Skip to content

This report is written by MaltSci based on the latest literature and research findings


How does CRISPR gene editing function?

Abstract

The emergence of CRISPR (Clustered Regularly Interspaced Short Palindromic Repeats) gene editing technology represents a groundbreaking advancement in the fields of genetics and molecular biology, enabling precise modifications of DNA sequences with unprecedented accuracy. Originally identified as a bacterial immune system, the CRISPR-Cas9 mechanism has been adapted for use in various organisms, facilitating significant advancements in therapeutic applications, agricultural biotechnology, and industrial processes. This review delves into the core mechanisms of CRISPR gene editing, detailing the operational principles of CRISPR-Cas systems, including the design of guide RNAs, the formation of ribonucleoprotein complexes, and the induction of double-strand breaks in target DNA. Following cleavage, the cellular DNA repair pathways, namely non-homologous end joining (NHEJ) and homology-directed repair (HDR), play critical roles in determining the outcomes of gene editing efforts. The review further explores the diverse applications of CRISPR technology, highlighting its potential to treat genetic disorders, enhance crop traits, and optimize industrial microbial strains. Despite its transformative potential, CRISPR technology faces ethical challenges, particularly regarding germline modifications and off-target effects, necessitating the establishment of comprehensive regulatory frameworks. In conclusion, this review emphasizes the need for ongoing research to refine CRISPR technology, enhance its precision, and address ethical considerations, ensuring its responsible application in research and clinical settings.

Outline

This report will discuss the following questions.

  • 1 Introduction
  • 2 Mechanisms of CRISPR Gene Editing
    • 2.1 Overview of CRISPR-Cas Systems
    • 2.2 Mechanism of Action: Targeting and Cleavage
    • 2.3 Repair Mechanisms: NHEJ and HDR
  • 3 Applications of CRISPR Technology
    • 3.1 Therapeutic Applications in Human Health
    • 3.2 Agricultural Biotechnology
    • 3.3 Industrial Biotechnology
  • 4 Ethical Considerations and Regulatory Challenges
    • 4.1 Ethical Implications of Gene Editing
    • 4.2 Regulatory Frameworks and Guidelines
    • 4.3 Public Perception and Acceptance
  • 5 Current Limitations and Future Directions
    • 5.1 Off-Target Effects and Precision
    • 5.2 Advances in CRISPR Technology
    • 5.3 Future Prospects in Research and Medicine
  • 6 Summary

1 Introduction

The advent of CRISPR (Clustered Regularly Interspaced Short Palindromic Repeats) gene editing has marked a pivotal shift in the landscape of genetics and molecular biology, offering unprecedented capabilities for precise DNA modifications across various organisms. Originally discovered as a bacterial immune mechanism, the CRISPR-Cas9 system has been repurposed into a powerful tool for genome editing, enabling researchers to add, delete, or alter specific genetic sequences with remarkable accuracy and efficiency [1][2]. This revolutionary technology not only facilitates fundamental biological research but also harbors significant potential for therapeutic applications, including the treatment of genetic disorders, cancer, and agricultural enhancements [3][4].

The significance of CRISPR technology lies in its ability to transform our understanding of gene function and its implications for human health. By enabling precise genetic modifications, CRISPR has opened new avenues for investigating the molecular underpinnings of diseases and developing targeted therapies. For instance, its applications in gene therapy have shown promise in correcting genetic defects and advancing cancer immunotherapy [2][3]. Moreover, CRISPR's utility extends to agricultural biotechnology, where it can be employed to create crops with improved traits, thereby addressing food security challenges in the face of climate change [5].

Despite its transformative potential, the implementation of CRISPR technology is not without challenges. Ethical considerations surrounding gene editing, particularly in human embryos and germline modifications, raise important questions about the implications of altering the genetic makeup of future generations [3]. Additionally, concerns regarding off-target effects, where unintended genetic alterations may occur, pose significant risks that must be carefully managed as the technology progresses [6][7]. As such, establishing robust regulatory frameworks and ensuring public acceptance of CRISPR applications are crucial steps toward its responsible use in both research and clinical settings [3].

This review aims to provide a comprehensive overview of the mechanisms underlying CRISPR gene editing, detailing its operational principles, including the CRISPR-Cas systems, the mechanisms of action such as targeting and cleavage, and the subsequent DNA repair pathways, namely non-homologous end joining (NHEJ) and homology-directed repair (HDR) [2]. Furthermore, we will explore the diverse applications of CRISPR technology across therapeutic, agricultural, and industrial domains, highlighting its transformative impact on human health and biotechnology [4][5]. The ethical considerations and regulatory challenges associated with CRISPR will also be discussed, providing insights into the ongoing discourse surrounding its societal implications [3]. Finally, we will address the current limitations of CRISPR technology, including off-target effects and the quest for improved precision, while outlining future directions that may enhance its efficacy and safety [6][7].

By elucidating the intricacies of CRISPR technology, this report seeks to inform ongoing research and development efforts, fostering a deeper understanding of its potential and limitations in the rapidly evolving field of gene editing.

2 Mechanisms of CRISPR Gene Editing

2.1 Overview of CRISPR-Cas Systems

CRISPR gene editing operates through a sophisticated mechanism that utilizes the CRISPR-Cas systems, which are derived from the adaptive immune responses of prokaryotes. This system allows for precise modifications to genomic DNA by introducing targeted double-strand breaks (DSBs) at specific locations in the genome. The process is primarily facilitated by the Cas (CRISPR-associated) proteins, most notably Cas9, which functions as an RNA-guided nuclease.

The fundamental steps involved in CRISPR gene editing can be summarized as follows:

  1. Guide RNA Design: The first step involves the design of a guide RNA (gRNA) that is complementary to the target DNA sequence. This gRNA is crucial as it directs the Cas protein to the precise location within the genome where the edit is to be made.

  2. Formation of the CRISPR-Cas Complex: The gRNA binds to the Cas9 protein, forming a ribonucleoprotein complex. This complex is then introduced into the target cells, where it will locate the corresponding DNA sequence.

  3. Target Recognition and Binding: The CRISPR-Cas complex scans the genome for the specific target sequence. Upon finding a match, the complex binds to the DNA. The binding is contingent upon the presence of a protospacer adjacent motif (PAM) sequence, which is essential for Cas9 to recognize and bind to the target DNA.

