Restriction Endonuclease Enzymes- Origin, Types and Function

Restriction Endonuclease Enzymes- Origin, Types and Function

Figure: Restriction Endonucleases acting as molecular scissor

1. What are Restriction Endonucleases (Restriction Endonuclease Definition)?

Inside the intricate world of DNA, there exists a group of enzymes known as endonucleases, which possess the remarkable ability to cleave or make cuts within DNA molecules. Some endonucleases have a preference for cleaving just one of the two strands in a DNA duplex, akin to creating tiny nicks in the DNA strand. For e.g., S1 nuclease derived from Aspergilla ary, exhibit this behavior. In contrast, a different class of endonucleases has the power to cut both strands of DNA molecules.

These endonucleases, like deoxyribonuclease I (DNase I) obtained from cow pancreas, do so at random sites along the DNA molecule. However, among the diverse range of endonucleases, there are the precise “restriction endonucleases.” These enzymes are known for their ability to cleave DNA molecules solely within or near specific sites with distinct base sequences.

These particular sites are termed recognition sequences or recognition sites. Remarkably, each type of restriction endonuclease recognizes its own distinct recognition sequence. There are over 3,600 restriction endonucleases identified, showcasing more than 250 distinct specificities. Out of these, over 3,000 have undergone comprehensive research, and a considerable number of them—more than 800—are readily accessible for purchase. These enzymes play a routine role in modifying DNA within laboratories and serve as an essential instrument in the realm of molecular cloning.

Types of nuclease activity

Figure: Types of nuclease activity (https://www.differencebetween.com/difference-between-endonuclease-and-vs-exonuclease/, CC BY-SA 4.0 <https://creativecommons.org/licenses/by-sa/4.0>, via Wikimedia Commons)

1.1. Definition of Restriction Endonucleases

“Restriction Endonucleases, also known as restriction enzymes or molecular scissors, restriction endonucleases are enzymes that cleave DNA molecules at specific recognition sites within the DNA sequence. These enzymes play a crucial role in molecular biology, allowing scientists to precisely cut DNA for various applications, such as DNA analysis, gene cloning, and genetic engineering. Their discovery revolutionized the field and paved the way for recombinant DNA technology, enabling the manipulation and modification of genetic material”

1.2. History of Restriction Endonucleases

The captivating term “restriction enzyme” owes its origin to the captivating world of phage λ—a virus that invades bacteria—and the intriguing dance of host-controlled restriction and modification of these bacterial phages, or bacteriophages. Delving into the early 1950s laboratories of Salvador Luria, Jean Weigle, and Giuseppe Bertani, this phenomenon came to light. Their pioneering work revealed that bacteriophage λ, thriving magnificently in one strain of Escherichia coli (E. coli C, for instance), can experience a significant plunge in yield, up to a staggering 3-5 orders of magnitude, when cultivated in another strain, like E. coli K.

This curiously restricting host, E. coli K in this case, possesses a remarkable talent for dampening the biological vigor of phage λ. Remarkably, if a phage flourishes in one strain, its growth becomes curtailed in others. This intriguing narrative took a fascinating turn in the 1960s, thanks to the groundbreaking work of Werner Arber and Matthew Meselson. They unearthed the truth behind this restriction: an enzymatic cleavage of phage DNA. The term “restriction enzyme” was thus born.

1.3. Who Discovered Restriction Endonuclease?

In their quest, Arber and Meselson examined type I restriction enzymes, which execute random DNA cleavage away from the recognition site. In a triumphant moment of scientific discovery, Hamilton O. Smith, Thomas Kelly, and Kent Wilcox isolated and scrutinized the inaugural type II restriction enzyme, HindII, extracted from the bacterium Haemophilus influenzae in 1970. These type II enzymes, profoundly valued in laboratory settings, cleave DNA right at their recognition sequence, becoming a staple tool in molecular biology. A pivotal breakthrough emerged when Daniel Nathans and Kathleen Danna unveiled the precise fragments resulting from simian virus 40 (SV40) DNA cleavage by restriction enzymes. This method of cleavage paved the way for mapping DNA using polyacrylamide gel electrophoresis, a groundbreaking revelation that expanded the horizons of restriction enzymes. This watershed moment was rewarded with the 1978 Nobel Prize for Physiology or Medicine, bestowed upon Werner Arber, Daniel Nathans, and Hamilton O. Smith for their awe-inspiring contribution to restriction enzyme discovery and characterization.

The enchanting journey into restriction enzymes brought forth a powerful elixir: recombinant DNA technology. This innovative marvel, arising from the manipulation of DNA, has unveiled a treasure trove of applications, including the monumental achievement of producing proteins like human insulin on a large scale, offering hope to countless diabetic patients. As we delve into the realm of restriction enzymes, we unearth the secret to shaping the very fabric of life—DNA—unlocking endless possibilities for science and medicine.

Table: History of Restriction Endonucleases

AspectDetails
Discovery of Restriction EnzymesOriginated from the study of bacteriophage λ and host-controlled restriction and modification. Pioneering work by Luria, Weigle, and Bertani revealed λ phage’s sensitivity to different E. coli strains.
Arber and Meselson’s Type I Enzyme ExplorationExamined type I restriction enzymes causing random DNA cleavage away from the recognition site. Arber and Meselson contributed to understanding the restriction phenomenon.
Discovery of Type II Restriction EnzymesHamilton O. Smith, Thomas Kelly, and Kent Wilcox isolated the first type II restriction enzyme, HindII, from Haemophilus influenzae in 1970. Type II enzymes became crucial tools in molecular biology due to their precise cleavage at recognition sites.
DNA Cleavage Mapping and ElectrophoresisDaniel Nathans and Kathleen Danna’s breakthrough in simian virus 40 (SV40) DNA cleavage using restriction enzymes led to DNA mapping using polyacrylamide gel electrophoresis.
Nobel Prize RecognitionWerner Arber, Daniel Nathans, and Hamilton O. Smith received the 1978 Nobel Prize in Physiology or Medicine for their significant contribution to restriction enzyme discovery and characterization.
Recombinant DNA Technology and ApplicationsThe journey into restriction enzymes led to recombinant DNA technology, enabling groundbreaking applications like producing human insulin through DNA manipulation.

