Wang, X.Q. Crosstalk among circRNA/lncRNA, miRNA, and mRNA in Osteoarthritis. Front. Cell Dev. Biol. 2021, 9, 774370. [Google Scholar] [CrossRef] [PubMed]
  24. Ng, W.L.; Marinov, G.K.; Liau, E.S.; Lam, Y.L.; Lim, Y.Y.; Ea, C.K. Inducible RasGEF1B circular RNA is a positive regulator of ICAM-1 in the TLR4/LPS pathway. RNA Biol. 2016, 13, 861–871. [Google Scholar] [CrossRef] [Green Version]
  25. Agirre, X.; Meydan, C.; Jiang, Y.; Garate, L.; Doane, A.S.; Li, Z.; Verma, A.; Paiva, B.; Martín-Subero, J.I.; Elemento, O.; et al. Long non-coding RNAs discriminate the stages and gene regulatory states of human humoral immune response. Nat. Commun. 2019, 10, 821. [Google Scholar] [CrossRef] [Green Version]
  26. Biomarkers Definitions Working Group. Biomarkers and surrogate endpoints: Preferred definitions and conceptual framework. Clin. Pharmacol. Ther. 2001, 69, 89–95. [Google Scholar] [CrossRef]
  27. Black, S.; Kushner, I.; Samols, D. C-reactive Protein. J. Biol. Chem. 2004, 279, 48487–48490. [Google Scholar] [CrossRef] [Green Version]
  28. Volanakis, J.E. Acute-phase proteins in rheumatic disease. In Arthritis and Allied Conditions: A Textbook of Rheumatology, 13th ed.; Koopman, W.J., Ed.; Williams & Wilkins: Baltimore, MD, USA, 1997; pp. 505–514. [Google Scholar]
  29. Larsen, F.F.; Petersen, J.A. Novel biomarkers for sepsis: A narrative review. Eur. J. Intern. Med. 2017, 45, 46–50. [Google Scholar] [CrossRef]
  30. van Vugt, S.F.; Broekhuizen, B.D.; Lammens, C.; Zuithoff, N.P.; de Jong, P.A.; Coenen, S.; Ieven, M.; Butler, C.C.; Goossens, H.; Little, P.; et al. GRACE consortium. Use of serum C reactive protein and procalcitonin concentrations in addition to symptoms and signs to predict pneumonia in patients presenting to primary care with acute cough: Diagnostic study. BMJ 2013, 346, f2450. [Google Scholar] [CrossRef] [Green Version]
  31. Simon, L.; Gauvin, F.; Amre, D.K.; Saint-Louis, P.; Lacroix, J. Serum procalcitonin and C-reactive protein levels as markers of bacterial infection: A systematic review and meta-analysis. Clin. Infect. Dis. 2004, 39, 206–217. [Google Scholar] [CrossRef] [Green Version]
  32. Liu, Y.; Hou, J.H.; Li, Q.; Chen, K.J.; Wang, S.N.; Wang, J.M. Biomarkers for diagnosis of sepsis in patients with systemic inflammatory response syndrome: A systematic review and meta-analysis. SpringerPlus 2016, 5, 2091. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Tan, M.; Lu, Y.; Jiang, H.; Zhang, L. The diagnostic accuracy of procalcitonin and C-reactive protein for sepsis: A systematic review and meta-analysis. J. Cell. Biochem. 2018, 120, 5852–5859. [Google Scholar] [CrossRef] [PubMed]
  34. Brunkhorst, F.M.; Eberhard, O.K.; Brunkhorst, R. Discrimination of infectious and noninfectious causes of early acute respiratory distress syndrome by procalcitonin. Crit. Care Med. 1999, 27, 2172–2176. [Google Scholar] [CrossRef]
  35. Uzzan, B.; Cohen, R.; Nicolas, P.; Cucherat, M.; Perret, G.Y. Procalcitonin as a diagnostic test for sepsis in critically ill adults and after surgery or trauma: A systematic review and meta-analysis. Crit. Care Med. 2006, 34, 1996–2003. [Google Scholar] [CrossRef] [PubMed]
  36. Schmit, X.; Vincent, J.L. The time course of blood C-reactive protein concentrations in relation to the response to initial antimicrobial therapy in patients with sepsis. Infection 2008, 36, 213–219. [Google Scholar] [CrossRef]
  37. Assicot, M.; Gendrel, D.; Carsin, H.; Raymond, J.; Guilbaud, J.; Bohuon, C. High serum procalcitonin concentrations in patients with sepsis and infection. Lancet 1993, 341, 515–518. [Google Scholar] [CrossRef]
  38. Tang, B.M.; Eslick, G.D.; Craig, J.C.; McLean, A.S. Accuracy of procalcitonin for sepsis diagnosis in critically ill patients: Systematic review and meta-analysis. Lancet Infect. Dis. 2007, 7, 210–217. [Google Scholar] [CrossRef]
  39. Wacker, C.; Prkno, A.; Brunkhorst, F.M.; Schlattmann, P. Procalcitonin as a diagnostic marker for sepsis: A systematic review and meta-analysis. Lancet Infect. Dis. 2013, 13, 426–435. [Google Scholar] [CrossRef]
  40. Memar, M.Y.; Baghi, H.B. Presepsin: A promising biomarker for the detection of bacterial infections. Biomed. Pharmacother. 2019, 111, 649–656. [Google Scholar] [CrossRef]
  41. Hung, S.K.; Lan, H.M.; Han, S.T.; Wu, C.C.; Chen, K.F. Current Evidence and Limitation of Biomarkers for Detecting Sepsis and Systemic Infection. Biomedicines 2020, 8, 494. [Google Scholar] [CrossRef]
  42. Zhang, X.; Liu, D.; Liu, Y.N.; Wang, R.; Xie, L.X. The accuracy of presepsin (sCD14-ST) for the diagnosis of sepsis in adults: A meta-analysis. Crit. Care 2015, 19, 323. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Zheng, Z.; Jiang, L.; Ye, L.; Gao, Y.; Tang, L.; Zhang, M. The accuracy of presepsin for the diagnosis of sepsis from SIRS: A systematic review and meta-analysis. Ann. Intensive Care 2015, 5, 48. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Zhang, J.; Hu, Z.D.; Song, J.; Shao, J. Diagnostic Value of Presepsin for Sepsis: A Systematic Review and Meta-Analysis. Medicine 2015, 94, e2158. [Google Scholar] [CrossRef]
