Next Article in Journal
A Novel Affordable and Reliable Framework for Accurate Detection and Comprehensive Analysis of Somatic Mutations in Cancer
Previous Article in Journal
The Influence of Sex Steroid Hormone Fluctuations on Capsaicin-Induced Pain and TRPV1 Expression
Previous Article in Special Issue
The Role of MicroRNAs in the Pathophysiology of Osteoarthritis
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Molecular Morbidity Score–Can MicroRNAs Assess the Burden of Disease?

1
Department of Surgery, Lambe Institute for Translational Research, University of Galway, H91 TK33 Galway, Ireland
2
Department of Surgery, University Hospital Galway, Newcastle Road, H91 YR71 Galway, Ireland
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2024, 25(15), 8042; https://doi.org/10.3390/ijms25158042
Submission received: 21 June 2024 / Revised: 17 July 2024 / Accepted: 18 July 2024 / Published: 24 July 2024
(This article belongs to the Special Issue The MicroRNAs in the Pathophysiology of Chronic Diseases)

Abstract

:
Multimorbidity refers to the presence of two or more chronic diseases and is associated with adverse outcomes for patients. Factors such as an ageing population have contributed to a rise in prevalence of multimorbidity globally; however, multimorbidity is often neglected in clinical guidelines. This is largely because patients with multimorbidity are systematically excluded from clinical trials. Accordingly, there is an urgent need to develop novel biomarkers and methods of prognostication for this cohort of patients. The hallmarks of ageing are now thought to potentiate the pathogenesis of multimorbidity. MicroRNAs are small, regulatory, noncoding RNAs which have been implicated in the pathogenesis and prognostication of numerous chronic diseases; there is a substantial body of evidence now implicating microRNA dysregulation with the different hallmarks of ageing in the aetiology of chronic diseases. This article proposes using the hallmarks of ageing as a framework to develop a panel of microRNAs to assess the prognostic burden of multimorbidity. This putative molecular morbidity score would have many potential applications, including assessing the efficacy of clinical interventions, informing clinical decision making and facilitating wider inclusion of patients with multimorbidity in clinical trials.

1. Introduction

Multimorbidity is a ubiquitous feature of clinical practice and is associated with adverse outcomes for patients, complex challenges for clinicians and increased costs for healthcare systems. Despite a reported global prevalence of at least 40%, which is inexorably increasing, multimorbidity is critically neglected in clinical trials and international guidelines [1]. There is a need to develop novel methods of predicting prognosis for patients with multimorbidity, which could facilitate personalised management strategies. To enable this, research is beginning to focus on identifying biomarkers which could enhance insight into the pathological mechanisms driving multimorbidity and offer innovative clinical utilities. MicroRNAs are small, regulatory, noncoding RNAs which control gene expression and cellular activity and have been implicated in the pathophysiology of many chronic diseases. It is unclear, however, how microRNA expression is associated with the burden of multimorbidity.

2. Aims

This study aims to demonstrate the urgent need to develop prognostic biomarkers to assess the burden of multimorbidity, then highlights the putative utility of microRNAs in this area and identifies specific microRNAs for use in future projects.

3. Multimorbidity

Comorbidity was originally defined by Feinstein 1970 as a distinct “clinical entity” which occurs in addition to an index disease [2]. Since then, a precise and universally accepted definition has not been agreed upon; it has been suggested that comorbidity can be conceptually divided into diagnostic, therapeutic and prognostic comorbidities which affect the diagnosis, treatment and prognosis of primary conditions, respectively [3]. This makes a crucial distinction between more acute conditions, such as minor musculoskeletal injuries or upper respiratory tract infections, and important chronic diseases which have broader clinical relevance [3]. Studies have demonstrated that comorbidity is associated with critical outcomes such as mortality, length of stay and readmission rates [4].
There is also a subtle distinction between comorbidity and multimorbidity, with the latter considering the co-occurrence of at least two chronic diseases [5] without considering an index disease [6]. Historically, there has been a greater emphasis on comorbidity, particularly in secondary and tertiary care settings, due to a more reductionist, disease-focused model of healthcare and biomedical research [7]. There is growing appreciation, however, of the value of the more wholistic and patient-centred perspective that the concept of multimorbidity offers, with authors acknowledging the false dichotomy between the two [6].
The prevalence of multimorbidity is increasing across the globe, though precise values vary based on the definition and methodology used by studies [5]. A recent systematic review and meta-analysis which looked at 193 studies found that the pooled prevalence of multimorbidity was 42.4%, with higher prevalences in older age groups but no significant difference between high- and low- or middle-income countries [1]. Factors such as improved diagnostic and management strategies for diseases, sedentary lifestyles predisposing to obesity and ageing populations are thought to contribute to this rising prevalence globally [8]. National populational studies have found that multimorbidity is more common in patients from lower socio-economic backgrounds, female patients and patients who smoke [9]; whilst it can occur at any age, multimorbidity is more common amongst older patients, and the number of comorbidities increases with age [9,10]. Moreover, having a single chronic disease at baseline increases the risk of developing subsequent comorbidities in a potentially self-propagating cascade [11]. As well as affecting high-income countries, the burden of chronic diseases has a significant effect on low-income and middle-income countries, with three-quarters of global mortality rates from noncommunicable diseases occurring in these settings [12,13].
This rising prevalence has serious implications for clinical practice. As well conferring the burden of composite diseases, multimorbidity is associated with factors such as increased hospitalisations, fragmentation of care and polypharmacy [5]. This creates more complex healthcare needs for patients and encumbers management for clinicians, which ultimately results in increased costs for healthcare systems [14,15,16]. Accordingly, multimorbidity has been associated with poor clinical outcomes for patients, including impaired physical and mental health functioning, higher mortality rates and frailty [17,18,19].
Despite the prolific effects of multimorbidity on clinical practice, there is a critical oversight of this in guidelines. Most guidelines focus on individual diseases and provide recommendations which are then applied cumulatively to patients with multimorbidity [20]; this is laborious and potentially harmful for patients, as these single-disease guidelines may provide conflicting and potentially contradictory recommendations [8]. There are logistical issues involved with providing recommendations for the many possible combinations of diseases [21], and guidelines are also often constructed using evidence from randomised control trials which have explicitly excluded older patients with comorbidities [8,22]. Indeed, a recent systematic review found that 80% of included randomised controlled trials excluded over half of the prospective patients, largely due to their age, comorbidities and coprescribing [23]. More specifically, clinical trials for heart failure often exclude patients with coronary heart disease, valvular disease and arrhythmias, as well as other commonly occurring and clinically important comorbidities [20]. Indeed, the exclusion of patients with clinically important comorbidities has been demonstrated in RCTs focusing on interventions for many chronic conditions, including stroke, diabetes, chronic obstructive pulmonary disease (COPD) and cancer [22,24]. This significantly limits the clinical applicability of results from these studies.
A National Institute of Clinical Excellence guideline has been developed which focuses on addressing multimorbidity; this recommends a wholistic and patient-centred approach to care which minimises the burden of treatment and disease for patients and optimises the coordination of care whilst proposing methods of identifying patients who have clinically significant multimorbidity [25]. Crucially, this guideline acknowledges the need for further research, particularly addressing issues such as the cost-effectiveness of different strategies for managing multimorbidity, as well developing tools to better predict outcomes for patients with multimorbidity [25]. More recently, authors have reiterated the urgent need for further research in this field, in particular exploring the ability to identify patients with clinically significant multimorbidity by developing superior methods of prognostication [8].
There are numerous tools which currently provide a summative assessment of the burden of multimorbidity for patients to predict clinical outcomes [1]. Sarfati divided these instruments into four categories based on whether they use a simple disease count from a prespecified reference list, assess the functionality of individual organ systems, apply differential weightings to individual conditions based on their clinical significance to generate indices or use alternative methods [26]. For each of these approaches, a list of chronic diseases affecting a patient is compiled and used to generate a prognostic score. A recent systematic review including 96 studies identified 33 different instruments that were used to assess multimorbidity [27]. The authors found that the most commonly used methods for assessing multimorbidity were simple disease counts and weighted disease indices such as the Charlson Comorbidity Index, Elders Risk Assessment and Elixhauser Index, with additional studies using case-mix or pharmaceutical-based strategies [27]. These instruments could successfully predict numerous clinical outcomes, including mortality, quality of life and hospitalisations, which have been identified as critical metrics for multimorbidity research by international consensus [28]. No consideration was given in these studies, however, to the potential use of molecular biomarkers for assessing multimorbidity.
Moreover, there are significant limitations associated with the current approaches. Direct counting methods can be cumbersome and inherently overlook the differences in clinical significance between the individual conditions [29]; weighted indices are often affected by factors such as patient populations, outcomes being investigated and overall context for assessment, which restricts how they can be applied in different clinical settings [27]. There are also significant challenges posed by data collection. Researchers are exploring the relative merits of different approaches including using chart review, discharge notes and administrative information [3]; however, there is currently no gold-standard approach for collecting chronic disease data [27]. Developing a theoretical understanding of comorbidity, including at a molecular level, could facilitate optimised and more generalisable methods of assessment.
Indeed, there has been growing research exploring the aetiological mechanisms of multimorbidity. It is thought that the biochemical hallmarks of ageing and chronic inflammation contribute to the aetiology of chronic diseases and resultant multimorbidity as the pathophysiological effects of these mechanisms accrue [8]. Genomic instability, epigenetic changes, telomere depletion, impaired proteostasis and cell signalling, mitochondrial dysfunction, cellular senescence and stem cell depletion have all been associated with the development of chronic, age-related diseases, including chronic lung diseases, neurodegenerative conditions, musculoskeletal conditions and malignancies [8]. There is a relative paucity of studies investigating how these factors specifically contribute to multimorbidity, but this is a growing area.
For example, Niedzwiedz et al. investigated the effect of telomere length on the incidence of chronic diseases [30]. They found that whilst telomere length was associated with specific chronic diseases, it had very limited associations with multimorbidity [30]. The study was limited by using a narrow set of reference diseases and did not use validated indices for assessing multimorbidity and its potential prognostic significance [30]. A separate study found that shortened absolute telomere length was associated with sarcopenia, frailty and adverse survival outcomes in patients with multimorbidity, though the actual impact on multimorbidity was not assessed [31]. Mitochondrial dysfunction in T cells was associated with secretion of inflammatory cytokines which induce senescence in a murine model of ageing and multimorbidity [32]. This also led to multisystem degeneration, with the mice exhibiting immunosuppression, heart failure, valvular abnormalities, sarcopenia and signs of neurological impairment, with associated changes in messenger RNA expression [32].
Accordingly, the pathophysiology of multimorbidity is intrinsically related to the geroscience hypothesis [8]. This proposes that the general mechanisms of deterioration associated with ageing lead to multisystem disease and so have inherent value as prospective therapeutic and clinical targets [33]. Indeed, a comprehensive literature review, which considered over 900,000 scientific abstracts, identified five hallmarks of ageing which had statistically significant associations with the presence of multimorbidity in patients [34].
Researchers are using this theoretical knowledge to identify clinically useful biomarkers for patients with multimorbidity [8]. Inflammatory and anabolic hormone biomarkers have been associated with higher levels of multimorbidity as assessed by the number of diagnosed diseases [35]. A different study used linear regression methods to identify a panel of 11 cardiovascular protein biomarkers which could predict the burden of multimorbidity out of 80 cardiovascular protein biomarkers included in the study. Interestingly, this panel was used to generate a “biomarker score” which was associated with prognostic outcomes, including mortality [36]. There is also extensive research focusing on how biomarkers can predict clusters of comorbidities, which could be used to target preventative interventions to at-risk populations [37]. There is a need for further studies to explore prospective biomarkers for patients with multimorbidity [38]. There appears to be a particular paucity of studies investigating how biomarkers can be used to assess the burden of multimorbidity by correlation with clinically validated indices.

4. MicroRNAs and Multimorbidity

MicroRNAs are small, regulatory, noncoding RNAs which control gene expression and cellular activity and have been implicated in the pathophysiology of many chronic diseases [39]. Consequently, research has shown that microRNA expression can guide the diagnosis, characterisation and prognosis of these conditions [40,41,42,43]. Specific properties enhance the putative utility of microRNA as a clinical biomarker. For example, microRNA is stable in circulation, and fixed samples and can be extracted by liquid biopsy from blood, urine and other bodily fluids [44].
Whilst there is a prodigious area of study investigating the clinical significance of microRNA in specific diseases, the implications of microRNA in the context of multimorbidity is comparatively under-researched. This is reflected in the findings of a historical review paper which found that among 59 papers which assessed the clinical significance of biomolecules in the context of comorbidity, only 5% considered microRNA [45]. At this time, there was growing recognition that molecular markers could be used to classify diseases and predict comorbidity relationships [46]. Subsequently, network models were developed which used microRNA to investigate disease–disease relationships [47]. Studies have found that including noncoding RNA such as microRNA improves the predictive accuracy of network models for disease associations, thus helping to elucidate cluster patterns for comorbidities [48].
Indeed, numerous studies have used microRNAs to explore associations between specific comorbidities. MicroRNAs have been identified as important mediators of the cardiovascular complications of rheumatoid arthritis and other systemic autoimmune disorders [49,50], whilst microRNAs have also been used to explore the mechanisms contributing to nonalcoholic steatohepatitis and periodontitis occurring as comorbidities for patients with psoriasis [51,52]. Similarly, extracellular vesicle-associated microRNAs are thought to contribute to the aetiology of multimorbidity in patients with chronic obstructive pulmonary disease [53]. Fascinating studies are using network medicine approaches, augmented by deep learning algorithms, to predict comorbidities for patients infected with SARS-CoV-2 by looking at microRNA expression profiles and identifying common molecular pathways [54]. In the field of neuropsychiatry, numerous studies have explored how microRNA can be used to predict the occurrence of diseases such as major depression, anxiety and substance use disorder as comorbidities [55,56]. A significant body of research is also investigating the clinical significance of microRNAs relating to obesity and how these can be used to predict metabolic syndrome and other related comorbidities [57,58,59].
There remains, however, a relative paucity of studies investigating how microRNAs can assess the prognostic burden of multimorbidity. This article will now identify specific microRNAs for use in future multimorbidity studies by establishing a panel of microRNAs associated with the hallmarks of ageing.

5. MicroRNAs and the Hallmarks of Ageing

The foregoing sections of the article established that the hallmarks of ageing are thought to be critical to the aetiology of multimorbidity. López-Otín et al. first proposed the nine original hallmarks of ageing in 2013 [60] to describe the mechanisms which cause the physiological deterioration and chronic diseases strongly associated with ageing. In 2023, Lopez-Otin et al. proposed three additional hallmarks—disabled macro-autophagy, chronic inflammation and dysbiosis—to account for further advances in the mechanistic theories of ageing [61]. These authors also categorised the expanded twelve hallmarks as primary, antagonistic or integrative, based, respectively, on whether they were definitively pathological, were pathological only in specific circumstances or reflected a failure of homeostatic mechanisms to compensate for accumulated physiological damage [61]. Substantial research has demonstrated how these hallmarks work synergistically to cause chronic age-related diseases [62]. Studies have also elucidated the molecular determinants of these hallmarks and have investigated how nucleic acids such as microRNAs provide transcriptional control [63].

