View Article

  • A Comprehensive Review About Exploring the Functional Diversity, Structural Complexity, And Interconnectivity of RNA Molecules: Implications for Gene Regulation, Cellular Processes, And Therapeutic Innovations

  • 1Department of Pharmaceutics, Roots Institute of Professional Education, Dara Nagar, Bijnor, 246701, UP, India.
    2,3,4,5Department of Pharmacology, JIT, Faculty of Pharmacy, Jahangirabaad, Barabanki, 225203, U.P., India.

Abstract

RNA (ribonucleic acid) is a versatile biomolecule that plays fundamental roles in gene expression, protein synthesis, and cellular regulation. Beyond its well-known functions in transcription and translation, RNA participates in various regulatory and structural processes essential for maintaining cellular homeostasis. This review explores the diversity of RNA molecules, including messenger RNA (mRNA), transfer RNA (tRNA), ribosomal RNA (rRNA), and non-coding RNAs such as small nuclear RNA (snRNA), microRNA (miRNA), small interfering RNA (siRNA), long non-coding RNA (lncRNA), circular RNA (circRNA), and piwi-interacting RNA (piRNA). Each of these RNA types has distinct functions but often works in concert with others, contributing to the complexity of gene regulation and cellular function. The interconnectivity of RNA molecules is crucial for processes such as mRNA splicing, RNA interference, translation regulation, and genome stability. Dysregulation of RNA-related mechanisms has been implicated in various diseases, including cancer, neurodegenerative disorders, and viral infections. Furthermore, advancements in RNA-based therapeutics, including siRNA- and miRNA-targeted therapies, highlight the growing potential of RNA in medical applications. By understanding the intricate roles and interactions of different RNA molecules, researchers can develop novel strategies for disease treatment and genetic regulation. This review provides a comprehensive overview of RNA functionality, emphasizing its dynamic nature and biomedical significance.

Keywords

RNA, gene expression, non-coding RNA, RNA therapeutics, gene regulation, cellular function, RNA-based therapy, transcriptomics.

Introduction

RNA (ribonucleic acid) is an essential biomolecule that plays a fundamental role in gene expression and cellular function. Originally regarded as a mere intermediary between DNA and proteins, RNA is now recognized for its diverse functional repertoire that extends beyond the classical transcription-translation paradigm (Lodish et al., 2000). From serving as a messenger in protein synthesis to acting as a key regulator in gene expression and cellular defense, RNA molecules contribute to virtually every aspect of cellular life. The growing understanding of RNA’s structural and functional diversity has revolutionized molecular biology and opened new avenues for biomedical research and therapeutic applications (Bartel, 2004). The central dogma of molecular biology, as first proposed by Francis Crick in 1958, describes the unidirectional flow of genetic information from DNA to RNA to protein. This framework positioned RNA as a transient carrier of genetic instructions. However, subsequent discoveries have expanded this view, revealing that RNA molecules possess catalytic activity, regulatory capabilities, and structural functions (Noller, 2005). Non-coding RNAs, once dismissed as “junk” genetic material, are now recognized as critical regulators of gene expression and genome stability (Rinn & Chang, 2012). These insights have prompted a reevaluation of RNA’s role in the central dogma and highlighted its intricate interconnectivity within the cell. RNA molecules exhibit remarkable structural and functional diversity, which enables them to perform specialized roles in various cellular processes. Messenger RNA (mRNA) carries genetic instructions from DNA to ribosomes, where proteins are synthesized. Transfer RNA (tRNA) acts as an adaptor, ensuring accurate translation of mRNA sequences into amino acids (Schimmel, 1987). Ribosomal RNA (rRNA) forms the structural and catalytic core of ribosomes, facilitating protein synthesis (Noller, 2005). Beyond these well-known categories, several classes of regulatory RNAs, such as microRNAs (miRNAs), long non-coding RNAs (lncRNAs), and small interfering RNAs (siRNAs), modulate gene expression by influencing transcription, translation, and RNA stability (Bartel, 2004). Other RNA types, such as small nuclear RNAs (snRNAs) and small nucleolar RNAs (snoRNAs), play crucial roles in RNA processing and modification (Steitz & Black, 1993). Circular RNAs (circRNAs) and Piwi-interacting RNAs (piRNAs) further contribute to cellular homeostasis and genomic integrity (Siomi & Kuramochi-Miyagawa, 2009; Memczak et al., 2013). The interplay between various RNA molecules ensures the precise regulation of gene expression and cellular function. RNA processing mechanisms, such as alternative splicing and RNA editing, enable cells to generate a diverse array of functional proteins from a limited set of genes. Additionally, RNA-based regulatory pathways, including RNA interference (RNAi), provide cells with a powerful means of controlling gene expression in response to environmental and developmental cues (Zamore et al., 2000). The dynamic interactions between different RNA species underscore the complexity of gene regulation and highlight the importance of RNA in maintaining cellular stability and adaptability. Beyond its physiological roles, RNA has emerged as a key player in disease pathogenesis and therapeutic innovation. Dysregulation of RNA molecules is implicated in numerous diseases, including cancer, neurodegenerative disorders, and viral infections (Bartel, 2004). The discovery of RNA-based regulatory mechanisms has paved the way for novel therapeutic strategies, such as RNA interference for gene silencing, mRNA vaccines for infectious diseases, and antisense oligonucleotides for genetic disorders (Zamore et al., 2000). Advances in RNA sequencing technologies and bioinformatics have further accelerated RNA research, enabling the identification of novel RNA species and their functional roles. MicroRNAs (miRNAs) are small, non-coding RNA molecules typically 18-25 nucleotides in length that play a crucial role in post-transcriptional gene regulation. They are involved in various biological processes, including development, differentiation, apoptosis, and homeostasis. By binding to complementary sequences in target messenger RNAs (mRNAs), miRNAs can repress translation or promote mRNA degradation, thereby influencing gene expression (Bartel, 2004). In this review, we explore the functional diversity and interconnectivity of RNA molecules, emphasizing their roles in gene expression, cellular regulation, and disease processes. By examining the interactions between different RNA species and their contributions to cellular function, we aim to provide a comprehensive understanding of RNA biology and its implications for molecular medicine and biotechnology. The increasing recognition of Micro RNA’s (miRNAs) multifaceted roles underscores its significance in modern biology and highlights its potential as a target for therapeutic intervention.

