Reviews:

Cell Stress, Vol. 8, No. 1, pp. 59 - 68; doi: 10.15698/cst2024.05.297

Saliva, a molecular reflection of the human body? Implications for diagnosis and treatment

Vincent Géli and Norbert Nabet

Download PDF download pdf
Show/hide additional information

    19 rue Riboud, 69003, Ziwig, Lyon, France.

Keywords: saliva, miRNA, diagnosis, cancer, endometriosis

Abbreviations:

circRNA - circular RNA, EV -

extracellular vesicle,

gDNA - genomic DNA,

lncRNA - long ncRNA,

mRNA - messenger RNA,

miRNA - micro RNA,

ncRNA - non-coding RNA,

piRNA - protein-interacting RNA.
Received originally: 14/03/2024 Received in revised form: 02/05/2024
Accepted: 03/05/2024 Published: 27/05/2024

Correspondence:
Vincent Géli, Phone: 33 (0) 676478900; vincentgeli1@gmail.com

Conflict of interest statement: From December 2023 to April 2024, V.G. was employed by the Ziwig Biotech company.
Please cite this article as: Vincent Géli, Norbert Nabet (2024). Saliva, a molecular reflection of the human body? Implications for diagnosis and treatment. Cell Stress 8: 59-68. doi: 10.15698/cst2024.05.297

Abstract

For many diseases, and cancer in particular, early diagnosis allows a wider range of therapies and a better disease management. This has led to improvements in diagnostic procedures, most often based on tissue biopsies or blood samples. Other biological fluids have been used to diagnose disease, and among them saliva offers a number of advantages because it can be collected non-invasively from large populations at relatively low cost. To what extent might saliva content reveal the presence of a tumour located at a distance from the oral cavity and the molecular information obtained from saliva be used to establish a diagnosis are current questions. This review focuses primarily on the content of saliva and shows how it potentially offers a source of diagnosis, possibly at an early stage, for pathologies such as cancers or endometriosis.

INTRODUCTION

The possibility of using biological fluids such as blood, urine, saliva and other biofluids to diagnose pathologies is a relatively old idea that has taken on a new lease of life in recent years. Thanks to a combination of technological advances, the first major progress was observed in genomics, proteomics and bioinformatics techniques, now including artificial intelligence, making it possible to establish observed differences between analysed samples with greater sensitivity and a higher degree of confidence. The second is the standardization of sample collection, processing, and storage, minimizing variations linked to sample collection and analysis, as well as to inter-individual variability. Finally, improved knowledge, particularly in molecular and cellular biology, makes it possible to give biological meaning to variations between samples. This is particularly decisive for the analysis of a particular class of RNAs, known as non-coding RNAs (ncRNAs). In recent years, understanding of the role and regulation of ncRNAs has progressed exponentially, particularly in terms of how ncRNAs respond to genetic and epigenetic perturbations, especially in pathological situations 1. A crucial element in the use of biological fluids for the discovery of disease-associated biomarkers is the characterization and understanding of a mode of intercellular communication based on the exchange of vesicles between vesicle-secreting cells, called extracellular vesicles (EVs), and receptor cells within a target tissue 2, 3. These EVs can act close to their site of secretion, promoting signaling in neighboring cells, or circulate in the blood or lymph to affect distant tissues. Their effect at distance depends on the concentration of the EVs and their content. A growing number of examples show that EVs found in biological fluids, and particularly in saliva, can reflect acute or chronic pathology located outside a patient’s oral cavity 4. In this review, we will describe the potential of saliva as a biological fluid, not only for the diagnosis of various diseases, but also for the establishment of possible therapeutic solutions.

EXTRACELLULAR VESICLES AND THEIR CONTENTS

As mentioned above, some of the differences in saliva content between healthy people and patients with a given pathology could be due to different concentrations of EVs, themselves of a different nature in patients. EVs differ in size, physical properties, mode of biosynthesis and content, which depends on the secreting cell 5. EVs are generated from invaginations of the cell membrane which, following various maturation processes, form multivesicular bodies containing intraluminal vesicles 6. These in turn can be degraded or secreted into the extracellular space notably as exosomes 7. The orientation of transmembrane proteins on the surface of exosomes is identical to that observed in the cell membrane, albeit with different steric constraints due to the greater radius of curvature in exosomes. Other, larger types of EVs can also form from buds on the cell surface, or from cells undergoing apoptosis, generating so-called apoptotic bodies 6. Over the past decade, considerable progress has been made in clarifying the content of the molecules transported by EVs, known as “cargoes”, and the role of EVs. Cargoes include proteins such as membrane receptors, key signalling proteins, major histocompatibility complexes, intracellular trafficking regulators and various RNA-binding proteins 8. Remarkably, many nucleic acids, particularly ncRNAs, are found within EVs, as are amino acids and metabolites. It is now accepted that the biogenesis mechanism of EVs can serve to actively capture and concentrate cargoes through the action of effectors 6, 9. Conversely, other cargoes can also be passively encapsulated within EVs. The secretion of EVs constitutes a genuine mechanism for the exchange of molecules and the transmission of signals between a secretory cell and the receptor cell, through paracrine effects 10. As mentioned above, the content of EVs depends on the cell type secreting them, and this content can be disrupted in stressful or pathological situations such as cancer 11, 12. Numerous studies have shown that in pathological contexts, notably cancers, the rate of secretion of EVs by tumour cells is higher than in normal cells 13. EVs in biological fluids may therefore reflect cell or tissue dysfunction.

 

To what extent EVs are found in different biological fluids and how their distribution in these fluids provides specific answers to their use as a tool for diagnosis are open questions.

SALIVA, A BIOLOGICAL FLUID FOR BIOMARKER DETECTION

Saliva is a biological fluid secreted by various salivary glands, which once poured into the oral cavity, is mixed with secretions from the epithelial cells of the oral wall. It also contains cellular infiltrates and many micro-organisms, including bacteria and viruses 14, 15. It is an abundant saliva secretion, estimated to a liter per day for a healthy person 16. Saliva, made up of 98% water, contains a variety of proteins (mucins, lactoferrin, histatins, various proteases, immunoglobulins) involved in protecting the oral cavity and in the initial stages of digestion 17, as well as growth factors, hormones, cytokines and metabolites 18. Proteomic studies have shown that most proteins are synthesized in the salivary glands 19, although some are transported from the blood or lymph into the saliva 20. Saliva also contains nucleic acids: nuclear DNA 21 and, above all, many different classes of ncRNA 22. More than a decade ago, the question was whether saliva vesicles and their contents originated solely from the salivary glands, possibly revealing oral pathologies, or whether they could be derived from cells belonging, for example, to a tumour (or their microenvironment) located at a distance from the oral cavity. Numerous studies have now shown that small ncRNAs found in the saliva of cancer patients are associated with tumour-derived cancer cells 23. The question then arose as to how EVs originating from tumour cells or associated with tumours find their way into the patient’s saliva. The prevailing idea is that EVs secreted by tumour or tumour-associated cells reach the salivary glands via the general circulation, or via the lymph. EVs are then taken up by the acinar cells of the salivary glands by membrane fusion or by an invagination mechanism (endocytosis). The salivary glands then secrete mixed EVs into the saliva, originating from both acinar cells and EVs derived from the tumour located at a distance from the salivary glands 23. This model explains why ncRNAs detected in saliva, particularly in pathological situations, will have a different profile to those found in other biological fluids, notably blood 24.

 

For certain pathologies, saliva can therefore serve as a biomarker of disease. The higher the number of samples, the more robust the biomarker. The fact that saliva sampling is easy, non-invasive, easy to handle, inexpensive and can be used on a large scale of patients 25 opens new prospects not only for the discovery of biomarkers for the diagnosis of pathologies, but also for the understanding of the pathologies studied.

DNA IN THE SALIVA

Saliva contains many different types of nucleic acid. Much of the DNA extracted from saliva comes from bacteria, but mainly from leukocytes and epithelial cells in the oral cavity 26. The proportion of cellular DNA will depend on how the saliva is collected, influencing the collection of cells in the saliva sample. Sequenced genomic DNA (gDNA) will provide information on the patient’s genetic identity but cannot be used to determine somatic alterations in the cells involved in the disease. In this sense, gDNA purified from saliva can be substituted for gDNA purified from blood leukocytes to perform whole genome sequencing 27 or large-scale analysis of gDNA methylation profiles 28. Average telomere length can even be measured by PCR from saliva samples 29. Nevertheless, in diagnostic terms, gDNA from oral cavity cells will not be informative, except for familial cancers 30, and cancers forming in the upper aerodigestive tract for which saliva contains cancer cells 31.

 

In cancer patients, saliva is likely to contain circular DNA consisting of short DNA sequences wrapped around one or more nucleosomes 32. Nevertheless, a significant fraction of saliva’s extracellular DNA is protected from nuclease action, suggesting that it is contained within secreted EVs, either by cells of the oral cavity or, in the case of cancers, by more distal tumour cells 33. The origin of extracellular DNA and gDNA-containing EVs is not fully elucidated. It could come from apoptotic bodies generated by cells undergoing apoptosis 6. Studies have shown that nuclear DNA is not found in small, highly purified EVs (exosomes) 8. However, in ovarian cancer patients, it has been shown that 10% of exosomes contain genomic DNA. Its presence in EVs is thought to be linked to micronuclei that characterize the genetic instability of cancer cells 34. Other mechanisms linked to the genetic instability of cancer cells may also explain the leakage of gDNA into the cytoplasm 35. These results are in line with various studies which have shown that mutations (KRAS, TP53, EGFR, and PIK3CA) associated with different types of cancer (pancreas, breast, lung) could be detected from EVs purified from patient blood, notably thanks to the use of digital droplet PCR enabling a region of DNA to be amplified with high sensitivity 36, 37, 38, 39. In saliva, extracellular DNA is present at lower concentrations than in blood 40. A better understanding of the origin of the extracellular DNA present in saliva is needed to consider the enrichment of EVs containing tumour DNA, and to evaluate the potential of saliva for the detection of cancer-associated mutations.

