Fact-checked by Grok 2 weeks ago

Restriction enzyme

Restriction enzymes, also known as restriction endonucleases, are proteins produced by bacteria as part of their defense system against invading foreign DNA, such as from bacteriophages, by recognizing and cleaving specific short nucleotide sequences within the DNA molecule. These enzymes typically target palindromic recognition sites—symmetric sequences like 4 to 8 base pairs long—and hydrolyze the phosphodiester bonds in the DNA backbone, generating either blunt ends or overhanging "sticky" ends that facilitate subsequent ligation in molecular applications. Over 4,000 restriction enzymes have been identified (as of 2024), with type II enzymes being the most commonly used due to their precise cleavage at or near the recognition site without requiring additional cofactors. The discovery of restriction enzymes stemmed from observations in the 1950s and 1960s of bacterial host-controlled restriction of phage growth, with the enzymatic basis first described in 1968 by researchers including and . The first type II restriction enzyme, HindII, was isolated in 1970 by Hamilton Smith, enabling the specific fragmentation of DNA and paving the way for recombinant DNA technology. This breakthrough earned Arber, Smith, and the 1978 in Physiology or Medicine for their work on restriction enzymes and their application to . Since their identification, restriction enzymes have become indispensable tools in , facilitating techniques such as DNA cloning, , polymerase chain reaction (PCR) analysis, and the creation of genetically modified organisms. They enable the precise cutting and joining of DNA segments from different sources, underpinning advancements in biotechnology, forensics, and medical diagnostics, while their bacterial origins highlight an ancient immune-like mechanism evolved to combat viral infections.

Discovery and History

Initial Discovery

The initial discovery of restriction enzymes stemmed from observations of host-specific variations in bacteriophage infectivity during the early 1950s. In 1952, Salvador E. Luria and Mary L. Human conducted phage infection experiments using T1 and T3 bacteriophages on different bacterial hosts, including strains of Shigella dysenteriae and Escherichia coli. They observed that phages propagated on one bacterial strain exhibited significantly reduced virulence—often by factors of 10^3 to 10^5—when infecting a different strain, a phenomenon termed "host-induced variation." This variation was non-hereditary, as progeny phages regained high efficiency upon passage through the original host, suggesting a reversible modification or degradation process mediated by the bacterial host. Similar findings were reported in 1953 by Giuseppe Bertani and Jean J. Weigle, who studied bacteriophage on E. coli strains B and K. Using plaque assays, they quantified the efficiency of plating (EOP), noting that lambda phages grown on strain B formed plaques on strain K with an EOP of approximately 10^{-4}, while the reverse showed near-normal efficiency. This host-controlled asymmetry indicated that strain K restricted phages adapted to B, with restriction occurring intracellularly during the early stages of infection. These experiments highlighted the phenomenon's specificity to bacterial-phage interactions, laying the groundwork for understanding it as a defense mechanism. In the 1960s, Werner Arber extended these observations by investigating the molecular basis of "host-controlled variation" in bacteriophage lambda using E. coli strains C, K, and B. Collaborating with Daisy Dussoix, Arber performed DNA transfection assays, extracting naked phage DNA and measuring its ability to produce progeny phages in permissive and non-permissive hosts. They found that DNA from lambda grown on one strain was inefficiently transfecting in another (EOP ~10^{-4}), but efficiency was restored after modification in the new host. These results demonstrated that host specificity resides in the DNA molecule itself, leading Arber to hypothesize the existence of bacterial enzymes that selectively degrade unmodified foreign DNA while sparing host-modified DNA—a concept central to restriction-modification systems. The first restriction enzyme was isolated in 1968 by and Robert Yuan from E. coli strain K12. Through of cell extracts and assays monitoring the degradation of lambda DNA from non-permissive hosts, they purified an ATP- and S-adenosylmethionine-dependent endonuclease (now known as EcoK) that specifically cleaved unmodified foreign DNA into fragments while leaving host DNA intact. This enzymatic activity was confirmed via sedimentation analysis, showing rapid breakdown of restricted DNA . The isolation relied on transformation efficiency tests with bacterial DNA, where enzyme-treated foreign DNA failed to transform recipient cells, establishing the enzyme's role in the observed restriction phenomenon.

Key Milestones and Nobel Recognition

In 1970, Hamilton O. Smith and Kent W. Wilcox purified and characterized HindII, the first Type II restriction endonuclease, from Rd, marking a pivotal advancement in enabling precise DNA cleavage at specific recognition sites. This isolation demonstrated the enzyme's ability to produce discrete DNA fragments, laying the groundwork for subsequent techniques. By 1972, Herbert W. Boyer and Robert B. Helling isolated from , a Type II enzyme that generates cohesive ends, facilitating the first successful experiments when combined with . This breakthrough, stemming from Boyer's lab at the , enabled the ligation of foreign DNA fragments into plasmids, revolutionizing . The profound impact of these discoveries was recognized in 1978, when the Nobel Prize in Physiology or Medicine was awarded jointly to Werner Arber, Daniel Nathans, and Hamilton O. Smith for their foundational work on restriction enzymes and their applications in understanding and manipulating DNA. Arber's earlier theoretical insights into restriction-modification systems, Nathans' mapping of viral genomes using these enzymes, and Smith's purification efforts were highlighted as transforming biology. Following these milestones, commercialization accelerated after 1980 through companies like New England Biolabs (NEB), which had begun supplying restriction enzymes in 1975 and expanded production to meet growing demand in recombinant DNA research. This led to the characterization of over 4,000 restriction enzymes by 2023, with REBASE documenting thousands more putative systems, enabling widespread adoption in genomics and biotechnology. A recent expansion came in 2023 with the identification of CoCoNuTs, a subclass of Type IV restriction systems predicted to target RNA in addition to DNA, broadening applications beyond traditional DNA manipulation.

Biological Origins and Function

Role in Bacterial Defense

Restriction enzymes function as a key component of the , providing defense against invading bacteriophages by recognizing and cleaving foreign DNA at specific sequences. This mechanism protects the from , thereby preventing the propagation of phage progeny within the host cell. In essence, these enzymes act as a primitive form of immunity, analogous to eukaryotic adaptive immune responses but operating through sequence-specific activity to degrade non-self DNA. The specificity of restriction enzymes targets unmethylated or differently modified foreign DNA, while the host's own DNA is spared through protective patterns established by companion methyltransferases in restriction-modification (R-M) systems. This distinction allows to differentiate self from non-self nucleic acids, ensuring that only invading phage genomes are fragmented and rendered non-functional. Evolutionarily, this confers significant advantages by reducing successful phage infections; studies on phage show that in recognition sites can increase probabilities by up to several orders of magnitude, but such adaptations often impose fitness costs on the phage, limiting their prevalence. For instance, phages evolving point in restriction sites demonstrate higher survival rates against host enzymes, highlighting the ongoing selective pressure driving bacterial-phage . Bacteriophages counter this defense through various strategies, including the production of anti-restriction proteins that inhibit enzyme activity or modifications to their DNA that mimic host methylation patterns. These countermeasures, such as phage-encoded nucleases or decoy substrates, enable some phages to bypass cleavage and establish infection, though they require rapid evolutionary tuning to match diverse host defenses. Despite these adaptations, R-M systems remain highly prevalent, found in over 80% of sequenced prokaryotic genomes as of 2022, underscoring their enduring role in microbial immunity.