  45. Zeng, Z.; Li, Y.; Pan, Y.; Lan, X.; Song, F.; Sun, J.; Zhou, K.; Liu, X.; Ren, X.; Wang, F.; et al. Cancer-derived exosomal miR-25-3p promotes pre-metastatic niche formation by inducing vascular permeability and angiogenesis. Nat. Commun. 2018, 9, 5395. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Zhang, H.; Zhu, L.; Bai, M.; Liu, Y.; Zhan, Y.; Deng, T.; Yang, H.; Sun, W.; Wang, X.; Zhu, K.; et al. Exosomal circRNA derived from gastric tumor promotes white adipose browning by targeting the miR-133/PRDM16 pathway. Int. J. Cancer 2019, 144, 2501–2515. [Google Scholar] [CrossRef] [PubMed]
  47. Li, L.; Li, C.; Wang, S.; Wang, Z.; Jiang, J.; Wang, W.; Li, X.; Chen, J.; Liu, K.; Li, C.; et al. Exosomes Derived from Hypoxic Oral Squamous Cell Carcinoma Cells Deliver miR-21 to Normoxic Cells to Elicit a Prometastatic Phenotype. Cancer Res. 2016, 76, 1770–1780. [Google Scholar] [CrossRef] [Green Version]
  48. Memczak, S.; Jens, M.; Elefsinioti, A.; Torti, F.; Krueger, J.; Rybak, A.; Maier, L.; Mackowiak, S.D.; Gregersen, L.H.; Munschauer, M.; et al. Circular RNAs are a large class of animal RNAs with regulatory potency. Nature 2013, 495, 333–338. [Google Scholar] [CrossRef]
  49. Shen, T.; Han, M.; Wei, G.; Ni, T. An intriguing RNA species—Perspectives of circularized RNA. Protein Cell 2015, 6, 871–880. [Google Scholar] [CrossRef] [Green Version]
  50. Salzman, J.; Gawad, C.; Wang, P.L.; Lacayo, N.; Brown, P.O. Circular RNAs are the predominant transcript isoform from hundreds of human genes in diverse cell types. PLoS ONE 2012, 7, e30733. [Google Scholar] [CrossRef] [Green Version]
  51. Enuka, Y.; Lauriola, M.; Feldman, M.E.; Sas-Chen, A.; Ulitsky, I.; Yarden, Y. Circular RNAs are long-lived and display only minimal early alterations in response to a growth factor. Nucleic Acids Res. 2016, 44, 1370–1383. [Google Scholar] [CrossRef]
  52. Harland, R.; Misher, L. Stability of RNA in developing Xenopus embryos and identification of a destabilizing sequence in TFIIIA messenger RNA. Development 1988, 102, 837–852. [Google Scholar] [CrossRef] [PubMed]
  53. Jeck, W.R.; Sharpless, N.E. Detecting and characterizing circular RNAs. Nat. Biotechnol. 2014, 32, 453–461. [Google Scholar] [CrossRef] [PubMed]
  54. Li, Z.; Kearse, M.G.; Huang, C. The nuclear export of circular RNAs is primarily defined by their length. RNA Biol. 2019, 16, 404–412. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Zhu, L.Y.; Zhu, Y.R.; Dai, D.J.; Wang, X.; Jin, H.C. Epigenetic regulation of alternative splicing. Am. J. Cancer Res. 2018, 8, 2346–2358. [Google Scholar]
  56. Chen, L.L.; Yang, L. Regulation of circRNA biogenesis. RNA Biol. 2015, 12, 381–388. [Google Scholar] [CrossRef] [PubMed]
  57. Li, J.; Sun, D.; Pu, W.; Wang, J.; Peng, Y. Circular RNAs in Cancer: Biogenesis, Function, and Clinical Significance. Trends Cancer 2020, 6, 319–336. [Google Scholar] [CrossRef]
  58. Zhang, X.O.; Dong, R.; Zhang, Y.; Zhang, J.L.; Luo, Z.; Zhang, J.; Chen, L.L.; Yang, L. Diverse alternative back-splicing and alternative splicing landscape of circular RNAs. Genome Res. 2016, 26, 1277–1287. [Google Scholar] [CrossRef] [Green Version]
  59. Kristensen, L.S.; Andersen, M.S.; Stagsted, L.; Ebbesen, K.K.; Hansen, T.B.; Kjems, J. The biogenesis, biology and characterization of circular RNAs. Nat. Rev. Genet. 2019, 20, 675–691. [Google Scholar] [CrossRef]
  60. Lei, M.; Zheng, G.; Ning, Q.; Zheng, J.; Dong, D. Translation and functional roles of circular RNAs in human cancer. Mol. Cancer 2020, 19, 30. [Google Scholar] [CrossRef]
  61. Huang, C.; Liang, D.; Tatomer, D.C.; Wilusz, J.E. A length-dependent evolutionarily conserved pathway controls nuclear export of circular RNAs. Genes Dev. 2018, 32, 639–644. [Google Scholar] [CrossRef]
  62. Piwecka, M.; Glažar, P.; Hernandez-Miranda, L.R.; Memczak, S.; Wolf, S.A.; Rybak-Wolf, A.; Filipchyk, A.; Klironomos, F.; Cerda Jara, C.A.; Fenske, P.; et al. Loss of a mammalian circular RNA locus causes miRNA deregulation and affects brain function. Science 2017, 357, eaam8526. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Zheng, Q.; Bao, C.; Guo, W.; Li, S.; Chen, J.; Chen, B.; Luo, Y.; Lyu, D.; Li, Y.; Shi, G.; et al. Circular RNA profiling reveals an abundant circHIPK3 that regulates cell growth by sponging multiple miRNAs. Nat. Commun. 2016, 7, 11215. [Google Scholar] [CrossRef] [PubMed]
  64. Du, W.W.; Zhang, C.; Yang, W.; Yong, T.; Awan, F.M.; Yang, B.B. Identifying and Characterizing circRNA-Protein Interaction. Theranostics 2017, 7, 4183–4191. [Google Scholar] [CrossRef]
