Fact-checked by Grok 2 weeks ago

RNA

Ribonucleic acid (RNA) is a linear polymer of ribonucleotides, each consisting of a ribose sugar, a phosphate group, and one of four nitrogenous bases—adenine (A), cytosine (C), guanine (G), or uracil (U)—that plays central roles in coding, decoding, regulation, and expression of genes in most living organisms and many viruses. Unlike deoxyribonucleic acid (DNA), RNA is typically single-stranded, allowing it to fold into complex three-dimensional structures that enable its diverse functions, though some RNA viruses contain double-stranded forms as their genetic material. The of RNA features a backbone formed by 5' to 3' phosphodiester bonds between the sugars and phosphates, with the 2' hydroxyl group on distinguishing it from DNA's and contributing to RNA's reactivity and flexibility. Bases pair via hydrogen bonds—A with U (two bonds) and C with G (three bonds)—creating secondary structures such as stems, loops, and hairpins, while tertiary folding into helices, bulges, and pseudoknots supports specific interactions with proteins, other RNAs, and small molecules. RNA exists in multiple types, each with specialized structures and roles; the three primary types involved in protein synthesis are messenger RNA (mRNA), which is transcribed from DNA and carries the genetic code to ribosomes as a single-stranded chain averaging 1,000–10,000 nucleotides; transfer RNA (tRNA), a cloverleaf-shaped molecule of about 70–90 nucleotides that decodes mRNA codons via its anticodon loop to deliver specific amino acids; and ribosomal RNA (rRNA), which comprises the structural and catalytic core of ribosomes (70S in prokaryotes, 80S in eukaryotes) and accounts for up to 80% of cellular RNA. Beyond these, non-coding RNAs include small nuclear RNAs (snRNAs), such as U1–U6, which form spliceosomes to process pre-mRNA by removing introns; small nucleolar RNAs (snoRNAs), which guide chemical modifications like methylation and pseudouridylation on rRNA, tRNA, and snRNA in the nucleolus; and regulatory RNAs like microRNAs (miRNAs) and small interfering RNAs (siRNAs), which mediate gene silencing through RNA interference. Other notable types encompass long non-coding RNAs (lncRNAs) that influence chromatin structure and transcription, circular RNAs that act as miRNA sponges or regulators, and ribozymes—catalytic RNAs capable of self-splicing or cleaving phosphodiester bonds. The core function of RNA is to bridge DNA's genetic information to functional proteins via transcription (DNA to mRNA) and translation (mRNA to polypeptide chains on ribosomes), a process essential for cellular and growth. RNAs also regulate by modulating transcription, mRNA stability, and translation efficiency, as seen in miRNA-mediated repression or lncRNA scaffolding of protein complexes. Certain RNAs exhibit enzymatic activity as ribozymes, facilitating reactions like formation in ribosomes or removal, while in RNA viruses, RNA serves directly as the heritable replicated by RNA-dependent RNA polymerases. Dysfunctions in RNA processing or structure underlie diseases such as and certain cancers, highlighting RNA's therapeutic potential, including in mRNA vaccines that encode antigens to elicit immune responses.

Structure and Composition

Nucleotide Components

RNA is a linear composed of repeating units known as , which are linked together to form the chain. Each consists of three primary components: a sugar called , a group, and one of four nitrogenous bases— (A), (G), (C), or uracil (U). The nitrogenous bases are heterocyclic aromatic compounds; and are purines with a fused double-ring structure, while and uracil are pyrimidines featuring a single six-membered ring. These bases attach to the sugar via an N-glycosidic bond at the C1' position of the sugar and the N9 (for purines) or N1 (for pyrimidines) of the base. The ribose sugar in RNA is specifically β-D-ribofuranose, existing in a five-membered furanose ring conformation with hydroxyl groups at the 2', 3', and 5' positions. This configuration arises from the furanose form of D-ribose, where the ring oxygen is between C1' and C4', and the β-anomer has the base attached above the plane of the ring. The distinctive 2'-hydroxyl (2'-OH) group on the ribose enhances RNA's chemical reactivity compared to deoxyribose in DNA, as it participates in nucleophilic attacks that facilitate processes like self-cleavage or enzymatic modifications. For instance, the chemical formula of adenosine monophosphate (AMP), the ribonucleotide containing adenine, is C_{10}H_{14}N_5O_7P, illustrating the integration of these components. Similar formulas apply to the other ribonucleotides: guanosine monophosphate (GMP, C_{10}H_{14}N_5O_8P), cytidine monophosphate (CMP, C_9H_{14}N_3O_8P), and uridine monophosphate (UMP, C_9H_{13}N_2O_9P). The phosphodiester backbone of RNA forms through covalent bonds between the phosphate group of one ribonucleotide and the sugar of the adjacent one, creating a directional chain. Specifically, the 3'-OH of the ribose in one nucleotide reacts with the 5'-phosphate attached to the adjacent nucleotide, forming a phosphodiester linkage and resulting in a polymer with 5' to 3' polarity. This asymmetry imparts directionality to the RNA strand, with the 5' end typically bearing a phosphate or triphosphate group and the 3' end terminating in a hydroxyl. The 2'-OH group contributes to RNA's relative instability, rendering it susceptible to hydrolysis under basic conditions; the hydroxyl acts as a nucleophile to attack the adjacent phosphodiester bond, forming a 2',3'-cyclic phosphate intermediate that leads to chain cleavage. This reactivity contrasts with the stability of DNA and underscores RNA's transient role in cellular processes.

Differences from DNA

One key chemical difference between RNA and DNA lies in the substitution of uracil (U) for (T) as one of the nitrogenous bases in RNA. Uracil pairs with (A) through two bonds, similar to the A-T pairing in DNA, whereas (G) pairs with (C) via three bonds in both molecules, contributing to the overall of base pairing. DNA uses instead of uracil to allow detection and repair of cytosine , which produces uracil; if DNA used uracil, such damage would be indistinguishable from normal bases. In RNA, uracil is used as it is energetically cheaper to synthesize (lacking the ) and RNA's short lifespan reduces the impact of mutations. Another fundamental distinction is the sugar component: RNA incorporates , which has a hydroxyl group (-OH) at the 2' position of the sugar ring, whereas DNA uses lacking this group. The 2'-OH group in RNA enhances the conformational flexibility of the single-stranded molecule, allowing it to adopt diverse shapes more readily than the more rigid DNA backbone. However, this same group renders RNA more vulnerable to enzymatic and chemical , as it can participate in nucleophilic attacks on the phosphodiester backbone, briefly tying back to the nucleotide's inherent reactivity. RNA is typically single-stranded, in contrast to DNA's double-helical structure, which impacts their respective and . The absence of a complementary strand in RNA reduces base-pairing protection, making it less stable and more prone to unfolding or , while DNA's double helix provides greater and resistance to environmental damage for long-term genetic storage. This single-stranded nature also facilitates RNA's role as a transient intermediary rather than a permanent . RNA molecules are generally much shorter in length than DNA, with most ranging from hundreds to thousands of , compared to DNA's genome-spanning millions. Consequently, RNA exhibits a higher turnover rate, with half-lives often ranging from minutes to hours depending on the type and cellular conditions, enabling rapid , whereas DNA persists stably across cell generations for archival purposes. Physicochemically, RNA demonstrates higher solubility in water than DNA, attributable to the polar 2'-OH group increasing hydrophilicity. This feature, combined with the single-stranded form, promotes RNA's tendency to form complex intramolecular folds through base pairing and stacking interactions, unlike the more uniform double helix of DNA. Additionally, RNA's susceptibility to degradation by ubiquitous ribonucleases (RNases) far exceeds DNA's vulnerability to deoxyribonucleases (DNases), underscoring its ephemeral nature in cellular environments.

Folding and Higher-Order Structures

The primary structure of RNA, defined by its linear of ribonucleotides, encodes the intrinsic potential for folding into functional three-dimensional conformations through specific interactions. This dictates the locations and types of complementary bases available for , influencing the and architecture of higher-order structures. Seminal studies have established that variations in primary can profoundly alter folding pathways and final structures, underscoring the as the foundational determinant of RNA's conformational landscape. RNA secondary structures emerge from intramolecular base pairing along the primary chain, forming double-stranded helices interspersed with single-stranded regions. Canonical Watson-Crick base pairs (A-U and G-C) provide the core stability via two or three hydrogen bonds, respectively, while non-canonical interactions, such as the G-U wobble pair, introduce flexibility and are nearly as thermodynamically stable as Watson-Crick pairs due to their isomorphic geometry and comparable hydrogen bonding. These pairings create characteristic motifs, including stem-loops (where a helical stem terminates in a loop), hairpins (short stems closed by loops of 3-7 nucleotides), bulges (unpaired bases on one side of a helix), and internal loops (unpaired regions on both sides). Such elements allow RNA to adopt compact, hierarchical architectures that serve as scaffolds for tertiary folding.00112-X) Tertiary structures result from the spatial organization of multiple secondary elements through long-range contacts, yielding compact globular forms essential for RNA function. Key motifs include pseudoknots, in which a single-stranded from one stem pairs with a distant to interlock helices; coaxial helices, where adjacent helical segments stack continuously without interruption; and kissing loops, involving reciprocal base pairing between the loops of two hairpins. These interactions enable complex topologies, as exemplified by the L-shaped tertiary fold in tRNA, which positions its acceptor and anticodon arms orthogonally, or the precisely organized active sites in ribozymes that catalyze formation. The driving forces behind RNA folding encompass a balance of enthalpic and entropic contributions from non-covalent interactions. Hydrogen bonding between base edges stabilizes paired regions, while base stacking—arising from van der Waals and hydrophobic forces between adjacent aromatic bases—provides the dominant stabilization in helical segments, contributing up to 50% of the in double-stranded RNA. Electrostatic repulsion from the negatively charged backbone is mitigated by divalent cations like Mg²⁺ ions, which bind specifically to facilitate contacts and neutralize charges, often increasing folding efficiency by orders of magnitude in physiological conditions.00112-X) Predicting RNA folding computationally relies on thermodynamic models that approximate minimization. Algorithms like mfold, introduced in seminal work using dynamic programming to enumerate suboptimal structures, and the ViennaRNA package, which implements partition function calculations for ensemble predictions, model secondary structures by scoring base pairs based on nearest-neighbor parameters derived from melting experiments. These tools excel for sequences up to several hundred but face limitations from kinetic traps—local energy minima that trap RNA in metastable states during folding, as opposed to the global minimum—leading to discrepancies between predicted and native structures . Advanced extensions incorporate barrier-crossing heuristics to address these kinetic effects.