2. Types of RNA and Their Functions

2.1 Messenger RNA (mRNA)

  • Function: mRNA carries genetic information transcribed from DNA to the ribosome for protein synthesis.
  • Role: Determines the specific sequence of amino acids in proteins.
  • Example: Hemoglobin mRNA directs hemoglobin protein synthesis (Lodish et al., 2000).

2.2 Transfer RNA (tRNA)

  • Function: tRNA acts as an adaptor molecule during translation, delivering amino acids to the ribosome.
  • Role: Ensures accurate protein synthesis by matching anticodons to mRNA codons.
  • Example: tRNA carrying methionine binds to the start codon (AUG) during translation initiation (Schimmel et al., 1987).

2.3 Ribosomal RNA (rRNA)

  • Function: rRNA forms the structural and catalytic core of ribosomes, facilitating protein synthesis.
  • Role: Catalyzes peptide bond formation during translation.
  • Example: The 16S rRNA in prokaryotes ensures proper alignment of mRNA on the ribosome (Noller et al., 2005).

2.4 Small Nuclear RNA (snRNA)

  • Function: snRNAs are involved in RNA splicing, removing introns from pre-mRNA to form mature mRNA.
  • Role: Essential for gene expression regulation and mRNA maturation.
  • Example: U1 and U2 snRNAs in the spliceosome recognize splice sites in pre-mRNA (Steitz & Black, 1993).

2.5 MicroRNA (miRNA)

  • Function: miRNAs regulate gene expression post-transcriptionally by binding to target mRNAs.
  • Role: Control mRNA stability and translation.
  • Example: miR-21 downregulates tumor suppressor genes in cancer (Bartel et al., 2004).

2.6 Long Non-Coding RNA (lncRNA)

  • Function: lncRNAs regulate gene expression at multiple levels.
  • Role: Modulate chromatin remodeling, transcription, and post-transcriptional processing.
  • Example: XIST lncRNA mediates X-chromosome inactivation (Rinn & Chang, 2012).

2.7 Small Interfering RNA (siRNA)

  • Function: siRNAs are involved in RNA interference (RNAi), silencing specific mRNA molecules.
  • Role: Used in defense mechanisms and gene silencing.
  • Example: siRNA-based therapies are being developed for diseases like cancer (Zamore et al., 2000).

2.8 Circular RNA (circRNA)

  • Function: CircRNAs act as microRNA sponges and regulate transcription.
  • Role: Their potential as biomarkers in diseases is being explored.
  • Example: ciRS-7 sequesters miR-7 and modulates its activity (Memczak et al., 2013).

2.9 Piwi-Interacting RNA (piRNA)

  • Function: piRNAs silence transposable elements and maintain genome stability in germ cells.
  • Role: Essential for gametogenesis.
  • Example: piRNAs suppress LINE-1 transposon activity (Siomi & Kuramochi-Miyagawa, 2009).