5.1. Primary Hallmarks

5.1.1. Genomic Instability

A myriad of deleterious factors, from both endogenous and exogenous sources, contribute to DNA damage, which is exacerbated by impairments in DNA repair networks [61]. This leads to genomic instability, which has been implicated in the pathophysiology of ageing and associated chronic diseases (see Table 1) [61].
Research has explored how microRNA dysregulation contributes to genomic instability and resultant disease [64]. Lämmerhirt et al. recently demonstrated that the loss of miR-101-3p in human melanoma cells confers genomic stability, which prevents apoptosis [65]. miR-101-3p was found to directly target lamin B1, ATP-dependent helicase ATRX, caspase3 and poly(ADP-ribose)-polymerase, which are important regulators of genomic integrity, and the re-expression of miR-101-3p led to an increase in DNA damage and induction of apoptosis [65]. In colorectal cancer cells, Wang et al. found that miR-653-3p inhibited sirtuin 1 (SIRT1) whilst promoting the phosphorylation of signal transducer and activator of transcription protein 3 (STAT3) and promoting Twist family bHLH transcription factor 1 (TWIST1) expression. This contributed to the chromosomal instability and increased DNA damage seen with the ectopic expression of miR-653-3p [66]. Interestingly, genomic instability which is mediated by microRNA dysregulation has been associated with the pathophysiological sequelae of chronic inflammatory conditions. For example, In the ascending colon of patients with primary sclerosing cholangitis, upregulated miR-155 expression levels was associated with high microsatellite instability and the inhibition of suppressors of cytokine signalling 1 (SOCS1), which resulted in STAT-3 induction [67]. Moreover, the absence of miR-155 in the sigmoid colon of these patients activated the interleukin-6 (IL-6)/sphingosine-1-phosphate receptor 1 signalling pathway and affected the IL-17/forkhead box O3 (FOXP3) ratio, which was indicative of chronic inflammation [67].
MicroRNA may also mediate the effects of exogenous factors on genomic stability. Interestingly, dysregulation of the hypoxia-inducible factor-1α/miR-210/RAD52 pathway leading to defects in DNA repair is thought to contribute to genomic instability and mediate the malignant transformation, which occurs secondary to the DNA damage induced by nickel nanoparticles [68]. miR-145 has also been shown to impair the repair of DNA double-strand breaks by impeding the classical nonhomologous end-joining pathway through the suppression of DNA-dependent protein kinase [69]. This led to an increase in DNA damage and hypersensitivity to ionising radiation [69].
Researchers are investigating how microRNAs which are associated with genome instability can be used to predict prognosis for patients. In 2024, Zhang et al. assessed features of genomic instability which were exhibited by patients with breast cancer [70]. As well as finding that genomic instability was associated with adverse outcomes for this cohort, the authors found that the expression levels of miR-105-5p and miR-767-5p were significantly associated with features of genomic instability and could be used to predict prognosis [70]. In a similar study, Xu et al. identified a panel of 12 genome instability microRNAs which generated a microRNA signature which could predict the prognosis of patients with gastric cancer [71].
Table 1. MicroRNA and the primary hallmarks of ageing.
Table 1. MicroRNA and the primary hallmarks of ageing.
HallmarkMicroRNAEffectModelTarget/MechanismFindingsReference
Genomic Instability
miR-101-3pHuman melanoma cells and melanocytesLamin B1, ATRX, CASP3 and PARPRe-expression of miR-101-3p led to an increase in DNA damage and induction of apoptosis[65]
miR-105-5p and miR-767-5pPatients with breast cancerUntested2 microRNA signature was associated with genomic instability and could predict prognosis[70]
miR-653-3pHuman colorectal cancer cellsSIRT1/TWIST1 signalling pathwayEctopic expression of miR-653-3p induced increased DNA damage and chromosomal instability but inhibited apoptosis[66]
Telomere Attrition
miR-340-5pMurine model of Alzheimer’s diseasePOT1miR-340-5p upregulated telomerase activity and increased cellular telomere length, improving Alzheimer’s disease symptoms[72]
Deleting miR-126aMurine model of cholestasisversicanDeleting miR-126a induced telomere shortening and associated inflammation and hepatic dysfunction[73]
miR-185Human cell linesPOT1miR-185 reduces P0T1 and overexpression increases telomere dysfunction-induced foci signals and cellular senescence[74]
Epigenetic Alterations
miR-148aPancreatic surgery specimensN/AHypermethylation of the DNA region encoding miR-148a differentiates chronic pancreatitis and pancreatic ductal adenocarcinoma[75]
miR-7Buccal epithelial samples from patients with COPDN/AmiR-7 methylated levels could differentiate COPD phenotypes[76]
miR-223-3pGastric cancer tissue specimensArid1aMir-2223-3p promotes the progression of gastric cancer[77]
Loss of Proteostasis
miR-34Drosophilia melanogasterH3K27me3, Lst8 subunit of TORC1Loss of miR-34 expression associated with increased protein accumulation, early ageing and neurodegeneration [78]
miR-9Multiple models for Hutchinson–Gilford progeria syndromeLamin A and progerin expressionmiR-9 inhibits lamin A and progerin expression in neural cells, mitigating toxic accumulation and protecting against neurodegeneration [79]
miR-320aColorectal cancer cellseIF2, unfolded protein responsemiR-320a regulates the unfolded protein response in colorectal cancer cells[80]
Disabled Macro-Autophagy
miR-33BAL cells from patients with idiopathic pulmonary fibrosisMitochondrial homeostasis and autophagy pathwaysInhibition of miR-33 ameliorates mitochondrial homeostasis and autophagy, decreasing inflammation after bleomycin exposure[81]
miR-125bThyroid surgical specimensMAPK and AKT/mTOR signallingmiR125 expression was associated with thyroid cancer invasion and BRAFV600E mutation status[82]
miR-494Rat model of diabetic cardiomyopathyPI3K/AKT/mTOR pathwayDecreased miR-494 expression reduced apoptosis and autophagy induced by hyperglycaemia[83]
↑ denotes positive association between microRNA and disease progression/hallmark of ageing; ↓ denotes negative association between microRNA and disease progression/hallmark of ageing; miR, microRNA; ATRX, ATP-dependent helicase ATRX; CASP3, caspase 3; PARP, poly(ADP-ribose)-polymerase; SIRT1, sirtuin 1; TWIST1, Twist family bHLH transcription factor 1; POT1, protection of telomere 1; COPD, chronic obstructive pulmonary disease; TORC1, target of rapamycin complex 1; eIF2, eukaryotic initiation factor 2; BAL, bronchoalveolar lavage; MAPK, mitogen-activated protein kinase; AKT, protein kinase B; mTOR, mechanistic target of rapamycin; PI3K, phosphoinositide 3-kinase; N/A, not available.

5.1.2. Telomere Attrition

The progressive shortening of telomeres occurs following cell division in the absence of telomerase and has been associated with ageing and numerous chronic diseases [61]. miR-340-5p has been shown to upregulate the activity of telomerase and increase cellular telomere length by targeting the telomere protein protection of telomere 1 (POT1) in a murine model of Alzheimer’s disease, which led to an improvement in dementia symptoms [72]. Another study found that miR-185 was negatively associated with POT1, such that overexpression of miR-185 increased telomere dysfunction-induced foci signals in human cancer cells and primary human fibroblasts [74]. Deleting miR-126a induced age-associated telomere shortening, which was associated with inflammation and hepatic dysfunction in a murine model of cholestasis; the authors found that versican was a direct target of miR-340-5p, which mediated these effects [73].

5.1.3. Epigenetic Alterations

Epigenetic changes mediated by changes in DNA methylation, histone post-translational modification and aberrant chromatin remodelling contribute to the pathology of ageing and age-related chronic diseases [61].
Aberrant breast DNA hypermethylation in archived breast tumour tissue has been associated with diminished expression levels of miR-29a, miR-29b, miR-26a, miR-26b, miR-148a and miR-148b. Moreover, microRNA itself can be a target of epigenetic alterations which are clinicopathologically significant. Indeed, the methylation of target microRNAs is thought to contribute to the neoplastic processes of chronic lymphocytic leukaemia and can distinguish chronic lymphocytic leukaemia samples from healthy controls [84]. As a more specific example, hypermethylation of the DNA region encoding miR-148a leads to a repression of this microRNA in pancreatic ductal adenocarcinoma and precursor pancreatic intraepithelial neoplasia lesions [75]; this has been proposed as a biomarker to differentiate pancreatic ductal adenocarcinoma from chronic pancreatitis [75]. More recently, Ma et al. demonstrated that DNA methyltransferase causes the hypermethylation of the promoter region for miR-34a, thereby repressing its expression [85]. This is important as the authors found that increased expression of miR-34a inhibits the activation of the Notch pathway, which improves the sensitivity of pancreatic cancer cells to molecular therapies [85].
The significance of microRNA-mediated changes in DNA hypermethylation extends beyond oncology. The methylation status of miR-7 has been shown to differentiate between different clinical phenotypes of patients with COPD [76]. Hypermethylation was more common in patients with emphysema compared to those who had frequent exacerbations, chronic bronchitis and asthma COPD overlap syndrome [76].
The interconnected relationship between microRNA expression and hypermethylation reflects the reciprocal pathways by which epigenetic alterations cause disease [86]. This can be seen in the mechanisms of chromatin remodelling. Switch/sucrose nonfermentable complexes (SWI/SNF) and imitation switch complexes (ISWI) effect chromatin remodelling and are influenced by microRNA expression. Arid1a is a subunit of the SWI/SNF complex, and Zhu et al. showed that miR-223-3p regulated the expression of Arid1a, which promoted the proliferation and invasion of gastric cancer cells [77]. Similarly, Wang et al. showed that MiR-146b-5p inhibited the SMARCA5 subunit of ISWI in glioma stem-like cells/mesenchymal stem cell fusion cells, which led to an inactivation of a transforming growth factor-β (TGF-β) pathway and impaired proliferation and invasion of the fusion cell, thus suggesting a critical role in malignant progression [87].

5.1.4. Loss of Proteostasis

Ageing and numerous ageing-associated chronic diseases such as Alzheimer’s are associated with the accumulation of pathologically altered proteins due to deficiencies in normal mechanisms of proteostasis [61]. This could be due to disease-specific mutations increasing the risk of protein misfolding, or, alternatively, changes in the pathways for translation of proteins can promote translational errors leading to disease [88]. Impaired proteostasis can also occur secondary to dysfunction of regulatory components which normally remove defunct proteins, such as the unfolded protein response of the endoplasmic reticulum [61]. Additionally, impaired degradation of proteins may be mediated by deficits in pathways such as chaperone-mediated autophagy, which is dependent on factors such as lysosome-associated membrane protein type 2A (LAMP2A) [61].
Researchers are currently exploring the putative effect of microRNAs on cellular proteostasis. Changes in the expression of miR-34 have been shown to affect proteostasis, leading to the accumulation of protein aggregation markers, early ageing and features of neurodegeneration in Drosophila melanogaster [78]. This effect was mediated by the dysregulated translation of proteins, with altered H3K27me3 and specific changes affecting the Lst8 subunit of target of rapamycin complex 1 (TORC1), identified as possible targets of miR-34 [78]. In another study, miR-71 was shown to promote ubiquitin-dependent protein turnover [89]. Interestingly the authors found that miR-71 targeted the Toll receptor domain protein TIR-1 in AWC olfactory neurons and that the effect of olfaction on this axis influenced proteostasis and longevity [89]. In laminopathy, dysregulated proteostasis affecting the nuclear matrix protein lamin A is associated with multisystem pathology and syndromes of progeria [90]. This is mediated by a confluence of additional hallmarks including cell senescence, apoptosis and unstable genome integrity [90]. Interestingly, microRNAs have been proposed as potential therapeutic agents for use in laminopathy [90]. Indeed, Nissan et al. showed that miR-9 inhibited the expression of lamin A and the precursor progerin, thus preventing the toxic accumulation of progerin in patients with Hutchinson–Gilford progeria syndrome [79].
Moreover, there is interest in how microRNAs affect specific mechanisms of proteostasis [91]. Researchers are investigating how microRNAs impact the unfolded protein response and what significance this relationship has clinically [92]. Fields et al. showed that miR-320a targets the eukaryotic initiation factor 2 (eIF2) signalling pathway to affect control of the unfolded protein response in colorectal cancer cells [80]. Similarly, microRNAs control chaperone-mediated autophagy; Let7b microRNA increases the expression of LAMP2A, which affects the degradation of the mutant huntingtin aggregates which cause Huntington’s disease [93]. Consequently, let7b microRNA is thought to play an important role in the pathophysiology of Huntington’s disease and has been suggestive as a candidate clinical biomarker for this condition [93].

5.1.5. Disabled Macro-Autophagy

Cytoplasmic material, including nonproteinaceous macromolecules and organelles, is sequestered in autophagosomes which subsequently fuse with lysosomes, leading to the destruction of the contents; dysfunction of this process is implicated in pathological ageing [61]. Researchers are endeavouring to elucidate the mechanisms by which microRNAs may regulate this process and thus contribute to disease [94].
Inhibiting miR-33 in lung macrophages was found to reduce fibrotic changes following bleomycin in a murine model of pulmonary fibrosis [81]. Interestingly, this was associated with upregulated signs of autophagy and mitophagy, which the authors proposed contributed to this therapeutic effect [81]. A separate study found that miR-125b regulated autophagy in patients with thyroid cancer and that decreased the expression of hsa-miR-125b could differentiate malignant from benign thyroid tumours [82]. Here, miR-125b was found to target the mitogen-activated protein kinase (MAPK) and AKT/mechanistic target of rapamycin (mTOR) signalling cascades, which may have mediated the effects of this microRNA on autophagy and resultant carcinogenesis [82]. Furthermore, Ning et al. demonstrated that the expression of miR-494 was upregulated in the myocardium of rats with diabetic cardiomyopathy [83]. By analysing the expression levels of BCl-2 and Bax, the authors showed that miR-494 upregulated autophagy and also regulated phosphorylation of the phosphoinositide 3-kinase (PI3K)/AKT/mTOR signalling pathway, which, the authors argued, provides valuable insights into the aetiological mechanisms driving diabetic cardiomyopathy [83].

5.2. Antagonistic Hallmarks

5.2.1. Cellular Senescence

Cellular senescence refers to the stable arrest of proliferation (see Table 2). Senescence occurs in response to tissue damage; it is strongly associated with ageing and occurs in all cell types [61]. This process relies on factors such as the activation of the tumour suppressors CDKN2A/p16 and tumour protein 53 and depletion of lamin B1 from the nuclear envelope [61]. Failure or bypass of senescence is implicated in carcinogenesis, whilst nonproliferative diseases such as pulmonary fibrosis, hepatic steatosis and metabolic syndrome are associated with the senescence-associated secretory phenotype [61]. The senescence-associated secretory phenotype is a heterogenous and complex pro-inflammatory mechanism which is thought to promote localised healing in response to tissue injury: however, this results in disease when it becomes dysregulated [61]. The molecular regulation of senescence pathways is a rapidly evolving area of research, and microRNAs are now understood to have an important role at both the transcriptional and post-transcriptional levels [95].
Indeed, significant research is focused on further understanding the regulatory role of microRNAs in senescence pathways and how this can be applied clinically [95]. Using human cell lines and murine models, Hui et al. showed that the expression of miR-3200-3p in tumour-derived exosomes promotes the senescence of regulatory T cells in non-small-cell lung cancer and potentiates carcinogenesis [96]. This was mediated by decreasing the expression of damage-specific DNA-binding protein 1 which consequently suppressed DCAF1/glutathione S-transferase P1 [96]. MicroRNA regulation of cell senescence has also been implicated in gastric cancer. For example, bromodomain-containing protein 4 (BRD4) has been shown to upregulate the E2F-mediated expression of miR-106b-5p, which suppresses p21 and thereby inhibits cell senescence [97]. Importantly, JQ1 is thought to mitigate the proliferation of gastric cancer cells by inhibiting BRD4 and the E2F/miR-106b-5p/p21 axis and thus inducing senescence [97]. This pathway has been identified as a potential target for future therapeutic targets.
There have been similar findings in nonproliferative chronic diseases. Lu et al. investigated the importance of microRNA regulation of cellular senescence in the pathogenesis of chronic obstructive pulmonary disorder [98]. Overexpression of mir-377-3p was shown to induce senescence in lung fibroblasts, which potentiated pathological changes such as airway remodelling. This occurred secondary to profibrotic effects of the senescence-associated secretory phenotype and suppression of the ZFP36L1 protein [98]. Furthermore, Zhao et al. considered how microRNA and senescence may contribute to age-associated cardiac dysfunction [99]. Firstly, they found that there was an age-associated fall in miR-203 expression which corresponded to increased cardiac dysfunction. The authors showed that miR-203 was important for regulating senescence in cardiomyocytes and, interestingly, old transgenic mice with systemic overexpression of miR-203 exhibited improved cardiac function compared to wild-type controls [99]. The authors demonstrated that this increase in miR-203 attenuated age-associated senescence and remodelling seen in the murine cardiac tissue. This was in part mediated by the actions of miR-203 in suppressing poly (ADP-ribose) polymerase 1, leading to an increase in nicotinamide adenine dinucleotide+ and a resultant improvement in mitochondrial function in senescent cardiomyocytes [99]. Similar findings have implicated microRNA regulation of senescence with other chronic conditions, such as Alzheimer’s disease [100].
Other studies are actively exploring how senescence-associated microRNAs can be targeted therapeutically. Zhang et al. found that increased miR-217-5p expression in exosomes secreted by senescent epithelial cells in a murine model was associated with the progression of pulmonary fibrosis following paraquat exposure [101]. The authors found that increased expression of miR-217-5p led to the inhibition of SIRT1, which resulted in increased levels of acetylated β-catenin and the activation of Wnt signalling. Accordingly, increased miR-217-5p expression levels mediated the activation of pulmonary fibroblasts such that suppression of miR-217-5p with an antagomir resulted in a reversal of the paraquat-induced pulmonary fibrosis [101].
Similar therapeutic strategies have been investigated in other studies. A recent study investigated the cartilaginous expression of miR-708-5p, which was shown to decline with age [102]. This was also associated with the development of more severe osteoarthritic lesions in response to mechanical stress in older rats compared to younger subjects. Crucially, the authors showed that local injection of an agonist for miR-708-5p could reduce degenerative changes and partially restore the chondrogenic differentiation capability of the tissue by suppressing TLR4 expression in the Toll-like receptor 4/nuclear factor-κB (NF-κB) pathway [102]. Similarly, dysregulated expression of specific microRNAs and the resultant effects on senescence have been implicated in the aetiology of diabetic ulcers [103]. Wei et al. developed a novel bioactive wound dressing using small extracellular vesicles which exhibited enriched expression levels of MiR-17-5p [104]. By targeting phosphatase and tensin homologue (PTEN) and p21, this was shown to inhibit senescence and promote angiogenesis and collagen deposition, with a resultant enhancement of diabetic wound healing [104].
The development of therapeutic strategies targeting senescence-associated microRNAs illustrates the centrality of these microRNAs to the aetiology of diseases. This may also provide indirect validation for the use of senescence-associated microRNAs to guide prognostic assessments, though more research is needed to confirm this.
Table 2. MicroRNAs and the antagonistic hallmarks of ageing.
Table 2. MicroRNAs and the antagonistic hallmarks of ageing.
HallmarkMicroRNAEffectModelTarget/MechanismFindingsReference
Cellular Senescence
miR-3200-3pCellular and murine modelDDB1 in Treg cellsInhibition of VEGFR2 upregulates miR-3200-3p which targets DDB1 in Treg cells to promote senescence in non-small-cell lung cancer[96]
miR-377-3pHuman patients and cellular and murine modelsZFP36L1miR-377-3p promotes lung fibroblast senescence and suppresses ZFP36L1 to exacerbate COPD[98]
miR-106b-5pHuman gastric cancer cell lines and patient gastric tissue samplesE2F/miR-106b-5p/p21 axisBRD4 modulates the proliferation of gastric cancer cells by controlling cellular senescence by targeting E2F/miR-106b-5p/p21 axis[97]
Mitochondrial Dysfunction
miR-128-3pMurine asthma modelSIX1miR-128-3p controls airway inflammation by targeting SIX1 and regulating mitochondrial function[105]
miR-181aMurine modelPDCD4miR-181a targets PDCD4 to modulate mitochondrial fission and apoptosis and preserve left ventricular function following myocardial infarction[106]
miR-328-5pin vitro and murine modelSirt1lncRNA Glis2 inhibited miR-328-5p to improve mitochondrial function and mitigate podocyte apoptosis and progression of diabetic nephropathy[107]
Deregulated Nutrient-Sensing
miR-221Breast cancer cell linePTEN/Akt/mTOR signallingmiR-221 mediated breast cancer cell proliferation and resistance to adriamycin by modulating PTEN/Akt/mTOR signalling[108]
miR-125bMurine modelRRAGD/mTOR/ULK1 pathwayAtherosclerotic progression was associated with reduced autophagy and downregulated expression of miR-125b[109]
miR-192-5pHuman patients with NAFLD and murine modelsRictor/Akt/FOX01Increased expression of miR-192-5p promotes hepatic macrophage activation and disease progression in NAFLD by modulating Rictor/Akt/Fox01 signalling[110]
↑ denotes positive association between microRNA and disease progression/hallmark of ageing; ↓ denotes negative association between microRNA and disease progression/hallmark of ageing; MiR, microRNA; DDB1, damage-specific DNA-binding protein 1; VEGFR2, vascular endothelial growth factor receptor 2; COPD, chronic obstructive pulmonary disease; BRD4, bromodomain-containing protein 4; SIX1, sine oculis homeobox homologue 1; PDCD4, programmed cell death protein 4; Sirt1, sirtuin1; lncRNA, long noncoding RNA; PTEN, phosphatase and tensin homologue; mTOR, mechanistic target of rapamycin; NAFLD, nonalcoholic fatty liver disease; Rictor, rapamycin-insensitive companion of mammalian target of rapamycin; Fox01, forkhead box transcription factor 01.

