Fact-checked by Grok 2 weeks ago

Transfection

Transfection is the deliberate introduction of foreign nucleic acids, such as DNA or RNA, into eukaryotic cells to modify their genetic content, enable gene expression, or study cellular functions. This technique refers to non-viral methods, distinct from viral transduction which uses infectious viral vectors like adenoviruses or lentiviruses to deliver genetic material. Developed as a cornerstone of molecular biology since the mid-20th century, transfection allows researchers to transiently or stably alter cell behavior, facilitating applications from basic gene function analysis to therapeutic interventions. Transfection methods are broadly classified into physical and chemical categories, each with varying efficiency, cytotoxicity, and suitability for different cell types. Physical approaches, such as —which uses electric pulses to create temporary pores in cell membranes—and , offer precise delivery but can damage cells due to mechanical stress. Chemical methods, including lipofection with cationic lipids like and calcium phosphate precipitation, form complexes with nucleic acids to facilitate uptake via , providing a balance of ease and moderate efficiency for adherent or suspension cells. In research and , transfection underpins key advancements, including the of recombinant proteins in lines like HEK293 and for biopharmaceuticals, RNA for with siRNA, and delivery of CRISPR-Cas9 components for . Transient transfection yields short-term expression ideal for rapid functional assays, while stable transfection integrates DNA into the host for long-term studies or cell line engineering, often selected via antibiotic resistance markers. Despite challenges like low efficiency in primary s and potential off-target effects, ongoing innovations in carriers and devices continue to enhance transfection's precision and applicability in for diseases such as cancer and genetic disorders.

Terminology and Fundamentals

Definition and Principles

Transfection is the process of introducing exogenous nucleic acids, such as DNA or RNA, into eukaryotic cells using non-viral methods to enable gene expression, functional studies, or genetic modification. This technique differs from viral transduction, which relies on viral vectors for delivery and often integrates genetic material into the host genome, potentially eliciting immune responses. In contrast to bacterial transformation, where prokaryotic cells uptake naked DNA for heritable changes, transfection targets eukaryotic cells and typically does not result in permanent genomic integration unless specifically designed for stable outcomes. The primary goal is to overcome cellular barriers to allow the nucleic acids to reach their functional sites, facilitating applications in research and therapy. The underlying principles of transfection revolve around cellular uptake, intracellular trafficking, and processing. Eukaryotic cells present formidable barriers, including the plasma membrane's , which repels hydrophilic s. For chemical methods, delivery aids form complexes with s to promote entry primarily via . Once internalized via , the s are often trapped in endosomes, where endosomal escape is crucial to prevent degradation by lysosomal enzymes and enable progression to the or . For DNA, successful expression requires nuclear entry, which can occur through complexes or during when the disassembles, influenced by DNA size and nuclear localization signals. , such as mRNA, bypasses nuclear entry and is directly translated by ribosomes in the , allowing faster onset of protein production. Efficiency of transfection is modulated by several factors, including —where adherent cell lines like HEK293 exhibit higher rates than primary or non-dividing cells—and characteristics, such as size, with larger constructs (>10 kb) reducing uptake and expression due to steric hindrance. Key metrics include transfection rate, defined as the percentage of cells successfully incorporating and expressing the , often quantified via , and expression level, measured using reporter genes like (GFP) to visualize fluorescent cells under or assess intensity proportional to protein output. For instance, uses electric pulses to transiently permeabilize the , allowing direct uptake into the and bypassing endocytic pathways. Outcomes can be transient, with episomal expression fading over divisions, or stable through selection for integration.