Table: Timeline Showing Major Milestones in the History of Restriction Enzymes

YearMilestone
1950s-1960sSalvador Luria, Jean Weigle, and Giuseppe Bertani discover that bacteriophage λ’s yield varies significantly when cultivated in different E. coli strains.
Werner Arber and Matthew Meselson begin exploring Type I restriction enzymes and their random DNA cleavage.
1970Hamilton O. Smith, Thomas Kelly, and Kent Wilcox isolate the first Type II restriction enzyme, HindII, from Haemophilus influenzae.
Daniel Nathans and Kathleen Danna uncover the precise fragments resulting from simian virus 40 (SV40) DNA cleavage by restriction enzymes.
1978Werner Arber, Daniel Nathans, and Hamilton O. Smith receive the Nobel Prize in Physiology or Medicine for their contributions to restriction enzyme discovery.
1980sDiscovery of various subtypes within Type II restriction enzymes.
Emergence of Type III restriction enzymes’ unique properties, including DNA cleavage and methylation functions.
1990sExploration of Type IV restriction enzymes that target modified DNA variants.
Continued research into DNA modification and methylation patterns.
2000sIntroduction of engineered restriction enzymes, such as Zinc Finger Nucleases (ZFNs) and TAL Effector Nucleases (TALENs), for genome editing.
Pioneering applications of CRISPR-Cas9 system for precise genome editing.
2010sRefinement of CRISPR-Cas9 technology for diverse genetic manipulations.
Rapid expansion of CRISPR-based genome editing tools and applications.
Advancements in understanding the structure and function of various restriction enzyme subtypes.
2020sOngoing research into novel types of restriction enzymes and their potential applications.
Continued refinement of genome editing techniques and applications in biotechnology, medicine, and scientific research.

2. Origin of Restriction Enzymes

The world of molecular biology unveils the intriguing source of restriction enzymes – bacterial cells. These enzymes earned their name, “restriction enzymes,” due to their pivotal role in shielding bacteria from certain viruses called bacteriophages. Inside bacterial cells, these enzymes orchestrate a tactical defense, selectively dismantling viral DNA while keeping the bacterial DNA untouched. This defense strategy preserves the bacterium’s integrity.

When foreign genetic material infiltrates a bacterial cell, the restriction enzyme stands guard. It doesn’t hesitate to identify and slice the invader’s DNA at multiple points along its structure. This precision defense mechanism forms a barrier against the invasion of foreign genetic content, securing the bacterial domain. Remarkably, each type of bacterium possesses its own collection of restriction enzymes, each tailored to recognize and sever specific DNA sequences.

Delving into the evolutionary history, it is hypothesized that restriction enzymes share a common ancestry. They then expanded their influence through a process known as horizontal gene transfer, which entails the exchange of genetic material between different organisms. In an intriguing twist, emerging evidence proposes that restriction endonucleases evolved as self-serving genetic entities. These enzymes harbor their own interests and actively propagate themselves within a genome.

In a nutshell, restriction enzymes emerge from the realm of bacterial cells, offering steadfast protection against viral onslaught by dismantling foreign DNA. Their uncanny ability to target and cleave specific DNA sequences grants them a pivotal role in molecular biology and genetic exploration.

3. Nomenclature (Naming) of Restriction Endonuclease

Figure: Restriction Enzyme Nomenclature

Ever wondered what those big names for restriction enzymes mean? Let’s break the restriction enzyme nomenclature down into simple parts. Think of this nomenclature like a puzzle made up of meaningful pieces that scientists have put together. Each piece gives us a clue about the enzyme’s source and what it does.

First Letters Reveal the Genus: The first letter in the name is like a clue that tells us the genus, which is like a family name for bacteria. It’s written in capital letters, so it stands out. For example, “Eco” is the hint for Escherichia Coli bacteria.

Special Italics for Species: After the genus hint, there are two more letters in italics. These letters represent the species of bacteria. They narrow down the family to a specific member. Like “Hin” in HindIII means Haemophilus influenzae bacteria.

Subscripts Add Details: Sometimes there are small letters below the main letters. These tell us extra details, like the specific type of bacteria or even if the enzyme comes from a plasmid. Think of them as extra notes to the main tune. For instance, “Ecox” means the enzyme comes from a plasmid of Escherichia Coli.

Roman Numerals for Different Enzymes: When a bacteria has more than one enzyme, Roman numerals come into play. These numbers show which enzyme is which. It’s like giving each enzyme a special name tag. For example, “Hinll” and “Hin” show there are different versions of Haemophilus influenzae enzymes.

The “R” Prefix: The Enzyme Hint: Before the enzyme name, you’ll often find an “R.” It’s like a signal that this name is for a restriction enzyme. It’s added to avoid confusion with similar molecules. Keeping Things Simple, In real life, we make things easier. The small letters for bacteria types are usually written next to the main name. And sometimes, we skip the “R” if everyone knows we’re talking about a restriction enzyme.

4. Recognition Site/ Sequences of Restriction Endonucleases

Figure: HindIII restriction nucleases cleaving at recognition site (Image Source: Helixitta, CC BY-SA 4.0 <https://creativecommons.org/licenses/by-sa/4.0>, via Wikimedia Commons)

In the realm of restriction enzymes, recognition sites are like specialized addresses that these molecular scissors recognize and latch onto within DNA strands. These spots are special because they showcase a particular symmetry pattern called palindromes. These palindromes are sequences that read the same way forwards and backwards.

For example, let’s delve into the palindrome GAATTC. On one side of the DNA strand, it reads in a 5′ to 3′ direction, while on the other side, it flips and reads in a 3′ to 5′ direction. But if we read both strands in the 5′ to 3′ way, the sequence remains unchanged. This gives us the recognition site:

5′ GAATTC 3′ 

3′ CTTAAG 5′

Inside this palindrome, there’s a focal point of symmetry, often right in the middle between specific base pairs (like AT/AT in our example). This symmetrical point is crucial because this is where the restriction enzymes take out their molecular scissors and cut DNA.

Now, let’s talk about the cutting styles. Some enzymes chop DNA right at the symmetry center, creating what’s known as blunt ends. This means both strands get chopped at the same position, leaving no dangling bits.

But there’s another type of cut that’s even more fascinating. Some enzymes snip DNA between the same two bases but not directly at the symmetry center. This creates a staggered cut, often called a “sticky” break. The ends that result have short, single-stranded tails that fit like puzzle pieces with each other. These puzzle-piece tails are the famous “sticky ends,” and they’re amazing at matching up with complementary ends from other DNA pieces. This unique trait makes them a treasure trove for molecular biologists, especially during DNA manipulation.

So, in a nutshell, recognition sites are like palindromic addresses where restriction enzymes make their precision cuts. These cuts can produce either blunt ends or the intriguing sticky ends, depending on the enzyme’s preferences. This remarkable ability of restriction enzymes is like having super-smart molecular scissors that scientists can wield with incredible precision.