Restriction-Modification Systems

Restriction-modification (R-M) systems consist of a restriction endonuclease that cleaves DNA at specific sequences and a site-specific DNA methyltransferase, also known as the modification enzyme, that methylates the same sequences to protect the host . The two enzymes typically function in tandem within bacterial cells, with the methyltransferase ensuring that host DNA is shielded from the endonuclease's activity. In the mechanism of R-M systems, the methyltransferase adds a methyl group to a specific base within the recognition sequence on the host DNA, thereby preventing the restriction endonuclease from binding and cleaving at that site. Unmethylated foreign DNA, such as that from invading bacteriophages, lacks this protective modification and is thus recognized and degraded by the endonuclease, providing a defense against viral infection. This selective degradation maintains genomic integrity while targeting exogenous nucleic acids. The modification enzymes in R-M systems catalyze three primary types of DNA methylation: N6-methyladenine (m6A), (m5C), or N4-methylcytosine (m4C), each occurring at defined positions within the recognition sequence. These modifications vary across different R-M systems and contribute to the specificity of host protection. R-M systems exhibit diversity, including phase-variable forms in where the expression of the restriction and modification components can switch on or off through mechanisms like slipped-strand mispairing in invertible promoter regions, enabling adaptive immune responses to changing environmental threats. This variability allows to modulate their defense strategies dynamically. Experimental evidence for the protective role of matching modification enzymes comes from transformation assays, where unmethylated plasmid DNA shows drastically reduced uptake and integration efficiency in host cells harboring active R-M systems, but efficiency is restored when the DNA is pre-methylated by the corresponding methyltransferase to match the host's modification pattern. For instance, in Synechococcus sp. PCC 7002, methylation with the Escherichia coli Dam methylase significantly enhanced transformation efficiency by evading restriction, demonstrating the necessity of sequence-specific protection. Similar results in Borrelia burgdorferi confirm that CpG methylation protects against host restriction, underscoring the system's specificity.

Molecular Recognition and Cleavage

Recognition Sequences

Restriction enzymes identify their target sites on double-stranded DNA through specific recognition sequences, which are typically short palindromic motifs of 4 to 8 base pairs in length. These sequences exhibit dyad symmetry, reading the same on both strands when oriented from 5' to 3', allowing symmetric binding by dimeric enzymes. The palindromic nature ensures that the enzyme can interact equivalently with both halves of the site, facilitating precise recognition. Sequence specificity is central to enzyme function, with examples including the 6-base pair sequence 5'-GAATTC-3' recognized by , isolated from . Isoschizomers are distinct enzymes from different bacterial sources that share the identical recognition sequence and cleavage pattern, such as SphI and BbuI, both targeting 5'-GCATGC-3'. This conservation highlights evolutionary convergence in DNA targeting mechanisms across species. Binding occurs primarily through enzyme dimers that form hydrogen bonds with bases in the major groove and van der Waals contacts with the sugar-phosphate backbone, enabling discrimination of the correct sequence from non-cognate sites. The structural basis of has been elucidated by of enzyme-DNA complexes, revealing intimate molecular interactions. For instance, in the EcoRI-DNA complex at 3 Å resolution, 12 bonds mediate specificity: Arg200 forms two bonds with , while Glu144 and Arg145 contribute four bonds to adjacent residues, collectively ensuring . Typically, type II restriction enzymes form 15-20 such bonds with the , supplemented by van der Waals interactions. Recognition efficiency is modulated by external factors, including salt concentration, which affects electrostatic interactions between the enzyme and DNA phosphate backbone; higher salt levels can reduce binding affinity for many enzymes. Temperature influences enzyme stability and kinetics, with optimal ranges often around 37°C but varying by enzyme. Additionally, the flanking sequence context around the recognition site can impact cleavage rates, as certain adjacent bases may alter DNA conformation or enzyme access.

Types of Cuts and Overhangs

Restriction enzymes cleave DNA at specific recognition sequences through hydrolysis of the phosphodiester backbone, producing either blunt ends or sticky ends depending on the enzyme's cleavage pattern. Blunt-end cleavage occurs when the enzyme cuts both strands at the same position within the recognition site, resulting in flush termini without overhangs; for example, SmaI recognizes CCCGGG and cuts between the third C and first G on each strand, yielding blunt-ended fragments. In contrast, sticky-end (or cohesive-end) cleavage involves staggered cuts offset by one to four nucleotides, generating single-stranded overhangs that facilitate base-pairing; EcoRI, for instance, recognizes GAATTC and cleaves between G and A on each strand, producing 5'-AATT overhangs. The cleavage reaction is a magnesium (Mg²⁺)-dependent , where a water , activated by the and Mg²⁺ cofactor, performs a nucleophilic attack on the scissile , leading to inversion of at the atom and producing a 5'-phosphate on one fragment and a 3'-hydroxyl group on the other. This mechanism ensures precise bond breakage following recognition of the , with the simplified reaction represented as: \text{DNA}-(\text{pN}) \rightarrow \text{DNA}-p + \text{HO}-N where "pN" denotes the phosphodiester linkage. Overhangs from sticky-end cuts are typically 1-4 nucleotides long, with 5' or 3' polarity depending on the enzyme; enzymes producing compatible overhangs, such as EcoRI (5'-AATT) and MfeI (also 5'-AATT from CAATTC), allow directional ligation of fragments despite differing recognition sites. Under suboptimal conditions, such as high enzyme concentration, elevated glycerol levels, or non-optimal pH and ionic strength, restriction enzymes may exhibit star activity, resulting in non-specific cleavage at sequences similar to the canonical site.

Classification of Restriction Enzymes

Type I Restriction Enzymes

Type I restriction enzymes are large, multisubunit components of bacterial restriction-modification (RM) systems, functioning primarily as a defense mechanism against invading foreign DNA such as bacteriophages. Unlike simpler enzymes, they combine DNA recognition, methylation, translocation, and cleavage activities within a single complex, requiring energy input for their full function. The holoenzyme forms a heteropentameric structure consisting of two restriction (R) subunits, two modification (M) subunits, and one DNA sequence-specificity (S) subunit, often denoted as R₂M₂S. The R subunits contain ATPase, helicase-like translocase, and endonuclease domains, enabling DNA movement and cleavage, while the M subunits handle site-specific methylation to protect host DNA. The S subunit, which determines sequence specificity, bridges the R and M subunits and features two target recognition domains (TRDs) connected by a central linker, allowing it to bind asymmetric, bipartite DNA sequences. The recognition process involves binding to specific, interrupted palindromic sequences, typically 15 base pairs long with a 6- to 8-nucleotide spacer, such as AAC(N₆)GTGC for the EcoKI enzyme from Escherichia coli K-12. Upon binding unmethylated foreign DNA, the enzyme initiates an ATP-dependent translocation mechanism, where the R subunits use ATP hydrolysis to reel in DNA from both directions at rates up to 4 kb/min, forming looped structures as the enzyme moves bidirectionally along the double helix. This translocation continues until the enzyme collides with another DNA-bound complex or encounters a barrier, triggering random double-strand cleavage at distant, variable positions (typically thousands of base pairs away) from the recognition site, without sequence specificity at the cut site. Essential cofactors include ATP for energy-dependent translocation and helicase activity, S-adenosylmethionine (SAM) for the methylation function of the M subunits, and Mg²⁺ ions to facilitate the endonucleolytic hydrolysis during cleavage. In their biological role, Type I enzymes like EcoKI and EcoAI contribute to bacterial immunity by destroying unmodified invading DNA while sparing methylated host genomes, though the process can sometimes lead to host DNA damage if not tightly regulated. Their complex, non-specific cutting pattern—resulting in blunt or short overhangs at unpredictable locations—makes them less practical for routine laboratory applications such as or DNA mapping, where precise, site-specific incisions are preferred; consequently, they are rarely used in workflows despite their foundational importance in RM system studies.

