Next Article in Journal
Generation and Validation of Normative, Age-Specific Reference Curves for Bone Strain Index in Women
Previous Article in Journal
Epithelial Cell Adhesion Molecule (EpCAM) Expression in Human Tumors: A Comparison with Pan-Cytokeratin and TROP2 in 14,832 Tumors
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

A Comprehensive Review on Circulating cfRNA in Plasma: Implications for Disease Diagnosis and Beyond

by
Pengqiang Zhong
1,†,
Lu Bai
1,†,
Mengzhi Hong
1,
Juan Ouyang
1,
Ruizhi Wang
1,
Xiaoli Zhang
2,* and
Peisong Chen
1,*
1
Department of Clinical Laboratory, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China
2
Department of Pediatrics, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Diagnostics 2024, 14(10), 1045; https://doi.org/10.3390/diagnostics14101045
Submission received: 14 April 2024 / Revised: 13 May 2024 / Accepted: 15 May 2024 / Published: 17 May 2024

Abstract

:
Circulating cfRNA in plasma has emerged as a fascinating area of research with potential applications in disease diagnosis, monitoring, and personalized medicine. Circulating RNA sequencing technology allows for the non-invasive collection of important information about the expression of target genes, eliminating the need for biopsies. This comprehensive review aims to provide a detailed overview of the current knowledge and advancements in the study of plasma cfRNA, focusing on its diverse landscape and biological functions, detection methods, its diagnostic and prognostic potential in various diseases, challenges, and future perspectives.

1. Introduction

In recent times, there has been a rapid advancement in liquid biopsy technology. In comparison to conventional tissue biopsy, liquid biopsy technology offers several advantages, including reduced invasiveness, enhanced repeatability, the ability to overcome tumor molecular spatial heterogeneity, and the capacity to dynamically reflect tumor changes [1]. Consequently, it has found extensive application in early cancer screening, guiding patients towards adjuvant therapy, evaluating the effectiveness of chemotherapy, and providing insights into tumor prognosis [2]. Liquid biopsy technology, a highly anticipated early-detection technology for cancer [1,3,4], encompasses markers such as circulating tumor cells (CTCs), extracellular DNA (cfDNA) and its methylation, extracellular cell-free RNA (cfRNA), and exosomal proteins [5]. Among the various liquid biopsy samples, plasma cell-free DNA (cfDNA) and plasma cfRNA in peripheral blood have garnered significant attention. These DNA or RNA molecules are products of normal cell metabolism and apoptosis, as well as tumor cell activity, and they hold substantial value for clinical applications.
cfRNA, also known as extracellular RNA (exRNA), is found in bodily fluids such as blood, urine, alveolar lavage fluid, and pleural fluid. As showed in Figure 1, it primarily consists of various types of RNA, including messenger RNA (mRNA), small RNA (miRNA), long non-coding RNAs (lncRNAs), and circular RNA (circRNA). Circulating free RNA is the predominant form of free RNA existing in blood. The exact source of cfRNA remains uncertain. Zhou et al. conducted an in vitro study which revealed that the concentration of exRNA can increase under conditions of hypoxia and increased cell metabolism [6]. This finding may offer a possible explanation for the elevated levels of cfRNA observed in cancer patients. However, this has not yet been validated through in vivo testing. cfRNA originates from cellular activities such as apoptosis, necrosis, or active secretion, and can be released from the nucleus, cytoplasm, or extracellular vesicles (e.g., exosomes). Circulating cfRNA, released into the extracellular environment and present in body fluids such as plasma, holds immense promise as a non-invasive source of genetic information. The analysis of plasma cfRNA offers a unique opportunity for early disease detection, monitoring treatment responses, and understanding disease progression.
As a biomarker, cfRNA is sensitive and functional. The expression of many RNAs, including those from human and microbial sources, is tissue-specific [7]. Changes in specific RNA expression profiles in different tumors can be reflected in plasma. Therefore, biomarkers based on cfRNA not only have many signals in patient plasma at the early stage of cancer [8], but also provide more functional information and can be used as an early diagnostic tool for a variety of diseases, including cancer [9]. Plasma cfRNA is derived from many tissues in the body [10], and its detection can help in indirectly observing pathogeneses in real-time and identifying physiological changes associated with the prephase of subtypes. RNA can more actively enter the environment outside of the cell through cellular efflux mechanisms, such as exosomes [11]. This aspect is particularly significant as it allows for liquid biopsies to serve as a potential means of indirectly observing the pathogenesis in real time and identifying physiological changes associated with the prephase of subtypes. Moreover, effective cfRNA sequencing targeting cancer signals has a lower cost than cfDNA and its methylation sequencing, making it more beneficial to the popularization of early screening and early diagnosis [12]. In addition, compared with cfDNA, cfRNA analysis provides more valuable information on gene expression, splicing, and post-transcriptional regulation. Under specific conditions, such as inflammation, an immune response caused by microbial infection or a tumor, changes in the microenvironment will produce plenty of specific cfRNA molecules [7].
Despite the advancements in high-throughput sequencing instruments, library preparation techniques, and bioinformatics pipelines documented over the last 15 years, and the evidence showing that cfRNA has great potential in cancer diagnosis [13,14], treatment [15], and personalized medicine [16], the implementation of cfRNA diagnostics in clinical environments remains limited. This can be attributed, at least in part, to the absence of a singular dominant technology that effectively, easily, and economically tackles all of the associated obstacles. In this review, we discuss the current knowledge and advancements in the study of plasma cfRNA, focusing on detection methods, its diagnostic and prognostic potential in various diseases, its biological functions, challenges, and future perspectives (Figure 1).

2. Diverse Landscape of Plasma cfRNA

Compared with urine, cerebrospinal fluid (CSF), and other body fluids, blood encompasses a rich composition of both universal and specialized characteristics. Its universal characteristics involve the facilitation of fluid transport within the circulatory system [17], the regulation of body temperature [18,19] and acid–base balance [20], and the transportation of oxygen and carbon dioxide [21,22]. Additionally, blood plays a crucial role in immune responses [23], demonstrating its essential function in maintaining physiological homeostasis. On the other hand, blood’s particularity includes specific cellular components, such as red blood cells, white blood cells, and platelets, each with unique functions. The red blood cells are designated for carrying oxygen, while leukocytes are engaged in immune responses, and platelets contribute significantly to the coagulation process. Another distinctive feature is the presence of plasma, which is the liquid component of blood containing water, electrolytes, and proteins [24,25]. Consequently, it plays a crucial role in nutrient and hormone transportation, as well as in the process of blood coagulation. Additionally, blood has a special coagulation mechanism allowing it to rapidly form blood clots in response to an injury or vascular damage, thanks to the involvement of coagulation factors, platelets, and related components. Lastly, blood is involved in nutrient metabolism by transporting nutrients such as glucose and amino acids, as well as by eliminating metabolites like urea and lactic acid, thereby participating in the overall nutrient metabolism of the body [26,27]. Blood is universal in terms of its circulation, oxygen transport, and immune defense. However, its unique cell composition, clotting mechanism, and nutrient transport function result in particularities that distinguish it from other body fluids. As a result, blood plays a distinctive and irreplaceable role in maintaining overall physiological balance and responding to external changes.
The plasma cfRNA landscape includes a diversity of RNA molecules, detection methods, and clinical roles, and covers various diseases. The diversity of cfRNA is characterized by the presence of various RNA molecules in the bloodstream, encompassing a range of RNA types and functions. Notably, this diversity constitutes a crucial attribute of cfRNA as a potential biomarker. One type of RNA found in plasma is messenger RNA (mRNA), which originates from different cells and holds information about the expression of specific genes. As a result, mRNA in plasma has the potential to serve as a valuable indicator of cellular activity in disease states and therapeutic responses [28]. Another class of RNA found in plasma is miRNA, typically composed of 20–22 nucleotides. miRNA plays a key role in regulating gene expression and can impact the level of gene expression by targeting specific mRNA. Therefore, miRNA present in plasma can be utilized as a potential marker for disease diagnosis and prognosis [29]. Additionally, long non-coding RNAs (lncRNAs), a type of long RNA molecule that does not code for proteins, are involved in the regulation of gene expression and cellular function. Certain lncRNAs detected in plasma are associated with the development and progression of diseases, such as tumors and cardiovascular diseases [30,31]. Another distinctive type of RNA present in plasma is circular RNA (circRNA), a special closed-loop RNA molecule. CircRNA found in plasma is believed to be closely linked to the occurrence and development of various diseases, particularly tumors [32]. Furthermore, several other types of RNA, including piRNA [33], snRNA [34], and snoRNA [35], have been identified in plasma. Although ongoing research is investigating their functionality and effects, these types of RNA may hold significant implications for the diagnosis and treatment of specific diseases. Collectively, the diverse array of cfRNA molecules reflects the intricate regulation of gene expression in cellular activity and disease states. Realizing their potential value in disease diagnosis, prognostic evaluation, and therapeutic monitoring depends on gaining an in-depth understanding of their diversity and functionality. Thus, further research is essential to discern their specific applications and significance in clinical practice.