  65. Zhang, Y.; Zhang, X.O.; Chen, T.; Xiang, J.F.; Yin, Q.F.; Xing, Y.H.; Zhu, S.; Yang, L.; Chen, L.L. Circular intronic long noncoding RNAs. Mol. Cell 2013, 51, 792–806. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Su, Q.; Lv, X. Revealing new landscape of cardiovascular disease through circular RNA-miRNA-mRNA axis. Genomics 2020, 112, 1680–1685. [Google Scholar] [CrossRef] [PubMed]
  67. Guo, X.; Tan, W.; Wang, C. The emerging roles of exosomal circRNAs in diseases. Clin. Transl. Oncol. 2021, 23, 1020–1033. [Google Scholar] [CrossRef] [PubMed]
  68. Palazzo, A.F.; Lee, E.S. Non-coding RNA: What is functional and what is junk? Front. Genet. 2015, 6, 2. [Google Scholar] [CrossRef] [Green Version]
  69. Mattick, J.S.; Makunin, I.V. Non-coding RNA. Hum. Mol. Genet. 2006, 15, R17–R29. [Google Scholar] [CrossRef] [Green Version]
  70. Esteller, M. Non-coding RNAs in human disease. Nat. Rev. Genet. 2011, 12, 861–874. [Google Scholar] [CrossRef]
  71. Bhan, A.; Soleimani, M.; Mandal, S.S. Long Noncoding RNA and Cancer: A New Paradigm. Cancer Res. 2017, 77, 3965–3981. [Google Scholar] [CrossRef] [Green Version]
  72. Ohtsuka, M.; Tanemura, M.; Akamatsu, H. Long noncoding RNAs regulate malignant phenotypes in colorectal cancer. Biotarget 2018, 2, 4. [Google Scholar] [CrossRef]
  73. Harrow, J.; Frankish, A.; Gonzalez, J.M.; Tapanari, E.; Diekhans, M.; Kokocinski, F.; Aken, B.L.; Barrell, D.; Zadissa, A.; Searle, S.; et al. GENCODE: The reference human genome annotation for The ENCODE Project. Genome Res. 2012, 22, 1760–1774. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Hu, G.; Tang, Q.; Sharma, S.; Yu, F.; Escobar, T.M.; Muljo, S.A.; Zhu, J.; Zhao, K. Expression and regulation of intergenic long noncoding RNAs during T cell development and differentiation. Nat. Immunol. 2013, 14, 1190–1198. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Quinn, J.; Chang, H. Unique features of long non-coding RNA biogenesis and function. Nat. Rev. Genet. 2016, 17, 47–62. [Google Scholar] [CrossRef]
  76. Wang, K.C.; Chang, H.Y. Molecular mechanisms of long noncoding RNAs. Mol. Cell 2011, 43, 904–914. [Google Scholar] [CrossRef] [Green Version]
  77. Dahariya, S.; Paddibhatla, I.; Kumar, S.; Raghuwanshi, S.; Pallepati, A.; Gutti, R.K. Long non-coding RNA: Classification, biogenesis and functions in blood cells. Mol. Immunol. 2019, 112, 82–92. [Google Scholar] [CrossRef]
  78. Wu, H.; Yang, L.; Chen, L.L. The Diversity of Long Noncoding RNAs and Their Generation. Trends Genet. 2017, 33, 540–552. [Google Scholar] [CrossRef]
  79. Bartel, D.P. MicroRNAs: Target recognition and regulatory functions. Cell 2009, 136, 215–233. [Google Scholar] [CrossRef] [Green Version]
  80. Ha, M.; Kim, V.N. Regulation of microRNA biogenesis. Nat. Rev. Mol. Cell Biol. 2014, 15, 509–524. [Google Scholar] [CrossRef]
  81. Lee, Y.; Ahn, C.; Han, J.; Choi, H.; Kim, J.; Yim, J.; Lee, J.; Provost, P.; Rådmark, O.; Kim, S.; et al. The nuclear RNase III Drosha initiates microRNA processing. Nature 2003, 425, 415–419. [Google Scholar] [CrossRef]
  82. Krol, J.; Loedige, I.; Filipowicz, W. The widespread regulation of microRNA biogenesis, function and decay. Nat. Rev. Genet. 2010, 11, 597–610. [Google Scholar] [CrossRef] [PubMed]
  83. Chousterman, B.G.; Swirski, F.K.; Weber, G.F. Cytokine storm and sepsis disease pathogenesis. Semin. Immunopathol. 2017, 39, 517–528. [Google Scholar] [CrossRef] [PubMed]
  84. Grondman, I.; Pirvu, A.; Riza, A.; Ioana, M.; Netea, M.G. Biomarkers of inflammation and the etiology of sepsis. Biochem. Soc. Trans. 2020, 48, 537–546. [Google Scholar] [CrossRef]
  85. Yang, L.; Zhao, L.; Zhang, H.; Chen, P. Up-regulation of TUG1 can regulate miR-494/PDK4 axis to inhibit LPS-induced acute lung injury caused by sepsis. Am. J. Transl. Res. 2021, 13, 12375–12385. [Google Scholar]
  86. Qiu, N.; Xu, X.; He, Y. LncRNA TUG1 alleviates sepsis-induced acute lung injury by targeting miR-34b-5p/GAB1. BMC Pulm. Med. 2020, 20, 49. [Google Scholar] [CrossRef] [PubMed]
  87. Dong, Y.; Fan, G.; Li, Y.; Zhou, Q. TUG1 Represses Apoptosis, Autophagy, and Inflammatory Response by Regulating miR-27a-3p/SLIT2 in Lipopolysaccharide-Treated Vascular Endothelial Cells. J. Surg. Res. 2020, 256, 345–354. [Google Scholar] [CrossRef] [PubMed]
  88. Zhao, S.; Chen, W.; Li, W.; Yu, W.; Li, S.; Rao, T.; Ruan, Y.; Zhou, X.; Liu, C.; Qi, Y.; et al. LncRNA TUG1 attenuates ischaemia-reperfusion-induced apoptosis of renal tubular epithelial cells by sponging miR-144-3p via targeting Nrf2. J. Cell. Mol. Med. 2021, 25, 9767–9783. [Google Scholar] [CrossRef] [PubMed]
  89. Xu, Y.; Niu, Y.; Li, H.; Pan, G. Downregulation of lncRNA TUG1 attenuates inflammation and apoptosis of renal tubular epithelial cell induced by ischemia-reperfusion by sponging miR-449b-5p via targeting HMGB1 and MMP2. Inflammation 2020, 43, 1362–1374. [Google Scholar] [CrossRef]
  90. Su, Q.; Liu, Y.; Lv, X.W.; Ye, Z.L.; Sun, Y.H.; Kong, B.H.; Qin, Z.B. Inhibition of lncRNA TUG1 upregulates miR-142-3p to ameliorate myocardial injury during ischemia and reperfusion via targeting HMGB1- and Rac1-induced autophagy. J. Mol. Cell. Cardiol. 2019, 133, 12–25. [Google Scholar] [CrossRef]
