Fact-checked by Grok 2 weeks ago

Point mutation

A point mutation is a genetic alteration in which a single base within sequence is changed, inserted, or deleted, representing one of the smallest-scale mutations possible in the . These mutations typically arise during or due to exposure to mutagens such as ultraviolet radiation or chemicals, though cellular repair mechanisms correct many instances. While trillions occur daily across the body's cells, most point mutations are benign and have no noticeable effect on an organism's function or . Point mutations are broadly categorized into substitutions, insertions, and deletions, each with distinct molecular consequences. A substitution replaces one with another and can be further classified as silent (no change in the encoded due to codon redundancy), missense (resulting in a different that may alter or ), or nonsense (premature termination of protein synthesis by creating a ). Insertions add one or more , and deletions remove them; when not a multiple of three bases, these shift the reading frame (), often leading to a completely altered and usually nonfunctional protein downstream. The biological impact of point mutations varies widely depending on their location—such as within coding regions, regulatory sequences, or non-coding areas—and can range from neutral contributions to to pathogenic outcomes. For instance, a well-known missense substitution in the beta-hemoglobin gene causes sickle cell anemia by altering a single , leading to abnormal shape and associated health complications. In evolutionary terms, point mutations serve as a primary source of novel , driving and over generations when they confer selective advantages. point mutations in non-reproductive cells can also contribute to diseases like cancer if they activate oncogenes or inactivate tumor suppressors.

Fundamentals

Definition and Scope

A point mutation is a type of genetic mutation involving an alteration at a single position in the DNA sequence, most commonly a substitution where one nucleotide base is replaced by another. This change affects only one base pair in the double-stranded DNA molecule, which consists of four nucleotide bases: adenine (A), thymine (T), cytosine (C), and guanine (G). These bases pair specifically—A with T via two hydrogen bonds, and C with G via three hydrogen bonds—forming the rungs of the DNA double helix. While substitutions represent the typical form, the term point mutation sometimes encompasses small insertions or deletions of a single base pair, though these can shift the reading frame during protein translation. Point mutations are distinguished from larger-scale genetic alterations, such as chromosomal aberrations (e.g., deletions or duplications of extensive DNA segments) or insertions/deletions (indels) spanning multiple base pairs, which impact broader genomic regions and often lead to more severe structural changes. In contrast, point mutations are localized to one site and may result in various outcomes depending on their location and nature, including silent mutations (no change), missense mutations (altered ), nonsense mutations (premature ), or frameshift mutations (from single-base indels). These effects arise from errors in or damage but are confined without disrupting the overall chromosomal architecture. The scope of point mutations extends across all genomic contexts and organisms, occurring in both prokaryotes and eukaryotes where they serve as a primary source of genetic variation. In eukaryotic genomes, which include both coding (exons) and non-coding regions (introns, regulatory elements, and intergenic spaces), point mutations can influence protein-coding sequences or modulate if they affect non-coding functional elements. Similarly, prokaryotic genomes, lacking extensive introns, experience point mutations primarily in their compact coding and regulatory regions, contributing to adaptive in . Overall, these mutations are fundamental to , with their prevalence shaped by repair mechanisms and selective pressures in both unicellular and multicellular life forms.

Molecular Context

Point mutations occur within the context of DNA's double-helical structure, where two antiparallel strands are stabilized by hydrogen bonds between complementary base pairs: (A) with (T) via two hydrogen bonds, and (G) with (C) via three hydrogen bonds. This specific base pairing ensures the structural integrity and functional fidelity of the , as the double helix allows for accurate unwinding and separation during cellular processes. A point mutation, involving the substitution of a single , disrupts this pairing by introducing a mismatch, such as replacing an A with a G, which can lead to instability in the helix or errors in subsequent molecular interactions. During , point mutations primarily arise from errors introduced by enzymes, which synthesize new strands by adding complementary to the . In eukaryotes, replicative polymerases such as DNA polymerase δ and ε incorporate with high selectivity, but intrinsic errors occur approximately once every 10^4 to 10^5 bases before . The overall replication fidelity is enhanced by the polymerase's 3'→5' activity and post-replicative mismatch repair, achieving an error rate of approximately 10^{-9} to 10^{-10} mutations per per . In the transcription process, point mutations within protein-coding genes alter the DNA template, leading to corresponding changes in the synthesized messenger RNA (mRNA) sequence, which can affect splicing, stability, or translation into proteins. These genomic mutations are distinct from RNA editing events, which involve post-transcriptional modifications to the mRNA itself, such as base conversions by enzymes like ADAR, without altering the underlying DNA. Point mutations can occur at various chromosomal locations within genes, including exons (coding regions that are retained in mature mRNA) and introns (non-coding intervening sequences removed during splicing), as well as in regulatory regions such as promoters upstream of the transcription start site. Mutations in exons directly impact the protein-coding sequence, while those in introns may disrupt splice sites, and alterations in promoters can impair transcription initiation by affecting binding or regulatory factor recruitment.

