Get Permission Chaudhari and Ranjan: Advances in CRISPR-based technologies for genome editing in microorganisms


Introduction

CRISPR, or clustered regularly interspaced short palindromic repeats, is a ground-breaking technology in molecular biology that has completely changed the way we can work with genetic material. CRISPR was first identified in bacteria as a component of their adaptive immune system. Since then, it has been used and modified to do precise genome editing in a variety of taxa, including microbes, plants, and animals. Its remarkable accuracy, effectiveness, and adaptability make it significant because they enable scientists to edit genes more precisely than they have in the past.1 The natural function of CRISPR in bacteria and archaea as an adaptive defensive mechanism against invasive viruses accounts for much of its significance. Together, the system's linked proteins (Cas proteins) and short, partly palindromic DNA sequences (CRISPRs) make up a potent molecular toolbox. A tiny portion of the viral DNA is retained by the bacteria in its CRISPR region when it overcomes a viral assault. This acts as a chemical memory, making it easier for the bacteria to identify and fight off viral infections in the future. The versatility of CRISPR for genome editing is what makes it revolutionary. 1 The CRISPR-Cas system has been repurposed by scientists to precisely target and change certain genes under controlled conditions. This skill has broad applications in biotechnology, health, agriculture, and environmental science, among other domains. 2 We shall examine the significant influence of CRISPR-based technologies in this review paper, presenting them as ground-breaking instruments in the field of molecular biology. This paper presents a historical account of the evolution of CRISPR from its discovery to its current position as a vital tool for precisely editing the genomes of microorganisms. The focus is on the special qualities that make CRISPR a revolutionary tool in the fields of molecular biology and microbiology.

Discussion

CRISPR systems in microorganisms

The complex yet interesting mosaic of CRISPR systems that are inherent to microorganisms reveals the advanced machinery that bacteria, archaea, and other microbial domains use as part of their adaptive defense mechanisms. The CRISPR-Cas (CRISPR-associated) system, a strong and adaptable weapon that microorganisms have evolved to ward off viral invaders, is at the center of this complex system. 2 Within the microbial realm, bacteria and archaea are not inert entities; rather, they are genetic designers with the ability to retain a molecular memory of previous interactions with viral attackers. This is the situation in which CRISPR-Cas systems become useful, functioning as a type of genetic immunological memory that enables microbes to identify and develop a response against certain viral threats. 2 The CRISPR array, a group of short, partly palindromic DNA sequences, and the Cas proteins, which act as molecular scissors or effectors, are the two primary parts of the CRISPR-Cas system. The CRISPR array functions as a sort of memory bank, holding onto bits of DNA from viruses that the microbe has already come into contact with. When a microorganism survives an encounter with a virus, it retains a small piece of the viral DNA within its CRISPR array. 3 This molecular memory enables the microorganism to recognize the virus upon subsequent infections. In response to a viral threat, the Cas proteins are deployed to search for and bind to the matching viral DNA sequences within the CRISPR array. Upon recognition, the Cas proteins function as molecular scissors, cleaving the viral DNA and neutralizing the viral threat. This process, known as "targeted cleavage" or "DNA interference," is a decisive and precise mechanism employed by Cas proteins. 3 The recognition is facilitated by the guide RNA, a molecular beacon that guides the Cas proteins to the specific DNA sequence corresponding to the viral invader stored in the CRISPR array. Once the Cas proteins reach the identified viral DNA sequence, they undergo a conformational change that activates their enzymatic activity. This activation transforms them into molecular scissors, capable of cleaving the viral DNA at the precise location dictated by the guide RNA. The cleavage disrupts the integrity of the viral genome, rendering it nonfunctional. 4 The cleavage of viral DNA serves a dual purpose: it neutralizes the immediate threat by preventing the virus from replicating within the microbial host, and it also reinforces the memory within the CRISPR array. The microbial host is left with a fragmented piece of the viral DNA, a molecular reminder of its prior encounter. 4 This ensures a heightened and more efficient defense in the event of a subsequent viral attack, as the microorganism can swiftly recognize and target the invader. Following the cleavage, cellular repair mechanisms come into play to fix the double-strand breaks induced by the Cas proteins. This repair process can be error-prone, leading to mutations or disruptions in the viral DNA. Over time, this adaptation contributes to the evolutionary dynamics of both microorganisms and viruses, influencing the ongoing arms race between them. 2 The versatility of Cas proteins in targeting and cleaving specific DNA sequences extends beyond viral defense. In the context of CRISPR-based genome editing technologies, researchers have repurposed this natural system to precisely modify genes in a variety of organisms. By designing synthetic guide RNAs, scientists can program Cas proteins to target and cleave specific genes of interest, enabling precise genetic modifications.