Chemical Modifications

RNA molecules undergo a diverse array of post-transcriptional chemical modifications that alter their structure and function, building upon the core components of , , , , and . Over 170 distinct types of these modifications have been identified as of 2025, with the majority occurring in eukaryotic organisms and prominently in (rRNA), (tRNA), and (mRNA).30638-4) These modifications include base methylations, such as N6-methyladenosine (m6A) on , pseudouridylation where is isomerized to (Ψ), and ribose 2'-O-methylation on the sugar backbone. Among these, m6A stands out as the most abundant internal modification in eukaryotic mRNA, often found in the DRACH (where D = A/G/U, R = A/G, H = A/C/U). The installation, removal, and interpretation of these modifications are mediated by enzymatic complexes known as writers, erasers, and readers, respectively. For m6A, the primary writer is the METTL3-METTL14-WTAP methyltransferase complex, which catalyzes the addition of a to the N6 position of . Erasers, such as the demethylase FTO, reverse this modification by oxidative demethylation, thereby dynamically regulating m6A levels. Readers, including YTH-domain-containing proteins like YTHDF2, recognize and bind to modified sites to influence downstream RNA processes, such as directing m6A-marked transcripts to decay pathways. Similar machinery exists for other modifications; for instance, pseudouridine synthases (e.g., enzymes) act as writers for Ψ without requiring erasers, while fibrillarin catalyzes site-specific 2'-O-methylations in rRNA. These chemical alterations profoundly impact RNA by enhancing against degradation, modulating base-pairing interactions, and fine-tuning processes like and efficiency. For example, m6A promotes mRNA decay through YTHDF2-mediated recruitment to , thereby reducing protein output, while 2'-O-methylation stabilizes RNA structures and improves translational fidelity in rRNA. Pseudouridylation enhances RNA flexibility and , facilitating proper tRNA anticodon recognition during and influencing site selection in pre-mRNA. Such modifications collectively regulate at multiple levels, with disruptions linked to diseases including cancer and neurological disorders. RNA modifications exhibit evolutionary conservation, particularly in essential RNAs like tRNA and rRNA, where core sites such as m6A in transcripts are preserved from to humans, underscoring their fundamental roles in cellular homeostasis.00451-2) Detection of these modifications has advanced through epitranscriptomics, employing techniques like for quantitative profiling of abundant RNAs and sequencing-based methods, such as m6A-seq or Pseudo-seq, which use pulldowns or chemical labeling to map modification sites genome-wide with single-nucleotide resolution. These approaches have revealed dynamic, context-dependent modification patterns that respond to cellular stresses and developmental cues.00147-7)

Synthesis and Processing

Transcription Mechanism

Transcription is the enzymatic process by which RNA is synthesized from template, involving the of ribonucleotides in the 5' to 3' direction to produce a complementary RNA strand. This DNA-directed synthesis uses the DNA as a , where the reads one strand (the template strand) and assembles RNA using triphosphates (NTPs) that match the complementary bases. In prokaryotes, a single , composed of core subunits and a for promoter recognition, catalyzes the transcription of all RNA types. In eukaryotes, three distinct RNA polymerases perform specialized roles: (Pol I) transcribes most ribosomal RNAs (rRNAs), (Pol II) synthesizes messenger RNAs (mRNAs) and small RNAs (snRNAs), and (Pol III) produces transfer RNAs (tRNAs) and 5S rRNA. The transcription process occurs in three main stages: initiation, elongation, and termination. Initiation begins with the binding of RNA polymerase to promoter elements on the DNA. In prokaryotes, the core promoter includes the -35 box (TTGACA consensus) and -10 box (TATAAT consensus), recognized by the sigma factor to unwind DNA and form the open complex. In eukaryotes, Pol II initiation involves the TATA box (TATAAA consensus, located 25-35 bases upstream of the start site), bound by the TATA-binding protein (TBP) as part of the transcription factor IID (TFIID) complex, which recruits additional factors and the polymerase. Eukaryotic promoters may also include enhancers, distal regulatory sequences that boost transcription rates. During elongation, the RNA polymerase moves along the DNA template, incorporating NTPs (ATP, GTP, CTP, UTP) complementary to the template bases, extending the RNA chain in the 5' to 3' direction at rates of about 20-50 per second in prokaryotes and 22-25 per second for Pol II in eukaryotes. The energy for this comes from the of the high-energy phosphoanhydride bonds in NTPs, releasing (PPi) and driving the irreversible addition of each . Fidelity is maintained through base-pairing selectivity and mechanisms; the initial misincorporation error rate is approximately 1 in 10^4 , improved by intrinsic cleavage activity in some polymerases, such as Pol III, which removes mismatched 3' termini via hydrolytic , enhancing accuracy by up to 10^3-fold. Overall transcription error rates reach about 10^{-5} per in like E. coli. Termination signals the end of RNA synthesis and release of the transcript. In prokaryotes, involves the formation of a GC-rich loop in the RNA followed by a run of uracils, causing pausing and dissociation, while rho-dependent termination uses the Rho helicase protein to unwind the RNA-DNA hybrid. Eukaryotic termination for Pol II occurs downstream of the signal, involving cleavage and release, though mechanisms vary by type.

Post-Transcriptional Processing

Post-transcriptional encompasses a series of modifications that transform the primary RNA transcript, known as pre-mRNA in eukaryotes, into mature, functional RNA molecules. These steps occur in the and are crucial for RNA stability, export to the , and proper . In prokaryotes, processing is minimal due to coupled transcription and translation, whereas eukaryotic processing is more elaborate to accommodate larger genomes and regulatory complexity. One of the initial modifications is 5' , which involves the addition of a 7-methylguanosine to the 5' end of the nascent pre-mRNA shortly after transcription initiation. This is covalently linked via a 5'-5' triphosphate bridge by the guanylyltransferase, followed by . The protects the RNA from 5' exonucleases, facilitates nuclear export through interactions with export factors like NXF1, and enhances translation initiation by recruiting the eIF4E. At the 3' end, occurs after of the pre-mRNA at a specific site defined by the AAUAAA signal, followed by the addition of a poly-A tail consisting of 200-250 residues in eukaryotes. This process is catalyzed by a multiprotein including cleavage and specificity factor (CPSF) and poly-A polymerase (). The poly-A tail increases mRNA stability by preventing degradation from 3' exonucleases and promotes export and translation efficiency via binding to poly-A binding proteins (PABPs). Splicing removes non-coding introns and joins coding to form mature mRNA, a process mediated by the in eukaryotes, which assembles from small nuclear ribonucleoproteins (snRNPs) U1 through U6. The recognizes conserved splice sites ( at the 5' end and at the 3' end of introns) and catalyzes two reactions to excise introns. Some introns, such as group I and II, can self-splice without proteins, relying on RNA . , where different exon combinations are selected, generates multiple protein isoforms from a single , expanding proteomic diversity. RNA editing introduces base changes post-transcriptionally, with adenosine-to-inosine (A-to-I) editing being prevalent in eukaryotes, performed by enzymes that deaminate to , which is read as during . This can alter codons, potentially changing or creating stop codons, thus modulating protein function and diversity. For instance, editing in transcripts affects calcium permeability in neurons. mechanisms ensure only properly processed RNAs proceed, with (NMD) targeting transcripts containing premature termination codons for degradation. NMD involves recognition by factors like UPF1, UPF2, and UPF3 during the pioneer round of , preventing accumulation of truncated proteins. This pathway degrades about 5-30% of human transcripts, highlighting its role in regulating . Secondary structures in the RNA can influence transcription termination signals, thereby affecting the substrate for these processing events.

Major Types and Functions

Protein-Coding RNAs

Protein-coding RNAs, primarily messenger RNAs (mRNAs), serve as the intermediary molecules that convey genetic information from DNA to ribosomes for protein synthesis, embodying a core aspect of the central dogma of molecular biology. In eukaryotic cells, mRNAs are typically monocistronic, encoding a single protein from one open reading frame, whereas prokaryotic mRNAs are often polycistronic, allowing multiple proteins to be translated from a single transcript organized into operons. The structure of mRNA includes a 5' untranslated region (UTR) that regulates translation initiation, a central coding sequence composed of nucleotide triplets known as codons that specify amino acid sequences, and a 3' UTR that influences mRNA stability, localization, and translation efficiency, often ending with a poly-A tail in eukaryotes. The of mRNA into proteins occurs in three main stages: , , and termination. During in eukaryotes, the small ribosomal subunit binds to the 5' cap of the mRNA with assistance from eukaryotic initiation factors (eIFs), scanning to the () recognized via the for efficient assembly of the full . follows as transfer RNAs (tRNAs) match their anticodons to successive mRNA codons in the 's A site, facilitating formation and translocation along the mRNA. Termination is triggered by stop codons (UAA, UAG, UGA) in the A site, prompting release factors to disassemble the and liberate the nascent polypeptide. Following transcription in the , mature eukaryotic mRNAs are exported to the through complexes, where they localize to specific cellular compartments for targeted , such as dendrites in neurons. mRNA stability is tightly regulated, with degradation initiated by deadenylation (shortening of the poly-A tail) followed by and exonucleolytic digestion, ensuring rapid turnover of transcripts in response to cellular needs. In terms of abundance, mRNAs constitute approximately 1-5% of total cellular RNA in eukaryotes, with half-lives ranging from minutes for short-lived transcripts like the proto-oncogene c-fos to several hours for more stable ones, reflecting their role in dynamic control. Evolutionarily, protein-coding RNAs are thought to trace back to an ancient , where self-replicating RNA molecules encoded rudimentary peptides via a primitive , laying the foundation for modern systems. Prior to translation, mRNA precursors undergo processing steps like capping, splicing, and to generate functional transcripts.