 

There are also stress or pathological conditions that cause mitochondrial DNA 41 to leak into the cytoplasm, inducing inflammatory-type responses initiated by the recognition of DNA found in the cytoplasm by specific sensors
42. Under these conditions, cytoplasmic mitochondrial DNA can be incorporated into EVs, contributing to the propagation of the inflammatory response 43. It has been shown that mitochondrial DNA amplicons can be directly amplified and quantified by qRT-PCR from saliva independently of the cellular content of the saliva 44. In head and neck cancers, the normalized amount of extracellular mitochondrial DNA present in saliva correlated with poorer patient survival indicating that the level of extracellular mitochondrial DNA in saliva can be used as a robust predictor of overall patient survival 45. It will be of interest to study whether quantification of mitochondrial DNA, and possibly mitochondrial gene sequence, established from saliva constitutes a novel biomarker for cancers 46.

miRNAs IN SALIVA

As aforementioned, saliva can reflect systemic processes, mainly through the EVs contained in it. Like other EVs, salivary EVs contain a variable proportion of ncRNAs corresponding to RNA molecules that are not translated into proteins. ncRNAs include small RNAs of around 22 nucleotides (nt) called microRNAs (miRNAs), long non-coding RNAs (lncRNAs) whose size exceeds 200 nt, and a particular class of Piwi protein-interacting RNAs (piRNAs) notably involved in the repression of transposable elements 4, 33. These ncRNAs are accompanied by single-stranded circular RNAs (circRNAs) produced by mRNA by back‐splicing of the upstream 5′ splice donor site with the upstream 3′splice acceptor 47. CircRNAs, whose abundance varies with tissue and age, are particularly stable and have been detected in plasma and saliva 47. They act as miRNA sponges, and as such represent a new source of biomarkers, notably in many cancers 48.

 

miRNAs were first identified in the C. elegans worm 49, 50 and then in vertebrates 51. Numerous studies have since clarified the mode of biosynthesis and mechanism of action of miRNAs 52. They are synthesized as precursors by RNA polymerase II and undergo an initial maturation step in the nucleus, generating miRNAs of around 70 nt in size, which are then exported to the cytoplasm 52. In the cytoplasm, these stem-loop miRNAs undergo the action of the DICER endoribonuclease, producing double-stranded RNA duplexes of around 22 nucleotides. One strand of the miRNA duplex (the other strand is degraded) forms a complex with the Argonaute protein (AGO), called RISC, whose action will switch off the expression of genes with sequences complementary to miRNA 53, either by destabilizing the RNA targeted by the miRNA, or by inhibiting its translation, depending on the degree of homology of the miRNA with its target RNA 54, 55.

 

It has been established that most mammalian messenger RNAs (mRNAs) can be targets for miRNAs 56. Over 2,600 different miRNAs have been identified, of which more than 1000 are annotated in databases 57. However, the number of protein-coding genes whose expression is effectively regulated by miRNAs remains to be determined. It is estimated that 60% of coding genes are regulated by miRNAs.

 

Although miRNAs found in biofluids are not necessarily EV-associated, it is now recognized that miRNAs secreted into EVs are key players in EV-mediated intercellular communication 58. It should be noted that the population of miRNAs released into EVs is distinct from the cellular population by the presence of export sequences (Exomotifs) favoring their concentration in EVs 59. miRNAs can also be secreted into biological fluids by binding to RNA-affine proteins such as Ago2 and lipoproteins 60. Extracellular miRNAs have been found in all biological fluids, including saliva 33. miRNAs are highly stable in biological fluids, both because of their structure and because they are released from EVs 61. A large number of studies have exploited the use of blood miRNAs as biomarkers of disease 62, but fewer have explored the utility of salivary miRNAs. Nevertheless, recent studies indicate that biomarkers based on salivary EVs could predict several pathologies other than those of the oral cavity 63.

SALIVARY miRNAs: BIOMARKERS OF CANCER

Although saliva has advantages over blood as a biological fluid for diagnosis, variations in its potential contamination and composition have been obstacles to its use. This limitation can be partially overcome by the standardization of samples and by the purification of EVs, which constitutes a step in the purification and enrichment of ncRNA contained in saliva 47. In addition to head and neck cancers, miRNAs from salivary EVs have recently been shown to be biomarkers for oesophageal cancers 64, colorectal cancers 65, 66, lung carcinomas 67, 68, hepatocarcinomas 69 and as potential biomarkers for pancreatic cancer 70, 71. Salivary miRNAs are also thought to be biomarkers for ovarian cancer, for which several miRNAs purified from patient blood have been associated with the proliferation and migration of epithelial cancer cells 72, 73, 74.

 

Various protocols have now been developed to purify EVs from saliva, including exosomes, opening new horizons for the use of saliva in diagnosis. In particular, salivary EVs can be concentrated by immuno-purification using antibodies directed against four-domain transmembrane proteins called tetraspanins (CD9, CD63, CD81) found on the surface of most exosomes 7. However, there are now several evidences in the EV-field that there may be a significant population of EVs that are negative for those markers 75. Mass spectrometry studies have also shown that exosomes derived from pancreatic tumours may contain CD151, another tetraspanin 76, making it possible to enrich specific exosomes associated with a particular type of cancer from saliva. Similarly, ovarian cancers overexpress a family of proteins called syndecans 77, 78 consisting of an extracellular domain, a transmembrane domain and a cytoplasmic domain which communicate with tetraspanins to regulate vesicular trafficking and the activity of associated receptors 79. It is conceivable that syndecans overexpressed in ovarian cancer could escape proteolytic cleavage of their extracellular domain, enabling the purification of specific exosomes with appropriate antibodies to facilitate diagnosis.

COMPLEXITY OF miRNA EXPRESSION REGULATION

While the mechanism of miRNA-mediated repression of gene expression has been extensively studied, the mechanisms leading to changes in miRNA expression, particularly in cancer cells, are much less well understood 80. miRNAs can be classified into intragenic and intergenic miRNAs. Most of the miRNAs deregulated in cancer (onco-miRNAs) are integrated into the introns of host genes. Using multidimensional omics databases to simultaneously study the expression of intragenic miRNAs and their host genes, it has been shown that, in several types of cancer, the expression of onco-miRNAs is often correlated with that of related host genes 81. It is reasonable to assume that this co-regulation may be linked to epigenetic modifications of either the host gene promoter or the microRNA promoter 82. There are numerous examples showing that, in cancer, the expression of onco-miRNAs can be derepressed via hypomethylation of the CpG islands of their promoter, while conversely, the expression of miRNAs with a tumour suppressor role is inhibited via hypermethylation of the CpG islands 83. Nevertheless, analysis of the links between onco-miRNAs and host genes is still far from complete. Nevertheless, the analysis of the links between DNA methylation and miRNA expression remains complicated because miRNAs, themselves regulated by epigenetic modifications, control the expression of genes encoding key DNA methylation factors, thus creating feedback loops
83. Moreover, it has recently been shown that methylation of host gene DNA regions bordering the miRNA facilitates ribonuclease-mediated maturation of pre-miRNAs 84. This highlights the complexity of miRNA expression regulation, which depends on transcriptional, epigenetic and post-transcriptional mechanisms involved in miRNA maturation and stability. This underscores the difficulty of establishing causal links between miRNA expression variations. Nevertheless, the co-regulation of several miRNAs may be indicative of an epigenetic modification with a causal link to a given cancer.

miRNAs: FROM MECHANISMS TO THERAPEUTIC SOLUTIONS

Beyond the diagnostic use of the signatures constituted by the expression profile of salivary miRNAs, the aberrant expression of miRNAs associated with cancers can be used to glimpse therapeutic solutions 85. As the functions of miRNAs are essentially defined by the identity of their targets, the action of miRNAs must be considered in the context of gene regulatory networks 86. In some cases, variation in the expression of one or more miRNAs promotes tumour transformation by influencing the expression of a key mediator of a signalling pathway 87 or a tumour suppressor 88. In other cases, a single miRNA may promote tumour development by altering anti-tumour immunity 85, 89 or by stimulating angiogenesis 90. Numerous strategies based on the inhibition of miRNA expression, or conversely on their restoration, have been envisioned 91. Recent progress, linked to the development of RNA vaccines, suggests that the introduction of miRNAs could be exploited therapeutically 92. In cases where cancer is accompanied by the extinction of expression of a key miRNA, it is conceivable to introduce, using lipid nanoparticles, synthetic double-stranded miRNAs (mimicking the miRNA) capable of forming a complex with RISC and targeting the mRNA encoding the overexpressed oncogene. Very recently, a new approach called RiboStrike was developed to identify molecules capable of inhibiting the activity of miRNA-21, which plays a key role in breast cancer development 93. This screen, based on different datasets and using cell-based screens and artificial intelligence, identified from millions of molecules three compounds with miRNA-21 inhibitory activity whose efficacy was validated in mouse models of breast cancer 93. These results demonstrate the power of new therapeutic approaches targeting miRNA activity.

lncRNAs AND piRNAs IN SALIVA: NOVEL BIOMARKERS?