  4. Induction of Double-Strand Breaks: Once bound, Cas9 induces a double-strand break in the DNA at the target site. This break activates the cell's natural DNA repair mechanisms, which are critical for the subsequent steps of the editing process.

  5. DNA Repair Mechanisms: The cell attempts to repair the double-strand break using one of two primary repair pathways:

    • Non-Homologous End Joining (NHEJ): This pathway often leads to insertions or deletions (indels) at the break site, which can result in gene disruption. It is a quick but error-prone method of repair.
    • Homology-Directed Repair (HDR): If a donor DNA template is provided along with the CRISPR-Cas components, the cell can utilize this template to make precise edits, such as correcting mutations or inserting new genetic material.
  6. Outcome of the Editing Process: The outcome of the CRISPR editing process can vary based on the repair pathway utilized. NHEJ may result in gene knockout, while HDR can facilitate precise modifications, allowing for targeted gene correction or the introduction of new genetic traits.

The versatility and efficiency of the CRISPR-Cas systems have led to their widespread adoption across various fields, including agriculture, biotechnology, and medicine. For instance, in plant genome editing, the CRISPR-Cas system has shown promise for improving crop traits and functional genomics studies [8]. In human disease research, CRISPR has been utilized to explore genetic diseases and develop potential therapeutic strategies [1], demonstrating its transformative impact on molecular biology and genetics.

Overall, the CRISPR-Cas systems represent a powerful tool for genome editing, enabling precise genetic modifications that have significant implications for research and therapeutic applications.

2.2 Mechanism of Action: Targeting and Cleavage

CRISPR gene editing, particularly through the CRISPR/Cas9 system, operates on a fundamental mechanism involving the recognition and cleavage of target DNA sequences. This process is primarily facilitated by the Cas9 endonuclease, which is guided by a specific RNA sequence known as the single-guide RNA (sgRNA). The mechanism can be broken down into several key steps: target recognition, binding, and cleavage.

Initially, the sgRNA is designed to be complementary to a specific DNA sequence within the genome. This sgRNA forms a complex with the Cas9 protein, creating a ribonucleoprotein complex that is essential for the targeting process. The first step involves the identification of the protospacer adjacent motif (PAM), a short, conserved sequence that is crucial for Cas9 binding. The PAM sequence is typically located adjacent to the target DNA sequence and is necessary for the Cas9 to successfully attach and initiate the editing process[9].

Once the Cas9-sgRNA complex binds to the target DNA, the next step is the formation of an R-loop, where the sgRNA hybridizes with the target DNA while displacing one of the DNA strands. This hybridization is critical as it ensures specificity in targeting, allowing the Cas9 protein to recognize the correct genomic locus for editing[10].

Following successful binding and R-loop formation, Cas9 induces a double-strand break (DSB) in the DNA at the target site. This cleavage is achieved through a two-metal-ion mechanism that is essential for the catalytic activity of Cas9. The active site of Cas9 coordinates two magnesium ions, which facilitate the cleavage of the phosphodiester bonds in the DNA, resulting in a DSB[7]. The DSB is then repaired by the cell's natural DNA repair mechanisms, predominantly through two pathways: non-homologous end joining (NHEJ) and homology-directed repair (HDR). NHEJ often leads to insertions or deletions (indels) that can disrupt gene function, while HDR can be utilized to introduce specific genetic modifications when a repair template is provided[2].

Moreover, recent studies have indicated that the efficiency of CRISPR-mediated cleavage is influenced by several factors, including the chromatin state of the target region, the GC content of the PAM proximal and distal regions, and the overall structure of the sgRNA. For instance, the presence of nucleosomes can inhibit cleavage efficiency, which suggests that the accessibility of the target site plays a significant role in the effectiveness of CRISPR gene editing[11][12].

In summary, CRISPR gene editing functions through a highly orchestrated mechanism involving target recognition via sgRNA, PAM-dependent binding, R-loop formation, and Cas9-mediated DNA cleavage, followed by cellular repair processes that facilitate the desired genetic modification. This system has not only transformed the landscape of genetic engineering but also holds immense potential for therapeutic applications in treating genetic disorders and various diseases.

2.3 Repair Mechanisms: NHEJ and HDR

CRISPR gene editing, particularly through the CRISPR/Cas9 system, operates by inducing site-specific double-strand breaks (DSBs) in the target DNA. The cellular response to these DSBs predominantly involves two distinct DNA repair pathways: non-homologous end joining (NHEJ) and homology-directed repair (HDR).

NHEJ is the primary mechanism for repairing DSBs in eukaryotic cells, especially in higher plants and mammalian systems. This pathway is characterized by its rapid action, wherein the broken ends of the DNA are directly ligated together without the need for a homologous template. However, NHEJ is prone to errors, often resulting in insertions or deletions (indels) that can disrupt the reading frame of the gene, leading to frameshift mutations. This can create genetic knockouts, which are useful for functional studies but may not provide the precision needed for targeted modifications [13].

In contrast, HDR is a more precise repair mechanism that utilizes a homologous template to guide the repair process. This pathway allows for accurate gene modifications, including targeted insertions, deletions, and substitutions. For HDR to occur, a donor template is typically provided, which can be a single-stranded oligonucleotide or a double-stranded DNA fragment. The challenge with HDR is its relatively low efficiency compared to NHEJ, particularly in postmitotic cells where cell cycle constraints further limit HDR activity [14].

To enhance HDR efficiency, various strategies have been explored. These include inhibiting key factors of the NHEJ pathway, optimizing donor templates, synchronizing cells to favor HDR-permissive phases, and employing engineered proteins that promote HDR [15]. For instance, recent advancements have shown that manipulating the balance between NHEJ and HDR can significantly increase the frequency of precise genome editing. Techniques such as the fusion of Cas9 to DNA repair factors or the use of small molecules to modulate repair pathways have demonstrated promising results in increasing HDR rates [16][17].

Moreover, researchers have identified specific proteins, such as RAD18, that can stimulate HDR by suppressing NHEJ activity at the site of DSBs. This targeted approach allows for a higher ratio of HDR products to NHEJ products, thus improving the overall efficiency of CRISPR-mediated genome editing [18].