5.2.2. Mitochondrial Dysfunction

Mitochondrial dysfunction has been implicated in pathological ageing [61]. Factors such as mutations in mitochondrial DNA and deficiencies in proteostasis which accrue over time contribute to a deterioration in the function of the organelle. Reactive oxygen species or mitochondrial DNA leak out of the mitochondria, which leads to a cascade of adverse physiological effects including inflammation and cell death [61]. The rise in reactive oxygen species is also exacerbated by the inherent decline in antioxidant defence, which is associated with ageing [111]. This results in increased oxidative stress, which has been associated with many of the hallmarks of ageing and is an area of ongoing research [111].
Dysregulated microRNA signalling is now recognised to have a significant impact on mitochondrial functionality, and this relationship is the focus of ongoing research [112]. Liu et al. found that miR-128-3p inhibited sine oculis homeobox homologue 1 (SIX1) in a murine model of asthma [105]. This affected processes of mitochondrial fission and fusion and alleviated mitochondrial dysfunction, which mitigated resultant apoptosis and the progression of airway inflammation. The miR-128-3p/SIX1 axis was thus recognised to be an important mechanism in the pathophysiology of asthma, and the authors posited that it could offer a new therapeutic target [105]. In a different study, Cheng et al. found that changes to miR-29a-3p/DNA methyltransferase 3A (DNMT3A) mediated the beneficial effects of curcumin in a murine model of pulmonary fibrosis by regulating fibrotic pathways and mitochondrial function [113]. Curcumin increased the expression of miR-29a-3p in pulmonary tissue, which led to a reduction in DNMT3A levels. This inhibited extracellular matrix remodelling and, interestingly, attenuated a fibrosis-associated increase in mitochondrial function [113].
Similar findings have been seen in studies involving numerous diseases. miR-181-a was shown to reduce both the size of infarction and the level of ischaemia-induced apoptosis following occlusion of the left-anterior-descending artery in mice and also preserved left ventricular functioning [106]. This was mediated by a direct downregulation of programmed cell death protein 4 and BID, which mitigated the apoptosis and mitochondrial fission occurring as a result of anoxia [106]. Similarly, MiR-124 overexpression was shown to alleviate oxidative stress and resultant apoptosis and improve mitochondrial function in an in vitro model of hypoxic ischaemic brain damage by inhibiting the expression of STAT3 [114]. These findings were corroborated in an in vivo model of hypoxic ischaemic brain damage using rats, and, importantly, treatment with a miR-124 agomir reduced cerebral infarct volume, local oedema and neuronal damage and improved neurological function [114].
Other studies have investigated chronic kidney diseases. Long noncoding RNA (lncRNA) Glis2 was found to inhibit the apoptosis of mouse podocytes in an in vitro model of diabetic nephropathy by ameliorating changes in mitochondrial function and morphology [107]. Significantly, lnc Glis2 was shown to act as a competing endogenous RNA for miR-328-5p, which mediated these beneficial effects; miR-328-5p directly inhibited the expression of Sirt1, which was shown to reduce podocyte apoptosis and improve mitochondrial function [107]. Crucially, the authors demonstrated that inducing lncRNA Glis2 overexpression in diabetic mice attenuated podocyte apoptosis and pathological changes associated with diabetic nephropathy, including podocyte foot process effacement and glomerular basement membrane thickening [107]. This validated the clinical utility of their findings. Indeed, the exosomal delivery of miR-204 was investigated as a novel therapeutic strategy to ameliorate mitochondrial dysfunction in a murine model of diabetic nephropathy [115]. This emphasises the potential therapeutic value of understanding microRNA-mediated regulation of mitochondrial function.

5.2.3. Deregulated Nutrient Sensing

Nutrient-sensing networks mediate the cellular response to nutritional status and physiological stress [61]. This involves insulins and insulin-like growth factors (IGFs) and their tyrosine kinase receptor targets, as well as numerous intracellular cascades. The somatotrophic axis, ALK-dependent signalling and mTOR are all important components of nutrient sensing and have been implicated in both longevity and numerous chronic diseases, including malignancies [61,116].
There is growing appreciation for the importance of microRNA regulation of mTOR for the aetiology of malignancies [117]. In 2016, Zhang et al. showed that miR-147 mitigated the malignant progression of a breast cancer cell line by inhibiting the Akt/mTOR pathway [118]. More recently, Yin et al. found that miR-221 mediated tumour cell proliferation and resistance to adriamycin by inhibiting the expression of PTEN, which upregulated the activation of the Akt/mTOR pathway in a breast cancer cell line [108]. Using renal cancer cell lines, Shen et al. demonstrated that miR-188-5p inhibits the proliferation and invasion of renal cancer cells by downregulating myristoylated alanine-rich C-kinase substrate (MARCKS) expression and inhibiting the AKT/mTOR signalling pathway [119]. Interestingly, in the study, miR-188-5p was then used therapeutically to successfully limit renal cancer growth in an in vivo murine model [119]. Similar findings highlighted the clinical significance of microRNA regulation of mTOR in prostate cancer [120].
Moreover, this pathway is implicated in nonmalignant chronic diseases. An interesting historical study found that inhibiting miR-7a activated mTOR signalling, which enhanced the proliferation of pancreatic islet cells in mice; the authors suggested that targeting the mir-7–mTOR axis may therefore have potential therapeutic value for treating diabetes [121]. In 2023, Ning et al. showed that exposure to high glucose upregulated the expression of miR-494, which led to increased autophagy and apoptosis in cardiomyocytes [83]. Crucially, the increased expression of miR-494 was associated with a decreased phosphorylation of the PI3K/AKT/mTOR pathway, which the authors proposed could potentiate the pathogenesis of diabetic cardiomyopathy [83]. In a separate study, atherosclerotic progression was associated with reduced autophagy and the decreased expression of miR-125b using a murine model [109]. Conversely, overexpression of miR-125b upregulated autophagy by modulating the RRAGD/mTOR/Unc-51-like autophagy, activating the kinase 1 (ULK1) pathway, which led to reduced atherosclerotic changes [109]. Likewise, miR-101 is thought to ameliorate the progression of liver fibrosis by inhibiting the PI3K/Akt/mTOR pathway, whilst the effect of miR-155 in suppressing this signalling pathway is also thought to facilitate the development of epilepsy [122,123].
Indeed, the microRNA regulation of additional nutrient-sensing pathways is similarly important for the aetiology of chronic diseases. Forkhead box transcription factor Os (FoxOs) are important transcription factors in nutrient-sensing pathways [61]; increased expression of miR-192-5p has a pro-inflammatory effect in nonalcoholic steatohepatitis and promotes hepatic macrophage activation and disease progression by targeting the rapamycin-insensitive companion of the mammalian target of rapamycin (Rictor)/Akt/FoxO1 signalling pathway [110]. In a different study, MiR-30a-3p was shown to reduce the proliferation and invasion of hepatocellular carcinoma cells by inhibiting IGF-1, which is an important extracellular ligand for nutrient sensing [61,124]. Interestingly, miR-221 was found to propagate androgenetic alopecia by suppressing IGF-1 expression, which downregulated the MAPK pathway in dermal papilla cells and downregulated the PI3K/AKT pathway in dermal sheath cells [125].
Studies are also investigating how microRNA expression is associated with the sequelae of overnutrition. Overnutrition leads to obesity, which increases the risk of numerous chronic diseases through factors such as pathological adipokine secretion, which potentiates a chronic inflammatory state [126,127]. MicroRNAs are now thought to mediate some of these adverse effects. For example, changes in the expression of MiR-34a in visceral murine adipose tissue induced pro-inflammatory macrophage infiltration and activation and mediated the development of metabolic dysfunction including insulin resistance in response to a high-fat diet [128]. Furthermore, there is additional interest in how changes in microRNA expression are implicated in the therapeutic benefit of dietary interventions such as intermittent fasting, which is an area of emerging interest [129].

5.3. Integrative Hallmarks

5.3.1. Stem Cell Exhaustion

Adequate stem cell function is essential for the repair and renewal processes which facilitate cellular response to injury; deterioration in this capacity is implicated in pathological ageing (see Table 3) [61]. Intrinsic to this is cellular reprogramming, wherein cells undergo dedifferentiation and simultaneously experience a physiological “rejuvenation” [61]. As a result, the cells are reset and lose adverse ageing-associated features, which is reflected in changes like the lengthening of telomeres [61]. Transcription factors such as octamer-binding transcription factor 4 (OCT4), sex-determining region Y-box 2 (SOX2), Krüppel-like factor 4 (KLF4) and MYC and forkhead box protein M1 (FOXM1) regulate this process [61].
Studies are exploring how microRNA expression levels affect stem cell function. In 2013, Yu et al. found that miR-141-3p downregulated ZMPSTE24, which led to an accumulation of prelamin A in the nuclear envelope and mediated cellular senescence using a model for ageing human mesenchymal stem cells [130]. More recently, Jani et al. showed that the mir-221/222 cluster promotes haematopoietic stem cell quiescence and multipotency by targeting the Fos/activator protein-1 (AP-1)/immediate early genes (IEG) pathway and reducing granulopoiesis [131]. The authors suggested that this could be used to optimise outcomes following bone marrow transplantation [131]. Interestingly, in a different study, miR-146a was found to regulate the functionality of haematopoietic stem cells as well as their progression through haematopoiesis in response to chronic inflammation in a murine model [132]. Here, Zhao et al. also demonstrated that these effects were mediated by changes in the miR-146a/tumour necrosis factor receptor associated factor 6 (TRAF6)/NF-κB/IL-6 pathway, which the authors suggested may have significant implications for the aetiology of myeloid malignancies [132]. Su et al. showed that miR-31 modulated the IL-34/Janus kinase (JAK)–STAT3 pathway to control the differentiation and functional reserve of satellite cells, thereby affecting the regenerative capacity of skeletal muscle [133]. Fascinating studies are now exploring how the microRNA regulation of stem cell function can be used to develop innovative therapeutic strategies [134,135].
Cellular reprogramming is a fascinating process by which the differentiation of stem cells can be reversed. This is regulated by transcription factors such as Oct3/4, Sox2, c-Myc and Klf4 [136]. MicroRNAs are now understood to have a central role in modulating these processes, and dysregulation of microRNA control of these mechanisms is implicated in chronic diseases including malignancies [137,138]. For example, Xie et al. showed that circRNA vacuole membrane protein 1 (CircVMP1) expression contributed to cisplatin resistance and tumour progression in non-small-cell lung cancer cells by downregulating miR-524-5p, which promoted the expression of methyltransferase 3 N6-adenosine-methyltransferase complex catalytic subunit and SOX2 [139]. Similarly, Chen et al. demonstrated that circRNA derived from the pleiotophin gene (circPTN) downregulates miR-145-5p and miR-330-5p in glioma cells to potentiate proliferation [140]. Interestingly, in the study, miR-145-5p was shown be critical for tumorigenesis by promoting the capacity of glioma cells for self-renewal; this was evidenced by factors such as an increase in stemness markers such as SOX2, SOX9 and Nestin [140]. Indeed, in a separate study, miR-122 was shown to reduce the stemness and chemoresistance of hepatic cancer cells by targeting Wnt/β-catenin signalling [141].
Table 3. MicroRNAs and the integrative hallmarks of ageing.
Table 3. MicroRNAs and the integrative hallmarks of ageing.
HallmarkMicroRNAEffectModelTarget/MechanismFindingsReference
Stem Cell Exhaustion
miR-31Murine modelIL34, JAK-STAT3 signallingmiR-31 modulates IL-34/JAK-STAT3 signalling to determine the differentiation and functional reserve of satellite cells, thus regulating the regenerative capacity of skeletal muscle[133]
miR-524-5pNSCLC cell linesmiR-524-5p-METTL3/SOX2 axiscircVMP1 potentiates NSCLC progression and DDP resistance by modulating miR-524-5p-METTL3/SOX2 axis[139]
miR-122Human hepatic cancer cell lineWnt/β-cateninmiR-122 reduces the stemness and chemoresistance of hepatic cancer cells by modulating Wnt/β-catenin signalling[141]
Altered Intercellular Communication
miR-322-5pRat model of myocardial infarctionSmurf2, TGF-β/Smad pathwaymiR-322-5p/Smurf2 axis modulates TGF-β/Smad signalling to potentiate myocardial injury following myocardial infarction [142]
miR-582-5pHuman NSCLC cell linesHippo-YAP/TAZmiR-582-5p induces tumour-suppressive changes in NSCLC cells by downregulating YAP/TAZ signalling[143]
miR-133bMurine model of atherosclerosisNotch signallingMiR-133b exacerbates atherosclerosis by activating Notch signalling[144]
Chronic Inflammation
miR-210Human patients with psoriasis vulgarisFOXP3Upregulated miR-210 modulates FOXP3 in CD4+ T cells to potentiate immune dysfunction in psoriasis vulgaris[145]
miR-181bHuman osteosarcoma tissue samplesIl-1β/NF-κBIL-1β/NF-κB signalling induces overexpression of miR-181b which promotes the proliferation of osteosarcoma cells[146]
miR-29a/29bHuman patients with cirrhosisN/AReduced miR-29a/miR-29b expression was associated with upregulated IL-6 and TNF-α and a more advanced grade of cirrhosis [147]
Dysbiosis
miR-582-3pPatients with NASH and in vitro modelsTMBIM1Gut microbiota affected expression of miR-582-3p which potentiated hepatic fibrosis[148]
miR-122Murine modelN/AIntestinal flora-produced butyrate downregulates miR-122 expression which ameliorates diet-induced hypercholesterolaemia[149]
miR-29aMurine modelN/AmiR-29a alleviated hepatic steatosis, altered the intestinal flora, reduced inflammation and improved lipid metabolism[150]
↑ denotes positive association between microRNA and disease progression/hallmark of ageing; ↓ denotes negative association between microRNA and disease progression/hallmark of ageing; miR, microRNA; IL34, interleukin 34; JAK-STAT3, Janus kinase-signal transducer and activator of transcription protein 3; NSCLC, non-small-cell lung cancer; METTL3, methyltransferase 3; SOX2, SRY-box transcription factor 2; circVMP1, circRNA vacuole membrane protein 1; DDP, cisplatin; Smurf2, Smad ubiquitin regulatory factor 2; TGF-β, transforming growth factor-β; YAP, yes-associated protein; TAZ, WW domain-containing transcription regulator protein 1; FOXP3, forkhead box P3; IL-1β, interleukin-1β; NF-κB, nuclear factor- κB; IL-6, interleukin-6; TNF-α, tumour necrosis factor-alpha; NASH, nonalcoholic steatohepatitis; TMBIM1, transmembrane BAX inhibitor motif-containing 1.