Other non-coding RNAs, apart from miRNAs, have been identified in saliva, broadening the potential spectrum of biomarkers 47. Among these, piRNAs, which range in size from 26 to 31 nucleotides 94, associate after maturation with Piwi proteins to form piRISC complexes involved in target gene silencing through transcriptional and post-transcriptional mechanisms 95. piRNAs are predominantly expressed in germ lines to protect their genome against mobilization of transposable elements by repressing transposon-encoded proteins 96. Their expression is controlled by genetic and epigenetic mechanisms at specific loci, which restrict their expression mainly to germ lines 97. Nevertheless, piRNAs are also expressed in somatic cells, particularly in tumours of different cancer types, in which their expression is correlated with aberrant DNA methylation profiles 98. As such, aberrant expression of piRNAs in cancers can also serve as a biomarker for diagnosis, and potentially some piRNAs could be targeted for therapeutic purposes 99, 100. The most abundant piRNAs are found in all biological fluids and circulate mainly in EVs 47. The concentration of specific piRNAs is particularly high in gastric, colorectal, renal, and prostate cancers 101. Each fluid has a specific piRNA signature indicating that cohorts can potentially be identified based on several classes of small ncRNAs.

 

LncRNAs are another class of RNAs whose expression is deregulated in cancers1, yet their concentration is lower in saliva. They are transcribed, modified, and matured in the cell using the canonical mechanisms of mRNA maturation
102. They act on gene transcription in multiple ways, notably by regulating the recruitment of factors associated with transcription, or by directly or indirectly modifying the state of chromatin or the architecture of the nucleus 103. They also act at the post-transcriptional level through multiple mechanisms, including regulating alternative splicing of mRNAs 104, serving as a platform to create molecular scaffolds 105 or acting as sponges to titrate miRNAs 106. They can also take part in phase separation processes to concentrate macromolecular machineries at their site of action 107.

 

Certain lncRNAs, such as HOTAIR, MALAT1, NEAT1, H19 and XIST1 101 are found deregulated in multiple cancers and are prognostic markers 108, 109, 110, 111, 112, 113, 114, 115. The H19 lncRNA, located on the H19 locus close to the IGF2 (Insulin-like growth factor 2) gene, is particularly interesting since this locus is subject to imprinting: the H19 lncRNA is expressed by the maternal allele, while IGF2 is transcribed by the paternal allele 116. Expression of the two genes is closely coordinated and controlled by methylation of an intergenic region located between the two genes 117. H19 lncRNA has been shown to be deregulated in gynecological cancers and also in endometriosis, suggesting a common etiology between these cancers and endometriosis. The presence of lncRNAs in saliva has not been explored in detail as in the case of miRNAs, notably because of their lower abundance and stability. In saliva, HOTAIR is found overexpressed in pancreatic cancers 118, while MALAT1 is a salivary biomarker of oral epithelial lining cancer 119. Further studies are needed to determine whether lncRNAs in saliva can be biomarkers of cancer or other pathologies.

USE OF SALIVA IN THE DIAGNOSIS OF ENDOMETRIOSIS

Endometriosis is a systemic, heterogeneous disease characterized by the presence of endometrial like tissue outside the uterus, causing intense pelvic pain and a high risk of infertility 120. Although this disease affects 5-10% of women of reproductive age, it is not diagnosed early, complicating the implementation of treatments, which are themselves limited. Endometriosis is classified into three subtypes: superficial peritoneal endometriosis, deep infiltrating endometriosis and endometrial cyst of the ovary 121. It causes systemic inflammation with consequences extending beyond the areas infiltrated by transformed endometrial cells 122. Various models have been proposed to explain the ectopic localization of endometrial cells, which must acquire new properties to migrate and establish themselves in a new environment 123. Endometriosis was initially associated with retrograde menstruation 124, but this condition is clearly not sufficient 122, 123. It is generally proposed that during retrograde menstruation, endometrial stem cells, along with other cells, are deposited on the peritoneum, ovaries and fallopian tubes 125. Genetic or epigenetic alterations in these stem cells 126, 127, combined with hormonal stimulation and environmental cues, contribute to their survival, proliferation, altered differentiation and adhesion to their new environment 128. Cells establish new interactions enabling the growth of endometriosis lesions 128. The proliferation of precursor cells and their differentiation in endometriosis depots is favored by an imbalance in hormonal regulation associated with elevated estrogen expression 120, 121, 122 and various mechanisms affecting the action of progesterone, which normally inhibits estrogen-dependent proliferation of endometrial cells, induces endometrial decidualization and acts against inflammation 129. These mechanisms may be genetic or epigenetic 130, 131. Several environmental signals may contribute to the physiopathology of endometriosis. For example, periodic and repeated bleeding drains erythrocytes whose macrophage-mediated lysis is likely to release heme and iron from hemoglobin degradation into the peritoneal environment 132, 133 thereby inducing the formation of reactive oxygen species leading to oxidative damage of macromolecules contributing to chronic inflammation 134. Notably, local excess of iron can induce ferroptosis, a form of cell death associated with lipid peroxidation 135. Like cancer cells, it is suggested that stromal cells in endometriosis lesions have increased resistance to ferroptosis 136, 137.

 

All this reveals the complexity of the mechanisms behind endometriosis lesions. Very recently, single-cell experiments allowing to assess cell types in normal endometrium 138, 139 and endometriosis lesions 140, 141 have shed new light on this pathology. These studies provide an insight into the specific microenvironment associated with endometrial lesions, consisting of epithelial and stromal cells, infiltrates of immune cells, fibroblasts, vascular and mesothelial cells, illustrating the fact that endometrial lesions are often fibrotic with increased angiogenic capacity 140, 141. They also reveal variations in gene expression between epithelial and stromal cells in endometriosis lesions and those of the normal endometrium 140, 141. Interestingly, stromal cells in endometriosis lesions are more responsive to estrogen than stromal cells in normal endometrium, whereas this difference has not been observed in epithelial cells 141. Moreover, for each cell type within the lesions, specific subtypes can be identified, with proportions varying between normal endometrium and endometriosis lesions 140, 141. For example, the relative abundance of monocyte and macrophage subpopulations and natural killer (NK) subtypes differs between normal endometrium and endometrial lesions, which may explain the tolerance of endometriosis lesions to the action of the immune system and the inflammatory responses associated with endometriosis 140, 141. Similarly, ectopic endometrial deposits show distinct subpopulations of venous endothelial cells and myofibroblasts 140.

 

It is quite clear that in many respects, endometriosis lesions have characteristics like those of tumours, and as such present biomarkers detectable in biological fluids. In normal endometrial function, stromal cells secrete EVs that contain multiple cargos including miRNA that mediate intercellular communication within the endometrium and condition physiological changes in the uterine environment 142. Not surprisingly, EVs, including exosomes, have been purified from plasma and peritoneal fluid in endometriosis patients and healthy controls that reveal unique EV/miRNA signatures in endometriosis patients 143. Variations in miRNA expression have since been observed in the endometrial tissues and various biological fluids of women with endometriosis 144. Several reviews summarize the function of miRNAS that are differentially regulated in samples from patients with endometriosis 145, 146. Although the biological role of certain miRNAs in endometriosis can be addressed 147, the question arises as to their use in diagnosis and as therapeutic targets.

 

A multicentric study with intermediate validation involving a cohort of 200 patients with confirmed endometriosis was conducted in the form of a collaboration between several endometriosis reference centers in France and the Core Facility (iGenSeq) of the Brain and Spinal Cord Institute (ICM, Paris) 148. Using deep-sequencing and machine-based learning algorithm 149 from saliva samples, this study identified a panel of 109 miRNAs constituting a signature with a positive predictive value of 95.1% and a negative predictive value of 86.7% 146. This study has led to the commercialization of a saliva test whose large-scale exploitation could constitute a decisive advance in the diagnosis of endometriosis (https://ziwig.com/endometriose/). Several concerns have been expressed regarding the large-scale use of this test for diagnostic purposes, for example, the consequences of a positive test in an asymptomatic patient, or conversely, a negative test in a symptomatic patient presenting a highly suggestive clinical picture 150. Clearly, the test is not intended for asymptomatic patients, and has a low false-negative rate compared with other diagnostic methods 151. Another interesting question concerns the acceleration of referral of patients with a positive test to specialized services 150. The answer to this question is that a positive test could immediately authorize second-line treatment by primary care physicians (midwives, general physicians, and gynecologists), thereby reducing the need for referral to an expert center and holistic patient management. Another issue is the applicability of the test outside France 150. Rather, the miRNA signature should reflect the consequence of endometriosis depots, irrespective of polymorphisms linked to different ethnicities. Nevertheless, the question is legitimate and deserves attention.