Figure 1

CRISPR technology

https://s3-us-west-2.amazonaws.com/typeset-prod-media-server/0aaabc5f-d109-43b3-83a9-cd3df8c0acf3image1.png

CRISPR-Cas9 and beyond

CRISPR-Cas9 is renowned for its ability to precisely target specific DNA sequences within a genome. 5 The Cas9 protein acts as a molecular scissor guided by a single-guide RNA (sgRNA) to recognize and cleave the targeted DNA sequence. The adaptability of CRISPR-Cas9 to target different genes in various organisms makes it a versatile tool in genetic engineering. 6 It has been successfully applied across a wide range of species, from bacteria and yeast to plants and animals. Designing a guide RNA for a specific target gene is relatively straightforward, allowing researchers to easily customize CRISPR-Cas9 for diverse applications. This simplicity contributes to the widespread adoption of CRISPR-Cas9 in laboratories worldwide.CRISPR-Cas9 is scalable, allowing simultaneous editing of multiple genes within a single organism. This feature is particularly advantageous for studies investigating complex genetic interactions. 7 Thus it holds tremendous promise for therapeutic interventions, such as gene therapy. It allows for the correction of genetic mutations associated with various diseases, offering potential cures for conditions that were once considered incurable.CRISPR-Cas9 provides a high degree of precision, enabling targeted modifications at specific genomic loci. 8 This specificity reduces the risk of off-target effects, enhancing the accuracy of genome editing. The efficiency of CRISPR-Cas9 in inducing DNA cleavage and subsequent genetic modifications is notable. This efficiency streamlines the genome editing process, making it a preferred choice for researchers and scientists. Compared to previous genome editing techniques, CRISPR-Cas9 is relatively cost-effective. 8 The ease of design, simplicity of the technology, and the availability of commercial CRISPR tools contribute to its cost-effectiveness. Further modifications to this have helped evolve CRISPR-Cas12 and CRISPR-Cas13 systems. CRISPR-Cas12 exemplified by Cas12a (formerly known as Cpf1), offer distinct advantages. Cas12 proteins have different protospacer adjacent motif (PAM) requirements compared to Cas9, expanding the range of targetable sequences. 9 Additionally, Cas12 exhibits collateral cleavage activity, enhancing its potential for diagnostic applications.CRISPR-Cas13 systems, identified in RNA-targeting applications, can specifically target and cleave RNA molecules. Cas13's ability to target RNA expands the CRISPR toolkit to address genetic information at the RNA level, offering potential applications in RNA interference and antiviral strategies. 9 While CRISPR-Cas9 primarily targets DNA, the newer Cas12 and Cas13 systems enable RNA targeting and open avenues for RNA editing applications. This expansion broadens the spectrum of genetic manipulations and therapeutic interventions. 10 The collateral cleavage activity of Cas12 has been harnessed for diagnostic applications, allowing the detection of specific DNA sequences. This capability has potential applications in point-of-care diagnostics.Cas12 and Cas13 systems enhance the potential for multiplex genome editing, enabling simultaneous modifications of multiple genes or RNA molecules within the same organism.