Historical Development

The concept of introducing exogenous DNA into mammalian cells emerged in the early , with Elisabeth Szybalska and Wacław Szybalski demonstrating the first DNA-mediated heritable of a biochemical trait in human cell lines deficient in (HGPRT), using DNA extracted from wild-type cells to restore enzymatic function. This pioneering work laid the groundwork for non-viral gene transfer, though efficiencies were low and limited to specific selectable markers. Concurrently, bacterial systems advanced with Mandel and Higa's 1970 discovery of treatment to enhance competence for DNA uptake in , enabling efficient that became a cornerstone for experiments. The 1970s marked a pivotal shift with the advent of recombinant DNA technology, pioneered by Stanley Cohen and Herbert Boyer's 1973 construction of biologically functional bacterial plasmids in vitro, which relied on transformation techniques to propagate hybrid DNA molecules and fueled the biotechnology revolution. In mammalian cells, Frank Graham and Arie van der Eb introduced the calcium phosphate precipitation method in 1973, allowing assay of adenovirus DNA infectivity by facilitating DNA uptake into human and monkey cell lines with improved efficiency over prior approaches, though still yielding only 1-2% transfection rates. Diethylaminoethyl (DEAE)-dextran, initially described by McCutchan and Pagano in 1968 for enhancing simian virus 40 DNA infectivity, gained widespread adoption in the 1980s for transient transfections due to its simplicity in promoting endocytosis of DNA complexes. The 1980s and 1990s saw diversification of physical methods, with Eberhard Neumann and colleagues reporting in 1982 the use of high-voltage electric pulses () to permeabilize mouse lymphoma cell membranes for DNA uptake, achieving up to 10-fold higher transformation than chemical methods and spurring refinements in pulse parameters for broader cell types by the 1990s. Chemical innovations included Philip Felgner's 1987 development of lipofection using synthetic cationic like DOTMA to form liposome-DNA complexes, enabling high-efficiency (up to 50%) transfection in diverse mammalian lines with reduced toxicity compared to precipitates. The 2000s expanded to RNA delivery following and Craig Mello's 1998 demonstration of (RNAi) via double-stranded RNA in C. elegans, which earned the 2006 and prompted protocols for synthetic siRNA transfection using or to silence genes transiently. Transfection evolved from low-efficiency, labor-intensive techniques to high-throughput methods compatible with and large-scale production, driven by recombinant DNA's demand for rapid screening. Post-2010 advances integrated CRISPR-, with and Emmanuelle Charpentier's 2012 programmable endonuclease enabling precise genome editing via or lipofection of guide RNAs and Cas9 plasmids in human cells. Regulatory milestones included the FDA's 2017 approvals of Luxturna (AAV-based retinal produced via transfection) and Kymriah (CAR-T cells transduced ), validating transfection in therapeutic manufacturing and accelerating clinical translation.