5.3.2. Altered Intercellular Communication

The dysregulation of intercellular communication is implicated in the aetiology of ageing and associated chronic diseases [61]. Deficiencies in numerous pathways accrue over time which overlap with several other hallmarks including chronic inflammation, dysbiosis and the senescence-associated secretory phenotype [61].
The microRNA regulation of intercellular communication is an emerging area of study. Specific factors in blood, including eotaxin (CCL11), TGF-β and the complement component 1q (C1q) have been associated with pathological ageing [61]. miR-629-3p has been shown to potentiate the effect of IL-13 in the pathogenesis of asthma by downregulating FOXA2 and phosphorylating PI3K and AKT, with the resultant effect of reducing cell viability, promoting apoptosis and increasing the secretion of inflammatory chemokines such as CCL11 [151].
Similarly, the microRNA regulation of TGF-β has clinicopathologic significance. In a recent clinical cohort study involving 164 patients, miR-29a and miR-29b expression levels could differentiate cirrhotic from healthy patients, and reduced expression was associated with higher-grade cirrhosis [147]. Importantly, reduced miR-29a and miR-29b expression was also associated with an increased serum concentration of the pathological cytokines IL-6, IGF-1 and TGF-β1, which are known to propagate an epithelial–mesenchymal transition [147]. Interestingly, Guo et al. found that overexpression miR-322-5p ameliorated changes in myocardial enzymes, oxidative stress markers, myocardial apoptosis and myocardial function in a murine model of myocardial infarction [142]. This effect was achieved by inhibiting Smad ubiquitin regulatory factor 2 (Smurf2) expression and thereby downregulating the TGF-β/Smad signalling pathway [142].
Dysregulated intercellular communication also features complex remodelling of the extracellular matrix, which potentiates pathological tissue fibrosis [61]. Here, transcription factors such as WW domain-containing transcription regulator protein 1 (TAZ) and yes-associated protein (YAP) upregulate the expression of profibrotic genes such as lysyl oxidase, which ultimately leads to a resultant secretion of matrix metalloproteinases and corresponding upregulation of connected profibrotic pathways including Notch [61]. A fascinating study employing in silico network analysis methods demonstrated that miR-130/131 family members regulated fibrotic pathways across numerous diseases and tissue beds [152]. Moreover, the authors demonstrated in vivo that the YAP/TAZ-miR-130/301 pathway is implicated in human pulmonary and hepatic diseases, including idiopathic pulmonary fibrosis and nonalcoholic steatohepatitis [152]. Interestingly, inhibiting the miR-130/301 family in murine models led to a downregulation of YAP/TAZ signalling and mitigated extracellular remodelling and disease progression of both pulmonary and liver fibrosis [152]. In a separate study, Zhu et al. showed that miR-582-5p downregulates YAP/TAZ signalling via phosphorylation and inhibiting the PIP5KC1 and NCKAP1 actin regulators in non-small-cell lung cancer cells [143]. This resulted in a tumour-suppressive effect characterised by reduced proliferation, upregulated apoptosis, and changes to the cellular cytoskeleton [143].
Similar findings are seen in nonmalignant chronic diseases. Hu et al. used human umbilical vascular endothelial cells to investigate the effect of oxidised low-density lipoproteins and potentially elucidate pathological mechanisms driving the progression of atherosclerosis [153]. Oxidised low-density lipoproteins upregulated the expression of miR-496 in the endothelial cells whilst downregulating the Hippo-YAP/ZAP pathway [153]. This induced apoptosis and dysfunction of the vascular epithelial cells [153]. Indeed, a separate study found that miR-133b overexpression caused the disease progression of atherosclerosis in a murine model by activating the Notch signalling pathway [144]. This illustrates the clinicopathologic significance of microRNA regulation of this pathway.

5.3.3. Chronic Inflammation

Ageing is associated with a chronic increase in inflammation which has been described as “inflammaging”. This is characterised by systemic factors, such as an increase in circulating inflammatory cytokines including Il-6 and Il-1β, and also features a progressive deterioration in immune function [61]. Chronic inflammation can both be exacerbated by and also itself potentiate the other hallmarks of ageing, which is again indicative of the reciprocal pathways by which these mechanisms cause disease [61].
MicroRNA regulatory pathways have been implicated in the aetiology of immune dysfunction and resultant diseases [154]. Indeed, miR-155 has been identified as an important regulator of inflammatory pathways, with dysregulated expression contributing to the pathogenesis of cancers, pulmonary disorders, multiple sclerosis and numerous other inflammatory autoimmune diseases [155,156,157]. More specifically, Zhao et al. found that miR-210 was overexpressed in CD4+ T cells isolated from patients with psoriasis vulgaris [145]. This was shown to potentiate pathological inflammatory changes in the T-cell function by inhibiting FOXP3 expression [145]. As well as elucidating the potential role of microRNA-mediated immune cell dysfunction in the aetiology of psoriasis, the authors suggested that this could provide a novel therapeutic target [145].
Many studies have demonstrated that microRNA expression levels have a significant relationship with crucial inflammatory cytokines. This is important in the aetiology of malignancies. For example, Guo et al. recently demonstrated that IL-6 promotes the malignant progression and invasion of epithelial ovarian cancer cells by downregulating miR-200c and Let-7c through changes in the STAT3/hypoxia-inducible factor-1α pathway [158]. In a separate study, Pei et al. found that Il-1β upregulated the expression of miR-181a by activating NF-kB [159]. This downregulated PTEN expression and thereby promoted the proliferation of colorectal cancer cells [159]. Similarly, miR-181b overexpression, as induced by IL-1β/NF-κB signalling, was shown to downregulate the expression of PTEN and thereby promote the proliferation of osteosarcoma cells [146].
MicroRNA also mediates the pathological effects of inflammatory cytokines in autoimmune diseases. An interesting network analysis identified numerous microRNAs and corresponding signalling pathways involving IL-1β and chemokine ligand 8, which were implicated in the pathogenesis of psoriasis [160]. Likewise there is emerging evidence linking this relationship with thyroiditis; Li et al. found that increased expression of miR-146a was associated with increased concentrations of IL-17, IL-23 and IL-6 but decreased concentrations of IL-10 in patients with Hashimoto’s thyroiditis [161]. Moreover, increased expression of miR-146a could predict the occurrence of the disease compared to healthy controls [161]. A separate study demonstrated that miR-195-5p modulated the pathological effects of TNF-α in ulcerative colitis by reducing claudin-2 (CLDN2) expression, controlling the structure of the intestinal tight junctions and thereby reducing the permeability of the intestinal epithelial cells [162].
MicroRNA expression and chronic inflammation are similarly implicated in many other chronic diseases. Indeed, reduced expression of miR-29a and miR-29b has been associated with increased concentration of IL-6 and TNF-α and more advanced grades in patients with cirrhosis [147]. Gao et al. found that miR-20a-5p reduced neuronal damage and apoptosis by targeting STAT3 in murine and cellular models of Parkinson’s disease [163]. Furthermore, miR-20a-5p ameliorated associated inflammatory changes, which was indicated by reduced expression of inflammatory cytokines such as IL-6 [163]. Similarly, microRNAs have been shown to potentiate chronic inflammatory changes in diseases such as asthma and many rheumatological conditions, including rheumatoid arthritis, osteoarthritis and intervertebral disc degeneration [164,165,166,167].
Chronic inflammation due to microRNA dysregulation also contributes to the complications of these chronic diseases. Liechty et al. showed that mir-21 is upregulated in diabetic wounds and induces pro-inflammatory macrophages, thus mediating a chronic inflammatory phenotype [168]. Moreover, the authors showed that hyperglycaemia significantly upregulates Mir-21, which inhibits pTEN and positively regulates PI3K and NOX2, thus leading to the formation of reactive oxygen species [168]. In a separate study, Rawal et al. found that the expression of miR-369-3p is reduced in diabetes-associated atherosclerosis [169]. The authors demonstrated that miR-369-3p reduces the metabolic inflammation induced by oxidised low-density lipoproteins and alleviated associated mitochondrial dysfunction; this effect was mediated by downregulating succinate-GPR91 signalling [169]. Interestingly, the succinate receptor (GPR91) expression was increased in patients with coronary artery disease and diabetes, and the authors showed that antagonising GPR91 signalling offers a promising therapeutic strategy [169]. Indeed, Zhang et al. showed that a pathological increase in myocardial infarction-associated transcript (MIAT) mediates neuronal damage, apoptosis and impaired function in ischaemic stroke by downregulating miR-874-3p, which results in increased inflammatory cytokines including IL1B [170].

5.3.4. Dysbiosis

The gut microbiome is an important component of organismal health. The balance and composition of the intestinal flora affects factors such as immune system development and function and response to intestinal infections and also produces bioactive molecules such as short-chain fatty acids [171]. Accordingly, dysregulation of the gut microbiome, or dysbiosis, has been implicated in the aetiology of numerous chronic diseases [61]. Specifically, the ecological diversity, uniqueness of the microbiome and colonisation status of specific organisms are acknowledged to be important clinicopathologic factors [61]. Investigations have demonstrated the efficacy of interventions such as faecal microbiota transplantation and probiotics which target the gut microbiome; this validates the clinical significance of the gut microbiome and shows how it is becoming an innovative therapeutic target [61].
Research has shown that microRNAs are crucial mediators of host–microbiome interactions [171]. Microbiome composition can affect the expression of circulating microRNAs in the host and thus cause chronic diseases including chronic inflammatory conditions and cancers [172]. Interestingly, it is also thought that microRNAs contained within extracellular vesicles may be released by host intestinal cells to regulate the gut microbiome [173]. As such, faecal microRNAs are thought to have an important role in regulating the gut microbiota [172]. Accordingly, the clinicopathologic significance of the interaction between microRNA expression and the microbiome is an area of ongoing research [174]. Studies have shown that this relationship potentiates the pathogenesis of numerous chronic diseases, including metabolic disorders such as obesity, malignancies, irritable bowel syndrome, inflammatory bowel disease and systemic autoimmune diseases [175,176,177,178,179,180].
More specifically, there is a significant link with inflammatory diseases. A fascinating study found that chlorogenic acid alleviates diabetic symptoms and intestinal inflammation in diabetic mice by upregulating microRNA-129-1-3p and microRNA-666-5p [181]. This promoted the intestinal growth of Lactobacillus johnsonii, which mediated these therapeutic effects and demonstrated the clinicopathologic significance of the microRNA–microbiome axis [181]. This effect on local inflammation is naturally important for the development of inflammatory bowel disease. Indeed, Ma et al. demonstrated that pathological changes in the gut microbiota were associated with changes in microRNA expression in a murine model of ulcerative colitis [182]. Moreover, Huang et al. showed that miR-582-3p expression was upregulated in patients with nonalcoholic steatohepatitis [148]. The authors found that inhibiting miR-582-3p attenuated the proliferation of hepatic satellite cells and promoted hepatic satellite cell apoptosis [148]. By downregulating transmembrane BAX inhibitor motif-containing 1 (TMBIM1), miR-582-3p upregulated factors such as α-smooth muscle actin (α-SMA) and Toll-like receptor 4 (TLR4), which are myofibroblast markers, thus potentiating hepatic fibrosis. Crucially, miR-582-3p expression corresponded to changes in gut microbiota, and the authors proposed that intestinal dysbiosis may precipitate dysregulated miR-582-3p expression, thus contributing to the pathogenesis of nonalcoholic steatohepatitis [148].
Similarly, the microRNA–microbiome axis has been implicated in numerous metabolic disorders. Das et al. used a murine model to investigate the clinicopathologic importance of microbiota-associated metabolites [149]. The authors showed that butyrate upregulates ARE/poly(U)-binding/degradation factor-1 (AUF-1) in mice, which inhibits Dicer-1 expression and thus reduces expression of miR-122; this effect ameliorated diet-induced hypercholesterolaemia [149]. Importantly, this butyrate was shown to be produced by the intestinal flora of the mice after the mice were treated with antibiotics and this beneficial effect was removed [149]. Another study showed that miR-29a alleviated hepatic steatosis and inflammatory changes in mice in response to a high-fat diet; overexpression of miR-29a also protected the integrity of the intestinal epithelial barrier from damage induced by the high-fat diet [150]. Interestingly, miR-29a overexpression led to beneficial changes in the intestinal flora which were associated with reduced inflammation and improved lipid metabolism [150].
Innovative studies are further exploring how the microRNA–microbiome axis affects health. Indeed, microRNA are now thought to mediate the pathological effects of intestinal dysbiosis, contributing to the development of neuropsychiatric diseases such as cognitive disturbance, anxiety and depression [183,184,185]. In this way, microRNAs may have a central role in the brain–gut axis. Moreover, there is growing interest in how the microRNA–microbiome axis can be targeted therapeutically, whilst the clinicopathologic significance of faecal microRNAs is also being investigated [186,187].

6. Discussion

Multimorbidity is a very common feature of clinical practice; it is associated with adverse outcomes for patients and significant challenges for healthcare providers. Crucially, patients with multimorbidity are routinely excluded from clinical trials, which limits the clinical applicability of their results. As such, there is an urgent need to develop new biomarkers to assess the prognostic burden of multimorbidity. Whilst microRNAs have been shown to predict common clusters of comorbidities, very few studies have considered how microRNA expression relates to the prognostic burden of multimorbidity. The hallmarks of ageing are important factors potentiating the pathogenesis of multimorbidity, and this study highlights the breadth of research showing how microRNAs mediate the pathological effects of these hallmarks in the aetiology of many chronic diseases. Accordingly, this study proposes that the hallmarks of ageing can be used as a framework to develop a panel of microRNAs to assess the prognostic burden of multimorbidity.
This is a novel approach for prognosticating multimorbidity. Existing molecular strategies have focused on using inflammatory or epigenetic markers to characterise pathological ageing [188,189]. Freitas et al. showed that the expression of microRNAs relating to cardiovascular comorbidities were associated with increased severity of obstructive sleep apnoea, but no attempt was made to correlate microRNA expression with the burden of comorbidity [190]. In a fascinating project, Sayed et al. developed an inflammatory ageing clock which uses inflammatory and immunological markers to characterise the physiological age of patients [188]. This model was associated with multimorbidity as assessed by a simple count of diseases. In this study, however, a comprehensive panel of microRNAs was not used, a detailed assessment of how this model relates to the prognostic burden of multimorbidity was not made and this study only used inflammatory and immunological biomarkers, which may overlook other physiological determinants of ageing and multimorbidity. Iannone et al. found that miR-181a was significantly associated with the burden of multimorbidity, as assessed by the CIRS comorbidity score [191]; the authors only investigated this one microRNA, however, which is known to regulate inflammatory molecules.
A separate study identified a panel of microRNAs which could identify high-risk patients as indicated by the haematopoietic cell transplantation comorbidity index. This study used a haematological-specific comorbidity index which limits the wider applicability of the results, and the authors acknowledged the need for more studies to investigate how microRNA can be used to predict mortality and prognosis [192]. Ultimately, much more research is needed to elucidate the potential use of microRNA in assessing the prognostic burden of multimorbidity. In contrast to these previous studies, the present article proposes adopting a multifaceted mechanistic approach to multimorbidity assessment which uses microRNA to provide a comprehensive molecular assessment of the different hallmarks of ageing to assess prognosis. This may broaden the clinical relevance of the model.
Developing a molecular morbidity score using microRNA has many potential applications. Firstly, by providing a summative molecular assessment of the hallmarks of ageing, it would enable research to further elucidate the aetiology of multimorbidity. Specifically, it could be used to explore how differences in the hallmarks of ageing correspond to different multimorbidity phenotypes. Exploring the mechanisms of multimorbidity is an area of ongoing research, and as well as positing clear academic merit, this has the potential to facilitate innovative new therapeutic strategies which target these mechanisms [8]. Indeed, advancements in theoretical understanding of ageing are currently being used to develop drugs which inhibit ageing processes in order to ameliorate multimorbidity [193]. As well as advancing the theoretical understanding of multimorbidity, a molecular morbidity score could also be used to assess the mechanistic effects of interventions as a composite molecular assessment of health.
Secondly, a molecular morbidity score would have clear clinical benefit, as it would enable precise prognostication of patients with multimorbidity. This could be used to identify clinically significant multimorbidity and target specific interventions for high-risk patients [8]. Furthermore, the proposed score would have the potential to guide clinical decision making, including ceiling-of-care decisions and fitness for surgery. As a more theoretical application, it could also provide molecular validation for other methods of prognostication for patients with multimorbidity, such as the syndrome of complex multimorbidity [194]. Similarly, this score could assess the molecular significance of more general prognostic metrics, such as sarcopenia [195].
Crucially, a molecular morbidity score would enable precise and objective characterisation of the burden of multimorbidity. This could facilitate wider integration of older patients with multimorbidity into clinical trials [23]. For example, the proposed molecular morbidity score could be used to randomise patient inclusion, or it could be included as a potential confounding variable in regression analyses. As such, it is potentially transformative.
Nevertheless, significant challenges must be acknowledged. Firstly, microRNA assessments can be expensive, and so the cost-effectiveness of this approach must be considered. Also, there are wider issues facing microRNA research, including lack of standardisation of research methods, which could potentially encumber the development of this idea [196]. A more detailed discussion of the practicalities of using microRNAs as clinical biomarkers has been outlined elsewhere [196,197].
Moreover, specific microRNAs may have many physiological effects, which vary based on tissue location and specific disease factors [198]. These potential effects may even oppose one another, and how this heterogeneity can be accounted for in the proposed molecular morbidity score should be a focus for further study [198]. Additionally, this article included studies using different species and disease models to illustrate the breadth of research linking microRNA expression with the hallmarks of ageing. Whilst admittedly, many of these microRNAs are known to be conserved between species, the actualisation of the proposed molecular morbidity score will require a systematic approach to developing a panel of suitable microRNAs which are clinically applicable for human patients. In these human patients, it will be necessary to consider how additional clinical factors, such as polypharmacy, may affect microRNA expression and thus confound analyses exploring its association with multimorbidity. This should be the focus of further research. Indeed, there is a clear academic and clinical mandate to explore this novel utility further.
Furthermore, the present study proposes the need for a paradigm shift in how ageing and chronic diseases are considered academically. There is a significant focus in the literature about the pathology of ageing [61]. Indeed, the geroscience hypothesis itself proposes that the mechanisms of ageing propagate the pathogenesis of chronic diseases, and there are many studies focused on developing “anti-ageing” medication [199,200]. The connection between ageing and chronic disease may be seen as an associative rather than causative relationship. Indeed, it is accepted that exogenous and endogenous factors contribute to pathological molecular changes that correspond to the hallmarks of ageing. At a single moment, there is a residual risk of exposure to these factors, and once a pathological change has occurred, there is an increased risk of chronic disease. Over time, by statistical inference, an individual is exposed to more factors and so more pathological changes accrue, which leads to an increased risk of disease.
As such, chronic disease may be described as a function of cumulative risk. This “cumulative risk” theory of chronic disease may be seen as a more mechanistic description for the intrinsic association between ageing and chronic disease. Importantly, it avoids pathologizing the process of getting older, which has the risk of stigmatising the elderly. Indeed, viewing ageing as inherently pathological seems like an overly reductionistic view of the human experience, and, especially from a clinician’s perspective, it belies the dignity of old age. The potential merit of a “cumulative risk” approach to chronic disease is an interesting area for further research.
In conclusion, this study proposes using the hallmarks of ageing as a framework for developing a microRNA panel to assess the prognostic burden of multimorbidity. This epitomises translational research and is a fertile and fascinating area for future study. The proposed molecular morbidity score places microRNAs at the furthest frontiers of the clinical science interface as part of an increasingly personalised and integrative approach to clinical practice.