Precision and efficiency improvements

Researchers have focused on enhancing the precision and efficiency of CRISPR-based genome editing by engineering and modifying Cas proteins. These modifications aim to improve target specificity, reduce off-target effects, and provide additional functionalities. 11 Variants of Cas9, such as eSpCas9 and HypaCas9, exhibit increased specificity, lowering the likelihood of unintended modifications. The design and optimization of sgRNAs play a crucial role in improving the precision of CRISPR-based editing. 12 Advances in sgRNA design algorithms and structure prediction tools enable the creation of highly specific and efficient guides. Additionally, synthetic sgRNAs with modified structures or chemical modifications can enhance stability, promoting reliable binding to target DNA sequences. Efficient delivery of CRISPR components into target cells is essential for successful genome editing. Recent advancements in delivery methods have focused on improving the targeted delivery of Cas proteins and guide RNAs. 13 Viral vectors, lipid nanoparticles, and electroporation techniques have been optimized to enhance delivery efficiency and reduce cellular toxicity. Tailoring Cas proteins for specific applications has become a recent trend. Engineered Cas proteins, such as enhanced versions of Cas12 (e.g., evoCas12a) and Cas13 (e.g., evolved Cas13 variants), exhibit improved features for precise genome editing and RNA targeting, respectively. 14 These optimized proteins enhance the overall efficiency of CRISPR-based technologies. Base editing and prime editing are innovative approaches that aim to achieve more precise modifications at the nucleotide level. Base editing allows the direct conversion of one DNA base pair into another without inducing double-strand breaks, while prime editing enables the targeted insertion or deletion of genetic material with minimal off-target effects. These advancements represent significant steps towards achieving highly accurate and controlled genome editing. 14 The development of CRISPR screens, such as pooled CRISPR libraries, has facilitated large-scale functional genomics studies. These screens enable the simultaneous targeting of multiple genes, providing insights into gene functions, pathways, and potential therapeutic targets. Optimizations in library design and screening methodologies contribute to the precision and reliability of functional genomics research. 5 Advances in high-throughput technologies and automation have streamlined the CRISPR-based genome editing process. Automated systems for CRISPR design, delivery, and screening enhance the efficiency of experiments and reduce variability, contributing to the reliability of results. 1 Machine learning algorithms are being employed to predict and optimize CRISPR target sites, guide RNA designs, and Cas protein variants. These computational approaches aid researchers in selecting the most effective components for their specific genome editing goals, contributing to increased precision.

Challenges in microbial genome editing efficiently

Delivering CRISPR components into microorganisms can be challenging, especially in complex microbial communities or environments. Traditional delivery methods may not effectively reach all target cells, leading to suboptimal editing efficiency. 15 Unintended modifications in non-targeted genomic regions, known as off-target effects, pose a significant challenge. These off-target effects can compromise the accuracy and safety of genome editing. 16 Microbial genomes often exhibit complexity, including repetitive sequences, high guanine-cytosine content, and unique structural features. These complexities can hinder the precise targeting and editing of specific genomic loci. In certain environments, microorganisms may reside in intricate niches or biofilms, making it challenging to deliver CRISPR components to every target cell. 17 Also, Microorganisms may develop resistance to CRISPR-based interventions over time, impacting the long-term effectiveness of genome editing strategies. 18 The ethical implications of genome editing in microorganisms, especially those used in industrial or environmental applications, raise concerns about unintended consequences and ecosystem impacts. However by adopting the certain strategies such as improved vectors and nanoparticle delivery the above challenges can be overcome.