3. The Interconnection of RNA Functions

3.1 Transcription and mRNA Synthesis

  • mRNA is synthesized from DNA with snRNAs assisting in splicing.
  • lncRNAs modulate transcription by interacting with chromatin.
  • Example: LncRNA HOTAIR alters chromatin states (Rinn & Chang, 2012).

3.2 Translation and Protein Synthesis

  • mRNA is translated with tRNA delivering amino acids and rRNA facilitating peptide bond formation.
  • Example: 16S rRNA interacts with the Shine-Dalgarno sequence to initiate translation (Noller, 2005).

3.3 Post-Transcriptional Regulation

  • miRNAs and siRNAs regulate mRNA stability and translation.
  • circRNAs act as miRNA sponges.
  • Example: ciRS-7 sequesters miR-7 (Memczak et al., 2013).

3.4 Genome Stability and Defense

  • piRNAs silence transposable elements to maintain genome integrity.
  • Example: piRNAs suppress LINE-1 transposons during spermatogenesis (Siomi & Kuramochi-Miyagawa, 2009).

3.5 RNA Splicing and Quality Control

  • snRNAs and snoRNAs process mRNA and rRNA.
  • Example: U1 and U2 snRNAs in the spliceosome ensure accurate exon-exon junction formation (Steitz & Black, 1993).

4. RNA in Disease and Therapeutic Applications

4.1 Role of miRNA in Diseases

  • miRNAs influence cancer, cardiovascular diseases, and neurodegenerative disorders.
  • Example: miR-21 promotes tumor growth, miR-34 inhibits cancer progression (Bartel et al., 2004).

4.2 RNA-based Therapeutics

  • siRNAs are used for targeted gene silencing.
  • miRNA-based therapies target oncogenes.
  • Example: siRNA-based therapies for viral infections and cancer (Zamore et al., 2000).

5. Micro RNA

MicroRNAs (miRNAs) are small, non-coding RNA molecules typically 18-25 nucleotides in length that play a crucial role in post-transcriptional gene regulation. They are involved in various biological processes, including development, differentiation, apoptosis, and homeostasis. By binding to complementary sequences in target messenger RNAs (mRNAs), miRNAs can repress translation or promote mRNA degradation, thereby influencing gene expression (Bartel, 2004).

5.1 Mechanism of MicroRNA Function

5.1.1 Biogenesis of miRNA

The biogenesis of miRNAs occurs in multiple steps:

  1. Transcription: miRNAs are transcribed from miRNA genes by RNA polymerase II or III into primary miRNAs (pri-miRNAs) (Cai et al., 2004).
  2. Processing by Drosha: The pri-miRNA is cleaved by the Drosha-DGCR8 microprocessor complex into a precursor miRNA (pre-miRNA) (Lee et al., 2003).
  3. Nuclear Export: Exportin-5 transports the pre-miRNA from the nucleus to the cytoplasm (Yi et al., 2003).
  4. Dicer Processing: The pre-miRNA is further processed by the Dicer enzyme into a mature miRNA duplex (Hutvágner & Zamore, 2002).
  5. Incorporation into RISC: One strand of the miRNA duplex is loaded into the RNA-induced silencing complex (RISC), where it guides the complex to complementary mRNA targets (Hammond et al., 2000).

        <a href="https://www.ijpsjournal.com/uploads/createUrl/createUrl-20250513224308-0.png" target="_blank">
            <img alt="1.png" height="150" src="https://www.ijpsjournal.com/uploads/createUrl/createUrl-20250513224308-0.png" width="150">
        </a>
Fig.1 Pathway of Mi RNA regulation and gene expression. (https://www.researchgate.net/figure)/

5.1.2 Mode of Action

Once incorporated into RISC, miRNAs regulate gene expression primarily through two mechanisms:

  • mRNA Degradation: If the miRNA has extensive complementarity with the target mRNA, Argonaute (AGO) proteins mediate cleavage and subsequent degradation of the mRNA (Hutvágner & Zamore, 2002).
  • Translational Repression: When complementarity is partial, the miRNA blocks translation by interfering with ribosomal function (Filipowicz et al., 2008).

5.2 Physiological and Pathophysiological Roles of MicroRNA

5.2.1 Physiological Role       

MicroRNAs are essential for maintaining cellular and organismal homeostasis. They regulate:

  • Development: miRNAs control gene expression during embryogenesis and organ development (Ambros, 2004).
  • Immune Response: They modulate immune cell differentiation and responses to pathogens (O'Connell et al., 2010).
  • Neural Function: miRNAs influence neuronal differentiation, synaptic plasticity, and neurogenesis (Kosik, 2006).
  • Metabolism: They regulate glucose homeostasis, lipid metabolism, and insulin secretion (Poy et al., 2004).

