Next Article in Journal
LncRNA-Dependent Mechanisms of Transforming Growth Factor-β: From Tissue Fibrosis to Cancer Progression
Next Article in Special Issue
The Role of miR-217-5p in the Puromycin Aminonucleoside-Induced Morphological Change of Podocytes
Previous Article in Journal
An Expanded Landscape of Unusually Short RNAs in 11 Samples from Six Eukaryotic Organisms
Previous Article in Special Issue
Modulation of MicroRNA Processing by Dicer via Its Associated dsRNA Binding Proteins
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Perspective

Non-Coding RNAs in Rheumatoid Arthritis: Implications for Biomarker Discovery

1
Unidad de Investigación Biomédica de Zacatecas, Instituto Mexicano del Seguro Social, Zacatecas 98000, Mexico
2
Cátedras CONACYT, Consejo Nacional de Ciencia y Tecnología, Cd. México 03940, Mexico
3
Departamento de Inmunología, Facultad de Medicina, Universidad Autónoma de Nuevo León, Monterrey 64460, Mexico
4
Instituto de Ciencias Biologicas, Universidad Autónoma de Guadalajara, Zapopan 45129, Mexico
*
Authors to whom correspondence should be addressed.
Non-Coding RNA 2022, 8(3), 35; https://doi.org/10.3390/ncrna8030035
Submission received: 25 February 2022 / Revised: 11 May 2022 / Accepted: 12 May 2022 / Published: 25 May 2022
(This article belongs to the Special Issue Current Trends in MicroRNA Research: From Basics to Applications)

Abstract

:
Recent advances in gene expression analysis techniques and increased access to technologies such as microarrays, qPCR arrays, and next-generation sequencing, in the last decade, have led to increased awareness of the complexity of the inflammatory responses that lead to pathology. This finding is also the case for rheumatic diseases, importantly and specifically, rheumatoid arthritis (RA). The coincidence in major genetic and epigenetic regulatory events leading to RA’s inflammatory state is now well-recognized. Research groups have characterized the gene expression profile of early RA patients and identified a group of miRNAs that is particularly abundant in the early stages of the disease and miRNAs associated with treatment responses. In this perspective, we summarize the current state of RNA-based biomarker discovery and the context of technology adoption/implementation due to the COVID-19 pandemic. These advances have great potential for clinical application and could provide preclinical disease detection, follow-up, treatment targets, and biomarkers for treatment response monitoring.

1. Novel Gene Expression Analysis Technologies for Biomarkers Discovery

Technologies that analyze gene expression, from qPCR machines to single-cell sequencing for single-cell transcriptomics, have been restricted mostly to research facilities [1]. The translation of these tools to clinical settings has been slow, perhaps due to the few tests being developed that have clinical value (e.g., detecting RNA or DNA from viruses such as HIV and HCV).
Recently, these tools have become available in many centers worldwide due to the COVID-19 pandemic, which required these centers to provide dynamic protocols for RNA detection and/or sequencing [2]. These resources could be used to further significant progress in areas such as inflammatory rheumatic diseases [3]. In the few pages ahead, we highlight the possible application of these technologies in the rheumatology setting using potential RNA-based biomarkers with special emphasis on RA.