Applications in biotechnology, medicine, and environmental science

CRISPR has revolutionized biotechnology by enabling the precise modification of microbial genomes for enhanced production of biofuels, pharmaceuticals, and industrial enzymes. The technology allows for the optimization of metabolic pathways and the creation of strains with improved characteristics for industrial processes. 16 CRISPR-based genome editing is employed to optimize microorganisms for enhanced biofuel production. Microbes, such as bacteria and yeast, can be engineered to improve their metabolic pathways, increase substrate utilization efficiency, and boost the overall yield of biofuels like ethanol and biodiesel. 17 In the medical field, CRISPR holds enormous promise for gene therapy and the treatment of genetic disorders.19 It allows scientists to correct or replace faulty genes associated with diseases, offering potential cures for conditions that were once considered incurable. CRISPR facilitates the development of genetically modified crops with improved traits such as disease resistance, drought tolerance, and enhanced nutritional content. 17 This has implications for global food security and sustainable agriculture. CRISPR is an invaluable tool in basic research, enabling scientists to study gene function by selectively activating, deactivating, or modifying specific genes. 15 This has accelerated the pace of biological research, leading to a deeper understanding of fundamental biological processes. CRISPR is being explored for therapeutic applications beyond genetic disorders, including the development of cancer immunotherapies, where it is used to engineer immune cells for more effective targeting of cancer cells. 16 CRISPR technologies can be employed in environmental applications, such as the engineering of microorganisms for pollution remediation and the modification of plants to thrive in challenging environmental conditions.

To identify the precise genetic mutations causing cancer and understand their mechanisms, CRISPR/Cas9 gene editing for tumor cells and immune cells holds great potential in addition to traditional therapeutic approaches like surgery, radiotherapy, chemotherapy, and immunotherapy. Because of the genetic precision that CRISPR/Cas9 provides, treating cancers effectively may now be approached from a different perspective 20, 21

Ethical considerations and regulatory frameworks

As we delve deeper into the molecular intricacies of the CRISPR Technology ethical considerations emerge. CRISPR-based genome editing in microorganisms may lead to unintended consequences, including off-target effects, ecological disturbances, or the unintended alteration of non-targeted genes. The potential for unintended outcomes raises ethical concerns about the unpredictable impacts on microbial communities and ecosystems. 12 The release of genetically modified microorganisms into the environment raises ethical questions about the potential for uncontrolled spread and ecological disruption. Containment measures and risk assessments are crucial to prevent unintended consequences and maintain environmental integrity. The dual-use nature of CRISPR technology raises ethical dilemmas, as the same techniques employed for beneficial purposes could potentially be misused for harmful applications. The intentional engineering of microorganisms for malicious purposes, such as bioweapons, underscores the need for ethical guidelines and responsible oversight. 13 The long-term effects of CRISPR-based genome editing on microbial ecosystems are not fully understood. Ethical considerations include the responsibility to conduct thorough risk assessments, monitor the persistence of modified organisms, and address any unforeseen consequences that may emerge over time. 17 The equitable access to CRISPR technologies and their benefits, especially in the context of agricultural or industrial applications, raises ethical questions. Ensuring that the benefits of genome editing are distributed fairly and do not exacerbate existing social or economic inequalities is a key consideration. addressing the ethical considerations surrounding CRISPR-based genome editing in microorganisms requires a multifaceted and adaptive approach. 17 Combining transparent communication, community engagement, regulatory frameworks, ethical impact assessments, and global collaboration can help navigate the ethical complexities associated with this powerful technology. A responsible and ethical use of CRISPR technologies contributes to their potential benefits while minimizing potential risks and unintended consequences.

Figure 2

Applications and limitations of CRISPR technology

https://s3-us-west-2.amazonaws.com/typeset-prod-media-server/0aaabc5f-d109-43b3-83a9-cd3df8c0acf3image2.png

Future perspectives

Ongoing research aims to enhance the precision of CRISPR-based genome editing in microorganisms. 22 This includes the development of advanced CRISPR systems with improved target specificity, reduced off-target effects, and enhanced control over the editing process at the nucleotide level. Researchers are exploring ways to expand the targeting range of CRISPR systems to address genomic regions that were previously challenging to modify. This includes the development of novel Cas proteins and engineering existing ones to recognize a broader range of DNA sequences. 15 Advancements in epigenome editing tools within the CRISPR framework hold promise for regulating gene expression without altering the underlying DNA sequence. This could enable fine-tuned control over microbial traits, metabolic pathways, and phenotypic characteristics. The development of high-throughput screening platforms using CRISPR technologies allows for the simultaneous assessment of multiple genetic modifications. This approach facilitates the rapid identification of optimal strains for various applications, including bioproduction and environmental remediation. Harnessing machine learning algorithms for the prediction and optimization of CRISPR target sites, guide RNA designs, and Cas protein variants is an emerging area of research. 17 Computational tools are expected to play a significant role in streamlining the design and optimization of CRISPR experiments. Future of CRISPR-based genome editing for microorganisms holds exciting prospects, with ongoing research focused on enhancing precision, expanding capabilities, and exploring novel applications. The integration of emerging technologies, interdisciplinary approaches, and a comprehensive understanding of microbial systems will contribute to breakthroughs that reshape our ability to engineer microorganisms for diverse purposes.