In summary, CRISPR gene editing harnesses the cellular DNA repair mechanisms of NHEJ and HDR to achieve its effects. While NHEJ offers rapid repair, it often results in undesirable mutations. HDR, although more precise, is less efficient and requires strategic enhancement to facilitate accurate gene modifications. The ongoing research aims to refine these repair pathways to improve the fidelity and efficiency of CRISPR-based editing technologies [19][20].

3 Applications of CRISPR Technology

3.1 Therapeutic Applications in Human Health

CRISPR (Clustered Regularly Interspaced Short Palindromic Repeats) technology represents a revolutionary advancement in genome editing, enabling precise modifications of DNA sequences within living organisms. The core mechanism of CRISPR involves the use of RNA-guided endonucleases, particularly the CRISPR-associated protein 9 (Cas9), which can create double-strand breaks at specific genomic locations. This precision is achieved through the design of a guide RNA (gRNA) that is complementary to the target DNA sequence, allowing Cas9 to bind and cleave the DNA at the desired site.

The applications of CRISPR technology in therapeutic contexts are extensive and varied. One of the most promising areas is the treatment of genetic disorders. For instance, CRISPR has been applied to correct mutations associated with diseases such as Duchenne muscular dystrophy, β-thalassemia, and cystic fibrosis. These applications have moved from proof-of-concept studies to clinical trials, showcasing the potential of CRISPR to address monogenic diseases by directly modifying the defective genes responsible for these conditions (Kofler & Kraschel, 2018; Luthra et al., 2021; Cai et al., 2016).

Moreover, CRISPR's utility extends beyond genetic diseases to include applications in oncology and infectious diseases. For example, it has been explored for use in cancer therapies by targeting oncogenes or enhancing the efficacy of chimeric antigen receptor (CAR) T-cell therapies. This approach can improve the ability of T-cells to recognize and destroy cancer cells, potentially leading to more effective treatments (El-Kenawy et al., 2019; Kavousinia et al., 2024). In the realm of infectious diseases, CRISPR has been investigated for its ability to target and deactivate viral genomes, including those of HIV and hepatitis B, thereby offering new avenues for treatment (Adlat et al., 2023).

The development of CRISPR technology has also led to innovations such as base editing and prime editing, which allow for even more precise modifications at the genetic level. These advancements aim to reduce off-target effects and enhance the overall safety and efficacy of gene editing in therapeutic contexts (Pacesa et al., 2024; Cetin et al., 2025).

Despite the remarkable potential of CRISPR technology, challenges remain. Issues such as off-target effects, delivery methods, and ethical considerations regarding germline editing necessitate ongoing research and regulatory oversight to ensure safe and responsible application in human health (Doudna, 2020; Hirakawa et al., 2020). Overall, the transformative nature of CRISPR in therapeutic applications underscores its promise in reshaping the future of medicine, providing new hope for the treatment of previously intractable diseases.

3.2 Agricultural Biotechnology

CRISPR gene editing operates through a mechanism that utilizes the CRISPR/Cas system, which is a revolutionary tool in agricultural biotechnology. This system allows for precise modifications of the genome by employing a guide RNA (gRNA) that is complementary to a specific DNA sequence in the target genome, along with the Cas9 protein, which acts as a molecular scissors to create double-strand breaks at designated sites. The process involves several critical steps:

  1. Design of Guide RNA: The first step in CRISPR gene editing is designing a gRNA that matches the target DNA sequence. This gRNA is responsible for guiding the Cas9 protein to the correct location in the genome.

  2. Formation of the CRISPR/Cas Complex: The gRNA is then combined with the Cas9 protein to form a ribonucleoprotein (RNP) complex. This complex is essential for the subsequent editing process.

  3. Introduction into the Target Cells: The RNP complex is introduced into the plant cells through various delivery methods, such as Agrobacterium-mediated transformation, biolistics, or direct transfection methods. The choice of delivery method can influence the efficiency of the gene editing process and is dependent on the specific crop and target gene.

  4. Targeted DNA Cleavage: Once inside the cell, the RNP complex locates the target DNA sequence. The Cas9 protein binds to the gRNA and uses it as a guide to identify the specific DNA region, where it introduces a double-strand break.

  5. DNA Repair Mechanisms: Following the introduction of a double-strand break, the cell’s natural DNA repair mechanisms are activated. The cell can repair the break through two primary pathways: non-homologous end joining (NHEJ) or homology-directed repair (HDR). NHEJ often results in insertions or deletions (indels) that can disrupt the target gene, effectively knocking it out. Alternatively, HDR can be used to introduce specific changes if a donor template is provided.

  6. Transgene-Free Editing: A significant advantage of the CRISPR/Cas system is its ability to generate transgene-free plants. By delivering the Cas9 protein and gRNA in a way that they are not integrated into the plant genome, researchers can create edited plants without foreign DNA, which addresses some regulatory concerns associated with genetically modified organisms (GMOs) [21].

The applications of CRISPR technology in agricultural biotechnology are extensive. This technology has enabled the development of crops with improved traits such as enhanced disease resistance, increased yield, and better tolerance to abiotic stresses like drought and salinity. For instance, CRISPR has been utilized to create rice varieties with improved nutritional profiles and increased resilience against pathogens [22].

Furthermore, CRISPR technology is not limited to simple gene knockouts; it also facilitates more complex genetic modifications, including the introduction of beneficial traits through precise editing. For example, researchers have employed CRISPR to enhance the nutritional quality of staple crops and to improve traits such as flowering time and fruit size in horticultural crops [23].

In summary, CRISPR gene editing works by leveraging a guide RNA and the Cas9 protein to introduce precise modifications in the plant genome, facilitating the development of crops that can better meet the challenges posed by climate change and increasing global food demand. The versatility and efficiency of this technology position it as a cornerstone of modern agricultural biotechnology, promising to transform crop improvement strategies and enhance food security worldwide [24].

3.3 Industrial Biotechnology

CRISPR gene editing operates through a sophisticated mechanism that leverages the natural defense system found in bacteria, specifically the clustered regularly interspaced short palindromic repeats (CRISPR) and CRISPR-associated proteins (Cas). This technology enables precise modifications to genomic DNA by utilizing guide RNA (gRNA) to direct the Cas protein to specific DNA sequences. Upon binding, the Cas protein induces a double-strand break in the target DNA, which is subsequently repaired by the cell's DNA repair mechanisms, leading to insertions, deletions, or substitutions at the target site.