3. Biological Functions of cfRNA

As showed in Figure 1, cfRNA represents a complex and actively researched area within biology. These RNA molecules exist as fragments of varying lengths and are associated with multiple potential biological functions. Firstly, cell-to-cell communication may occur via cfRNA, which acts as a vector transmitting RNA fragments through the bloodstream to affect the biological processes of distant cells. Extensive research has shown that cfRNA can be transmitted between cells through mechanisms such as urinary bubble RNA-binding proteins, which serve as carriers of information. This intercellular communication serves the purpose of regulating cellular function at locations far from where the cells were produced, such as during damage repair, disease states, and immune responses [36,37]. However, the exact mechanisms and influence of cfRNA in intercellular communication remain subject to ongoing investigation. Therefore, further experiments and studies are necessary to uncover its detailed molecular mechanisms and biological function.
Secondly, plasma cfRNA can directly participate in gene regulation in target cells, with miRNA, for example, inhibiting gene translation or inducing gene degradation by binding to the mRNA of target genes, thereby impacting the functional immune regulation of the target cells [38,39]. The gene regulation function of cfRNA is an actively evolving field of research, characterized by continuous discoveries and insights. Plasma cfRNA molecules serve a critical role in intercellular communication and gene regulation. miRNA, for example, acts through a mechanism wherein it binds to the mRNA of target genes, leading to the inhibition of gene translation or the promotion of mRNA degradation [40]. Exosome bubbles or binding proteins facilitate the release of RNA into the extracellular environment, where they exert a regulatory influence on distant cells. The interactions of RNAs with target genes in plasma play a pivotal role in various biological processes such as cell proliferation [41], apoptosis [42], differentiation [39], metabolism [43], and immune responses [44], thereby significantly impacting disease pathogenesis and progression. On the other hand, long non-coding RNAs (lncRNAs) are characterized as long-length non-coding RNAs that impact gene expression through various regulatory mechanisms, including chromatin modification [45] and transcriptional regulatory protein stability [46]. Similar to other RNAs, lncRNAs can be transferred to other cells in plasma through extracellular vesicles, such as exosomes, to exert a distant regulatory influence. Notably, lncRNA is instrumental in tumor development, the regulation of the cell cycle, and other key cellular processes [46,47]. Additionally, small RNA fragments present in plasma may regulate protein translation by binding to mRNA, thereby influencing gene expression. These small RNA fragments are capable of being transferred between cells, affecting cellular function by regulating gene expression levels [48,49]. Ultimately, the regulatory functions of cfRNA molecules have far-reaching implications in various physiological and pathological processes, underpinning their significance in cellular and molecular biology research.
Furthermore, several studies indicate that cfRNA may influence immune system responses by modulating the activity of immune cells, thereby affecting immune responses to combat infection and control tumors [50,51]. cfRNA has an immunomodulatory function that impacts the regulation of immune cells. miRNA in plasma can be released into the environment through exosomes or bubbles and taken up by other cells, including immune cells [52]. Once internalized, RNAs have the potential to regulate the expression of target genes, affect the development and differentiation of immune cells, and inhibit inflammation. Some RNAs are particularly important in inhibiting the inflammatory response by reducing the inflammatory response and maintaining the immune system balance through the regulation of inflammation-related genes [53,54]. Furthermore, plasma miRNA’s effects on the immune system extend to T-cell regulation, wherein it may influence the antiviral immunity by regulating the function of T-cells, including promoting or inhibiting their proliferation, differentiation, and apoptosis [55,56,57]. Certain RNAs may play a key role in regulating antiviral immunity by influencing the expression level of antiviral genes and immune cells’ responses to viruses, thereby affecting the overall function of the immune system by regulating the expression of immunoregulatory factors, such as interferon chemokines [58,59]. In addition, cfRNA can impact the polarization of immune cells, such as M1/M2 macrophage polarization, thereby affecting the balanced immune tolerance of inflammatory responses and immune regulation [60]. Furthermore, studies have indicated that cfRNA molecules are associated with T-cell tolerance by affecting T-cell development and function, regulating the immune system’s tolerance to autoantigens, and preventing the occurrence of autoimmune diseases [61]. Abnormal expressions of cfRNA are linked to the occurrence and development of autoimmune diseases, such as rheumatoid arthritis [62] and systemic lupus erythematosus [63].
Additionally, certain cfRNA molecules may also be involved in apoptosis and cell survival, which are crucial for tissue homeostasis. The regulation of apoptosis by miRNA is a multifaceted process that encompasses both the promotion and inhibition of cell death. Some RNAs have been identified as proponents of apoptosis, achieving this by either inhibiting anti-apoptosis genes or augmenting the expression of apoptosis-inducing factors [64]. These specific RNAs are present in plasma and may be transported to target cells through exosomes or other carriers, resulting in a direct impact on cell survival. Moreover, the interaction of RNAs with apoptosis-related genes such as Bcl-2 [65] and Caspases [66] serves to modulate the expression levels of these genes, subsequently influencing the susceptibility of cells to apoptosis. Conversely, there are RNAs that exhibit anti-apoptotic properties, exerting their effect by promoting the expression of anti-apoptosis genes or hampering the activity of apoptosis-inducing factors to sustain cell survival. This dichotomy highlights the role of cfRNA in delicately regulating the balance between cell survival and apoptosis [67]. Additionally, cfRNA can influence cell survival states through the modulation of survival signaling pathways, including the PI3K/AKT and MAPK pathways [68,69]. Beyond miRNA, other small RNA fragments in plasma may also partake in the regulation of cell survival and apoptosis by interacting with mRNA or proteins to modulate intracellular signaling pathways. This underscores the intricate nature of miRNA and other small RNA fragments in orchestrating the delicate intertwined processes of cell survival and apoptosis regulation [70,71,72].
Additionally, alterations in cfRNA may serve as potential biomarkers for diseases, notably apoptosis and survival in cancer and other diseases, reflecting biological changes in cells and tissues. Moreover, some studies have shown that they may regulate pathways related to angiogenesis [73], impacting the development and maintenance of the vascular system, as well as playing a regulatory role in neurodevelopmental, neuroprotection, and neurodegenerative diseases [74,75]. Importantly, it is crucial to recognize that the specific function of cfRNA may vary depending on its species, and different types of RNA may play distinct roles in the body. Consequently, ongoing research is imperative to uncover the numerous unknowns and fully understand the biological function of cfRNA, given its complex nature and the need for thorough and continuous investigation.

4. Methods of cfRNA Detection

Various techniques have been developed to detect and analyze cfRNA in plasma. The detection of cfRNA involves several methods, beginning with RNA extraction as the initial step. Due to the low concentration of RNA in plasma, efficient extraction methods such as the phenol/chloroform silica gel column and magnetic bead methods are commonly employed to ensure sufficient RNA yields. Following extraction, reverse transcription (RT) is essential to convert the single-stranded RNA in plasma into complementary DNA (cDNA), a key step in the detection process. Common RT methods include reverse transcription polymerase chain reaction (RT-PCR) and reverse transcriptase chain amplification (RT-qPCR). Subsequently, quantitative PCR, including real-time fluorescent quantitative PCR (qPCR), is employed to amplify and detect specific RNA molecules, enabling the quantification of mRNA and miRNA in plasma. Additionally, the traditional method of Northern Blotting, though less commonly used than PCR techniques, remains valuable in specific instances for separating RNAs via electrophoresis and detecting specific RNAs via membrane transfer and probe hybridization. Next-generation sequencing (NGS) technology provides comprehensive information about different RNA species and their relative abundance in plasma, thereby facilitating the discovery of new RNA markers and understanding of the overall diversity of cfRNA [76]. Furthermore, flow cytometry, a cytological technique, can rapidly and accurately quantify specific cfRNA molecules by combining them with a fluorescently labeled probe [77]. When selecting the detection method, consideration must be given to the sensitivity, specificity, and availability of laboratory technology and equipment. In general, a combination of detection approaches may provide a more holistic understanding of the diversity and abundance of free RNA in plasma.
As showed in Table 1, various methods are used for the detection of cfRNA, each with distinct advantages and limitations. Real-time fluorescence quantitative PCR (RT-qPCR) is a widely utilized method due to its high sensitivity, good quantification, and suitability for the detection of specific RNA sequences [78]. However, it is limited to the detection of known RNA sequences and requires prior knowledge of the target RNA’s sequence information [79,80]. Next-generation sequencing (NGS) provides the ability to comprehensively detect RNA sequencing (RNA-seq) in plasma with high throughput, including known and unknown sequences, making it suitable for the discovery of new RNA markers and offering detailed transcriptomic information [10,81]. Despite these advantages, it comes with relatively high costs and data analysis difficulties, particularly in large-scale epidemiological studies [76,82]. Polyadenylation ligation-mediated sequencing (PALM-Seq), also known as RNA sequencing, is a technique used to study RNA molecule sequences and is often associated with the presence of Polyadenosine (PolyA) in RNA. This technique offers numerous advantages and disadvantages that are important for researchers to consider. PALM-Seq offers several advantages, such as the ability to quantify transcription and discover new transcripts, as well as the ability to detect variable splicing events and study RNA modifications. However, it is important to consider the associated drawbacks, including the high cost and stringent RNA quality and integrity requirements. Furthermore, PALM-Seq does not provide absolute gene expression levels and necessitates the use of complex bioinformatics tools and algorithms for data analysis [83,84]. Digital PCR, specifically droplet digital PCR (ddPCR), is a widely used technology for achieving more accurate detection. It falls under the third-generation PCR category, and it offers four primary application directions, including absolute quantification [85], rare copy detection [86], copy number variation analysis [87], and the determination of gene expression levels [88]. ddPCR provides more accurate quantitative information, particularly for low-copy-number RNA detection, making it suitable for rare events [89,90]. On the other hand, mass spectrometry can be used to detect a variety of RNA modifications and provide detailed RNA structure information [91,92], albeit with the requirement of complex instruments, professional techniques, high costs, and a limited scope of application [93,94]. Flow cytometry can be applied to detect cfRNA, which includes the analysis of RNA expressions of specific cell subsets. This technique can also be combined with appropriate antibodies or fluorescent probes to identify modifications on RNAs, such as m6A (N6-methyadenylate), as well as for investigating intracellular RNA localization and dynamics [95]. Flow cytometry is primarily advantageous in the detection of cell surface markers and proteins, whereas its applications in RNA detection are somewhat limited. When compared to other RNA analysis techniques like RT-qPCR or RNA sequencing, flow cytometry may present some drawbacks, particularly for complex RNA expression analyses [43,96]. Furthermore, the limitations of RNA distribution in tissues highlight the need for suitable methods to study such distributions, as the detection of free RNA in plasma may not be sensitive enough and is more appropriate for tissue sample selection. Ultimately, selecting the appropriate method depends on the specific purpose of the study, the budget, the sample size, and the available laboratory equipment and technology. Combining multiple methods can provide a more comprehensive understanding of free RNA in plasma. We summarized the methods of cfRNA detection in recent research (Table 2).

5. Circulating cfRNA in Disease Diagnosis

Plasma cfRNA has shown great promise as a diagnostic tool in various diseases. In cancer, cfRNA analysis enables the detection of tumor-specific genetic alterations, facilitating early diagnosis and the monitoring of treatment responses [81]. In certain instances, cancer cells release specific RNA or miRNA molecules that can be identified in plasma. This makes cfRNA a potential marker for early cancer diagnosis, even in small tumors without clear symptoms [76]. The analysis of free RNA has emerged as a valuable tool with significant potential in cancer diagnosis, cancer detection, the prediction of tumor origin tissues, and the identification of cancer subtypes [97,98]. This promising analyte offers a unique opportunity to uncover tissue- and subtype-specific biomarkers in breast and lung cancer patients [101]. It is essential to establish a baseline cell-free transcriptome in the absence of cancer, facilitating the identification of tissue specificity and subtype specificity in breast and lung cancer patients [16]. After cancer treatment, monitoring changes in cfRNA can help detect cancer recurrence earlier. Regular testing of plasma samples from patients can lead to the timely treatment of potential recurrences. One study found that plasma miR-4442 levels were associated with colorectal cancer (CRC) recurrence and showed an incremental increase with earlier recurrence dates. Furthermore, miR-4442 demonstrated high sensitivity and specificity as a potential biomarker for early CRC recurrence. Subsequent analyses indicated that the expression of miR-4442 in cancer tissues of patients with metastatic liver cancer from CRC was higher than that in normal liver, CRC, and normal colorectal tissues. Notably, the overexpression of miR-4442 promoted the proliferative, migratory, and invasive activities of CRC cells, while also resulting in decreased levels of RBMS1 and E-cadherin, and increased levels of N-cadherin and Snail1. These findings underscore the clinical utility of plasma miR-4442 as a biomarker for predicting the early recurrence of CRC [104]. In addition, cfRNA has shown to have a potential biomarker role in a variety of cancers. The study of cfRNA has made significant progress in the field of breast cancer, and several miRNA and long-stranded RNA molecules in cfRNA have been found to be abnormally expressed in breast cancer patients [105,106]. These RNAs have the potential for use in the diagnosis of early breast cancer, prediction of patient prognoses, and monitoring of treatment responses [107,108]. Furthermore, changes in the expression levels of specific miRNA molecules in the plasma of lung cancer patients have been linked to the occurrence and development of lung cancer, indicating potential clinical applications for early diagnosis [102,103]. Additionally, some miRNA and mRNA expression levels in plasma exhibit significant variation in patients with gastric cancer [109,110] and liver cancer [111,112], presenting promising prospects for early diagnosis and treatment monitoring of these cancers. Similarly, research focusing on prostate cancer has been seeking potential biomarkers to aid in diagnostic and therapeutic monitoring [113,114], while studies on colorectal cancer have indicated associations between certain miRNA molecules in plasma and the development and progression of this disease [7,115]. Moreover, scientists are actively exploring the potential application of cfRNA as a biomarker in other cancer types, emphasizing the need for further in-depth research and validation in this area. It is important to note that despite promising research results, the application of these markers in clinical practice still requires additional validation and standardization.
In infectious diseases, the presence of pathogen-specific RNAs in plasma cfRNA can aid in the identification of the causative agent [116,117]. Furthermore, cfRNA analysis has potential applications in cardiovascular diseases [47,118,119] and neurodegenerative conditions [120]. The detection and analysis of cfRNA as a potential biomarker in disease diagnosis can provide valuable insights into the development and treatment responses of diseases. Changes in the expression levels of cfRNA are associated with the onset and progression of various diseases, including cancer, cardiovascular diseases, infectious diseases, and neurological diseases. Identifying these changes enables the identification of potential biomarkers for early diagnosis and disease monitoring in diverse conditions. Notably, in cancer diagnosis, cfRNA released by tumor cells can be detected in the blood, making it a non-invasive cancer marker. Studies have explored the potential application of cfRNA in various cancer types, such as breast cancer, lung cancer, and gastrointestinal tumors. Moreover, the analysis of cfRNA in detecting infectious diseases, such as viral infections, has potential clinical applications for early diagnosis and monitoring [99]. Additionally, the alteration of neurosystem-related RNAs in neurological diseases, such as Alzheimer’s [121] and Parkinson’s disease [122,123], provides potential biomarkers for diagnosis and therapeutic response monitoring. It is essential to acknowledge that while cfRNA holds potential in disease diagnosis, further research and validation are needed. Consideration of the sensitivity and specificity of the technology available is crucial to ensure its reliability in clinical practice as this field continues to evolve.