2. Gene Expression Profiling of Rheumatic Diseases: Focus on RA

Rheumatoid arthritis is a complex disease involving several metabolic pathways, gene polymorphisms, and epigenetic alterations [4]. In this regard, identifying its gene expression profiles is a meaningful strategy to better understand the mechanisms of pathogenicity, disease course and progression, assess therapeutic response, and even treatment selection. Transcriptional profiling is a powerful technology capable of providing an integrative view of pathophysiological pathways. It evaluates thousands of molecules in one experiment [5]. Microarrays (MA) based on high-throughput platforms are among the most important tools for discovering new genetic, transcriptomic, or epigenetic disease alterations and identifying key future biomarkers for diagnosis, prognosis and response to therapy [6,7]. Microarray technology has been applied to compare RA patients’ gene expression profiles in a specific disease stage to healthy controls, other diseases, or another disease status of the same disease [8]. Molecular patterns identified by microarrays can be so specific that they differentiate particularities among patients with the same disease.
Transcriptomic assays supply vast information, which must be simplified to provide some context in systems biology. Many studies in the field use multiple strategies to simplify the metadata. One strategy to simplify gene expression data involves assessing changes in molecular patterns in a specific cell population. For example, dendritic cells (DCs) are important immune cells associated with the genesis of some autoimmune diseases. There is a specific transcriptional signature in DCs identified in three different autoimmune diseases, namely RA, systemic lupus erythematosus, and type 1 diabetes [9,10]. This transcriptomic profile in DCs is associated with two essential characteristics, the heterogeneity of the autoimmune disease and the disease activity level. These immune gene expression profiles provide essential clues to molecular pathways that can serve as determinants for therapy prognosis and disease course. These gene expression profiles can help to identify new molecular targets and treatment development.
We previously reported a specific transcriptional signature in peripheral blood cells associated with first-degree relatives negative to anti-cyclic citrullinated peptide antibodies (ACPA) that differentiate relatives positive to ACPA, and a specific transcriptional signature for treatment-naïve newly diagnosed RA patients [4]. Identifying these gene expression profiles brings new molecular pathways associated with disease course and diagnostic/prognostic candidate biomarkers for RA. One example is identifying the peripheral blood cell biomarkers SUMO1 (Small ubiquitin-like modifier 1) and POU2AF1 (POU domain class 2-associating Factor 1), which have decreased expression in patients with RA compared to first-degree relatives and negatively correlate with disease progression and ACPA levels [11]. Relatives with mild to low levels of these biomarkers can help identify patients with recent-onset RA in the “window of opportunity”, when treatment could have a beneficial effect on disease progression, even leading more effectively to clinical remission of RA [12].
Several studies have already focused on transcriptomics in RA, and have indeed provided plentiful data, some reporting important cell signatures associated with treatment response [6], the participation of T cells in early RA, ACPA [13], molecular differences between first-degree relatives with or without ACPA [4], and many others with substantial information in RA (Table 1). However, it is not currently practical to evaluate the gene expression profile for every patient with RA for diagnosis or to determine treatment failure or success. However, molecular markers obtained by different transcriptomic studies can help identify successful diagnostic biomarkers in clinical settings.

3. The Identification of mRNA, LncRNAs, and miRNAs as Biomarkers in RA

As mentioned earlier, technological advances in biomedicine, functional genomics, and systems biology have resulted in the identification of a growing number of biomarkers with potential use in clinical practice. They can help identify patients with early RA, identify treatment failure, differentiate other diseases, and evaluate disease course. Some of these biomarkers come from omics assays, which evaluate thousands of molecules, working with metadata and summarizing information to provide specific differentially expressed molecules as new biomarkers for RA (Table 2). These still require extensive validation in a real-world clinical setting to demonstrate their efficacy.

4. Implications and Perspective of Clinical Applicability in RA

Autoimmune diseases are heterogeneous, and RA is pleomorphic in its clinical presentation, disease course, and response to treatment. In this context, identifying RNA biomarkers among patients in different disease stages provides essential information about pathophysiological pathways, functional genetics, and molecular markers for diagnosis, prognosis, and therapy individualization. Due to the advent of RNA-based detection of SARS-CoV-2, the necessary infrastructure needed to detect these RNA species by RT-qPCR is now a reality in most places [21,22].
Ethical, social, and legal implications also need to be considered for the broad adoption of these technologies, such as (1) the anonymity of genetic testing results, (2) data privacy, and (3) the adoption of strict policies and rules regarding information gathering, storage, and access by authorized labs and the biotech industry. These aspects have been widely reviewed in detail elsewhere in order to balance clinical testing usefulness and the prediction of future clinically important conditions against the right to privacy of a patient in the ELSI declaration [23,24]. There has been great advancement in this field given that there are institutions in Mexico, such as the National Institute for Access to Information (INAI, in Spanish), responsible for issuing policies, requirements, storage conditions, and access to personal data that companies, or public entities gather from citizens (genetic data included) and access to non-sensitive government information. In the future, these institutional advances will allow for fast, reliable, and state-of-the-art genetic testing with adequate management of the generated data for the benefit of the patient, investigation, and research.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Acknowledgments