CONCLUSIONS

Saliva is a liquid biopsy with many diagnostic potentialities and advantages. It is easy to collect and store, non-invasive, and contains a large number of molecules, including many miRNAs that have been validated as biomarkers in numerous pathologies. Nevertheless, beyond endometriosis, salivary diagnostics is still in its infancy, and many challenges remain to be overcome, notably to increase the concentration and specificity of biomarker molecules and the sensitivity of molecule detection. Moreover, there is a need to integrate and cross-reference saliva data from patients with different cancers on a large scale, combine several salivary biomarkers, and reduce the heterogeneity associated with the study of the same disease, which sometimes leads to disparate results for the same type of cancer from saliva samples. To increase specificity, the researchers set out to analyze the biomarkers contained in saliva EVs. A growing body of evidence indicates that EVs associated with cancer cells have specific characteristics, and that these EVs are found in saliva. Clinically applicable purification of EVs may represent a decisive step forward in the use of saliva for diagnosis. Finally, beyond diagnosis, the greatest challenge remains to transform omics data from saliva into biological information to understand the mechanisms of pathological situations and identify targets for therapeutic purposes.

References

  1. Slack F J, Chinnaiyan A M (2019). The Role of Non-coding RNAs in Oncology. Cell
    179
    (5)
    : 10331055. doi:10.1016/j.cell.2019.10.017
  2. Xie S, Zhang Q, Jiang L (2022). Current Knowledge on Exosome Biogenesis, Cargo-Sorting Mechanism and Therapeutic Implications. Membranes 12(5): 498. doi:10.3390/membranes12050498
  3. Record M, Carayon K, Poirot M, Silvente-Poirot S (2014). Exosomes as new vesicular lipid transporters involved in cell-cell communication and various pathophysiologies. Biochim Biophys Acta 1841(1): 108120. doi:10.1016/j.bbalip.2013.10.004
  4. Bonne N J, Wong D T (2012). Salivary biomarker development using genomic, proteomic and metabolomic approaches. Genome Med 4(10): 82. doi:10.1186/gm383
  5. Van Niel G V, DAngelo G, Raposo G (2018). Shedding light on the cell biology of extracellular vesicles. Nat Rev Mol Cell Biol 19(4): 213228. doi:10.1038/nrm.2017.125
  6. Dixson A C, Dawson T R, Di Vizio D, Weaver A M (2023). Context-specific regulation of extracellular vesicle biogenesis and cargo selection. Nat Rev Mol Cell Biol 24(7): 454476. doi:10.1038/s41580-023-00576-0
  7. Kalluri R, Lebleu V S (2020). The biology , function , and biomedical applications of exosomes. Science 367(6478): 6977. doi:10.1126/science.aau6977
  8. Jeppesen D K, Fenix A M, Franklin J L, Higginbotham J N, Zhang Q, Zimmerman L J, Liebler D C, Ping J, Liu Q, Evans R, Fissell W H, Patton J G, Rome L H, Burnette D T, Coffey R J (2019). Reassessment of Exosome Composition. Cell 177(2): 428445. doi:10.1126/science.aau6977
  9. Raposo G, Stoorvogel W (2013). Extracellular vesicles: exosomes, microvesicles, and friends. J Cell Biol
    200
    (4)
    : 373383. doi:10.1083/jcb.201211138
  10. Mathieu M, Martin-Jaular L, Lavieu G, Théry C (2019). Specificities of secretion and uptake of exosomes and other extracellular vesicles for cell-to-cell communication. Nat Cell Biol 21(1): 917. doi:10.1038/s41556-018-0250-9
  11. Balaj L, Lessard R, Dai L, Cho Y-J, Pomeroy S L, Breakefield X O, Skog J (2011). Tumour microvesicles contain retrotransposon elements and amplified oncogene sequences. Nat Commun 2(1): 180. doi:10.1038/ncomms1180
  12. Lau C, Kim Y, Chia D, Spielmann N, Eibl G, Elashoff D, Wei F, Lin Y-L, Moro A, Grogan T, Chiang S, Feinstein E, Schafer C, Farrell J, Wong DTW (2013). Role of Pancreatic Cancer-derived Exosomes in Salivary Biomarker Development. J Biol Chem 288(37): 2688826897. doi:10.1074/jbc.M113.452458
  13. Lebleu V S, Kalluri R (2020). Exosomes as a Multicomponent Biomarker Platform in Cancer. Trends Cancer
    6
    (9)
    : 767774. doi:10.1016/j.trecan.2020.03.007
  14. Chiappin S, Antonelli G, Gatti R, De Palo E F (2007). Saliva specimen: a new laboratory tool for diagnostic and basic investigation. Clin Chim Acta 383(1-2): 3040. doi:10.1016/j.cca.2007.04.011
  15. Humphrey S P, Williamson R T (2001). A review of saliva: normal composition, flow, and function. J Prosthet Dent 85(2): 162169. doi:10.1067/mpr.2001.113778
  16. Chicharro J L, Lucía A, Pérez M, Vaquero A F, Ureña R (1998). Saliva composition and exercise. Sports Med
    26
    (1)
    : 1727. doi:10.2165/00007256-199826010-00002
  17. Carpenter G H (2013). The Secretion, Components, and Properties of Saliva. Annu Rev Food Sci Technol
    4
    (1)
    : 267276. doi:10.1146/annurev-food-030212-182700
  18. O’sullivan E M, Dowling P, Swandulla D, Ohlendieck K (2023). Proteomic Identification of Saliva Proteins as Noninvasive Diagnostic Biomarkers. Methods Mol Biol 2596: 147167. doi:10.1146/annurev-food-030212-182700
  19. Drake R R, Cazare L H, Semmes O J, Wadsworth J T (2005). Serum, salivary and tissue proteomics for discovery of biomarkers for head and neck cancers. Expert Rev Mol Diagn 5(1): 93100. doi:10.1586/14737159.5.1.93
  20. Yeh C-K, Christodoulides N J, Floriano P N, Miller C S, Ebersole J L, Weigum S E, Mcdevitt J, Redding S W (2010). Current development of saliva/oral fluid-based diagnostics. Tex Dent J 127(7): 20737986. PMID: 20737986.
  21. Rylander-Rudqvist T, Håkansson N, Tybring G, Wolk A (2006). Quality and Quantity of Saliva DNA Obtained from the Self-administrated Oragene Method-A Pilot Study on the Cohort of Swedish Men. Cancer Epidemiol Biomarkers Prev 15(9): 17421745. doi:10.1158/1055-9965.EPI-05-0706
  22. Urbizu A, Arnaldo L, Beyer K (2023). Obtaining miRNA from Saliva-Comparison of Sampling and Purification Methods. IJMS 24(3): 2386. doi:10.3390/ijms24032386
  23. Nonaka T, Wong DTW (2017). Saliva-Exosomics in Cancer: Molecular Characterization of Cancer-Derived Exosomes in Saliva. Enzymes 42: 125151. doi:10.1016/bs.enz.2017.08.002
  24. Liu A, Hefley B, Escandon P, Nicholas S E, Karamichos D (2023). Salivary Exosomes in Health and Disease: Future Prospects in the Eye. IJMS 24(7): 6363. doi:10.3390/ijms24076363
  25. Pfaffe T, Cooper-White J, Beyerlein P, Kostner K, Punyadeera C (2011). Diagnostic Potential of Saliva: Current State and Future Applications. Clin Chem 57(5): 675687. doi:10.1373/clinchem.2010.153767
  26. James C, Iwasiow R M, Birnboim H C (2011). Human genomic DNA content of saliva samples collected with the Oragene® self-collection kit. DNA Genotek. PD-WP-011
  27. Rylander-Rudqvist T, Håkansson N, Tybring G, Wolk A (2006). Quality and Quantity of Saliva DNA Obtained from the Self-administrated Oragene Method-A Pilot Study on the Cohort of Swedish Men. Cancer Epidemiol Biomarkers Prev 15(9): 17421745. doi:10.1158/1055-9965.EPI-05-0706
  28. Nishitani S, Parets S E, Haas B W, Smith A K (2018). DNA methylation analysis from saliva samples for epidemiological studies. Epigenetics 13(4): 352362. doi:10.1080/15592294.2018.1461295
  29. Han X, Kubota R, Tanaka K, Hayashi H, Seki M, Sakai N, Kawaguchi-Ihara N, Arakawa K, Murohashi I (2021). The correlation of salivary telomere length and single nucleotide polymorphisms of the ADIPOQ, SIRT1 and FOXO3A genes with lifestyle-related diseases in a Japanese population. PLoS ONE 16(1): 243745. doi:10.1371/journal.pone.0243745