6. Circulating cfRNA in Disease Prognosis

cfRNA plays a crucial role in disease prognosis assessments, serving as a prognostic marker, monitoring treatment responses, and providing insights into disease progression. Specifically, in diseases like cancer, alterations in cfRNA levels can be indicative of disease prognosis. For instance, the types and quantity of tumor-associated free RNA in a patient’s plasma can be linked to the risk of disease progression and recurrence, as well as the overall prognosis [60]. Furthermore, monitoring changes in cfRNA during treatment can offer valuable information regarding a treatment’s effectiveness and a disease’s progression. Treatment-induced fluctuations in specific RNA levels can be utilized to evaluate a patient’s response to treatment and forecast disease progression rates and severity [2]. By scrutinizing cfRNA, medical professionals can gain a deeper understanding of a patient’s pathophysiological state and disease characteristics, and can tailor individualized treatment plans based on RNA marker variations. Despite the potential of cfRNA in disease prognosis, extensive research and clinical validation are imperative to identify the most predictive RNA molecules for specific diseases and ensure the accuracy and reliability of corresponding detection methods. This comprehensive approach will enable the optimization of treatment strategies and enhance the overall treatment effectiveness.
The examination of cfRNA’s role in disease prognosis, particularly miRNA and other RNA molecules influencing disease therapeutic responses and patient prognoses, is currently a focal point of research. Specific levels of cfRNA are associated with the clinical features, survival, and treatment responses of tumors, presenting a crucial role in tumor prognosis. The analysis of miRNA expression in plasma through survival analysis provides valuable information for patient prognosis, with particular RNAs indicating a favorable prognosis and others signaling a worsening prognosis. Tumor-associated RNAs, found in plasma, are crucial for tumor prognosis. Several studies have linked specific miRNA expression levels to tumor clinical features, survival, and responses to treatment [55,74]. cfRNA also substantially impacts the prognosis of cardiovascular diseases. Changes in the expression of certain RNAs in plasma following myocardial infarction are closely linked to a patient’s prognosis and can predict cardiovascular events, enabling the use of specific cfRNA molecules to forecast the risk of cardiovascular events and facilitate the early intervention and treatment of neurological outcomes [47,119]. Furthermore, alterations in miRNA expression in plasma are associated with the prognosis of neurological diseases and patient outcomes, including the development of neurodegenerative diseases. cfRNA molecules are also biomarkers for assessing a patient’s prognosis post stroke and have the potential to serve as biomarkers for predicting the prognoses of infectious diseases, reflecting immune responses and disease progression [74]. Additionally, cfRNA is correlated with the treatment responses and progression of immune system diseases, offering insights into predicting long-term patient outcomes. The role of cfRNA in disease prognosis spans various categories, including tumors, cardiovascular diseases, neurological diseases, infectious diseases, and immune system diseases [23,46]. Consequently, changes in the expression patterns and levels of these RNA molecules hold the potential to serve as biomarkers for enhancing prognostic assessments of diseases, guiding treatment decisions, and supporting personalized medicine. Nevertheless, additional research and validation are required to elucidate their precise mechanisms and specific clinical applications.

7. Circulating cfRNA in Disease Treatment

The potential role of cfRNA in disease treatment encompasses several crucial aspects. Firstly, certain expression patterns of cfRNA bear substantial implications for the prognosis and therapeutic response of specific diseases. This analysis aids in the prediction of disease progression, formulation of treatment plans, and assessment of treatment effectiveness. Moreover, alterations in cfRNA during treatment can function as a biomarker of therapeutic effects, affording valuable insights into therapeutic responses and guiding the adaptation of treatment protocols. Importantly, the potential utility of cfRNA extends beyond diagnosis, serving as a tool to identify new therapeutic targets linked to diseases [100]. Systematic analyses of cfRNA can yield information conducive to the discovery of novel drugs or treatments that enhance the efficacy of disease-specific treatments. Furthermore, the examination of cfRNA contributes to an enhanced understanding of disease biology. By scrutinizing the expression patterns and function of these RNA molecules, researchers can garner insights into the biological mechanisms of diseases, thereby informing the development of more efficacious treatment strategies [15,106].
cfRNA plays a pivotal role in therapeutic monitoring, fulfilling various functions in this process. Firstly, the evaluation of therapeutic responses relies on the expression pattern or level of cfRNA, which may change as treatment progresses. By regularly testing cfRNA, doctors can assess a patient’s response to treatment. In cases where treatment is effective, the RNA associated with a particular disease may decrease or disappear, while its expression may increase if treatment is less effective or if the disease relapses. Consequently, this monitoring aids in tracking treatment effectiveness and disease progression. Moreover, cfRNA analysis facilitates the early prediction of treatment effects. Specifically, it allows doctors to anticipate a treatment’s efficacy in the initial stage, offering insights into whether the patient has positively responded to the treatment. This early knowledge enables adjustments to the treatment plan, thereby enhancing the likelihood of treatment success. Notably, individual variations in patients’ responses to the same treatment due to differing pathophysiological characteristics make it essential to monitor cfRNA [19]. This approach enables doctors to grasp the patient’s condition better and tailor their treatment plan based on individual differences, thereby enhancing treatment targeting and effectiveness. Furthermore, monitoring cfRNA serves as a useful way to detect disease recurrence after treatment completion. Some diseases may experience remission post treatment, and continuous monitoring of RNA changes helps doctors identify signs of disease recurrence promptly. This timely detection enables necessary measures to be taken to mitigate disease resurgence. Lastly, the monitoring of cfRNA minimizes the likelihood of unnecessary treatments being administered for patients unsuitable for or not benefiting from a specific treatment. By adjusting treatment plans based on monitoring results, doctors can better manage patients’ treatment, reducing unnecessary side effects and costs [95].
cfRNA plays an important role in the discovery of drug targets and new therapies, as reflected in several key aspects. Firstly, changes in cfRNA are closely related to the development and progression of diseases, making it possible to identify biomarkers associated with specific disease states through the analysis of free RNA in the plasma. These biomarkers can serve as potential drug targets or indicators of therapeutic responses, facilitating a better understanding of the molecular mechanisms of diseases [124]. Additionally, studying the abnormal expression of free RNA in disease states enables researchers to identify key molecules associated with disease progression that may become new drug targets. This understanding helps in developing more targeted and effective pathophysiological drug mechanisms [41]. Furthermore, analyzing the expression profile of cfRNA in different disease states provides insights into the molecular basis of diseases, revealing their key signaling pathways and biological processes. This understanding supports the design of new therapeutic strategies and individualized treatment plans based on the expression patterns of specific RNAs in a patient’s plasma, aiming to improve treatment targeting, reduce side effects, and increase treatment success rates. Lastly, the analysis of cfRNA can be utilized to assess the efficacy and safety of drugs, enabling the full assessment of drug effectiveness and the timely detection of potential adverse reactions or toxicity by monitoring changes in the RNAs in a patient’s plasma during treatment [15,106].
Despite its potential applications in disease treatment, the application of cfRNA is fraught with challenges related to the stability of standardized RNAs obtained from samples, standardized analytical techniques, and clinical validation. Nonetheless, with the continuous advancement of technology and comprehensive research, the potential role of cfRNA as a biomarker in disease treatment will attract considerable attention. We have summarized the clinical value of cfRNA and methods for its detection according to recent research in Table 2.