Causes

Spontaneous Causes

Spontaneous point mutations arise from intrinsic biochemical processes within the , independent of external agents, and represent a fundamental source of . These mutations occur at low but measurable rates during DNA maintenance and replication, primarily due to the chemical instability of DNA bases and the inherent limitations of enzymatic . Although most such errors are corrected by cellular repair mechanisms, those that persist can lead to base substitutions or small insertions/deletions. Key processes include hydrolytic reactions, tautomeric shifts, polymerase inaccuracies, and oxidative damage from metabolic byproducts. Depurination involves the spontaneous hydrolysis of the N-glycosidic bond linking a purine base (adenine or guanine) to the deoxyribose sugar in the DNA backbone, resulting in an apurinic (AP) site. This occurs at a rate of approximately 10,000 to 20,000 events per mammalian cell per day under physiological conditions, primarily driven by water molecules acting as nucleophiles. During subsequent DNA replication, the AP site can cause transversion mutations if the polymerase inserts an adenine opposite the vacancy, leading to a purine-to-pyrimidine substitution in the daughter strand. The chemical reaction can be represented as: \ce{N-glycosidic\ bond\ hydrolysis:\ Purine-DNA + H2O -> AP\ site + Purine} Deamination, another hydrolytic process, entails the removal of an amino group from a base, most commonly cytosine, converting it to uracil. This reaction proceeds via nucleophilic attack by water on the C4 amino group, yielding uracil and ammonia at a rate of about 100 to 500 cytosine residues per human cell per day. If unrepaired, uracil pairs with adenine during replication, resulting in a C-to-T transition mutation. The process is depicted as: \ce{C + H2O -> U + NH3} Less frequent deaminations affect adenine (to hypoxanthine, pairing with cytosine) and guanine (to xanthine, which still pairs with cytosine but may stall replication), but cytosine deamination predominates due to its higher susceptibility. Tautomerization refers to the reversible isomerization of DNA bases between their common keto or amino forms and rare enol or imino forms, facilitated by proton shifts under physiological conditions. This transient shift alters hydrogen-bonding patterns, promoting non-standard base pairing during DNA replication. For instance, the enol tautomer of thymine can form two hydrogen bonds with guanine instead of adenine, potentially causing a T-to-C transition if the error persists. Such events are rare, occurring at frequencies around 10^{-4} to 10^{-5} per base pair, but contribute to spontaneous mutagenesis without external triggers. Replication errors stem from the intrinsic infidelity of s, which occasionally insert incorrect due to base slippage or wobble pairing, particularly in repetitive sequences. High-fidelity polymerases like δ and ε exhibit base insertion error rates of about 10^{-5} to 10^{-7} per , exacerbated by slippage in microsatellites where the nascent strand temporarily dissociates and realigns, leading to small indels. by the polymerase's 3'→5' activity enhances fidelity by 100- to 1,000-fold, excising mismatched bases, yet deficiencies or overwhelming error loads allow some mismatches to evade correction, contributing to point mutations at an overall rate of approximately 10^{-9} to 10^{-10} per . Endogenous reactive oxygen species (ROS), generated as byproducts of cellular metabolism such as mitochondrial respiration, induce oxidative lesions in DNA that manifest as point mutations. Superoxide radicals, hydrogen peroxide, and hydroxyl radicals attack guanine preferentially, forming 8-oxoguanine (8-oxoG), one of the most abundant oxidative adducts, at rates estimated at 100 to 1,000 lesions per human cell per day. During replication, 8-oxoG mispairs with adenine, yielding G-to-T transversions if not repaired by base excision repair pathways. This process underscores how normal metabolic activity inadvertently promotes genomic instability.

Induced Causes

Induced point mutations arise from exposure to external agents that chemically or physically alter DNA bases, leading to errors during replication or repair. These mutagens include chemicals and , which can be encountered environmentally or applied deliberately in laboratory settings. Unlike spontaneous mutations from endogenous processes, induced ones often result from deliberate chemical modifications or energy deposition that targets specific DNA components. Chemical mutagens are among the most studied inducers of point mutations, primarily through or base mimicry. Alkylating agents, such as (EMS), react with bases to form O6-alkylguanine adducts, which mispair with during replication, predominantly causing G-to-A transitions. EMS is highly effective due to its ability to alkylate DNA at multiple sites, resulting in a high of up to 10^{-3} per locus in treated organisms. Another class, base analogs like 5-bromouracil (5-BU), incorporates into DNA in place of but exists in a tautomeric form that pairs with , leading to A-T to G-C transitions. The mutagenic potential of 5-BU stems from its shifted enol-keto equilibrium, increasing mispairing frequency compared to natural bases. Radiation exposure also induces point mutations by damaging DNA bases or generating reactive species. (UV) light, particularly UVB wavelengths, forms cyclobutane (CPDs) between adjacent or bases, which, if unrepaired or processed via error-prone translesion synthesis, result in C-to-T or CC-to-TT substitutions—commonly known as UV signature mutations. , such as X-rays or gamma rays, produces that cause oxidative base modifications, including , which mispairs with to yield G-to-T transversions, alongside direct strand breaks that can lead to base substitutions during repair. These effects are dose-dependent, with mutation frequencies increasing linearly with exposure levels across cell types. In experimental contexts, induced point mutations are harnessed for genetic studies using model organisms. EMS mutagenesis, for instance, is widely applied in forward and reverse genetics screens in species like Arabidopsis thaliana, Caenorhabditis elegans, and rice, where soaked seeds or larvae are treated to generate libraries of mutants with random point mutations for phenotypic analysis. This approach has facilitated the identification of thousands of genes involved in development and stress responses, with mutation rates tailored by EMS concentration (typically 0.1-1% solutions). Such techniques enable high-throughput sequencing to map induced variants, contrasting with natural mutation rates by orders of magnitude. Environmental exposures contribute to induced point mutations through chronic low-level contact with mutagens. Cigarette smoke contains polycyclic aromatic hydrocarbons and nitrosamines that act as alkylating agents, inducing G-to-T transversions in genes like TP53 and KRAS, which are hallmarks of smoking-related lung cancers. Similarly, industrial pollutants such as benzene derivatives function as alkylators, elevating point mutation rates in exposed populations via base alkylation akin to EMS. These agents often bypass standard repair mechanisms, amplifying mutation accumulation over time.

Classification

Substitution Types

Point mutations involving substitutions replace one nucleotide base with another without altering the DNA sequence length. These substitutions are categorized into two main types based on the chemical properties of the bases involved: transitions and transversions. Transitions occur when a base is substituted for another purine ( [A] to [G] or vice versa) or a base for another (cytosine [C] to [T] or vice versa). These changes involve bases with similar chemical structures and shapes, which facilitates tautomeric shifts during replication and contributes to their higher occurrence compared to other substitutions. A common example is the C-to-T transition resulting from the spontaneous of cytosine to uracil, which is often not repaired and leads to a mismatch during replication. Another frequent transition is G-to-A, particularly at hotspots in CpG dinucleotides where cytosine increases deamination rates, effectively yielding this substitution on the complementary strand. Transversions, in contrast, involve the substitution of a for a or vice versa, such as A-to-C, A-to-T, G-to-C, or G-to-T. These exchanges occur between bases with dissimilar shapes and chemical properties, making them less likely during normal replication errors and often associated with more severe DNA damage from external agents. In many genomes, including the , transitions outnumber transversions, with a typical of approximately 2:1. This arises from both mutational processes and selective pressures, influencing the overall pattern of by favoring certain synonymous changes in coding regions.