5. Types of Restriction Endonucleases

Classification of naturally occurring restriction endonucleases, commonly known as restriction enzymes, encompasses five primary types (Type I, Type II, Type III, Type IV, and Type V), distinguished by factors like their constitution, cofactor prerequisites, nature of target sequences, and the position of cleavage sites. It’s important to acknowledge that the analysis of DNA sequences has unveiled a broader spectrum of diversity, hinting at the possibility of further unexplored types within this category.

Outlined below is a compendium elucidating the distinct categories of restriction enzymes:

Type I Restriction Endonucleases: These enzymes effectuate DNA cleavage at locations remote from their recognition sequences. Their optimal function necessitates both ATP and S-adenosyl-L-methionine as essential cofactors. Type I enzymes possess multifaceted attributes, embodying restriction digestion as well as methylase activities within their structure.

Type II Restriction Endonucleases: Characterized by their capacity to cleave DNA at or proximate to their recognition sites, Type II enzymes predominantly depend on magnesium as a cofactor. Diverging from Type I enzymes, Type II enzymes are specialized single-function agents that operate independently from methylase activity.

Type III Restriction Endonucleases: These enzymes execute DNA cleavage in close proximity to their recognition sequences. While they rely on ATP (albeit non-hydrolyzed) and can be prompted by S-adenosyl-L-methionine, the latter is not an imperative component for their functionality. Type III enzymes operate in concert with a modification methylase within a composite molecular structure.

Type IV Restriction Endonucleases: Targeting modified DNA variants like methylated, hydroxymethylated, and glucosyl-hydroxymethylated DNA, Type IV enzymes discern specific modifications along the DNA molecule and incise at corresponding sites.

Type V Restriction Endonucleases: Type V enzymes harness guide RNAs (gRNAs) to identify their designated target sequences. The gRNA serves as a navigational tool, directing the enzyme toward the precise DNA sequence, thus enabling targeted cleavage at the desired position.

Table: Types of Restriction Endonucleases

Type of Restriction Restriction EndonucleasesKey Characteristics of Restriction EndonucleasesRepresentative Examples of Restriction EndonucleasesNotable Features of Restriction Endonucleases
Type I Restriction Endonucleases– Cleaves DNA at locations remote from recognition sequences.- Requires ATP and S-adenosyl-L-methionine as cofactors.- Has restriction and methylase activities.EcoKIRecognizes complex recognition sequences. Operates with DNA translocation mechanism.
Type II Restriction Endonucleases– Cleaves DNA at or proximate to recognition sites.- Depends on magnesium as a cofactor.- Single-function enzymes.HindII, EcoRI, BamHIProvides precision cleavage at recognition sites. Commonly used in molecular biology techniques.
Type III Restriction Endonucleases– Cleaves DNA close to recognition sequences.- Utilizes ATP and S-adenosyl-L-methionine (non-hydrolyzed) as cofactors.- Works with a modification methylase in a complex.EcoP15Involves recognition of two inverse, non-palindromic sequences. Uses two essential cofactors for DNA methylation and restriction.
Type IV Restriction Endonucleases– Targets modified DNA variants like methylated, hydroxymethylated, and glucosyl-hydroxymethylated DNA.- Cuts at corresponding sites of modifications.McrBC, Mrr (E. coli)Detects and cleaves modified DNA. Differentiates between modified and unmodified DNA, serving as a defense mechanism.
Type V Restriction Endonucleases– Uses guide RNAs (gRNAs) to target specific DNA sequences.- Employs gRNA to direct enzyme to precise DNA sequence for cleavage.CRISPR-Cas9 systemOffers programmable DNA targeting. Allows for highly specific genome editing.
Artificial Restriction Endonucleases– Engineered enzymes with DNA-binding and nuclease parts.- Designed for precision genome editing.- Flexible in target sequence recognition.Zinc Finger Nucleases (ZFNs), TAL Effector Nucleases (TALENs), CRISPR-Cas9Customizable DNA-binding parts combined with nuclease activity. Pioneering tools in genome editing and manipulation.
PNAzymesEngineered ribonucleases for RNA cleavage.

5.1. Type I Restriction Endonucleases

Emerged as the vanguards of their kind, being the very first class of restriction enzymes unveiled to the scientific world. This significant discovery was made in various strains of Escherichia coli (E. coli), most notably the K-12 and B strains. These enzymes wield a distinctive ability to cleave DNA, albeit at an intriguingly remote and random location—typically a substantial distance of at least 1000 base pairs away from their designated recognition sequence.

What sets the cleavage process of Type I restriction enzymes apart is their DNA translocation mechanism, effectively casting them in the role of molecular motors. These enzymes exhibit an unmatched precision in targeting their recognition sequence, which is a composition of two distinct segments. A specific portion, with 3-4 nucleotides, harmonizes with another section of 4-5 nucleotides. The two portions elegantly intertwine, separated by a non-specific spacer that spans roughly 6-8 nucleotides.

Type I restriction enzymes are true polymaths, seamlessly juggling between two integral functions—restriction digestion and modification. Their prowess in this dual role hinges on the methylation status of the DNA target. In the face of foreign DNA, they enact their cutting action, thwarting potential threats. Alternatively, when dealing with the host’s DNA, they deftly append methyl groups, effectively guarding it against their own cleaving activity—a truly remarkable act of self-preservation.

Behind the scenes of this intricate dance of DNA manipulation lie three essential cofactors—S-adenosyl methionine (AdoMet), hydrolyzed adenosine triphosphate (ATP), and magnesium ions (Mg2+). AdoMet takes center stage in DNA methylation, fortifying the DNA against the enzyme’s cleaving propensity. The hydrolysis of ATP fuels the enzyme’s motor-like translocation, while the magnesium ions orchestrate a seamless performance of this complex biochemical ballet.

Ensemble cast is crucial in this saga, with Type I restriction enzymes comprising three vital subunits: HsdR, HsdM, and HsdS. The HsdR subunit leads the charge, orchestrating the restriction digestion, deftly cleaving the DNA at the predetermined recognition site. HsdM takes up the mantle of DNA modification, skillfully augmenting the host DNA with methyl groups through its methyltransferase activity. Finally, the HsdS subunit takes on a pivotal role, fine-tuning the specificity of the recognition site, while also participating in both DNA cleavage and methyltransferase activities.

In summation, Type I restriction enzymes have earned their place as the trailblazers among restriction enzymes. Their ability to cleave DNA at distant, seemingly random sites and their unique DNA translocation mechanism add to their mystique. These versatile enzymes, guided by specific cofactors, are comprised of subunits that masterfully wield roles in restriction digestion, DNA modification, and the precise recognition of their DNA target. As we delve into the world of molecular machinery, we uncover not only the intricacies of Type I restriction enzymes but also the potential they hold in shaping the landscape of genetic engineering and molecular exploration.