Regulatory Non-Coding RNAs

Regulatory non-coding RNAs (ncRNAs) are a diverse class of RNA molecules that do not encode proteins but play crucial roles in modulating at transcriptional, post-transcriptional, and epigenetic levels. These RNAs, ranging from short 20-30 species to long transcripts exceeding 200 nucleotides, interact with DNA, RNA, or proteins to fine-tune cellular processes such as development, differentiation, and response to stress. Unlike protein-coding RNAs, their primary function lies in regulation rather than translation, enabling precise control over genome activity without altering the genetic code directly. MicroRNAs (miRNAs) are small endogenous ncRNAs approximately 21-25 in length that primarily repress post-transcriptionally. They are initially transcribed as primary miRNAs (pri-miRNAs) with stem-loop structures, which are processed in the by the complex containing and DGCR8 to form precursor miRNAs (pre-miRNAs). These precursors are then exported to the cytoplasm and cleaved by into mature miRNAs, which are loaded into the (RISC) containing proteins. Within RISC, miRNAs typically bind to the 3' untranslated regions (UTRs) of target mRNAs through partial base-pairing, leading to translational repression or mRNA destabilization and decay. This mechanism allows a single miRNA to regulate hundreds of targets, influencing processes like and ; for instance, the founding miRNA lin-4 was discovered in C. elegans where it negatively regulates LIN-14 protein levels during development. Small interfering RNAs (siRNAs) are structurally similar to miRNAs, also 20-25 nucleotides long and processed by , but they arise primarily from exogenous double-stranded RNA (dsRNA) precursors and mediate sequence-specific (RNAi) for . Unlike miRNAs, siRNAs often exhibit perfect complementarity to their targets, triggering direct cleavage by Argonaute-2 in the RISC complex rather than translational repression. This pathway was first demonstrated in C. elegans, where injection of dsRNA corresponding to specific genes led to potent and heritable silencing, far more effective than single-stranded RNA. siRNAs play key roles in antiviral defense and transposon suppression, with applications in experimental across eukaryotes. Long non-coding RNAs (lncRNAs), defined as transcripts longer than 200 nucleotides, exhibit diverse regulatory functions including modification, transcriptional interference, and post-transcriptional modulation. Many lncRNAs act as scaffolds for protein complexes, recruiting histone modifiers like Polycomb repressive complex 2 (PRC2) to specific genomic loci. A prominent example is , a 17-19 kb lncRNA essential for X-chromosome inactivation in female mammals, where it coats the inactive in cis, leading to epigenetic silencing through recruitment of silencing factors and compaction. Another well-studied lncRNA, HOTAIR (HOX transcript antisense RNA), is a 2.2 kb transcript from the HOXC locus that represses HOXD genes in trans by interacting with PRC2 and LSD1 to promote H3K27 methylation and H3K4 demethylation, respectively, thereby establishing repressive domains. lncRNAs like these are implicated in developmental patterning and cancer progression when dysregulated. Enhancer RNAs (eRNAs) are short, often bidirectional ncRNAs transcribed from enhancer regions, typically 50-2000 long, that facilitate enhancer-promoter interactions to activate transcription. eRNAs promote by stabilizing loops, recruiting and complexes, or interacting with transcription factors like YY1 to enhance activity at target promoters. Their discovery stemmed from genome-wide mapping of nascent transcripts, revealing pervasive enhancer transcription in active states. For example, eRNAs from the β-globin locus enhancers loop to interact with the promoter, boosting expression during . eRNA levels correlate with enhancer activity, providing a dynamic readout of regulatory potential. Piwi-interacting RNAs (piRNAs), 24-31 nucleotides long, form complexes with proteins to silence transposons primarily in cells, protecting integrity from mutagenic insertions. Unlike miRNAs and siRNAs, piRNAs are generated from long single-stranded precursors via a Dicer-independent pathway involving endonuclease, and they exhibit a bias for at the 5' end. In animals, piRNAs guide to transposon loci, inducing formation through H3K9 methylation or transcriptional repression. The ping-pong amplification cycle, where primary piRNAs direct cleavage of sense transcripts to produce secondary piRNAs, amplifies the response. Seminal studies in mice identified piRNAs bound to MIWI and , clustered in germline-specific loci, underscoring their role in and transposon control.

Structural and Catalytic RNAs

Structural and catalytic RNAs encompass a diverse class of non-coding RNAs that provide essential scaffold and enzymatic functions within the , most prominently in the machinery and RNA processing pathways. These molecules, including ribosomal RNAs (rRNAs) and transfer RNAs (tRNAs), form the core structural components of the , facilitating protein , while ribozymes demonstrate RNA's capacity for independent of proteins. Unlike protein-coding or regulatory RNAs, structural and catalytic RNAs primarily enable constitutive cellular processes through their architectural and reactive properties. Ribosomal RNA (rRNA) constitutes the majority of the ribosome's mass and serves as its structural and functional backbone. In prokaryotes, the small ribosomal subunit contains 16S rRNA, while the large subunit includes 23S and 5S rRNAs; in eukaryotes, these correspond to 18S rRNA in the small subunit and 28S, 5.8S, and 5S rRNAs in the large subunit. These rRNAs fold into complex three-dimensional structures that position ribosomal proteins and create functional sites for . A highly conserved region within the 23S/28S rRNA forms the peptidyl transferase center (PTC), the site responsible for catalyzing formation during protein synthesis. Transfer RNA (tRNA) molecules act as adapters , linking to their corresponding codons on through specific . The canonical tRNA secondary adopts a cloverleaf conformation, characterized by an acceptor , D-, anticodon , and T-, which folds into an L-shaped . The anticodon loop, located at one end of the L-shape, contains a three-nucleotide anticodon sequence that base-pairs with mRNA codons to ensure accurate selection. At the opposite end, the 3' terminus serves as the attachment site for the , a process catalyzed by aminoacyl-tRNA synthetases that recognize specific tRNA identity elements to achieve high-fidelity charging. Ribozymes represent RNA molecules with intrinsic catalytic activity, exemplified by self-splicing introns and RNase P. Group I self-splicing introns excise themselves from precursor RNAs using a nucleotide or its derivatives as a cofactor, initiating reactions that join the flanking exons without protein assistance. In contrast, undergo self-splicing via two steps, resulting in a lariat intermediate where the intron's 5' end branches to a bulged , mirroring the mechanism of spliceosomal introns. RNase P, a ribonucleoprotein , processes the 5' leader sequence of precursor tRNAs to generate mature tRNAs; its RNA subunit alone exhibits catalytic activity , cleaving pre-tRNA substrates in the presence of monovalent and divalent cations. The itself functions as a , with its activity residing entirely within the rRNA component of the large subunit. Biochemical and structural studies have shown that the PTC, composed of rRNA without direct involvement of ribosomal proteins, catalyzes the nucleophilic attack of the aminoacyl-tRNA's alpha-amino group on the peptidyl-tRNA's ester linkage to form a . This RNA-based underscores the ancient evolutionary origins of the , predating protein synthesis machinery. Post-transcriptional modifications enhance the stability and functionality of structural RNAs like tRNAs and rRNAs. In tRNAs, hypermodifications such as wybutosine at position 37 (adjacent to the anticodon) stabilize the codon-anticodon interaction through base-stacking, preventing frameshifting and promoting efficient fidelity. Similar modifications in rRNAs, including pseudouridylation and 2'-O-methylation in the PTC and decoding regions, fine-tune ribosomal and catalytic .

Genetic and Evolutionary Roles

RNA Genomes and Viruses

RNA serves as the hereditary material in numerous viruses, where it functions as the genome rather than DNA, enabling rapid replication and evolution in host cells. This contrasts with the DNA-based genomes of cellular organisms and most organelles, highlighting RNA's versatility in genetic systems. The Baltimore classification system categorizes viruses into seven groups based on their genome type (DNA or RNA, single- or double-stranded, sense or antisense) and replication strategy, with groups III through VI encompassing those with RNA genomes. Developed in 1971, this framework underscores how RNA viruses exploit host machinery while relying on virus-encoded polymerases for genome replication, often leading to high genetic diversity. Positive-sense single-stranded RNA (+ssRNA) viruses, classified in group IV, possess genomes that directly serve as (mRNA) upon entry into host cells, allowing immediate of proteins including (RdRp). For example, (a ) uses its ~7.5 +ssRNA genome to produce RdRp, which then synthesizes a complementary negative-sense strand as a template for new +ssRNA genomes, facilitating efficient replication in cytoplasmic membrane-bound compartments. Negative-sense single-stranded RNA (-ssRNA) viruses, in group V, carry their genome in an antisense and package RdRp within the virion to first transcribe positive-sense mRNAs for protein before full genome replication can occur. , with its segmented ~13.5 -ssRNA genome, exemplifies this: the complex initiates transcription in the , producing mRNAs capped by host factors, which are translated to support subsequent replication of full-length antigenomic intermediates. Double-stranded RNA (dsRNA) viruses, grouped in class III, feature genomes composed of 10–12 linear segments encapsidated within the virion, providing stability against host nucleases. Reoviruses, such as mammalian orthoreovirus, have a ~24 kb dsRNA divided into large, medium, and small segments, each encoding specific proteins; replication occurs in cytoplasmic viral factories where endogenous RdRp transcribes and replicates segments without free RNA intermediates, enabling genetic reassortment during co-infection. Retroviruses, in group VI, maintain single-stranded RNA genomes (~9 kb) that are reverse-transcribed into DNA , which integrate into the host as stable hereditary elements. Human immunodeficiency virus (HIV-1), for instance, forms a double-stranded DNA via its RNA template, allowing persistent infection and propagation with host DNA during . A key challenge in RNA virus replication stems from the error-prone nature of RdRp enzymes, which lack proofreading mechanisms found in DNA polymerases, resulting in mutation rates of approximately 10^{-4} to 10^{-5} errors per nucleotide per replication cycle—orders of magnitude higher than DNA-based systems. This quasispecies diversity drives rapid viral evolution, immune evasion, and adaptation to antiviral therapies, but also imposes fitness costs on progeny virions. In organelles like mitochondria and chloroplasts, genetic material is primarily circular DNA (e.g., ~16 kb mitochondrial DNA in humans), yet in animal cells, RNA transcripts from mitochondrial genomes can constitute a significant portion (up to 30%) of the cellular mRNA pool in metabolically active tissues like heart muscle, supporting organelle function and indirectly influencing hereditary traits through post-transcriptional regulation. Evolutionarily, RNA genomes in viruses suggest ancient origins, potentially mirroring primordial RNA-world scenarios, and their high mutability facilitates diversification across host species, contributing to zoonotic emergences.