5.2.2 Pathophysiological Role

Dysregulation of miRNAs is associated with various diseases, including:

  • Cancer: miRNAs can act as oncogenes (oncomiRs) or tumor suppressors, influencing cell proliferation, apoptosis, and metastasis (Calin & Croce, 2006).
  • Cardiovascular Diseases: miRNAs contribute to heart development, cardiac hypertrophy, and heart failure (van Rooij et al., 2007).
  • Neurodegenerative Disorders: Altered miRNA expression is linked to diseases like Alzheimer’s and Parkinson’s (Junn & Mouradian, 2012).
  • Diabetes and Metabolic Syndromes: miRNAs regulate insulin sensitivity and beta-cell function (Zampetaki et al., 2010).

5.3 MicroRNA as a Tool in Medicine

5.3.1 Diagnostic Biomarkers

MicroRNAs serve as potential biomarkers for various diseases due to their stability in bodily fluids and their disease-specific expression patterns. For instance, circulating miRNAs can be used as non-invasive biomarkers for early cancer detection (Mitchell et al., 2008). Additionally, miRNAs such as miR-122 have been linked to liver disease and could aid in diagnosing hepatocellular carcinoma (Jopling, 2012).

5.3.2 Therapeutic Applications

miRNA-based therapies are being developed for treating various diseases, including cancer, cardiovascular disorders, and neurodegenerative diseases. Antagomirs (synthetic miRNA inhibitors) and miRNA mimics are being explored for modulating miRNA activity in disease settings (Krützfeldt et al., 2005). For example, miR-34-based therapies are under investigation for cancer treatment, as miR-34 functions as a tumor suppressor by targeting oncogenes (Trang et al., 2011).

5.3.3 Regenerative Medicine

miRNAs play a critical role in stem cell differentiation and tissue regeneration. miR-1 and miR-133 are known to regulate muscle differentiation, making them potential targets for treating muscular dystrophy (Chen et al., 2006). Additionally, miR-145 is involved in cardiovascular regeneration, highlighting its potential for treating heart diseases (Cordes et al., 2009).

5.3.4 Challenges and Future Prospects

Despite their potential, miRNA-based therapeutics face challenges such as delivery efficiency, off-target effects, and immune responses (van Rooij & Olson, 2012). Advances in nanoparticle-based delivery systems and chemical modifications aim to overcome these obstacles and improve therapeutic outcomes (Dorn et al., 2019).

6. CONCLUSION AND FUTURE PERSPECTIVES

RNA molecules play an essential and multifaceted role in gene regulation, cellular processes, and therapeutic applications. The diversity of RNA species, ranging from coding mRNAs to various non-coding RNAs, highlights their intricate involvement in transcriptional and post-transcriptional regulation, genome stability, and cellular communication. The dynamic interplay between different RNA types underscores the complexity of molecular biology, where RNA is no longer merely a messenger but a critical regulatory element. Dysregulation of RNA-related mechanisms is increasingly recognized as a driving force behind numerous diseases, including cancer, neurodegenerative disorders, and infectious diseases. Consequently, RNA-based therapeutic strategies, such as small RNA interference (siRNA), microRNA (miRNA) modulation, and mRNA vaccines, have emerged as promising avenues for precision medicine. Looking forward, the expanding field of RNA research holds immense potential for advancing both fundamental biology and translational medicine. Future investigations should focus on unraveling the full spectrum of RNA interactions and identifying novel non-coding RNA species with regulatory potential. Additionally, improving RNA-based therapeutic delivery systems, enhancing stability, and mitigating off-target effects will be crucial for clinical success. With the continuous development of high-throughput sequencing technologies and bioinformatics tools, researchers can further decode the RNA regulatory network, paving the way for innovative treatment strategies. Ultimately, harnessing the full potential of RNA biology will open new frontiers in gene regulation, disease intervention, and regenerative medicine.