Author Contributions

T.B.: Conceptualisation, methodology, investigation, writing—original draft, writing—review and editing, visualisation. M.G.D.: methodology, investigation, visualisation, writing—review and editing, supervision. M.J.K.: conceptualisation, methodology, writing—review and editing, visualisation, supervision. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Acknowledgments

The authors would like to gratefully acknowledge the support and encouragement of Nicola Miller during the development of this project.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Ho, I.S.-S.; Azcoaga-Lorenzo, A.; Akbari, A.; Davies, J.; Hodgins, P.; Khunti, K.; Kadam, U.; Lyons, R.; McCowan, C.; Mercer, S.W. Variation in the estimated prevalence of multimorbidity: Systematic review and meta-analysis of 193 international studies. BMJ Open 2022, 12, e057017. [Google Scholar] [CrossRef] [PubMed]
  2. Feinstein, A.R. The pre-therapeutic classification of co-morbidity in chronic disease. J. Chronic Dis. 1970, 23, 455–468. [Google Scholar] [CrossRef] [PubMed]
  3. Charlson, M.E.; Carrozzino, D.; Guidi, J.; Patierno, C. Charlson comorbidity index: A critical review of clinimetric properties. Psychother. Psychosom. 2022, 91, 8–35. [Google Scholar] [CrossRef] [PubMed]
  4. Librero, J.; Peiró, S.; Ordiñana, R. Chronic comorbidity and outcomes of hospital care: Length of stay, mortality, and readmission at 30 and 365 days. J. Clin. Epidemiol. 1999, 52, 171–179. [Google Scholar] [CrossRef] [PubMed]
  5. Chua, Y.P.; Xie, Y.; Lee, P.S.S.; Lee, E.S. Definitions and prevalence of multimorbidity in large database studies: A scoping review. Int. J. Environ. Res. Public Health 2021, 18, 1673. [Google Scholar] [CrossRef] [PubMed]
  6. Harrison, C.; Fortin, M.; van den Akker, M.; Mair, F.; Calderon-Larranaga, A.; Boland, F.; Wallace, E.; Jani, B.; Smith, S. Comorbidity versus Multimorbidity: Why It Matters; SAGE Publications Sage UK: London, UK, 2021; Volume 11, p. 2633556521993993. [Google Scholar]
  7. Banerjee, A.; Hurst, J.; Fottrell, E.; Miranda, J.J. Multimorbidity: Not just for the West. Glob. Heart 2020, 15, 45. [Google Scholar] [CrossRef]
  8. Skou, S.T.; Mair, F.S.; Fortin, M.; Guthrie, B.; Nunes, B.P.; Miranda, J.J.; Boyd, C.M.; Pati, S.; Mtenga, S.; Smith, S.M. Multimorbidity. Nat. Rev. Dis. Primers 2022, 8, 48. [Google Scholar] [CrossRef] [PubMed]
  9. Xiao, X.; Beach, J.; Senthilselvan, A. Prevalence and determinants of multimorbidity in the Canadian population. PLoS ONE 2024, 19, e0297221. [Google Scholar] [CrossRef]
  10. St John, P.D.; Menec, V.; Tyas, S.L.; Tate, R.; Griffith, L. Multimorbidity in Canadians living in the community: Results from the Canadian Longitudinal Study of Aging. Can. Fam. Physician 2021, 67, 187–197. [Google Scholar] [CrossRef]
  11. Kudesia, P.; Salimarouny, B.; Stanley, M.; Fortin, M.; Stewart, M.; Terry, A.; Ryan, B.L. The incidence of multimorbidity and patterns in accumulation of chronic conditions: A systematic review. J. Multimorb. Comorbidity 2021, 11, 26335565211032880. [Google Scholar] [CrossRef]
  12. Asogwa, O.A.; Boateng, D.; Marzà-Florensa, A.; Peters, S.; Levitt, N.; van Olmen, J.; Klipstein-Grobusch, K. Multimorbidity of non-communicable diseases in low-income and middle-income countries: A systematic review and meta-analysis. BMJ Open 2022, 12, e049133. [Google Scholar] [CrossRef] [PubMed]
  13. Williams, J.; Allen, L.; Wickramasinghe, K.; Mikkelsen, B.; Roberts, N.; Townsend, N. A systematic review of associations between non-communicable diseases and socioeconomic status within low-and lower-middle-income countries. J. Glob. Health 2018, 8, 020409. [Google Scholar] [CrossRef] [PubMed]
  14. Nguyen, H.; Manolova, G.; Daskalopoulou, C.; Vitoratou, S.; Prince, M.; Prina, A.M. Prevalence of multimorbidity in community settings: A systematic review and meta-analysis of observational studies. J. Comorbidity 2019, 9, 2235042X19870934. [Google Scholar] [CrossRef] [PubMed]
  15. Soley-Bori, M.; Ashworth, M.; Bisquera, A.; Dodhia, H.; Lynch, R.; Wang, Y.; Fox-Rushby, J. Impact of multimorbidity on healthcare costs and utilisation: A systematic review of the UK literature. Br. J. Gen. Pract. 2021, 71, e39–e46. [Google Scholar] [CrossRef] [PubMed]
  16. Mair, A.; Wilson, M.; Dreischulte, T. Addressing the challenge of polypharmacy. Annu. Rev. Pharmacol. Toxicol. 2020, 60, 661–681. [Google Scholar] [CrossRef] [PubMed]
  17. Sullivan, M.K.; Rankin, A.J.; Jani, B.D.; Mair, F.S.; Mark, P.B. Associations between multimorbidity and adverse clinical outcomes in patients with chronic kidney disease: A systematic review and meta-analysis. BMJ Open 2020, 10, e038401. [Google Scholar] [CrossRef] [PubMed]
  18. Rizzuto, D.; Melis, R.J.; Angleman, S.; Qiu, C.; Marengoni, A. Effect of chronic diseases and multimorbidity on survival and functioning in elderly adults. J. Am. Geriatr. Soc. 2017, 65, 1056–1060. [Google Scholar] [CrossRef] [PubMed]
  19. Vetrano, D.L.; Palmer, K.; Marengoni, A.; Marzetti, E.; Lattanzio, F.; Roller-Wirnsberger, R.; Lopez Samaniego, L.; Rodríguez-Mañas, L.; Bernabei, R.; Onder, G. Frailty and multimorbidity: A systematic review and meta-analysis. J. Gerontol. Ser. A 2019, 74, 659–666. [Google Scholar] [CrossRef] [PubMed]
  20. Kuan, V.; Denaxas, S.; Patalay, P.; Nitsch, D.; Mathur, R.; Gonzalez-Izquierdo, A.; Sofat, R.; Partridge, L.; Roberts, A.; Wong, I.C. Identifying and visualising multimorbidity and comorbidity patterns in patients in the English National Health Service: A population-based study. Lancet Digit. Health 2023, 5, e16–e27. [Google Scholar] [CrossRef]
  21. Guthrie, B.; Payne, K.; Alderson, P.; McMurdo, M.E.; Mercer, S.W. Adapting clinical guidelines to take account of multimorbidity. Bmj 2012, 345, e6341. [Google Scholar] [CrossRef]
  22. Du Vaure, C.B.; Dechartres, A.; Battin, C.; Ravaud, P.; Boutron, I. Exclusion of patients with concomitant chronic conditions in ongoing randomised controlled trials targeting 10 common chronic conditions and registered at ClinicalTrials. gov: A systematic review of registration details. BMJ Open 2016, 6, e012265. [Google Scholar] [CrossRef] [PubMed]
  23. He, J.; Morales, D.R.; Guthrie, B. Exclusion rates in randomized controlled trials of treatments for physical conditions: A systematic review. Trials 2020, 21, 228. [Google Scholar] [CrossRef]
  24. Unger, J.M.; Hershman, D.L.; Fleury, M.E.; Vaidya, R. Association of patient comorbid conditions with cancer clinical trial participation. JAMA Oncol. 2019, 5, 326–333. [Google Scholar] [CrossRef] [PubMed]
  25. Farmer, C.; Fenu, E.; O’Flynn, N.; Guthrie, B. Clinical assessment and management of multimorbidity: Summary of NICE guidance. BMJ 2016, 354, i4843. [Google Scholar] [CrossRef] [PubMed]
  26. Sarfati, D. How do we measure comorbidity? In Cancer and Chronic Conditions: Addressing the Problem of Multimorbidity in Cancer Patients and Survivors; Springer: Berlin/Heidelberg, Germany, 2016; pp. 35–70. [Google Scholar]
  27. Lee, E.S.; Koh, H.L.; Ho, E.Q.-Y.; Teo, S.H.; Wong, F.Y.; Ryan, B.L.; Fortin, M.; Stewart, M. Systematic review on the instruments used for measuring the association of the level of multimorbidity and clinically important outcomes. BMJ Open 2021, 11, e041219. [Google Scholar] [CrossRef] [PubMed]
  28. Smith, S.M.; Wallace, E.; Salisbury, C.; Sasseville, M.; Bayliss, E.; Fortin, M. A core outcome set for multimorbidity research (COSmm). Ann. Fam. Med. 2018, 16, 132–138. [Google Scholar] [CrossRef] [PubMed]
  29. Aslam, F.; Khan, N.A. Tools for the assessment of comorbidity burden in rheumatoid arthritis. Front. Med. 2018, 5, 39. [Google Scholar] [CrossRef]
  30. Niedzwiedz, C.L.; Katikireddi, S.V.; Pell, J.P.; Smith, D.J. Sex differences in the association between salivary telomere length and multimorbidity within the US Health & Retirement Study. Age Ageing 2019, 48, 703–710. [Google Scholar] [PubMed]
  31. Bernabeu-Wittel, M.; Gómez-Díaz, R.; González-Molina, Á.; Vidal-Serrano, S.; Díez-Manglano, J.; Salgado, F.; Soto-Martín, M.; Ollero-Baturone, M.; Researchers, P. Oxidative stress, telomere shortening, and apoptosis associated to sarcopenia and frailty in patients with multimorbidity. J. Clin. Med. 2020, 9, 2669. [Google Scholar] [CrossRef]
  32. Desdín-Micó, G.; Soto-Heredero, G.; Aranda, J.F.; Oller, J.; Carrasco, E.; Gabandé-Rodríguez, E.; Blanco, E.M.; Alfranca, A.; Cussó, L.; Desco, M. T cells with dysfunctional mitochondria induce multimorbidity and premature senescence. Science 2020, 368, 1371–1376. [Google Scholar] [CrossRef]
  33. Hornsby, P.J. The nature of aging and the geroscience hypothesis. In Handbook of the Biology of Aging; Elsevier: Amsterdam, The Netherlands, 2021; pp. 69–76. [Google Scholar]
  34. Fraser, H.C.; Kuan, V.; Johnen, R.; Zwierzyna, M.; Hingorani, A.D.; Beyer, A.; Partridge, L. Biological mechanisms of aging predict age-related disease co-occurrence in patients. Aging Cell 2022, 21, e13524. [Google Scholar] [CrossRef]
  35. Fabbri, E.; An, Y.; Zoli, M.; Simonsick, E.M.; Guralnik, J.M.; Bandinelli, S.; Boyd, C.M.; Ferrucci, L. Aging and the burden of multimorbidity: Associations with inflammatory and anabolic hormonal biomarkers. J. Gerontol. Ser. A Biomed. Sci. Med. Sci. 2015, 70, 63–70. [Google Scholar] [CrossRef]
  36. Wessman, T.; Tofik, R.; Ruge, T.; Melander, O. Associations between biomarkers of multimorbidity burden and mortality risk among patients with acute dyspnea. Intern. Emerg. Med. 2022, 17, 559–567. [Google Scholar] [CrossRef]
  37. Vázquez-Fernández, A.; Lana, A.; Struijk, E.A.; Vega-Cabello, V.; Cárdenas-Valladolid, J.; Salinero-Fort, M.Á.; Rodríguez-Artalejo, F.; Lopez-Garcia, E.; Caballero, F.F. Cross-sectional association between plasma biomarkers and multimorbidity patterns in older adults. J. Gerontol. Ser. A 2024, 79, glad249. [Google Scholar] [CrossRef] [PubMed]
  38. Ferreira, G.D.; Simoes, J.A.; Senaratna, C.; Pati, S.; Timm, P.F.; Batista, S.R.; Nunes, B.P. Physiological markers and multimorbidity: A systematic review. J. Comorbidity 2018, 8, 2235042X18806986. [Google Scholar] [CrossRef]
  39. Peters, L.J.; Floege, J.; Biessen, E.A.; Jankowski, J.; van der Vorst, E.P. MicroRNAs in chronic kidney disease: Four candidates for clinical application. Int. J. Mol. Sci. 2020, 21, 6547. [Google Scholar] [CrossRef] [PubMed]
  40. Davey, M.G.; Feeney, G.; Annuk, H.; Paganga, M.; Holian, E.; Lowery, A.J.; Kerin, M.J.; Miller, N. Identification of a Five-MiRNA Expression Assay to Aid Colorectal Cancer Diagnosis. Gastrointest. Disord. 2022, 4, 190–204. [Google Scholar] [CrossRef]
  41. Davey, M.G.; McGuire, A.; Casey, M.C.; Waldron, R.M.; Paganga, M.; Holian, E.; Newell, J.; Heneghan, H.M.; McDermott, A.M.; Keane, M.M. Evaluating the role of circulating microRNAs in predicting long-term survival outcomes in breast cancer: A prospective, multicenter clinical trial. J. Am. Coll. Surg. 2023, 236, 317–327. [Google Scholar] [CrossRef]
  42. Bouz Mkabaah, L.; Davey, M.G.; Lennon, J.C.; Bouz, G.; Miller, N.; Kerin, M.J. Assessing the Role of MicroRNAs in Predicting Breast Cancer Recurrence—A Systematic Review. Int. J. Mol. Sci. 2023, 24, 7115. [Google Scholar] [CrossRef]
  43. Davey, M.G.; Abbas, R.; Kerin, E.P.; Casey, M.C.; McGuire, A.; Waldron, R.M.; Heneghan, H.M.; Newell, J.; McDermott, A.M.; Keane, M.M. Circulating microRNAs can predict chemotherapy-induced toxicities in patients being treated for primary breast cancer. Breast Cancer Res. Treat. 2023, 202, 73–81. [Google Scholar] [CrossRef]
  44. Condrat, C.E.; Thompson, D.C.; Barbu, M.G.; Bugnar, O.L.; Boboc, A.; Cretoiu, D.; Suciu, N.; Cretoiu, S.M.; Voinea, S.C. miRNAs as biomarkers in disease: Latest findings regarding their role in diagnosis and prognosis. Cells 2020, 9, 276. [Google Scholar] [CrossRef] [PubMed]
  45. Pouladi, N.; Li, H.; Berghout, J.; Kenost, C.; Gonzalez-Garay, M.; Lussier, Y. Biomechanisms of comorbidity: Reviewing integrative analyses of multi-omics datasets and electronic health records. Yearb. Med. Inform. 2016, 25, 194–206. [Google Scholar] [CrossRef] [PubMed]
  46. Liu, C.-C.; Tseng, Y.-T.; Li, W.; Wu, C.-Y.; Mayzus, I.; Rzhetsky, A.; Sun, F.; Waterman, M.; Chen, J.J.; Chaudhary, P.M. DiseaseConnect: A comprehensive web server for mechanism-based disease–disease connections. Nucleic Acids Res. 2014, 42, W137–W146. [Google Scholar] [CrossRef] [PubMed]
  47. Jin, S.; Zeng, X.; Fang, J.; Lin, J.; Chan, S.Y.; Erzurum, S.C.; Cheng, F. A network-based approach to uncover microRNA-mediated disease comorbidities and potential pathobiological implications. NPJ Syst. Biol. Appl. 2019, 5, 41. [Google Scholar] [CrossRef]
  48. Morselli Gysi, D.; Barabási, A.-L. Noncoding RNAs improve the predictive power of network medicine. Proc. Natl. Acad. Sci. USA 2023, 120, e2301342120. [Google Scholar] [CrossRef] [PubMed]
  49. Tanase, D.M.; Gosav, E.M.; Petrov, D.; Teodorescu, D.-S.; Buliga-Finis, O.N.; Ouatu, A.; Tudorancea, I.; Rezus, E.; Rezus, C. MicroRNAs (miRNAs) in cardiovascular complications of rheumatoid arthritis (RA): What is new? Int. J. Mol. Sci. 2022, 23, 5254. [Google Scholar] [CrossRef]