In the context of industrial biotechnology, CRISPR technology has shown remarkable potential for enhancing microbial strains used in various applications, including the production of biopharmaceuticals, biofuels, and other industrially relevant metabolites. For instance, the CRISPR/Cas system has been effectively employed to optimize metabolic pathways in microorganisms such as Escherichia coli, thereby increasing the yield and productivity of desired compounds. By controlling metabolic flux through targeted gene editing, researchers can achieve significant improvements in the synthesis of biochemical products that are of high industrial significance[25].

Moreover, CRISPR technology has facilitated the development of advanced genetic engineering techniques in industrial microorganisms, allowing for the creation of complex and programmable gene circuits. This versatility enhances the ability to engineer strains that can efficiently produce a wide range of products, from biofuels to pharmaceuticals[26]. The system's ease of use and adaptability make it an attractive option for researchers aiming to improve industrial bioprocesses.

In agriculture, CRISPR has been utilized to develop crops with enhanced traits, such as improved nutritional value, resistance to pests, and tolerance to abiotic stresses. These advancements not only promise to increase crop yields but also contribute to sustainability in food production, addressing the growing global demand for food[27].

Overall, the application of CRISPR technology in industrial biotechnology represents a transformative approach that enables precise genetic modifications, leading to improved efficiency and productivity in various sectors, including agriculture and pharmaceuticals. As the technology continues to evolve, it holds the potential to unlock new avenues for innovation and efficiency in industrial applications.

4 Ethical Considerations and Regulatory Challenges

4.1 Ethical Implications of Gene Editing

CRISPR-Cas9 gene editing is a revolutionary technique that allows for precise modifications to DNA. This technology utilizes an RNA guide molecule that binds to complementary DNA sequences, which subsequently recruits the endonuclease Cas9 to introduce double-stranded breaks at targeted locations in the DNA. The double-stranded breaks can then be repaired by the cell's natural repair mechanisms, allowing for the modification or removal of specific DNA bases. The advantages of CRISPR-Cas9 include its affordability, speed, and ease of use, making it a popular choice in laboratories for various applications, including the development of complex animal models and potential therapies for genetic and non-genetic diseases (Cribbs & Perera, 2017) [28].

Despite its potential, the application of CRISPR technology raises significant ethical and regulatory challenges. One of the primary ethical concerns involves human germline modifications, which could lead to unforeseen consequences and moral dilemmas, such as the potential for eugenics. Issues such as informed consent, the risk of unintended effects, and the broader implications of altering human genetics are at the forefront of discussions surrounding CRISPR (Shinwari et al., 2018) [29]. There is a consensus among experts that stringent regulations and guidelines are essential to ensure the responsible use of CRISPR technology. An extensive dialogue involving scientists, ethicists, and policymakers is critical to navigate these ethical waters and to establish a regulatory framework that addresses both the benefits and risks associated with genome editing (Vassena et al., 2016) [30].

The controversy surrounding the "CRISPR babies" case further highlights the urgency of establishing robust ethical standards and regulatory oversight. The actions taken by researchers in this instance raised questions about compliance with ethical norms and the need for stronger governance mechanisms in the face of rapidly advancing technology (Kleiderman & Ogbogu, 2019) [31]. The potential for misuse of CRISPR technology, such as germ-line enhancement and unequal access to genetic modifications, underscores the necessity for a globally coordinated approach to governance that prioritizes ethical considerations and societal implications (Biswas, 2025) [32].

In conclusion, while CRISPR-Cas9 gene editing presents groundbreaking opportunities for scientific advancement and therapeutic interventions, it also poses significant ethical and regulatory challenges. Addressing these challenges requires a concerted effort from the scientific community, ethicists, and policymakers to ensure that the technology is developed and applied in a manner that is safe, ethical, and beneficial to society.

4.2 Regulatory Frameworks and Guidelines

CRISPR gene editing operates through a sophisticated mechanism that leverages a bacterial defense system to precisely modify DNA sequences in various organisms. The core of this technology is the CRISPR/Cas system, which includes the Cas9 protein, an RNA-guided endonuclease that creates double-strand breaks at specific locations in the genome. This process is initiated by the introduction of a guide RNA (gRNA) that is complementary to the target DNA sequence. Upon binding to the target DNA, the Cas9 protein is activated to induce a double-strand break, which the cell attempts to repair through two primary mechanisms: homology-directed repair (HDR) and non-homologous end joining (NHEJ). HDR can facilitate precise edits, while NHEJ often results in insertions or deletions that may disrupt gene function [2][3].

The application of CRISPR technology has raised significant ethical considerations and regulatory challenges. As gene editing capabilities expand, particularly concerning human and agricultural applications, there is a pressing need for regulatory frameworks that ensure safety and efficacy while addressing public concerns. CRISPR technology has prompted many countries to rethink their regulatory approaches to genetically modified organisms (GMOs), leading to discussions about the flexibility of regulations surrounding gene-edited crops. This shift reflects a growing recognition of the potential benefits of CRISPR applications in enhancing food security and agricultural sustainability [5][27].

The regulatory landscape for CRISPR technologies is complex and varies widely across jurisdictions. For instance, in the European Union, gene-edited organisms may be subject to the same stringent regulations as traditional GMOs, which can hinder research and development efforts. Conversely, in countries like the United States, there is a more permissive approach that differentiates between gene editing and transgenic methods, potentially streamlining the approval process for CRISPR-modified organisms [5].

Ethical concerns also extend to the implications of germline editing, which involves changes that can be inherited by future generations. This aspect raises questions about the long-term consequences of such modifications, including unintended effects on genetic diversity and potential societal inequalities. As CRISPR technology continues to evolve, ongoing dialogue among scientists, ethicists, policymakers, and the public will be crucial to navigate these challenges effectively [1][33].

In summary, while CRISPR gene editing presents groundbreaking opportunities for advancements in medicine, agriculture, and biotechnology, it also necessitates careful consideration of ethical implications and the establishment of robust regulatory frameworks to guide its responsible use.

4.3 Public Perception and Acceptance

CRISPR-Cas9 gene editing is a revolutionary technique that enables precise modifications of DNA sequences. The technology utilizes an RNA guide molecule that binds to complementary DNA sequences, facilitating the recruitment of the Cas9 endonuclease. This process results in the introduction of double-stranded breaks in the target DNA, which can subsequently be repaired, allowing for the modification or removal of specific DNA bases. The method is characterized by its cost-effectiveness, speed, and ease of use, making it a popular choice in laboratories worldwide. Its applications extend beyond basic research to include in vivo modifications for the development of complex animal model systems, indicating its potential as a therapeutic tool for both genetic and non-genetic diseases, although it remains in the early stages of clinical application (Cribbs & Perera, 2017) [28].