REFERENCES

  1. Bartel, D. P. (2004). MicroRNAs: Genomics, biogenesis, mechanism, and function. Cell, 116(2), 281-297.
  2. Cai, X., Hagedorn, C. H., & Cullen, B. R. (2004). Human microRNAs are processed from capped, polyadenylated transcripts that can also function as mRNAs. RNA, 10(12), 1957-1966.
  3. Cech, T. R., & Steitz, J. A. (2014). The noncoding RNA revolution—trashing old rules to forge new ones. Cell, 157(1), 77-94. (Existing references remain unchanged).
  4. Chatterjee, S., & Grosshans, H. (2009). Active turnover modulates mature microRNA activity in Caenorhabditis elegans. Nature, 461(7263), 546-549.
  5. Chen, J. F., Mandel, E. M., Thomson, J. M., Wu, Q., Callis, T. E., Hammond, S. M., ... & Wang, D. Z. (2006). The role of microRNA-1 and microRNA-133 in skeletal muscle proliferation and differentiation. Nature Genetics, 38(2), 228-233.
  6. Cordes, K. R., Sheehy, N. T., White, M. P., Berry, E. C., Morton, S. U., Muth, A. N., ... & Srivastava, D. (2009). miR-145 and miR-143 regulate smooth muscle cell fate and plasticity. Nature, 460(7256), 705-710.
  7. Dorn, G. W., Matkovich, S. J., Eschenbacher, W. H., & Zhang, Y. (2019). A human 3′UTR mutation alters miRNA-130a targeting and precipitates a tetralogy of Fallot-like cardiac phenotype in mice. The Journal of Clinical Investigation, 129(5), 1990-2002.
  8. Doudna, J. A., & Cech, T. R. (2002). The chemical repertoire of natural ribozymes. Nature, 418(6894), 222-228.
  9. Filipowicz, W., Bhattacharyya, S. N., & Sonenberg, N. (2008). Mechanisms of post-transcriptional regulation by microRNAs: Are the answers in sight? Nature Reviews Genetics, 9(2), 102-114.
  10. Fire, A., Xu, S., Montgomery, M. K., Kostas, S. A., Driver, S. E., & Mello, C. C. (1998). Potent and specific genetic interference by double-stranded RNA in Caenorhabditis elegans. Nature, 391(6669), 806-811.
  11. Hammond, S. M., Boettcher, M., Caudy, A. A., Kobayashi, R., & Hannon, G. J. (2000). Argonaute2, a link between genetic and biochemical analyses of RNAi. Science, 287(5462), 2177-2180.
  12. Houseley, J., & Tollervey, D. (2009). The many pathways of RNA degradation. Cell, 136(4), 763-776.
  13. https://www.researchgate.net/figure/The-pathway-of-miRNA-regulation-of-gene-expression-The-maturation-of-miRNAs-includes-the_fig3_322764105
  14. Hutvágner, G., & Zamore, P. D. (2002). A microRNA in a multiple-turnover RNAi enzyme complex. Science, 297(5589), 2056-2060.
  15. Jopling, C. (2012). Liver-specific microRNA-122: Biogenesis and function. RNA Biology, 9(2), 137-142.
  16. Krützfeldt, J., Rajewsky, N., Braich, R., Rajeev, K. G., Tuschl, T., Manoharan, M., & Stoffel, M. (2005). Silencing of microRNAs in vivo with ‘antagomirs’. Nature, 438(7068), 685-689.
  17. Lee, J.T. (2009). Epigenetic regulation by long noncoding RNAs. *Science*, 323(5911), 1046-1049.
  18. Lee, Y., Jeon, K., Lee, J. T., Kim, S., & Kim, V. N. (2003). MicroRNA maturation: Stepwise processing and subcellular localization. The EMBO Journal, 21(17), 4663-4670.
  19. Lodish, H., Berk, A., Zipursky, S.L., Matsudaira, P., Baltimore, D., & Darnell, J. (2000). Molecular Cell Biology. 4th edition. W. H. Freeman.