Transfection Methods

Physical Methods

Physical methods of transfection rely on mechanical or electrical forces to permeabilize cell membranes and facilitate the entry of nucleic acids, bypassing the need for chemical carriers or viral vectors. These approaches are particularly valuable for transfecting difficult cell types, such as primary s or tissues with thick extracellular matrices, where other methods may fail. Key techniques include , , biolistics, and emerging laser-based variants like optoporation. Electroporation involves applying short, high-voltage electric pulses to cells in suspension, creating transient pores in the plasma membrane through which DNA or RNA can enter. The process was first demonstrated in 1982 by Neumann and colleagues, who showed that electric fields of 8 kV/cm for 5 microseconds dramatically enhanced DNA uptake in mouse lyoma cells. Typical parameters include voltages of 100-1000 V across a cuvette gap of 0.1-0.4 cm, resulting in field strengths of 250-2000 V/cm, and pulse durations of 1-10 ms, often using exponential decay waveforms where the time constant τ is determined by τ = RC (R being the resistance of the medium and C the capacitance of the electroporator). For optimal efficiency, cells are prepared in a low-conductivity buffer to minimize heating, harvested at 70-90% confluency, and resuspended at 10^5-10^7 cells/mL; post-transfection, cells are immediately diluted in recovery medium and incubated for 24-48 hours to allow membrane resealing and expression. This method achieves transfection efficiencies of 50-90% in hard-to-transfect cells like primary neurons, but high voltages can reduce viability to below 50% due to irreversible electroporation and Joule heating. Unlike chemical methods, electroporation leaves no residues, making it suitable for downstream applications sensitive to contaminants. Microinjection delivers nucleic acids directly into the cell cytoplasm or nucleus using a fine glass micropipette under microscopic guidance, ensuring precise targeting of individual cells. Pioneered for genetic transformation in the late 1970s, a seminal application was reported in 1980 by Gordon et al., who microinjected purified DNA into mouse embryos to achieve stable integration. The technique requires immobilizing cells on a chamber, inserting a needle with an inner diameter of 0.5-1 μm filled with 1-10 μg/μL DNA, and injecting 1-10 pL per cell at pressures of 10-100 hPa to avoid bursting. Post-injection, cells are returned to culture medium for recovery, often with serum supplementation to mitigate stress. While nearly 100% efficient for single-cell transfection, it is labor-intensive, limited to low throughput (hundreds of cells per hour), and carries risks of mechanical damage leading to 20-50% cell death, though it excels for rare or precious samples like oocytes. Biolistics, or particle bombardment, accelerates DNA-coated microprojectiles into cells using a gene gun, penetrating cell walls and membranes via kinetic energy. Developed in the mid-1980s by Sanford and colleagues at Cornell University, the method was first detailed in a 1987 Nature paper by Klein et al., demonstrating delivery of nucleic acids into living plant and animal cells with particles fired at velocities of 300-600 m/s. Gold or tungsten particles (0.5-3 μm diameter) are coated with 1-5 μg DNA/mg, loaded into a cartridge, and propelled by helium pressure (200-600 psi); target cells are plated on Petri dishes, and post-bombardment, excess particles are washed away before incubation in antibiotic-free medium for 24-72 hours. Efficiencies range from 10-50% in adherent cells, with advantages in transfecting thick tissues like skin or plant leaves without dissociation, but disadvantages include variable penetration depth causing uneven delivery and potential tissue trauma from high-impact particles. Recent advancements include laser-based optoporation, which uses focused laser pulses (typically 800 nm, 100 fs duration) to induce localized membrane poration without electrodes or needles, achieving single-cell precision. Reviews from the highlight efficiencies of 50-80% with viabilities above 70% in mammalian cells, as shown in studies like Stevenson et al. (2013) using titanium-sapphire lasers for delivery. These physical methods collectively offer high specificity and broad applicability but require optimization to balance efficiency and .

Chemical Methods

Chemical methods of transfection utilize synthetic or biochemical agents to facilitate the entry of nucleic acids into through the formation of protective complexes that interact with cellular membranes. These approaches rely on electrostatic interactions between positively charged carriers and negatively charged nucleic acids, promoting and subsequent intracellular release. Unlike physical methods, which may induce higher cell stress, chemical strategies emphasize and ease of use in laboratory settings. One of the most established chemical techniques is lipofection, introduced by Felgner et al. in 1987, which employs cationic lipids such as DOTAP (1,2-dioleoyl-3-trimethylammonium-propane) to form liposomes or lipid nanoparticles that encapsulate DNA or RNA. These lipids self-assemble into vesicles via hydrophobic interactions, with the positively charged head groups condensing nucleic acids at an optimal nitrogen-to-phosphate (N/P) ratio of 2-6, where N represents moles of protonatable nitrogens in the lipid and P denotes moles of phosphates in the nucleic acid. The complexes fuse with or are endocytosed by the cell membrane, releasing the cargo in the cytoplasm, though endosomal entrapment remains a challenge. Complex assembly typically involves mixing lipids and nucleic acids in serum-free media for 15-30 minutes at room temperature, followed by adding the mixture to cells for 2-4 hours before replacing with complete media to enhance efficiency. Calcium phosphate precipitation, pioneered by Graham and van der Eb in 1973, represents another foundational chemical method, where DNA is co-precipitated with calcium ions in a buffered phosphate solution at pH 6.8-7.4 to form microcrystals that are taken up by cells via endocytosis or phagocytosis. The protocol entails dissolving DNA in a calcium chloride solution, adding it dropwise to a HEPES-buffered phosphate buffer to generate the precipitate, which is then applied to cells for 4-16 hours in a controlled environment to avoid excessive precipitation that could damage cells. This technique is particularly suited for adherent cells and large-scale transfections due to its simplicity and low cost. Polyfection using polymers like branched polyethylenimine (PEI, typically 25 kDa) offers high efficiency through the formation of polyplexes that protect nucleic acids and promote endosomal escape via the proton sponge effect, where PEI's multiple amines buffer endosomal acidification, causing osmotic swelling and rupture to release the cargo. First demonstrated by Boussif et al. in 1995, PEI complexes are prepared by mixing polymer and DNA at an N/P ratio of 5-10, incubating for 15-30 minutes, and exposing cells for 2-4 hours in serum-free conditions to optimize uptake. To mitigate PEI's cytotoxicity from high charge density, biodegradable variants or lower doses are employed, balancing efficiency with cell viability. Overall, chemical methods are cost-effective and scalable for high-throughput applications, achieving transfection efficiencies of 10-90% depending on and optimization, though variability arises from endosomal barriers and potential . Advances in nanoparticles post-2020, as seen in formulations, have improved stability and , reducing compared to viral alternatives; recent developments as of 2024 include ionizable nanoparticles achieving >90% efficiency in diverse types for therapeutic mRNA .