8. Challenges and Future Perspectives

Despite the potential clinical applications of cfRNA in disease diagnosis and prognostic assessments, practical challenges hinder its effective implementation. One such challenge is the sourcing and handling of samples. The content of cfRNA is relatively low and is susceptible to interference from external factors. Hence, ensuring the integrity of RNA molecules requires meticulous attention when selecting the conditions for sample collection, processing, and storage. Improper treatment can result in RNA degradation, undermining the reliability of subsequent analyses. Various types of RNases are present in the blood, such as endonucleases and exonucleases. These enzymes can significantly affect the detection of mRNA. RNase enzymes in the blood are responsible for breaking down free RNA molecules, including mRNA. Consequently, if blood samples become contaminated with RNases during collection, processing, or storage, the mRNA contained within them can undergo rapid degradation, leading to compromised experimental accuracy [125]. To mitigate the detrimental effects of RNases on mRNA integrity, a range of protective measures are typically implemented during RNA sample handling and storage. These precautions may include incorporating RNase inhibitors, storing the samples at low temperatures, and minimizing the samples’ exposure to room temperature. Blood samples should be processed promptly following collection, and precautions should be taken to prevent contact with RNase-containing materials during sample handling in order to minimize RNA sample degradation attributable to RNase activity [126,127].
Moreover, the lack of standardized sample processing and analysis methods presents a hurdle, as different laboratories and research teams employ varied techniques. This diversity complicates result comparison and integration, emphasizing the need for standardized experimental procedures. Furthermore, the biological diversity across populations contributes to significant variations in cfRNA expression levels among individuals and over time. Therefore, large-scale clinical studies must account for these differences to establish biomarkers with reliable specificity and sensitivity. In this context, the criticality of ensuring assay specificity and sensitivity in early disease diagnosis and prognostic assessments becomes evident. The accurate detection of small RNA changes is crucial for precise results.
When performing RNA sequencing, the fragmented nature of circulating nucleic acids poses a significant challenge, particularly for shorter miRNA and other small non-coding RNA fragments. This nature of fragmentation may potentially affect the efficacy of certain RNA types, such as mRNA and miRNA, as detected with sequencing technology [55,67]. To address this challenge and enhance RNA detection, various strategies can be implemented. These strategies include optimizing sample processing and extraction methods to minimize the fragmentation of circulating nucleic acids. Additionally, utilizing specific RNA extraction kits or methods can help safeguard the integrity of RNA molecules. Selecting appropriate sequencing technologies and analytical methods is crucial for maximizing the sensitivity and accuracy of circulating nucleic acid detection. Moreover, integrating bioinformatics analyses with existing knowledge of RNA structures and functions can aid in the interpretation of sequencing data, leading to the more precise identification and quantification of circulating nucleic acids [35]. Various methods exist for extracting free RNA from plasma or serum samples, as these samples contain free RNA molecules. One common method is the plasma/serum total RNA extraction method, which is known for its versatility in extracting RNAs of different lengths, including small and long RNA molecules. This method boasts a high extraction efficiency, making it suitable for larger sample sizes. Commercial kits designed for this purpose are readily available in the market, enabling the extraction of RNAs suitable for a range of downstream applications, such as RT-qPCR and RNA sequencing. However, the drawbacks of this method include the relatively high costs of the kits, the complexity of the operation process, the requirement for specialized laboratory equipment, and the need for advanced technology. Another specialized method is the miRNA extraction method, specifically tailored for extracting small RNA molecules, such as miRNA, from plasma or serum samples. This method is particularly advantageous for capturing low-abundance miRNA. Nonetheless, it is characterized by its high cost and limitations on the types and lengths of RNAs it can extract, making it unsuitable for the extraction of long RNA molecules [1]. The magnetic bead separation method is also commonly employed due to its use of magnetic bead binding technology, which facilitates easy operation and fast extraction. This method is suitable for extracting RNA molecules of varying lengths from plasma or serum samples. However, it requires the use of special equipment, such as a magnetic bead separator, leading to increased experimental costs. Additionally, some methods may exhibit limitations in terms of the purity and extraction efficiency of the RNA molecules obtained. For those seeking a rapid extraction process, there is the rapid extraction method, characterized by its simplicity and quick extraction of free RNA. This method is ideal for extracting RNAs from small sample volumes, but it may not achieve extraction efficiencies comparable to those achieved with other methods and comes with a higher cost [128]. In summary, each extraction method has its own set of advantages and disadvantages. Researchers must carefully assess their experimental requirements, sample characteristics, and laboratory capabilities to select the most suitable method for their specific needs.
Moreover, interpreting the biological significance of cfRNA and linking it to specific disease states poses a complex challenge, necessitating independent validation of the discovered biomarkers and clinical validation in large-scale studies. Ethical and regulatory considerations add another layer of complexity, encompassing issues such as privacy and lawful sample use. Furthermore, the cost of detecting cfRNA in practical applications may be a limiting factor, underscoring the need for affordable assays for wider clinical use. Notwithstanding these challenges, ongoing scientific efforts are aiming to address these issues, fueling advancements in the application of cfRNA as a biomarker for disease diagnosis and prognosis assessments. With advancing technology and increased clinical validation, cfRNA is anticipated to emerge as a valuable tool in future clinical practice.
When analyzing circulating free RNA, the choice between using plasma or serum as the sample source significantly impacts research and diagnostic outcomes. Both have distinct characteristics affecting the analysis, composition, and handling. Plasma offers advantages in terms of its high circulating cell-free RNA (cfRNA) integrity due to the reduced RNA enzyme content resulting from the removal of cellular components like platelets during processing [2]. Furthermore, due to cell lysis, platelet activation, and various proteins and factors changing during coagulation, more nucleic acids are released, and the target cfRNA may be degraded, which can lead to more data noise and analytical complexity in serum compared to plasma [7,74]. cfRNA in plasma is more stable as it is isolated in the presence of an anticoagulant, minimizing coagulation. However, processing plasma can be complex, requiring the addition of anticoagulants that may complicate subsequent analysis steps. On the other hand, serum preparation is simpler as it does not necessitate anticoagulants and the serum can be separated via centrifugation post natural coagulation. However, serum poses a greater risk of cfRNA degradation due to the potentially higher RNA enzyme levels, which can impact analysis outcomes [12]. Alternatively, platelets may act as a carrier, carrying a certain amount of cfRNA and further decreasing the cfRNA in platelet-free serum during centrifugation. In conclusion, the choice between plasma and serum for cfRNA analyses hinges on various factors such as the sample handling, research objectives, and anticipated results, requiring careful consideration from investigators. The potential of cfRNA as a biomarker for disease diagnosis and prognosis has a wide-ranging impact on the future of clinical practice. The following are some potential future applications and directions for this promising biomarker. Early diagnosis and screening represent a key area where cfRNA holds great promise. By detecting subtle RNA changes, this biomarker can identify a patient’s risk prior to disease onset or the manifestation of symptoms, thus enabling earlier interventions and treatments. Personalized therapy is another valuable application of cfRNA analysis. By examining a patient’s RNA profile, healthcare professionals can tailor treatment plans, choose personalized and more effective strategies, and enhance treatment success rates. Additionally, cfRNA has the potential to monitor treatment responses in real time, facilitating timely adjustments to treatment plans, improving treatment outcomes, and minimizing treatment-related adverse events. In large-scale epidemiological studies, the analysis of cfRNA can be beneficial for simultaneously detecting and differentiating multiple diseases, given the shared molecular biomarkers across various diseases. The analysis of cfRNA plays a crucial role in advancing precision medicine, which can provide personalized prevention, diagnostic, and treatment options based on an individual’s genetic, physiological, and environmental characteristics [1]. A comprehensive understanding of cfRNA can improve the awareness of disease pathogeneses, disease progression patterns, and individual treatment responses, thereby optimizing the accuracy and effectiveness of healthcare. Specifically, the benefits of cfRNA analysis may include personalized diagnoses and treatment planning, the prediction of therapeutic responses and adverse effects, the promotion of targeted therapies and drug development, and the facilitation of interdisciplinary collaboration and data integration. Personalized diagnoses and treatment planning involve a closer examination of individual cfRNA profiles, allowing a deeper understanding of disease-specific biology [19]. This enables customizable diagnoses and treatment options in order to achieve improved therapeutic efficacy, unlike traditional medical methods that often follow a “one-size-fits-all” strategy, neglecting individual variations in genetic composition, physiological conditions, and environmental influences. Predicting therapeutic responses and adverse effects through changes in cfRNA levels can guide treatment decisions by indicating a patient’s responses to specific therapies, therapeutic effects, and potential adverse effects. This analysis can help in predicting patient responses to treatment modalities, optimizing treatment selection, and minimizing unnecessary treatments and adverse effects. cfRNA analysis supports targeted therapy and drug development by identifying molecular targets for diseases and offering insights for designing targeted therapeutic drugs to enhance the precision and efficacy of treatments. Evaluating drug efficacy and safety through cfRNA analysis accelerates the development and clinical application of new therapies. Furthermore, promoting interdisciplinary collaboration and data integration in cfRNA analysis is essential for realizing precision medicine. Collaboration across interdisciplinary fields such as biology, medicine, and bioinformatics is necessary, involving experts from different domains. Integrating various clinical, genomic, and epigenomic datasets establishes a robust data platform for precision medicine, facilitating personalized diagnostic and therapeutic strategies [129,130,131].
The wide adoption of cfRNA could serve as a valuable tool for these studies, offering insights into disease pathogenesis and influential factors. Continued technological advancements, including the use of high-throughput sequencing and bioinformatics tools, are likely to enhance the sensitivity and accuracy of cfRNA detection, addressing concerns around standardization and repeatability, and advancing its application in clinical practice. As clinical validation and trials progress, the integration of cfRNA with other diagnostic and monitoring methods may offer more comprehensive patient management. Ultimately, cfRNA is poised to be a pivotal tool in disease management, supporting precise and personalized medicine. However, the widespread utilization of cfRNA in clinical practice warrants addressing challenges such as standardizing ethical and regulatory protocols, an initiative that scientists and medical professionals are actively working on to realize its broader application.

9. Conclusions

cfRNA has garnered considerable attention regarding its potential for diagnosing, treating, and assessing the prognoses of diseases, positioning it to serve as a biomarker and regulator that can drive the advancement of precision medicine. Despite its promise, the complexity of cfRNA underscores the need for extensive and ongoing research to unravel its many unknown aspects and attain a comprehensive understanding of its biological function. Simultaneously, the continuous development in this field necessitates considering the sensitivity and specificity of detection technology to ensure its reliability in clinical applications. As clinical validation and trials progress, the combination of cfRNA with other diagnostic and monitoring methods holds promise for bolstering patient management and facilitating a more comprehensive approach to disease management, thereby supporting the progress of precision and personalized medicine. To enable the widespread clinical use of cfRNA, the challenges related to regulatory ethics and protocols must be addressed. Scientists and medical professionals are actively pursuing initiatives aimed at surmounting these challenges and expanding the implementation of cfRNA analysis methods in clinical practice. This review comprehensively discusses the role of cfRNA in various diseases and biological processes to establish a systematic framework of knowledge that can advance the utilization of cfRNA molecules as biomarkers or therapeutic targets by researchers.