The ethical considerations surrounding CRISPR technology are multifaceted and often contentious, particularly regarding human germline modification. This technology poses significant ethical dilemmas related to safety, efficacy, human dignity, and the implications of human genetic enhancement. Concerns arise from the potential for heritable changes in the human genome, which necessitates thorough discussion and consensus across various ethical and cultural perspectives. For instance, Islamic ethical frameworks highlight principles such as the preservation of human life and dignity, suggesting that germline editing may be permissible under specific medical circumstances but should not proceed until safety and efficacy are assured (Isa et al., 2020) [34].

Furthermore, there are societal implications tied to the use of CRISPR technology. The power of CRISPR raises urgent questions regarding governance, equity, and access to the technology. Concerns have been raised about the potential for germline enhancement and eugenic practices, particularly if access to such technologies becomes stratified along socioeconomic lines. The case of embryo editing exemplifies how unregulated applications can erode public trust, highlighting the necessity for robust ethical guidelines and regulations to prevent misuse (Biswas, 2025) [32].

Public perception and acceptance of CRISPR technology are influenced by these ethical and regulatory challenges. As the technology progresses, it is crucial for medical professionals to engage in informed discussions with patients regarding the nuances, limitations, and potential barriers to access related to CRISPR applications. The need for transparent, collaborative, and regulated research frameworks is paramount to navigate the societal implications of this powerful technology, ensuring that its benefits are realized without compromising ethical standards or human rights (Kofler & Kraschel, 2018) [35].

In conclusion, while CRISPR-Cas9 gene editing holds immense promise for transforming therapeutic strategies, particularly for heritable diseases, it also necessitates careful consideration of ethical, regulatory, and societal dimensions to foster public trust and ensure responsible use of the technology.

5 Current Limitations and Future Directions

5.1 Off-Target Effects and Precision

CRISPR gene editing operates primarily through the CRISPR/Cas9 system, which is derived from the bacterial immune response. This system utilizes a guide RNA (gRNA) that is designed to target a specific DNA sequence within the genome. The Cas9 endonuclease is then directed by the gRNA to create double-strand breaks (DSBs) at the target site. Following this, the cell's endogenous DNA repair mechanisms can either insert, delete, or substitute genetic material, thus facilitating precise genetic modifications.

Despite the transformative potential of CRISPR/Cas9, several limitations impede its widespread application, particularly concerning off-target effects. Off-target effects refer to unintended mutations that occur at sites other than the intended target, potentially leading to genomic instability and adverse outcomes. These effects pose significant challenges for the safety and efficacy of CRISPR technologies, especially in clinical settings. Studies have reported that off-target activity can exceed 50%, raising concerns about the reliability of CRISPR for therapeutic applications (Zhang et al., 2015) [36].

Current strategies to mitigate off-target effects include the engineering of high-fidelity Cas9 variants, optimized gRNA design, and the development of alternative editing techniques such as base editing and prime editing. Enhanced delivery mechanisms, including the use of nanoparticles, have also been explored to improve the specificity and efficiency of CRISPR applications [37][38].

Future directions in CRISPR research emphasize the need for advanced computational models and multi-modal detection systems to accurately assess off-target effects in vivo. There is also a pressing need for standardized methodologies to evaluate off-target activity, which could facilitate more reliable and reproducible results across different studies [39][40]. Moreover, integrating CRISPR with other therapeutic modalities, such as photodynamic therapy and stem cell therapies, is being investigated to enhance the precision and effectiveness of cancer treatments while addressing off-target concerns [37].

In conclusion, while CRISPR/Cas9 technology has revolutionized gene editing, addressing off-target effects remains a critical challenge. Ongoing research is focused on refining CRISPR systems to improve specificity, enhance detection methods, and ensure the safe application of this powerful technology in therapeutic contexts.

5.2 Advances in CRISPR Technology

CRISPR gene editing operates primarily through the CRISPR/Cas9 system, which is a powerful tool derived from the adaptive immune systems of bacteria. This technology utilizes a guide RNA (gRNA) to direct the Cas9 nuclease to a specific location in the genome, where it introduces a double-strand break (DSB) in the DNA. The cell's natural repair mechanisms then kick in, either repairing the break through non-homologous end joining (NHEJ), which can lead to insertions or deletions (indels), or homology-directed repair (HDR) if a repair template is provided. This allows for precise modifications of the genome, making it a valuable tool for both research and therapeutic applications[3].

Despite its revolutionary potential, CRISPR technology is not without limitations. Key challenges include off-target effects, where unintended regions of the genome are edited, and issues related to delivery systems that can hinder the efficiency of CRISPR applications in vivo. Moreover, the long-term safety and ethical implications of germline editing pose significant concerns. Current research is focused on overcoming these obstacles, with innovations aimed at enhancing the specificity and controllability of CRISPR systems. For instance, advancements in CRISPR interference (CRISPRi) and CRISPR activation (CRISPRa) have been developed to facilitate reversible gene regulation without permanent alterations to the DNA[41].

Future directions in CRISPR technology include the development of third-generation gene editing tools, such as base editors and prime editors, which offer greater precision by enabling targeted base substitutions without generating DSBs. These advancements aim to minimize the risks associated with traditional CRISPR methods and expand the scope of possible applications, including in gene therapy for complex diseases. Researchers are also exploring conditional control strategies, employing chemically modified oligonucleotides to achieve spatiotemporal regulation of CRISPR activity, thereby enhancing its therapeutic potential[42][43].

In summary, CRISPR gene editing has transformed the landscape of genetic manipulation, offering precise editing capabilities that are continuously being refined to address existing limitations. The ongoing advancements in CRISPR technology promise to unlock new therapeutic avenues, potentially leading to significant breakthroughs in the treatment of genetic disorders and other diseases[44][45].