Viral Methods

Viral methods of transfection utilize genetically modified viruses as vectors to deliver nucleic acids into cells, exploiting the natural mechanisms of viruses for efficient gene transfer. These vectors are engineered to carry therapeutic or experimental DNA or RNA while minimizing pathogenicity, allowing for targeted expression in various cell types. Unlike non-viral approaches, viral vectors can achieve high rates by leveraging viral replication machinery and , though they require careful handling. Common viral vectors include adenoviral, lentiviral, and (AAV) systems, each with distinct properties suited to different applications. Adenoviral vectors are non-integrating, remaining episomal in , which enables without risking permanent genetic alteration. They can achieve high titers up to 10^12 plaque-forming units per milliliter (PFU/mL) and efficiently infect both dividing and non-dividing s due to their broad . Lentiviral vectors, derived from HIV-1, integrate the into via reverse transcription of their payload, providing stable, long-term expression ideal for applications requiring persistent gene activity. These vectors are often pseudotyped with the vesicular stomatitis virus glycoprotein (VSV-G) envelope to confer broad , allowing infection of a wide range of cell types, including non-dividing cells like neurons. AAV vectors, which are parvoviruses requiring helper viruses for replication, persist primarily as episomes with low integration rates, supporting long-term expression—up to five years in some tissues like muscle—without eliciting strong immune responses. AAV s 1 through 9 vary in tissue , with 2 commonly used for its efficiency in dividing cells and 9 for hepatic targeting. Production of viral vectors typically involves transient transfection of packaging cells, such as HEK293 cells, which express necessary viral proteins to assemble infectious particles from the vector genome. The multiplicity of infection (), defined as the ratio of viral particles to target cells (ranging from 1 to 100), is optimized to maximize yield while minimizing during propagation. Post-production, vectors are purified using cesium chloride (CsCl) density gradient to remove contaminants and achieve high purity, ensuring safety for downstream applications. Viral methods offer advantages such as near-100% efficiency in permissive cells and inherent cell-type specificity through natural , making them superior for delivery compared to non-replicating non-viral systems. However, they carry risks including from genomic integration (particularly with lentiviruses), which can disrupt host genes and potentially lead to oncogenesis, and immune responses triggered by viral proteins that may limit repeat dosing. Most viral vectors, excluding those with replication-competent elements, are handled under 2 (BSL-2) conditions to mitigate risks. The foundational use of viral transduction dates to the 1970s, when retroviruses were first demonstrated to transfer cellular genes, laying the groundwork for engineered vectors. Advances in delivery have extended to CRISPR-Cas9 systems, with early clinical explorations in the mid-2010s enabling precise ; for instance, CRISPR-Cas9 editing entered clinical trials by 2018 for applications like treatment. Immunogenicity in adenoviral vectors is mitigated through "gutless" or helper-dependent designs, which eliminate most genes to reduce capsid-associated immune activation while maintaining high-capacity delivery.