Type II Restriction Enzymes

Type II restriction enzymes constitute the largest and most widely utilized class of restriction endonucleases, prized for their precise, ATP-independent cleavage of DNA at or near specific recognition sequences. Unlike Type I and Type III enzymes, which rely on ATP hydrolysis and exhibit translocation activity, Type II enzymes perform direct hydrolysis of phosphodiester bonds in the DNA backbone, requiring only Mg²⁺ as a cofactor. This simplicity enables their routine use in DNA manipulation, where they generate defined fragments essential for cloning, mapping, and sequencing. Structurally, orthodox Type II enzymes typically function as homodimers, with each subunit contributing to the recognition of a short, symmetric palindromic DNA sequence, usually 4 to 8 base pairs long. For instance, the enzyme EcoRI from Escherichia coli recognizes the sequence 5'-GAATTC-3' and cleaves between the G and A residues on both strands, producing 5' sticky ends. The dimeric structure allows the enzyme to bind across the DNA double helix, positioning catalytic residues from each monomer to attack opposite strands simultaneously, ensuring coordinated cleavage. Subtypes deviate from this archetype: Type IIS enzymes, such as FokI, feature separate domains for DNA recognition and catalysis, recognizing asymmetric sequences and cutting at fixed positions offset from the site, often generating 5' or 3' overhangs. In contrast, Type IIG enzymes, like BcgI, integrate restriction and methylation activities into a single polypeptide, allowing coupled cleavage and modification. Type II enzymes are further classified by cleavage patterns: conventional Type IIP enzymes cut within the palindromic recognition site to yield either sticky (cohesive) or blunt ends, while Type IIS variants enable more flexible strategies due to their offset cuts. Over 3,500 Type II restriction enzymes have been isolated and characterized, primarily cataloged in the REBASE database, with hundreds commercially available for laboratory applications. Notably, heat-labile mesophilic variants are complemented by thermostable enzymes from thermophilic , such as TaqI from , which withstand conditions and facilitate high-temperature reactions without denaturation.

Type III Restriction Enzymes

Type III restriction enzymes constitute a distinct class within bacterial restriction-modification (RM) systems, serving as a defense mechanism against foreign DNA invasion by cleaving unmethylated DNA at specific sites. These enzymes are less common than Type II variants, with only a limited number of well-characterized examples identified across bacterial genomes, reflecting their specialized role in advanced host protection strategies. Unlike Type II enzymes, which cleave directly at or near their recognition sequences, Type III enzymes exhibit a more complex, energy-dependent process involving DNA translocation. The structural organization of Type III enzymes features a heterodimeric arrangement of two distinct subunits: the Mod (modification) subunit, which handles sequence-specific DNA binding and adenine methylation for host protection, and the Res (restriction) subunit, which confers the endonucleolytic activity. The functional holoenzyme assembles as a heterotetramer (Mod₂Res₂), where the Mod subunits each contain a target recognition domain (TRD) for binding asymmetric sequences, and the Res subunits provide ATPase and nuclease functionalities. This modular architecture enables bidirectional DNA tracking, distinguishing Type III systems from simpler RM setups. Mechanistically, Type III enzymes recognize non-palindromic, asymmetric DNA sequences of 5–6 base pairs, such as the 5'-CAGCAG-3' site targeted by EcoP15I from . Efficient cleavage requires two such unmethylated sites in inverse (head-to-head) orientation, separated by 1–25 base pairs, with the enzyme binding one site and using to translocate short-range along the DNA until colliding with the second site, triggering a double-strand break. Cuts occur asymmetrically, approximately 25–27 nucleotides downstream of the recognition sites on opposite strands, generating 2-base 5' overhangs. The process is ATP- and Mg²⁺-dependent, proceeding more slowly than in Type I systems due to limited processivity and reliance on site collision rather than long-range looping. This translocation-based activation enhances specificity in dense bacterial genomes while minimizing off-target cuts.

Type IV Restriction Enzymes

Type IV restriction enzymes represent a distinct of modification-dependent restriction endonucleases that specifically recognize and cleave DNA harboring post-replicative modifications, such as (5mC) or N6-methyladenine (6mA), without requiring a paired methyltransferase as in classical restriction-modification systems. Unlike Types I-III, which target unmodified palindromic sequences, Type IV enzymes exhibit low sequence selectivity and focus on epigenetic marks that foreign DNA, like from invading phages, may carry to evade host defenses. These enzymes are typically encoded within bacterial defense islands, genomic loci enriched for anti-phage systems, highlighting their integration into broader prokaryotic immune strategies. Structurally, Type IV enzymes often comprise multi-subunit complexes with dedicated domains for modification sensing and activity. For instance, the prototypical McrBC system in consists of the McrB subunit, which includes an N-terminal HNH domain, a central helicase-like domain for DNA translocation, and a C-terminal methylcytosine-binding domain, paired with the accessory McrC subunit that enhances specificity. Recent structural studies on the GmrSD-family enzymes, such as the 2021 X-ray (PDB: 7P9M) of BrxU, reveal a dimeric architecture where the GmrS specificity subunit forms an aromatic to bind modified bases like (5hmC), while the GmrD endonuclease delivers the cleavage. These structures underscore evolutionary adaptations for probing DNA modifications, often without strict ATP dependence, contrasting with the energy-intensive translocation in other restriction types. The mechanism of Type IV enzymes involves modification-triggered DNA binding followed by cleavage at or near the altered site, frequently via a looping or translocation process to amplify destructive potential. In McrBC, recognition occurs at purine-methylcytosine (RmC) motifs separated by 40–3,000 base pairs; GTP hydrolysis powers bidirectional DNA unwinding and looping, culminating in non-specific double-strand breaks approximately 30 bp from one of the recognition sites. This process is modification-specific but sequence-tolerant, enabling broad-spectrum defense against methylated phage genomes that might otherwise bypass unmethylation-dependent systems. Biologically, these enzymes fortify bacterial immunity by targeting the epigenetic signatures of foreign nucleic acids, as evidenced by their prevalence in diverse prokaryotes and role in restricting phages like λ carrying dam or dcm methylation patterns. Key examples include McrA, which cleaves DNA at 5mC residues positioned near the fragment center in a length-dependent manner, and McrBC, widely studied for its GTPase-driven activity in E. coli K-12 strains. The GmrSD family, identified in , exemplifies emerging variants that target glucosylated 5hmC, with 2024–2025 studies revealing their potential for selective epigenetic cleavage in contexts. These systems have independently evolved multiple times, reflecting their adaptive value in microbial conflict.