Functional Classifications

Point mutations are functionally classified based on their effects on and protein function, primarily arising from substitutions in or regulatory regions. This includes silent, missense, and mutations within exons, which influence the protein sequence through changes in codons, as well as regulatory mutations that alter non-coding elements like promoters and splice sites. These categories highlight how a single change can range from to severely disruptive, depending on the genetic code's degeneracy and the mutation's location. Silent mutations occur when a nucleotide substitution changes a codon but does not alter the encoded amino acid, due to the degeneracy of the genetic code where multiple codons specify the same amino acid. For example, changing CGU to CGC both code for arginine, resulting in no change to the protein sequence. Such mutations are typically neutral in terms of protein function, though they may subtly affect translation efficiency in some contexts. Missense mutations involve a nucleotide change that results in a codon specifying a different , leading to a single substitution in the protein. An illustrative case is the substitution of CGU () to CAU (), which alters the protein's chemical properties. A well-known example is the GAG to GTG change in the beta-globin gene, replacing with and causing sickle cell anemia. Nonsense mutations convert a codon for an into a premature (UAA, UAG, or UGA), truncating the protein and often rendering it nonfunctional. For instance, CAG () to TAG (stop) at codon 161 in the low-density lipoprotein receptor gene leads to a shortened protein. The impact depends on the position, with early stops causing more severe loss of function. Regulatory mutations affect non-coding regions, such as promoters that influence transcription initiation or sites that direct pre-mRNA processing, thereby altering levels or mRNA isoform production without changing the protein sequence. Examples include mutations in donor sites, like c.1845+1G>A in the , which disrupts recognition and causes in type 1. Promoter variants can similarly reduce transcription rates by impairing binding. To illustrate how substitutions map to these functional classes, consider the following examples from the standard genetic code, which exhibits degeneracy primarily at the third codon position:
Original CodonMutationNew CodonAmino Acid ChangeFunctional ClassExample Amino Acid
CGUU to CCGCNoneSilentArginine to Arginine
CGUG to ACAUArg to HisMissenseArginine to Histidine
CAGC to TTAGGln to StopNonsenseGlutamine to Stop
GAG (beta-globin)A to TGTGGlu to ValMissenseGlutamic acid to Valine
This table references key positions where third-position changes often yield silent outcomes due to wobble pairing, while first- or second-position alterations typically cause missense or nonsense effects.

Small Indels as Point Mutations

Small insertions or deletions (indels) of a single nucleotide are considered a type of point mutation, distinct from larger structural variants, and they characteristically produce frameshift mutations by altering the reading frame of the downstream genetic sequence in protein-coding regions. These frameshifts occur because the genetic code is read in triplets (codons), and the addition or removal of one base disrupts this grouping, leading to a cascade of incorrect amino acid incorporations during translation. In contrast to base substitutions, which involve swapping one nucleotide for another without changing the overall length of the DNA sequence, small indels modify the sequence length itself, often resulting in a higher mutagenic potential due to the extensive alteration of the protein product. The mechanism of small indels typically involves errors during , such as polymerase slippage, where the enzyme temporarily dissociates and reassociates, adding or omitting a single . For example, an insertion might add an extra (A) within a codon, shifting the so that subsequent bases are grouped differently—e.g., the sequence ATG-CGT-AAA becoming ATG-ACG-TAA-A..., producing a garbled chain and potentially introducing a premature . Deletions operate similarly by removing one base, compressing the and altering all downstream codons. These changes often lead to nonsense-like effects, where the protein is truncated or nonfunctional, akin to nonsense mutations but through frame disruption rather than a direct creation. Small indels frequently arise from replication slippage, particularly in repetitive sequences. Representative examples illustrate their impact: a single adenine insertion (+A) at codon 18 of the β-globin (HBB) causes a frameshift, generating a premature and resulting in β-thalassemia major due to absent functional . Similarly, a single-base deletion in the leads to a frameshift in Tay-Sachs disease, causing premature termination of the α-subunit of β-hexosaminidase A and accumulation of GM2 gangliosides. Such mutations are common in repetitive sequences like microsatellites, where they can expand or contract during replication. In terms of frequency, small s constitute the second most common form of after base substitutions across genomes, but they occur at lower rates overall—approximately 15-25% as frequent as substitutions. However, their prevalence rises significantly in microsatellites, where slippage mechanisms amplify indel rates, making them a key driver of in those loci.

Effects

Biochemical Consequences

Point mutations, which can involve a single nucleotide substitution, insertion, or deletion in the DNA sequence, often arise during replication when a polymerase incorporates an incorrect base, creating a mismatch between the template strand and the nascent strand. If not corrected by DNA mismatch repair mechanisms, this mismatch becomes permanently incorporated after the next round of replication, potentially altering the genetic code. Such mismatches can impede replication fork progression, causing transient stalling of DNA polymerase as it struggles to extend from the mismatched base, which may activate downstream repair or recombination pathways to resume synthesis. At the structural level, point mutations can influence DNA duplex stability by altering base-pairing strength; for instance, transitions from G-C to A-T pairs reduce the number of hydrogen bonds from three to two, lowering the melting temperature and overall thermodynamic stability of the . When a point mutation occurs in a , it alters the mRNA sequence during transcription, changing one or more codons and thereby affecting the read by ribosomes. For example, a might shift a codon from encoding a hydrophobic like (GUU) to a hydrophilic one like serine (UCU), disrupting the mRNA's role in specifying protein composition. Single-base insertions or deletions cause frameshift mutations, altering all downstream codons and typically leading to a garbled amino acid sequence with a high likelihood of premature termination. mutations introduce premature stop codons (e.g., UAG), leading to truncated transcripts that terminate early and produce incomplete polypeptides, while silent mutations may subtly influence mRNA secondary structure or splicing efficiency, indirectly impacting rates. At the protein level, substitutions resulting from point mutations frequently cause structural perturbations, such as misfolding due to changes in hydrophobicity; replacing a buried hydrophobic residue with a hydrophilic one can expose nonpolar cores to solvent, destabilizing the native fold and promoting aggregation. Frameshift mutations generally produce nonfunctional proteins due to the altered . These alterations often lead to loss-of-function, where the protein loses enzymatic activity or binding affinity, or gain-of-function, where novel interactions emerge, such as enhanced or aberrant signaling. Misfolded proteins accumulating in the trigger cellular stress responses, notably the unfolded protein response (UPR), which activates sensors like IRE1, PERK, and ATF6 to halt , upregulate chaperones, and enhance pathways to restore . Persistent activation of UPR from -induced misfolding can shift from adaptive to pro-apoptotic if unresolved, though this is mitigated by repair if the is detected early. Point mutation rates in eukaryotic genomes typically range from 10^{-9} to 10^{-8} per per , reflecting the balance of replication fidelity and efficiency. In , the probability of fixation for a beneficial point mutation with small positive s approximates $2s in large populations, as derived from diffusion models, while deleterious mutations have near-zero fixation probability unless under .