Reverse Transcription and Retroelements

Reverse transcription is the process by which DNA is synthesized from an RNA template, a mechanism first discovered in 1970 by Howard Temin and Satoshi Mizutani, who identified an RNA-dependent in virions of , and independently by in RNA tumor viruses, challenging the . This enzyme, known as (RT), was recognized for its role in retroviral replication, earning Temin and Baltimore the 1975 in or . Reverse transcriptase is a multifunctional encoded by the pol gene in retroviruses, such as HIV-1, where it is produced as part of the Gag-Pol polyprotein and exhibits both activity for RNA-templated synthesis and RNase H activity to degrade the RNA strand in RNA-DNA . In HIV-1, RT forms a heterodimer consisting of p66 (catalytic subunit) and p51 (structural subunit), with the polymerase domain in p66 responsible for nucleotide addition and the RNase H domain cleaving RNA in hybrid duplexes. The enzyme's error-prone nature, with a rate of approximately 1 error per 10,000 incorporated, contributes to high mutation rates in retroviral genomes, driving viral diversity and . The mechanism of reverse transcription begins upon retroviral entry into , where uses a tRNA primer annealed to the viral RNA genome's primer to synthesize a strand, forming an RNA-DNA hybrid. RNase H activity then partially degrades the RNA template, allowing the newly synthesized DNA to fold back via repeat sequences to prime second-strand synthesis, ultimately yielding a double-stranded DNA (dsDNA) flanked by long terminal repeats (LTRs) generated through template switching during the process. These LTRs, identical direct repeats at both ends of the proviral DNA, contain promoter and enhancer elements essential for viral gene expression after integration into genome. Retroelements are that propagate via RNA intermediates and reverse transcription, comprising a significant portion of eukaryotic genomes. Endogenous retroviruses (ERVs), ancient integrations of retroviral proviruses, account for about 8% of the , with many retaining LTRs but lacking functional gag, pol, and env genes due to over evolutionary time. Non-LTR retrotransposons include (LINEs), such as LINE-1, which encode their own RT and are autonomously mobile, and short interspersed nuclear elements (SINEs), like Alu elements, which are non-autonomous and rely on LINE-1 machinery for retrotransposition via RNA intermediates. Alu elements, the most abundant in with over one million copies, amplify through transcription into RNA, reverse transcription, and reintegration, influencing genome structure and sometimes contributing to genetic disorders. Telomerase represents a cellular application of reverse transcription, consisting of the (TERT) protein subunit with activity and the telomerase RNA component (TERC), which provides the sequence for adding telomeric repeats to ends. In humans, TERC's 3'-CAC-5' directs TERT to extend the 3' overhang of telomeres by adding TTAGGG repeats, counteracting replicative shortening and maintaining genomic stability in and cancer cells. This ribonucleoprotein complex exemplifies how reverse transcription mechanisms, originally identified in viruses, are co-opted for essential eukaryotic functions.

Double-Stranded and Circular RNAs

Double-stranded RNA (dsRNA) serves as a critical intermediate in the replication of many RNA viruses, where it is generated by viral RNA-dependent RNA polymerases (RdRps) during the synthesis of complementary strands from positive-sense RNA templates. This biogenesis process is essential for viral genome amplification, as RdRps use the dsRNA as a template to produce progeny genomic RNA. In cellular contexts, dsRNA also arises during RNA interference (RNAi) pathways, where double-stranded precursors are processed into small interfering RNAs (siRNAs) to silence gene expression. dsRNA plays a pivotal role in triggering innate immune responses, primarily through activation of the protein kinase R (PKR) pathway, which phosphorylates 2α (eIF2α) to inhibit global protein translation and induce stress responses. This activation leads to the of type I interferons (IFNs) via downstream signaling, enhancing antiviral defenses by upregulating interferon-stimulated genes. Additionally, dsRNA serves as a trigger for RNAi-mediated antiviral immunity, where enzymes cleave it into siRNAs that guide proteins to degrade viral RNA. In viruses with RNA genomes, these dsRNA intermediates are key targets for host recognition, linking them to broader antiviral mechanisms. Evolutionarily, dsRNA structures are conserved in innate immune pathways across eukaryotes, suggesting ancient origins in antiviral defense. Circular RNAs (circRNAs) are a class of non-coding RNAs formed through back-splicing, a process where a downstream donor joins an upstream acceptor, often facilitated by complementary sequences in flanking introns or . Biogenesis can involve lariat intermediates from canonical splicing, where introns are removed and the lariat's branch point enables to form the circular structure. This covalent closure renders circRNAs highly resistant to degradation, conferring greater stability compared to linear RNAs. circRNAs exert regulatory functions, notably as microRNA (miRNA) sponges that sequester miRNAs and prevent their interaction with target mRNAs; a prominent example is ciRS-7 (also known as CDR1as), which harbors over 70 binding sites for miR-7 and modulates neuronal . In select cases, circRNAs undergo translation to produce proteins, particularly when containing internal ribosome entry sites (IRES) or undergoing cap-independent initiation, as observed in some and cellular circRNAs. Detection of circRNAs typically involves enrichment with RNase R, an that digests linear RNAs but spares circular forms, followed by RNA sequencing to identify back-spliced junctions. In certain cell types, such as neurons, circRNAs can comprise 10-20% of the , reflecting their high abundance and stability. Recent studies since 2020 have highlighted circRNAs' involvement in neurodegeneration, where dysregulated circRNAs in Alzheimer's and Parkinson's diseases influence synaptic function and amyloid-beta accumulation through miRNA sponging and protein interactions. For dsRNA, post-2020 research has elucidated its role in enhancing antiviral transcriptional responses independent of sequence-specific recognition, bolstering innate immunity against emerging pathogens like SARS-CoV-2. Evolutionarily, , emerging from , represent an evolutionary innovation in higher organisms, with over 100,000 identified in humans, potentially enhancing regulatory diversity without genomic expansion.

Historical and Fundamental Discoveries

Early Identification and Characterization

In 1869, Swiss biochemist Friedrich Miescher isolated a phosphorus-rich substance he termed "nuclein" from the nuclei of white blood cells obtained from discarded surgical bandages, marking the first identification of nucleic acids, which include both DNA and RNA. Miescher's extraction involved treating the cells with pepsin to remove proteins, followed by alkali to precipitate the nuclein, revealing its acidic nature and high phosphorus content, distinct from known proteins or lipids. Although Miescher did not distinguish between DNA and RNA at the time, his work laid the groundwork for recognizing nucleic acids as fundamental cellular components. The identification of RNA as a distinct emerged in the late 1930s through studies on the (TMV). In 1936, Roy Markham and Northrop demonstrated the presence of a in purified TMV preparations, and by 1937, Bawden and Pirie confirmed it contained , indicating a composition. Further analysis in 1939 by Bawden and Pirie established that the was ribonucleic acid (RNA), not deoxyribonucleic acid, based on its susceptibility to hydrolysis and base composition. Wendell Stanley, who had crystallized TMV in 1935, collaborated with Bawden and Pirie, and by 1944, their collective work proposed RNA's potential role in viral heredity, challenging the protein-centric views of inheritance prevalent at the time. In the , the emerging field of began elucidating RNA's functional roles in protein synthesis. proposed the "adaptor hypothesis" in 1955, suggesting that molecules act as intermediaries to translate the sequences of a genetic into chains, addressing the mismatch between the four-letter code and the twenty . This idea, detailed in Crick's 1958 paper "On Protein Synthesis," posited that these adaptors—later identified as transfer RNAs (tRNAs)—recognize specific codons via base-pairing while carrying attached , thus serving as the bridge in the . The hypothesis provided a conceptual framework for RNA's intermediary function, influencing subsequent experiments on genetic coding. The concept of messenger RNA (mRNA) was experimentally validated in 1961 through studies by Sydney Brenner, François Jacob, and Matthew Meselson using T4 bacteriophage infection in Escherichia coli. Their pulse-labeling experiments with radioactive uracil demonstrated the existence of a short-lived, unstable RNA species that rapidly incorporates genetic information from newly synthesized DNA and directs protein synthesis at ribosomes. Published in Nature as "An Unstable Intermediate Carrying Information from Genes to Ribosomes for Protein Synthesis," the work showed that this RNA turns over quickly, with a half-life of about 2-3 minutes, confirming its role as a transient messenger between DNA and ribosomes. This discovery resolved debates about how genetic information flows in bacteria and established mRNA as the key intermediary in gene expression. The 1970s brought structural insights into RNA components and the revelation of gene organization complexities. In 1974, Alexander Rich and Sung-Hou Kim, along with colleagues, determined the three-dimensional crystal structure of yeast phenylalanine tRNA at 3.0 Å resolution, revealing its L-shaped tertiary fold with stacked helices and a cloverleaf secondary structure stabilized by modified bases and magnesium ions. This structure, resolved using diffraction on crystals grown from purified tRNA, confirmed Crick's adaptor by showing the anticodon loop positioned to interact with mRNA and the acceptor stem for attachment. Concurrently, in 1977, Phillip and Richard Roberts independently discovered introns—non-coding sequences interrupting eukaryotic s—through electron microscopy of adenovirus RNA hybrids, revealing looped-out regions where introns are transcribed but spliced out during mRNA maturation. 's team at and Roberts' at Cold Spring Harbor used heteroduplex mapping to show that the beta-globin and s contain intervening sequences, fundamentally altering views of gene continuity. These findings, awarded the 1993 in Physiology or Medicine, highlighted RNA's role in post-transcriptional processing.

Milestones in RNA Function and Regulation

In 1982, Thomas Cech's laboratory discovered the self-splicing capability of the ribosomal RNA intron from Tetrahymena thermophila, demonstrating that RNA could catalyze its own excision without protein assistance, thus identifying the first ribozyme. This breakthrough challenged the prevailing view that only proteins function as enzymes and paved the way for understanding RNA's catalytic potential. Independently, in 1983, Sidney Altman's group showed that the RNA component of RNase P from Escherichia coli performs the catalytic cleavage of tRNA precursors, confirming RNA's enzymatic role in vivo. Their discoveries, recognized with the 1989 Nobel Prize in Chemistry, established ribozymes as key players in RNA processing and regulation, influencing fields from splicing mechanisms to synthetic biology. The late 1990s brought further revelations in RNA-mediated gene silencing with the 1998 identification of RNA interference (RNAi) by Andrew Fire and Craig Mello, who demonstrated that double-stranded RNA triggers potent, sequence-specific degradation of homologous mRNAs in Caenorhabditis elegans. This work elucidated the core RNAi pathway involving small interfering RNAs (siRNAs) and microRNAs (miRNAs), which guide Argonaute proteins to target transcripts for cleavage or translational repression, thereby regulating gene expression at the post-transcriptional level. Awarded the 2006 Nobel Prize in Physiology or Medicine, RNAi revolutionized functional genomics, enabling targeted gene knockdown and inspiring therapeutic applications like siRNA drugs for viral infections and genetic disorders. The and marked an explosion in recognizing regulatory non-coding RNAs, particularly long non-coding RNAs (lncRNAs), with early examples like —first sequenced in 1991 but functionally characterized in the mid- for its role in X-chromosome inactivation through coating and silencing. By 2012, the project's GENCODE consortium cataloged over 9,000 human lncRNA loci, revealing their widespread expression and diverse regulatory functions, such as epigenetic modulation and transcriptional interference, far beyond initial annotations. This systematic annotation, building on computational pipelines from the late , highlighted lncRNAs' prevalence—comprising up to 80% of the non-coding —and spurred genome-wide studies into their roles in development and disease. Advancements in epitranscriptomics emerged in the 2010s, with Kate D. Meyer's 2012 development of MeRIP-seq enabling transcriptome-wide mapping of N⁶-methyladenosine (m⁶A), the most abundant internal mRNA modification, enriched near stop codons and in 3' UTRs to influence , stability, and translation. Concurrently, Julia Salzman's 2012 analysis uncovered circular RNAs (circRNAs) as predominant isoforms from thousands of human genes, formed via back-splicing and functioning as miRNA sponges or regulators of parental , challenging linear RNA paradigms. These discoveries expanded RNA to include chemical modifications and topologies, with m⁶A "writers" like METTL3 and circRNA abundance in neural tissues underscoring their tissue-specific impacts. From 2020 onward, research has illuminated RNA's role in biomolecular condensates, particularly within stress granules—cytoplasmic assemblies that sequester mRNAs during cellular stress to halt and promote . A pivotal 2020 study revealed G3BP1 as a core driver, where RNA binding induces its conformational switch to trigger liquid-liquid , dynamically partitioning RNAs for selective protection or degradation. Single-cell RNA sequencing (scRNA-seq) has further resolved regulatory networks, with tools like IReNA (2022) integrating scRNA-seq and scATAC-seq to infer cell-type-specific interactions, uncovering dynamic modules in heterogeneous tissues like tumors. The accelerated RNA research through mRNA vaccines, which by 2021 demonstrated scalable production and immune efficacy, spurring over 200 clinical trials for non-viral applications like cancer immunotherapies and boosting lipid nanoparticle delivery innovations. These developments, from 2020 to 2025, have integrated phase-separated RNA dynamics with high-resolution profiling, transforming regulatory insights and therapeutic pipelines.