Type V Restriction Enzymes

Type V restriction enzymes represent a distinct class of programmable DNA endonucleases within bacterial defense systems, primarily exemplified by CRISPR-associated (Cas) proteins such as from the system. Unlike traditional restriction enzymes that rely on fixed recognition sites, Type V enzymes utilize guide RNAs to achieve sequence-specific targeting of foreign DNA, enabling adaptive immunity against phages and plasmids. These enzymes were first characterized as part of Class 2 systems, where a single effector protein performs both recognition and cleavage functions. The structure of Type V enzymes like Cas9 consists of a single polypeptide chain, approximately 1368 amino acids in length for Streptococcus pyogenes Cas9 (SpCas9), which folds into bilobed architecture with recognition (REC) and nuclease (NUC) lobes. This protein forms a ribonucleoprotein complex with CRISPR RNA (crRNA) and trans-activating crRNA (tracrRNA), or a single-guide RNA (sgRNA) fusion for simplified use. The REC lobe binds the guide RNA and target DNA, while the NUC lobe houses the catalytic domains responsible for cleavage. Crystal structures reveal that the guide RNA scaffolds the DNA duplex into the active site, forming an R-loop where the target strand hybridizes with the crRNA spacer sequence. The mechanism of Type V enzymes involves RNA-guided DNA recognition followed by double-strand break (DSB) formation. Target interrogation begins with (PAM) recognition, a short cis-element required for binding; for SpCas9, this is typically 5'-NGG-3', where N is any and G is , located 3-4 base pairs downstream of the target site. Upon PAM binding, the guide RNA's 20-nucleotide spacer sequence pairs with complementary target DNA, displacing the non-target strand to form a . occurs via two distinct domains: the RuvC domain cuts the non-target strand, and the HNH domain cleaves the target strand, generating blunt-ended DSBs approximately 3 base pairs upstream of the PAM. This process requires divalent metal ions, primarily Mg²⁺, as cofactors to coordinate the catalytic residues in both s. The programmable stems from the crRNA, which can be engineered to direct cleavage to virtually any genomic locus adjacent to a suitable PAM. Recognition by Type V enzymes is highly specific yet flexible due to the guide RNA's role in defining the target sequence, contrasting with the rigid palindromic sites of other restriction types. The PAM acts as a safety checkpoint to distinguish self from non-self DNA, as CRISPR arrays lack corresponding PAMs. Variations in PAM requirements exist across orthologs; for instance, Staphylococcus aureus Cas9 (SaCas9) prefers 5'-NNGRRT-3', expanding targetable sites. Mismatches in the seed region (positions 1-12 of the spacer) abolish activity, ensuring precision, while distal mismatches are more tolerated. Recent advances from 2023 to 2025 have leveraged Type V scaffolds, particularly deactivated Cas9 (dCas9) or nickase variants (nCas9), to develop base editors and prime editors for precise genome modification without DSBs. Base editors fuse deaminases (e.g., cytidine deaminase for C-to-T or adenine deaminase for A-to-G conversions) to nCas9, enabling single-base changes with efficiencies up to 50-70% in mammalian cells, as optimized in 2024 variants with improved PAM compatibility and reduced bystander editing. Prime editors, introduced earlier but refined in this period, combine nCas9 with a reverse transcriptase and prime editing guide RNA (pegRNA) to install insertions, deletions, or all 12 base-to-base transitions directly from an extended RNA template, achieving up to 20-50% efficiency in human cells with 2025 iterations incorporating AI-designed Cas9 mutants for enhanced specificity and reduced off-target effects. These tools have facilitated therapeutic applications, such as correcting sickle cell mutations in preclinical models.

Engineered Restriction Enzymes

Artificial and Synthetic Variants

Artificial and synthetic variants of restriction enzymes represent engineered endonucleases designed in laboratories to recognize and cleave DNA sequences absent in natural enzymes, thereby broadening applications in precise DNA manipulation. These variants are developed through targeted modifications to achieve novel recognition specificities, often starting from existing enzyme scaffolds like type II restriction endonucleases or homing endonucleases. Rational design approaches focus on structure-guided mutations to alter DNA-binding interfaces. For example, in the MmeI family of type II restriction endonucleases, single or paired substitutions can alter specificity at specific positions in the recognition sequence, such as changing TCCRAC to TCGRAC, leveraging crystallographic data to predict changes without disrupting catalytic activity. This method enables the creation of isoschizomers or neoschizomers for specific experimental needs. Combinatorial library methods generate diversity by simultaneously varying multiple residues in the recognition domain. For example, combinatorial mutagenesis of the I-CreI homing endonuclease, a natural rare cutter, involved assembling libraries of variants with altered DNA-binding residues, followed by selection for cleavage of chosen targets like the COMB1 sequence. This yielded artificial enzymes with altered specificity for new sites, demonstrating the power of library-based engineering for custom rare cutters. Directed evolution complements these strategies by iteratively mutating and selecting variants under selective pressure. Early applications include the evolution of BstYI, a type II enzyme recognizing R^GATCY (R = A or G; Y = C or T), where error-prone and selection increased specificity to cut only AGATCT with a 12-fold preference over off-target sites like AGATCC or GGATCT. More recently, directed evolution of NlaIV, which cuts GGN/NCC, produced variants with enhanced specificity for blunt-end cutting through and functional assays. In 2024, methodological advances have accelerated the development of synthetic rare cutters via . A drop-based microfluidic platform enables of restriction endonuclease libraries, detecting cleavage activity with single-drop sensitivity and processing up to 10^5 variants per hour, overcoming bottlenecks in traditional assays for novel specificity engineering. TALENs exemplify synthetic variants with modular design, employing transcription activator-like effector (TALE) proteins—originally from plant pathogens—for customizable DNA binding, paired with a domain to achieve targeted cleavage at user-specified sites of 12–20 base pairs. This allows programming for rare-cutting profiles not achievable with natural type II enzymes. Despite these advances, synthetic variants often face fidelity challenges relative to natural restriction enzymes, including higher rates of off-target cleavage due to incomplete specificity tuning and potential non-specific nuclease activity, which can complicate applications requiring high precision. Ongoing refinements in selection stringency and structural optimization aim to mitigate these issues. These artificial enzymes preview expanded roles in genome editing, offering custom rare cutters for large-scale manipulations like chromosomal rearrangements, surpassing the limitations of natural enzymes in versatility and target range. They served as precursors to more advanced tools like CRISPR-Cas systems for programmable DNA cleavage.

Fusion Enzymes and Meganucleases

Fusion enzymes represent engineered hybrids that combine DNA-binding domains with nuclease modules to achieve precise , distinct from traditional restriction enzymes by their customizable specificity. nucleases (ZFNs) exemplify this approach, consisting of engineered proteins fused to the non-specific cleavage domain of the restriction enzyme. This modular fusion allows ZFNs to recognize specific DNA sequences of 9-18 base pairs, with the domain providing the cutting activity only upon dimerization of two ZFN monomers bound to adjacent sites. The seminal design of ZFNs was reported in , enabling targeted double-strand breaks for applications in and . Meganucleases, derived from homing endonucleases, are compact enzymes that naturally recognize long DNA sequences of 12-40 base pairs, offering inherently high specificity due to their extended contact interfaces. A prominent example is I-SceI, a LAGLIDADG-family homing endonuclease from Saccharomyces cerevisiae mitochondria that cleaves an 18-base-pair site. Engineering efforts have further refined meganuclease specificity through directed evolution and rational redesign, altering recognition patterns while preserving cleavage efficiency. These modifications make meganucleases valuable for therapeutic genome editing, where their large target sites minimize off-target effects compared to shorter-recognition tools. The design of both ZFNs and meganuclease fusions emphasizes modular assembly to target desired loci, with dimerization playing a key role in reducing off-target cleavage. In ZFNs, the FokI domain requires dimerization for activation, as monomeric FokI lacks nuclease activity; two FokI units must associate to form the catalytically competent complex: \text{FokI} + \text{FokI} \rightleftharpoons (\text{FokI})_2 \quad \text{(active nuclease)} This requirement ensures cleavage occurs only when two ZFNs bind in close proximity on opposite DNA strands, separated by a short spacer (typically 5-6 bp), thereby enhancing specificity. Mutations at the FokI dimer interface have further optimized this process, reducing toxicity from unintended cuts. Meganuclease fusions, such as megaTALs, integrate the compact homing endonuclease domain with TALE (transcription activator-like effector) binding modules to create hyper-specific, single-chain nucleases suitable for viral delivery in therapies. Recent advancements as of 2025 highlight compact meganuclease variants for clinical applications, including the engineered RHO1-2 meganuclease designed to correct the P23H mutation in , promoting rejuvenation and cone preservation (preprint, September 2025). These variants maintain the small size of native homing endonucleases (around 300-400 ) while achieving therapeutic efficacy in preclinical models, underscoring their potential in for inherited diseases. Ongoing refinements focus on delivery compatibility and further specificity tuning to advance toward human trials.