5.3 Future Prospects in Research and Medicine

CRISPR gene editing technology, specifically the CRISPR/Cas9 system, has revolutionized the field of genetic engineering due to its remarkable efficiency and programmability. The mechanism involves the use of a guide RNA (gRNA) that directs the Cas9 nuclease to a specific sequence of DNA, allowing for precise cuts in the genome. Once the DNA is cleaved, the cell's natural repair mechanisms can be harnessed to introduce specific changes, such as gene knockout, insertion, or modification. This technology has been instrumental in advancing our understanding of genetic diseases and developing therapeutic interventions.

However, despite its transformative potential, CRISPR technology faces several current limitations. Key issues include off-target effects, where unintended parts of the genome are altered, and a lack of control over editing activity. These limitations pose significant challenges, particularly for therapeutic applications in humans. For instance, the efficiency of DNA repair outcomes and the delivery of CRISPR components to target cells remain problematic, with inadequate delivery performance being a notable barrier to clinical efficacy (Wegrzyn et al., 2018; Lee & Kim, 2019). Furthermore, immune responses against the CRISPR components can complicate treatment and lead to adverse effects (Hirakawa et al., 2020).

Looking ahead, the future prospects of CRISPR in research and medicine are promising, with ongoing advancements aimed at overcoming existing limitations. Innovations in CRISPR technology, such as base editing and prime editing, enhance the precision and flexibility of gene modifications, potentially reducing off-target effects and improving safety profiles (Hillary et al., 2025). Additionally, efforts are being made to develop more effective delivery systems for CRISPR components, which are crucial for successful in vivo applications (Huang et al., 2022). As research progresses, the integration of CRISPR technology into clinical settings is expected to expand, with applications ranging from cancer therapies to genetic disease treatments (Cetin et al., 2025).

In summary, while CRISPR gene editing holds tremendous potential for both basic research and therapeutic applications, addressing its limitations through ongoing technological innovations and enhanced delivery methods will be essential for unlocking its full capabilities in medicine. The landscape of gene editing is rapidly evolving, and with it, the promise of CRISPR to transform healthcare practices and improve patient outcomes.

6 Conclusion

The CRISPR gene editing technology has ushered in a new era in genetics, characterized by its precision and versatility in modifying DNA across various organisms. This review has highlighted several key findings regarding the operational mechanisms of CRISPR-Cas systems, including the intricate processes of guide RNA design, target recognition, and the subsequent DNA repair pathways (NHEJ and HDR). Notably, CRISPR's therapeutic applications have shown promise in addressing genetic disorders, enhancing cancer treatments, and improving agricultural traits, thereby underscoring its transformative potential in multiple fields. However, the technology is not without its challenges, particularly concerning off-target effects and ethical implications surrounding germline editing. As the regulatory landscape evolves, it is imperative to establish robust frameworks that address these ethical concerns while fostering public trust in CRISPR applications. Looking ahead, future research should focus on enhancing the precision and safety of CRISPR technologies, exploring innovative delivery methods, and expanding its applications in both therapeutic and industrial contexts. By addressing current limitations and embracing ethical considerations, CRISPR can continue to revolutionize the fields of medicine and biotechnology, ultimately improving human health and food security.