  30. Vysotskaia V S, Hogan G J, Gould G M, Wang X, Robertson A D, Haas K R, Theilmann M R, Spurka L, Grauman P V, Lai H H, Jeon D, Haliburton G, Leggett M, Chu C S, Iori K, Maguire J R, Ready K, Evans E A, Kang H P, Haque I S (2017). Development and validation of a 36-gene sequencing assay for hereditary cancer risk assessment. PeerJ 5: 3046. doi:10.7717/peerj.3046
  31. Kumar P, Gupta S, Das BC (2024). Saliva as a potential non-invasive liquid biopsy for early and easy diagnosis/prognosis of head and neck cancer. Transl Oncol 40: 101827. doi:10.1016/j.tranon.2023.101827
  32. Mouliere F (2018). Enhanced detection of circulating tumor DNA by fragment size analysis. Sci Transl Med 10(466): 4921. doi:10.1126/scitranslmed.aat4921
  33. Huang Z, Yang X, Huang Y, Tang Z, Chen Y, Liu H, Huang M, Qing L, Li L, Wang Q, Jie Z, Jin X, Jia B (2023). Saliva – a new opportunity for fluid biopsy. Clin Chem Lab Med 61(1): 432. doi:10.1515/cclm-2022-0793
  34. Yokoi A, Villar-Prados A, Oliphint P A, Zhang J, Song X, De Hoff P, Morey R, Liu J, Roszik J, Clise-Dwyer K, Burks J K , O’Halloran T J, Laurent L C, Sood A K (2019). Mechanisms of nuclear content loading to exosomes. Sci Adv 5(11): 8849. doi:10.1126/sciadv.aax8849
  35. Takahashi A, Okada R, Nagao K, Kawamata Y, Hanyu A, Yoshimoto S, Takasugi M, Watanabe S, Kanemaki M T, Obuse C, Hara E (2017). Exosomes maintain cellular homeostasis by excreting harmful DNA from cells. Nat Commun 8: 15287. doi:10.1038/ncomms15287
  36. Allenson K, Castillo J, San Lucas F A, Scelo G, Kim D U, Bernard V, Davis G, Kumar T, Katz M, Overman M JForetova L, Fabianova E, Holcatova I,Janout, V, Meric-Bernstam F, Gascoyne P, Wistuba I, Varadhachary G, Brennan P, Hanash S, Li D, Maitra A, Alvarez, H (2017). High prevalence of mutant KRAS in circulating exosome-derived DNA from early-stage pancreatic cancer patients. Ann Oncol 28(4): 741747. doi:10.1093/annonc/mdx004
  37. Yang S, Che Spy, Kurywchak P, Tavormina J L, Gansmo L B, Correa De Sampaio P, Tachezy M, Bockhorn M, Gebauer F, Haltom A R, Melo S A, Lebleu V S, Kalluri R (2017). Detection of mutant KRAS and TP53 DNA in circulating exosomes from healthy individuals and patients with pancreatic cancer. Cancer Biol Ther 18(3): 158165. doi:10.1080/15384047.2017.1281499
  38. Castellanos-Rizaldos E, Grimm D G, Tadigotla V, Hurley J, Healy J, Neal P L, Sher M, Venkatesan R, Karlovich C, Raponi M, Krug A, Noerholm M, Tannous J, Tannous B A, Raez L E, Skog J K (2018). Exosome-Based Detection of EGFR T790M in Plasma from Non-Small Cell Lung Cancer Patients. Clin Cancer Res 24(12): 29442950. doi:10.1158/1078-0432.CCR-17-3369
  39. Nakai M, Yamada T, Sekiya K, Sato A, Hankyo M, Kuriyama S, Takahashi G, Kurita T, Yanagihara K, Yoshida H, Ohashi R, Takei H (2022). Use of Liquid Biopsy to Detect PIK3CA Mutation in Metastatic Breast Cancer. J Nippon Med Sch 89(1): 6671. doi:10.1272/jnms.JNMS.2022_89-107
  40. Hyun K-A, Gwak H, Lee J, Kwak B, Jung H-I (2018). Salivary Exosome and Cell-Free DNA for Cancer Detection. Micromachines 9(7): 340. doi:10.3390/mi9070340
  41. Ock F J, Tait SWG (2020). Mitochondria as multifaceted regulators of cell death. Nat Rev Mol Cell Biol 21(2): 85100. doi:10.1038/s41580-019-0173-8
  42. White M J, Mcarthur K, Metcalf D, Lane R M, Cambier J C, Herold M J, Van Delft M F, Bedoui S, Lessene G, Ritchie M E, Huang DCS, Kile BT (2014). Apoptotic caspases suppress mtDNA-induced STING-mediated type I IFN production. Cell 159(7): 15491562. doi:10.1016/j.cell.2014.11.036
  43. Konaka H, Kato Y, Hirano T, Tsujimoto K, Park J, Koba T, Aoki W, Matsuzaki Y, Taki M, Koyama S, Itotagawa E, Jo T, Hirayama T, Kawai T, Ishii K J, Ueda M, Yamaguchi S, Akira S, Morita T, Maeda Y, Nishide M,bNishida S, Shima Y, Narazaki M, Takamatsu H, Kumanogoh A (2023). Secretion of mitochondrial DNA via exosomes promotes inflammation in Behçet’s syndrome. EMBO J 42(20): 112573. doi:10.15252/embj.2022112573
  44. Trumpff C, Rausser S, Haahr R, Karan K R, Gouspillou G, Puterman E, Kirschbaum C, Picard M (2022). Dynamic behavior of cell-free mitochondrial DNA in human saliva. Psychoneuroendocrinology 143: 105852. doi:10.1016/j.psyneuen.2022.105852
  45. Sayal L, Hamadah O, Almasri A, Idrees M, Kassem I, Habbal W, Alsalamah B, Kujan O (2023). Salivary-Based Cell-Free Mitochondrial DNA Level Is an Independent Prognostic Biomarker for Patients with Head and Neck Squamous Cell Carcinoma. JPM 13(2): 301. doi:10.3390/jpm13020301
  46. Malik A N, Czajka A (2013). Is mitochondrial DNA content a potential biomarker of mitochondrial dysfunction? Mitochondrion 13: 481492. doi:10.3390/jpm13020301
  47. Bahn J H, Zhang Q, Li F, Chan T-M, Lin X, Kim Y, Wong DTW, Xiao X (2015). The landscape of microRNA, Piwi-interacting RNA, and circular RNA in human saliva. Clin Chem 61(1): 221230. doi:10.1373/clinchem.2014.230433
  48. Cui J, Chen M, Zhang L, Huang S, Xiao F, Zou L (2022). Circular RNAs: Biomarkers of cancer. Cancer Innov 1(3): 197206. doi:10.1002/cai2.28
  49. Lau N C, Lim L P, Weinstein E G, Bartel D P (2001). An abundant class of tiny RNAs with probable regulatory roles in Caenorhabditis elegans. Science 294(5543): 858862. doi:10.1126/science.1065062
  50. Lee R C, Ambros V (2001). An extensive class of small RNAs in Caenorhabditis elegans. Science 294(5543): 862864. doi:10.1126/science.1065329
  51. Lagos-Quintana M, Rauhut R, Lendeckel W (2001). Identification of novel genes coding for small expressed RNAs. Science 294(5543): 853858. doi:10.1126/science.1064921
  52. Bartel D P (2018). Metazoan MicroRNAs. Cell 173(1): 2051. doi:10.1016/j.cell.2018.03.006
  53. Treiber T, Treiber N, Meister G (2019). Regulation of microRNA biogenesis and its crosstalk with other cellular pathways. Nat Rev Mol Cell Biol 20(1): 520. doi:10.1038/s41580-018-0059-1
  54. Doench J G, Sharp P A (2004). Specificity of microRNA target selection in translational repression. Genes Dev 18(5): 504511. doi:10.1101/gad.1184404
  55. Hum Humphreys D T, Westman B J , Martin DIK, Preiss T (2005). MicroRNAs control translation initiation by inhibiting eukaryotic initiation factor 4E/cap and poly(A) tail function. Proc Natl Acad Sci U S A
    102
    (47)
    : 1696116966. doi:10.1073/pnas.0506482102
  56. Friedman R C, Farh KK-H, Burge C B, Bartel D P (2009). Most mammalian mRNAs are conserved targets of microRNAs. Genome Res 19(1): 92105. doi:10.1101/gr.082701.108
  57. Alles J, Fehlmann T, Fischer U, Backes C, Galata V, Minet M, Hart M, Abu-Halima M, Grässer F A, Lenhof H-P, Keller A, Meese E (2019). An estimate of the total number of true human miRNAs. Nucleic Acids Res 47(7): 33533364. doi:10.1093/nar/gkz097
  58. Mori M A, Ludwig R G, Garcia-Martin R, Brandão B B, Kahn C R (2019). Extracellular miRNAs: From Biomarkers to Mediators of Physiology and Disease. Cell Metab 30(4): 656673. doi:10.1016/j.cmet.2019.07.011
  59. Garcia-Martin R, Wang G, Brandão B B, Zanotto T M, Shah S, Kumar Patel S, Schilling B, Kahn C R (2022). MicroRNA sequence codes for small extracellular vesicle release and cellular retention. Nature 601(7893): 446451. doi:10.1038/s41586-021-04234-3
  60. O’Brien J, Hayder H, Zayed Y, Peng C (2018). Overview of MicroRNA Biogenesis, Mechanisms of Actions, and Circulation. Front Endocrinol 9: 402. doi:10.3389/fendo.2018.00402
  61. Gallo A, Tandon M, Alevizos I, Illei G G (2012). The majority of microRNAs detectable in serum and saliva is concentrated in exosomes. PLoS One 7(3): 30679. doi:10.1371/journal.pone.0030679