Nomenclature and Specific Examples

Naming Conventions

The standardized for restriction enzymes was first proposed in 1973 to systematically name these enzymes based on their bacterial origin, ensuring clarity and traceability in . This system uses an italicized three- or four-letter abbreviation derived from the first letters of the and names of the source organism, followed by one or two letters or numbers designating the strain or , and concluding with a Roman numeral indicating the order of discovery from that strain. For instance, EcoRI refers to the first restriction enzyme isolated from strain RY13. This convention was refined in 2003 to accommodate the growing complexity of enzyme discoveries, including provisions for related proteins like methyltransferases. A key aspect of the distinguishes between isoschizomers and neoschizomers, which are enzymes that recognize identical or similar DNA sequences but originate from different organisms. Isoschizomers catalyze cleavage at the same position within the same recognition sequence; the first identified serves as the prototype, and subsequent ones retain their organism-specific names while being noted as isoschizomers of the prototype—for example, EcoRI and RsrI both recognize the sequence GAATTC and cleave identically. Neoschizomers, in contrast, recognize the same sequence but cleave at different positions, allowing for nuanced functional comparisons across species. These distinctions facilitate the cataloging of variants without implying functional equivalence. In commercial applications, suppliers such as or often assign proprietary catalog codes or numbers for practical distribution, but the adheres strictly to the organism-based to maintain and link enzymes to their biological context. The REBASE database, curated by , serves as the central repository for this information, tracking over 4,000 biochemically characterized restriction enzymes and their variants as of 2025, with regular updates reflecting new isolations and genomic data.

Notable Restriction Enzymes

EcoRI, isolated from Escherichia coli RY13, is one of the first restriction enzymes discovered and widely adopted for molecular cloning. It recognizes the palindromic 6-base pair sequence 5'-GAATTC-3' and cleaves between the G and A residues, generating 5' sticky ends with a 4-nucleotide AATT overhang. This cohesive end facilitates efficient ligation in recombinant DNA construction, marking EcoRI as a foundational tool in genetic engineering since its characterization in the early 1970s. BamHI, derived from Bacillus amyloliquefaciens H, targets the 6-base pair sequence 5'-GGATCC-3' and cuts after the first G on each strand, producing 5' sticky ends with a 4-nucleotide GATC overhang. Its compatibility with multiple cloning sites in expression vectors has made it a staple for inserting genes into plasmids, enabling high-yield in bacterial systems. SmaI, from , is a prototypical blunt-end cutter that recognizes the 6-base pair sequence 5'-CCCGGG-3' and cleaves symmetrically in the center between the third C and G on each strand. This produces flush ends ideal for seamless without overhang compatibility issues, though it requires higher concentrations for efficiency in blunt-end applications. FokI, a Type IIS from Flavobacterium okeanokoites, exemplifies enzymes that cleave outside their asymmetric (5'-GGATG-3', 9/13), generating 4-nucleotide 5' overhangs at variable distances from the site. Its dimeric domain, activated by binding two sites, has been fused to proteins to create nucleases (ZFNs) for targeted . Cas9, an RNA-guided endonuclease classified under Type V systems and derived from Streptococcus pyogenes, represents a paradigm shift in programmable DNA cleavage. It forms a complex with CRISPR RNA (crRNA) and trans-activating crRNA (tracrRNA), or a single-guide RNA (sgRNA), to recognize a (PAM) sequence (5'-NGG-3') and induce double-strand breaks 3-4 base pairs upstream. This versatility has revolutionized by enabling precise, multiplexed modifications across diverse organisms, far surpassing traditional restriction enzymes in specificity and ease of targeting.

Applications in Molecular Biology

Recombinant DNA Technology

Recombinant DNA technology relies on restriction enzymes to facilitate the precise assembly of DNA molecules from different sources, enabling the creation of hybrid DNA constructs for genetic engineering. The core process involves treating both a vector DNA, such as a plasmid, and the insert DNA of interest with the same restriction enzyme to generate compatible ends. For instance, enzymes like EcoRI or HindIII produce sticky ends—single-stranded overhangs that can base-pair with complementary sequences—allowing the fragments to anneal spontaneously before being covalently joined by DNA ligase. This ligation seals the phosphodiester bonds, forming a stable recombinant molecule that can be propagated in host cells. The technique of sticky-end cloning exemplifies the utility of these enzymes, as the overhangs ensure directional insertion of the insert into the vector, minimizing the formation of incorrect orientations or self-ligated vectors. Enzymes such as EcoRI, which recognizes the sequence GAATTC and cuts to leave a 5' overhang of AATT, and HindIII, which cuts AAGCTT to produce a 5' overhang of AGCT, are commonly used for this purpose due to their frequent occurrence in DNA and the specificity of their cuts. This method offers high efficiency compared to blunt-end ligation, with annealing of sticky ends increasing the local concentration of reactive groups and facilitating ligation rates up to 10-100 times higher. Overhang compatibility, as seen in these enzymes, further enhances precision by allowing selective joining of fragments from diverse sources. Plasmid vectors like pBR322, developed in the late 1970s, incorporate multiple unique restriction sites clustered in non-essential regions, often as polylinkers, to accommodate inserts without disrupting replication or selection markers. pBR322 features sites for over 40 restriction enzymes, including EcoRI, HindIII, and PstI, positioned within genes for ampicillin and tetracycline resistance to enable insertional inactivation screening. These polylinkers allow flexibility in choosing enzymes for cloning, supporting the insertion of foreign DNA while maintaining selectable phenotypes for recombinant identification. The advent of restriction enzyme-based in the 1970s, pioneered by experiments combining and viral DNA fragments, revolutionized by enabling isolation, expression, and manipulation on an unprecedented scale. Stanley Cohen and Herbert Boyer's 1973 demonstration of in vitro construction using marked the birth of this technology, leading to widespread applications in and foundational advances in genetic research.

Genome Mapping and Analysis

Restriction mapping is a fundamental technique in genome analysis that utilizes restriction enzymes to determine the positions of their recognition sites on a DNA molecule. By performing single and double digests with different enzymes, the resulting fragment sizes—separated via —reveal the relative order and distances between cut sites, enabling the construction of physical maps. This approach was pioneered in the early 1970s through the of viral , where enzymes like HindII were used to cleave and map the simian virus 40 (SV40) DNA into specific fragments. Type II restriction enzymes, with their precise sequence specificity, are particularly suited for generating detailed maps of smaller DNA regions, as detailed in the classification of these enzymes. Restriction fragment length polymorphism (RFLP) extends restriction mapping by detecting natural variations in DNA sequences that alter restriction sites, producing polymorphic fragment patterns useful for genetic fingerprinting and linkage studies. In RFLP analysis, genomic DNA is digested with restriction enzymes, the fragments are separated by , and variations in band patterns indicate polymorphisms, such as insertions, deletions, or single changes affecting cut sites. This method revolutionized by enabling the construction of linkage maps for gene localization, as demonstrated in early applications to track hereditary conditions like . RFLP has been instrumental in and paternity testing, providing a basis for identifying individuals based on unique DNA profiles. Southern blotting complements restriction-based mapping by allowing the detection of specific DNA sequences within complex digests. The process involves digesting DNA with restriction enzymes, separating the fragments on an agarose gel, transferring them to a nitrocellulose membrane, and hybridizing with a labeled probe to visualize target regions via autoradiography or chemiluminescence. Developed in the mid-1970s, this technique facilitated gene cloning and mutation detection by confirming the presence of restriction fragments corresponding to known sequences. It remains valuable for verifying restriction maps and analyzing gene structure in contexts like viral integration or copy number variations. For mapping large eukaryotic , pulsed-field gel electrophoresis (PFGE) addresses the limitations of conventional gels by resolving DNA fragments exceeding 50 kb, often generated by rare-cutting restriction enzymes like NotI or SfiI. In PFGE, an alternating electric field prevents mega-fragments from migrating anomalously, producing resolvable bands that aid in ordering contigs and constructing high-resolution physical maps. Introduced in the 1980s, PFGE was crucial for projects like the yeast sequencing, where it helped assemble chromosome-sized maps from restriction digests. This method has been applied to microbial , such as typing bacterial strains via macro-restriction profiles. Recent advancements have integrated Type IV restriction enzymes—modification-dependent endonucleases that target methylated or otherwise modified DNA—with next-generation sequencing (NGS) for epigenome mapping. These enzymes selectively cleave modified regions, enriching NGS libraries for sites of epigenetic marks like 5-methylcytosine, enabling base-resolution profiling without bisulfite conversion's DNA damage. In 2024, studies highlighted their utility in bacterial epigenome analysis, revealing modification patterns that influence gene regulation and phage defense, with applications extending to eukaryotic systems for non-destructive methylation detection.