References

  • [1] Yuanyuan Xu;Zhanjun Li. CRISPR-Cas systems: Overview, innovations and applications in human disease research and gene therapy.. Computational and structural biotechnology journal(IF=4.1). 2020. PMID:33005303. DOI: 10.1016/j.csbj.2020.08.031.
  • [2] Jeong H Joo;Soogene Lee;Keun P Kim. Precision gene editing: The power of CRISPR-Cas in modern genetics.. Molecular therapy. Nucleic acids(IF=6.1). 2025. PMID:41210586. DOI: 10.1016/j.omtn.2025.102733.
  • [3] Debarati Ghosh;Prabhadevi Venkataramani;Saikat Nandi;Sonali Bhattacharjee. CRISPR-Cas9 a boon or bane: the bumpy road ahead to cancer therapeutics.. Cancer cell international(IF=6.0). 2019. PMID:30636933. DOI: 10.1186/s12935-019-0726-0.
  • [4] Salah Adlat;Alexandra M Vázquez Salgado;Markcus Lee;Dingzi Yin;Kirk J Wangensteen. Emerging and potential use of CRISPR in human liver disease.. Hepatology (Baltimore, Md.)(IF=15.8). 2025. PMID:37607734. DOI: 10.1097/HEP.0000000000000578.
  • [5] Marina Martín-Valmaseda;Sama Rahimi Devin;Germán Ortuño-Hernández;Cristian Pérez-Caselles;Sayyed Mohammad Ehsan Mahdavi;Geza Bujdoso;Juan Alfonso Salazar;Pedro Martínez-Gómez;Nuria Alburquerque. CRISPR/Cas as a Genome-Editing Technique in Fruit Tree Breeding.. International journal of molecular sciences(IF=4.9). 2023. PMID:38068981. DOI: 10.3390/ijms242316656.
  • [6] Nadja Bischoff;Sandra Wimberger;Marcello Maresca;Cord Brakebusch. Improving Precise CRISPR Genome Editing by Small Molecules: Is there a Magic Potion?. Cells(IF=5.2). 2020. PMID:32466303. DOI: 10.3390/cells9051318.
  • [7] Lorenzo Casalino;Łukasz Nierzwicki;Martin Jinek;Giulia Palermo. Catalytic Mechanism of Non-Target DNA Cleavage in CRISPR-Cas9 Revealed by Ab Initio Molecular Dynamics.. ACS catalysis(IF=13.1). 2020. PMID:33520346. DOI: 10.1021/acscatal.0c03566.
  • [8] Vinay Kumar;Mukesh Jain. The CRISPR-Cas system for plant genome editing: advances and opportunities.. Journal of experimental botany(IF=5.7). 2015. PMID:25371501. DOI: 10.1093/jxb/eru429.
  • [9] Shaked Bergman;Tamir Tuller. Strong association between genomic 3D structure and CRISPR cleavage efficiency.. PLoS computational biology(IF=3.6). 2024. PMID:38848440. DOI: 10.1371/journal.pcbi.1012214.
  • [10] Jeongmin Lee;Cherlhyun Jeong. Single-molecule perspectives of CRISPR/Cas systems: target search, recognition, and cleavage.. BMB reports(IF=3.3). 2025. PMID:39701024. DOI: .
  • [11] Robert M Yarrington;Surbhi Verma;Shaina Schwartz;Jonathan K Trautman;Dana Carroll. Nucleosomes inhibit target cleavage by CRISPR-Cas9 in vivo.. Proceedings of the National Academy of Sciences of the United States of America(IF=9.1). 2018. PMID:30201707. DOI: 10.1073/pnas.1810062115.
  • [12] Won Jun Jung;Soo-Ji Park;Seongkwang Cha;Kyoungmi Kim. Factors affecting the cleavage efficiency of the CRISPR-Cas9 system.. Animal cells and systems(IF=3.2). 2024. PMID:38440123. DOI: 10.1080/19768354.2024.2322054.
  • [13] Hao Chen;Matthew Neubauer;Jack P Wang. Enhancing HR Frequency for Precise Genome Editing in Plants.. Frontiers in plant science(IF=4.8). 2022. PMID:35592579. DOI: 10.3389/fpls.2022.883421.
  • [14] Sibtain Haider;Claudio Mussolino. Fine-Tuning Homology-Directed Repair (HDR) for Precision Genome Editing: Current Strategies and Future Directions.. International journal of molecular sciences(IF=4.9). 2025. PMID:40362308. DOI: 10.3390/ijms26094067.
  • [15] Lupeng Ye;Chengkun Wang;Lingjuan Hong;Ninghe Sun;Danyang Chen;Sidi Chen;Feng Han. Programmable DNA repair with CRISPRa/i enhanced homology-directed repair efficiency with a single Cas9.. Cell discovery(IF=12.5). 2018. PMID:30062046. DOI: 10.1038/s41421-018-0049-7.
  • [16] Antonio Carusillo;Sibtain Haider;Raul Schäfer;Manuel Rhiel;Daniel Türk;Kay O Chmielewski;Julia Klermund;Laura Mosti;Geoffroy Andrieux;Richard Schäfer;Tatjana I Cornu;Toni Cathomen;Claudio Mussolino. A novel Cas9 fusion protein promotes targeted genome editing with reduced mutational burden in primary human cells.. Nucleic acids research(IF=13.1). 2023. PMID:37070192. DOI: 10.1093/nar/gkad255.
  • [17] Tomoko Kato-Inui;Gou Takahashi;Szuyin Hsu;Yuichiro Miyaoka. Clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated protein 9 with improved proof-reading enhances homology-directed repair.. Nucleic acids research(IF=13.1). 2018. PMID:29672770. DOI: 10.1093/nar/gky264.
  • [18] Tarun S Nambiar;Pierre Billon;Giacomo Diedenhofen;Samuel B Hayward;Angelo Taglialatela;Kunheng Cai;Jen-Wei Huang;Giuseppe Leuzzi;Raquel Cuella-Martin;Andrew Palacios;Anuj Gupta;Dieter Egli;Alberto Ciccia. Stimulation of CRISPR-mediated homology-directed repair by an engineered RAD18 variant.. Nature communications(IF=15.7). 2019. PMID:31363085. DOI: 10.1038/s41467-019-11105-z.
  • [19] Aimee Malzahn;Levi Lowder;Yiping Qi. Plant genome editing with TALEN and CRISPR.. Cell & bioscience(IF=6.2). 2017. PMID:28451378. DOI: 10.1186/s13578-017-0148-4.
  • [20] Katherine S Pawelczak;Navnath S Gavande;Pamela S VanderVere-Carozza;John J Turchi. Modulating DNA Repair Pathways to Improve Precision Genome Engineering.. ACS chemical biology(IF=3.8). 2018. PMID:29210569. DOI: 10.1021/acschembio.7b00777.
  • [21] S Antony Ceasar;S Ignacimuthu. CRISPR/Cas genome editing in plants: Dawn of Agrobacterium transformation for recalcitrant and transgene-free plants for future crop breeding.. Plant physiology and biochemistry : PPB(IF=5.7). 2023. PMID:36812799. DOI: 10.1016/j.plaphy.2023.02.030.
  • [22] Workie Anley Zegeye;Mesfin Tsegaw;Yingxin Zhang;Liyong Cao. CRISPR-Based Genome Editing: Advancements and Opportunities for Rice Improvement.. International journal of molecular sciences(IF=4.9). 2022. PMID:35457271. DOI: 10.3390/ijms23084454.
  • [23] Qiang Li;Manoj Sapkota;Esther van der Knaap. Perspectives of CRISPR/Cas-mediated cis-engineering in horticulture: unlocking the neglected potential for crop improvement.. Horticulture research(IF=8.5). 2020. PMID:32194972. DOI: 10.1038/s41438-020-0258-8.
  • [24] Mohamed Atia;Wenjun Jiang;Khalid Sedeek;Haroon Butt;Magdy Mahfouz. Crop bioengineering via gene editing: reshaping the future of agriculture.. Plant cell reports(IF=4.5). 2024. PMID:38494539. DOI: 10.1007/s00299-024-03183-1.