  62. Chen X, Ba Y, Ma L, Cai X, Yin Y, Wang K, Guo J, Zhang Y, Chen J, Guo X, Li Q, Li X, Wang W, Zhang Y, Wang J, Jiang X, Xiang Y, Xu C, Zheng P, Zhang J, Li R, Zhang H, Shang X, Gong T, Ning G, Wang J, Zen K, Zhang JZhang C-Y (2008). Characterization of microRNAs in serum: a novel class of biomarkers for diagnosis of cancer and other diseases. Cell Res 18(10): 9971006. doi:10.1038/cr.2008.282
  63. Han Y, Jia L, Zheng Y, Li W (2018). Salivary Exosomes: Emerging Roles in Systemic Disease. Int J Biol Sci
    14
    (6)
    : 633643. doi:10.7150/ijbs.25018
  64. Li F, Yoshizawa J M, Kim K-M, Kanjanapangka J, Grogan T R, Wang X, Elashoff D E, Ishikawa S, Chia D, Liao W, Akin D, Yan X, Lee M-S, Choi R, Kim S-M, Kang S-Y, Bae J-M, Sohn T-S, Lee J-H, Choi M-G, Min B-H, Lee J-H, Kim J J, Kim Y, Kim S, Wong DTW (2018). Discovery and Validation of Salivary Extracellular RNA Biomarkers for Noninvasive Detection of Gastric Cancer. Clin Chem 64(10): 15131521. doi:10.1373/clinchem.2018.290569
  65. Sazanov A A, Kiselyova E V, Zakharenko A A, Romanov M N, Zaraysky M I (2017). Plasma and saliva miR-21 expression in colorectal cancer patients. J Appl Genet 58(2): 231237. doi:10.1007/s13353-016-0379-9
  66. Rapado-González Ó, Majem B, Álvarez-Castro A, Díaz-Peña R, Abalo A, Suárez-Cabrera L, Gil-Moreno A, Santamaría A, López-López R, Muinelo-Romay L, Suarez-Cunqueiro MM (2019). A Novel Saliva-Based miRNA Signature for Colorectal Cancer Diagnosis. J Clin Med 8(12): 2029. doi:10.3390/jcm8122029
  67. Liu M, Yu X, Bu J, Xiao Q, Ma S, Chen N, Qu C (2023). Comparative analyses of salivary exosomal miRNAs for patients with or without lung cancer. Front Genet 14: 1249678. doi:10.3389/fgene.2023.1249678
  68. Raczkowska J, Bielska A, Krętowski A, Niemira M (2023). Extracellular circulating miRNAs as potential non-invasive biomarkers in non-small cell lung cancer patients. Front Oncol 13: 1209299. doi:10.3389/fonc.2023.1209299
  69. Mariam A, Miller-Atkins G, Moro A, Rodarte A I, Siddiqi S, Acevedo-Moreno L-A, Brown J M, Allende D S, Aucejo F, Rotroff D M (2022). Salivary miRNAs as non-invasive biomarkers of hepatocellular carcinoma: a pilot study. PeerJ 10: 12715. doi:10.7717/peerj.12715
  70. Humeau M, Vignolle-Vidoni A, Sicard F, Martins F, Bournet B, Buscail L, Torrisani J, Cordelier P (2015). Salivary MicroRNA in Pancreatic Cancer Patients. PLoS ONE 10(6): 130996. doi:10.1371/journal.pone.0130996
  71. Xie Z, Yin X, Gong B, Nie W, Wu B, Zhang X, Huang J, Zhang P, Zhou Z, Li Z (2015). Salivary microRNAs show potential as a noninvasive biomarker for detecting resectable pancreatic cancer. Cancer Prev Res
    8
    (2)
    : 165173. doi:10.1371/journal.pone.0130996
  72. Robotti M, Scebba F, Angeloni D (2023). Circulating Biomarkers for Cancer Detection: Could Salivary microRNAs Be an Opportunity for Ovarian Cancer Diagnostics? Biomedicines 11(3): 652. doi:10.3390/biomedicines11030652
  73. Fanale D, Corsini L R, Bono M, Randazzo U, Barraco N, Brando C, Cancelliere D, Contino S, Giurintano A, Magrin L, Pedone E, Perez A, Piraino P, Pivetti A, Giovanni E D, Russo TDB, Prestifilippo O, Gennusa V, Pantuso G, Russo A, Bazan V (2024). Clinical relevance of exosome-derived microRNAs in Ovarian Cancer: Looking for new tumor biological fingerprints. Crit Rev Oncol Hematol 193: 104220. doi:10.1016/j.critrevonc.2023.104220
  74. Dabi Y, Favier A, Razakamanantsoa L, Delbos L, Poilblanc M, Descamps P, Golfier F, Touboul C, Bendifallah S,
    Daraï E  (2023). Insight on Non-Coding RNAs from Biofluids in Ovarian Tumors. Cancers 15(5): 1539. doi:10.3390/cancers15051539
  75. Castro-Cruz M, Hyka L, Daaboul G, Leblanc R, Meeussen S, Lembo F, Oris A, Van Herck L Van, Granjeaud S, David G, Zimmermann P (2023). PDZ scaffolds regulate extracellular vesicle production, composition, and uptake. Proc Natl Acad Sci U S A 120(38): 2310914120. doi:10.1073/pnas.2310914120
  76. Castillo J, Bernard V, San Lucas F A, Allenson K, Capello M, Kim D U, Gascoyne P, Mulu F C, Stephens B M, Huang J, Wang H, Momin A A, Jacamo R O, Katz M, Wolff R, Javle M, Varadhachary G, Wistuba I I, Hanash S, Maitra A, Alvarez H (2018). Surfaceome profiling enables isolation of cancer-specific exosomal cargo in liquid biopsies from pancreatic cancer patients. Ann Oncol 29(1): 223229. doi:10.1093/annonc/mdx542
  77. Baietti M F, Zhang Z, Mortier E, Melchior A, Degeest G, Geeraerts A, Ivarsson Y, Depoortere F, Coomans C, Vermeiren E, Zimmermann P, David G (2012). Syndecan-syntenin-ALIX regulates the biogenesis of exosomes. Nat Cell Biol 14(7): 67785. doi:10.1038/ncb2502
  78. Ghossoub R, Chéry M, Audebert S, Leblanc R, Egea-Jimenez A L, Lembo F, Mammar S, Le Dez F, Camoin L, Borg J P, Rubinstein E, David G, Zimmermann P (2020). Tetraspanin-6 negatively regulates exosome production. Proc Natl Acad Sci U S A 117(11): 59135922. doi:10.1073/pnas.1922447117
  79. Oto J, Le Q-K, Schäfer S D, Kiesel L, Marí-Alexandre J, Gilabert-Estellés J, Medina P, Götte M (2023). Role of Syndecans in Ovarian Cancer: New Diagnostic and Prognostic Biomarkers and Potential Therapeutic Targets. Cancers 15(12): 3125. doi:10.3390/cancers15123125
  80. Gulyaeva L F, Kushlinskiy N E (2016). Regulatory mechanisms of microRNA expression. J Transl Med 14(1): 143. doi:10.1186/s12967-016-0893-x
  81. Liu B, Shyr Y, Cai J, Liu Q (2019). Interplay between miRNAs and host genes and their role in cancer. Brief Funct Genomics 18(4): 255266. doi:10.1093/bfgp/elz002
  82. Saviana M, Le P, Micalo L, Del Valle-Morales D, Romano G, Acunzo M, Li H, Nana-Sinkam P (2023). Crosstalk between miRNAs and DNA Methylation in Cancer. Genes 14(5): 1075. doi:10.3390/genes14051075
  83. Wang S, Wu W, Claret F X (2017). Mutual regulation of microRNAs and DNA methylation in human cancers. Epigenetics 12(3): 187197. doi:10.1080/15592294.2016.1273308
  84. Glaich O, Parikh S, Bell R E, Mekahel K, Donyo M, Leader Y, Shayevitch R, Sheinboim D, Yannai S, Hollander D, Melamed Z, Lev-Maor G, Ast G, Levy C (2019). DNA methylation directs microRNA biogenesis in mammalian cells. Nat Commun 10(1): 5657. doi:10.1038/s41467-019-13527-1
  85. Nemeth K, Bayraktar R, Ferracin M, Calin G A (2024). Non-coding RNAs in disease: from mechanisms to therapeutics. Nat Rev Genet 25(3): 211232. doi:10.1038/s41467-019-13527-1
  86. Duchaine T F, Fabian M R (2019). Mechanistic Insights into MicroRNA-Mediated Gene Silencing. Cold Spring Harb Perspect Biol 11(3): 32771. doi:10.1101/cshperspect.a032771
  87. Selvakumar S C, Preethi K A, Sekar D (2023). MicroRNAs as important players in regulating cancer through PTEN/PI3K/AKT signalling pathways. Biochim Biophys Acta Rev Cancer 1878(3): 188904. doi:10.1016/j.bbcan.2023.188904
  88. Fu J, Imani S, Wu M-Y, Wu R-C (2019). MicroRNA-34 Family in Cancers: Role, Mechanism, and Therapeutic Potential. Cancers 15: 4723. doi:10.1016/j.bbcan.2023.188904
  89. Kalkusova K, Taborska P, Stakheev D, Smrz D (2022). The Role of miR-155 in Antitumor Immunity. Cancers
    14
    (21)
    : 5414. doi:10.3390/cancers14215414
  90. Landskroner-Eiger S, Moneke I, Sessa W C (2013). miRNAs as modulators of angiogenesis. Cold Spring Harb Perspect Med 3(2): 6643. doi:10.3390/cancers14215414
  91. Winkle M, El-Daly S M, Fabbri M, Calin G A (2021). Noncoding RNA therapeutics – challenges and potential solutions. Nat Rev Drug Discov 20(8): 629651. doi:10.1038/s41573-021-00219-z