References

  1. [1]
    Restriction Enzyme - National Human Genome Research Institute
    A restriction enzyme is a protein from bacteria that cleaves DNA at specific sites, producing fragments with known sequences. They cut any DNA, not ...Missing: mechanism | Show results with:mechanism
  2. [2]
    DNA Restriction & Nucleic Acid Analysis - Molecular Biology
    Restriction enzymes, or endonucleases, cut DNA at specific sequences, which are palindromic restriction sites. These enzymes are derived from bacteria.
  3. [3]
    GENETIC ENGINEERING – BIOLOGY BASICS
    Restriction enzymes are critical in producing recombinant DNA. Restriction enzymes were discovered in bacteria living in old faithful. More than 3000 ...
  4. [4]
    1968: First Restriction Enzymes Described
    Apr 26, 2013 · Restriction enzymes recognize and cut specific short sequences of DNA. They're found in bacteria, which use the enzymes to digest invading DNA.Missing: definition function
  5. [5]
    Highlights of the DNA cutters: a short history of the restriction enzymes
    This review traces the discovery of restriction enzymes and their continuing impact on molecular biology and medicine.
  6. [6]
    How restriction enzymes became the workhorses of molecular biology
    Apr 26, 2005 · Restriction enzymes have proved to be invaluable for the physical mapping of DNA. They offer unparalleled opportunities for diagnosing DNA sequence content.
  7. [7]
    Restriction enzymes – Molecular Biology and ... - Eagle Pubs
    Restriction enzymes, or restriction endonucleases, are proteins produced by bacteria to defend against foreign DNA by cutting it into nonfunctional pieces.
  8. [8]
    Hamilton Smith, MD - J. Craig Venter Institute
    In 1970, he discovered the first Type II restriction enzyme, HindII. He received the Nobel Prize in Medicine in 1978 for that work. From 1975 to 1998, he ...
  9. [9]
    [13] Purification and properties of HindII and HindIII endonucleases ...
    This chapter discusses the purification and properties of HindII and HindIII endonucleases from Haemophilus influenzae Rd. Haemophilus influenzae Rd is the ...
  10. [10]
    [PDF] Hamilton O. Smith - Nobel Lecture
    At the same time, Meselson and Yuan ('7) reported more extensive experiments with a highly purified restriction endonuclease from E. coli K. Using sucrose ...
  11. [11]
    Highlights of the DNA cutters: a short history of the restriction enzymes
    Preliminary descriptions of the phenomenon of R-M were published by Luria and Human (1952) (13), Anderson and Felix (1952) (14) and Bertani and Weigle (1953) ( ...
  12. [12]
    Herbert W. Boyer and Stanley N. Cohen | Science History Institute
    In 1972 researchers, including Boyer, realized that the enzyme EcoRI, which had actually been discovered in Boyer's UCSF lab, cut DNA in such a way that the ...
  13. [13]
    The Nobel Prize in Physiology or Medicine 1978 - NobelPrize.org
    The Nobel Prize in Physiology or Medicine 1978 was awarded jointly to Werner Arber, Daniel Nathans and Hamilton O. Smith for the discovery of restriction ...
  14. [14]
    The Nobel Prize in Physiology or Medicine 1978 - Press release
    Smith verified Arber's hypothesis with a purified bacterial restriction enzyme and showed that this enzyme cuts DNA in the middle of a specific symmetrical ...
  15. [15]
  16. [16]
    REBASE: a database for DNA restriction and modification: enzymes ...
    The previous description of REBASE in the 2015 NAR Database Issue (1) described 6,804 biochemically or genetically characterized restriction-modification ...
  17. [17]
    CoCoNuTs: A diverse subclass of Type IV restriction systems ...
    Feb 11, 2024 · These CoCoNut systems are predicted to target RNA as well as DNA and to utilise defence mechanisms with some similarity to type III CRISPR-Cas ...
  18. [18]
    Defense and anti-defense mechanisms of bacteria and ...
    In this paper, we summarize recent research on bacterial phage resistance and phage anti-defense mechanisms, as well as collaborative win-win systems involving ...
  19. [19]
    Bacteriophage strategies for overcoming host antiviral immunity
    In these systems, restriction endonucleases (REases) are used to cut invading foreign DNA at specific recognition sites, while the host DNA would not be ...Introduction · Inactivation of the immune... · Genes modifications or... · Conclusion
  20. [20]
    Systematic evasion of the restriction-modification barrier in bacteria
    May 16, 2019 · Found in ∼90% of sequenced bacterial genomes, RM systems enable bacteria to distinguish self- from nonself-DNA via two enzymatic activities: a ...
  21. [21]
    Restriction-modification systems have shaped the evolution and ...
    Within R-M systems, Type II are the most abundant, present in 39.2% of bacterial genomes ( ... We use percentage amino acid identity scores of 50% for ...
  22. [22]
    Bacterial restriction-modification systems: mechanisms of defense ...
    Here, we review the molecular basis of R-M systems and the diverse strategies used by bacteriophages to evade these defenses. Furthermore, we highlight the co- ...
  23. [23]
    Effects of mutations in phage restriction sites during escape from ...
    Dec 13, 2017 · We show that while mutation effects at different restriction sites are unequal, they are independent. As a result, the probability of bacteriophage escape ...
  24. [24]
    Effects of mutations in phage restriction sites during escape ... - NIH
    Dec 13, 2017 · Our results demonstrate that point mutations in phage restriction sites can substantially increase the probability of phages escaping ...Missing: adaptation | Show results with:adaptation
  25. [25]
    Review Evolution and ecology of anti-defence systems in phages ...
    Jan 6, 2025 · In this review, we describe the diverse strategies that phage and plasmid anti-defence systems employ to escape host defence systems.
  26. [26]
    [PDF] Restriction-Modification and CRISPR-Cas Systems
    Generally, these systems provide defense by coordinating the activities of two distinct enzymes: a restriction endonuclease and a methyltransferase. Both ...
  27. [27]
    [PDF] Overcoming DNA Restriction-Modification systems to enable ...
    Of the four classes of RM systems, Types I, II, and III are typically comprised of two primary components, a restriction enzyme and a methyltransferase. The ...
  28. [28]
    Diverse Functions of Restriction-Modification Systems in Addition to ...
    R-M systems are classified mainly into four different types based on their subunit composition, sequence recognition, cleavage position, cofactor requirements, ...
  29. [29]
    The Role of DNA Restriction-Modification Systems in the Biology of ...
    Jan 22, 2016 · The RE degrades DNA molecules possessing unmethylated target sequences ... Methylation and DNA cleavage reactions occur simultaneously.
  30. [30]
    A mobile restriction–modification system provides phage defence ...
    Mar 14, 2022 · Therefore, the bacterial genome is protected from cleavage due to methylation, whereas unmethylated intruders are degraded by the REase (22).
  31. [31]
    Restriction-Modification Systems as Mobile Epigenetic Elements
    DNA methyltransferases methylate specific bases in recognition sequences and generate three types of modified base: 5-methylcytosine (m5C), N4-methylcytosine ( ...
  32. [32]
    Bacterial N4-methylcytosine as an epigenetic mark in eukaryotic DNA
    Feb 28, 2022 · In eukaryotes, modifications predominantly involve C5-methylcytosine (5mC) and occasionally N6-methyladenine (6mA), while bacteria frequently ...
  33. [33]
    Evolutionary ecology of prokaryotic innate and adaptive immune ...
    Oct 20, 2020 · Phase-variable methylation and epigenetic regulation by type I restriction-modification systems. FEMS Microbiology Reviews. 2017;41:S3–S15 ...