Conclusion

In the rapidly evolving landscape of CRISPR-based genome editing for microorganisms, the journey from its inception to the current state reflects a remarkable trajectory of innovation, discovery, and transformative potential. The review paper has delved into the key advancements, challenges, and ethical considerations surrounding CRISPR technologies, highlighting their diverse applications across fields such as bioproduction, medicine, environmental remediation, and beyond. As we stand at the cusp of the next phase in CRISPR research, the future promises to unfold unprecedented breakthroughs that will redefine our understanding and utilization of these powerful tools. The pursuit of precision genome editing, expanded targeting capabilities, and integration with emerging technologies like RNA editing and epigenome editing holds the key to unlocking new dimensions of genetic control and manipulation. The journey ahead is marked by a commitment to responsible and ethical use. As CRISPR technologies become more intricate, our ethical compass must remain steadfast, navigating the complex terrains of unintended consequences, environmental release, and societal implications. Continuous engagement with stakeholders, transparent communication, and adaptive regulatory frameworks will be pivotal in ensuring the ethical governance of CRISPR-based genome editing. In this era of interdisciplinary collaboration, the convergence of CRISPR with synthetic biology, machine learning, and high-throughput screening platforms heralds a new era of possibilities. The integration of CRISPR into diagnostics, biosensors, and in vivo applications showcases its versatility and potential to revolutionize diverse fields, from healthcare to environmental monitoring.23 As researchers embark on endeavors to engineer the microbiome, optimize microbial communities, and venture into multi-omics integration, the boundaries of CRISPR applications continue to expand. These pursuits not only advance our scientific understanding but also raise the bar for responsible innovation, ensuring that the benefits of CRISPR technologies are realized while minimizing potential risks. In conclusion, the review paper encapsulates the current state of CRISPR-based genome editing for microorganisms, offering a comprehensive exploration of its advancements, challenges, ethical considerations, and future directions. The journey from the foundational CRISPR-Cas9 system to the frontiers of precision editing, environmental applications, and beyond is a testament to the dynamic nature of scientific inquiry. As the narrative unfolds, researchers, policymakers, and society at large must collaboratively navigate the ethical and scientific complexities to harness the full potential of CRISPR technologies for the betterment of humanity and the environment. The next chapter in the CRISPR saga holds the promise of unlocking unprecedented possibilities, and the journey continues with anticipation and responsibility.

Source of Funding

None.

Conflict of Interest

None.

References

1 

M Jinek K Chylinski I Fonfara M Hauer JA Doudna E Charpentier A programmable dual-RNA-guided DNA endonuclease in adaptive bacterial immunityScience2012337609681621

2 

JA Doudna E Charpentier Genome Editing: The New Frontier of Genome Engineering with CRISPR-Cas9Science20143466213125809610.1126/science.1258096

3 

B Zetsche JS Gootenberg OO Abudayyeh IM Slaymaker KS Makarova P Essletzbichler Cpf1 is a single RNA-guided endonuclease of a class 2 CRISPR-Cas systemCell2015163375971

4 

PD Hsu DA Scott JA Weinstein FA Ran S Konermann V Agarwala DNA targeting specificity of RNA-guided Cas9 nucleasesNat Biotechnol201331982732

5 

P Mali L Yang KM Esvelt J Aach M Guell JE Dicarlo RNA-guided human genome engineering via Cas9Science201333961218236

6 

S Shmakov A Smargon D Scott D Cox N Pyzocha W Yan Diversity and evolution of class 2 CRISPR-Cas systemsNat Rev Microbiol201715316982