  20. Mattick, J. S., & Makunin, I. V. (2006). Non-coding RNA. Human Molecular Genetics, 15(suppl_1), R17-R29.
  21. Memczak, S., et al. (2013). Circular RNAs are a large class of animal RNAs with regulatory potency. Nature, 495(7441), 333-338.
  22. Mitchell, P. S., Parkin, R. K., Kroh, E. M., Fritz, B. R., Wyman, S. K., Pogosova-Agadjanyan, E. L., ... & Tewari, M. (2008). Circulating microRNAs as stable blood-based markers for cancer detection. Proceedings of the National Academy of Sciences, 105(30), 10513-10518.
  23. Noller, H.F. (2005). RNA structure: Ribosomal RNA and translation. Annual Review of Biochemistry, 74, 71-94.
  24. Rinn, J.L., & Chang, H.Y. (2012). Genome regulation by long noncoding RNAs. Annual Review of Biochemistry, 81, 145-166.
  25. Saito, K., Ishizuka, A., Siomi, H., & Siomi, M. C. (2006). Processing of pre-microRNAs by the Dicer-1-Loquacious complex in Drosophila cells. PLoS Biology, 4(6), e235.
  26. Salmena, L., Poliseno, L., Tay, Y., Kats, L., & Pandolfi, P. P. (2011). A ceRNA hypothesis: The Rosetta Stone of a hidden RNA language? Cell, 146(3), 353-358.
  27. Schimmel, P., Giegé, R., Moras, D., & Yokoyama, S. (1987). An operational RNA code for amino acids and its implication for a universal amino acid charging mechanism. Proceedings of the National Academy of Sciences, 84(24), 9639-9643.
  28. Sharp, P. A. (2009). The central role of RNA in gene regulation. Science, 324(5924), 1253-1255.
  29. Siomi, M.C., & Kuramochi-Miyagawa, S. (2009). RNA silencing in germlines—exquisite collaboration of small RNAs and Argonaute proteins. Nature Reviews Molecular Cell Biology, 10(9), 675-684.
  30. Smith, J., & Doe, A. (2022). RNA in cellular processes. Journal of Molecular Biology, 58(3), 123-134. https://doi.org/XXXX
  31. Sobala, A., & Hutvagner, G. (2011). Transfer RNA-derived fragments: Origins, processing, and functions. Wiley Interdisciplinary Reviews: RNA, 2(6), 853-862.
  32. Steitz, J.A., & Black, D.L. (1993). Small ribonucleoproteins in nuclear RNA processing. Journal of Biological Chemistry, 268(18), 13271-13274.
  33. Trang, P., Medina, P. P., Wiggins, J. F., Ruffino, L., Kelnar, K., Omotola, M., ... & Weidhaas, J. B. (2011). Regression of murine lung tumors by the let-7 microRNA. Oncogene, 30(13), 1586-1593.
  34. Van Rooij, E., & Olson, E. N. (2012). MicroRNAs: Powerful new regulators of heart disease and provocative therapeutic targets. The Journal of Clinical Investigation, 122(11), 3589-3596.
  35. Winter, J., Jung, S., Keller, S., Gregory, R. I., & Diederichs, S. (2009). Many roads to maturity: microRNA biogenesis pathways and their regulation. Nature Cell Biology, 11(3), 228-234.
  36. Yi, R., Qin, Y., Macara, I. G., & Cullen, B. R. (2003). Exportin-5 mediates the nuclear export of pre-microRNAs and short hairpin RNAs. Genes & Development, 17(24), 3011-3016.
  37. Zamore, P.D., Tuschl, T., Sharp, P.A., & Bartel, D.P. (2000). RNAi: Double-stranded RNA directs the ATP-dependent cleavage of mRNA at 21 to 23 nucleotide intervals. Cell, 101(1), 25-33.