  34. [34]
    [PDF] The phage-host arms-race: Shaping the evolution of microbes
    Another interesting example of exaptation of RM systems is evident in phase- variable Type III RM systems [94], whose genes can be reversibly inactivated due to.<|control11|><|separator|>
  35. [35]
    Improving the Transformation Efficiency of Synechococcus sp. PCC ...
    Aug 4, 2025 · transformation efficiency · genetic engineering · DNA methylation · restriction modification ... To determine if DNA methylation would protect ...
  36. [36]
    In Vitro CpG Methylation Increases the Transformation Efficiency of ...
    These results indicate that lp56, but not BBE02, is required for in vitro CpG methylation to have a dramatic effect on transformation efficiency and suggest ...
  37. [37]
    How to Use Restriction Enzymes: A Resource Guide
    In 1968, Arber and Linn demonstrated nuclease activity of Eco B restriction enzyme and Meselson and Yuan purified a similar enzyme from E. ... first base ...
  38. [38]
  39. [39]
    Structure of the DNA-Eco RI Endonuclease Recognition Complex at ...
    Arg200 forms two hydrogen bonds with guanine while Glu144 and Arg145 form four hydrogen bonds to adjacent adenine residues. These interactions discriminate the ...
  40. [40]
    Structure and function of type II restriction endonucleases
    Typically ∼15–20 hydrogen bonds are formed between a dimeric restriction enzyme and the bases of the recognition sequence, in addition to numerous van der Waals ...
  41. [41]
    Restriction Endonuclease - an overview | ScienceDirect Topics
    The first restriction endonucleases were discovered at Cold Spring Harbor Laboratory, but they were first commercialized by the company New England Biolabs in ...
  42. [42]
    Unidirectional translocation from recognition site and a necessary ...
    The sequence context of recognition sites has been suggested to influence the site of cleavage by Type III restriction enzymes (13). We have analysed the ...
  43. [43]
    Restriction enzymes & DNA ligase (article) - Khan Academy
    Many restriction enzymes make staggered cuts, producing ends with single-stranded DNA overhangs. However, some produce blunt ends. DNA ligase is a DNA-joining ...
  44. [44]
    Restriction Enzymes - Science Primer |
    These are referred to as blunt end cuts. An example of an enzyme that leaves blunt ends is SmaI. SmaI recognizes the six base sequence: CCCGGG. When cut the ...
  45. [45]
    Restriction Enzyme - an overview | ScienceDirect Topics
    Cleavage of DNA by a restriction enzyme may be blocked or impaired when a particular base in the recognition site is modified. For example, MspI and HpaII ...
  46. [46]
    Structure and function of type II restriction endonucleases - PMC
    Type IIB restriction endonucleases cleave DNA at both sides of the recognition sequence, for example BcgI which recognizes an asymmetric sequence, or BplI which ...
  47. [47]
    Site-specific DNA transesterification catalyzed by a restriction enzyme
    In the presence of Mg2+, type II restriction endonucleases (REases) hydrolyze specific phosphodiester bonds in DNA to yield 5′-phosphate and 3′-hydroxyl termini ...
  48. [48]
    EcoRV restriction endonuclease: a catalytic role - Oxford Academic
    Phosphodiester hydrolysis by EcoRV proceeds with inversion of configuration at the target phosphorus and thus involves the direct attack of a water molecule in ...
  49. [49]
    DB code: S00403 - EzCatDB
    According to the paper [6], cleavage of DNA by restriction endonucleases yields 3'-OH and 5'-phosphate ends, where hydrolysis of the phosphodiester bonds by ...
  50. [50]
    Utilization of Type IIS Endonucleases in Molecular Cloning
    The MfeI site (CAATTC) is compatible with the EcoRI site (GAATTC); both enzymes create the 5'- AATT overhang. No enzymes however, are compatible with HindIII ( ...
  51. [51]
  52. [52]
  53. [53]
    What is restriction enzyme star activity - Promega Corporation
    This loss of fidelity or increase in cleavage at sites similar to the cognate site is commonly referred to as star activity. A number of reaction parameters can ...
  54. [54]
    The Fidelity Index provides a systematic quantitation of star activity ...
    Many restriction endonucleases show relaxed sequence recognition, called star activity, as an inherent property under various digestion conditions including the ...
  55. [55]
    Type I restriction enzymes and their relatives - Oxford Academic
    Type I restriction enzymes (REases) are large pentameric proteins with separate restriction (R), methylation (M) and DNA sequence-recognition (S) subunits.
  56. [56]
    Type I Restriction Systems: Sophisticated Molecular Machines (a ...
    A type I RM enzyme behaves like a molecular motor, translocating vast stretches of DNA towards itself before eventually breaking the DNA molecule.<|control11|><|separator|>
  57. [57]
    On the structure and operation of type I DNA restriction enzymes
    Type I DNA restriction enzymes are large, molecular machines possessing DNA methyltransferase, ATPase, DNA translocase and endonuclease activities.
  58. [58]
    Type II restriction endonucleases: structure and mechanism - PubMed
    Abstract. Type II restriction endonucleases are components of restriction modification systems that protect bacteria and archaea against invading foreign DNA.Missing: paper | Show results with:paper
  59. [59]
    Type II restriction endonucleases—a historical perspective and more
    Five years after EcoRI was purified to homogeneity in 1976, the amino acid sequences of the EcoRI REase and MTase were determined by cloning the EcoRI R–M ...
  60. [60]
    Structures, activity and mechanism of the Type IIS restriction ... - NIH
    Mar 29, 2023 · Type IIS restriction endonucleases contain separate DNA recognition and catalytic domains and cleave their substrates at well-defined distances outside their ...Missing: paper | Show results with:paper
  61. [61]
    Structural and evolutionary classification of Type II restriction ...
    Type II enzymes that exhibit structural and functional peculiarities (requirement of more than one target site for cleavage, cleavage at a distance from the ...
  62. [62]
    Restriction Endonuclease Basics | Thermo Fisher Scientific - ES
    Today about 4,000 restriction enzymes have been characterized, and over 600 of those are commercially available. REBASE is a useful, browsable resource for ...
  63. [63]
    Type II restriction endonucleases—a historical perspective and more
    Number of publications for EcoRI and EcoRV per year from 1972 ... Isolation of gram quantities of EcoRI restriction and modification enzymes from an overproducing ...
  64. [64]
    Type III restriction-modification enzymes: a historical perspective
    Type III R–M enzymes need to interact with two separate unmethylated DNA sequences in inversely repeated head-to-head orientations for efficient cleavage.
  65. [65]
    Type III restriction-modification enzymes: a historical perspective
    This review focuses on the small group of well-characterized Type III R–M enzymes (Table 1), and most knowledge comes from studies on the enzymes from the ...Missing: Loenen | Show results with:Loenen
  66. [66]
    [PDF] Mechanistic insights into type III restriction enzymes - IMR Press
    This review will discuss the biochemical and biophysical properties of the Type III restriction enzymes and highlight their mechanism of action. 2.1. Subunit ...
  67. [67]
    Structural Insights into the Assembly and Shape of Type III ...
    To begin to understand the action mechanism by Type III restriction enzymes at a more structural level, we undertook small angle X-ray scattering (SAXS) and ...
  68. [68]
  69. [69]
    Type III restriction enzymes cleave DNA by long-range ... - PNAS