Author Contributions

Writing—original draft preparation, P.Z.; editing, L.B.; writing—review, M.H., J.O. and R.W.; supervision, P.C. and X.Z. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Nikanjam, M.; Kato, S.; Kurzrock, R. Liquid biopsy: Current technology and clinical applications. J. Hematol. Oncol. 2022, 15, 131. [Google Scholar] [CrossRef] [PubMed]
  2. Zhou, H.; Zhu, L.; Song, J.; Wang, G.; Li, P.; Li, W.; Luo, P.; Sun, X.; Wu, J.; Liu, Y.; et al. Liquid biopsy at the frontier of detection, prognosis and progression monitoring in colorectal cancer. Mol. Cancer 2022, 21, 86. [Google Scholar] [CrossRef]
  3. Alix-Panabières, C.; Pantel, K. Liquid Biopsy: From Discovery to Clinical Application. Cancer Discov. 2021, 11, 858–873. [Google Scholar] [CrossRef] [PubMed]
  4. Schwarzenbach, H.; Hoon, D.S.; Pantel, K. Cell-free nucleic acids as biomarkers in cancer patients. Nat. Rev. Cancer 2011, 11, 426–437. [Google Scholar] [CrossRef] [PubMed]
  5. Kalluri, R. The biology and function of exosomes in cancer. J. Clin. Investig. 2016, 126, 1208–1215. [Google Scholar] [CrossRef] [PubMed]
  6. Zhou, H.; Xu, W.; Qian, H.; Yin, Q.; Zhu, W.; Yan, Y. Circulating RNA as a novel tumor marker: An in vitro study of the origins and characteristics of extracellular RNA. Cancer Lett. 2008, 259, 50–60. [Google Scholar] [CrossRef] [PubMed]
  7. Kan, C.M.; Pei, X.M.; Yeung, M.H.Y.; Jin, N.; Ng, S.S.M.; Tsang, H.F.; Cho, W.C.S.; Yim, A.K.; Yu, A.C.; Wong, S.C.C. Exploring the Role of Circulating Cell-Free RNA in the Development of Colorectal Cancer. Int. J. Mol. Sci. 2023, 24, 11026. [Google Scholar] [CrossRef] [PubMed]
  8. Speicher, M.R.; Pantel, K. Tumor signatures in the blood. Nat. Biotechnol. 2014, 32, 441–443. [Google Scholar] [CrossRef] [PubMed]
  9. Tzimagiorgis, G.; Michailidou, E.Z.; Kritis, A.; Markopoulos, A.K.; Kouidou, S. Recovering circulating extracellular or cell-free RNA from bodily fluids. Cancer Epidemiol. 2011, 35, 580–589. [Google Scholar] [CrossRef]
  10. Chen, S.; Jin, Y.; Wang, S.; Xing, S.; Wu, Y.; Tao, Y.; Ma, Y.; Zuo, S.; Liu, X.; Hu, Y.; et al. Cancer type classification using plasma cell-free RNAs derived from human and microbes. eLife 2022, 11, e75181. [Google Scholar] [CrossRef]
  11. Koh, W.; Pan, W.; Gawad, C.; Fan, H.C.; Kerchner, G.A.; Wyss-Coray, T.; Blumenfeld, Y.J.; El-Sayed, Y.Y.; Quake, S.R. Noninvasive in vivo monitoring of tissue-specific global gene expression in humans. Proc. Natl. Acad. Sci. USA 2014, 111, 7361–7366. [Google Scholar] [CrossRef]
  12. Mader, S.; Pantel, K. Liquid Biopsy: Current Status and Future Perspectives. Oncol. Res. Treat. 2017, 40, 404–408. [Google Scholar] [CrossRef]
  13. Liang, Y.; Liu, Y.; Li, Q. Lung Cancer Feature Recognition Method Based on Whole-Transcriptome Sequencing of Circulating RNA in Plasma, Involves Constructing and Training Predictive Model According to Top-Level Feature Information, and Using Trained Prediction Model to Identify Features of Non-Small Cell Lung Cancer. Patent CN116312800A, 23 June 2023. [Google Scholar]
  14. Liu, Y.; Liang, Y.; Li, Q.; Li, Q. Comprehensive analysis of circulating cell-free RNAs in blood for diagnosing non-small cell lung cancer. Comput. Struct. Biotechnol. J. 2023, 21, 4238–4251. [Google Scholar] [CrossRef]
  15. Raez, L.; Danenberg, K.; Huang, E.; Usher, J.; Danenberg, P.; Sumarriva, D. P14.04 cfRNA from liquid biopsies is more abundant than cfDNA, informs treatment outcome and is concordant with tissue. J. Thorac. Oncol. 2021, 16, S330. [Google Scholar] [CrossRef]
  16. Larson, M.H.; Pan, W.; Kim, H.J.; Mauntz, R.E.; Stuart, S.M.; Pimentel, M.; Zhou, Y.; Knudsgaard, P.; Demas, V.; Aravanis, A.M.; et al. A comprehensive characterization of the cell-free transcriptome reveals tissue- and subtype-specific biomarkers for cancer detection. Nat. Commun. 2021, 12, 2357. [Google Scholar] [CrossRef]
  17. Sun, H.; Wu, C.; Lu, F.; Tan, X.; Huang, X. Method for Observing and Analyzing Blood Transport, Involves Comparing Color of Skin Grafting Body Ring and Color of Blood Ring in Blood Image, and Outputting Blood Transport Contrast Data to Determine Blood Transport State of Doctor. Patent CN115568838A, 6 January 2023. [Google Scholar]
  18. Ijima, E.; Kodera, S.; Hirata, A.; Hikage, T.; Matsumoto, A.; Ishitake, T.; Masuda, H. Excessive whole-body exposure to 28 GHz quasi-millimeter wave induces thermoregulation accompanied by a change in skin blood flow proportion in rats. Front. Public Health 2023, 11, 1225896. [Google Scholar] [CrossRef] [PubMed]
  19. Shan, C.; Hu, J.; Zou, J.; Zhang, A. Wearable Personal Core Body Temperature Measurement Considering Individual Differences and Dynamic Tissue Blood Perfusion. IEEE J. Biomed. Health Inform. 2022, 26, 2158–2168. [Google Scholar] [CrossRef] [PubMed]
  20. Colombo, R.; Wu, M.A.; Castelli, A.; Fossali, T.; Rech, R.; Ottolina, D.; Cogliati, C.; Catena, E. The effects of severe hemoconcentration on acid -base equilibrium in critically ill patients: The forgotten role of buffers in whole blood. J. Crit. Care 2020, 57, 177–184. [Google Scholar] [CrossRef] [PubMed]
  21. Cavaliere, F.; Bevilacqua, F.; Cesana, B.M.; Calabrese, M.; Arlotta, G.; Antoniucci, M.E.; Corsi, F.; Corrado, M.; De Paulis, S.; Scapigliati, A. Carbon dioxide partial pressure and oxygen saturation in venous blood from the upper body compared with mixed venous blood. Br. J. Anaesth. 2021, 126, E177–E179. [Google Scholar] [CrossRef]
  22. Zhou, Y.; Li, F.; Chen, X.; Tan, J.; Zhou, L.; Zhang, H.; Cai, X.; Huang, G.; Li, Z.; Li, R. Blood Oxygen Heart Rate Detecting System Has Communication Module for Sending Blood Oxygen Data and Heart Rate Data to Server, and Data Processor Electrically Connected with Carbon Dioxide Sensor and Bluetooth Module. Patent CN112353373A, 12 February 2021. [Google Scholar]
  23. Belizaire, R.; Wong, W.J.; Robinette, M.L.; Ebert, B.L. Clonal haematopoiesis and dysregulation of the immune system. Nat. Rev. Immunol. 2023, 23, 595–610. [Google Scholar] [CrossRef]
  24. Samukange, W.T.; Gardarsdottir, H.; Leufkens HG, M.; Mantel-Teeuwisse, A.K. Selection of Blood, Blood Components, and Blood Products as Essential Medicines in 105 Low- and Middle-Income Countries. Transfus. Med. Rev. 2020, 34, 94–100. [Google Scholar] [CrossRef]
  25. Farley, A.; Hendry, C.; McLafferty, E. Blood components. Nurs. Stand. 2012, 27, 35–42. [Google Scholar] [CrossRef]
  26. Reinfeld, B.I.; Madden, M.Z.; Wolf, M.M.; Chytil, A.; Bader, J.E.; Patterson, A.R.; Sugiura, A.; Cohen, A.S.; Ali, A.; Do, B.T.; et al. Cell-programmed nutrient partitioning in the tumour microenvironment. Nature 2021, 593, 282–288. [Google Scholar] [CrossRef] [PubMed]
  27. Nemeth, E.; Ganz, T. Hepcidin and Iron in Health and Disease. Annu. Rev. Med. 2023, 74, 261–277. [Google Scholar] [CrossRef]
  28. Yi, K.; Wang, F. Biomolecular Marker for the Diagnosis of Liver Cancer, Which Is a Plasma Exosome mRNA Including HMMR and B4GALT2. Patent CN116121374A, 15 March 2023. [Google Scholar]
  29. Wang, Z.; Han, J.; Deng, X.; Wei, M.; Liu, S.; Zhang, Y.; Yang, X. Use of Plasma Exosome microRNA Marker and/or Substance for Detecting Plasma Exosome microRNA Marker in Preparation of Colorectal Cancer Liver Metastasis Detection Marker and Product. Patent CN116622847A, 22 August 2023. [Google Scholar]
  30. Gu, X.; Zhang, Y.; Xiong, H. Correlation between plasma lncRNA CASC11 and malignancy in lung adenocarcinoma patients and the prognostic value of lncRNA CASC11. Pers. Med. 2023, 20, 167–173. [Google Scholar] [CrossRef]
  31. Shen, W.; Xie, X.; Liu, M.; Wang, L. Diagnostic Value of lncRNA ROR in Differentiating Ovarian Cancer Patients. Clin. Lab. 2020, 66, 1261–1267. [Google Scholar] [CrossRef] [PubMed]
  32. Zheng, F.; Tan, L.; Zhang, F.; Li, S.; Lai, Z.; Xu, H.; Xiong, Z.; Dai, Y. The circRNA-miRNA-mRNA regulatory network in plasma and peripheral blood mononuclear cells and the potential associations with the pathogenesis of systemic lupus erythematosus. Clin. Rheumatol. 2023, 42, 1885–1896. [Google Scholar] [CrossRef] [PubMed]
  33. Jing, Z.; Xi, Y.; Yin, J.; Shuwen, H. Biological roles of piRNAs in colorectal cancer. Gene 2021, 769, 145063. [Google Scholar] [CrossRef]
  34. Kim, N.; Kang, H.; Jo, A.; Yoo, S.A.; Lee, H.O. Perspectives on single-nucleus RNA sequencing in different cell types and tissues. J. Pathol. Transl. Med. 2023, 57, 52–59. [Google Scholar] [CrossRef]
  35. Xiao, L.; Wang, J.; Ju, S.; Cui, M.; Jing, R. Disorders and roles of tsRNA, snoRNA, snRNA and piRNA in cancer. J. Med. Genet. 2022, 59, 623–631. [Google Scholar] [CrossRef]
  36. Murillo, O.D.; Thistlethwaite, W.; Rozowsky, J.; Subramanian, S.L.; Lucero, R.; Shah, N.; Jackson, A.R.; Srinivasan, S.; Chung, A.; Laurent, C.D.; et al. exRNA Atlas Analysis Reveals Distinct Extracellular RNA Cargo Types and Their Carriers Present across Human Biofluids. Cell 2019, 177, 463–477. [Google Scholar] [CrossRef] [PubMed]
  37. Das, S.; Extracellular RNA Communication Consortium; Ansel, K.M.; Bitzer, M.; Breakefield, X.O.; Charest, A.; Galas, D.J.; Gerstein, M.B.; Gupta, M.; Milosavljevic, A.; et al. The Extracellular RNA Communication Consortium: Establishing Foundational Knowledge and Technologies for Extracellular RNA Research. Cell 2019, 177, 231–242. [Google Scholar] [CrossRef] [PubMed]
  38. Papanota, A.M.; Karousi, P.; Kontos, C.K.; Artemaki, P.I.; Liacos, C.I.; Papadimitriou, M.A.; Bagratuni, T.; Eleutherakis-Papaiakovou, E.; Malandrakis, P.; Ntanasis-Stathopoulos, I.; et al. A Cancer-Related microRNA Signature Shows Biomarker Utility in Multiple Myeloma. Int. J. Mol. Sci. 2021, 22, 13144. [Google Scholar] [CrossRef] [PubMed]