5.2. Type II Restriction Endonucleases

Stand apart from their Type I counterparts in several distinctive ways, marking a paradigm shift in our understanding of DNA manipulation. These enzymes come together in the form of homodimers, an arrangement where identical subunits form a functional unit. Their recognition sites, often seen as the genetic “bullseye,” are commonly undivided and palindromic, typically spanning 4 to 8 nucleotides. Unlike their Type I counterparts, these enzymes possess the unique ability to recognize and precisely cleave DNA at the very same site. Their molecular machinery operates without the need for the energy-rich ATP or AdoMet, as their efficiency is finely tuned by the presence of magnesium ions (Mg2+)—the biochemical conductors of their cellular symphony.

At the heart of their catalytic prowess lies their capacity to cleave the phosphodiester bonds of the DNA double helix. This action can result in either a blunt cut at the center of both DNA strands or a staggered cut that begets single-stranded overhangs known as “sticky ends.” These molecular artisans are commonly referred to as the “workhorses” of restriction enzymes, owing to their widespread availability and extensive use.

However, the narrative of Type II enzymes took an intriguing turn in the late 20th century and the early years of the 21st century. Novel members of this enzyme family emerged, defying the conventional boundaries of their kin. These outliers prompted the scientific community to develop a new subfamily classification, marked by a letter suffix, to better capture their unique features.

Let’s embark on a journey through the subgroups that adorn the landscape of Type II restriction enzymes:

Type IIB Restriction Endonucleases: Among them are illustrious figures like BcgI and BplI. These enzymes, unlike their peers, emerge as multimers, featuring multiple subunits. Their modus operandi involves a symmetrical cleavage on either side of their recognition sequence, effectively excising it from the DNA landscape. To perform their biological magic, they demand not just a single cue, but a duo—both AdoMet and Mg2+—to activate their enzymatic orchestra.

Type IIE Restriction Endonucleases: Here, NaeI takes the spotlight. These enzymes exhibit a penchant for double vision, interacting with not one, but two copies of their recognition sequence. One site takes center stage as the cleavage target, while its counterpart plays the role of an allosteric conductor, orchestrating an enhancement in the efficiency of the cleavage dance.

Type IIF Restriction Endonucleases: In the spotlight stands NgoMIV. This subgroup, too, aligns itself with the “twin” philosophy, engaging with two recognition sequences. However, in this case, the symphony takes on a simultaneous note, as both sites experience cleavage in harmony.

Type IIG Restriction Endonucleases: A unique ensemble led by RM.Eco57I, these enzymes stand as single-subunit entities akin to classical Type II enzymes. However, their performance calls for a partner—AdoMet—as a cofactor to grace the stage. This cofactor lends its weight to the elegance of their activity.

Type IIM Restriction Endonucleases: DpnI takes center stage here, wielding the ability to recognize and cleave DNA that wears the cloak of methylation.

Type IIS Restriction Endonucleases: Enter FokI, an enzyme that cleaves DNA with precision, distanced from non-palindromic, asymmetric recognition sites. This signature move is frequently choreographed for in-vitro cloning techniques like Golden Gate cloning. These enzymes often dance in pairs, as dimers, carrying out their cellular performance.

Type IIT Restriction Endonucleases: This category boasts figures like Bpu10I and BslI. What sets them apart is their composition—two different subunits that collaborate in harmony. Some within this group possess a penchant for palindromic sequences, while others favor asymmetric recognition sites.

In the grand tapestry of scientific exploration, Type II restriction enzymes don’t just play a role; they are the architects of genetic engineering, the maestros of molecular manipulation. Their nuanced characteristics and subgroups have unlocked new avenues for DNA manipulation, gene cloning, and biotechnology.

5.3. Type III Restriction Endonucleases: Guardians of DNA Integrity

DNA manipulation and protection harbors a fascinating cast of characters, each with a distinct role to play. Among these are the enigmatic Type III restriction enzymes, exemplified by EcoP15, which occupy a unique niche in the realm of molecular biology. These enzymes, akin to vigilant sentinels, wield a two-fold power: recognition and defense against foreign DNA intrusion.

Type III restriction enzymes boast an unusual duality—recognizing two separate non-palindromic DNA sequences that stand in an inverse orientation. This distinctive feature sets them apart, guiding their scissors to make a precise cut about 20 to 30 base pairs downstream from the recognition site. It is this cut that serves as a bulwark against genetic invaders.

Comprising multiple subunits, Type III enzymes orchestrate their molecular symphony with the accompaniment of two essential cofactors—AdoMet (S-Adenosyl methionine) and ATP. These cofactors act as the fuel that drives two essential roles: DNA methylation and restriction digestion. In the intricate dance of cellular processes, AdoMet plays a key role in the modification of DNA, offering a protective cloak of methylation to the genetic material. Meanwhile, ATP acts as a conductor, orchestrating the precision of restriction digestion.

The partnership between subunits within Type III enzymes is both elegant and vital. Two characters take the spotlight: Res and Mod. The Mod subunit emerges as a modification methyltransferase, much like the M and S subunits of their Type I counterparts. It bestows its protective embrace upon the DNA, adorning it with methyl groups like the jewels of a crown. Res, on the other hand, plays a role crucial for the integrity of DNA. Although it wields no enzymatic power of its own, it is the catalyst for restriction digestion, ensuring the DNA remains pristine and untampered.

Type III enzymes recognize a distinctive DNA signature—a short, asymmetric sequence spanning 5 to 6 base pairs. Their cut is precise, occurring approximately 25 to 27 base pairs downstream, yielding short, single-stranded protrusions at the 5′ end. But their operation is not solitary; they demand a dual presence of recognition sites, inversely oriented and devoid of methylation, for their precision cut to unfold.

Interestingly, Type III enzymes display a form of DNA methylase activity that is unique—they methylate only one strand of the DNA, specifically at the N-6 position of adenyl residues. This asymmetry in methylation serves as a safeguard, protecting the newly synthesized DNA from the looming threat of restriction digestion.

Within the grand genetic narrative, Type III restriction enzymes are part of the beta-subfamily of N6 adenine methyltransferases—a group characterized by nine defining motifs. These motifs are the language of their activity, including the AdoMet binding pocket (FXGXG) found in motif I, and the catalytic region (S/D/N (PP) Y/F) nestled in motif IV.