RNA in Abiogenesis and Prebiotic Chemistry

The RNA world hypothesis posits that RNA served as both the genetic material and catalyst in the earliest stages of life on Earth, preceding the emergence of DNA and proteins. Proposed by Walter Gilbert in 1986, this model suggests that self-replicating RNA molecules capable of catalyzing their own replication and basic metabolic reactions formed the foundation of prebiotic evolution. In this scenario, RNA's dual functionality—storing information like DNA and performing enzymatic roles like proteins—allowed it to bootstrap the complexity of life without requiring more sophisticated biopolymers initially. Prebiotic synthesis pathways for RNA components remain a central focus, with research exploring plausible geochemical environments. , the building blocks of RNA, could have formed in settings such as formamide-rich pools or hydrothermal vents, where simple precursors like and react under mild conditions to yield sugars and nucleobases. A landmark achievement came in , when Matthew Powner and colleagues demonstrated the synthesis of ribonucleotides—such as and derivatives—from simple prebiotic molecules like , , and , bypassing the unstable free and requiring only wet-dry cycles for activation. This pathway, conducted under conditions mimicking , produced activated with 3'-5' phosphodiester linkages, addressing a key hurdle in RNA . Hydrothermal vents provide another proposed site, where surfaces catalyze assembly from CO2 and H2 under high-temperature gradients. Despite these advances, significant challenges persist in reconstructing a fully RNA-based prebiotic . One major issue is the preferential formation of non-standard 2'-5' phosphodiester linkages during non-enzymatic , which destabilize RNA duplexes and hinder template-directed replication compared to the biologically relevant 3'-5' bonds. These aberrant linkages arise because prebiotic reactions often activate the 2'-hydroxyl group on , leading to branched polymers that are less stable and prone to . experiments, such as Sol Spiegelman's 1967 work with Qβ phage RNA replicase, illustrate the dynamics of RNA simplification under selective pressure; serial transfer in test tubes produced "Spiegelman's monster," a truncated 218-nucleotide RNA that replicated rapidly but lost non-essential genetic information, highlighting the ease of evolutionary regression without stabilizing mechanisms. Supporting evidence for the RNA world includes ribozymes that mimic primitive metabolic functions, demonstrating RNA's catalytic versatility. For instance, in vitro-selected ribozymes have been engineered to perform reactions akin to glycolysis intermediates, such as carbon-carbon bond formation, suggesting that early RNA networks could sustain basic without proteins. Extraterrestrial delivery of RNA precursors further bolsters the hypothesis; the , which fell in 1969, contains a suite of nucleobases including , , , uracil, and , with isotopic signatures indicating abiotic synthesis in space. These compounds, detected at concentrations up to 70 , could have seeded Earth's prebiotic soups with ready-made building blocks. Recent computational and experimental studies from 2023 to 2025 have refined models of prebiotic RNA pathways using and co-evolution simulations. In 2024, studies on vesicle-RNA co-evolution demonstrated that vesicles encapsulate short RNA oligomers, enhancing their stability and enabling template-directed in dilute prebiotic conditions, with encapsulated RNAs showing up to 10-fold faster replication rates compared to free molecules. These models suggest that membranes and RNA co-emerged, facilitating the transition from abiotic chemistry to Darwinian . , as seen in modern structural RNAs, provides a brief analog for such ancient functions, where self-splicing introns hint at RNA processing capabilities.

Applications in Medicine and Biotechnology

Therapeutic RNA Molecules

Therapeutic RNA molecules represent a rapidly advancing class of pharmaceuticals that leverage RNA's natural roles in and regulation to treat diseases. These include messenger RNA (mRNA) vaccines, antisense oligonucleotides (), small interfering RNAs (siRNAs), and aptamers, each designed to modulate specific biological processes such as protein production, , or protein binding. Unlike traditional small-molecule drugs, offer high specificity and the potential for rapid development, particularly in response to emerging threats like infectious diseases or genetic disorders. Their clinical success has been enabled by innovations in chemical modifications and delivery systems to overcome inherent RNA vulnerabilities. mRNA vaccines, a breakthrough in prophylactic and therapeutic applications, instruct host cells to produce antigenic proteins that trigger immune responses. The Pfizer-BioNTech vaccine (BNT162b2), approved by the U.S. (FDA) in December 2020 under , and the Moderna vaccine (mRNA-1273), authorized shortly thereafter, both encode the within nucleoside-modified mRNA encapsulated in nanoparticles (LNPs) for efficient cellular uptake and from . Upon , the mRNA is translated by ribosomes into the , eliciting neutralizing antibodies and T-cell immunity without using live virus. These vaccines demonstrated over 90% efficacy in preventing symptomatic in phase 3 trials, marking the first widespread deployment of mRNA technology in humans. Antisense oligonucleotides (ASOs) function by hybridizing to target RNA sequences to alter splicing, block , or induce , providing precise control over . Nusinersen (Spinraza), an ASO approved by the FDA in December 2016 for (), binds to an intronic splicing silencer site in SMN2 pre-mRNA, promoting of 7 to increase full-length survival (SMN) protein production. Administered intrathecally, it has shown significant improvements in motor function for infants and children with SMA in clinical trials, with sustained benefits observed over multiple years. Similarly, siRNA therapeutics exploit to silence disease-causing genes. Patisiran (Onpattro), approved by the FDA in August 2018 for hereditary transthyretin-mediated (hATTR) , is a lipid nanoparticle-formulated siRNA conjugated to (GalNAc) for hepatocyte-specific targeting via the . It reduces hepatic (TTR) production by over 80% in patients, alleviating symptoms as evidenced in the APOLLO phase 3 trial. Aptamers, single-stranded RNA or DNA ligands selected for high-affinity binding to target proteins, offer a non-immunogenic alternative for inhibiting protein function. Pegaptanib (Macugen), the first FDA-approved aptamer in December 2004 for neovascular (wet) age-related macular degeneration (AMD), is a 27-nucleotide RNA molecule pegylated for stability that specifically binds the vascular endothelial growth factor (VEGF165) isoform, preventing its interaction with receptors and reducing pathological angiogenesis in the retina. Intravitreal injections slowed vision loss in about 70% of treated patients in pivotal trials, establishing aptamers as viable therapeutics despite later competition from protein-based anti-VEGFs. Key challenges in RNA therapeutics include rapid enzymatic degradation and innate immune activation, which can limit efficacy and cause adverse reactions. To enhance , modifications such as (Ψ) are incorporated, as in mRNA vaccines, where Ψ substitution reduces recognition, lowers immunogenicity, and boosts translation efficiency by up to 10-fold compared to unmodified RNA. LNPs and GalNAc conjugates further address delivery barriers by facilitating endosomal escape and tissue-specific uptake, though off-target effects and manufacturing scalability remain hurdles. As of 2025, mRNA platforms are expanding into , with BioNTech's individualized neoantigen-specific mRNA vaccines (e.g., autogene cevumeran) showing promising immune activation and tumor reduction in phase 2 trials for pancreatic and cancers, with phase 3 studies planned or initiated in 2025. Recent approvals include donidalorsen in August 2025 for , further expanding the portfolio of RNA-based treatments. These developments underscore RNA's potential for , with over a dozen RNA drugs now FDA-approved.

Diagnostic and Research Tools

(RT-PCR) and quantitative PCR (qPCR) are foundational techniques for RNA detection in diagnostics, involving the conversion of RNA to followed by amplification to quantify or levels. These methods gained prominence during the for detection, where RT-qPCR served as the gold standard due to its high (detecting as few as 10-100 RNA copies) and specificity exceeding 99%, enabling rapid identification of infected individuals from nasopharyngeal swabs. Limitations include potential false negatives from low loads or sample degradation, but optimizations like one-step RT-qPCR have improved throughput for large-scale testing. RNA sequencing () represents a high-throughput approach for comprehensive analysis, capturing the full spectrum of RNA molecules to profile , , and novel transcripts in research and disease diagnostics. By sequencing cDNA libraries from RNA samples, RNA-seq provides quantitative data on thousands of genes simultaneously, outperforming earlier methods in and resolution, with applications in identifying biomarkers for cancers like through differential expression patterns. Single-cell RNA-seq (scRNA-seq) extends this by resolving cellular heterogeneity, isolating transcripts from individual cells to map rare subpopulations, such as tumor-infiltrating immune cells, which bulk methods obscure; for instance, scRNA-seq has revealed subtype-specific gene signatures in heterogeneity with over 10,000 cells profiled per sample. Microarrays and Northern blots offer targeted tools for RNA expression profiling, though they have been largely supplanted by sequencing in modern workflows. DNA microarrays hybridize labeled RNA or cDNA to immobilized probes on a , enabling parallel assessment of up to 50,000 genes to detect expression changes, as validated in studies of inflammatory responses where fold-changes correlated with clinical outcomes. Northern blots, a classical gel-based , separate RNA by size via , transfer it to a , and detect specific transcripts using radiolabeled or chemiluminescent probes, providing size confirmation and quantification for validation, such as confirming levels in developmental tissues with sensitivity down to 1-5 pg of target RNA. In situ hybridization (ISH) enables spatial visualization of RNA localization within intact tissues, using labeled probes to bind target sequences and reveal expression patterns at cellular resolution. RNA-ISH, often fluorescent (FISH), has been pivotal in for mapping mRNA distribution in sections, identifying localized transcripts like those for neurotransmitters with single-molecule sensitivity, and in for diagnosing viral infections or oncogenic fusions in tumor biopsies. Advances like branched DNA amplification in ISH platforms enhance signal detection in formalin-fixed tissues, achieving multiplexed analysis of up to 48 RNA targets simultaneously. Spatial transcriptomics, exemplified by the Visium platform, integrates with tissue imaging to map at near-single-cell resolution, addressing limitations of dissociated samples by preserving spatial context. Introduced in 2019 and refined by 2024 with high-definition versions capturing 2-micron pixels across 1 cm² sections, Visium has elucidated tumor microenvironments in , identifying spatially segregated immune niches with over 18,000 genes profiled per spot. These methods support research into tissue architecture in diseases like , where zonal gene gradients inform . Post-2023 integrations of (AI) with RNA diagnostics have enhanced analysis of complex datasets, such as using on RNA-seq outputs for predictive modeling in . AI algorithms, like deep neural networks trained on scRNA-seq data, classify cancer subtypes with 95% accuracy by detecting subtle expression patterns missed by traditional statistics, as demonstrated in pancreatic ductal adenocarcinoma diagnostics. In viral diagnostics, AI-optimized RT-qPCR interpretation reduces false positives by 20% through in amplification curves, accelerating outbreak responses.