  91. Song, T.; Wang, P.; Xin, L. LncRNA TUG1 Contributes to Hypoxia-Induced Myocardial Cell Injury through Downregulating miR-29a-3p in AC16 Cells. J. Cardiovasc. Pharmacol. 2020, 76, 533–539. [Google Scholar] [CrossRef]
  92. Cai, X.; Wang, S.; Hong, L.; Yu, S.; Li, B.; Zeng, H.; Yang, X.; Zhang, P.; Shao, L. Long Noncoding RNA Taurine-Upregulated Gene 1 Knockdown Protects Cardiomyocytes Against Hypoxia/Reoxygenation-induced Injury through Regulating miR-532-5p/Sox8 Axis. J. Cardiovasc. Pharmacol. 2020, 76, 556–563. [Google Scholar] [CrossRef] [PubMed]
  93. Wu, Z.; Zhao, S.; Li, C.; Liu, C. LncRNA TUG1 serves an important role in hypoxia-induced myocardial cell injury by regulating the miR-145-5p-Binp3 axis. Mol. Med. Rep. 2018, 17, 2422–2430. [Google Scholar] [CrossRef]
  94. Wu, X.; Liu, Y.; Mo, S.; Wei, W.; Ye, Z.; Su, Q. LncRNA TUG1 competitively binds to miR-340 to accelerate myocardial ischemia-reperfusion injury. FASEB J. 2021, 35, e21163. [Google Scholar] [CrossRef]
  95. Wei, S.; Liu, Q. Long noncoding RNA MALAT1 modulates sepsis-induced cardiac inflammation through the miR-150-5p/NF-κB axis. Int. J. Clin. Exp. Pathol. 2019, 12, 3311–3319. [Google Scholar] [PubMed]
  96. Gao, F.; Chen, R.; Xi, Y.; Zhao, Q.; Gao, H. Long noncoding RNA MALAT1 regulates sepsis in patients with burns by modulating miR-214 with TLR5. Mol. Med. Rep. 2019, 19, 3756–3766. [Google Scholar] [CrossRef] [PubMed]
  97. Wang, S.M.; Liu, G.Q.; Xian, H.B.; Si, J.L.; Qi, S.X.; Yu, Y.P. LncRNA NEAT1 alleviates sepsis-induced myocardial injury by regulating the TLR2/NF-κB signaling pathway. Eur. Rev. Med. Pharmacol. Sci. 2019, 23, 4898–4907. [Google Scholar] [CrossRef] [PubMed]
  98. Yang, Y.; Xue, J.; Qin, L.; Zhang, J.; Liu, J.; Yu, J. LncRNA NEAT1 Promotes Inflammatory Response in Sepsis via the miR-31-5p/POU2F1 Axis. Inflammation 2021, 44, 1518–1528. [Google Scholar] [CrossRef]
  99. Liu, B.; Ren, H.; Chen, J. LncRNA NEAT1 correlates with Th1 and Th17 and could serve as an assistant biomarker in sepsis. Biomark. Med. 2021, 15, 1177–1186. [Google Scholar] [CrossRef]
  100. Iro, M.A.; Soundara Pandi, S.P. Clinical application of non-coding RNAs in sepsis. Curr. Opin. Infect. Dis. 2020, 33, 530–539. [Google Scholar] [CrossRef]
  101. Ghafouri-Fard, S.; Khoshbakht, T.; Hussen, B.M.; Taheri, M.; Arefian, N. Regulatory Role of Non-Coding RNAs on Immune Responses during Sepsis. Front. Immunol. 2021, 12, 798713. [Google Scholar] [CrossRef]
  102. Wei, B.; Yu, L. Circular RNA PRKCI and microRNA-545 relate to sepsis risk, disease severity and 28-day mortality. Scand. J. Clin. Lab. Investig. 2020, 80, 659–666. [Google Scholar] [CrossRef] [PubMed]
  103. Tian, C.; Liu, J.; Di, X.; Cong, S.; Zhao, M.; Wang, K. Exosomal hsa_circRNA_104484 and hsa_circRNA_104670 may serve as potential novel biomarkers and therapeutic targets for sepsis. Sci. Rep. 2021, 11, 14141. [Google Scholar] [CrossRef] [PubMed]
  104. Hong, X.; Li, S.; Wang, J.; Zhao, Z.; Feng, Z. Circular RNA circFADS2 is overexpressed in sepsis and suppresses LPS-induced lung cell apoptosis by inhibiting the maturation of miR-15a-5p. BMC Immunol. 2021, 22, 29. [Google Scholar] [CrossRef] [PubMed]
  105. Zeng, Q.; Wu, J.; Yang, S. Circulating lncRNA ITSN1-2 is upregulated, and its high expression correlates with increased disease severity, elevated inflammation, and poor survival in sepsis patients. J. Clin. Lab. Anal. 2019, 33, e22836. [Google Scholar] [CrossRef] [Green Version]
  106. Xu, Y.; Shao, B. Circulating long noncoding RNA ZNFX1 antisense RNA negatively correlates with disease risk, severity, inflammatory markers, and predicts poor prognosis in sepsis patients. Medicine 2019, 98, e14558. [Google Scholar] [CrossRef]
  107. Gui, F.; Peng, H.; Liu, Y. Elevated circulating lnc-ANRIL/miR-125a axis level predicts higher risk, more severe disease condition, and worse prognosis of sepsis. J. Clin. Lab. Anal. 2019, 33, e22917. [Google Scholar] [CrossRef]
  108. Geng, F.; Liu, W.; Yu, L. Potential role of circulating long noncoding RNA MALAT1 in predicting disease risk, severity, and patients’ survival in sepsis. J. Clin. Lab. Anal. 2019, 33, e22968. [Google Scholar] [CrossRef] [Green Version]
  109. Wang, Y.; Fu, X.; Yu, B.; Ai, F. Long non-coding RNA THRIL predicts increased acute respiratory distress syndrome risk and positively correlates with disease severity, inflammation, and mortality in sepsis patients. J. Clin. Lab. Anal. 2020, 33, e22882. [Google Scholar] [CrossRef]
  110. Li, M.; Zhang, Z.; Liu, B.; Chen, L.; Wang, M. LncRNA GAS5 upregulates miR-214 through methylation to participate in cell apoptosis of sepsis. Arch. Physiol. Biochem. 2020, 1–6. [Google Scholar] [CrossRef]
  111. Wu, X.; Chen, D.; Yu, L. The value of circulating long non-coding RNA maternally expressed gene 3 as a predictor of higher acute respiratory distress syndrome risk and 28-day mortality in sepsis patients. J. Clin. Lab. Anal. 2020, 34, e23488. [Google Scholar] [CrossRef]