In the symphony of prokaryotic DNA restriction-modification systems, Type III enzymes emerge as pivotal players. Their unique blend of methylation and restriction activities, along with their inverted recognition mechanism, enlivens the intricate tale of cellular guardianship. Each cut they make, every methyl group they place, is a note in the symphony of cellular harmony—a harmonious interplay of molecules that safeguards the sanctity of DNA in a world brimming with genetic intricacies.

5.4. Type IV Restriction Endonucleases: Guardians of Genetic Identity

In the intricate tapestry of molecular biology, Type IV restriction enzymes emerge as unique sentinels, primed to recognize and safeguard the integrity of modified DNA. Their role, exemplified by the McrBC and Mrr systems of E. coli, unveils a specialized defense strategy within the bacterial realm.

Distinct Recognition of Modified DNA

Type IV restriction enzymes form a distinct class, finely attuned to the nuances of DNA modification, particularly methylation. Their recognition mechanism transcends the norm, as they don’t seek out specific DNA sequences but rather specific modifications on the DNA molecule. This feature serves as a powerful marker, distinguishing between the host’s own DNA and foreign genetic material.

Unveiling the McrBC System

E. coli unveils the McrBC system—a vigilant guardian against foreign DNA invasion. This system’s ability to discriminate between methylated and unmethylated DNA is its cornerstone. It functions as a cleaver, detecting methylated DNA and slicing it at its designated recognition sites. This strategic fragmentation destroys foreign DNA that lacks the required methylation patterns while allowing the host DNA, safeguarded by proper methylation, to remain unscathed. The McrBC system, thus, is a sentinel protecting the sanctity of the bacterial genome.

The Mrr System: A Unique Weapon

The Mrr system of E. coli emerges as yet another example of Type IV restriction enzymes. This system’s prowess lies in its precision—the ability to target DNA molecules adorned with specific modified bases. The Mrr enzyme acts as a molecular sword, cleaving DNA either directly at or in proximity to these modified bases. In doing so, it effectively curtails the spread of DNA embellished with distinct modifications, reinforcing the bacterial defense against genetic intruders.

Preservers of Genetic Integrity

Type IV restriction enzymes are the unsung heroes of bacterial protection, upholding the sanctity of genetic identity. By homing in on modified DNA, they become the gatekeepers of the genome. Their intrinsic ability to differentiate between modified and unmodified DNA provides an ingenious strategy for bacterial cells to discern friend from foe, thus shielding themselves from potentially detrimental foreign genetic material.

In the grand orchestra of cellular defense mechanisms, Type IV restriction enzymes are a unique melody—a specialized countermeasure against the infiltration of modified DNA. The McrBC and Mrr systems, among others, showcase their prowess in precision cleavage and selective recognition. As the realm of molecular biology continues to unveil hidden complexities, these enzymes stand as emblematic guardians, preserving the genetic legacy of bacterial life.

5.5. Type V Restriction Endonucleases: A Revolution in Genome Engineering

In the realm of molecular biology, a groundbreaking class of enzymes emerges—Type V restriction enzymes. These enzymatic marvels wield guide RNAs (gRNAs) as their allies, honing in on specific non-palindromic DNA sequences that often characterize invasive organisms. One shining exemplar of this class is the Cas9-gRNA complex derived from CRISPR systems—a herald of unprecedented genetic manipulation possibilities.

A New Dimension of Precision

Type V restriction enzymes chart a new course, capitalizing on guide RNAs to target and cleave non-palindromic DNA sequences. The distinctive feature here is the synergy between the Cas9 protein and the guide RNA. This two-component system functions in concert, with the guide RNA containing a sequence perfectly matched to the target DNA. This molecular compass guides the Cas9 protein to the precise cleavage site, affording a level of precision that other restriction enzymes can’t emulate.

The CRISPR-Cas9 Symphony

Among the shining stars in this constellation is the CRISPR-Cas9 system—a Type V restriction enzyme of profound impact. Cas9, the protein powerhouse, dances with its partner, the guide RNA, orchestrating a symphony of targeted DNA cleavage. Unlike traditional enzymes tethered to specific sequences, the programmability of Type V enzymes shines through. The gRNA can be tailored to navigate a diverse array of DNA sequences, enabling scientists to edit, delete, or insert genetic information with unparalleled finesse.

DNA Breaks and Genetic Masterpieces

The Cas9-gRNA complex embarks on a journey of double-strand DNA breaks at the designated target site. This breach calls upon the cell’s DNA repair machinery—deploying non-homologous end joining (NHEJ) or homology-directed repair (HDR)—to mend the rupture. The revolutionary feat lies in the precision of these repairs, ushering in the era of genome editing. Genes can be modified, diseases can be modeled, and therapeutic interventions beckon as possibility.

Unveiling New Horizons

The allure of Type V restriction enzymes, especially the CRISPR-Cas9 system, extends far beyond the confines of the lab. From fundamental research to biotechnological innovation and medical breakthroughs, their impact knows no bounds. As research continues to illuminate the nuanced intricacies of these enzymes, the realms of efficiency, specificity, and adaptability continue to expand. This trailblazing journey sets the stage for novel applications in diverse realms, driving the realms of science and medicine forward.

A Transformative Age

Type V restriction enzymes have ushered in an era of transformative genetic engineering. The Cas9-gRNA complex resonates as a beacon of precision, enabling the sculpting of genetic destinies. As the symphony of DNA manipulation continues to unfold, the echoes of Type V enzymes reverberate through the corridors of progress, unraveling the mysteries of genetics and opening vistas of possibility.

5.6. Artificial Restriction Endonucleases

Artificial Restriction Endonucleases have become superheroes of genetic engineering and molecular biology. These enzyme wonders are created by combining a DNA-binding part with a nuclease part, often taken from a type IIS restriction enzyme called FokI. This fusion brings together the DNA-binding accuracy and the cutting power of the nuclease, allowing scientists to design these enzymes to go after specific DNA sequences they’re interested in. Unlike natural restriction enzymes that have a thing for palindrome DNA sequences, these artificial champs are super flexible. They can be engineered to lock onto any DNA sequence you want, even the long ones, up to 36 base pairs.

Zinc finger nuclease (ZFN): Meet the zinc finger nuclease (ZFN), a rock star among artificial restriction enzymes. These ZFNs are a fusion of a DNA-binding part made of zinc finger motifs and the FokI nuclease part. What’s cool is that you can customize the DNA-binding part by tweaking the zinc finger domains to recognize whatever DNA sequence you fancy. ZFNs are like precision gene-editing machines, allowing scientists to home in on specific genes or parts of the genome and make changes.

TAL effector nucleases (TALENs): These are another cool class of artificial restriction enzymes. These TALENs come from a DNA-binding part found in TAL effectors, which are like genetic influencers. When you fuse the TAL effector’s DNA-binding part with the FokI nuclease, you get TALENs. Just like ZFNs, TALENs can be engineered to aim at specific DNA sequences for super accurate genome editing.