Phenotypic Outcomes

Point mutations can result in neutral phenotypic effects when they occur in non-coding regions or as silent mutations that do not alter the sequence of proteins, thereby contributing to without impacting organismal . These neutral changes accumulate through and serve as a substrate for future evolutionary processes, maintaining polymorphism in populations. Deleterious point mutations often reduce by inactivating essential enzymes or disrupting protein function, leading to phenotypes such as decreased metabolic efficiency or in homozygous individuals. For instance, a single substitution can abolish catalytic activity in enzymes critical for cellular processes, resulting in impaired growth or viability. Beneficial point mutations confer adaptive advantages, such as enhanced survival under selective pressures; a notable example is the single base change in the TEM-1 β-lactamase enzyme that increases bacterial resistance to antibiotics by improving substrate . These mutations can rapidly spread in populations exposed to antibiotics, demonstrating how point changes drive adaptive in microbes. In evolutionary terms, point mutations are primary drivers of , with the neutral theory positing that the majority are selectively neutral and fixed primarily by rather than . Proposed by Kimura, this framework explains the observed of neutral substitutions accumulating at a constant rate across lineages. Within , point mutations introduce new whose frequencies shift via drift, , or selection, influencing evolutionary trajectories. In cases of , such as the sickle-cell providing resistance to carriers, these mutations maintain balanced polymorphisms, stabilizing frequencies despite homozygous disadvantages.

Role in Diseases

Cancer-Associated Mutations

Point mutations play a central role in oncogenesis by altering key regulatory genes, primarily through changes acquired during tumorigenesis rather than inherited variants. These mutations are detected via tumor sequencing and are distinct from mutations, which are present in all cells and contribute to hereditary cancer syndromes. In cancer genomes, point mutations drive the transformation of normal cells into malignant ones by activating oncogenes or inactivating tumor suppressors, often accumulating over multiple steps in . Activating point mutations in oncogenes, such as those in the family, promote uncontrolled . A prominent example is the G12D missense mutation, which substitutes with at codon 12 (changing GGT to GAT), locking the protein in an active GTP-bound state and hyperactivating downstream signaling. This mutation occurs in over 40% of pancreatic ductal adenocarcinomas (PDAC), where it initiates and sustains tumor growth. Similarly, mutations in other isoforms contribute to oncogenesis across various cancers, with alterations present in approximately 30% of human tumors. In tumor suppressor genes like TP53, inactivating point mutations disrupt DNA damage response and , allowing genomic instability to propagate. mutations in TP53 introduce premature stop codons, leading to truncated, nonfunctional proteins, while missense mutations at hotspots such as R175 and R248 alter the , abolishing transcriptional activity. These hotspots, including R175H and R248Q, are recurrent in diverse cancers and compromise p53's guardian function, with TP53 mutations identified in over 50% of tumors cataloged in the COSMIC database. Alterations in the RAS-MAPK signaling pathway exemplify how point mutations converge to drive multistep . Activating mutations aberrantly stimulate the MAPK/ERK cascade, promoting cell survival and proliferation independent of growth factors. This pathway's deregulation, often via somatic point mutations in or upstream regulators, is a hallmark of many cancers and facilitates the sequential accumulation of additional mutations required for full malignancy. According to the Catalogue of Somatic Mutations in Cancer (COSMIC), point mutations in driver genes like and TP53 are implicated in the majority of tumors, with over 90% of cancer cases harboring at least one such alteration in key pathways.

Inherited Disorders

Point mutations in the can lead to monogenic inherited disorders by altering protein function in a heritable manner, often following patterns. These mutations are transmitted from parents to offspring and manifest in affected individuals, contrasting with somatic mutations that are not inherited. In autosomal dominant disorders, a single heterozygous point mutation suffices to cause disease due to or dominant-negative effects. type 1 (NF1), for instance, arises from nonsense mutations in the NF1 gene on chromosome 17q11.2, such as the R1947X variant, which introduces a premature and produces a truncated neurofibromin protein, reducing its tumor suppressor activity. This results in , leading to symptoms like café-au-lait spots, neurofibromas, and learning disabilities; the disorder affects approximately 1 in 3,000 individuals worldwide and exhibits nearly complete but variable expressivity influenced by modifier genes. Autosomal recessive disorders require biallelic mutations, with carriers typically asymptomatic. Sickle-cell anemia exemplifies this, caused by the homozygous HBB Glu6Val missense mutation (also known as HbS) in the beta-globin gene on chromosome 11, substituting glutamic acid with valine at position 6 and promoting hemoglobin polymerization under low oxygen, leading to red blood cell sickling, vaso-occlusive crises, and chronic hemolysis. The carrier frequency reaches about 1 in 10 among African Americans, reflecting heterozygote advantage against malaria in endemic regions. Tay-Sachs disease similarly follows autosomal recessive inheritance, with point mutations in the HEXA gene on chromosome 15 causing hexosaminidase A deficiency and GM2 ganglioside accumulation in neurons; common variants include the 4-bp insertion (c.1274_1277dupTATC) in exon 11 that shifts the reading frame and the G-to-C transversion at the splice site (c.1421+1G>C). Carrier screening has reduced incidence in high-risk Ashkenazi Jewish populations, where the carrier frequency is about 1 in 27. X-linked recessive disorders predominantly affect males, with females as carriers. Hemophilia A results from point mutations in the F8 gene on , encoding coagulation factor VIII; examples include nonsense mutations like R2307X, which truncate the protein and abolish its procoagulant function, causing severe bleeding tendencies. This hotspot at CpG sites leads to a of about 1 in 5,000 males globally. Even identical point mutations can exhibit incomplete penetrance, where not all carriers develop symptoms, or variable expressivity, where phenotypes range from mild to severe due to genetic modifiers, epigenetic factors, or environmental influences. In NF1, monozygotic twins with the same NF1 mutation show differing tumor burdens and cognitive impacts, highlighting expressivity variation. Similarly, in sickle-cell , the Glu6Val mutation's severity varies with levels modulated by BCL11A variants. and carrier screening, such as newborn testing for sickle-cell trait, enable early intervention and in at-risk populations.