We thank Sergio Lozano-Rodríguez from the Universidad Autónoma de Nuevo León for reviewing and editing the English version of the manuscript.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Koboldt, D.C.; Steinberg, K.M.; Larson, D.E.; Wilson, R.K.; Mardis, E.R. The next-generation sequencing revolution and its impact on genomics. Cell 2013, 155, 27–38. [Google Scholar] [CrossRef] [Green Version]
  2. Gharizadeh, B.; Yue, J.; Yu, M.; Liu, Y.; Zhou, M.; Lu, D.; Zhang, J. Navigating the pandemic response life cycle: Molecular diagnostics and immunoassays in the context of COVID-19 management. IEEE Rev. Biomed. Eng. 2020, 14, 30–47. [Google Scholar] [CrossRef]
  3. Sachinidis, A.; Garyfallos, A. Double Negative (DN) B cells: A connecting bridge between rheumatic diseases and COVID-19? Mediterr. J. Rheumatol. 2021, 32, 192. [Google Scholar] [CrossRef]
  4. Macías-Segura, N.; Castañeda-Delgado, J.E.; Bastian, Y.; Santiago-Algarra, D.; Castillo-Ortíz, J.D.; Aleman-Navarro, A.L.; Jaime-Sánchez, E.; Gomez-Moreno, M.; Saucedo-Toral, C.; Lara-Ramírez, E.E. Transcriptional signature associated with early rheumatoid arthritis and healthy individuals at high risk to develop the disease. PLoS ONE 2018, 13, e0194205. [Google Scholar] [CrossRef]
  5. Wainberg, M.; Sinnott-Armstrong, N.; Mancuso, N.; Barbeira, A.N.; Knowles, D.A.; Golan, D.; Ermel, R.; Ruusalepp, A.; Quertermous, T.; Hao, K. Opportunities and challenges for transcriptome-wide association studies. Nat. Genet. 2019, 51, 592–599. [Google Scholar] [CrossRef]
  6. Plant, D.; Maciejewski, M.; Smith, S.; Nair, N.; Maximising Therapeutic Utility in Rheumatoid Arthritis Consortium, the RAMS Study Group; Hyrich, K.; Ziemek, D.; Barton, A.; Verstappen, S. Profiling of gene expression biomarkers as a classifier of methotrexate nonresponse in patients with rheumatoid arthritis. Arthritis Rheumatol. 2019, 71, 678–684. [Google Scholar] [CrossRef]
  7. Romo-García, M.; Bastian, Y.; Zapata-Zuñiga, M.; Macías-Segura, N.; Castillo-Ortiz, J.; Lara-Ramírez, E.; Fernández-Ruiz, J.; Berlanga-Taylor, A.; González-Amaro, R.; Ramos-Remus, C. Identification of putative miRNA biomarkers in early rheumatoid arthritis by genome-wide microarray profiling: A pilot study. Gene 2019, 720, 144081. [Google Scholar] [CrossRef]
  8. Li, W.C.; Bai, D.L.; Xu, Y.; Chen, H.; Ma, R.; Hou, W.B.; Xu, R.J. Identification of differentially expressed genes in synovial tissue of rheumatoid arthritis and osteoarthritis in patients. J. Cell. Biochem. 2019, 120, 4533–4544. [Google Scholar] [CrossRef]
  9. Ashton, M.P.; Eugster, A.; Dietz, S.; Loebel, D.; Lindner, A.; Kuehn, D.; Taranko, A.E.; Heschel, B.; Gavrisan, A.; Ziegler, A.G. Association of Dendritic Cell Signatures With Autoimmune Inflammation Revealed by Single-Cell Profiling. Arthritis Rheumatol. 2019, 71, 817–828. [Google Scholar] [CrossRef]
  10. Yuan, M.; Wang, S.; Yu, L.; Qu, B.; Xu, L.; Liu, L.; Sun, H.; Li, C.; Shi, Y.; Liu, H. Long noncoding RNA profiling revealed differentially expressed lncRNAs associated with disease activity in PBMCs from patients with rheumatoid arthritis. PLoS ONE 2017, 12, e0186795. [Google Scholar] [CrossRef] [Green Version]
  11. Romo-García, M.F.; Nava-Ramírez, H.S.; Zapata-Zúñiga, M.; Macías-Segura, N.; Santiago-Algarra, D.; Castillo-Ortiz, J.D.; Bastián, Y.; Ramos-Remus, C.; Enciso-Moreno, J.A.; Castañeda-Delgado, J.E. Evaluation of SUMO1 and POU2AF1 in whole blood from rheumatoid arthritis patients and at risk relatives. Int. J. Immunogenet. 2019, 46, 59–66. [Google Scholar] [CrossRef]