And let’s not forget the game-changer – CRISPR-Cas9. It started as a prokaryotic defense system but got transformed into a top-tier genome-editing tool. The nuclease part, called Cas9, gets guided to its target by a short RNA buddy, the guide RNA. This dynamic duo inspired the creation of artificial restriction enzymes based on CRISPR-Cas9. These enzyme buddies offer simplicity, efficiency, and versatility in the world of genome editing.

But wait, there’s more! Scientists have also made artificial ribonucleases to act as restriction enzymes for RNA. One example is PNAzymes, which mimic ribonucleases. These PNAzymes have a fancy Cu(II)-2,9-dimethylphenanthroline tag that helps them snip RNA at specific spots, like RNA bulges. This opens up doors for tinkering with RNA and even RNA-based treatments.

Thanks to these artificial restriction enzymes and ribonucleases, our toolbox for DNA and RNA tinkering has exploded. They offer customization, pinpoint accuracy, and endless possibilities for studying genes, editing genomes, and crafting treatments. And as scientists keep pushing the boundaries, you can bet on more exciting innovations in the world of genetic engineering and molecular biology.

Table: Difference Between Types of Restriction Endonucleases

Types of Restriction EndonucleasesType IType IIType IIIType IVType VArtificial Endonucleases
Recognition SiteAsymmetric, often not palindromicPalindromicNon-palindromicVariableNon-palindromicEngineered DNA recognition
Cleavage PatternRandom, distant from recognition siteSymmetric, within or near recognition siteVariable, distant from recognition siteVariableVariablePrecise and customizable
Cofactor RequirementATP-dependentNot requiredATP-dependentNot requiredNot requiredNot required
ExamplesEcoKI, EcoAIEcoRI, HindIIEcoP15, EcoP1McrBCCpf1, C2c2Zinc Finger Nucleases (ZFNs), TALEN, CRISPR-Cas Systems
ExplanationType I enzymes are large, multisubunit complexes that recognize asymmetric DNA sequences. They cleave DNA at a variable distance away from the recognition site and require ATP for their activity.Type II enzymes recognize specific palindromic DNA sequences and cleave at or near their recognition sites. They are widely used in molecular biology due to their simple cleavage patterns.Type III enzymes recognize non-palindromic DNA sequences. They cleave DNA at a variable distance away from the recognition site and require ATP for their activity.Type IV enzymes recognize modified DNA sequences, such as methylated or hydroxymethylated DNA. They cleave DNA at variable positions to protect the host from foreign DNA.Type V enzymes, like Cpf1 and C2c2, are non-palindromic endonucleases that recognize distinct sequences and have potential applications in genome editing.Artificial endonucleases, such as ZFNs, TALENs, and CRISPR-Cas systems, are engineered to target specific DNA sequences, allowing precise genome editing and customization.

6. Restriction Endonucleases: Mechanism of Action

The fascinating world of DNA recognition and cleavage by restriction endonucleases unfolds through a variety of mechanisms that depend on the enzyme type and the DNA sequence they’re targeting. We’ll delve into the workings of different types like IIP, IIF, IIE, IIS, and IIB to understand how they bind and slice those phosphodiester bonds.

Let’s start with the type IIP enzymes – EcoRI, EcoRV, NotI, and BglI. They usually team up as identical subunits, forming dimers. These enzymes have a thing for palindromic DNA sequences, and they grip onto these symmetrical recognition sites. Each subunit interacts with DNA the same way, and each dimeric enzyme packs two active sites primed to cut one phosphodiester bond in each DNA strand.

But hold on, there’s more to it. Some enzymes handling palindromic sites go solo or roll in as tetramers. Imagine MvaI going solo – it likely latches onto its symmetrical recognition spot in one direction, slices one DNA strand, steps back, and then dives in again from the opposite side to slice the other strand.

Now, let’s talk about the tetrameric players like SfiI, SgrAI, and Ecl18kI. They use a dynamic duo – two subunits – to lock onto one copy of the symmetrical recognition sequence, a bit like their dimeric pals. But here’s the twist: when they’re only holding onto one target sequence, they’re in low gear. For the full show, they need both of their subunits to buddy up with a second copy of the recognition sequence. Only then, with both sites occupied, do they slice both strands at both sites before parting ways with the DNA.

Shifting our attention to type IIE enzymes – the dimeric stars like EcoRII and NaeI. They’re all about multitasking, binding to two or more copies of the recognition sequence at different spots within their protein structure. In one spot, they do the cool catalytic stuff for phosphodiester slicing, while in another spot, they act as the rule enforcers. But here’s the kicker – they only pull off their slicer role when the regulatory spots are hanging out with the right DNA buddy. It’s like they need a thumbs-up from their DNA friend to swing into action.

Now, let’s chat about those tricky asymmetric sequences. Homodimeric enzymes – where both subunits do the same thing – can’t crack this code. So, enter the stage, monomeric proteins or oligomers with diverse subunits. These guys team up to conquer the whole DNA sequence or different chunks of it. For example, check out BbvCI – it’s an oligomeric champ that uses its subunits like specialized players to tackle various parts of the asymmetrical sequence.

Now, when enzymes need to team up with two sites, they can do it in two ways: cis or trans. In the cis scenario, the enzyme camps out across two sites in the same DNA, causing the space between them to bend. In the trans situation, the enzyme links arms with sites on separate DNA molecules, practically gluing them together. But guess what? Cis interactions are the cooler kids in town. Why? Because having two sites close together in one DNA molecule is more favorable than having sites on separate DNAs. Enzymes that need two sites, like type I and type III systems, are like double agents – they’re more active on DNA chunks with multiple copies of the recognition sequence. It’s all about the neighborhood vibes – the higher concentration of two sites in cis beats the concentration in trans.

In a nutshell, the DNA recognition and cleavage dance of restriction endonucleases is a multifaceted spectacle. Their tactics are influenced by their structure, whether they’re solo or part of an ensemble, and the unique traits of the DNA sequence they’re after.