  50. Lopez-Pedrera, C.; Barbarroja, N.; Patiño-Trives, A.M.; Luque-Tévar, M.; Torres-Granados, C.; Aguirre-Zamorano, M.A.; Collantes-Estevez, E.; Pérez-Sánchez, C. Role of microRNAs in the development of cardiovascular disease in systemic autoimmune disorders. Int. J. Mol. Sci. 2020, 21, 2012. [Google Scholar] [CrossRef]
  51. Xian, N.; Bai, R.; Guo, J.; Luo, R.; Lei, H.; Wang, B.; Zheng, Y. Bioinformatics analysis to reveal the potential comorbidity mechanism in psoriasis and nonalcoholic steatohepatitis. Ski. Res. Technol. 2023, 29, e13457. [Google Scholar] [CrossRef]
  52. Lei, H.; Chen, X.; Wang, Z.; Xing, Z.; Du, W.; Bai, R.; He, K.; Zhang, W.; Wang, Y.; Zheng, Y. Exploration of the underlying comorbidity mechanism in psoriasis and periodontitis: A bioinformatics analysis. Hereditas 2023, 160, 7. [Google Scholar] [CrossRef]
  53. Reid, L.V.; Spalluto, C.M.; Watson, A.; Staples, K.J.; Wilkinson, T. The role of extracellular vesicles as a shared disease mechanism contributing to multimorbidity in patients with COPD. Front. Immunol. 2021, 12, 754004. [Google Scholar] [CrossRef]
  54. Duman, E.T.; Tuna, G.; Ak, E.; Avsar, G.; Pir, P. Optimized network based natural language processing approach to reveal disease comorbidities in COVID-19. Sci. Rep. 2024, 14, 2325. [Google Scholar] [CrossRef] [PubMed]
  55. Chen, F.; Xu, Y.; Shi, K.; Zhang, Z.; Xie, Z.; Wu, H.; Ma, Y.; Zhou, Y.; Chen, C.; Yang, J. Multi-omics study reveals associations among neurotransmitter, extracellular vesicle-derived microRNA and psychiatric comorbidities during heroin and methamphetamine withdrawal. Biomed. Pharmacother. 2022, 155, 113685. [Google Scholar] [CrossRef] [PubMed]
  56. Chen, Y.-H.; Wang, H. The association between depression and gastroesophageal reflux based on phylogenetic analysis of miRNA biomarkers. Curr. Med. Chem. 2020, 27, 6536–6547. [Google Scholar] [CrossRef] [PubMed]
  57. Naguib, M.; Magdy, M.; Yousef, O.A.E.; Ibrahim, W.; Gharib, D.M. Circulating MicroRNA-30a, Beclin1 and Their Association with Different Variables in Females with Metabolically Healthy/Unhealthy Obesity. Diabetes Metab. Syndr. Obes. 2023, 16, 3065–3074. [Google Scholar] [CrossRef] [PubMed]
  58. Ghafouri-Fard, S.; Taheri, M. The expression profile and role of non-coding RNAs in obesity. Eur. J. Pharmacol. 2021, 892, 173809. [Google Scholar] [CrossRef] [PubMed]
  59. Hutny, M.; Hofman, J.; Zachurzok, A.; Matusik, P. MicroRNAs as the promising markers of comorbidities in childhood obesity—A systematic review. Pediatr. Obes. 2022, 17, e12880. [Google Scholar] [CrossRef] [PubMed]
  60. López-Otín, C.; Blasco, M.A.; Partridge, L.; Serrano, M.; Kroemer, G. The hallmarks of aging. Cell 2013, 153, 1194–1217. [Google Scholar] [CrossRef] [PubMed]
  61. López-Otín, C.; Blasco, M.A.; Partridge, L.; Serrano, M.; Kroemer, G. Hallmarks of aging: An expanding universe. Cell 2023, 186, 243–278. [Google Scholar] [CrossRef] [PubMed]
  62. Lettieri-Barbato, D.; Aquilano, K.; Punziano, C.; Minopoli, G.; Faraonio, R. MicroRNAs, long non-coding RNAs, and circular RNAs in the redox control of cell senescence. Antioxidants 2022, 11, 480. [Google Scholar] [CrossRef]
  63. Victoria, B.; Lopez, Y.O.N.; Masternak, M.M. MicroRNAs and the metabolic hallmarks of aging. Mol. Cell. Endocrinol. 2017, 455, 131–147. [Google Scholar] [CrossRef]
  64. Cardoso, A.P.F.; Banerjee, M.; Nail, A.N.; Lykoudi, A.; States, J.C. miRNA dysregulation is an emerging modulator of genomic instability. Semin. Cancer Biol. 2021, 76, 120–131. [Google Scholar] [CrossRef]
  65. Lämmerhirt, L.; Kappelmann-Fenzl, M.; Fischer, S.; Meier, P.; Staebler, S.; Kuphal, S.; Bosserhoff, A.-K. Loss of miR-101-3p in melanoma stabilizes genomic integrity, leading to cell death prevention. Cell. Mol. Biol. Lett. 2024, 29, 29. [Google Scholar] [CrossRef] [PubMed]
  66. Wang, H.; Liang, Y.; Zhao, L.; Deng, J.; Li, Y.; Zhao, H.; Zhang, X.; Zou, F. miR-653-3p promotes genomic instability of colorectal cancer cells via targeting SIRT1/TWIST1 signaling pathway. Biochim. Et Biophys. Acta (BBA)-Mol. Basis Dis. 2023, 1869, 166821. [Google Scholar] [CrossRef] [PubMed]
  67. Adamowicz, M.; Stukan, I.; Milkiewicz, P.; Bialek, A.; Milkiewicz, M.; Kempinska-Podhorodecka, A. Modulation of mismatch repair and the SOCS1/p53 axis by microRNA-155 in the colon of patients with primary sclerosing cholangitis. Int. J. Mol. Sci. 2022, 23, 4905. [Google Scholar] [CrossRef] [PubMed]
  68. Mo, Y.; Zhang, Y.; Zhang, Y.; Yuan, J.; Mo, L.; Zhang, Q. Nickel nanoparticle-induced cell transformation: Involvement of DNA damage and DNA repair defect through HIF-1α/miR-210/Rad52 pathway. J. Nanobiotechnology 2021, 19, 370. [Google Scholar] [CrossRef] [PubMed]
  69. Sudhanva, M.S.; Hariharasudhan, G.; Jun, S.; Seo, G.; Kamalakannan, R.; Kim, H.H.; Lee, J.-H. MicroRNA-145 impairs classical non-homologous end-joining in response to ionizing radiation-induced DNA double-strand breaks via targeting DNA-PKcs. Cells 2022, 11, 1509. [Google Scholar] [CrossRef] [PubMed]
  70. Zhang, Y.; Chen, Y.; Liu, T. Genomic instability-associated two-miRNA signature as a novel prognostic biomarker in breast cancer. J. Gene Med. 2024, 26, e3604. [Google Scholar] [CrossRef]
  71. Xu, J.; Song, J.; Chen, X.; Huang, Y.; You, T.; Zhu, C.; Shen, X.; Zhao, Y. Genomic instability-related twelve-microRNA signatures for predicting the prognosis of gastric cancer. Comput. Biol. Med. 2023, 155, 106598. [Google Scholar] [CrossRef]
  72. Li, X.; Zhang, J.; Yang, Y.; Wu, Q.; Ning, H. MicroRNA-340-5p increases telomere length by targeting telomere protein POT1 to improve Alzheimer’s disease in mice. Cell Biol. Int. 2021, 45, 1306–1315. [Google Scholar] [CrossRef] [PubMed]
  73. Yan, Y.; Qin, D.; Hu, B.; Zhang, C.; Liu, S.; Wu, D.; Huang, W.; Huang, X.; Wang, L.; Chen, X. Deletion of miR-126a promotes hepatic aging and inflammation in a mouse model of cholestasis. Mol. Ther. Nucleic Acids 2019, 16, 494–504. [Google Scholar] [CrossRef]
  74. Li, T.; Luo, Z.; Lin, S.; Li, C.; Dai, S.; Wang, H.; Huang, J.; Ma, W.; Songyang, Z.; Huang, Y. MiR-185 targets POT1 to induce telomere dysfunction and cellular senescence. Aging 2020, 12, 14791. [Google Scholar] [CrossRef] [PubMed]
  75. Hanoun, N.; Delpu, Y.; Suriawinata, A.A.; Bournet, B.; Bureau, C.; Selves, J.; Tsongalis, G.J.; Dufresne, M.; Buscail, L.; Cordelier, P. The silencing of microRNA 148a production by DNA hypermethylation is an early event in pancreatic carcinogenesis. Clin. Chem. 2010, 56, 1107–1118. [Google Scholar] [CrossRef]
  76. Rosas-Alonso, R.; Galera, R.; Sánchez-Pascuala, J.J.; Casitas, R.; Burdiel, M.; Martínez-Cerón, E.; Vera, O.; Rodriguez-Antolin, C.; Pernía, O.; De Castro, J. Hypermethylation of anti-oncogenic microRNA 7 is increased in emphysema patients. Arch. Bronconeumol. 2020, 56, 506–513. [Google Scholar] [CrossRef] [PubMed]
  77. Zhu, Y.; Li, K.; Yan, L.; He, Y.; Wang, L.; Sheng, L. miR-223-3p promotes cell proliferation and invasion by targeting Arid1a in gastric cancer. Acta Biochim. Biophys. Sin. 2020, 52, 150–159. [Google Scholar] [CrossRef] [PubMed]
  78. Srinivasan, A.R.; Tran, T.T.; Bonini, N.M. Loss of miR-34 in Drosophila dysregulates protein translation and protein turnover in the aging brain. Aging Cell 2022, 21, e13559. [Google Scholar] [CrossRef] [PubMed]
  79. Nissan, X.; Blondel, S.; Navarro, C.; Maury, Y.; Denis, C.; Girard, M.; Martinat, C.; De Sandre-Giovannoli, A.; Levy, N.; Peschanski, M. Unique preservation of neural cells in Hutchinson-Gilford progeria syndrome is due to the expression of the neural-specific miR-9 microRNA. Cell Rep. 2012, 2, 1–9. [Google Scholar] [CrossRef] [PubMed]
  80. Fields, C.J.; Li, L.; Hiers, N.M.; Li, T.; Sheng, P.; Huda, T.; Shan, J.; Gay, L.; Gu, T.; Bian, J. Sequencing of Argonaute-bound microRNA/mRNA hybrids reveals regulation of the unfolded protein response by microRNA-320a. PLoS Genet. 2021, 17, e1009934. [Google Scholar]
  81. Ahangari, F.; Price, N.L.; Malik, S.; Chioccioli, M.; Bärnthaler, T.; Adams, T.S.; Kim, J.; Pradeep, S.P.; Ding, S.; Cosmos, C., Jr. microRNA-33 deficiency in macrophages enhances autophagy, improves mitochondrial homeostasis, and protects against lung fibrosis. JCI Insight 2023, 8, e158100. [Google Scholar] [CrossRef] [PubMed]
  82. Spirina, L.V.; Kovaleva, I.V.; Chizhevskaya, S.Y.; Chebodaeva, A.V.; Tarasenko, N.V. Autophagy-Related MicroRNA: Tumor miR-125b and Thyroid Cancers. Genes 2023, 14, 685. [Google Scholar] [CrossRef]
  83. Ning, S.; Zhang, S.; Guo, Z. MicroRNA-494 regulates high glucose-induced cardiomyocyte apoptosis and autophagy by PI3K/AKT/mTOR signalling pathway. ESC Heart Fail. 2023, 10, 1401–1411. [Google Scholar] [CrossRef]
  84. Baer, C.; Claus, R.; Frenzel, L.P.; Zucknick, M.; Park, Y.J.; Gu, L.; Weichenhan, D.; Fischer, M.; Pallasch, C.P.; Herpel, E. Extensive promoter DNA hypermethylation and hypomethylation is associated with aberrant microRNA expression in chronic lymphocytic leukemia. Cancer Res. 2012, 72, 3775–3785. [Google Scholar] [CrossRef] [PubMed]
  85. Ma, Y.; Chai, N.; Jiang, Q.; Chang, Z.; Chai, Y.; Li, X.; Sun, H.; Hou, J.; Linghu, E. DNA methyltransferase mediates the hypermethylation of the microRNA 34a promoter and enhances the resistance of patient-derived pancreatic cancer cells to molecular targeting agents. Pharmacol. Res. 2020, 160, 105071. [Google Scholar] [CrossRef] [PubMed]
  86. Bure, I.V.; Nemtsova, M.V. Mutual regulation of ncRNAs and chromatin remodeling complexes in normal and pathological conditions. Int. J. Mol. Sci. 2023, 24, 7848. [Google Scholar] [CrossRef] [PubMed]
  87. Wang, H.; Tan, L.; Dong, X.; Liu, L.; Jiang, Q.; Li, H.; Shi, J.; Yang, X.; Dai, X.; Qian, Z. MiR-146b-5p suppresses the malignancy of GSC/MSC fusion cells by targeting SMARCA5. Aging 2020, 12, 13647. [Google Scholar] [CrossRef] [PubMed]
  88. Shcherbakov, D.; Nigri, M.; Akbergenov, R.; Brilkova, M.; Mantovani, M.; Petit, P.I.; Grimm, A.; Karol, A.A.; Teo, Y.; Sanchón, A.C. Premature aging in mice with error-prone protein synthesis. Sci. Adv. 2022, 8, eabl9051. [Google Scholar] [CrossRef] [PubMed]
  89. Finger, F.; Ottens, F.; Springhorn, A.; Drexel, T.; Proksch, L.; Metz, S.; Cochella, L.; Hoppe, T. Olfaction regulates organismal proteostasis and longevity via microRNA-dependent signalling. Nat. Metab. 2019, 1, 350–359. [Google Scholar] [CrossRef] [PubMed]
  90. Lee, Y.-K.; Jiang, Y.; Ran, X.-R.; Lau, Y.-M.; Ng, K.-M.; Lai, W.-H.K.; Siu, C.-W.; Tse, H.-F. Recent advances in animal and human pluripotent stem cell modeling of cardiac laminopathy. Stem Cell Res. Ther. 2016, 7, 139. [Google Scholar] [CrossRef] [PubMed]
  91. Zhou, Y.; Pang, B.; Xiao, Y.; Zhou, S.; He, B.; Zhang, F.; Liu, W.; Peng, H.; Li, P. The protective microRNA-199a-5p-mediated unfolded protein response in hypoxic cardiomyocytes is regulated by STAT3 pathway. J. Physiol. Biochem. 2019, 75, 73–81. [Google Scholar] [CrossRef] [PubMed]
  92. Azizi, M.; Salehi-Mazandarani, S.; Nikpour, P.; Andalib, A.; Rezaei, M. The role of unfolded protein response-associated miRNAs in immunogenic cell death amplification: A literature review and bioinformatics analysis. Life Sci. 2023, 314, 121341. [Google Scholar] [CrossRef]
  93. Choi, S.H.; Cho, K. LAMP2A-mediated autophagy involved in Huntington’s disease progression. Biochem. Biophys. Res. Commun. 2021, 534, 561–567. [Google Scholar] [CrossRef]
  94. Sermersheim, M.A.; Park, K.H.; Gumpper, K.; Adesanya, T.A.; Song, K.; Tan, T.; Ren, X.; Yang, J.-M.; Zhu, H. MicroRNA regulation of autophagy in cardiovascular disease. Front. Biosci. (Landmark Ed.) 2017, 22, 48. [Google Scholar]
  95. Ma, X.; Zheng, Q.; Zhao, G.; Yuan, W.; Liu, W. Regulation of cellular senescence by microRNAs. Mech. Ageing Dev. 2020, 189, 111264. [Google Scholar] [CrossRef] [PubMed]
  96. Hui, K.; Dong, C.; Hu, C.; Li, J.; Yan, D.; Jiang, X. VEGFR affects miR-3200-3p-mediated regulatory T cell senescence in tumour-derived exosomes in non-small cell lung cancer. Funct. Integr. Genom. 2024, 24, 31. [Google Scholar] [CrossRef] [PubMed]
  97. Dong, X.; Hu, X.; Chen, J.; Hu, D.; Chen, L.-F. BRD4 regulates cellular senescence in gastric cancer cells via E2F/miR-106b/p21 axis. Cell Death Dis. 2018, 9, 203. [Google Scholar] [CrossRef]
  98. Lu, F.; Yao, L.-P.; Gao, D.-D.; Alinejad, T.; Jiang, X.-Q.; Wu, Q.; Zhai, Q.-C.; Liu, M.; Zhu, S.-M.; Qian, M.-X. MicroRNA-377-3p exacerbates chronic obstructive pulmonary disease through suppressing ZFP36L1 expression and inducing lung fibroblast senescence. Respir. Res. 2024, 25, 67. [Google Scholar] [CrossRef] [PubMed]
  99. Zhao, L.; Tang, P.; Lin, Y.; Du, M.; Li, H.; Jiang, L.; Xu, H.; Sun, H.; Han, J.; Sun, Z. MiR-203 improves cardiac dysfunction by targeting PARP1-NAD+ axis in aging murine. Aging Cell 2024, 23, e14063. [Google Scholar] [CrossRef] [PubMed]
  100. Rochín-Hernández, L.J.; Rochín-Hernández, L.S.; Padilla-Cristerna, M.L.; Duarte-García, A.; Jiménez-Acosta, M.A.; Figueroa-Corona, M.P.; Meraz-Ríos, M.A. Mesenchymal Stem Cells from Familial Alzheimer’s Patients Express MicroRNA Differently. Int. J. Mol. Sci. 2024, 25, 1580. [Google Scholar] [CrossRef] [PubMed]