  39. Kassambara, A.; Herviou, L.; Ovejero, S.; Jourdan, M.; Thibaut, C.; Vikova, V.; Pasero, P.; Elemento, O.; Moreaux, J. RNA-sequencing data-driven dissection of human plasma cell differentiation reveals new potential transcription regulators. Leukemia 2021, 35, 1451–1462. [Google Scholar] [CrossRef] [PubMed]
  40. Joshi, G.K.; Deitz-McElyea, S.; Liyanage, T.; Lawrence, K.; Mali, S.; Sardar, R.; Korc, M. Label-Free Nanoplasmonic-Based Short Noncoding RNA Sensing at Attomolar Concentrations Allows for Quantitative and Highly Specific Assay of MicroRNA-10b in Biological Fluids and Circulating Exosomes. ACS Nano 2015, 9, 11075–11089. [Google Scholar] [CrossRef] [PubMed]
  41. Lin, X.; Zhuang, S.; Chen, X.; Du, J.; Zhong, L.; Ding, J.; Wang, L.; Yi, J.; Hu, G.; Tang, G.; et al. lncRNA ITGB8-AS1 functions as a ceRNA to promote colorectal cancer growth and migration through integrin-mediated focal adhesion signaling. Mol. Ther. 2022, 30, 688–702. [Google Scholar] [CrossRef] [PubMed]
  42. Tan, C.; Li, J.; Yuan, Z.; Mu, Y. Circular RNA ciRs-126 promotes hypoxia/reoxygenation cardiac injury possibly through miR-21. Thromb. J. 2022, 20, 2. [Google Scholar] [CrossRef] [PubMed]
  43. Rossi, A.; Pacella, I.; Piconese, S. RNA Flow Cytometry for the Study of T Cell Metabolism. Int. J. Mol. Sci. 2021, 22, 3906. [Google Scholar] [CrossRef] [PubMed]
  44. Zhuang, J.; Ibarra, A.; Acosta, A.; Karns, A.P.; Aballi, J.; Nerenberg, M.; Sninsky, J.J.; Quake, S.R.; Toden, S. Survey of extracellular communication of systemic and organ-specific inflammatory responses through cell free messenger RNA profiling in mice. EBioMedicine 2022, 83, 104242. [Google Scholar] [CrossRef]
  45. Kaur, G.; Singh, K.; Maremanda, K.P.; Li, D.; Chand, H.S.; Rahman, I. Differential plasma exosomal long non-coding RNAs expression profiles and their emerging role in E-cigarette users, cigarette, waterpipe, and dual smokers. PLoS ONE 2020, 15, e0243065. [Google Scholar] [CrossRef]
  46. Liu, J.; Zhou, W.Y.; Luo, X.J.; Chen, Y.X.; Wong, C.W.; Liu, Z.X.; Bo Zheng, J.; Yu Mo, H.; Chen, J.Q.; Li, J.J.; et al. Long noncoding RNA Regulating ImMune Escape regulates mixed lineage leukaemia protein-1-H3K4me3-mediated immune escape in oesophageal squamous cell carcinoma. Clin. Transl. Med. 2023, 13, e1410. [Google Scholar] [CrossRef]
  47. Videira, R.F.; da Costa Martins, P.A.; Falcão-Pires, I. Non-Coding RNAs as Blood-Based Biomarkers in Cardiovascular Disease. Int. J. Mol. Sci. 2020, 21, 9285. [Google Scholar] [CrossRef]
  48. Verbeek, M.W.C.; Erkeland, S.J.; van der Velden, V.H.J. Dysregulation of Small Nucleolar RNAs in B-Cell Malignancies. Biomedicines 2022, 10, 1229. [Google Scholar] [CrossRef]
  49. Li, D.; Xie, X.; Yin, N.; Wu, X.; Yi, B.; Zhang, H.; Zhang, W. tRNA-derived small RNAs: A Novel Regulatory Small Non-coding RNA in renal diseases. Kidney Dis. 2023, 10, 1–11. [Google Scholar] [CrossRef]
  50. Su, Y.; Zhang, X.; Liang, Y.; Sun, J.; Lu, C.; Huang, Z. Integrated analysis of single-cell RNA-seq and bulk RNA-seq to unravel the molecular mechanisms underlying the immune microenvironment in the development of intestinal-type gastric cancer. Biochim. Biophys. Acta-Mol. Basis Dis. 2024, 1870, 166849. [Google Scholar] [CrossRef]
  51. Yang, L.; Han, B.; Zhang, Z.; Wang, S.; Bai, Y.; Zhang, Y.; Tang, Y.; Du, L.; Xu, L.; Wu, F.; et al. Extracellular Vesicle-Mediated Delivery of Circular RNA SCMH1 Promotes Functional Recovery in Rodent and Nonhuman Primate Ischemic Stroke Models. Circulation 2020, 142, 556–574. [Google Scholar] [CrossRef]
  52. Caner, V.; Cetin, G.O.; Hacioglu, S.; Baris, I.C.; Tepeli, E.; Turk, N.S.; Bagci, G.; Yararbas, K.; Cagliyan, G. The miRNA content of circulating exosomes in DLBCL patients and in vitro influence of DLBCL-derived exosomes on miRNA expression of healthy B-cells from peripheral blood. Cancer Biomark. 2021, 32, 519–529. [Google Scholar] [CrossRef]
  53. Li, Y.; Zhou, Y.; Zhao, M.; Zou, J.; Zhu, Y.; Yuan, X.; Liu, Q.; Cai, H.; Chu, C.Q.; Liu, Y. Differential Profile of Plasma Circular RNAs in Type 1 Diabetes Mellitus. Diabetes Metab. J. 2020, 44, 854–865. [Google Scholar] [CrossRef]
  54. Suen, A.O.; Chen, F.; Wang, S.; Li, Z.; Zhu, J.; Yang, Y.; Conn, O.; Lopez, K.; Cui, P.; Wechsler, L.; et al. Extracellular RNA Sensing Mediates Inflammation and Organ Injury in a Murine Model of Polytrauma. J. Immunol. 2023, 210, 1990–2000. [Google Scholar] [CrossRef]
  55. Jiang, Q.; Wang, Q.; Tan, S.; Cai, J.; Ye, X.; Su, G.; Yang, P. Effects of Plasma-Derived Exosomal miRNA-19b-3p on Treg/T Helper 17 Cell Imbalance in Behcet’s Uveitis. Investig. Ophthalmol. Vis. Sci. 2023, 64, 28. [Google Scholar] [CrossRef]
  56. Deng, J.N.; Li, Y.Q.; Liu, Y.; Li, Q.; Hu, Y.; Xu, J.Q.; Sun, T.Y.; Xie, L.X. Exosomes derived from plasma of septic patients inhibit apoptosis of T lymphocytes by down-regulating bad via hsa-miR-7-5p. Biochem. Biophys. Res. Commun. 2019, 513, 958–966. [Google Scholar] [CrossRef]
  57. Wang, Y.; Guo, Y.; Zhang, X.; Zhao, H.; Zhang, B.; Wu, Y.; Zhang, J. The role and mechanism of miR-557 in inhibiting the differentiation and maturation of megakaryocytes in immune thrombocytopenia. RNA Biol. 2021, 18, 1953–1968. [Google Scholar] [CrossRef]
  58. Iacob, D.G.; Rosca, A.; Ruta, S.M. Circulating microRNAs as non-invasive biomarkers for hepatitis B virus liver fibrosis. World J. Gastroenterol. 2020, 26, 1113–1127. [Google Scholar] [CrossRef]
  59. Huang, Y.; Zeng, G.; Randhawa, P.S. Detection of BKV encoded mature MicroRNAs in kidney transplant patients: Clinical and biologic insights. J. Clin. Virol. 2019, 119, 6–10. [Google Scholar] [CrossRef]
  60. Tang, Y.; Hu, S.; Li, T.; Qiu, X. Tumor cells-derived exosomal circVCP promoted the progression of colorectal cancer by regulating macrophage M1/M2 polarization. Gene 2023, 870, 147413. [Google Scholar] [CrossRef]
  61. Wu, X.; Wang, Z.; Wang, J.; Tian, X.; Cao, G.; Gu, Y.; Shao, F.; Yan, T. Exosomes Secreted by Mesenchymal Stem Cells Induce Immune Tolerance to Mouse Kidney Transplantation via Transporting LncRNA DANCR. Inflammation 2022, 45, 460–475. [Google Scholar] [CrossRef]
  62. Cieśla, M.; Kolarz, B.; Majdan, M.; Darmochwał-Kolarz, D. Plasma micro-RNA-22 is associated with disease activity in well-established rheumatoid arthritis. Clin. Exp. Rheumatol. 2020, 40, 945–951. [Google Scholar] [CrossRef]
  63. Luo, Q.; Ye, Y.; Zhang, L.; Gao, Y.; Rao, J.; Guo, Y.; Huang, Q.; Huang, Z.; Li, J. Hsa_circ_0044235 and hsa_circ_0001947 as novel biomarkers in plasma of patients with new-onset systemic lupus erythematosus. J. Immunotoxicol. 2023, 20, 2196453. [Google Scholar] [CrossRef]
  64. Li, J.; Salvador, A.M.; Li, G.; Valkov, N.; Ziegler, O.; Yeri, A.; Yang Xiao, C.; Meechoovet, B.; Alsop, E.; Rodosthenous, R.S.; et al. Mir-30d Regulates Cardiac Remodeling by Intracellular and Paracrine Signaling. Circ. Res. 2021, 128, E1–E23. [Google Scholar] [CrossRef]
  65. Deng, M.; Yuan, H.; Liu, S.; Hu, Z.; Xiao, H. Exosome-transmitted LINC00461 promotes multiple myeloma cell proliferation and suppresses apoptosis by modulating microRNA/BCL-2 expression. Cytotherapy 2019, 21, 96–106. [Google Scholar] [CrossRef]
  66. Chen, A.; Lu, D.; Yang, Z.; Che, X.; Xia, Y.; Shao, X.; Chen, Z.; Qian, J.; Ge, J. Association between NLRP3 inflammasome and periprocedural myocardial injury following elective PCI. Heliyon 2023, 9, e19269. [Google Scholar] [CrossRef]
  67. Li, J.; Wang, Y.; Wu, T.; Li, S.; Sun, Y.N.; Liu, Z.H. Baicalein suppresses high glucose-induced inflammation and apoptosis in trophoblasts by targeting the miRNA-17-5p-Mfn1/2-NF-κB pathway. Placenta 2022, 121, 126–136. [Google Scholar] [CrossRef]
  68. Liang, G.; Wang, Q.; Zhang, G.; Li, Z.; Wang, Q. Differentially expressed miRNAs and potential therapeutic targets for asthenospermia. Andrologia 2022, 54, e14265. [Google Scholar] [CrossRef]
  69. Lu, T.; Wang, Y.; Liu, F.; Zhang, L.; Huang, S.; Zhou, Y.; Wu, H.; Mao, Y.; Jin, C.; Song, W. Synergistic Inhibitory Effect of Berberine and Low-Temperature Plasma on Non-Small-Cell Lung Cancer Cells via PI3K-AKT-Driven Signaling Axis. Molecules 2023, 28, 7797. [Google Scholar] [CrossRef]
  70. Yao, J.; Wu, D.C.; Nottingham, R.M.; Lambowitz, A.M. Identification of protein-protected mRNA fragments and structured excised intron RNAs in human plasma by TGIRT-seq peak calling. eLife 2020, 9, e60743. [Google Scholar] [CrossRef]
  71. Solaguren-Beascoa, M.; Gámez-Valero, A.; Escaramís, G.; Herrero-Lorenzo, M.; Ortiz, A.M.; Minguet, C.; Gonzalo, R.; Bravo, M.I.; Costa, M.; Martí, E. Phospho-RNA-Seq Highlights Specific Small RNA Profiles in Plasma Extracellular Vesicles. Int. J. Mol. Sci. 2023, 24, 11653. [Google Scholar] [CrossRef]
  72. Giraldez, M.D.; Spengler, R.M.; Etheridge, A.; Goicochea, A.J.; Tuck, M.; Choi, S.W.; Galas, D.J.; Tewari, M. Phospho-RNA-seq: A modified small RNA-seq method that reveals circulating mRNA and lncRNA fragments as potential biomarkers in human plasma. EMBO J. 2019, 38, e101695. [Google Scholar] [CrossRef]
  73. Guo, X.; Chang, X.; Wang, Z.; Jiang, C.; Wei, Z. CircRNAs: Promising factors for regulating angiogenesis in colorectal cancer. Clin. Transl. Oncol. 2022, 24, 1673–1681. [Google Scholar] [CrossRef]
  74. Nie, C.; Sun, Y.; Zhen, H.; Guo, M.; Ye, J.; Liu, Z.; Yang, Y.; Zhang, X. Differential Expression of Plasma Exo-miRNA in Neurodegenerative Diseases by Next-Generation Sequencing. Front. Neurosci. 2020, 14, 438. [Google Scholar] [CrossRef]
  75. Szelenberger, R.; Kacprzak, M.; Saluk-Bijak, J.; Zielinska, M.; Bijak, M. Plasma MicroRNA as a novel diagnostic. Clin. Chim. Acta 2019, 499, 98–107. [Google Scholar] [CrossRef]
  76. Metzenmacher, M.; Váraljai, R.; Hegedüs, B.; Cima, I.; Forster, J.; Schramm, A.; Scheffler, B.; Horn, P.A.; Klein, C.A.; Szarvas, T.; et al. Plasma Next Generation Sequencing and Droplet Digital-qPCR-Based Quantification of Circulating Cell-Free RNA for Noninvasive Early Detection of Cancer. Cancers 2020, 12, 353. [Google Scholar] [CrossRef]