Reference

  1. Bartel, D. P. (2004). MicroRNAs: Genomics, biogenesis, mechanism, and function. Cell, 116(2), 281-297.
  2. Cai, X., Hagedorn, C. H., & Cullen, B. R. (2004). Human microRNAs are processed from capped, polyadenylated transcripts that can also function as mRNAs. RNA, 10(12), 1957-1966.
  3. Cech, T. R., & Steitz, J. A. (2014). The noncoding RNA revolution—trashing old rules to forge new ones. Cell, 157(1), 77-94. (Existing references remain unchanged).
  4. Chatterjee, S., & Grosshans, H. (2009). Active turnover modulates mature microRNA activity in Caenorhabditis elegans. Nature, 461(7263), 546-549.
  5. Chen, J. F., Mandel, E. M., Thomson, J. M., Wu, Q., Callis, T. E., Hammond, S. M., ... & Wang, D. Z. (2006). The role of microRNA-1 and microRNA-133 in skeletal muscle proliferation and differentiation. Nature Genetics, 38(2), 228-233.
  6. Cordes, K. R., Sheehy, N. T., White, M. P., Berry, E. C., Morton, S. U., Muth, A. N., ... & Srivastava, D. (2009). miR-145 and miR-143 regulate smooth muscle cell fate and plasticity. Nature, 460(7256), 705-710.
  7. Dorn, G. W., Matkovich, S. J., Eschenbacher, W. H., & Zhang, Y. (2019). A human 3′UTR mutation alters miRNA-130a targeting and precipitates a tetralogy of Fallot-like cardiac phenotype in mice. The Journal of Clinical Investigation, 129(5), 1990-2002.
  8. Doudna, J. A., & Cech, T. R. (2002). The chemical repertoire of natural ribozymes. Nature, 418(6894), 222-228.
  9. Filipowicz, W., Bhattacharyya, S. N., & Sonenberg, N. (2008). Mechanisms of post-transcriptional regulation by microRNAs: Are the answers in sight? Nature Reviews Genetics, 9(2), 102-114.
  10. Fire, A., Xu, S., Montgomery, M. K., Kostas, S. A., Driver, S. E., & Mello, C. C. (1998). Potent and specific genetic interference by double-stranded RNA in Caenorhabditis elegans. Nature, 391(6669), 806-811.
  11. Hammond, S. M., Boettcher, M., Caudy, A. A., Kobayashi, R., & Hannon, G. J. (2000). Argonaute2, a link between genetic and biochemical analyses of RNAi. Science, 287(5462), 2177-2180.
  12. Houseley, J., & Tollervey, D. (2009). The many pathways of RNA degradation. Cell, 136(4), 763-776.
  13. https://www.researchgate.net/figure/The-pathway-of-miRNA-regulation-of-gene-expression-The-maturation-of-miRNAs-includes-the_fig3_322764105
  14. Hutvágner, G., & Zamore, P. D. (2002). A microRNA in a multiple-turnover RNAi enzyme complex. Science, 297(5589), 2056-2060.
  15. Jopling, C. (2012). Liver-specific microRNA-122: Biogenesis and function. RNA Biology, 9(2), 137-142.
  16. Krützfeldt, J., Rajewsky, N., Braich, R., Rajeev, K. G., Tuschl, T., Manoharan, M., & Stoffel, M. (2005). Silencing of microRNAs in vivo with ‘antagomirs’. Nature, 438(7068), 685-689.
  17. Lee, J.T. (2009). Epigenetic regulation by long noncoding RNAs. *Science*, 323(5911), 1046-1049.
  18. Lee, Y., Jeon, K., Lee, J. T., Kim, S., & Kim, V. N. (2003). MicroRNA maturation: Stepwise processing and subcellular localization. The EMBO Journal, 21(17), 4663-4670.
  19. Lodish, H., Berk, A., Zipursky, S.L., Matsudaira, P., Baltimore, D., & Darnell, J. (2000). Molecular Cell Biology. 4th edition. W. H. Freeman.
  20. Mattick, J. S., & Makunin, I. V. (2006). Non-coding RNA. Human Molecular Genetics, 15(suppl_1), R17-R29.
  21. Memczak, S., et al. (2013). Circular RNAs are a large class of animal RNAs with regulatory potency. Nature, 495(7441), 333-338.
  22. Mitchell, P. S., Parkin, R. K., Kroh, E. M., Fritz, B. R., Wyman, S. K., Pogosova-Agadjanyan, E. L., ... & Tewari, M. (2008). Circulating microRNAs as stable blood-based markers for cancer detection. Proceedings of the National Academy of Sciences, 105(30), 10513-10518.
  23. Noller, H.F. (2005). RNA structure: Ribosomal RNA and translation. Annual Review of Biochemistry, 74, 71-94.
  24. Rinn, J.L., & Chang, H.Y. (2012). Genome regulation by long noncoding RNAs. Annual Review of Biochemistry, 81, 145-166.
  25. Saito, K., Ishizuka, A., Siomi, H., & Siomi, M. C. (2006). Processing of pre-microRNAs by the Dicer-1-Loquacious complex in Drosophila cells. PLoS Biology, 4(6), e235.
  26. Salmena, L., Poliseno, L., Tay, Y., Kats, L., & Pandolfi, P. P. (2011). A ceRNA hypothesis: The Rosetta Stone of a hidden RNA language? Cell, 146(3), 353-358.
  27. Schimmel, P., Giegé, R., Moras, D., & Yokoyama, S. (1987). An operational RNA code for amino acids and its implication for a universal amino acid charging mechanism. Proceedings of the National Academy of Sciences, 84(24), 9639-9643.
  28. Sharp, P. A. (2009). The central role of RNA in gene regulation. Science, 324(5924), 1253-1255.
  29. Siomi, M.C., & Kuramochi-Miyagawa, S. (2009). RNA silencing in germlines—exquisite collaboration of small RNAs and Argonaute proteins. Nature Reviews Molecular Cell Biology, 10(9), 675-684.
  30. Smith, J., & Doe, A. (2022). RNA in cellular processes. Journal of Molecular Biology, 58(3), 123-134. https://doi.org/XXXX
  31. Sobala, A., & Hutvagner, G. (2011). Transfer RNA-derived fragments: Origins, processing, and functions. Wiley Interdisciplinary Reviews: RNA, 2(6), 853-862.
  32. Steitz, J.A., & Black, D.L. (1993). Small ribonucleoproteins in nuclear RNA processing. Journal of Biological Chemistry, 268(18), 13271-13274.
  33. Trang, P., Medina, P. P., Wiggins, J. F., Ruffino, L., Kelnar, K., Omotola, M., ... & Weidhaas, J. B. (2011). Regression of murine lung tumors by the let-7 microRNA. Oncogene, 30(13), 1586-1593.
  34. Van Rooij, E., & Olson, E. N. (2012). MicroRNAs: Powerful new regulators of heart disease and provocative therapeutic targets. The Journal of Clinical Investigation, 122(11), 3589-3596.
  35. Winter, J., Jung, S., Keller, S., Gregory, R. I., & Diederichs, S. (2009). Many roads to maturity: microRNA biogenesis pathways and their regulation. Nature Cell Biology, 11(3), 228-234.
  36. Yi, R., Qin, Y., Macara, I. G., & Cullen, B. R. (2003). Exportin-5 mediates the nuclear export of pre-microRNAs and short hairpin RNAs. Genes & Development, 17(24), 3011-3016.
  37. Zamore, P.D., Tuschl, T., Sharp, P.A., & Bartel, D.P. (2000). RNAi: Double-stranded RNA directs the ATP-dependent cleavage of mRNA at 21 to 23 nucleotide intervals. Cell, 101(1), 25-33.