Emerging Synthetic and Editing Technologies

Emerging advancements in RNA synthetic and editing technologies have revolutionized and by enabling precise manipulation of genetic material at the RNA level. The CRISPR-Cas systems, originally derived from bacterial immune defenses, utilize single-guide RNAs (sgRNAs) to direct nucleases for targeted DNA cleavage and editing, allowing for insertions, deletions, or base substitutions in eukaryotic genomes. In parallel, Cas13 variants target RNA directly, facilitating transient knockdown or cleavage without altering the underlying DNA sequence, which is particularly useful for studying function or degrading viral RNAs. RNA editing technologies have advanced through the integration of components with endogenous enzymes like , which naturally catalyze adenosine-to-inosine (A-to-I) in RNA transcripts. For instance, CRISPRoff employs a catalytically dead fused to a repressor domain to achieve epigenetic silencing of DNA targets via , offering reversible repression without permanent mutations.31502-6) Building on ADAR's mechanism, the REPAIR system, developed in 2017, uses a catalytically inactive Cas13 fused to an evolved ADAR deaminase to enable programmable A-to-I edits in target RNAs, demonstrating up to 30% editing efficiency in cellular transcripts with minimal off-target effects. In , RNA molecules serve as programmable building blocks for regulatory circuits and nanostructures. Toehold switches, short RNA sequences that form metastable hairpins, act as riboregulators by activating translation upon binding complementary trigger RNAs, enabling logic-gated in and mammalian cells with sensitivities rivaling protein-based sensors.00864-0) RNA nanostructures, such as those created via RNA techniques, fold into complex two- and three-dimensional shapes through computational design of base-pairing motifs, achieving nanoscale assemblies stable under physiological conditions for potential use in . Aptamer evolution through the Systematic Evolution of Ligands by EXponential enrichment (SELEX) process generates high-affinity RNA ligands that bind specific targets, such as proteins or small molecules, with dissociation constants in the nanomolar range. For vaccine development, mRNA incorporates modifications like to reduce and enhance stability, as exemplified in the rapid deployment of mRNA vaccines that elicited robust immune responses in clinical trials. As of 2025, frontiers in emphasize self-assembling RNA particles for , where lipid-RNA nanoparticles encapsulate therapeutics to improve and reduce systemic toxicity in cancer therapies. Recent Cas13-based antiviral trials in 2024 demonstrated prophylactic efficacy against in animal models by degrading viral RNA , paving the way for RNA-targeted antivirals. Additionally, quantum dot-RNA sensors have emerged for detection of RNA biomarkers, leveraging energy transfer to achieve single-molecule sensitivity in diagnostic applications.