Table: Some of the important, most commonly used restriction enzyme, their source organism, recognition sequences and cut sites: 

Restriction EnzymeSource OrganismRecognition SequenceCut Sites
EcoRIEscherichia coli5′-GAATTC-3′3′ overhang
HindIIIHaemophilus influenzae5′-AAGCTT-3′5′ overhang
BamHIBacillus amyloliquefaciens5′-GGATCC-3′5′ overhang
PstIProvidencia stuartii5′-CTGCAG-3′5′ overhang
EcoRVEscherichia coli5′-GATATC-3′Blunt ends
NotINocardia otitidis-caviarum5′-GCGGCCGC-3′5′ overhang
SmaISerratia marcescens5′-CCCGGG-3′Blunt ends
XbaIXanthomonas badrii5′-TCTAGA-3′5′ overhang
KpnIKlebsiella pneumoniae5′-GGTACC-3′3′ overhang
BglIIBacillus licheniformis5′-AGATCT-3′3′ overhang
NcoINeisseria meningitidis5′-CCATGG-3′4-base overhang
AluIArthrobacter luteus5′-AGCT-3′4-base overhang
HaeIIIHaemophilus aegyptius5′-GGCC-3′Blunt ends
SacIStreptomyces achromogenes5′-GAGCTC-3′5′ overhang
XhoIXanthomonas oryzae5′-CTCGAG-3′5′ overhang
SpeISphingomonas species5′-ACTAGT-3′5′ overhang
SalIStreptomyces albus G5′-GTCGAC-3′5′ overhang
EcoRVEscherichia coli5′-GATATC-3′Blunt ends
Acc65IAcidaminococcus fermentans5′-GTMKAC-3′Blunt ends
BsaIBacillus stearothermophilus5′-GGTCTC-3′5′ overhang
ClaIPseudomonas aeruginosa5′-ATCGAT-3′5′ overhang
BglIBeggiatoa leptomitoformis5′-GCCNNNNNGGC-3′Cleaves in the middle
XhoIIXanthomonas holcicola5′-RGATCY-3′5′ overhang
DraIDeinococcus radiodurans5′-TTTAAA-3′4-base overhang
MaeIMicrococcus luteus5′-CTAG-3′4-base overhang
XmnIXanthomonas manihotis5′-GAANNNNTTC-3′Cleaves in the middle
HinfIHaemophilus influenzae5′-GANTC-3′4-base overhang
AvaIAnabaena variabilis5′-CYCGRG-3′5′ overhang
HpaIHaemophilus parainfluenzae5′-GTTAAC-3′5′ overhang
BstEIIBacillus stearothermophilus5′-RGATCY-3′5′ overhang
PvuIProteus vulgaris5′-CGATCG-3′3′ overhang
AseIAquifex aeolicus5′-ATTAAT-3′Blunt ends

7. Application/Significance of Restriction Endonucleases:

The application of restriction enzymes, also known as restriction endonucleases, encompasses a wide array of scientific, medical, and biotechnological endeavors. These remarkable molecular scissors play a pivotal role in genetic manipulation, enabling researchers to precisely cleave DNA at specific recognition sites. This precision has revolutionized various fields, such as genetic engineering, where restriction enzymes are instrumental in creating targeted gene modifications, deletions, or insertions. In molecular cloning, these enzymes act as essential tools for assembling DNA fragments, facilitating the construction of recombinant DNA molecules that hold the key to understanding gene function. Furthermore, restriction enzymes find utility in disease diagnosis, aiding in the identification of specific genetic mutations associated with various disorders. In forensic science, they contribute to DNA fingerprinting, allowing accurate determination of genetic relatedness and individual identity. The power of restriction enzymes also extends to studying gene expression, recombinant protein production, and genome mapping, providing crucial insights into regulatory mechanisms, biotechnological advancements, and genetic landscapes. As technology advances, including the emergence of genome editing techniques like CRISPR-Cas9, restriction enzymes continue to be foundational tools that unlock the secrets of genetics, offering limitless potential for scientific innovation and discovery.

Table: Applications of Restriction Endonucleases: Unveiling the Power of Precision DNA Tools

ApplicationDescription
Genetic Engineering: Precision Genome EditingRestriction enzymes enable precise gene editing by creating defined breaks in DNA. Genetic changes, deletions, or insertions can be introduced for tailored traits or functions.
Molecular Cloning: Assembling DNA Jigsaw PuzzlesRestriction enzymes act as molecular scissors, producing sticky ends for seamless joining of DNA fragments with complementary ends. Recombinant DNA molecules aid in genetic studies.
Disease Diagnosis: Unveiling Genetic SecretsRestriction enzymes target disease-associated DNA mutations. Restriction Fragment Length Polymorphism (RFLP) analysis aids in disease diagnosis and genetic profiling.
DNA Fingerprinting: Unmasking IdentityRestriction enzymes play a pivotal role in DNA fingerprinting for forensic science and paternity testing. Unique fragment patterns serve as identifying markers.
Studying Gene Expression: Probing Genetic ActivityRestriction enzymes selectively cut DNA at specific sites to probe gene expression regulatory mechanisms. Insights into gene on/off control are gained.
Recombinant Protein Production: Crafting Molecular FactoriesRestriction enzymes aid in creating precise DNA fragments for engineering cells to produce desired proteins. Biotechnology and pharmaceutical production benefit.
Genome Mapping: Navigating the Genetic LandscapeRestriction enzymes contribute to creating genetic maps by digesting genomic DNA. The maps offer navigational tools for exploring genetic relationships and disease-related genes.
Synthetic Biology: Engineering LifeRestriction enzymes facilitate DNA manipulation for synthetic biology, enabling the creation of synthetic organisms, metabolic pathways, and novel biological functions.
Epigenetic Investigations: Unraveling Gene RegulationRestriction enzymes aid in studying DNA methylation and chromatin structure, shedding light on epigenetic modifications that influence gene expression and identity.
Advanced Techniques: Beyond Traditional CleavageBeyond traditional DNA cleavage, enzymes like Cas9 are used for precision genome editing. Novel enzymes with tailored specificities expand genetic manipulation possibilities.
Conclusion: A Landscape of Boundless PotentialRestriction enzymes redefine genetic exploration, from engineering genomes to studying gene regulation. Their applications continue to evolve, shaping scientific innovation.
Beyond the Horizon: A Promising FutureRestriction enzymes remain at the forefront of innovation, from genetic disorder therapies to biofuel development. Their precision guides us through genetic complexity.

8. Questions (FAQs) on Restriction Endonucleases

Which restriction endonuclease enzyme is first discovered?

The first discovered restriction endonuclease enzyme is HindII, which was isolated from the bacterium Haemophilus influenzae in 1970.

How does restriction endonuclease function?

Restriction endonucleases function by recognizing specific DNA sequences (recognition sites) and cleaving the DNA at or near these sites. They break the phosphodiester bonds that hold the DNA strands together, resulting in the formation of DNA fragments.

Do eukaryotic cells have restriction endonuclease?

Eukaryotic cells do not have restriction endonucleases in the same way as bacteria do. However, eukaryotic cells have similar enzymes called DNA endonucleases that play roles in DNA repair and recombination.