Repeat-Induced Point Mutation

Mechanism

Repeat-induced point mutation (RIP) is a genome defense mechanism that operates during the premeiotic stage of the sexual cycle in certain fungi, particularly , where it targets duplicated DNA sequences to introduce mutations that inactivate repetitive elements such as transposons. This process occurs in haploid nuclei shortly after fertilization but before and , specifically during or immediately following premeiotic , allowing detection of homologous sequences in the paired nuclei. RIP induces a high frequency of C-to-T (or G-to-A on the complementary strand) transitions, often resulting in the loss of up to 50% of G·C base pairs in targeted duplicates over successive generations. These mutations are densely clustered and strand-specific, effectively mutating both copies of the duplicated sequence in a bilateral manner to prevent proliferation of . The targeting of RIP is highly specific to duplicated or repetitive sequences, requiring a minimum homology length of approximately 400 base pairs with at least 80% identity, though activity can extend to shorter repeats (down to ~150 bp) at reduced efficiency. The process scans the genome for such duplications regardless of their chromosomal location—whether linked or unlinked—and applies mutagenesis symmetrically to both homologs, ensuring comprehensive inactivation without relying on recombination. This homology-dependent recognition is thought to involve direct comparison of intact double-stranded DNA molecules during the premeiotic phase, distinguishing RIP from random mutational processes. At the enzymatic level, RIP begins with the recognition and of in the duplicated regions, primarily mediated by the RID (RIP-defective) protein, a cytosine methyltransferase homologue, in conjunction with DIM-2, another methyltransferase responsible for formation. Recent studies as of have further confirmed RID's essential role in both RIP mutagenesis and associated inactivation processes. This leads to dense, asymmetric (5mC) marks on both strands of the repeats. Subsequent of these methylated to generates the characteristic C-to-T transitions, a mechanism inferred from the and supported by the near-exclusive production of A·T pairs from original G·C sites. While the specific deaminase remains unidentified, a sexual-stage-specific deaminase has been proposed based on the process. RIP exhibits strong sequence specificity, with mutations preferentially occurring at cytosines in a 5'-CpA-3' (or 5'-TA-3' on the complementary strand) dinucleotide context, accounting for the majority of changes and contributing to the AT-rich degeneration of targeted sequences. This bias amplifies the mutagenic effect, as repeated rounds of can further mutate the already AT-biased products, leading to progressive silencing through associated and assembly.

Evolutionary and Biological Roles

Repeat-induced point mutation (RIP) functions primarily as a genome defense mechanism in fungi, targeting repetitive DNA sequences to suppress the proliferation of transposable elements (TEs) and prevent the spread of selfish genetic elements that could destabilize the genome. By inducing C-to-T transitions in duplicated sequences during the premeiotic stage, RIP hypermutates these elements, rendering them non-functional and limiting their accumulation, as observed in species like Neurospora crassa where it effectively curbs TE copy number increases. This process acts as a homology-dependent safeguard, diversifying repetitive sequences and reducing the risk of ectopic recombination, thereby maintaining genomic integrity against invasive DNA. In terms of evolutionary impact, RIP contributes to fungal architecture by generating AT-rich islands from mutated repeats, which are non-coding regions enriched in adenine and bases due to the bias toward A/T mutations. This mechanism promotes genome shrinkage, as RIP-active fungal lineages exhibit significantly smaller s compared to those lacking it; for instance, the loss of RIP-associated genes correlates with a 30-fold increase in genome size in certain leotiomycetes lineages approximately 120 million years ago. Furthermore, RIP influences by compartmentalizing genomic regions, altering gene density, and restricting TE-driven expansions, thereby shaping evolutionary trajectories in fungi. The biological consequences of RIP extend to nearby genes and duplicated sequences, where it can mutate loci in close proximity to repeats, potentially inactivating redundant copies and facilitating subfunctionalization—the evolutionary process by which gene duplicates partition ancestral functions. In fungal genomes, this reduces the number of paralogous genes, hindering broad gene duplication events that might otherwise promote evolutionary novelty, as evidenced by lower paralog retention rates in RIP-proficient species. Such targeted mutagenesis thus balances genome stability with adaptive evolution by selectively disabling superfluous elements. Comparatively, is predominantly found in Ascomycetes, such as and Magnaporthe, with bioinformatic signatures indicating activity in some Basidiomycetes, but it is absent in other eukaryotic kingdoms like and animals, which rely on alternative defenses. This fungal-specific trait parallels other silencing mechanisms, including (RNAi), in recognizing homologous sequences and promoting formation, though RIP uniquely employs direct mutagenesis rather than RNA-guided cleavage. The enrichment of RIP-like patterns in underscores its role in phylum-specific evolution, distinct from broader eukaryotic silencing pathways. Over the long term, RIP-mutated sequences often evolve into non-functional pseudogenes, as the accumulation of mutations erodes their coding potential, contributing to a compacted, streamlined fungal with reduced genetic redundancy. This process reinforces genome defense by permanently silencing once-repetitive elements, while also influencing broader evolutionary patterns such as decreased in TE-impacted regions. In like Podospora anserina, these pseudogenes represent relics of past RIP events, highlighting the mechanism's enduring impact on fungal biology.

Laboratory Applications

In the fungus , repeat-induced point mutation (RIP) serves as a targeted tool for inactivation, where introducing a duplicate copy of a via triggers extensive C-to-T during the sexual cycle, effectively knocking out the endogenous and facilitating mutant screens for functional studies. This approach, pioneered as one of the earliest methods for in Neurospora, enables high-throughput identification of with phenotypes such as altered growth or pigmentation, though it often results in multiple per , limiting precision for subtle changes.

Historical Context

Early Discoveries

The discovery of point mutations began with observations of spontaneous changes in fruit flies () in the early 1910s, which helped distinguish small-scale genetic alterations from larger chromosomal rearrangements. In 1910, identified the first sex-linked recessive mutation, the white-eyed trait, demonstrating inheritance patterns consistent with a localized gene change rather than gross chromosomal abnormalities. Calvin Bridges extended this work in the 1910s through studies of using white-eyed flies, providing cytological evidence that such mutations affected specific loci on chromosomes without disrupting their overall structure, thus laying the groundwork for separating point-like gene mutations from chromosomal mutations like deletions or duplications. A pivotal advance came in 1927 when Hermann J. Muller demonstrated that mutations could be artificially induced, proving their physical basis. By exposing sperm to X-rays, Muller observed a dramatic increase in lethal and visible mutations—about 150 times higher than spontaneous rates—many of which were heritable changes at specific gene loci, such as eye color or wing shape, without accompanying chromosomal breaks. This experiment established that mutations result from physical alterations to genes, shifting the view from vague hereditary variations to tangible, inducible events. In 1941, George W. Beadle and Edward L. Tatum built on these foundations by using X-ray-induced mutants in the fungus to propose the "one gene-one enzyme" hypothesis. They isolated auxotrophic mutants, such as those unable to synthesize (e.g., arg-1 requiring ), showing that each mutation disrupted a single enzymatic step in a biochemical pathway, linking specific genes to discrete biochemical functions. This work emphasized point mutations as precise disruptions within genes. The spontaneous nature of point mutations was further clarified in 1943 by Salvador E. Luria and through their fluctuation test in bacteria. By growing parallel cultures of and exposing them to bacteriophage T1, they found high variance in resistant mutants across cultures—indicating mutations arose randomly before selection—rather than uniformly in response to the virus, with jackpot events from early mutations amplifying clones. This confirmed pre-adaptive, spontaneous point mutations as a general mechanism across organisms.