  92. Matsui M, Corey D R (2017). Non-coding RNAs as drug targets. Nat Rev Drug Discov 16(3): 167179. doi:
    10.1038/nrd.2016.117
  93. A Keshavarzi Arshadi, Salem M, Karner H, Garcia K, Arab A, Yuan J S, Goodarzi H (2024). Functional microRNA-targeting drug discovery by graph-based deep learning. Patterns 5(1): 100909. doi:10.1016/j.patter.2023.100909
  94. Sai Lakshmi S, Agrawal S (2008). piRNABank: a web resource on classified and clustered Piwi-interacting RNAs. Nucleic Acids Res 36: D1737. doi:10.1093/nar/gkm696
  95. Ramat A, Simonelig M (2021). Functions of PIWI Proteins in Gene Regulation: New Arrows Added to the piRNA Quiver. Trends Genet 37(2): 188200. doi:10.1016/j.tig.2020.08.011
  96. Czech B, Munafò M, Ciabrelli F, Eastwood E L, Fabry M H, Kneuss E, Hannon G J (2018). piRNA-Guided Genome Defense: From Biogenesis to Silencing. Annu Rev Genes 52(1): 131157. doi:10.1146/annurev-genet-120417-031441
  97. Yu T, Fan K, Özata D M, Zhang G, Fu Y, Theurkauf W E, Zamore P D, Weng Z (2021). Long first exons and epigenetic marks distinguish conserved pachytene piRNA clusters from other mammalian genes. Nat Commun 12(1): 73. doi:10.1038/s41467-020-20345-3
  98. Wu Z, Yu X, Zhang S, He Y, Guo W (2023). Novel roles of PIWI proteins and PIWI-interacting RNAs in human health and diseases. Cell Commun Signal 21(1): 343. doi:10.1186/s12964-023-01368-x
  99. Taverna S, Masucci A, Cammarata G (2023). PIWI-RNAs Small Noncoding RNAs with Smart Functions: Potential Theranostic Applications in Cancer. Cancers 15(15): 3912. doi:10.3390/cancers15153912
  100. El-Mogy M, Lam B, Haj-Ahmad T A, Mcgowan S, Yu D, Nosal L, Rghei N, Roberts P, Haj-Ahmad Y (2018). Diversity and signature of small RNA in different bodily fluids using next generation sequencing. BMC Genomics 19(1): 408. doi:10.1186/s12864-018-4785-8
  101. Pardini B, Sabo A A, Birolo G, Calin G A (2019). Noncoding RNAs in Extracellular Fluids as Cancer Biomarkers: The New Frontier of Liquid Biopsies. Cancers 11(8): 1170. doi:10.3390/cancers11081170
  102. Iyer M K, Niknafs Y S, Malik R, Singhal U, Sahu A, Hosono Y, Barrette T R, Prensner J R, Evans J R, Zhao S, Poliakov A, Cao X, Dhanasekaran S M, Wu Y-M, Robinson D R, Beer D G, Feng F Y, Iyer H K, Chinnaiyan A M (2015). The landscape of long noncoding RNAs in the human transcriptome. Nat Genet 47(3): 199208. doi:10.1038/ng.3192
  103. Long Y, Wang X, Youmans D T, Cech T R (2017). How do lncRNAs regulate transcription? Sci Adv 3(9): 2110. doi:10.1126/sciadv.aao2110
  104. Malakar P, Shukla S, Mondal M, Kar R K, Siddiqui J A (2024). The nexus of long noncoding RNAs, splicing factors, alternative splicing and their modulations. RNA Biology 21(1): 120. doi:10.1080/15476286.2023.2286099
  105. Obuse C, Hirose T (2023). Functional domains of nuclear long noncoding RNAs: Insights into gene regulation and intracellular architecture. Curr Opin Cell Biol 85: 102250. doi:10.1016/j.ceb.2023.102250
  106. Bhattacharjee R, Prabhakar N, Kumar L, Bhattacharjee A, Kar S, Malik S, Kumar D, Ruokolainen J, Negi A, Jha N K, Kesari K K (2023). Crosstalk between long noncoding RNA and microRNA in Cancer. Cell Oncol 46(4): 885908. doi:10.1007/s13402-023-00806-9
  107. Mattick J S, Amaral P P, Carninci P, Carpenter S, Chang H Y, Chen L-L, Chen R, Dean C, Dinger M E, Fitzgerald K A, Gingeras T R, Guttman M, Hirose T, Huarte M, Johnson R, Kanduri C, Kapranov P, Lawrence J B, Lee J T, Mendell J T, Mercer T R, Moore K J, Nakagawa S, Rinn J L, Spector D L, Ulitsky I, Wan Y, Wilusz J E,Wu M (2023). Long non-coding RNAs: definitions, functions, challenges and recommendations. Nat Rev Mol Cell Biol 24(6): 430447. doi:10.1038/s41580-022-00566-8
  108. An C, Wang I, Li X, Xia R, Deng F (2022). Long non-coding RNA in prostate cancer. Am J Clin Exp Urol 10(3): 35874287. PMID:35874287.
  109. Shetty A, Venkatesh T, Kabbekodu S P, Tsutsumi R, Suresh P S (2022). LncRNA-miRNA-mRNA regulatory axes in endometrial cancer: a comprehensive overview. Arch Gynecol Obstet 306(5): 14311447. doi:10.1007/s00404-022-06423-5
  110. Cao Z, Oyang L, Luo X, Xia L, Hu J, Lin J, Tan S, Tang Y, Zhou Y, Cao D, Liao Q (2022). The roles of long non-coding RNAs in lung cancer. J Cancer 13(1): 174183. doi:10.7150/jca.65031
  111. Goyal B, Yadav SRM, Awasthee N, Gupta S, Kunnumakkara A B, Gupta S C (2021). Diagnostic, prognostic, and therapeutic significance of long non-coding RNA MALAT1 in cancer. Biochim Biophys Acta Rev Cancer 1875(2): 188502. doi:10.1016/j.bbcan.2021.188502
  112. Chowdhury P R, Salvamani S, Gunasekaran B, Peng H B, Ulaganathan V (2023). H19: An Oncogenic Long Non-coding RNA in ColorectalCancer. Yale J Biol Med 96(4): 495509. doi:10.59249/TDBJ7410
  113. Richart L, Picod-Chedotel M-L, Wassef M, Macario M, Aflaki S, Salvador M A, Héry T, Dauphin A, Wicinski J, Chevrier V, Pastor S, Guittard G, Le Cam S, Kamhawi H, Castellano R, Guasch G, Charafe-Jauffret E, Heard E, Margueron R, Ginestier C (2022). XIST loss impairs mammary stem cell differentiation and increases tumorigenicity through Mediator hyperactivation. Cell 185(12): 21642183. doi:10.1016/j.cell.2022.04.034
  114. Zhou H, Wang Y, Liu Z, Zhang Z, Xiong L, Wen Y (2022). Recent advances of NEAT1-miRNA interactions in cancer. ABBS 54(2): 153162. doi:10.3724/abbs.2021022
  115. Raju G S R, Pavitra E, Bandaru S S, Varaprasad G L, Nagaraju G P, Malla R R, Huh Y S, Han Y-K (2023). HOTAIR: a potential metastatic, drug-resistant and prognostic regulator of breast cancer. Mol Cancer 22(1): 65. doi:10.1186/s12943-023-01765-3
  116. Gabory A, Jammes H, Dandolo L (2010). The H19 locus: role of an imprinted non-coding RNA in growth and development. Bioessays 32(6): 473480. doi:10.1002/bies.200900170
  117. Lopes S, Lewis A, Hajkova P, Dean W, Oswald J, Forné T, Murrell A, Constância M, Bartolomei M, Walter J, Reik W (2003). Epigenetic modifications in an imprinting cluster are controlled by a hierarchy of DMRs suggesting long-range chromatin interactions. Hum Mol Genet 12(3): 295305. doi:10.1093/hmg/ddg022
  118. Xie Z, Chen X, Li J, Guo Y, Li H, Pan X, Jiang J, Liu H, Wu B (2016). Salivary HOTAIR and PVT1 as novel biomarkers for early pancreatic cancer. Oncotarget 7(18): 2540825419. doi:10.18632/oncotarget.8323
  119. Shalaby R, Ibrahim S, Kotb AAW, Baz S, Hafed L, Shaker O, Afifi S (2023). MALAT1 as a potential salivary biomarker in oral squamous cell carcinoma through targeting miRNA-124. Oral Dis. doi:10.1111/odi.14730
  120. Chapron C, Marcellin L, Borghese B, Santulli P (2019). Rethinking mechanisms, diagnosis and management of endometriosis. Nat Rev Endocrinol 15(11): 666682. doi:10.1038/s41574-019-0245-z
  121. Wang Y, Nicholes K, Shih I-M (2020). The Origin and Pathogenesis of Endometriosis. Annu Rev Pathol Mech Dis 15(1): 7195. doi:10.1146/annurev-pathmechdis-012419-032654
  122. Taylor H S, Kotlyar A M, Flores V A (2021). Endometriosis is a chronic systemic disease: clinical challenges and novel innovations. Lancet 397: 839852. doi:10.1016/S0140-6736(21)00389-5
  123. Zondervan K T, Becker C M, Koga K, Missmer S A, Taylor R N, Viganò P (2018). Endometriosis. Nat Rev Dis Primers 4(1): 9. doi:10.1038/s41572-018-0008-5
  124. Halme J, Hammond M G, Hulka J F, Raj S G, Talbert L M (1984). Retrograde menstruation in healthy women and in patients with endometriosis. Obstet Gynecol 64(2): 6234483. PMID:6234483.