  112. Chen, J.; He, Y.; Zhou, L.; Deng, Y.; Si, L. Long non-coding RNA MALAT1 serves as an independent predictive biomarker for the diagnosis, severity and prognosis of patients with sepsis. Mol. Med. Rep. 2020, 21, 1365–1373. [Google Scholar] [CrossRef] [PubMed]
  113. Liu, W.; Geng, F.; Yu, L. Long non-coding RNA MALAT1/microRNA 125a axis presents excellent value in discriminating sepsis patients and exhibits positive association with general disease severity, organ injury, inflammation level, and mortality in sepsis patients. J. Clin. Lab. Anal. 2020, 34, e23222. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Yang, Y.; Yang, L.; Liu, Z.; Wang, Y.; Yang, J. Long noncoding RNA NEAT 1 and its target microRNA-125a in sepsis: Correlation with acute respiratory distress syndrome risk, biochemical indexes, disease severity, and 28-day mortality. J. Clin. Lab. Anal. 2020, 34, e23509. [Google Scholar] [CrossRef] [PubMed]
  115. Wang, H.; Feng, Q.; Wu, Y.; Feng, L.; Yuan, H.; Hou, L.; Wei, P.; Wang, C.; Wang, J. Association of circulating long non-coding RNA HULC expression with disease risk, inflammatory cytokines, biochemical index levels, severity-assessed scores, and mortality of sepsis. J. Clin. Lab. Anal. 2021, 35, e23656. [Google Scholar] [CrossRef] [PubMed]
  116. Liu, Y.; Peng, H.; Gui, F. Long noncoding plasmacytoma variant translocation 1 facilitates the surveillance of acute respiratory distress syndrome and mortality prediction in sepsis. Biomark. Med. 2021, 15, 401–412. [Google Scholar] [CrossRef] [PubMed]
  117. Wang, J.F.; Yu, M.L.; Yu, G.; Bian, J.J.; Deng, X.M.; Wan, X.J.; Zhu, K.M. Serum miR-146a and miR-223 as potential new biomarkers for sepsis. Biochem. Biophys. Res. Commun. 2010, 394, 184–188. [Google Scholar] [CrossRef] [PubMed]
  118. Wang, H.; Zhang, P.; Chen, W.; Feng, D.; Jia, Y.; Xie, L.X. Evidence for serum miR-15a and miR-16 levels as biomarkers that distinguish sepsis from systemic inflammatory response syndrome in human subjects. Clin. Chem. Lab. Med. 2012, 50, 1423–1428. [Google Scholar] [CrossRef]
  119. Ma, Y.; Vilanova, D.; Atalar, K.; Delfour, O.; Edgeworth, J.; Ostermann, M.; Hernandez-Fuentes, M.; Razafimahatratra, S.; Michot, B.; Persing, D.H.; et al. Genome-wide sequencing of cellular microRNAs identifies a combinatorial expression signature diagnostic of sepsis. PLoS ONE 2013, 8, e75918. [Google Scholar] [CrossRef] [Green Version]
  120. Wang, L.; Wang, H.C.; Chen, C.; Zeng, J.; Wang, Q.; Zheng, L.; Yu, H.D. Differential expression of plasma miR-146a in sepsis patients compared with non-sepsis-SIRS patients. Exp. Ther. Med. 2013, 5, 1101–1104. [Google Scholar] [CrossRef] [Green Version]
  121. Wu, Y.; Li, C.; He, Y.; Li, Q.; Wang, G.; Wen, P.; Yang, W.; Yang, Y. Relationship between expression of microRNA and inflammatory cytokines plasma level in pediatric patients with sepsis. Zhonghua Er Ke Za Zhi 2014, 52, 28–33. [Google Scholar]
  122. Yao, L.; Liu, Z.; Zhu, J.; Li, B.; Chai, C.; Tian, Y. Clinical evaluation of circulating microRNA-25 level change in sepsis and its potential relationship with oxidative stress. Int. J. Clin. Exp. Pathol. 2015, 8, 7675–7684. [Google Scholar] [PubMed]
  123. Liu, J.; Shi, K.; Chen, M.; Xu, L.; Hong, J.; Hu, B.; Yang, X.; Sun, R. Elevated miR-155 expression induces immunosuppression via CD39+ regulatory T-cells in sepsis patient. Int. J. Infect. Dis. 2015, 40, 135–141. [Google Scholar] [CrossRef] [Green Version]
  124. Han, Y.; Dai, Q.C.; Shen, H.L.; Zhang, X.W. Diagnostic value of elevated serum miRNA-143 levels in sepsis. J. Int. Med. Res. 2016, 44, 875–881. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. Yang, Y.; He, J.; Xiao, Z. Value of Micro RNA-499 in prediction of myocardial damage in sepsis. Zhonghua Wei Zhong Bing Ji Jiu Yi Xue 2015, 27, 218–220. [Google Scholar] [CrossRef] [PubMed]
  126. Wu, X.; Yang, J.; Yu, L.; Long, D. Plasma miRNA-223 correlates with risk, inflammatory markers as well as prognosis in sepsis patients. Medicine 2018, 97, e11352. [Google Scholar] [CrossRef]
  127. Lan, C.; Shi, X.; Guo, N.; Pei, H.; Zhang, H. Value of serum miR-155-5p and miR-133a-3p expression for the diagnosis and prognosis evaluation of sepsis. Zhonghua Wei Zhong Bing Ji Jiu Yi Xue 2016, 28, 694–698. [Google Scholar] [CrossRef]
  128. Sun, B.; Luan, C.; Guo, L.; Zhang, B.; Liu, Y. Low expression of microRNA-328 can predict sepsis and alleviate sepsis-induced cardiac dysfunction and inflammatory response. Braz. J. Med. Biol. Res. 2020, 53, e9501. [Google Scholar] [CrossRef]
  129. Guo, H.; Tang, L.; Xu, J.; Lin, C.; Ling, X.; Lu, C.; Liu, Z. MicroRNA-495 serves as a diagnostic biomarker in patients with sepsis and regulates sepsis-induced inflammation and cardiac dysfunction. Eur. J. Med. Res. 2019, 24, 37. [Google Scholar] [CrossRef]