Key Developments and Milestones

One of the earliest identifications of a point mutation at the protein level occurred in 1957, when Vernon Ingram demonstrated that sickle-cell anemia results from a single in the beta-globin chain of , replacing with at position 6 due to a to codon change. This discovery provided the first direct evidence of how a could alter and function, marking a pivotal step in linking to . The foundational understanding of point mutations was advanced by the 1953 elucidation of DNA's double-helix structure by and , which explained the mechanisms of base substitutions during replication and their potential to cause heritable changes without disrupting the overall DNA framework. This model highlighted how a single replacement could lead to mismatched base pairing and propagate errors, laying the groundwork for subsequent research. In the 1960s, the cracking of the by Marshall Nirenberg and revealed the triplet nature of codons and demonstrated how point mutations—such as transitions or transversions—could result in missense, , or silent effects by altering specific specifications. Nirenberg's 1961 cell-free experiment using synthetic polynucleotides identified the first codon assignments, while Khorana's synthesis of repeating copolymers in the mid-1960s enabled decoding of all 64 codons, showing precisely how single-base changes disrupt . These breakthroughs, recognized with the 1968 in Physiology or Medicine, transformed the study of point mutations from phenomenological observations to predictable biochemical events. The advent of DNA sequencing in 1977 by Frederick Sanger and colleagues introduced chain-termination methods that allowed direct detection of point mutations by resolving nucleotide sequences up to several hundred bases long, revolutionizing mutation analysis beyond protein-level inferences. This technique, which earned Sanger a second Nobel Prize, enabled precise identification of base substitutions in genes, facilitating studies of mutational spectra and evolutionary rates. In the 1980s, Eric Selker's work in Neurospora crassa uncovered repeat-induced point mutation (RIP), a premeiotic process that systematically mutates duplicated DNA sequences through extensive C-to-T transitions, linking point mutations to epigenetic silencing and genome defense mechanisms. Selker's 1987 demonstration that RIP generates A/T-rich sequences prone to DNA methylation provided insights into how organisms suppress repetitive elements, influencing later research on transposable element evolution. The 2000s brought next-generation sequencing (NGS) technologies, starting with 454 in 2005, which enabled high-throughput profiling of point mutations across entire genomes at unprecedented scale and speed compared to Sanger methods. By the late 2000s, platforms like Illumina's sequencing-by-synthesis allowed simultaneous detection of millions of variants, transforming point mutation studies in cancer genomics and by revealing and landscapes.