7 

SW Cho S Kim JM Kim JS Kim Targeted genome engineering in human cells with the Cas9 RNA-guided endonucleaseNat Biotechnol20133132302

8 

R Barrangou C Fremaux H Deveau M Richards P Boyaval S Moineau CRISPR provides acquired resistance against viruses in prokaryotesScience20073155819170912

9 

M Jinek A East A Cheng S Lin E Ma J Doudna RNA-programmed genome editing in human cellsElife20132e0047110.7554/eLife.00471

10 

F Zhang Y Wen X Guo CRISPR/Cas9 for genome editing: progress, implications and challengesHum Mol Genet201423R1406

11 

L Cong FA Ran D Cox S Lin R Barretto N Habib Multiplex genome engineering using CRISPR/Cas systemsScience2013339612181923

12 

WY Hwang Y Fu D Reyon ML Maeder SQ Tsai JD Sander Efficient genome editing in zebrafish using a CRISPR-Cas systemNat Biotechnology20133132279

13 

P Mali J Aach PB Stranges KM Esvelt M Moosburner S Kosuri CAS9 transcriptional activators for target specificity screening and paired nickases for cooperative genome engineeringNat Biotechnol20133198338

14 

H Nishimasu FA Ran PD Hsu S Konermann SI Shehata N Dohmae Crystal structure of Cas9 in complex with guide RNA and target DNACell2014156593549

15 

SW Cho S Kim Y Kim J Kweon HS Kim S Bae Analysis of off-target effects of CRISPR/Cas-derived RNA-guided endonucleases and nickasesGenome Res201424113241

16 

W Jiang D Bikard D Cox F Zhang LA Marraffini RNA-guided editing of bacterial genomes using CRISPR-Cas systemsNat Biotechnol20133132339

17 

H Kim JS Kim A guide to genome engineering with programmable nucleasesNat Rev Genet201415532124

18 

S Shmakov OO Abudayyeh KS Makarova YI Wolf JS Gootenberg E Semenova Discovery and functional characterization of diverse class 2 CRISPR-Cas systemsMolecular Cell201560338597

19 

BA Davidson AX Miranda SC Reed R E Bergman JDJ Kemp AP Reddy An in vitro CRISPR screen of cell-free DNA identifies apoptosis as the primary mediator of cell-free DNA releaseCommun Biol20247144110.1038/s42003-024-06129-1

20 

Y Du Y Liu J Hu X Peng Z Liu CRISPR/Cas9 systems: Delivery technologies and biomedical applicationsAsian J Pharm Sci202318610085410.1016/j.ajps.2023.100854

21 

JG Doench N Fusi M Sullender M Hegde EW Vaimberg KF Donovan Optimized sgRNA design to maximize activity and minimize off-target effects of CRISPR-Cas9Nat Biotechnol201634218491

22 

EV Koonin KS Makarova F Zhang Diversity, classification and evolution of CRISPR-Cas systemsCurr Opin Microbiol201737677810.1016/j.mib.2017.05.008

23 

T Li Y Yang H Qi W Cui L Zhang X Fu CRISPR/Cas9 therapeutics: progress and prospectsSig Transduct Target Ther202383610.1038/s41392-023-01309-7



jats-html.xsl


This is an Open Access (OA) journal, and articles are distributed under the terms of the Creative Commons Attribution-NonCommercial-ShareAlike 4.0 License, which allows others to remix, tweak, and build upon the work non-commercially, as long as appropriate credit is given and the new creations are licensed under the identical terms.

  • Article highlights
  • Article tables
  • Article images

Article History

Received : 24-01-2024

Accepted : 23-03-2024


View Article

PDF File   Full Text Article


Copyright permission

Get article permission for commercial use

Downlaod

PDF File   XML File   ePub File


Digital Object Identifier (DOI)

Article DOI

https://doi.org/10.18231/j.ijmmtd.2024.003


Article Metrics






Article Access statistics

Viewed: 470

PDF Downloaded: 120



Medical Abbreviation List