  77. Lässer, C.; Alikhani, V.S.; Ekström, K.; Eldh, M.; Paredes, P.T.; Bossios, A.; Sjöstrand, M.; Gabrielsson, S.; Lötvall, J.; Valadi, H. Human saliva, plasma and breast milk exosomes contain RNA: Uptake by macrophages. J. Transl. Med. 2011, 9, 9. [Google Scholar] [CrossRef] [PubMed]
  78. Jin, N.; Kan, C.M.; Pei, X.M.; Cheung, W.L.; Ng, S.S.M.; Wong, H.T.; Cheng, H.Y.; Leung, W.W.; Wong, Y.N.; Tsang, H.F.; et al. Cell-free circulating tumor RNAs in plasma as the potential prognostic biomarkers in colorectal cancer. Front. Oncol. 2023, 13, 1134445. [Google Scholar] [CrossRef]
  79. Weiner, C.P.; Weiss, M.L.; Zhou, H.; Syngelaki, A.; Nicolaides, K.H.; Dong, Y. Detection of Embryonic Trisomy 21 in the First Trimester Using Maternal Plasma Cell-Free RNA. Diagnostics 2022, 12, 1410. [Google Scholar] [CrossRef] [PubMed]
  80. Yang, S.P.; Koh, L.C.W.; Kong, K.W.; Parameswaran, R.; Loke, K.S.H.; Ngiam, K.Y.; Tan, W.B.; Loh, T.; Ng, D.C.E.; Goh, B.C.; et al. A Multiplex Thyroid-Specific Assay for Quantification of Circulating Thyroid Cell-Free RNA in Plasma of Thyroid Cancer Patients. Front. Genet. 2021, 12, 721832. [Google Scholar] [CrossRef]
  81. Roskams-Hieter, B.; Kim, H.J.; Anur, P.; Wagner, J.T.; Callahan, R.; Spiliotopoulos, E.; Kirschbaum, C.W.; Civitci, F.; Spellman, P.T.; Thompson, R.F.; et al. Plasma cell-free RNA profiling distinguishes cancers from pre-malignant conditions in solid and hematologic malignancies. NPJ Precis. Oncol. 2022, 6, 28. [Google Scholar] [CrossRef]
  82. Jin, H.; Zhang, Y.; Fan, Z.; Wang, X.; Rui, C.; Xing, S.; Dong, H.; Wang, Q.; Tao, F.; Zhu, Y. Identification of novel cell-free RNAs in maternal plasma as preterm biomarkers in combination with placental RNA profiles. J. Transl. Med. 2023, 21, 256. [Google Scholar] [CrossRef]
  83. Zhou, S.; Li, J.; Yang, W.; Xue, P.; Yin, Y.; Wang, Y.; Tian, P.; Peng, H.; Jiang, H.; Xu, W.; et al. Noninvasive preeclampsia prediction using plasma cell-free RNA signatures. Am. J. Obstet. Gynecol. 2023, 229, 553.e1–553.e16. [Google Scholar] [CrossRef]
  84. Wang, Y.; Li, J.; Zhang, L.; Sun, H.X.; Zhang, Z.; Xu, J.; Xu, Y.; Lin, Y.; Zhu, A.; Luo, Y.; et al. Plasma cell-free RNA characteristics in COVID-19 patients. Genome Res. 2022, 32, 228–241. [Google Scholar] [CrossRef]
  85. Li, T.; Shao, Y.; Fu, L.; Xie, Y.; Zhu, L.; Sun, W.; Yu, R.; Xiao, B.; Guo, J. Plasma circular RNA profiling of patients with gastric cancer and their droplet digital RT-PCR detection. J. Mol. Med. 2018, 96, 85–96. [Google Scholar] [CrossRef]
  86. Vargas, D.Y.; Marras, S.A.E.; Tyagi, S.; Kramer, F.R. Suppression of Wild-Type Amplification by Selectivity Enhancing Agents in PCR Assays that Utilize SuperSelective Primers for the Detection of Rare Somatic Mutations. J. Mol. Diagn. JMD 2018, 20, 415–427. [Google Scholar] [CrossRef] [PubMed]
  87. Shebanits, K.; Günther, T.; Johansson, A.C.V.; Maqbool, K.; Feuk, L.; Jakobsson, M.; Larhammar, D. Copy number determination of the gene for the human pancreatic polypeptide receptor NPY4R using read depth analysis and droplet digital PCR. BMC Biotechnol. 2019, 19, 31. [Google Scholar] [CrossRef]
  88. Taylor, S.C.; Laperriere, G.; Germain, H. Droplet Digital PCR versus qPCR for gene expression analysis with low abundant targets: From variable nonsense to publication quality data. Sci. Rep. 2017, 7, 2409. [Google Scholar] [CrossRef]
  89. Lak, N.S.M.; Seijger, A.; van Zogchel, L.M.J.; Gelineau, N.U.; Javadi, A.; Zappeij-Kannegieter, L.; Bongiovanni, L.; Andriessen, A.; Stutterheim, J.; van der Schoot, C.E.; et al. Cell-Free RNA from Plasma in Patients with Neuroblastoma: Exploring the Technical and Clinical Potential. Cancers 2023, 15, 2108. [Google Scholar] [CrossRef]
  90. van Zogchel, L.M.J.; Lak, N.S.M.; Verhagen, O.J.H.M.; Tissoudali, A.; Gussmalla Nuru, M.; Gelineau, N.U.; Zappeij-Kannengieter, L.; Javadi, A.; Zijtregtop, E.A.M.; Merks, J.H.M.; et al. Novel Circulating Hypermethylated RASSF1A ddPCR for Liquid Biopsies in Patients with Pediatric Solid Tumors. JCO Precis. Oncol. 2021, 5, 1738–1748. [Google Scholar] [CrossRef]
  91. Lauman, R.; Garcia, B.A. Unraveling the RNA modification code with mass spectrometry. Mol. Omics 2020, 16, 305–315. [Google Scholar] [CrossRef]
  92. You, X.J.; Liu, T.; Ma, C.J.; Qi, C.B.; Tong, Y.; Zhao, X.; Yuan, B.F.; Feng, Y.Q. Determination of RNA Hydroxylmethylation in Mammals by Mass Spectrometry Analysis. Anal. Chem. 2019, 91, 10477–10483. [Google Scholar] [CrossRef]
  93. Ammann, G.; Berg, M.; Dalwigk, J.F.; Kaiser, S.M. Pitfalls in RNA Modification Quantification Using Nucleoside Mass Spectrometry. Acc. Chem. Res. 2023, 56, 3121–3131. [Google Scholar] [CrossRef] [PubMed]
  94. Ray, J.; Kruse, A.; Ozer, A.; Kajitani, T.; Johnson, R.; MacCoss, M.; Heck, M.; Lis, J.T. RNA aptamer capture of macromolecular complexes for mass spectrometry analysis. Nucleic Acids Res. 2020, 48, e90. [Google Scholar] [CrossRef] [PubMed]
  95. Yoo, H.B.; Park, S.R.; Hong, K.S.; Yang, I. Precise RNA Quantification by Counting Individual RNA Molecules Using High-Sensitivity Capillary Flow Cytometry. Anal. Chem. 2022, 94, 1752–1759. [Google Scholar] [CrossRef]
  96. Duckworth, A.D.; Gherardini, P.F.; Sykorova, M.; Yasin, F.; Nolan, G.P.; Slupsky, J.R.; Kalakonda, N. Multiplexed profiling of RNA and protein expression signatures in individual cells using flow or mass cytometry. Nat. Protoc. 2019, 14, 901–920. [Google Scholar] [CrossRef] [PubMed]
  97. Ita, M.I.; Wang, J.H.; Toulouse, A.; Lim, C.; Fanning, N.; O’Sullivan, M.; Nolan, Y.; Kaar, G.F.; Redmond, H.P. The utility of plasma circulating cell-free messenger RNA as a biomarker of glioma: A pilot study. Acta Neurochir. 2022, 164, 723–735. [Google Scholar] [CrossRef] [PubMed]
  98. Moufarrej, M.N.; Vorperian, S.K.; Wong, R.J.; Campos, A.A.; Quaintance, C.C.; Sit, R.V.; Tan, M.; Detweiler, A.M.; Mekonen, H.; Neff, N.F.; et al. Early prediction of preeclampsia in pregnancy with cell-free RNA. Nature 2022, 602, 689–694. [Google Scholar] [CrossRef] [PubMed]
  99. Pan, W.; Ngo, T.T.M.; Camunas-Soler, J.; Song, C.X.; Kowarsky, M.; Blumenfeld, Y.J.; Wong, R.J.; Shaw, G.M.; Stevenson, D.K.; Quake, S.R. Simultaneously Monitoring Immune Response and Microbial Infections during Pregnancy through Plasma cfRNA Sequencing. Clin. Chem. 2017, 63, 1695–1704. [Google Scholar] [CrossRef] [PubMed]
  100. Ita, M.I.; Wang, J.H.; Fanning, N.; Kaar, G.; Lim, C.; Redmond, H.P. Plasma circulating cell free messenger RNA as a potential biomarker of melanoma. Acta Oncol. 2021, 60, 1201–1209. [Google Scholar] [CrossRef] [PubMed]
  101. Hasegawa, N.; Kohsaka, S.; Kurokawa, K.; Shinno, Y.; Takeda Nakamura, I.; Ueno, T.; Kojima, S.; Kawazu, M.; Suehara, Y.; Ishijima, M.; et al. Highly sensitive fusion detection using plasma cell-free RNA in non-small-cell lung cancers. Cancer Sci. 2021, 112, 4393–4403. [Google Scholar] [CrossRef] [PubMed]
  102. Seneviratne, C.; Shetty, A.C.; Geng, X.; McCracken, C.; Cornell, J.; Mullins, K.; Jiang, F.; Stass, S. A Pilot Analysis of Circulating cfRNA Transcripts for the Detection of Lung Cancer. Diagnostics 2022, 12, 2897. [Google Scholar] [CrossRef]
  103. Mullins, K.E.; Seneviratne, C.; Shetty, A.C.; Jiang, F.; Christenson, R.; Stass, S. Proof of concept: Detection of cell free RNA from EDTA plasma in patients with lung cancer and non-cancer patients. Clin. Biochem. 2023, 118, 110583. [Google Scholar] [CrossRef]
  104. Shibamoto, J.; Arita, T.; Konishi, H.; Kataoka, S.; Furuke, H.; Takaki, W.; Kiuchi, J.; Shimizu, H.; Yamamoto, Y.; Komatsu, S.; et al. Roles of miR-4442 in Colorectal Cancer: Predicting Early Recurrence and Regulating Epithelial-Mesenchymal Transition. Genes 2023, 14, 1414. [Google Scholar] [CrossRef]
  105. Cheng, X.; Murthy, S.R.K.; Zhuang, T.; Ly, L.; Jones, O.; Basadonna, G.; Keidar, M.; Kanaan, Y.; Canady, J. Canady Helios Cold Plasma Induces Breast Cancer Cell Death by Oxidation of Histone mRNA. Int. J. Mol. Sci. 2021, 22, 9578. [Google Scholar] [CrossRef]
  106. Tian, Y.; Zhang, Z.; Zhang, Z.; Dai, X. Hsa_circRNA_0040462: A sensor of cells’ response to CAP treatment with double-edged roles on breast cancer malignancy. Int. J. Med. Sci. 2022, 19, 640–650. [Google Scholar] [CrossRef]
  107. Wang, M.; Liu, H.; Wu, W.; Zhao, J.; Song, G.; Chen, X.; Wang, R.; Shao, C.; Li, J.; Wang, H.; et al. Identification of Differentially Expressed Plasma lncRNAs As Potential Biomarkers for Breast Cancer. Clin. Breast Cancer 2022, 22, E135–E141. [Google Scholar] [CrossRef]
  108. Lin, L.; Cai, G.X.; Zhai, X.M.; Yang, X.X.; Li, M.; Li, K.; Zhou, C.L.; Liu, T.C.; Han, B.W.; Liu, Z.J.; et al. Plasma-Derived Extracellular Vesicles Circular RNAs Serve as Biomarkers for Breast Cancer Diagnosis. Front. Oncol. 2021, 11, 752651. [Google Scholar] [CrossRef]
  109. Han, L.; Zhang, X.; Wang, A.; Ji, Y.; Cao, X.; Qin, Q.; Yu, T.; Huang, H.; Yin, L. A Dual-Circular RNA Signature as a Non-invasive Diagnostic Biomarker for Gastric Cancer. Front. Oncol. 2020, 10, 184. [Google Scholar] [CrossRef]
  110. Yu, X.; Song, X.; Xie, Y.; Zhang, S.; Guo, J. Establishment of an Absolute Quantitative Method to Detect a Plasma tRNA-Derived Fragment and Its Application in the Non-Invasive Diagnosis of Gastric Cancer. Int. J. Mol. Sci. 2023, 24, 322. [Google Scholar] [CrossRef] [PubMed]
  111. Rankovic, B.; Hauptman, N. Circulating microRNA Panels for Detection of Liver Cancers and Liver-Metastasizing Primary Cancers. Int. J. Mol. Sci. 2023, 24, 15451. [Google Scholar] [CrossRef] [PubMed]
  112. Zhu, Y.; Wang, S.; Xi, X.; Zhang, M.; Liu, X.; Tang, W.; Cai, P.; Xing, S.; Bao, P.; Jin, Y.; et al. Integrative analysis of long extracellular RNAs reveals a detection panel of noncoding RNAs for liver cancer. Theranostics 2021, 11, 181–193. [Google Scholar] [CrossRef] [PubMed]