  125. Hufnagel D, Li F, Cosar E, Krikun G, Taylor H S (2015). The Role of Stem Cells in the Etiology and Pathophysiology of Endometriosis. Semin Reprod Med 33(5): 333340. doi:10.1055/s-0035-1564609
  126. Lalami I, Abo C, Borghese B, Chapron C, Vaiman D (2021). Genomics of Endometriosis: From Genome Wide Association Studies to Exome Sequencing. IJMS 22(14): 7297. doi:10.3390/ijms22147297
  127. Pais A S, Almeida-Santos T (2023). Recent insights explaining susceptibility to endometriosis-From genetics to environment. WIREs Mech Dis 15(6): 1624. doi:10.1002/wsbm.1624
  128. Hong I-S (2024). Endometrial Stem Cells: Orchestrating Dynamic Regeneration of Endometrium and Their Implications in Diverse Endometrial Disorders. Int J Biol Sci 20(3): 864879. doi:10.7150/ijbs.89795
  129. Zhang P, Wang G (2023). Progesterone Resistance in Endometriosis: Current Evidence and Putative Mechanisms. IJMS 24(8): 6992. doi:10.3390/ijms24086992
  130. Zukiewicz D, Stangret A, Ruiz-Ruiz C, Olivares E G, Soriţău O, Suşman S, Szewczyk G (2021). Estrogen- and Progesterone (P4)-Mediated Epigenetic Modifications of Endometrial Stromal Cells (EnSCs) and/or Mesenchymal Stem/Stromal Cells (MSCs) in the Etiopathogenesis of Endometriosis. Stem Cell Rev and Rep
    17
    (4)
    : 11741193. doi:10.1007/s12015-020-10115-5
  131. Rossi M, Seidita I, Vannuccini S, Prisinzano M, Donati C, Petraglia F (2023). Epigenetics, endometriosis and sex steroid receptors: An update on the epigenetic regulatory mechanisms of estrogen and progesterone receptors in patients with endometriosis. Vitam Horm 171191. doi:10.1016/bs.vh.2023.01.007
  132. Van Langendonckt A, Casanas-Roux F, Donnez J (2002). Iron overload in the peritoneal cavity of women with pelvic endometriosis. Fertil Steril 78(4): 712718. doi:10.1016/s0015-0282(02)03346-0
  133. Van Langendonckt A, Casanas-Roux F, Dolmans M-M, Donnez J (2002). Potential involvement of hemoglobin and heme in the pathogenesis of peritoneal endometriosis. Fertil Steril 77(3): 561570. doi:10.1016/s0015-0282(01)03211-3
  134. Donnez J, Binda M M, Donnez O, Dolmans M-M (2016). Oxidative stress in the pelvic cavity and its role in the pathogenesis of endometriosis. Fertil Steril 106(5): 10111017. doi:10.1016/j.fertnstert.2016.07.1075
  135. Wyatt J, Fernando S M, Powell S G, Hill C J, Arshad I, Probert C, Ahmed S, Hapangama D K (2023). The role of iron in the pathogenesis of endometriosis: a systematic review. Human Reproduction Open 2023(3): 33. doi:10.1093/hropen/hoad033
  136. Chen B, Zhao L, Yang R, Xu T (2023). The recent advancements of ferroptosis in the diagnosis, treatment and prognosis of ovarian cancer. Front Genet 14: 1275154. doi:10.3389/fgene.2023.1275154
  137. Wu Q, Liang Z, Jiang J, Feng X, Liu J, Zhang Z, Wang H, Wang N, Gou Y, Li Z, Cao Y (2023). Macrophages originated IL-33/ST2 inhibits ferroptosis in endometriosis via the ATF3/SLC7A11 axis. Cell Death Dis 14(10): 668. doi:10.1038/s41419-023-06182-4
  138. Wang W, Vilella F, Alama P, Moreno I, Mignardi M, Isakova A, Pan W, Simon C, Quake S R (2020). Single-cell transcriptomic atlas of the human endometrium during the menstrual cycle. Nat Med 26(10): 16441653. doi:10.1038/s41591-020-1040-z
  139. Garcia-Alonso L (2021). Mapping the temporal and spatial dynamics of the human endometrium in vivo and in vitro. Nat Genet 53(12): 16981711. doi:10.1038/s41588-021-00972-2
  140. Tan Y, Flynn W F, Sivajothi S, Luo D, Bozal S B, Davé M, Luciano A A, Robson P, Luciano D E, Courtois E T (2022). Single-cell analysis of endometriosis reveals a coordinated transcriptional programme driving immunotolerance and angiogenesis across eutopic and ectopic tissues. Nat Cell Biol 24(8): 13061318. doi:10.1038/s41556-022-00961-5
  141. Shin S, Chung Y, Moon S W, Choi E J, Kim M, Chung Y, Lee S H (2023). Single-cell profiling identifies distinct hormonal, immunologic, and inflammatory signatures of endometriosis-constituting cells. J Pathol 261(3): 323334. doi:10.1002/path.6178
  142. Beal J R, Ma Q, Bagchi I C, Bagchi M K (2023). Role of Endometrial Extracellular Vesicles in Mediating Cell-to-Cell Communication in the Uterus: A Review. Cells 12(22): 2584. doi:10.3390/cells12222584
  143. Khalaj K, Miller J E, Lingegowda H, Fazleabas A T, Young S L, Lessey B A, Koti M, Tayade C (2019). Extracellular vesicles from endometriosis patients are characterized by a unique miRNA-lncRNA signature. JCI Insight 4(18): 128846. doi:10.1172/jci.insight.128846
  144. Begum MIA, Chuan L, Hong S-T, Chae H-S (2023). The Pathological Role of miRNAs in Endometriosis. Biomedicines 11(11): 3087. doi:10.3390/biomedicines11113087
  145. Ghafouri-Fard S, Shoorei H, Anamag F T, Taheri M (2020). The Role of Non-Coding RNAs in Controlling Cell Cycle Related Proteins in Cancer Cells. Front Oncol 10: 608975. doi:10.3389/fonc.2020.608975
  146. Zhang Y, Sun X, Li Z, Han X, Wang W, Xu P, Liu Y, Xue Y, Wang Z, Xu S, Wang X, Li G, Tian Y, Zhao Q (2024). Interactions between miRNAs and the Wnt/β-catenin signaling pathway in endometriosis. Biomed Pharmacother 171: 116182. doi:10.1016/j.biopha.2024.116182
  147. Nazri H M, Greaves E, Quenby S, Dragovic R, Tapmeier T T, Becker C M (2023). The role of small extracellular vesicle-miRNAs in endometriosis. Hum Reprod 38(12): 22962311. doi:10.1093/humrep/dead216
  148. Ben Bendifallah S, Dabi Y, Suisse S, Delbos L, Spiers A, Poilblanc M, Golfier F, Jornea L, Bouteiller D, Fernandez H, Madar A, Petit E, Perotte F, Fauvet R, Benjoar M, Akladios C, Lavoué V, Darnaud T, Merlot B, Roman H, Touboul C, Descamps P (2023). Validation of a Salivary miRNA Signature of Endometriosis – Interim Data. NEJM Evidence 2(7): 2200282. doi:10.1056/EVIDoa2200282
  149. Bendifallah S, Dabi Y, Suisse S, Jornea L, Bouteiller D, Touboul C, Puchar A, Daraï E (2022). A Bioinformatics Approach to MicroRNA-Sequencing Analysis Based on Human Saliva Samples of Patients with Endometriosis. IJMS 23(14): 8045. doi:10.3390/ijms23148045
  150. Scheck S M, Henry C, Bedford N, Abbott J, Wynn-Williams M, Yazdani A, Mcdowell S (2023). Non-invasive tests for endometriosis are here; how reliable are they, and what should we do with the results? Aust N Z J Obstet Gynaecol 64(2): 168170. doi:10.1111/ajo.13765
  151. Pascoal E, Wessels J M, Aas-Eng M K, Abrao M S, Condous G, Jurkovic D, Espada M, Exacoustos C, Ferrero S, Guerriero S, Hudelist G, Malzoni M, Reid S, Tang S, Tomassetti C, Singh S S,  T Van Den Bosch, Leonardi M (2022). Strengths and limitations of diagnostic tools for endometriosis and relevance in diagnostic test accuracy research. Ultrasound Obstet Gynecol 60(3): 309327. doi:10.1002/uog.24892

 

ACKNOWLEDGMENTS

We would like to thank Professor Emile Darai for his critical reading of the manuscript and his advice.

COPYRIGHT

© 2024

Creative Commons License
Saliva, a molecular reflection of the human body? Implications for diagnosis and treatment by Géli and Nabet is licensed under a Creative Commons Attribution 4.0 International License.

By continuing to use the site, you agree to the use of cookies. more information

The cookie settings on this website are set to "allow cookies" to give you the best browsing experience possible. If you continue to use this website without changing your cookie settings or you click "Accept" below then you are consenting to this. Please refer to our "privacy statement" and our "terms of use" for further information.

Close