  12. Burgers, L.E.; Raza, K.; Van Der Helm-Van, A.H. Window of opportunity in rheumatoid arthritis–definitions and supporting evidence: From old to new perspectives. RMD Open 2019, 5, e000870. [Google Scholar] [CrossRef] [Green Version]
  13. Pratt, A.G.; Swan, D.C.; Richardson, S.; Wilson, G.; Hilkens, C.M.; Young, D.A.; Isaacs, J.D. A CD4 T cell gene signature for early rheumatoid arthritis implicates interleukin 6-mediated STAT3 signalling, particularly in anti-citrullinated peptide antibody-negative disease. Ann. Rheum. Dis. 2012, 71, 1374–1381. [Google Scholar] [CrossRef] [Green Version]
  14. Zhang, R.; Yang, X.; Wang, J.; Han, L.; Yang, A.; Zhang, J.; Zhang, D.; Li, B.; Li, Z.; Xiong, Y. Identification of potential biomarkers for differential diagnosis between rheumatoid arthritis and osteoarthritis via integrative genome-wide gene expression profiling analysis. Mol. Med. Rep. 2019, 19, 30–40. [Google Scholar] [CrossRef] [Green Version]
  15. Cheng, Q.; Chen, X.; Wu, H.; Du, Y. Three hematologic/immune system-specific expressed genes are considered as the potential biomarkers for the diagnosis of early rheumatoid arthritis through bioinformatics analysis. J. Transl. Med. 2021, 19, 18. [Google Scholar] [CrossRef]
  16. Huang, Y.; Zheng, S.; Wang, R.; Tang, C.; Zhu, J.; Li, J. CCL5 and related genes might be the potential diagnostic biomarkers for the therapeutic strategies of rheumatoid arthritis. Clin. Rheumatol. 2019, 38, 2629–2635. [Google Scholar] [CrossRef]
  17. Fernandez-Ruiz, J.C.; Ramos-Remus, C.; Sánchez-Corona, J.; Castillo-Ortiz, J.D.; Castaneda-Sanchez, J.J.; Bastian, Y.; Romo-Garcia, M.F.; Ochoa-González, F.; Monsivais-Urenda, A.E.; González-Amaro, R. Analysis of miRNA expression in patients with rheumatoid arthritis during remission and relapse after a 5-year trial of tofacitinib treatment. Int. Immunopharmacol. 2018, 63, 35–42. [Google Scholar] [CrossRef]
  18. Luo, Q.; Liu, J.; Fu, B.; Zhang, L.; Guo, Y.; Huang, Z.; Li, J. Circular RNAs Hsa_circ_0002715 and Hsa_circ_0035197 in Peripheral Blood Are Novel Potential Biomarkers for New-Onset Rheumatoid Arthritis. Dis. Markers 2019, 2019, 2073139. [Google Scholar] [CrossRef]
  19. Castañeda-Delgado, J.E.; Bastián-Hernandez, Y.; Macias-Segura, N.; Santiago-Algarra, D.; Castillo-Ortiz, J.D.; Alemán-Navarro, A.L.; Martínez-Tejada, P.; Enciso-Moreno, L.; Lira, G.-D.; Olguín-Calderón, D. Type I interferon gene response is increased in early and established rheumatoid arthritis and correlates with autoantibody production. Front. Immunol. 2017, 8, 285. [Google Scholar] [CrossRef] [Green Version]
  20. Wright, H.L.; Cox, T.; Moots, R.J.; Edwards, S.W. Neutrophil biomarkers predict response to therapy with tumor necrosis factor inhibitors in rheumatoid arthritis. J. Leukoc. Biol. 2017, 101, 785–795. [Google Scholar] [CrossRef]
  21. Da Silva, R.G.L.; Chammas, R.; Novaes, H.M.D. Rethinking approaches of science, technology, and innovation in healthcare during the COVID-19 pandemic: The challenge of translating knowledge infrastructures to public needs. Health Res. Policy Syst. 2021, 19, 104. [Google Scholar] [CrossRef] [PubMed]
  22. Dodds, K.; Broto, V.C.; Detterbeck, K.; Jones, M.; Mamadouh, V.; Ramutsindela, M.; Varsanyi, M.; Wachsmuth, D.; Woon, C.Y. The COVID-19 pandemic: Territorial, political and governance dimensions of the crisis. Territ. Politics Gov. 2020, 8, 289–298. [Google Scholar] [CrossRef]
  23. Ascencio-Carbajal, T.; Saruwatari-Zavala, G.; Navarro-Garcia, F.; Frixione, E. Genetic/genomic testing: Defining the parameters for ethical, legal and social implications (ELSI). BMC Med. Ethics 2021, 22, 156. [Google Scholar] [CrossRef]