References

  1. [1]
    Point Mutation - National Human Genome Research Institute
    A point mutation occurs in a genome when a single base pair is added, deleted or changed. While most point mutations are benign, they can also have various ...
  2. [2]
    Mutation, Repair and Recombination - Genomes - NCBI Bookshelf
    Point mutations are also called simple mutations or single-site mutations. They are sometimes described as substitution mutations but this risks confusion ...
  3. [3]
    Types of mutations - Understanding Evolution
    There are many different ways that DNA can be changed, resulting in different types of mutation. Here is a quick summary of a few of these.
  4. [4]
    Point Mutation - MeSH - NCBI - NIH
    Point Mutation. A mutation caused by the substitution of one nucleotide for another. This results in the DNA molecule having a change in a single base pair.
  5. [5]
    The Structure and Function of DNA - Molecular Biology of the Cell
    Each molecule of DNA is a double helix formed from two complementary strands of nucleotides held together by hydrogen bonds between G-C and A-T base pairs.
  6. [6]
    Point Mutation - an overview | ScienceDirect Topics
    Point mutation refers to a single-nucleotide change in a gene as a consequence of base substitution, insertion, or deletion.
  7. [7]
    Genetics, DNA Damage and Repair - StatPearls - NCBI Bookshelf
    One must first make the distinction between large-scale mutations, where a whole chunk of a chromosome is lost, relocated, or rearranged, vs. point mutation.
  8. [8]
  9. [9]
    How the Muta... - Hello. Pleas... | Learn Science at Scitable - Nature
    Sometimes, point mutations have little or no effect. This can be true if they occur in noncoding regions. Even if a point mutation occurs within the coding ...
  10. [10]
    Studying Mutation and Its Role in the Evolution of Bacteria - PMC - NIH
    2012). Prokaryotes that tend to have less coding sequences in total, provide a smaller target for the origin of deleterious mutations than eukaryotic genomes.
  11. [11]
  12. [12]
    Biochemistry, Mutation - StatPearls - NCBI Bookshelf - NIH
    Point mutations have different consequences depending on which codon and what part of it they affect.[13] Silent mutations can occur when a point mutation ...
  13. [13]
    DNA Replication Fidelity and Cancer - PMC - NIH
    Determinants of DNA replication fidelity. Normal cells replicate their DNA with extraordinary fidelity (~10−10 mutations per base pair per cell division) [52].
  14. [14]
    DNA polymerase ε and δ proofreading suppress discrete mutator ...
    Oct 6, 2009 · ... rate of ≈10−10 per base pair per cell division (1). Eukaryotes have 2 nuclear DNA polymerases with intrinsic proofreading activity: Pol ε ...Dna Polymerase ε And δ... · Sign Up For Pnas Alerts · Results And Discussion<|separator|>
  15. [15]
    Site-directed RNA editing: recent advances and open challenges
    Up to 60% of all genetic disorders are caused by point mutations [1,2]. These can affect transcript stability, translation, or RNA-splicing. Moreover, in ...
  16. [16]
    When you can't trust the DNA: RNA editing changes transcript ...
    RNA editing describes targeted sequence alterations in RNAs so that the transcript sequences differ from their DNA template.<|separator|>
  17. [17]
    Differential Gene Transcription - Developmental Biology - NCBI - NIH
    Anatomy of the gene: Exons and introns. There are two fundamental differences distinguishing most eukaryotic genes from most prokaryotic genes.
  18. [18]
    Gene promoters show chromosome-specificity and ... - BMC Genomics
    Apr 24, 2013 · A chromatin-dependent distribution of point mutations can lead to a gradual shift in gene expression. Gene promoters located mainly inside ...
  19. [19]
    The genome-wide rate and spectrum of EMS-induced heritable ...
    Oct 19, 2021 · As an alkylating agent, EMS induces chemical modifications of nucleotides. It was first demonstrated by Brookes and Lawley (1961) that EMS ...
  20. [20]
    Mechanisms of UV-induced mutations and skin cancer - PMC - NIH
    UV radiation causes DNA damage, especially cyclobutane pyrimidine dimers (CPDs), leading to mutations and skin cancer. CPDs are slow to repair, and deamination ...Uv-Induced Dna Damage... · Mechanisms Of Uv Mutagenesis · Mutations In Skin Cancer...
  21. [21]
    Radiation-induced point mutations, deletions and micronuclei in lacI ...
    Jun 1, 1994 · Ionizing radiation induces gene mutations (point mutations, deletions and insertions) as well as chromosome damage in mammalian cells.
  22. [22]
    Quantitative and qualitative mutational impact of ionizing radiation ...
    Feb 14, 2024 · The study reveals mutational rates proportional to IR dose and consistent across cell types, underscoring IR's universal mutagenic impact on ...
  23. [23]
    Current trends and insights on EMS mutagenesis application to ...
    Treatment with EMS is inexpensive, easy to implement, and induces point mutations at high rates and with good agreement in most species with different genetic ...
  24. [24]
    Next-generation sequencing for identification of EMS-induced ... - NIH
    Forward genetic analysis using chemical mutagenesis in model organisms is a powerful tool for investigation of molecular mechanisms in biological systems.
  25. [25]
    Cancer - How Tobacco Smoke Causes Disease - NCBI - NIH
    These types of mutations are frequently observed in the KRAS oncogene in lung cancer and in the TP53 gene in a variety of cancers induced by cigarette smoke.
  26. [26]
    Tobacco Smoke: Chemical Carcinogenesis and Genetic Lesions - NIH
    DNA is mutated by cigarette pollutants in a very deliberate fashion that differs, in general, from the mechanism of mutation by endogenous factors. Endogenous ...
  27. [27]
    Mutational Signatures as Sensors of Environmental Exposures
    Sep 27, 2022 · In summary, the mutational signatures observed in LUAD can be divided into three groups: smoking-associated (SBS4, SBS5, SBS40), immune-related ...
  28. [28]
    Transitions vs transversions
    Transitions involve purine-purine or pyrimidine-pyrimidine changes, while transversions involve purine-pyrimidine changes. Transitions are more frequent and ...Missing: ratio sources
  29. [29]
    Oxidized, deaminated cytosines are a source of C → T transitions in ...
    A model for C → T oxidative mutagenesis is suggested in which initial cytosine oxidation is followed by deamination to a poorly repaired uracil derivative.
  30. [30]
    Preservation of methylated CpG dinucleotides in human CpG islands
    Mar 22, 2016 · The methylated CpG dinucleotides can lead to a high rate of C to T substitution at these sites. It is known that a high mutation rate can be ...<|control11|><|separator|>
  31. [31]
    Error-prone polymerase activity causes multinucleotide mutations in ...
    First, they are enriched for transversions, with a transition:transversion ratio of 1:1 in contrast to the 2:1 genome-wide average. Second, we find that ...
  32. [32]
    On the Causes of Evolutionary Transition:Transversion Bias
    A pattern in which nucleotide transitions are favored several fold over transversions is common in molecular evolution.
  33. [33]
    Frameshift Mutation - an overview | ScienceDirect Topics
    Frameshift mutations are defined as insertions or deletions in the nucleic acid sequence that shift the translation mechanism from one reading frame to another, ...
  34. [34]
    Insertions and Deletions: Computational Methods, Evolutionary ...
    After point mutations, insertions and deletions (a.k.a. indels) constitute the second most important source of genomic variation in populations and species.
  35. [35]
    The shared frameshift mutation landscape of microsatellite-unstable ...
    MMR deficiency leads to insertion/deletion (indel) mutations at coding microsatellites (cMS) and to neoantigen-inducing translational frameshifts. Here, we ...
  36. [36]
    A Novel Frameshift Mutation (+A) at Codon 18 of the β-Globin Gene ...
    Jan 30, 2008 · The novel mutation causes a frameshift with the generation of a UGA stop codon. Analysis of the parent's DNA demonstrates that the A insertion ...Dna Sequencing · Results · Analysis Of Globin Mrna...
  37. [37]
    A frameshift mutation in a patient with Tay-Sachs disease causes ...
    Dec 15, 1989 · A frameshift mutation in a patient with Tay-Sachs disease causes premature termination and defective intracellular transport of the alpha ...Missing: example | Show results with:example
  38. [38]
    Real-time dynamics of mutagenesis reveal the chronology of DNA ...
    Jun 26, 2018 · The mismatch can either be successfully repaired by MMR or turn into a stable mutation during the next round of replication. (B) Single-cell ...
  39. [39]