  130. Zhang, W.; Jia, J.; Liu, Z.; Si, D.; Ma, L.; Zhang, G. Circulating microRNAs as biomarkers for Sepsis secondary to pneumonia diagnosed via Sepsis 3.0. BMC Pulm. Med. 2019, 19, 93. [Google Scholar] [CrossRef] [Green Version]
  131. Xu, H.; Liu, X.; Ni, H. Clinical significance of miR-19b-3p in patients with sepsis and its regulatory role in the LPS-induced inflammatory response. Eur. J. Med. Res. 2020, 25, 9. [Google Scholar] [CrossRef]
  132. Lin, R.; Hu, H.; Li, L.; Chen, G.; Luo, L.; Rao, P. The potential of microRNA-126 in predicting disease risk, mortality of sepsis, and its correlation with inflammation and sepsis severity. J. Clin. Lab. Anal. 2020, 34, e23408. [Google Scholar] [CrossRef] [PubMed]
  133. Rahmel, T.; Schäfer, S.T.; Frey, U.H.; Adamzik, M.; Peters, J. Increased circulating microRNA-122 is a biomarker for discrimination and risk stratification in patients defined by sepsis-3 criteria. PLoS ONE 2018, 13, e0197637. [Google Scholar] [CrossRef] [PubMed]
  134. Zheng, G.; Qiu, G.; Ge, M.; Meng, J.; Zhang, G.; Wang, J.; Huang, R.; Shu, Q.; Xu, J. miR-10a in Peripheral Blood Mononuclear Cells Is a Biomarker for Sepsis and Has Anti-Inflammatory Function. Mediat. Inflamm. 2020, 2020, 4370983. [Google Scholar] [CrossRef] [PubMed]
  135. Zhu, X. MiR-125b but not miR-125a is upregulated and exhibits a trend to correlate with enhanced disease severity, inflammation, and increased mortality in sepsis patients. J. Clin. Lab. Anal. 2020, 34, e23094. [Google Scholar] [CrossRef] [Green Version]
  136. Karam, R.A.; Zidan, H.E.; Karam, N.A.; Abdel Rahman, D.M.; El-Seifi, O.S. Diagnostic and prognostic significance of serum miRNA-146-a expression in Egyptian children with sepsis in a pediatric intensive care unit. J. Gene Med. 2019, 21, e3128. [Google Scholar] [CrossRef]
  137. Liu, G.; Liu, W.; Guo, J. Clinical significance of miR-181a in patients with neonatal sepsis and its regulatory role in the lipopolysaccharide-induced inflammatory response. Exp. Ther. Med. 2020, 19, 1977–1983. [Google Scholar] [CrossRef] [Green Version]
  138. Salim, R.F.; Sobeih, A.A.; Abd El Kareem, H.M. Evaluation of the clinical value of circulating miR-101, miR-187 and miR-21 in neonatal sepsis diagnosis and prognosis. Egypt. J. Med. Hum. Genet. 2020, 21, 12. [Google Scholar] [CrossRef] [Green Version]
  139. Wang, H.F.; Li, Y.; Wang, Y.Q.; Li, H.J.; Dou, L. MicroRNA-494-3p alleviates inflammatory response in sepsis by targeting TLR6. Eur. Rev. Med. Pharmacol. Sci. 2019, 23, 2971–2977. [Google Scholar] [CrossRef]
  140. Abou El-Khier, N.T.; Zaki, M.E.; Alkasaby, N.M. Study of MicroRNA-122 as a Diagnostic Biomarker of Sepsis. Egypt. J. Immunol. 2019, 26, 105–116. [Google Scholar]
  141. Na, L.; Ding, H.; Xing, E.; Zhang, Y.; Gao, J.; Liu, B.; Yu, J.; Zhao, Y. The predictive value of microRNA-21 for sepsis risk and its correlation with disease severity, systemic inflammation, and 28-day mortality in sepsis patients. J. Clin. Lab. Anal. 2020, 34, e23103. [Google Scholar] [CrossRef]
  142. Wang, Q.; Feng, Q.; Zhang, Y.; Zhou, S.; Chen, H. Decreased microRNA 103 and microRNA 107 predict increased risks of acute respiratory distress syndrome and 28-day mortality in sepsis patients. Medicine 2020, 99, e20729. [Google Scholar] [CrossRef] [PubMed]
  143. Chen, L.; Yu, L.; Zhang, R.; Zhu, L.; Shen, W. Correlation of microRNA-146a/b with disease risk, biochemical indices, inflammatory cytokines, overall disease severity, and prognosis of sepsis. Medicine 2020, 99, e19754. [Google Scholar] [CrossRef] [PubMed]
  144. Zhao, D.; Li, S.; Cui, J.; Wang, L.; Ma, X.; Li, Y. Plasma miR-125a and miR-125b in sepsis: Correlation with disease risk, inflammation, severity, and prognosis. J. Clin. Lab. Anal. 2020, 34, e23036. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  145. Lin, Y.; Ding, Y.; Song, S.; Li, M.; Wang, T.; Guo, F. Expression patterns and prognostic value of miR-210, miR-494, and miR-205 in middle-aged and old patients with sepsis-induced acute kidney injury. Bosn. J. Basic Med. Sci. 2019, 19, 249–256. [Google Scholar] [CrossRef]
  146. Liu, Z.; Yang, D.; Gao, J.; Xiang, X.; Hu, X.; Li, S.; Wu, W.; Cai, J.; Tang, C.; Zhang, D.; et al. Discovery and validation of miR-452 as an effective biomarker for acute kidney injury in sepsis. Theranostics 2020, 10, 11963–11975. [Google Scholar] [CrossRef]
  147. Li, S.; Zhao, D.; Cui, J.; Wang, L.; Ma, X.; Li, Y. Correlation of microRNA-125a/b with acute respiratory distress syndrome risk and prognosis in sepsis patients. J. Clin. Lab. Anal. 2020, 34, e23098. [Google Scholar] [CrossRef]
  148. Zhang, B.; Yu, L.; Sheng, Y. Clinical value and role of microRNA-29c-3p in sepsis-induced inflammation and cardiac dysfunction. J. Clin. Lab. Anal. 2021, 26, 90. [Google Scholar] [CrossRef]
Figure 1. MicroRNAs (miRNAs) in sepsis. Regulatory mechanisms of the pro-inflammatory state and interaction with host defense mechanisms that may substantiate their role as biomarkers.