References

  1. [1]
    Biochemistry, RNA Structure - StatPearls - NCBI Bookshelf - NIH
    Jul 29, 2023 · The primary function of RNA is to create proteins via translation. RNA carries genetic information that is translated by ribosomes into various ...Introduction · Fundamentals · Cellular Level · Function
  2. [2]
    From DNA to RNA - Molecular Biology of the Cell - NCBI Bookshelf
    All of these guide RNAs are members of a large class of RNAs called small nucleolar RNAs (or snoRNAs), so named because these RNAs perform their functions in a ...
  3. [3]
    Science Snippet: RNA's Remarkable Roles
    May 18, 2022 · Transfer RNA (tRNA) works with mRNA to make sure the right amino acids are inserted into the forming protein. Ribosomal RNA (rRNA), together ...
  4. [4]
    An RNA Structure Primer
    RNA is a polymer of ribonucleoside-phosphates. It's backbone is comprised of alternating ribose and phosphate groups. Ribose is a five carbon sugar that is ...
  5. [5]
    Nitrogenous Bases - HyperPhysics Concepts
    The larger bases adenine and guanine are purines which differ in the kinds of atoms that are attached to their double ring. The other bases cytosine, uracil, ...
  6. [6]
    [PDF] Working with Molecular Genetics Chapter 2. Structures of Nucleic ...
    ribose (β-D-ribofuranose) in RNA. 2-deoxyribose (β-D-2-deoxyribofuranose in DNA). Figure 2.7. The purine or pyrimidine base is connected to the (deoxy)ribose ...
  7. [7]
    Adenosine Monophosphate | C10H14N5O7P | CID 6083 - PubChem
    Adenosine Monophosphate | C10H14N5O7P | CID 6083 - structure, chemical names, physical and chemical properties, classification, patents, literature, ...
  8. [8]
    Nucleic Acid Structure
    Primary Structure: the sequence of bases along the pentose-phosphodiester backbone of a DNA molecule (or an RNA molecule) · 5 end to the 3 end--directionality ...
  9. [9]
    Primordial soup or vinaigrette: did the RNA world evolve at acidic pH?
    While RNA is prone to alkaline hydrolysis at pH > 6 [15], the 2'-deoxy sugar of the DNA backbone is much more resistant [27]. The presence of the vicinal 2',3'- ...
  10. [10]
    The third Bond - Nature
    Feb 1, 2006 · It is a truth universally acknowledged that a guanine–cytosine (GC) base pair has three hydrogen bonds whereas adenine–thymine (AT) has two.
  11. [11]
    The RNA World and the Origins of Life - Molecular Biology of the Cell
    The other differences between RNA and DNA—the double-helical structure of DNA and the use of thymine rather than uracil—further enhance DNA stability by making ...
  12. [12]
  13. [13]
    Rules governing the orientation of the 2′-hydroxyl group in RNA
    The presence of the 2′-hydroxyl group on the RNA pentose sugar ring is at the origin of the profound structural and dynamical differences observed between RNA ...Regular Article · Abstract · Introduction<|control11|><|separator|>
  14. [14]
    The life and death of RNA across temperatures - PMC
    Integrity of RNA molecules at high temperatures​​ RNA is more susceptible to hydrolysis than DNA since the 2′OH group, present only in RNAs, can interact with ...
  15. [15]
  16. [16]
    Comparison of DNA-, PMA-, and RNA-based 16S rRNA Illumina ...
    Jul 18, 2017 · Compared to DNA, RNA degrades more rapidly in the environment. The estimated turnover time or half-lives of prokaryotic RNA is about few minutes ...
  17. [17]
    RNA Structural Dynamics As Captured by Molecular Simulations
    The 2′-OH group of the ribose moiety gives rise to profound differences between DNA and RNA. Primarily, this is because it makes RNA less chemically stable than ...
  18. [18]
    Factors Affecting Stability of RNA – Temperature, Length ...
    Comparing DNA and RNA, the latter is more prone to non-enzymatic degradation due to the presence of a 2′‑hydroxyl group.
  19. [19]
    Folding and Finding RNA Secondary Structure - PMC - NIH
    Methods are reviewed that automate folding and discovery of RNAs with algorithms that couple thermodynamics with chemical mapping, NMR, and/or sequence ...
  20. [20]
    The G·U wobble base pair: A fundamental building block of RNA ...
    The G·U wobble base pair is a fundamental unit of RNA secondary structure that is present in nearly every class of RNA from organisms of all three phylogenetic ...
  21. [21]
    [PDF] Tertiary Motifs in RNA Structure and Folding - Doudna Lab
    kissing loops, the coaxial stacking ob- served in the pseudoknot requires either. Mg2‡ ions or a high concentration of Na‡ ions for complete stabilization ...
  22. [22]
    Annotation of tertiary interactions in RNA structures reveals ...
    The seven tertiary motifs we search for are coaxial helix, A-minor, ribose zipper, pseudoknot, kissing hairpin, tRNA D-loop/T-loop, and tetraloop–tetraloop ...
  23. [23]
    Critical Role of Mg2+ Ions in RNA Folding Transitions
    Sep 2, 2025 · Magnesium ions (Mg2+) play a crucial role in stabilizing various RNA tertiary motifs, such as pseudoknots, G-quadruplexes, kissing loops, ...
  24. [24]
    ViennaRNA Package 2.0 | Algorithms for Molecular Biology | Full Text
    Nov 24, 2011 · The main secondary structure prediction tool is RNAfold, which computes the minimum free energy (MFE) and backtraces an optimal secondary ...
  25. [25]
    (PDF) Computing the Partition Function for Kinetically Trapped RNA ...
    Aug 6, 2025 · For the first time, locally optimal structures (kinetic traps in the Turner energy model) can be rapidly generated for long RNA sequences, ...<|separator|>
  26. [26]
    RNA modifications in cellular metabolism: implications for ... - Nature
    Mar 27, 2024 · In this review, we characterize the distribution features, modifying mechanisms and biological functions of 8 RNA modifications.
  27. [27]
    Biological roles of the RNA m6A modification and its implications in ...
    Nov 29, 2022 · The N6-Methyladenosine (m6A) modification of RNA transcripts is the most prevalent and abundant internal modification in eukaryotic ...
  28. [28]
    writers, erasers, readers and functions in RNA metabolism - Nature
    May 22, 2018 · This review describes the current understanding of the m 6 A modification, particularly the functions of its writers, erasers, readers in RNA metabolism.
  29. [29]
    The emerging role of RNA modifications in the regulation of mRNA ...
    Mar 24, 2020 · In this review, we discuss our current understanding of the molecular mechanisms underlying the regulation of mammalian mRNA stability by various RNA ...
  30. [30]
    Functions and therapeutic applications of pseudouridylation - Nature
    May 20, 2025 · In this Review, we discuss the emerging functions of pseudouridylation in gene regulation, focusing on how pseudouridine in mRNA, tRNA and ribosomal RNA (rRNA) ...
  31. [31]
    Detection technologies for RNA modifications - Nature
    Oct 21, 2022 · In this review, we summarize the current knowledge about these RNA modification detection technologies and discuss the challenges for the existing detection ...
  32. [32]
    DNA Transcription | Learn Science at Scitable - Nature
    In eukaryotes, RNA pol I transcribes the genes that encode most of the ribosomal RNAs (rRNAs), and RNA pol III transcribes the genes for one small rRNA, plus ...
  33. [33]
    RNA Transcription by RNA Polymerase: Prokaryotes vs Eukaryotes
    In all species, transcription begins with the binding of the RNA polymerase complex (or holoenzyme) to a special DNA sequence at the beginning of the gene known ...
  34. [34]
    Selectivity and proofreading both contribute significantly to the ... - NIH
    The expression of genetic material is also highly accurate. In E. coli cells under standard growth conditions, the error rate of transcription is 10−5.
  35. [35]
    Structure and function of ribosomal RNA - PMC - PubMed Central
    Identification of a site on 23S ribosomal RNA located at the peptidyl transferase center. ... Refined secondary structure models for the 16S and 23S ribosomal RNA ...
  36. [36]
    The Lifecycle of Ribosomal RNA in Bacteria - NCBI
    Biogenesis of rRNA in bacteria. The primary transcript comprising the three rRNAs (16S, 23S and 5S), as well as external and internal transcribed spacers (ETS ...4.1. Ribosomal Dna Promoters... · 6. Bacterial Rrna... · 9. Rrna Quality Control<|separator|>
  37. [37]
    The Ancient History of Peptidyl Transferase Center Formation as ...
    Aug 5, 2020 · The peptidyl transferase center (PTC) is the catalytic center of the ribosome and forms part of the 23S ribosomal RNA.
  38. [38]
    Systematic Analysis of the Binding Surfaces between tRNAs and ...
    Jan 7, 2018 · The secondary structure of almost all tRNA molecules is a cloverleaf fold with four arms (the acceptor arm, D-arm, anticodon arm, and TΨ-arm) ...Missing: attachment | Show results with:attachment
  39. [39]
    Aminoacyl-tRNA synthetases - PMC - PubMed Central
    The aminoacyl-tRNA synthetases are an essential and universally distributed family of enzymes that plays a critical role in protein synthesis.
  40. [40]
    [PDF] Thomas R. Cech - Nobel Lecture
    ... group II intervening sequence was self-splicing in vitro. The reaction did not require guanosine, and occurred by formation of a branched “lariat” RNA. The ...
  41. [41]
    Evolution of group II introns | Mobile DNA | Full Text - BioMed Central
    Apr 1, 2015 · Excised group II introns in yeast mitochondria are lariats and can be formed by self-splicing in vitro. Cell. 1986;44:225–34. PubMed Google ...
  42. [42]
    The discovery of a catalytic RNA within RNase P and its legacy - PMC
    Sidney Altman's discovery of the processing of one RNA by another RNA that acts like an enzyme was revolutionary in biology.
  43. [43]
    The Peptidyl Transferase Center: a Window to the Past - PMC
    The PTC is responsible for peptide bond formation during protein synthesis and is usually considered to be the oldest part of the modern ribosome.
  44. [44]
    Biosynthesis of wybutosine, a hyper-modified nucleoside in ... - NIH
    These modifications play a critical role in the stabilization of codon–anticodon pairing through base–stacking interactions and function to maintain the reading ...<|control11|><|separator|>
  45. [45]
    The Baltimore Classification of Viruses 50 Years Later
    SUMMARY. Fifty years ago, David Baltimore published a brief conceptual paper delineating the classification of viruses by the routes of genome expression.
  46. [46]
    Structural insights into RNA polymerases of negative-sense ... - Nature
    Jan 25, 2021 · In this Review, we compare recent high-resolution X-ray and cryoelectron microscopy structures of RNA polymerases of negative-sense RNA viruses.
  47. [47]
    The structure of a 12-segmented dsRNA reovirus - Research journals
    Apr 21, 2023 · We determined the structure of mud crab reovirus (MCRV), a 12-segmented dsRNA virus that is a putative member of Reovirales in the non-turreted Sedoreoviridae ...
  48. [48]
    Integration - Retroviruses - NCBI Bookshelf - NIH
    Once integrated, however, the provirus is replicated along with host-cell DNA and genetically transmitted as an integral element of the host genome. Integration ...
  49. [49]
    The not-so-infinite malleability of RNA viruses: Viral and cellular ...
    Apr 27, 2017 · RNA viruses replicate with the highest known mutation rates, which are estimated to range between 10 −6 and 10 −4 substitutions per nucleotide per cell ...
  50. [50]
    Organellar transcripts dominate the cellular mRNA pool ... - PNAS
    Despite the minimal gene content of chloroplast and mitochondrial genomes, their transcripts comprise most of the mRNA pool in leaf cells.
  51. [51]
    Why are RNA virus mutation rates so damn high? | PLOS Biology
    Aug 13, 2018 · RNA viruses have high mutation rates—up to a million times higher than their hosts—and these high rates are correlated with enhanced virulence ...<|separator|>
  52. [52]
    The Nobel Prize in Physiology or Medicine 1975 - Press release
    Karolinska institutet has decided to award the Nobel Prize in Physiology or Medicine for 1975 jointly to David Baltimore, Renato Dulbecco and Howard Temin.
  53. [53]
    P03369 · POL_HV1A2 - UniProt
    Jan 23, 2007 · This enzyme displays a DNA polymerase activity that can ... Note: Binds 2 magnesium ions for reverse transcriptase polymerase activity.
  54. [54]
    Structure of HIV-1 reverse transcriptase cleaving RNA in an ... - PNAS
    Jan 2, 2018 · HIV-1 reverse transcriptase (RT) contains both DNA polymerase and RNase H activities to convert the viral genomic RNA to dsDNA in infected host ...
  55. [55]
    Reverse Transcriptase Properties | Thermo Fisher Scientific - US
    MMLV-based reverse transcriptases are reported to have an error rate in the range of one in 15,000 to 27,000 nucleotides synthesized, with AMV reverse ...DNA polymerase activity · RNase H activity · Thermostability · Processivity
  56. [56]
    Overview of Reverse Transcription - Retroviruses - NCBI Bookshelf
    Reverse transcription begins when the viral particle enters the cytoplasm of a target cell. The viral RNA genome enters the cytoplasm as part of a ...
  57. [57]
    Understanding Retroviral Life Cycle and its Genomic RNA Packaging
    Retroviral reverse transcription of viral RNA into double stranded DNA (dsDNA) involves several steps (Figure 6; summarized in75) In the first step, a short ...
  58. [58]
    Human endogenous retroviruses in development and disease - PMC
    Nov 2, 2021 · Human endogenous retroviruses (HERVs) represent ∼8% of human genome, deriving from exogenous retroviral infections of germ line cells ...1. Introduction · 3. Hervs In Cancer · Table 1
  59. [59]
    Retrotransposons as regulators of gene expression - PMC - NIH
    Feb 12, 2016 · LINEs and SINEs are retrotransposons; that is, they transpose via an RNA intermediate. We discuss how LINEs and SINEs have expanded in ...
  60. [60]
    Links Between Repeated Sequences - PMC - NIH