    Supercoiling and looping promote DNA base accessibility ... - Nature
    Sep 28, 2021 · Positive supercoiling exposes DNA bases​​ Unless this positive supercoiling is efficiently relaxed, the polymerases will stall, and replication ...
  40. [40]
    Translation: DNA to mRNA to Protein | Learn Science at Scitable
    Point mutations close to the AUG initiator codon affect the efficiency of translation of rat preproinsulin in vivo. Nature 308, 241–246 (1984) doi ...
  41. [41]
    Protein Misfolding and Degenerative Diseases - Nature
    In a properly folded protein, hydrophobic amino acid residues are together ... Disease-causing mutations seem to confer a toxic gain of function.Missing: loss | Show results with:loss
  42. [42]
    Loss-of-function, gain-of-function and dominant-negative mutations ...
    Jul 6, 2022 · For example, with the dominant-negative (DN) effect, the expression of a mutant protein interferes with the activity of a wild-type protein.
  43. [43]
  44. [44]
    An experimental test on the probability of extinction of new genetic ...
    Sep 13, 2013 · In 1927, J.B.S. Haldane reasoned that the probability of fixation of new beneficial alleles is twice their fitness effect.Missing: formula | Show results with:formula
  45. [45]
    The population genetics of mutations: good, bad and indifferent - PMC
    There are (i) point mutations that change only a single letter and lead to so-called 'single nucleotide polymorphisms' in populations, (ii) insertions and ...
  46. [46]
    Proteins' Evolution upon Point Mutations | ACS Omega
    Apr 14, 2022 · This work contributes to our general understanding of the effects of point mutations on proteins and may spur significant progress.
  47. [47]
    Computed structures of point deletion mutants and their enzymatic ...
    Point deletions in enzymes can vary in effect from negligible to complete loss of activity, however, these effects are not generally predictable.
  48. [48]
    Quantifying the Adaptive Potential of an Antibiotic Resistance Enzyme
    Jun 28, 2012 · We study the distribution of mutation effects on cefotaxime (Ctx) resistance and fitness of 48 unique beneficial mutations in the bacterial enzyme TEM-1 β- ...
  49. [49]
    Patterns of Epistasis between Beneficial Mutations in an Antibiotic ...
    May 15, 2013 · We take a systematic approach by quantifying epistasis in two sets of four beneficial mutations in the antibiotic resistance enzyme TEM-1 β-lactamase.
  50. [50]
    Heterozygote Advantage Is a Common Outcome of Adaptation in ...
    In the homozygous state, the HbS mutation overshoots the optimal phenotype, leading to severe anemia causing the often fatal sickle-cell disease (Kwiatkowski ...
  51. [51]
    Somatic Mutation vs. Germline Mutation - Cleveland Clinic
    May 24, 2022 · Germline mutations are DNA changes inherited during conception. Somatic mutations happen after conception to cells other than the egg and ...
  52. [52]
    Exploring the link between the germline and somatic genome in ...
    The analysis focused on the association between germline genotype and the frequency of somatic mutations in 138 well established cancer genes. The sample ...
  53. [53]
    KRAS G12D Mutation Subtype in Pancreatic Ductal Adenocarcinoma
    Oct 10, 2022 · KRAS G12D mutation subtype is present in over 40% of pancreatic ductal adenocarcinoma (PDAC), one of the leading global causes of cancer death.
  54. [54]
    MRTX1133 Targets Tumors with KRAS G12D Mutations - NCI
    Jan 12, 2023 · The G12D mutation is the most common in pancreatic cancer, present in approximately 35% of people diagnosed with the disease.Missing: point | Show results with:point<|separator|>
  55. [55]
    Emerging strategies to target RAS signaling in human cancer therapy
    Jul 23, 2021 · RAS mutations (HRAS, NRAS, and KRAS) are among the most common oncogenes, and around 19% of patients with cancer harbor RAS mutations.
  56. [56]
    TP53 Mutations in Human Cancers: Origins, Consequences ... - NIH
    Hotspot mutations are at residues involved either in making contacts with DNA or in supporting the structure of the DNA-binding surface. Mutant proteins have ...
  57. [57]
    Why are there hotspot mutations in the TP53 gene in human cancers?
    Nov 3, 2017 · TP53 missense mutations are the most common mutation in human cancers. ... These positions include R175, V157F, Y220C, G245, R248, R249, R273 and ...
  58. [58]
    Oncogenic mutant RAS signaling activity is rescaled by the ERK ...
    Activating mutations in RAS are present in ~ 30% of human tumors, and the resulting aberrations in ERK/MAPK signaling play a central role in oncogenesis.
  59. [59]
    CMC - Cancer Mutation Census - COSMIC
    The Cancer Mutation Census (CMC) project is an undertaking to classify coding mutations in COSMIC and identify variants driving different types of cancer.
  60. [60]
    Repeat-induced G-C to A-T Mutations in Neurospora - Science
    The process (termed RIP) produces exclusively GC to AT mutations. Changes occur principally at sites where adenine is 3′ of the changed cytosine.
  61. [61]
    Recurrence of repeat-induced point mutation (RIP) in Neurospora ...
    Duplicate DNA sequences in the genome of Neurospora crassa can be detected and mutated in the sexual phase of the life cycle by a process termed RIP.Missing: mechanism | Show results with:mechanism
  62. [62]
    Repeat-induced point mutation in Neurospora crassa causes the ...
    Jun 16, 2020 · Repeat-induced point (RIP) mutation in Neurospora crassa degrades transposable elements by targeting repeats with C→T mutations.
  63. [63]
    Repeat-Induced Point Mutation and Other Genome Defense ... - NIH
    In Neurospora vegetative cells, homology-directed heterochromatic silencing could play a backup role and/or could come into play in situations where repetitive ...
  64. [64]
    Partition of Repeat-Induced Point Mutations Reveals Structural ...
    Aug 21, 2018 · A premeiotic process known as repeat-induced point mutation (RIP) can accurately identify and mutate nearly all gene-sized DNA repeats present in the haploid ...
  65. [65]
    A cytosine methyltransferase homologue is essential for repeat ...
    During sexual development, Neurospora crassa inactivates genes in duplicated DNA segments by a hypermutation process, repeat-induced point mutation (RIP).Missing: rate | Show results with:rate
  66. [66]
    DNA methylation associated with repeat-induced point mutation in ...
    Repeat-induced point mutation (RIP) is a process that efficiently detects DNA duplications prior to meiosis in Neurospora crassa and peppers them with G:C to A ...Missing: AID- | Show results with:AID-
  67. [67]
    Repeat-Induced Point Mutation and the Population Structure of ... - NIH
    Repeat-induced point mutation (RIP) is a genome defense in fungi that hypermutates repetitive DNA and is suggested to limit the accumulation of transposable ...
  68. [68]
    Phylogenomic signatures of repeat-induced point mutations across ...
    In fungi, repeat-induced point mutation (RIP) targets TEs by recognizing repetitive sequences and inducing mutagenesis. However, the prevalence of RIP across ...
  69. [69]
    Neurospora crassa, a Model System for Epigenetics Research - PMC
    Thus, RIP may indeed limit evolution through gene duplication in Neurospora. ... RIP provided the first simple method to knock out genes in Neurospora and ...
  70. [70]
    [PDF] Use of RIP to inactivate genes in Neurospora crassa.
    In order to detect changes in the primary structure of DNA due to RIP, it is useful to employ restriction enzymes insensitive to cytosine methylation. We have ...
  71. [71]
  72. [72]
    Programmable editing of a target base in genomic DNA ... - Nature
    Apr 20, 2016 · Here we report the development of 'base editing', a new approach to genome editing that enables the direct, irreversible conversion of one ...
  73. [73]
    Editing the Sickle Cell Disease Mutation in Human Hematopoietic ...
    Aug 7, 2019 · Sickle cell disease (SCD) is an ideal model to investigate the potential use of gene editing to transvert a single point mutation at the β- ...
  74. [74]
    Precise base editing for the in vivo study of developmental signaling ...
    Feb 12, 2021 · In our work, we took advantage of base editors to generate C-to-T point mutations at unprecedented high efficiencies (up to 91%) without ...<|separator|>
  75. [75]
    Artificial Transmutation of the Gene - Science
    Artificial Transmutation of the Gene. H. J. MullerAuthors Info & Affiliations. Science. 22 Jul 1927. Vol 66, Issue 1699. pp. 84-87. DOI: 10.1126/science.66.1699 ...
  76. [76]
    a structure for deoxyribose nucleic acid - PubMed
    1953 Apr 25;171(4356):737-8. doi: 10.1038/171737a0. Authors. J D WATSON, F H CRICK. PMID: 13054692; DOI: 10.1038/171737a0. No abstract available. MeSH terms.