  101. Zhang, M.; Xue, X.; Lou, Z.; Lin, Y.; Li, Q.; Huang, C. Exosomes from senescent epithelial cells activate pulmonary fibroblasts via the miR-217-5p/Sirt1 axis in paraquat-induced pulmonary fibrosis. J. Transl. Med. 2024, 22, 310. [Google Scholar] [CrossRef] [PubMed]
  102. Xu, L.; Zhang, Y.; Yu, J.; Huo, W.; Xu, J.; Yang, H.; Zhang, M.; Yu, S.; Wu, Y.; Wang, M. miR-708-5p deficiency involves the degeneration of mandibular condylar chondrocytes via the TLR4/NF-κB pathway. Osteoarthr. Cartil. 2024, 32, 666–679. [Google Scholar] [CrossRef]
  103. Liang, L.; Stone, R.C.; Stojadinovic, O.; Ramirez, H.; Pastar, I.; Maione, A.G.; Smith, A.; Yanez, V.; Veves, A.; Kirsner, R.S. Integrative analysis of miRNA and mRNA paired expression profiling of primary fibroblast derived from diabetic foot ulcers reveals multiple impaired cellular functions. Wound Repair Regen. 2016, 24, 943–953. [Google Scholar] [CrossRef]
  104. Wei, Q.; Su, J.; Meng, S.; Wang, Y.; Ma, K.; Li, B.; Chu, Z.; Huang, Q.; Hu, W.; Wang, Z. MiR-17-5p-engineered sEVs Encapsulated in GelMA Hydrogel Facilitated Diabetic Wound Healing by Targeting PTEN and p21. Adv. Sci. 2024, 11, 2307761. [Google Scholar] [CrossRef]
  105. Liu, X.; Cui, H.; Bai, Q.; Piao, H.; Song, Y.; Yan, G. miR-128-3p alleviates airway inflammation in asthma by targeting SIX1 to regulate mitochondrial fission and fusion. Int. Immunopharmacol. 2024, 130, 111703. [Google Scholar] [CrossRef]
  106. Zhu, J.; Wang, Q.; Zheng, Z.; Ma, L.; Guo, J.; Shi, H.; Ying, R.; Gao, B.; Chen, S.; Yu, S. MiR-181a protects the heart against myocardial infarction by regulating mitochondrial fission via targeting programmed cell death protein 4. Sci. Rep. 2024, 14, 6638. [Google Scholar] [CrossRef]
  107. Wang, T.; Chen, Y.; Liu, Z.; Zhou, J.; Li, N.; Shan, Y.; He, Y. Long noncoding RNA Glis2 regulates podocyte mitochondrial dysfunction and apoptosis in diabetic nephropathy via sponging miR-328-5p. J. Cell. Mol. Med. 2024, 28, e18204. [Google Scholar] [CrossRef]
  108. Yin, Y.; Wang, X.; Li, T.; Ren, Q.; Li, L.; Sun, X.; Zhang, B.; Wang, X.; Han, H.; He, Y. MicroRNA-221 promotes breast cancer resistance to adriamycin via modulation of PTEN/Akt/mTOR signaling. Cancer Med. 2020, 9, 1544–1552. [Google Scholar] [CrossRef]
  109. Chen, X.; Cao, Y.; Guo, Y.; Liu, J.; Ye, X.; Li, H.; Zhang, L.; Feng, W.; Xian, S.; Yang, Z. microRNA-125b-1-3p mediates autophagy via the RRAGD/mTOR/ULK1 signaling pathway and mitigates atherosclerosis progression. Cell. Signal. 2024, 118, 111136. [Google Scholar] [CrossRef]
  110. Liu, X.-L.; Pan, Q.; Cao, H.-X.; Xin, F.-Z.; Zhao, Z.-H.; Yang, R.-X.; Zeng, J.; Zhou, H.; Fan, J.-G. Lipotoxic hepatocyte-derived exosomal miR-192–5p activates macrophages via Rictor/Akt/FoxO1 signaling in NAFLD. Hepatology (Baltim. Md.) 2020, 72, 454. [Google Scholar] [CrossRef]
  111. Maldonado, E.; Morales-Pison, S.; Urbina, F.; Solari, A. Aging hallmarks and the role of oxidative stress. Antioxidants 2023, 12, 651. [Google Scholar] [CrossRef]
  112. Li, X.; Han, Y.; Meng, Y.; Yin, L. Small RNA-big impact: Exosomal miRNAs in mitochondrial dysfunction in various diseases. RNA Biol. 2024, 21, 1–20. [Google Scholar] [CrossRef]
  113. Cheng, M.-H.; Kuo, H.-F.; Chang, C.-Y.; Chang, J.-C.; Liu, I.-F.; Hsieh, C.-C.; Hsu, C.-H.; Li, C.-Y.; Wang, S.-C.; Chen, Y.-H. Curcumin regulates pulmonary extracellular matrix remodeling and mitochondrial function to attenuate pulmonary fibrosis by regulating the miR-29a-3p/DNMT3A axis. Biomed. Pharmacother. 2024, 174, 116572. [Google Scholar] [CrossRef]
  114. Geng, J.; Feng, J.; Ke, F.; Fang, F.; Jing, X.; Tang, J.; Fang, C.; Zhang, B. MicroRNA-124 negatively regulates STAT3 to alleviate hypoxic-ischemic brain damage by inhibiting oxidative stress. Aging 2024, 16, 2828. [Google Scholar] [CrossRef]
  115. Jin, J.; Shang, Y.; Zheng, S.; Dai, L.; Tang, J.; Bian, X.; He, Q. Exosomes as nanostructures deliver miR-204 in alleviation of mitochondrial dysfunction in diabetic nephropathy through suppressing methyltransferase-like 7A-mediated CIDEC N6-methyladenosine methylation. Aging 2024, 16, 3302. [Google Scholar] [CrossRef]
  116. Fu, W.; Wu, G. Targeting mTOR for anti-aging and anti-cancer therapy. Molecules 2023, 28, 3157. [Google Scholar] [CrossRef]
  117. Nazari, N.; Jafari, F.; Ghalamfarsa, G.; Hadinia, A.; Atapour, A.; Ahmadi, M.; Dolati, S.; Rostamzadeh, D. The emerging role of microRNA in regulating the mTOR signaling pathway in immune and inflammatory responses. Immunol. Cell Biol. 2021, 99, 814–832. [Google Scholar] [CrossRef]
  118. Zhang, Y.; Zhang, H.; Liu, Z. MicroRNA-147 suppresses proliferation, invasion and migration through the AKT/mTOR signaling pathway in breast cancer. Oncol. Lett. 2016, 11, 405–410. [Google Scholar] [CrossRef]
  119. Shen, H.; Jin, J.; Wang, H.; Yu, N.; Liu, T.; Sheng, H.; Wan, Z.; Feng, C.; Huang, Y.; Gao, F. Integrated analysis identifies microRNA-188-5p as a suppressor of AKT/mTOR pathway in renal cancer. Cancer Sci. 2023, 114, 3128–3143. [Google Scholar] [CrossRef]
  120. Niture, S.; Tricoli, L.; Qi, Q.; Gadi, S.; Hayes, K.; Kumar, D. MicroRNA-99b-5p targets mTOR/AR axis, induces autophagy and inhibits prostate cancer cell proliferation. Tumor Biol. 2022, 44, 107–127. [Google Scholar] [CrossRef]
  121. Wang, Y.; Liu, J.; Liu, C.; Naji, A.; Stoffers, D.A. MicroRNA-7 regulates the mTOR pathway and proliferation in adult pancreatic β-cells. Diabetes 2013, 62, 887–895. [Google Scholar] [CrossRef]
  122. Lei, Y.; Wang, Q.-L.; Shen, L.; Tao, Y.-Y.; Liu, C.-H. MicroRNA-101 suppresses liver fibrosis by downregulating PI3K/Akt/mTOR signaling pathway. Clin. Res. Hepatol. Gastroenterol. 2019, 43, 575–584. [Google Scholar] [CrossRef]
  123. Duan, W.; Chen, Y.; Wang, X.R. MicroRNA-155 contributes to the occurrence of epilepsy through the PI3K/Akt/mTOR signaling pathway. Int. J. Mol. Med. 2018, 42, 1577–1584. [Google Scholar] [CrossRef]
  124. Wei, Y.-Y.; Ren, T.-L. MicroRNA-30a-3p inhibits malignant progression of hepatocellular carcinoma through regulating IGF1. Eur. Rev. Med. Pharmacol. Sci. 2020, 24, 12144–12152. [Google Scholar]
  125. Li, K.; Sun, Y.; Liu, S.; Zhou, Y.; Qu, Q.; Wang, G.; Wang, J.; Chen, R.; Fan, Z.; Liu, B. The AR/miR-221/IGF-1 pathway mediates the pathogenesis of androgenetic alopecia. Int. J. Biol. Sci. 2023, 19, 3307. [Google Scholar] [CrossRef]
  126. Sarma, S.; Sockalingam, S.; Dash, S. Obesity as a multisystem disease: Trends in obesity rates and obesity-related complications. Diabetes Obes. Metab. 2021, 23, 3–16. [Google Scholar] [CrossRef]
  127. Taylor, E.B. The complex role of adipokines in obesity, inflammation, and autoimmunity. Clin. Sci. 2021, 135, 731–752. [Google Scholar] [CrossRef]
  128. Pan, Y.; Hui, X.; Hoo, R.L.C.; Ye, D.; Chan, C.Y.C.; Feng, T.; Wang, Y.; Lam, K.S.L.; Xu, A. Adipocyte-secreted exosomal microRNA-34a inhibits M2 macrophage polarization to promote obesity-induced adipose inflammation. J. Clin. Investig. 2019, 129, 834–849. [Google Scholar] [CrossRef] [PubMed]
  129. Saini, S.K.; Singh, A.; Saini, M.; Gonzalez-Freire, M.; Leeuwenburgh, C.; Anton, S.D. Time-restricted eating regimen differentially affects circulatory miRNA expression in older overweight adults. Nutrients 2022, 14, 1843. [Google Scholar] [CrossRef]
  130. Yu, K.-R.; Lee, S.; Jung, J.-W.; Hong, I.-S.; Kim, H.-S.; Seo, Y.; Shin, T.-H.; Kang, K.-S. MicroRNA-141-3p plays a role in human mesenchymal stem cell aging by directly targeting ZMPSTE24. J. Cell Sci. 2013, 126, 5422–5431. [Google Scholar] [CrossRef]
  131. Jani, P.K.; Petkau, G.; Kawano, Y.; Klemm, U.; Guerra, G.M.; Heinz, G.A.; Heinrich, F.; Durek, P.; Mashreghi, M.-F.; Melchers, F. The miR-221/222 cluster regulates hematopoietic stem cell quiescence and multipotency by suppressing both Fos/AP-1/IEG pathway activation and stress-like differentiation to granulocytes. PLoS Biol. 2023, 21, e3002015. [Google Scholar] [CrossRef]
  132. Zhao, J.L.; Rao, D.S.; O’Connell, R.M.; Garcia-Flores, Y.; Baltimore, D. MicroRNA-146a acts as a guardian of the quality and longevity of hematopoietic stem cells in mice. eLife 2013, 2, e00537. [Google Scholar] [CrossRef]
  133. Su, Y.; Yu, Y.; Liu, C.; Zhang, Y.; Liu, C.; Ge, M.; Li, L.; Lan, M.; Wang, T.; Li, M. Fate decision of satellite cell differentiation and self-renewal by miR-31-IL34 axis. Cell Death Differ. 2020, 27, 949–965. [Google Scholar] [CrossRef] [PubMed]
  134. Zhang, Y.; Deng, W.; Wang, W.; Song, A.; Mukama, O.; Deng, S.; Han, X.; De Dieu Habimana, J.; Peng, K.; Ni, B. MicroRNA-206 down-regulated human umbilical cord mesenchymal stem cells alleviate cognitive decline in D-galactose-induced aging mice. Cell Death Discov. 2022, 8, 304. [Google Scholar] [CrossRef] [PubMed]
  135. Mas-Bargues, C.; Sanz-Ros, J.; Román-Domínguez, A.; Gimeno-Mallench, L.; Inglés, M.; Viña, J.; Borrás, C. Extracellular vesicles from healthy cells improves cell function and stemness in premature senescent stem cells by miR-302b and HIF-1α activation. Biomolecules 2020, 10, 957. [Google Scholar] [CrossRef] [PubMed]
  136. Takahashi, K.; Yamanaka, S. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell 2006, 126, 663–676. [Google Scholar] [CrossRef] [PubMed]
  137. Pascale, E.; Caiazza, C.; Paladino, M.; Parisi, S.; Passaro, F.; Caiazzo, M. MicroRNA roles in cell reprogramming mechanisms. Cells 2022, 11, 940. [Google Scholar] [CrossRef] [PubMed]
  138. Mirzaei, S.; Saebfar, H.; Gholami, M.H.; Hashemi, F.; Zarrabi, A.; Zabolian, A.; Entezari, M.; Hushmandi, K.; Samarghandian, S.; Aref, A.R. MicroRNAs regulating SOX2 in cancer progression and therapy response. Expert Rev. Mol. Med. 2021, 23, e13. [Google Scholar] [CrossRef] [PubMed]
  139. Xie, H.; Yao, J.; Wang, Y.; Ni, B. Exosome-transmitted circVMP1 facilitates the progression and cisplatin resistance of non-small cell lung cancer by targeting miR-524-5p-METTL3/SOX2 axis. Drug Deliv. 2022, 29, 1257–1271. [Google Scholar] [CrossRef] [PubMed]
  140. Chen, J.; Chen, T.; Zhu, Y.; Li, Y.; Zhang, Y.; Wang, Y.; Li, X.; Xie, X.; Wang, J.; Huang, M. circPTN sponges miR-145-5p/miR-330-5p to promote proliferation and stemness in glioma. J. Exp. Clin. Cancer Res. 2019, 38, 398. [Google Scholar] [CrossRef] [PubMed]
  141. Dai, J.; Hao, Y.; Chen, X.; Yu, Q.; Wang, B. miR-122/SENP1 axis confers stemness and chemoresistance to liver cancer through Wnt/β-catenin signaling. Oncol. Lett. 2023, 26, 390. [Google Scholar] [CrossRef] [PubMed]
  142. Guo, L.; Li, K.; Ma, Y.; Niu, H.; Li, J.; Shao, X.; Li, N.; Sun, Y.; Wang, H. MicroRNA-322-5p targeting Smurf2 regulates the TGF-β/Smad pathway to protect cardiac function and inhibit myocardial infarction. Hum. Cell 2024, 37, 972–985. [Google Scholar] [CrossRef]
  143. Zhu, B.; V, M.; Finch-Edmondson, M.; Lee, Y.; Wan, Y.; Sudol, M.; DasGupta, R. miR-582-5p is a tumor suppressor microRNA targeting the Hippo-YAP/TAZ signaling pathway in non-small cell lung cancer. Cancers 2021, 13, 756. [Google Scholar] [CrossRef]
  144. Han, B.; Li, T.; Zheng, S. MicroRNA-133b aggravates atherosclerosis by activating the Notch signaling pathway. Mol. Med. Rep. 2020, 22, 1621–1630. [Google Scholar] [CrossRef] [PubMed]
  145. Zhao, M.; Wang, L.-T.; Liang, G.-P.; Zhang, P.; Deng, X.-J.; Tang, Q.; Zhai, H.-Y.; Chang, C.C.; Su, Y.-W.; Lu, Q.-J. Up-regulation of microRNA-210 induces immune dysfunction via targeting FOXP3 in CD4+ T cells of psoriasis vulgaris. Clin. Immunol. 2014, 150, 22–30. [Google Scholar] [CrossRef] [PubMed]
  146. Wang, W.; Wang, Z.; Chen, S.; Zang, X.; Miao, J. Interleukin-1β/nuclear factor-ĸB signaling promotes osteosarcoma cell growth through the microRNA-181b/phosphatase and tensin homolog axis. J. Cell. Biochem. 2019, 120, 1763–1772. [Google Scholar] [CrossRef] [PubMed]
  147. Khalaf, S.E.; Abdelfattah, S.N.; Khaliefa, A.K.; Daoud, S.A.; Yahia, E.; Hasona, N.A. Expression of PVT-1 and miR-29a/29b as reliable biomarkers for liver cirrhosis and their correlation with the inflammatory biomarkers profile. Hum. Exp. Toxicol. 2024, 43, 09603271241251451. [Google Scholar] [CrossRef] [PubMed]
  148. Huang, S.; Xiao, X.; Wu, H.; Zhou, F.; Fu, C. MicroRNA-582-3p knockdown alleviates non-alcoholic steatohepatitis by altering the gut microbiota composition and moderating TMBIM1. Ir. J. Med. Sci. 2024, 193, 909–916. [Google Scholar] [CrossRef] [PubMed]
  149. Das, O.; Kundu, J.; Ghosh, A.; Gautam, A.; Ghosh, S.; Chakraborty, M.; Masid, A.; Gauri, S.S.; Mitra, D.; Dutta, M. AUF-1 knockdown in mice undermines gut microbial butyrate-driven hypocholesterolemia through AUF-1–Dicer-1–mir-122 hierarchy. Front. Cell. Infect. Microbiol. 2022, 12, 1011386. [Google Scholar] [CrossRef] [PubMed]
  150. Yang, Y.L.; Huang, Y.H.; Wang, F.S.; Tsai, M.C.; Chen, C.H.; Lian, W.S. MicroRNA-29a Compromises Hepatic Adiposis and Gut Dysbiosis in High Fat Diet-Fed Mice via Downregulating Inflammation. Mol. Nutr. Food Res. 2023, 67, 2200348. [Google Scholar] [CrossRef] [PubMed]
  151. Jian, G.; Yangli, J.; Chao, Z.; Kun, W.; Xiaomin, Z. MicroRNA-629-3p Promotes Interleukin-13-Induced Bronchial Epithelial Cell Injury and Inflammation by Targeting FOXA2. Cell Biochem. Biophys. 2022, 80, 457–466. [Google Scholar] [CrossRef] [PubMed]