What determines where a restriction endonuclease will cut?

The sequence of bases in the DNA, known as the recognition site, determines where a restriction endonuclease will cut. If the DNA sequence matches the specific recognition sequence for that enzyme, it will cleave the DNA at that site.

What is the function of restriction endonucleases in bacteria?

In bacteria, restriction endonucleases function as a defense mechanism against foreign DNA, such as viral DNA. By cleaving foreign DNA at specific recognition sites, they protect the bacterium from potential threats.

How do restriction endonucleases work?

Restriction endonucleases work by scanning DNA sequences for specific recognition sites. Once a recognition site is found, the enzyme binds to the DNA and cleaves the phosphodiester bonds, resulting in the formation of DNA fragments.

What is the source of restriction endonucleases?

Restriction endonucleases are commonly found in bacteria, where they serve as a part of the bacterial defense system. They are also used extensively in genetic engineering and molecular biology research.

Can restriction endonuclease/enzyme cut RNA?

No, restriction endonucleases specifically recognize and cleave DNA sequences. They do not cleave RNA molecules.

What is the role of restriction endonucleases in genetic engineering?

In genetic engineering, restriction endonucleases are used to cut DNA at specific sites, allowing for the insertion, deletion, or modification of genetic material. This is a crucial step in creating recombinant DNA molecules.

How do restriction endonucleases contribute to DNA fingerprinting?

Restriction endonucleases are used in DNA fingerprinting to cleave DNA at specific recognition sites. The resulting fragment patterns are unique to individuals, allowing for identification and forensic analysis.

Can restriction endonucleases be used for targeted gene editing?

Yes, along with other tools like CRISPR-Cas9, restriction endonucleases can be used for targeted gene editing. They help create precise breaks in DNA, which can be repaired with desired genetic modifications.

What are isoschizomers and neoschizomers in relation to restriction enzymes?

Isoschizomers are different restriction enzymes that recognize the same DNA sequence and produce the same cut pattern. Neoschizomers recognize the same sequence but produce different cut patterns.

Are there any natural inhibitors of restriction endonucleases?

Yes, some bacteria produce DNA methyltransferases that methylate recognition sites to protect their own DNA from cleavage by restriction endonucleases.

How are restriction endonucleases named?

Restriction endonucleases are often named after the bacterial species they are derived from. For example, EcoRI comes from Escherichia coli.

What is the significance of palindromic sequences in restriction endonuclease recognition?

Palindromic sequences are recognized by many restriction endonucleases. They form specific recognition sites that allow the enzyme to bind and cleave DNA at a specific position.

How are restriction endonucleases used in recombinant DNA technology?

In recombinant DNA technology, restriction endonucleases are used to cut both the target DNA and a vector DNA. This allows for the insertion of the target DNA into the vector, creating recombinant DNA.

What are the challenges and limitations of using restriction endonucleases in genetic manipulation?

Some limitations include the need for specific recognition sites, potential off-target cleavage, and difficulties in designing enzymes for non-palindromic sequences.

9. References

https://upload.wikimedia.org/wikipedia/commons/a/a4/Difference-Between-Endonuclease-and-Exonuclease-3.webp

https://upload.wikimedia.org/wikipedia/commons/e/e0/HindIII_Restriction_site_and_sticky_ends_vector.svg

Pingoud, A., Alves, J. and Geiger, R., 1993. Restriction enzymes. Enzymes of molecular biology, pp.107-200.

Di Felice, F., Micheli, G. and Camilloni, G., 2019. Restriction enzymes and their use in molecular biology: An overview. Journal of biosciences, 44(2), p.38.

Robinson, D., Walsh, P.R. and Bonventre, J.A., 2001. Restriction endonucleases. Molecular biology problem solver: a laboratory guide, pp.225-266.

Raleigh, E.A. and Brooks, J.E., 1998. Restriction modification systems: where they are and what they do. In Bacterial genomes: physical structure and analysis (pp. 78-92). Boston, MA: Springer US.

Pingoud, A., Fuxreiter, M., Pingoud, V. and Wende, W., 2005. Type II restriction endonucleases: structure and mechanism. Cellular and molecular life sciences62, pp.685-707.

Pingoud, A. and Jeltsch, A., 1997. Recognition and cleavage of DNA by type‐II restriction endonucleases. European Journal of Biochemistry246(1), pp.1-22.

Janscak, P., Sandmeier, U., Szczelkun, M.D. and Bickle, T.A., 2001. Subunit assembly and mode of DNA cleavage of the type III restriction endonucleases EcoP1I and EcoP15I. Journal of molecular biology306(3), pp.417-431.

Meisel, A., Mackeldanz, P., Bickle, T.A., Krüger, D.H. and Schroeder, C., 1995. Type III restriction endonucleases translocate DNA in a reaction driven by recognition site‐specific ATP hydrolysis. The EMBO Journal14(12), pp.2958-2966.

Katada, H. and Komiyama, M., 2009. Artificial restriction DNA cutters as new tools for gene manipulation. ChemBioChem10(8), pp.1279-1288.

Carroll, D., 2011. Genome engineering with zinc-finger nucleases. Genetics188(4), pp.773-782.

Porteus, M.H. and Carroll, D., 2005. Gene targeting using zinc finger nucleases. Nature biotechnology23(8), pp.967-973.

Bibikova, M., Beumer, K., Trautman, J.K. and Carroll, D., 2003. Enhancing gene targeting with designed zinc finger nucleases. Science300(5620), pp.764-764.

Joung, J.K. and Sander, J.D., 2013. TALENs: a widely applicable technology for targeted genome editing. Nature reviews Molecular cell biology14(1), pp.49-55.

Gaj, T., Gersbach, C.A. and Barbas, C.F., 2013. ZFN, TALEN, and CRISPR/Cas-based methods for genome engineering. Trends in biotechnology31(7), pp.397-405.

Gupta, R.M. and Musunuru, K., 2014. Expanding the genetic editing tool kit: ZFNs, TALENs, and CRISPR-Cas9. The Journal of clinical investigation124(10), pp.4154-4161.

LaFountaine, J.S., Fathe, K. and Smyth, H.D., 2015. Delivery and therapeutic applications of gene editing technologies ZFNs, TALENs, and CRISPR/Cas9. International journal of pharmaceutics494(1), pp.180-194.Zhang, H.X., Zhang, Y. and Yin, H., 2019. Genome editing with mRNA encoding ZFN, TALEN, and Cas9. Molecular Therapy27(4), pp.735-746.

Leave a Reply

Your email address will not be published. Required fields are marked *