    Most of these repeat sequences are derived from retrotransposons, which transpose through RNA intermediates. L1 and Alu elements are the most successful ...
  61. [61]
    Architecture of human telomerase RNA - PNAS
    Telomerase is a unique reverse transcriptase that catalyzes the addition of telomere DNA repeats onto the 3′ ends of linear chromosomes.
  62. [62]
    TERT - Telomerase reverse transcriptase - Homo sapiens (Human)
    Telomerase is a ribonucleoprotein enzyme essential for the replication of chromosome termini in most eukaryotes. Active in progenitor and cancer cells.
  63. [63]
    Telomerase RNA Component - an overview | ScienceDirect Topics
    It consists of a protein (TERT) which is a reverse transcriptase, and this RNA component (TERC) creates a template for telomere formation through insertions of ...
  64. [64]
    Double-Stranded RNA Is Produced by Positive-Strand RNA Viruses ...
    Double-stranded RNA (dsRNA) longer than 30 bp is a key activator of the innate immune response against viral infections. It is widely assumed that the ...
  65. [65]
    RNAi, a new therapeutic strategy against viral infection | Cell Research
    Dec 1, 2004 · RNA interference (RNAi) is an adaptive defense mechanism triggered by double-stranded RNA (dsRNA). It is a powerful reverse genetic tool ...
  66. [66]
    The response of mammalian cells to double-stranded RNA - PMC
    Double-stranded RNA (dsRNA) has long been recognized as a central component of the interferon (IFN) system. It was originally characterized as a key mediator of ...
  67. [67]
    Review The Biogenesis, Functions, and Challenges of Circular RNAs
    Aug 2, 2018 · Covalently closed circular RNAs (circRNAs) are produced by precursor mRNA back-splicing of exons of thousands of genes in eukaryotes.
  68. [68]
    Circular RNA biogenesis can proceed through an exon-containing ...
    Jun 9, 2015 · Direct biochemical detection of backsplicing intermediates supports the existence of an exon-containing lariat precursor. We considered two ...
  69. [69]
    Past, present, and future of circRNAs | The EMBO Journal
    Jul 25, 2019 · Exonic circular RNAs (circRNAs) are covalently closed RNA molecules generated by a process named back‐splicing. circRNAs are highly abundant ...From Past To Present--A... · The Making Of A Circrna · Circrna Properties And...
  70. [70]
    Molecular mechanisms of circular RNA translation - Nature
    Jun 14, 2024 · CircRNAs are predominantly generated through a back-splicing process within the nucleus. CircRNAs have long been considered non-coding RNAs ...
  71. [71]
    Best practice standards for circRNA research - PMC - NIH
    The inclusion of an RNase R-treated sample can help identify the band that corresponds to the circRNA. C) Direct circRNA detection using nanoString. This ...
  72. [72]
    Cell-Type Specific Features of Circular RNA Expression
    Thousands of loci in the human and mouse genomes give rise to circular RNA transcripts; at many of these loci, the predominant RNA isoform is a circle.
  73. [73]
    Circular RNAs in neurological conditions – computational ... - Nature
    Feb 17, 2025 · This review focuses on recent developments in circRNA research as they related to neuropsychiatric and neurodegenerative conditions.
  74. [74]
    Double-stranded RNA induces antiviral transcriptional response ...
    Jul 24, 2024 · Studies show that double-stranded RNAs (dsRNAs) also induce sequence-independent antiviral protection in arthropods (3, 4). Available ...
  75. [75]
    1869: DNA First Isolated - National Human Genome Research Institute
    Apr 22, 2013 · Miescher isolated a material rich in phosphorus from the cells and called it nuclein. He found nuclein in other types of cells as well, ...<|separator|>
  76. [76]
    Before Watson and Crick in 1953 Came Friedrich Miescher in 1869
    In 1869, the young Swiss biochemist Friedrich Miescher discovered the molecule we now refer to as DNA, developing techniques for its extraction.
  77. [77]
    Friedrich Miescher and the discovery of DNA - ScienceDirect.com
    Feb 15, 2005 · On February 26, 1869, he reported the discovery of this mysterious substance in a letter to Wilhelm His (Miescher, 1869a), “In my experiments ...
  78. [78]
    The discovery of the chemical nature of tobacco mosaic virus
    In their short paper, Bawden and co-operators demonstrated that TMV had a ribonucleoprotein nature, a result that was confirmed in the following years for ...Missing: RNA 1939-1944
  79. [79]
    Milestones in the research on tobacco mosaic virus - PMC - NIH
    In the molecular biological phase of research, TMV RNA was the first plant virus genome to be sequenced completely, its genes were found to be expressed by ...Missing: Bawden Pirie 1939-1944
  80. [80]
    60 years ago, Francis Crick changed the logic of biology - PMC
    Sep 18, 2017 · In September 1957, Francis Crick gave a lecture in which he outlined key ideas about gene function, in particular what he called the central dogma.Missing: 1950s | Show results with:1950s
  81. [81]
    Crick's Adaptor Hypothesis and the Discovery of Transfer RNA
    Apr 30, 2022 · This paper traces the parallel histories of Crick's adaptor hypothesis and of the experimental discovery of tRNA and compares their relative impacts on the ...
  82. [82]
    1961: mRNA Ferries Information
    Apr 26, 2013 · Sydney Brenner, Francois Jacob, and Matthew Meselson discovered that mRNA is the molecule that takes information from DNA in the nucleus to the protein-making ...Missing: T4 phage primary source
  83. [83]
    Lessons from bacteriophages part 1: Deriving utility from protein ...
    May 17, 2018 · ... T4 bacteriophage. Brenner and colleagues discovered that mRNA acts as the messenger in relaying the information from DNA to ribosomes with ...
  84. [84]
    Single Crystals of Transfer RNA: An X-Ray Diffraction Study - Science
    Single Crystals of Transfer RNA: An X-Ray Diffraction Study. Sung-Hou Kim and Alexander RichAuthors Info & Affiliations. Science. 20 Dec 1968. Vol 162, Issue ...
  85. [85]
    The Nobel Prize in Physiology or Medicine 1993 - Press release
    Roberts and Phillip A. Sharp in 1977 independently discovered that genes could be discontinuous, that is, a given gene could be present in the genetic material ...
  86. [86]
    Press release: The 1989 Nobel Prize in Chemistry - NobelPrize.org
    This year's Nobel Prize in chemistry has been awarded to Sidney Altman, USA and Thomas Cech, USA for their discovery that RNA (ribonucleic acid) in living ...
  87. [87]
    Press release: The Nobel Prize in Physiology or Medicine 2006
    Oct 2, 2006 · The discovery of RNA interference. Andrew Fire and Craig Mello were investigating how gene expression is regulated in the nematode worm ...
  88. [88]
    Synthesis of activated pyrimidine ribonucleotides in prebiotically ...
    May 14, 2009 · Powner, M. W. et al. On the prebiotic synthesis of ribonucleotides: photoanomerisation of cytosine nucleosides and nucleotides revisited.
  89. [89]
    Functional RNAs exhibit tolerance for non-heritable 2′–5′ vs. 3
    Apr 14, 2013 · However, it has been known for some time that 2′–5′ linkages destabilize RNA duplexes with respect to thermal denaturation. Here, we show that 2 ...Missing: challenges | Show results with:challenges
  90. [90]
    Catalytic promiscuity in the RNA World may have aided the ...
    Jan 26, 2021 · Here, we analyse the possibility that this “promiscous” nature of prebiotic ribozymes could have helped the maintenance of early replicator ...Missing: mimicking evidence
  91. [91]
    Identifying the wide diversity of extraterrestrial purine and pyrimidine ...
    Apr 26, 2022 · This study demonstrates that a diversity of meteoritic nucleobases could serve as building blocks of DNA and RNA on the early Earth.
  92. [92]
    AI-Driven Strategies in Prebiotic Research: Addressing Challenges ...
    Jun 17, 2025 · AI-Driven Strategies in Prebiotic Research: Addressing Challenges and Advancing Human Health. April 2025. DOI:10.1109/ICCIAA65327.2025.11013650.
  93. [93]
    Protocell Effects on RNA Folding, Function, and Evolution
    Jul 15, 2024 · Thus, these results suggested that, in a prebiotic soup containing both RNA and lipids, (66) RNAs that happened to be fortuitously encapsulated ...
  94. [94]
    Protocell Effects on RNA Folding, Function, and Evolution - PMC
    Minimal cells might therefore consist of simple vesicles enclosing a prebiotic RNA metabolism. The internal volume of a vesicle is a distinctive environment due ...
  95. [95]
    A comprehensive review of SARS-CoV-2 vaccines: Pfizer, Moderna ...
    The Pfizer and Moderna vaccines express similar mechanisms of action. The vaccine contains a nucleoside-modified mRNA that encodes the SARS-CoV-2 spike ...
  96. [96]
    Lipid nanoparticles in the development of mRNA vaccines for ...
    Jun 28, 2022 · This article focuses on the potential application of LNPs in the development and delivery of mRNA vaccines for COVID-19.
  97. [97]
    Role of nanotechnology behind the success of mRNA vaccines for ...
    It is the first time in history that two mRNA-based vaccines developed using lipid nanoparticles (LNPs) have been given emergency use authorization (EUA) by the ...
  98. [98]
    The Antisense Oligonucleotide Nusinersen for Treatment of Spinal ...
    The FDA approved nusinersen in December 2016 to treat SMA associated with SMN1 gene mutation. ... SMA as the CNS tissues are nusinersen's main site of action.
  99. [99]
    Nusinersen (Spinraza®) – Spinal Muscular Atrophy (SMA)
    Dec 3, 2024 · Nusinersen, marketed in the US as Spinraza ® (Biogen) is the first therapy approved for the treatment of SMA.
  100. [100]
    Delivery of Oligonucleotides to the Liver with GalNAc
    Indeed, patisiran, developed for the treatment of hereditary TTR amyloidosis, was the first siRNA drug to receive regulatory approval in 2018 and utilizes an ...
  101. [101]
    Small Interfering RNA (siRNA) Therapy - StatPearls - NCBI Bookshelf
    The first siRNA agent, patisiran, received US Food Drug Administration (FDA) approval in 2018. To date, the FDA has approved 6 siRNA agents: patisiran ...
  102. [102]
    The Critical Contribution of Pseudouridine to mRNA COVID-19 ... - NIH
    Aside from the delivery problem discussed above, therapeutic mRNA had at least two additional big challenges: 1) the in vitro transcribed (IVT) mRNA would be ...
  103. [103]
  104. [104]
    Current Progress and Future Perspectives of RNA-Based Cancer ...
    Jun 4, 2025 · The period from 2024 to 2025 has witnessed unprecedented clinical advances in RNA cancer vaccine development, establishing this therapeutic ...
  105. [105]
    mRNA Vaccines: Current Applications and Future Directions - PMC
    Oct 30, 2025 · Clinical trials have demonstrated that personalized mRNA cancer vaccines are safe and well‐tolerated [94, 95]. Companies like BioNTech and ...
  106. [106]
    Advances and Challenges in SARS-CoV-2 Detection: A Review of ...
    Feb 29, 2024 · Although RT-qPCR is considered the gold standard for detecting SARS-CoV-2 due to its high sensitivity and specificity, it does have some ...
  107. [107]
    An Open One-Step RT-qPCR for SARS-CoV-2 detection - PMC
    Jan 25, 2024 · Large-scale implementation of pooled RNA extraction and RT-PCR for SARS-CoV-2 detection. Clinical Microbiology and Infection. 2020;26(9): ...
  108. [108]
    RNA-Seq: a revolutionary tool for transcriptomics - PMC - NIH
    RNA-Seq is a recently developed approach to transcriptome profiling that uses deep-sequencing technologies.
  109. [109]
    Dissecting Cellular Heterogeneity Using Single-Cell RNA Sequencing
    Single-cell RNA sequencing (scRNA-seq) allows the quantitative and unbiased characterization of cellular heterogeneity by providing genome-wide molecular ...
  110. [110]
    Overview of mRNA expression profiling using DNA microarrays
    DNA microarray technology allows simultaneous measurement of the mRNA levels of thousands of genes. This powerful technology has applications in addressing ...
  111. [111]
    Improved northern blot method for enhanced detection of small RNA
    Jun 5, 2008 · Northern blot analysis involves the separation of RNA molecules by denaturing gel electrophoresis followed by transfer and cross-linking of the ...
  112. [112]
    In Situ Hybridization (ISH) - NCBI - NIH
    Nov 9, 2017 · In Situ Hybridization (ISH) is a technique that allows for precise localization of a specific segment of nucleic acid within a histologic section.
  113. [113]
    RNAscope: A Novel in Situ RNA Analysis Platform for Formalin ... - NIH
    We describe RNAscope, a novel RNA ISH technology with a unique probe design strategy that allows simultaneous signal amplification and background suppression.Materials And Methods · Rnascope Design Strategy · Single Rna Molecule...
  114. [114]
    High-definition spatial transcriptomic profiling of immune cell ...
    Jun 5, 2025 · Visium HD enables transcriptome-wide spatial gene expression analysis at single-cell scale. Gene expression is measured by the capture of ...
  115. [115]
    Interpretable spatially aware dimension reduction of spatial ... - Nature
    Oct 15, 2024 · Spatial transcriptomics produces high-dimensional gene expression measurements with spatial context. Obtaining a biologically meaningful ...
  116. [116]
    Integrating AI and RNA biomarkers in cancer - NIH
    Oct 10, 2025 · However, this study shows the interactive integration of AI and RNA biomarkers in cancer diagnostics and therapies, emphasizing advanced ...
  117. [117]
    Integrating Artificial Intelligence in Next-Generation Sequencing
    Jun 19, 2025 · The integration of AI into NGS applications is revolutionizing the landscape of genomics, epigenomics, transcriptomics, and clinical diagnostics ...3. Ai In Ngs Data Analysis · 4. Ai In Ngs Applications · 4.1. Genomics And...<|control11|><|separator|>