Figure 1. MicroRNAs (miRNAs) in sepsis. Regulatory mechanisms of the pro-inflammatory state and interaction with host defense mechanisms that may substantiate their role as biomarkers.
Diagnostics 12 01355 g001
Table 1. Main findings of the study regarding lncRNAs addressing the issue of their reliability as biomarker and prognostic tool in clinical settings of sepsis in humans.
Table 1. Main findings of the study regarding lncRNAs addressing the issue of their reliability as biomarker and prognostic tool in clinical settings of sepsis in humans.
Ref.YearlncRNANovel/ValidationPattern of ExpressionSample SizeDiagnostic PowerOther Results
Sepsis CasesControl1. Sensitivity (%)2. Specificity (%)AUC
[105]2019lncRNA ITSN1-2ValidationUpregulated309HC30059.586.30.777Positive correlation with APACHE II, CRP, TNF-α, IL-6, il-8; Negative correlation with IL-10
[106]2019lncRNA ZFAS1NovelDownregulated202HC200NRNR0.814Negative correlation with APACHE II, CRP, TNF-α, IL-6; Positive correlation with IL-10; predicts survivor from non-survivor
[107]2019lncRNA
ANRIL
NovelUpregulated126HC126NRNR0.800Positive correlation with CRP, PCT, APACHE II, SOFA, TNF-α, IL-8
[108]2019lncRNA
MALAT1
NovelUpregulated190HC190NRNR0.823Positive correlation with PCT, Scr, WBC, CRP, SOFA and APACHE II; predict 28-day mortality
[109]2020lncRNA THRILNovelUpregulated32 ARDS +sepsisnonARDS-sepsis77NRNR0.706Positive correlation with CRP, PCT, TNF-α, IL-1β
[110]2020lncRNA GAS5NovelDownregulated60HC60NRNRNRPositive correlation with miRNA-214
[111]2020lncRNA MEG3ValidationUpregulated112HC10077.7940.893Predictive role for ARDS-sepsis
[112]2020lncRNA MALAT1ValidationUpregulated120HC60NRNR0.910Positive correlation with PCT, Lactate levels, SOFA and APACHE II
[113]2020lncRNA MALAT1ValidationUpregulated196HC19691.378.60.931Negative correlation with miR125a and albumin; positive correlation with APACHE II, SOFA, Scr, CRP, IL-6, IL-8, IL-1β, TNF-α
[114]2020lncRNA NEAT1ValidationUpregulated102HC100NRNR0.992Negative correlation with miR-125a
[115]2021lncRNA HULCNovelUpregulated174HC10078.7970.939Positive correlation with TNF-α, IL-6, IL-17, ICAM1, and VCAM1 APACHE II, SOFA Score,
[116]2021lncRNA PVT1ValidationUpregulated109HC100NRNRNRPredictive role for ARDS and 28-day mortality, positive correlation with disease severity;
Novel: lncRNA identified for the first time; Validation: confirmation of a finding already reported in the literature. Ref: reference; NR: Not Report; HC: Healthy controls; IL: Interleukin; TNF-α: Tumor necrosis factor-α; SOFA: Sequential Organ Failure Assessment; APACHE II: Acute Physiologic Assessment and Chronic Health Evaluation II; PCT: procalcitonin; CRP: C-reactive protein; Scr: serum creatinine; WBC: white blood count; AUC: Area under the curve; miR: microRNA; ARDS: Acute respiratory Distress Syndrome.
Table 2. Main findings of the study regarding miRNA addressing the issue of their reliability as biomarkers and prognostic tools in clinical settings of sepsis in humans.
Table 2. Main findings of the study regarding miRNA addressing the issue of their reliability as biomarkers and prognostic tools in clinical settings of sepsis in humans.
Ref.YearmiRNANovel/ValidationPattern of ExpressionSample SizeDiagnostic PowerOther Results
Sepsis CasesControlNumberSensitivity (%)Specificity (%)AUC
[117]2009miR-146aNovelDownregulated50SIRS + HCs30 + 20NRNR0.804N/A
[117]2009miR-223NovelDownregulated50SIRS + HCs30 + 20NRNR0.858N/A
[118]2012miR-15aNovelDownregulated166SIRS3268.394.40.858N/A
[119]2013miR-150NovelUpregulated23SIRS2272.785.70.830N/A
[119]2013miR-4772-5p-isoNovelDownregulated23SIRS2268.271.40.760N/A
[120]2013miR-146aValidationDownregulated14SIRS146087.50.813N/A
[121]2013miR-146aValidationUpregulated40SIRS2077.5770.815Positive correlation with miR-223, IL-10, TNF-α
[121]2013miR-123NovelUpregulated40SIRS2077.5550.678Positive correlation with miR-146a, IL-10, TNF-α
[122]2014miR-25NovelDownregulated70SIRS30NRNR0.806Negative correlation with SOFA, PCT, CRP.