  152. Bertero, T.; Cottrill, K.A.; Annis, S.; Bhat, B.; Gochuico, B.R.; Osorio, J.C.; Rosas, I.; Haley, K.J.; Corey, K.E.; Chung, R.T. A YAP/TAZ-miR-130/301 molecular circuit exerts systems-level control of fibrosis in a network of human diseases and physiologic conditions. Sci. Rep. 2015, 5, 18277. [Google Scholar] [CrossRef] [PubMed]
  153. Hu, J.; Liu, T.; Zhang, Z.; Xu, Y.; Zhu, F. Oxidized low-density lipoprotein promotes vascular endothelial cell dysfunction by stimulating miR-496 expression and inhibiting the Hippo pathway effector YAP. Cell Biol. Int. 2019, 43, 528–538. [Google Scholar] [CrossRef]
  154. Garo, L.P.; Murugaiyan, G. Contribution of MicroRNAs to autoimmune diseases. Cell. Mol. Life Sci. 2016, 73, 2041–2051. [Google Scholar] [CrossRef] [PubMed]
  155. Mahesh, G.; Biswas, R. MicroRNA-155: A master regulator of inflammation. J. Interferon Cytokine Res. 2019, 39, 321–330. [Google Scholar] [CrossRef] [PubMed]
  156. McCoy, C.E. miR-155 dysregulation and therapeutic intervention in multiple sclerosis. Regul. Inflamm. Signal. Health Dis. 2017, 1024, 111–131. [Google Scholar]
  157. Xu, W.-D.; Feng, S.-Y.; Huang, A.-F. Role of miR-155 in inflammatory autoimmune diseases: A comprehensive review. Inflamm. Res. 2022, 71, 1501–1517. [Google Scholar] [CrossRef] [PubMed]
  158. Guo, Q.Y.; Song, J.N.; Chen, Y.M.; Yuan, H.N.; Xue, W.S.; Sun, Y.; Wang, Y.; Chen, X. IL-6 regulates epithelial ovarian cancer EMT, invasion, and metastasis by modulating Let-7c and miR-200c through the STAT3/HIF-1α pathway. Med. Oncol. 2024, 41, 1–11. [Google Scholar] [CrossRef] [PubMed]
  159. Pei, H.P.; Tan, F.B.; Liu, L.; Yu, N.H.; Zhu, H. IL-1β/NF-kb signaling promotes colorectal cancer cell growth through miR-181a/PTEN axis. Arch. Biochem. Biophys. 2016, 604, 20–26. [Google Scholar]
  160. Shu, X.; Chen, X.-X.; Kang, X.-D.; Ran, M.; Wang, Y.-L.; Zhao, Z.-K.; Li, C.-X. Identification of potential key molecules and signaling pathways for psoriasis based on weighted gene co-expression network analysis. World J. Clin. Cases 2022, 10, 5965. [Google Scholar] [CrossRef]
  161. Li, Y.-n.; Shen, J.; Feng, Y.; Zhang, Y.; Wang, Y.; Ren, X. The relationship of peripheral blood lncRNA-PVT1 and miR-146a levels with Th17/Treg cytokines in patients with Hashimoto’s thyroiditis and their clinical significance. Biomol. Biomed. 2024. ahead of print. [Google Scholar] [CrossRef] [PubMed]
  162. Scalavino, V.; Piccinno, E.; Bianco, G.; Schena, N.; Armentano, R.; Giannelli, G.; Serino, G. The increase of miR-195-5p reduces intestinal permeability in ulcerative colitis, modulating tight junctions’ expression. Int. J. Mol. Sci. 2022, 23, 5840. [Google Scholar] [CrossRef]
  163. Gao, Y.; Sheng, D.; Chen, W. Regulatory mechanism of miR-20a-5p in neuronal damage and inflammation in lipopolysaccharide-induced BV2 cells and MPTP-HCl-induced Parkinson’s disease mice. Psychogeriatrics 2024, 24, 752–764. [Google Scholar] [CrossRef]
  164. Lin, Z.; Bao, R.; Niu, Y.; Kong, X. KLF5-mediated pyroptosis of airway epithelial cells leads to airway inflammation in asthmatic mice through the miR-182–5p/TLR4 axis. Mol. Immunol. 2024, 170, 9–18. [Google Scholar] [CrossRef] [PubMed]
  165. Yang, J.; Wei, Z.; Li, H.; Lv, S.; Fu, Y.; Xiao, L. Paeoniflorin inhibits the inflammation of rheumatoid arthritis fibroblast-like synoviocytes by downregulating hsa_circ_009012. Heliyon 2024, 10, e30555. [Google Scholar] [CrossRef] [PubMed]
  166. Lv, H.; Liu, P.; Hu, H.; Li, X.; Li, P. MiR-98-5p plays suppressive effects on IL-1β-induced chondrocyte injury associated with osteoarthritis by targeting CASP3. J. Orthop. Surg. Res. 2024, 19, 239. [Google Scholar] [CrossRef] [PubMed]
  167. Li, C.; Sun, Z.; Lou, H.; Sun, Y.; Li, C.; Gao, X. Effects of miR-129-5p on inflammation and nucleus pulposus cell apoptosis in rats with intervertebral disc degeneration through JNK signaling pathway. Cell. Mol. Biol. 2024, 70, 164–168. [Google Scholar] [PubMed]
  168. Liechty, C.; Hu, J.; Zhang, L.; Liechty, K.W.; Xu, J. Role of microRNA-21 and its underlying mechanisms in inflammatory responses in diabetic wounds. Int. J. Mol. Sci. 2020, 21, 3328. [Google Scholar] [CrossRef]
  169. Rawal, S.; Randhawa, V.; Rizvi, S.H.M.; Sachan, M.; Wara, A.K.; Pérez-Cremades, D.; Weisbrod, R.M.; Hamburg, N.M.; Feinberg, M.W. miR-369-3p ameliorates diabetes-associated atherosclerosis by regulating macrophage succinate-GPR91 signaling. Cardiovasc. Res. 2024, cvae102. [Google Scholar] [CrossRef]
  170. Zhang, S.; Zhang, Y.; Wang, N.; Wang, Y.; Nie, H.; Zhang, Y.; Han, H.; Wang, S.; Liu, W.; Bo, C. Long non-coding RNA MIAT impairs neurological function in ischemic stroke via up-regulating microRNA-874-3p-targeted IL1B. Brain Res. Bull. 2021, 175, 81–89. [Google Scholar] [CrossRef]
  171. Fardi, F.; Khasraghi, L.B.; Shahbakhti, N.; Naseriyan, A.S.; Najafi, S.; Sanaaee, S.; Alipourfard, I.; Zamany, M.; Karamipour, S.; Jahani, M. An interplay between non-coding RNAs and gut microbiota in human health. Diabetes Res. Clin. Pract. 2023, 201, 110739. [Google Scholar] [CrossRef]
  172. Nikolaieva, N.; Sevcikova, A.; Omelka, R.; Martiniakova, M.; Mego, M.; Ciernikova, S. Gut microbiota–microRNA interactions in intestinal homeostasis and cancer development. Microorganisms 2022, 11, 107. [Google Scholar] [CrossRef]
  173. Du, X.; Ley, R.; Buck, A. MicroRNAs and extracellular vesicles in the gut: New host modulators of the microbiome? microLife 2021, 2, uqab010. [Google Scholar] [CrossRef]
  174. Yan, X.-Y.; Yao, J.-P.; Li, Y.-Q.; Zhang, W.; Xi, M.-H.; Chen, M.; Li, Y. Global trends in research on miRNA–microbiome interaction from 2011 to 2021: A bibliometric analysis. Front. Pharmacol. 2022, 13, 974741. [Google Scholar] [CrossRef] [PubMed]
  175. Prukpitikul, P.; Sirivarasai, J.; Sutjarit, N. The molecular mechanisms underlying gut microbiota-miRNA interaction in metabolic disorders. Benef. Microbes 2024, 15, 83–96. [Google Scholar] [CrossRef] [PubMed]
  176. Izdebska, W.M.; Daniluk, J.; Niklinski, J. Microbiome and MicroRNA or Long Non-Coding RNA—Two Modern Approaches to Understanding Pancreatic Ductal Adenocarcinoma. J. Clin. Med. 2023, 12, 5643. [Google Scholar] [CrossRef] [PubMed]
  177. Dothel, G.; Barbaro, M.R.; Di Vito, A.; Ravegnini, G.; Gorini, F.; Monesmith, S.; Coschina, E.; Benuzzi, E.; Fuschi, D.; Palombo, M. New insights into irritable bowel syndrome pathophysiological mechanisms: Contribution of epigenetics. J. Gastroenterol. 2023, 58, 605–621. [Google Scholar] [CrossRef] [PubMed]
  178. Oliveira, E.C.S.d.; Quaglio, A.E.V.; Magro, D.O.; Di Stasi, L.C.; Sassaki, L.Y. Intestinal microbiota and miRNA in IBD: A narrative review about discoveries and perspectives for the future. Int. J. Mol. Sci. 2023, 24, 7176. [Google Scholar] [CrossRef] [PubMed]
  179. Mousa, W.K.; Chehadeh, F.; Husband, S. Microbial dysbiosis in the gut drives systemic autoimmune diseases. Front. Immunol. 2022, 13, 906258. [Google Scholar] [CrossRef] [PubMed]
  180. Kopczyńska, J.; Kowalczyk, M. The potential of short-chain fatty acid epigenetic regulation in chronic low-grade inflammation and obesity. Front. Immunol. 2024, 15, 1380476. [Google Scholar] [CrossRef] [PubMed]
  181. Yan, Y.; Li, Q.; Yang, F.; Shen, L.; Guo, K.; Zhou, X. Chlorogenic acid ameliorates intestinal inflammation via miRNA-microbe axis in db/db mice. FASEB J. 2024, 38, e23665. [Google Scholar] [CrossRef] [PubMed]
  182. Ma, L.; Hou, C.; Yang, H.; Chen, Q.; Lyu, W.; Wang, Z.; Wang, J.; Xiao, Y. Multi-omics analysis reveals the interaction of gut microbiome and host microRNAs in ulcerative colitis. Ann. Med. 2023, 55, 2261477. [Google Scholar] [CrossRef]
  183. Huang, H.; Zhao, T.; Li, J.; Shen, J.; Xiao, R.; Ma, W. Gut microbiota regulation of inflammatory cytokines and microRNAs in diabetes-associated cognitive dysfunction. Appl. Microbiol. Biotechnol. 2023, 107, 7251–7267. [Google Scholar] [CrossRef]
  184. Li, Q.; Zhang, J.; Gao, Z.; Zhang, Y.; Gu, J. Gut microbiota-induced microRNA-206-3p increases anxiety-like behaviors by inhibiting expression of Cited2 and STK39. Microb. Pathog. 2023, 176, 106008. [Google Scholar] [CrossRef] [PubMed]
  185. Han, K.; Ji, L.; Wang, C.; Shao, Y.; Chen, C.; Liu, L.; Feng, M.; Yang, F.; Wu, X.; Li, X. The host genetics affects gut microbiome diversity in Chinese depressed patients. Front. Genet. 2023, 13, 976814. [Google Scholar] [CrossRef] [PubMed]
  186. Datta, N.; Johnson, C.; Kao, D.; Gurnani, P.; Alexander, C.; Polytarchou, C.; Monaghan, T.M. MicroRNA-based therapeutics for inflammatory disorders of the microbiota-gut-brain axis. Pharmacol. Res. 2023, 194, 106870. [Google Scholar] [CrossRef] [PubMed]
  187. Chen, H.-M.; Chung, Y.-C.E.; Chen, H.-C.; Liu, Y.-W.; Chen, I.-M.; Lu, M.-L.; Hsiao, F.S.-H.; Chen, C.-H.; Huang, M.-C.; Shih, W.-L. Exploration of the relationship between gut microbiota and fecal microRNAs in patients with major depressive disorder. Sci. Rep. 2022, 12, 20977. [Google Scholar] [CrossRef] [PubMed]
  188. Sayed, N.; Huang, Y.; Nguyen, K.; Krejciova-Rajaniemi, Z.; Grawe, A.P.; Gao, T.; Tibshirani, R.; Hastie, T.; Alpert, A.; Cui, L. An inflammatory aging clock (iAge) based on deep learning tracks multimorbidity, immunosenescence, frailty and cardiovascular aging. Nat. Aging 2021, 1, 598–615. [Google Scholar] [CrossRef] [PubMed]
  189. Horvath, S.; Raj, K. DNA methylation-based biomarkers and the epigenetic clock theory of ageing. Nat. Rev. Genet. 2018, 19, 371–384. [Google Scholar] [CrossRef] [PubMed]
  190. Freitas, L.S.; Silveira, A.C.; Martins, F.C.; Costa-Hong, V.; Lebkuchen, A.; Cardozo, K.H.; Bernardes, F.M.; Bortolotto, L.A.; Lorenzi-Filho, G.; Oliveira, E.M. Severe obstructive sleep apnea is associated with circulating microRNAs related to heart failure, myocardial ischemia, and cancer proliferation. Sleep Breath. 2020, 24, 1463–1472. [Google Scholar] [CrossRef] [PubMed]
  191. Iannone, F.; Crocco, P.; Dato, S.; Passarino, G.; Rose, G. Circulating miR-181a as a novel potential plasma biomarker for multimorbidity burden in the older population. BMC Geriatr. 2022, 22, 772. [Google Scholar] [CrossRef]
  192. Sorror, M.L.; Gooley, T.A.; Maclean, K.H.; Hubbard, J.; Marcondes, M.A.; Torok-Storb, B.J.; Tewari, M. Pre-transplant expressions of microRNAs, comorbidities, and post-transplant mortality. Bone Marrow Transplant. 2019, 54, 973–979. [Google Scholar] [CrossRef] [PubMed]
  193. Ermogenous, C.; Green, C.; Jackson, T.; Ferguson, M.; Lord, J.M. Treating age-related multimorbidity: The drug discovery challenge. Drug Discov. Today 2020, 25, 1403–1415. [Google Scholar] [CrossRef]
  194. Singer, L.; Green, M.; Rowe, F.; Ben-Shlomo, Y.; Kulu, H.; Morrissey, K. Trends in multimorbidity, complex multimorbidity and multiple functional limitations in the ageing population of England, 2002–2015. J. Comorbidity 2019, 9, 2235042X19872030. [Google Scholar] [CrossRef] [PubMed]
  195. Barnes, M.E.; Elliott, J.A.; McIntyre, T.V.; Boyle, E.A.; Gillis, A.E.; Ridgway, P.F. Sarcopenia and obesity among patients with soft tissue sarcoma–Association with clinicopathologic characteristics, complications and oncologic outcome: A systematic review and meta-analysis. Eur. J. Surg. Oncol. 2021, 47, 2237–2247. [Google Scholar] [CrossRef]
  196. Saliminejad, K.; Khorram Khorshid, H.R.; Ghaffari, S.H. Why have microRNA biomarkers not been translated from bench to clinic? Future Oncol. 2019, 15, 801–803. [Google Scholar] [CrossRef] [PubMed]
  197. Davey, M.G.; Davies, M.; Lowery, A.J.; Miller, N.; Kerin, M.J. The role of microRNA as clinical biomarkers for breast cancer surgery and treatment. Int. J. Mol. Sci. 2021, 22, 8290. [Google Scholar] [CrossRef]
  198. Mohr, A.M.; Mott, J.L. Overview of microRNA biology. Semin. Liver Dis. 2015, 35, 003–011. [Google Scholar] [CrossRef]
  199. Austad, S.N. The geroscience hypothesis: Is it possible to change the rate of aging? In Advances in Geroscience; Springer: Berlin/Heidelberg, Germany, 2016; pp. 1–36. [Google Scholar]
  200. Ding, X.; Ma, X.; Meng, P.; Yue, J.; Li, L.; Xu, L. Potential Effects of Traditional Chinese Medicine in Anti-Aging and Aging-Related Diseases: Current Evidence and Perspectives. Clin. Interv. Aging 2024, 19, 681–693. [Google Scholar] [CrossRef] [PubMed]
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

Butler, T.; Davey, M.G.; Kerin, M.J. Molecular Morbidity Score–Can MicroRNAs Assess the Burden of Disease? Int. J. Mol. Sci. 2024, 25, 8042. https://doi.org/10.3390/ijms25158042

AMA Style

Butler T, Davey MG, Kerin MJ. Molecular Morbidity Score–Can MicroRNAs Assess the Burden of Disease? International Journal of Molecular Sciences. 2024; 25(15):8042. https://doi.org/10.3390/ijms25158042

Chicago/Turabian Style

Butler, Thomas, Matthew G. Davey, and Michael J. Kerin. 2024. "Molecular Morbidity Score–Can MicroRNAs Assess the Burden of Disease?" International Journal of Molecular Sciences 25, no. 15: 8042. https://doi.org/10.3390/ijms25158042

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