  113. Martens-Uzunova, E.S.; Kusuma, G.D.; Crucitta, S.; Lim, H.K.; Cooper, C.; Riches, J.E.; Azad, A.; Ochiya, T.; Boyle, G.M.; Southey, M.C.; et al. Androgens alter the heterogeneity of small extracellular vesicles and the small RNA cargo in prostate cancer. J. Extracell. Vesicles 2021, 10, e12136. [Google Scholar] [CrossRef] [PubMed]
  114. Spieler, B.; Khodor, Y.; Chakrabortty, S.; Fischer, C.; Tadigotla, V.; Stoyanova, R.; Abramowtiz, M.; Punnen, S.; Pollack, A.; Yu, S.; et al. Plasma Exosomal RNA Biomarkers of High-Risk Prostate Cancer. Int. J. Radiat. Oncol. Biol. Phys. 2021, 111, S14–S15. [Google Scholar] [CrossRef]
  115. Zheng, R.; Zhang, K.; Tan, S.; Gao, F.; Zhang, Y.; Xu, W.; Wang, H.; Gu, D.; Zhu, L.; Li, S.; et al. Exosomal circLPAR1 functions in colorectal cancer diagnosis and tumorigenesis through suppressing BRD4 via METTL3-eIF3h interaction. Mol. Cancer 2022, 21, 49. [Google Scholar] [CrossRef]
  116. Kalantar, K.L.; Neyton, L.; Abdelghany, M.; Mick, E.; Jauregui, A.; Caldera, S.; Serpa, P.H.; Ghale, R.; Albright, J.; Sarma, A.; et al. Integrated host-microbe plasma metagenomics for sepsis diagnosis in a prospective cohort of critically ill adults. Nat. Microbiol. 2022, 7, 1805–1816. [Google Scholar] [CrossRef]
  117. Jerome, H.; Taylor, C.; Sreenu, V.B.; Klymenko, T.; Filipe, A.D.S.; Jackson, C.; Davis, C.; Ashraf, S.; Wilson-Davies, E.; Jesudason, N.; et al. Metagenomic next-generation sequencing aids the diagnosis of viral infections in febrile returning travellers. J. Infect. 2019, 79, 383–388. [Google Scholar] [CrossRef]
  118. Ward, Z.; Schmeier, S.; Pearson, J.; Cameron, V.A.; Frampton, C.M.; Troughton, R.W.; Doughty, R.N.; Richards, A.M.; Pilbrow, A.P. Identifying Candidate Circulating RNA Markers for Coronary Artery Disease by Deep RNA-Sequencing in Human Plasma. Cells 2022, 11, 3191. [Google Scholar] [CrossRef]
  119. Vanhaverbeke, M.; Attard, R.; Bartekova, M.; Ben-Aicha, S.; Brandenburger, T.; de Gonzalo-Calvo, D.; Emanueli, C.; Farrugia, R.; Grillari, J.; Hackl, M.; et al. Peripheral blood RNA biomarkers for cardiovascular disease from bench to bedside: A position paper from the EU-CardioRNA COST action CA17129. Cardiovasc. Res. 2022, 118, 3183–3197. [Google Scholar] [CrossRef]
  120. Sosanya, N.M.; Kumar, R.; Clifford, J.L.; Chavez, R.; Dimitrov, G.; Srinivasan, S.; Gautam, A.; Trevino, A.V.; Williams, M.; Hammamieh, R.; et al. Identifying Plasma Derived Extracellular Vesicle (EV) Contained Biomarkers in the Development of Chronic Neuropathic Pain. J. Pain 2020, 21, 82–96. [Google Scholar] [CrossRef]
  121. Wang, T.; Chen, K.; Li, H.; Dong, S.; Su, N.; Liu, Y.; Cheng, Y.; Dai, J.; Yang, C.; Xiao, S. The Feasibility of Utilizing Plasma MiRNA107 and BACE1 Messenger RNA Gene Expression for Clinical Diagnosis of Amnestic Mild Cognitive Impairment. J. Clin. Psychiatry 2015, 76, 135–141. [Google Scholar] [CrossRef]
  122. Rezaei, O.; Nateghinia, S.; Estiar, M.A.; Taheri, M.; Ghafouri-Fard, S. Assessment of the role of non-coding RNAs in the pathophysiology of Parkinson’s disease. Eur. J. Pharmacol. 2021, 896, 173914. [Google Scholar] [CrossRef]
  123. Shi, Q.; Kang, W.; Liu, Z.; Zhu, X. The role of exosomes in the diagnosis of Parkinson’s disease. Heliyon 2023, 9, e20595. [Google Scholar] [CrossRef]
  124. Xu, Y.X.; Pu, S.D.; Li, X.; Yu, Z.W.; Zhang, Y.T.; Tong, X.W.; Shan, Y.Y.; Gao, X.Y. Exosomal ncRNAs: Novel therapeutic target and biomarker for diabetic complications. Pharmacol. Res. 2022, 178, 106135. [Google Scholar] [CrossRef]
  125. Lee, H.H.; Wang, Y.N.; Hung, M.C. Functional roles of the human ribonuclease A superfamily in RNA metabolism and membrane receptor biology. Mol. Asp. Med. 2019, 70, 106–116. [Google Scholar] [CrossRef] [PubMed]
  126. Borgelt, L.; Wu, P. Targeting Ribonucleases with Small Molecules and Bifunctional Molecules. ACS Chem. Biol. 2023, 18, 2101–2113. [Google Scholar] [CrossRef] [PubMed]
  127. Mino, T.; Takeuchi, O. Regnase-1-related endoribonucleases in health and immunological diseases. Immunol. Rev. 2021, 304, 97–110. [Google Scholar] [CrossRef]
  128. Sun, Y.; Yu, H.; Han, S.; Ran, R.; Yang, Y.; Tang, Y.; Wang, Y.; Zhang, W.; Tang, H.; Fu, B.; et al. Method for the extraction of circulating nucleic acids based on MOF reveals cell-free RNA signatures in liver cancer. Natl. Sci. Rev. 2024, 11, nwae022. [Google Scholar] [CrossRef]
  129. Freitas, A.J.A.; Causin, R.L.; Varuzza, M.B.; Calfa, S.; Hidalgo Filho, C.M.T.; Komoto, T.T.; Souza, C.P.; Marques, M.M.C. Liquid Biopsy as a Tool for the Diagnosis, Treatment, and Monitoring of Breast Cancer. Int. J. Mol. Sci. 2022, 23, 9952. [Google Scholar] [CrossRef]
  130. Deleu, J.; Schoofs, K.; Decock, A.; Verniers, K.; Roelandt, S.; Denolf, A.; Verreth, J.; De Wilde, B.; Van Maerken, T.; De Preter, K.; et al. Digital PCR-based evaluation of nucleic acid extraction kit performance for the co-purification of cell-free DNA and RNA. Hum. Genom. 2022, 16, 73. [Google Scholar] [CrossRef] [PubMed]
  131. Loy, C.; Ahmann, L.; De Vlaminck, I.; Gu, W. Liquid Biopsy Based on Cell-Free DNA and RNA. Annu. Rev. Biomed. Eng. 2024. [Google Scholar] [CrossRef]
Figure 1. An overview of cfRNA in plasma for medicine. cfRNA in plasma includes various types of RNA such as miRNA, mRNA, lncRNA, and circRNA. Furthermore, various techniques like RT-qPCR have been developed to detect and analyze cfRNA in plasma, as we show in the first part of this paper. Also, cfRNA represents a complex and actively researched area within biology. These RNA molecules exist as fragments of varying lengths and are associated with multiple potential biological functions. Due to its abundant biological functions, this group of cfRNA could be applied in various diseases to diagnose patients, predict their progression, and assess their prognosis.
Figure 1. An overview of cfRNA in plasma for medicine. cfRNA in plasma includes various types of RNA such as miRNA, mRNA, lncRNA, and circRNA. Furthermore, various techniques like RT-qPCR have been developed to detect and analyze cfRNA in plasma, as we show in the first part of this paper. Also, cfRNA represents a complex and actively researched area within biology. These RNA molecules exist as fragments of varying lengths and are associated with multiple potential biological functions. Due to its abundant biological functions, this group of cfRNA could be applied in various diseases to diagnose patients, predict their progression, and assess their prognosis.
Diagnostics 14 01045 g001
Table 1. The advantages and limitations of different methods for detecting cfRNA in plasma.
Table 1. The advantages and limitations of different methods for detecting cfRNA in plasma.
Detection MethodAdvantagesLimitations
RT-qPCRReal-time fluorescence Quantitative PCRHigh sensitivity
Good quantification
Detects specific RNA sequences
Limited by unknown RNA sequences
NGSNext-generation sequencingHigh throughput
Includes known and unknown sequences
High costs
Data analysis difficulties
PALM-SeqPolyadenylation ligation-mediated sequencingQuantifies transcription
Discovers new transcripts
Variable splicing events
RNA modifications
High costs
Stringent RNA quality
Integrity requirements
Less gene-expression levels
ddPCRDroplet digital PCRAbsolute quantification
Detects a variety of RNA modifications
Provides detailed RNA structure information
Complex instruments
Professional techniques
High costs
Limited scope of application
Flow cytometry Detects cell surface markers and proteinsLess suitable for complex RNA expression analyses
Table 2. Summary of detection methods for cfRNA in recent research.
Table 2. Summary of detection methods for cfRNA in recent research.
Detection MethodBiomarker TypeDiseaseValueReference
qRT-PCRmRNAHCC/MM and related pre-malignant diseasesIdentification[81]
mRNAColorectal cancerMonitoring progression[78]
mRNAPreterm birth (PTB)Predication[82]
mRNAThyroid cancerQuantification of thyroid volume
Recurrence predication
[80]
mRNAGliomaIdentification[97]
cfRNAPreeclampsia Predication[98]
mRNAPregnancy infectionsMonitoring immune response and
microbial infections during pregnancy
[99]
mRNAMelanomaIdentification of novel therapeutic targets or biomarkers[100]
qRT-PCR and
PCF RNA testing
mRNA
miRNA
Embryonic trisomy 21 in the first trimesterDiagnosis[79]
NGSmRNA
rRNA
Non-small-cell lung cancersMonitoring progression
Disease prognosis prediction
[101]
Total cfRNALung cancerEarly diagnosis[102]
cfRNALung cancerDiagnosis[103]
NGS
RT-ddPCR
Solid cancersEarly diagnosis[76]
PALM-SeqmiRNA
tRNA
mRNA
COVID-19Prediction[84]
mRNA
miRNA
PreeclampsiaPredication[83]
ddPCRmRNANeuroblastomaDiagnosis[89]
mRNA
ctDNA
Pediatric solid tumorsPotential biomarker identification[90]
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

Zhong, P.; Bai, L.; Hong, M.; Ouyang, J.; Wang, R.; Zhang, X.; Chen, P. A Comprehensive Review on Circulating cfRNA in Plasma: Implications for Disease Diagnosis and Beyond. Diagnostics 2024, 14, 1045. https://doi.org/10.3390/diagnostics14101045

AMA Style

Zhong P, Bai L, Hong M, Ouyang J, Wang R, Zhang X, Chen P. A Comprehensive Review on Circulating cfRNA in Plasma: Implications for Disease Diagnosis and Beyond. Diagnostics. 2024; 14(10):1045. https://doi.org/10.3390/diagnostics14101045

Chicago/Turabian Style

Zhong, Pengqiang, Lu Bai, Mengzhi Hong, Juan Ouyang, Ruizhi Wang, Xiaoli Zhang, and Peisong Chen. 2024. "A Comprehensive Review on Circulating cfRNA in Plasma: Implications for Disease Diagnosis and Beyond" Diagnostics 14, no. 10: 1045. https://doi.org/10.3390/diagnostics14101045

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