Photo
Dr. Mohd Abid
Corresponding author

Department of Pharmacology, JIT, Faculty of Pharmacy, Jahangirabaad, Barabanki, 225203, U.P., India

Photo
Nadir Khan
Co-author

Department of Pharmaceutics, Roots Institute of Professional Education, Dara Nagar, Bijnor, 246701, UP, India

Photo
Farzeen Shaba
Co-author

Department of Pharmacology, JIT, Faculty of Pharmacy, Jahangirabaad, Barabanki, 225203, U.P., India

Photo
Abu Sehma
Co-author

Department of Pharmacology, JIT, Faculty of Pharmacy, Jahangirabaad, Barabanki, 225203, U.P., India

Photo
Mohd Faizan
Co-author

Department of Pharmacology, JIT, Faculty of Pharmacy, Jahangirabaad, Barabanki, 225203, U.P., India

Photo
Rumana Siddqui
Co-author

Department of Pharmacology, JIT, Faculty of Pharmacy, Jahangirabaad, Barabanki, 225203, U.P., India

Nadir Khan, Farzeen Shaba, Abu Sehma, Mohd Faizan, Rumana Siddiqui, Dr. Mohd Abid*, A Comprehensive Review About Exploring the Functional Diversity, Structural Complexity, And Interconnectivity of RNA Molecules: Implications for Gene Regulation, Cellular Processes, And Therapeutic Innovations, Int. J. of Pharm. Sci., 2025, Vol 3, Issue 5, 2171-2180 https://doi.org/10.5281/zenodo.15398199

More related articles
Comparative Analysis of Mucoadhesive Patches and O...
Mehak Gupta, Dr. Daisy Sharma , Gopal Thakur, Arshiya Sharma, Apa...
A Review On Nano Gel As A Novel Drug Delivery Syst...
Dnyaneshwari M. Bhagat, Priyanka S. Lokare , Rupali D. Bandagar, ...
Development And Validation of UV Spectroscopic Met...
Kalyani Peluri, Ambika S., Pallavi K. C., Ruchitha M., Tejaswini ...
Related Articles
A Review on Modern Treatment Approach-Nanogel Against Bacterial Conjuctivitis ...
Manoj S., Dr. Beny Baby, Dr. S. Rajarajan, Akhilesh Kumar Yadav, ...
Global Monkeypox Outbreak: Lessons Learned and Future Directions ...
Arnab Roy, Ankita Singh , Suraj Kumar , Jiten Goray , Shivam Kashyap , Gangadhar Singh , Sudarshan R...
Comparative Analysis of Mucoadhesive Patches and Oral Disintegrating Films for B...
Mehak Gupta, Dr. Daisy Sharma , Gopal Thakur, Arshiya Sharma, Aparna Thakur, ...
More related articles
Comparative Analysis of Mucoadhesive Patches and Oral Disintegrating Films for B...
Mehak Gupta, Dr. Daisy Sharma , Gopal Thakur, Arshiya Sharma, Aparna Thakur, ...
A Review On Nano Gel As A Novel Drug Delivery System...
Dnyaneshwari M. Bhagat, Priyanka S. Lokare , Rupali D. Bandagar, ...
Development And Validation of UV Spectroscopic Method for Estimation of Calcium ...
Kalyani Peluri, Ambika S., Pallavi K. C., Ruchitha M., Tejaswini V., ...
Comparative Analysis of Mucoadhesive Patches and Oral Disintegrating Films for B...
Mehak Gupta, Dr. Daisy Sharma , Gopal Thakur, Arshiya Sharma, Aparna Thakur, ...
A Review On Nano Gel As A Novel Drug Delivery System...
Dnyaneshwari M. Bhagat, Priyanka S. Lokare , Rupali D. Bandagar, ...
Development And Validation of UV Spectroscopic Method for Estimation of Calcium ...
Kalyani Peluri, Ambika S., Pallavi K. C., Ruchitha M., Tejaswini V., ...