  24. Marchant, G.; Barnes, M.; Evans, J.P.; LeRoy, B.; Wolf, S.M. From genetics to genomics: Facing the liability implications in clinical care. J. Law Med. Ethics 2020, 48, 11–43. [Google Scholar] [CrossRef]
Table 1. Example of studies using transcriptomics to obtain candidate biomarkers with potential clinical use in RA.
Table 1. Example of studies using transcriptomics to obtain candidate biomarkers with potential clinical use in RA.
ReferenceTechnology Group(s)Tissue SampleKey FindingsClinical Use
[14]Microarray RA vs OASynovium Candidate biomarkers used together: IL7R + STAT1 (93.94% Sens; 80.77% Spec)Diagnostic
[15]Microarray RA (early and stablished) vs OASynovium Three candidate biomarkers accordingly to their AUC: GZMA (0.906), PRC1 (0.809) and TTK (0.793)Diagnostic
[16]Microarray RA vs HCSynovium Gene modules characterized by the gene expression of CCL5, CCL6, CCL9, CCL10, CCL13, and ADCY2 are potential BM for RA diagnosisDiagnostic
[4]Microarray RA vs FDRWhole bloodGene expression profiles associated with RA in high risk relatives, and gene expression of BCL2, SERPINB9, MS4A1, ETS1, EGR1, CX3CL1 and MEF2A are potential BM for RA diagnosticDiagnostic
[6]Microarray RA responders to MTX vs RA nonresponders to MTXWhole bloodTheoretical model was able to detect ~50% of nonresponders at the expense of a false negative rate of ~20%Treatment response
[17]Firefly miRNA detectionResponse to tofacitinib treatmentplasmamiRNA signature detection in plasma samples associated with clinical remission or RA flareTreatment response
[7]miRNA MicroarrayEarly RA detectionWhole bloodIdentification of early RA cases is possible due to a massive expression of miRNAs in the early phases of diseaseDiagnostic
[10]LncRNAsMicroarrayRA detectionPBMCsIdentification of the transcriptional patterns of expression associated with disease. Among these LncRNAs ENST00000456270 and NR_002838 are promising Diagnostic
RA rheumatoid arthritis, HC healthy controls, OA osteoarthritis, BM biomarkers, FDR first degree relatives, MTX methotrexate, AUC area under curve.
Table 2. Examples of biomarkers in literature with potential clinical use in RA.
Table 2. Examples of biomarkers in literature with potential clinical use in RA.
Biomarker AUCP% Sensitivity % Specificity Reference
PCNT0.742<0.000171.20%68.60%[18]
AFF20.7090.000750.90%88.60%
SIAE0.7130.000654.20%82.90%
RSAD20.750.04475.00%100.00%[19,20]
LY6E0.690.058150.00%100.00%
IFI60.710.083262.50%100.00%[20]
0.820.00570.00%94.74%[4]
WIF10.920.00187.50%92.86%[4]
MXA0.810.00580.00%80.00%
SOSTDC10.93<0.00187.50%92.86%
Values of the biomarkers where obtained when compared to healthy controls. (AUC) Area under the ROC curve.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Castañeda-Delgado, J.E.; Macias-Segura, N.; Ramos-Remus, C. Non-Coding RNAs in Rheumatoid Arthritis: Implications for Biomarker Discovery. Non-Coding RNA 2022, 8, 35. https://doi.org/10.3390/ncrna8030035

AMA Style

Castañeda-Delgado JE, Macias-Segura N, Ramos-Remus C. Non-Coding RNAs in Rheumatoid Arthritis: Implications for Biomarker Discovery. Non-Coding RNA. 2022; 8(3):35. https://doi.org/10.3390/ncrna8030035

Chicago/Turabian Style

Castañeda-Delgado, Julio Enrique, Noé Macias-Segura, and Cesar Ramos-Remus. 2022. "Non-Coding RNAs in Rheumatoid Arthritis: Implications for Biomarker Discovery" Non-Coding RNA 8, no. 3: 35. https://doi.org/10.3390/ncrna8030035

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