  • [25] Suhyung Cho;Jongoh Shin;Byung-Kwan Cho. Applications of CRISPR/Cas System to Bacterial Metabolic Engineering.. International journal of molecular sciences(IF=4.9). 2018. PMID:29621180. DOI: 10.3390/ijms19041089.
  • [26] Shangjie Zhang;Feng Guo;Wei Yan;Zhongxue Dai;Weiliang Dong;Jie Zhou;Wenming Zhang;Fengxue Xin;Min Jiang. Recent Advances of CRISPR/Cas9-Based Genetic Engineering and Transcriptional Regulation in Industrial Biology.. Frontiers in bioengineering and biotechnology(IF=4.8). 2019. PMID:32047743. DOI: 10.3389/fbioe.2019.00459.
  • [27] Guilherme Souza Prado;Dhiôvanna Corrêia Rocha;Lucas Nascimento Dos Santos;Danyel Fernandes Contiliani;Paula Macedo Nobile;Juliana Camargo Martinati-Schenk;Lilian Padilha;Mirian Perez Maluf;Greice Lubini;Tiago Campos Pereira;Claudia Barros Monteiro-Vitorello;Silvana Creste;Raquel Luciana Boscariol-Camargo;Marco Aurélio Takita;Mariângela Cristofani-Yaly;Alessandra Alves de Souza. CRISPR technology towards genome editing of the perennial and semi-perennial crops citrus, coffee and sugarcane.. Frontiers in plant science(IF=4.8). 2023. PMID:38259920. DOI: 10.3389/fpls.2023.1331258.
  • [28] Adam P Cribbs;Sumeth M W Perera. Science and Bioethics of CRISPR-Cas9 Gene Editing: An Analysis Towards Separating Facts and Fiction.. The Yale journal of biology and medicine(IF=3.9). 2017. PMID:29259526. DOI: .
  • [29] Zabta Khan Shinwari;Faouzia Tanveer;Ali Talha Khalil. Ethical Issues Regarding CRISPR Mediated Genome Editing.. Current issues in molecular biology(IF=3.0). 2018. PMID:28879860. DOI: 10.21775/cimb.026.103.
  • [30] R Vassena;B Heindryckx;R Peco;G Pennings;A Raya;K Sermon;A Veiga. Genome engineering through CRISPR/Cas9 technology in the human germline and pluripotent stem cells.. Human reproduction update(IF=16.1). 2016. PMID:26932460. DOI: 10.1093/humupd/dmw005.
  • [31] Erika Kleiderman;Ubaka Ogbogu. Realigning gene editing with clinical research ethics: What the "CRISPR Twins" debacle means for Chinese and international research ethics governance.. Accountability in research(IF=4.0). 2019. PMID:31068009. DOI: 10.1080/08989621.2019.1617138.
  • [32] Irfan Biswas. Ethical dimensions and societal implications: ensuring the social responsibility of CRISPR technology.. Frontiers in genome editing(IF=4.4). 2025. PMID:40909132. DOI: 10.3389/fgeed.2025.1593172.
  • [33] Pegah Kavousinia;Mohammad Hossein Ahmadi;Hamid Sadeghian;Mahdi Hosseini Bafghi. Therapeutic potential of CRISPR/CAS9 genome modification in T cell-based immunotherapy of cancer.. Cytotherapy(IF=3.2). 2024. PMID:38466263. DOI: 10.1016/j.jcyt.2024.02.014.
  • [34] Noor Munirah Isa;Nurul Atiqah Zulkifli;Saadan Man. Islamic Perspectives on CRISPR/Cas9-Mediated Human Germline Gene Editing: A Preliminary Discussion.. Science and engineering ethics(IF=3.0). 2020. PMID:30830592. DOI: 10.1007/s11948-019-00098-z.
  • [35] Natalie Kofler;Katherine L Kraschel. Treatment of heritable diseases using CRISPR: Hopes, fears, and reality.. Seminars in perinatology(IF=3.2). 2018. PMID:30420296. DOI: 10.1053/j.semperi.2018.09.012.
  • [36] Xiao-Hui Zhang;Louis Y Tee;Xiao-Gang Wang;Qun-Shan Huang;Shi-Hua Yang. Off-target Effects in CRISPR/Cas9-mediated Genome Engineering.. Molecular therapy. Nucleic acids(IF=6.1). 2015. PMID:26575098. DOI: 10.1038/mtna.2015.37.
  • [37] J P Jose Merlin;Heidi Abrahamse. Optimizing CRISPR/Cas9 precision: Mitigating off-target effects for safe integration with photodynamic and stem cell therapies in cancer treatment.. Biomedicine & pharmacotherapy = Biomedecine & pharmacotherapie(IF=7.5). 2024. PMID:39332185. DOI: 10.1016/j.biopha.2024.117516.
  • [38] Muna Alariqi;Mohamed Ramadan;Lu Yu;Fengjiao Hui;Amjad Hussain;Xiaofeng Zhou;Yu Yu;Xianlong Zhang;Shuangxia Jin. Enhancing Specificity, Precision, Accessibility, Flexibility, and Safety to Overcome Traditional CRISPR/Cas Editing Challenges and Shape Future Innovations.. Advanced science (Weinheim, Baden-Wurttemberg, Germany)(IF=14.1). 2025. PMID:40548648. DOI: 10.1002/advs.202416331.
  • [39] Nechama Kalter;Carla Fuster-García;Alfredo Silva;Víctor Ronco-Díaz;Stefano Roncelli;Giandomenico Turchiano;Jan Gorodkin;Toni Cathomen;Karim Benabdellah;Ciaran Lee;Ayal Hendel. Off-target effects in CRISPR-Cas genome editing for human therapeutics: Progress and challenges.. Molecular therapy. Nucleic acids(IF=6.1). 2025. PMID:40777742. DOI: 10.1016/j.omtn.2025.102636.
  • [40] Ying-Ying Xu;Sheng-Mei Zhou;Lu-Yan Wang;Rong Zhang;Kai Li;Zhi-Yuan Qian;Li Xiao. Methods for detecting off-target effects of CRISPR/Cas9.. Biotechnology advances(IF=12.5). 2026. PMID:41213415. DOI: 10.1016/j.biotechadv.2025.108750.
  • [41] Lingling Jiao;Qi Zhou;Dongchang Sun. CRISPR-Based Regulation for High-Throughput Screening.. ACS synthetic biology(IF=3.9). 2025. PMID:40401794. DOI: 10.1021/acssynbio.5c00076.
  • [42] Pushpendra K Gupta;Sourabh Kumar. Third-generation novel technologies for gene editing.. Trends in biotechnology(IF=14.9). 2025. PMID:40738762. DOI: 10.1016/j.tibtech.2025.07.012.
  • [43] Busra Cetin;Fulya Erendor;Yunus Emre Eksi;Ahter D Sanlioglu;Salih Sanlioglu. Advancing CRISPR genome editing into gene therapy clinical trials: progress and future prospects.. Expert reviews in molecular medicine(IF=5.5). 2025. PMID:40160040. DOI: 10.1017/erm.2025.10.
  • [44] Martin Pacesa;Oana Pelea;Martin Jinek. Past, present, and future of CRISPR genome editing technologies.. Cell(IF=42.5). 2024. PMID:38428389. DOI: 10.1016/j.cell.2024.01.042.
  • [45] Renee D Wegrzyn;Andrew H Lee;Amy L Jenkins;Colby D Stoddard;Anne E Cheever. Genome Editing: Insights from Chemical Biology to Support Safe and Transformative Therapeutic Applications.. ACS chemical biology(IF=3.8). 2018. PMID:28992411. DOI: 10.1021/acschembio.7b00689.

MaltSci Intelligent Research Services

Search for more papers on MaltSci.com

CRISPR gene editing · CRISPR-Cas9 mechanism · gene therapy · agricultural biotechnology · ethical challenges


© 2025 MaltSci