Predictive role in 28-day mortality risk (AUC: 0.756)
[123]2014miR-155NovelUpregulated60HCs30NRNRNRPositive correlation with SOFA; predictive role in 28-days mortality risk (AUC: 763)
[124]2014miR-143NovelUpregulated103SIRS9578.691.60.910Positive correlation with SOFA, APACHE II
[125]2015miR-499NovelUpregulated112HCs2086.790.80.838N/A
[126]2016miR-223ValidationUpregulated187HCs18656.686.60.754Positive correlation with CRP, TNF-α, IL-1β, IL-6, IL-8 and negatively with IL-10; predicts survivor from non-survivor
[127]2016miR-155-5pValidationUpregulated105HCs3585.380.60.855N/A
[127]2016miR-133a-3pNovelUpregulated105HCs3597.954.80.769N/A
[128]2017miR-328NovelUpregulated110HCs8987.686.40.926Positive correlation with Scr, WBC, CRP, PTC, APACHE II, SOFA,
[129]2017miR-495NovelDownregulated105HCs10089.5830.915Distinguishes sepsis from sepsis shock (Sen: 85.3%; Spec: 87.3; AUC 0.885);
Negative correlation with Scr, WBC, CRP, PCT, APACHE II, SOFA
[130]2017miR-7110-5pNovelUpregulated44Non sepsis pneumonia + HC9684.290.50.883N/A
[130]2017miR-223-3pValidationUpregulated44Non sepsis pneumonia + HCs9682.91000.964N/A
[131]2017miR-19b-3pNovelDownregulated103HCs9885.485.70.921Independent prognostic factor for 28-days survival; Negative correlation with IL-6, TNF-α
[132]2018miR-126NovelUpregulated208HCs210NRNR0.726Positive correlation with APACHE II, ICU stay, MCD, Scr, CRP, TNF-α, IL-6, IL-8 and negative with IL-10
[133]2018miR-122ValidationUpregulated108Non sepsis infection2058.3950.760Independent prognostic factor for 30-days mortality (HR: 4.3)
[134]2018miR-10aNovelDownregulated62HCs20NRNR0.804Negative correlation with APACHE II, SOFA, CRP, PCT;
predictive role in 28-days mortality risk (AUC: 0.795)
[135]2018miR-125bNovelUpregulated120HCs12049.2800.658Positive correlation with APACHE II, SOFA, Scr, CRP, PCT, TNF-α, IL-6; Independent factor for mortality risk. In this study miR-125a upregulation was not associated with enhanced disease severity, inflammation, and increased mortality in sepsis patients
[136]2018miR-146aValidationDownregulated55HCs6086.656.60.803Negative correlation with CRP, PCT, IL-6, TNF-α
[137]2018miR-181aNovelDownregulated102Local infection5083.3840.893N/A
[138]2018miR-101NovelUpregulated50SIRS3084840.908N/A
[138]2018miR-187NovelUpregulated50SIRS3072760.789N/A
[138]2018miR-21NovelUpregulated50SIRS3064660.711N/A
[139]2019miR-494-3pNovelDownregulatedNRHCsNRNRNR0.837N/A
[140]2019miR-122NovelUpregulated25LWI251001001.000Higher AUC than CRP and WBC;
56% of accuracy as a prognostic biomarker
[141]2019miR-21ValidationDownregulated219HCs219NRNR0.801Negative correlation with APACHE II, SOFA, Scr, CRP, TNF-α, IL-1β, IL-6, IL-17;
[142]2019miR-103NovelDownregulated196HCs196NRNRNRNegative correlation with APACHE II, SOFA, Scr, CRP, TNF, IL-1β, IL-6, IL-8 positive with albumin;
predicted high ARDS risk (AUC: 0.727) and increased 28-days mortality risk (AUC: 0.704)
[142]2019miR-107NovelDownregulated196HC196NRNRNRNegative correlation with APACHE II, SOFA, Scr, CRP, TNF, IL-1β, IL-6, IL-8 positive with albumin;
predicted high ARDS risk (AUC: 0.694) and increased 28-days mortality risk (AUC: 0.649)
[143]2019miR-146aValidationUpregulated180HCs180NRNR0.774Positive correlation with APACHE II, SOFA, Scr, CRP, TNF-α, IL-1β, IL-6, IL17 and negative with albumin
[143]2019miR-146bNovelUpregulated180HCs180NRNR0.897Good predictive value in 28-days mortality risk (AUC: 0.703);Positive correlation with APACHE II, SOFA, Scr, CRP, TNF-α, IL-1β, IL-6, IL17 and negative with albumin
[144]2019miR-125aNovelUpregulated150HCs150NRNR0.749Positive correlation with APACHE II, SOFA. Not correlates with level of inflammation, disease severity, and 28-day mortality risk in sepsis patients
[144]2019miR-125bValidationUpregulated150HCs150NRNR0.839Positive correlation with APACHE II, SOFA, CRP, TNF-α, IL-6, IL-17, IL-23; predictive role in 28-days mortality risk (AUC: 0.699)
[145]2019miR-210NovelUpregulated125HCs1108180.90.852Positive correlation with BUN, Scr, CysC
[145]2019miR-494ValidationUpregulated125HCs11080.972.10.847Positive correlation with BUN, Scr, CysC
[145]2019miR-205NovelUpregulated125HCs11078.690.50.860Negative correlation with BUN, Scr, CysC
[146]2020miR-452NovelUpregulated97HCs89NRNRNRHigh efficacy in distinguishing AKI in sepsis patients
[147]2020miR-125aValidationUpregulated41noARDS-sepsis109NRNR0.650Positive correlation with Scr, APACHE II, SOFA
[147]2020miR-125bValidationUpregulated41noARDS-sepsis109NRNR0.739Positive correlation with with Scr, APACHE II, SOFA
[148]2021miR-29c-3pNovelUpregulated86HCs8580.281.10.872Positive correlation with APACHE II, SOFA, CRP, PCT
NOVEL (identified for the first time); VALIDATION (confirmation of a finding already reported in the literature). Ref: reference; NR: data not reported; N/A: not applicable; HC: Healthy controls; LWI: local wound infection; MCD: mechanical ventilation duration; SIRS: systemic inflammatory response syndrome; IL: Interleukin; TNF-α: Tumor necrosis factor-α; SOFA: Sequential Organ Failure Assessment; APACHE II: Acute Physiologic Assessment and Chronic Health Evaluation II; PCT: procalcitonin; CRP: C-reactive protein; Scr: serum creatinine; WBC: white blood count; BUN: blood urea nitrogen; CysC: Cystatin C; AUC: Area under the curve; Sen; sensibility; Spec: Specificity; miR: microRNA; ICU: Intensive Care Unit; HR: Hazard Ratio; ARDS: Acute respiratory Distress Syndrome; AKI: Akute Kidney Injury.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Di Raimondo, D.; Pirera, E.; Rizzo, G.; Simonetta, I.; Musiari, G.; Tuttolomondo, A. Non-Coding RNA Networks as Potential Novel Biomarker and Therapeutic Target for Sepsis and Sepsis-Related Multi-Organ Failure. Diagnostics 2022, 12, 1355. https://doi.org/10.3390/diagnostics12061355

AMA Style

Di Raimondo D, Pirera E, Rizzo G, Simonetta I, Musiari G, Tuttolomondo A. Non-Coding RNA Networks as Potential Novel Biomarker and Therapeutic Target for Sepsis and Sepsis-Related Multi-Organ Failure. Diagnostics. 2022; 12(6):1355. https://doi.org/10.3390/diagnostics12061355

Chicago/Turabian Style

Di Raimondo, Domenico, Edoardo Pirera, Giuliana Rizzo, Irene Simonetta, Gaia Musiari, and Antonino Tuttolomondo. 2022. "Non-Coding RNA Networks as Potential Novel Biomarker and Therapeutic Target for Sepsis and Sepsis-Related Multi-Organ Failure" Diagnostics 12, no. 6: 1355. https://doi.org/10.3390/diagnostics12061355

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