    Apr 30, 2010 · Type III restriction enzymes cleave DNA by long-range interaction between sites in both head-to-head and tail-to-tail inverted repeat.
  70. [70]
    The other face of restriction: modification-dependent enzymes
    Type IV enzymes recognize modified DNA with low sequence selectivity and have emerged many times independently during evolution.
  71. [71]
    Biology of host-dependent restriction-modification in prokaryotes
    Aug 26, 2025 · Host-dependent (HD) restriction-modification (RM) systems (HDRM) are present in most prokaryotic genomes (8). They regulate the fate of entering ...
  72. [72]
    A diverse subclass of Type IV restriction systems predicted to target ...
    Type IV restriction enzymes contain at least two components: 1) a dedicated specificity domain that recognizes modified DNA, and 2) an endonuclease domain that ...
  73. [73]
    A Type IV modification dependent restriction nuclease that targets ...
    The McrBC enzyme complex utilizes GTP-driven DNA translocation, and a length-determining looping mechanism to create non-specific fragments of DNA of a ...Missing: examples | Show results with:examples
  74. [74]
    Modified DNA substrate selectivity by GmrSD-family Type IV ...
    Sep 4, 2025 · Type IV restriction enzymes have evolved to recognize and cleave modified DNA. We have recently characterized and solved the first structure for ...
  75. [75]
    Modified DNA substrate selectivity by GmrSD-family Type IV ... - NIH
    Sep 4, 2025 · Type IV restriction enzymes have evolved to recognize and cleave modified DNA. We have recently characterized and solved the first structure for ...
  76. [76]
    Rational engineering of type II restriction endonuclease DNA ...
    Using simple and predictable mutagenesis in this family it is now possible to create hundreds of unique new type II restriction endonuclease specificities.
  77. [77]
    A combinatorial approach to create artificial homing endonucleases ...
    In this report, we describe the first artificial HEs whose specificity has been entirely redesigned to cleave a naturally occurring sequence.
  78. [78]
    Directed Evolution of Restriction Endonuclease BstYI to Achieve ...
    Our objective was to investigate the substrate specificity of BstYI by attempting to increase the specificity to recognition of only AGATCT.
  79. [79]
    Crystal Structure and Directed Evolution of Specificity of NlaIV ...
    Here, we present the results of a directed evolution approach to the engineering of a dimeric, blunt end cutting restriction enzyme NlaIV (GGN/NCC).
  80. [80]
    TALENs: a widely applicable technology for targeted genome editing
    Recently, transcription activator-like effector nucleases (TALENs) have rapidly emerged as an alternative to ZFNs for genome editing and introducing targeted ...
  81. [81]
    TALENs—an indispensable tool in the era of CRISPR: a mini review
    Aug 21, 2021 · This review provides significant insights into the pros and cons of the two most popular genome editing tools TALENs and CRISPRs.Genome Editing · Crispr · Talen
  82. [82]
    Recent advances in the use of ZFN-mediated gene editing for ... - NIH
    We reported the creation of ZFNs by fusing modular zinc finger proteins (ZFPs) to the non-specific cleavage domain of FokI restriction enzyme in 1996 [3]. ZFPs ...
  83. [83]
    Homing endonucleases from mobile group I introns: discovery to ...
    Mar 3, 2014 · Homing endonucleases are highly specific DNA cleaving enzymes that are encoded within genomes of all forms of microbial life including phage and eukaryotic ...
  84. [84]
    Directed Evolution and Substrate Specificity Profile of Homing ...
    Together these tools were used to successfully evolve mutant I-SceI homing endonucleases with altered DNA cleavage specificities. The most highly evolved enzyme ...
  85. [85]
    Engineering altered protein–DNA recognition specificity
    Here, we summarize the creation of novel DNA specificities for zinc finger proteins, meganucleases, TAL effectors, recombinases and restriction endonucleases.
  86. [86]
    FokI dimerization is required for DNA cleavage - PNAS
    3. These plots were used to calculate the initial velocity (vo = d[P]/dt) of each reaction catalyzed by a given FokI concentration ...Missing: equation | Show results with:equation
  87. [87]
    Off‐target effects of engineered nucleases - Yee - 2016 - FEBS Press
    May 21, 2016 · As dimerization of the Fok1 nuclease domain is required for the generation of DSB, two ZFNs are designed to recognize the genomic target with ...
  88. [88]
    megaTALs: a rare-cleaving nuclease architecture for therapeutic ...
    This architecture allows the generation of extremely active and hyper-specific compact nucleases that are compatible with all current viral and nonviral cell ...
  89. [89]
    Halting Recombinant Adeno-Associated Virus Transgene ...
    Our findings underscore the potential of meganucleases as a viable safety mechanism in rAAV gene therapies, marking a significant advance toward safer long-term ...Missing: compact | Show results with:compact
  90. [90]
    A nomenclature for restriction enzymes, DNA methyltransferases ...
    They proposed that the enzyme names should begin with a three-letter acronym in which the first letter was the first letter of the genus from which the enzyme ...
  91. [91]
    How restriction enzymes became the workhorses of molecular biology
    Simultaneously, Matt Meselson and Bob Yuan also isolated a restriction enzyme from Escherichia coli K ( 10). The systems that Arber and Meselson characterized ...
  92. [92]
    Structure of restriction endonuclease BamHI phased at 1.95 å ...
    The BamHI recognition sequence differs by only one base pair in each half site from the EcoRI sequence 5′ -GAATTC-3′ .
  93. [93]
    Restriction Endonucleases 101: Basics to Golden Gate Cloning
    Sep 19, 2025 · Sticky ends: These are single-stranded overhangs created when restriction enzymes make staggered cuts in the DNA. · Blunt ends: These occur when ...<|control11|><|separator|>
  94. [94]
    An efficient method for blunt-end ligation of PCR products - PubMed
    In the example presented, PCR products were blunt-end ligated to a SmaI-cut vector, in the presence of SmaI endonuclease.
  95. [95]
  96. [96]
    Binding of two zinc finger nuclease monomers to two specific sites is ...
    The two-site model proposed for the mechanism of DNA cleavage by FokI restriction enzyme [32] best explains the double-strand cleavage by ZFNs as well. First, ...
  97. [97]
    A Programmable Dual-RNA–Guided DNA Endonuclease ... - Science
    Jun 28, 2012 · Efficient genome editing of bovine in vitro derived zygotes via Cas9 RNP-electroporation using extended-stored bovine ovaries ...
  98. [98]
    CRISPR/Cas9 therapeutics: progress and prospects - Nature
    Jan 16, 2023 · By changing the nucleotide sequence of a small segment of guide RNA, CRISPR/Cas9 allows the accurate targeting of almost any desired genomic ...
  99. [99]
    Construction of Biologically Functional Bacterial Plasmids In Vitro
    Abstract. The construction of new plasmid DNA species by in vitro joining of restriction endonuclease-generated fragments of separate plasmids is described.
  100. [100]
  101. [101]
    11.1: Recombinant DNA and Gene Cloning - Biology LibreTexts
    Mar 17, 2025 · If we treat any other sample of DNA, e.g., from human cells, with EcoRI, fragments with the same sticky ends will be formed.Making Recombinant DNA... · Examples of Plasmids · pAMP · Ligation Possibilities
  102. [102]
    Construction and characterization of new cloning vehicles. II. A ...
    The vector pBR322 was constructed in order to have a plasmid with a single PstI site, located in the ampicillin-resistant gene (Apr), in addition to four unique ...