Types of Transfection Outcomes

Transient Transfection

Transient transfection refers to the temporary introduction of nucleic acids, such as DNA or mRNA, into eukaryotic cells, resulting in short-term without integration into the host . This process enables the expression of transgenes from extrachromosomal elements, typically lasting from 1 to 7 days, and is widely used for rapid functional studies in . Unlike stable transfection, which involves genomic incorporation for long-term , transient transfection relies on non-integrative delivery methods that prioritize speed and ease over persistence. The ephemerality of transient transfection arises primarily from extrachromosomal expression, where delivered plasmids remain as episomes in the nucleus and are diluted during cell division, leading to a half-life of approximately 24-48 hours in dividing cells. Additionally, nucleases in the cellular environment degrade the introduced nucleic acids over time, further limiting expression duration; for instance, plasmid DNA is progressively lost without replication signals, while mRNA is subject to rapid turnover by exonucleases. Detection of transient expression often employs reporter genes like luciferase, where activity peaks between 24 and 72 hours post-transfection, allowing quantification via luminescence assays that correlate with transgene levels. Protocols for transient transfection typically involve high doses of nucleic acids, ranging from 1 to 10 μg per 10^6 cells, to achieve sufficient expression levels despite the short window, with optimal timing for analysis set around 24-48 hours to capture peak activity. These methods are particularly suited for applications, such as in 96-well plate formats, where lipofection or delivers constructs en masse for functional assays like protein studies. Efficiency can be enhanced by incorporating elements like the enhancer, which promotes episomal replication in certain cell types, thereby extending expression slightly without stable integration. A key advantage of transient transfection is its rapid readout, facilitating quick functional validation in research settings, though it suffers from low persistence, with expression often undetectable after one week due to dilution and . In therapeutic contexts, transient mRNA transfection has shown superiority over DNA-based approaches in , particularly in post-2018 CAR-T cell engineering, where of mRNA yields transient receptor expression that minimizes tonic signaling and enhances safety profiles in clinical trials.

Stable Transfection

Stable transfection refers to the process by which exogenous nucleic acids, typically DNA, are integrated into the host cell's genome, enabling long-term, heritable expression of the transgene across subsequent cell divisions. This contrasts with transient transfection by achieving permanent genomic incorporation, primarily through two mechanisms: homologous recombination, which allows precise insertion at targeted loci using homology arms, or random insertion, often facilitated by non-homologous end joining (NHEJ) or transposon systems. Transposon-based methods, such as the Sleeping Beauty system—a synthetic transposon reconstructed from inactive fish elements—promote efficient cut-and-paste integration, typically resulting in 1-10 copies per cell, which influences expression levels and potential genotoxicity. The first demonstrations of stable transfection in mammalian cells occurred in the early 1970s using calcium phosphate-mediated DNA uptake, leading to the establishment of transformed lines like HEK293. More recently, CRISPR-Cas9 technologies from the Zhang laboratory have advanced precise stable knock-ins via homology-directed repair (HDR), as detailed in protocols for genome engineering published in 2013. Protocols for stable transfection begin with the delivery of linear or circular plasmids containing the and a , such as the neomycin phosphotransferase gene conferring resistance to (Geneticin). Following transfection, cells are cultured in selective media containing 200-800 μg/mL for 2-4 weeks to eliminate non-integrated cells, allowing only those with stable integrations to proliferate. Clonal isolation is then achieved through limiting dilution or fluorescence-activated (FACS) to obtain monoclonal populations, followed by verification of integration via analysis, which confirms copy number and site specificity. Recent CRISPR-mediated approaches enhance specificity by co-delivering , , and donor templates, achieving knock-in efficiencies up to several percent in optimized systems, though overall stable transfection efficiency remains below 1% without enrichment strategies like FACS. The primary advantage of stable transfection is the generation of heritable cell lines for sustained and functional studies, essential for applications like recombinant therapeutics. However, it carries risks including position effect variegation, where transgene silencing occurs due to spreading at random integration sites, and potential oncogenesis from disrupting proto-oncogenes or tumor suppressors. These drawbacks necessitate careful site selection and validation to mitigate variability in expression stability.

RNA-Specific Transfection

Endogenous vs. Exogenous RNA Delivery

Endogenous RNA molecules are produced within cells through transcription by , followed by co- and post-transcriptional processing that includes the addition of a 5' cap structure (7-methylguanosine) to protect against exonucleases and facilitate initiation, as well as the attachment of a 3' poly(A) tail by poly(A) polymerase to enhance stability and export from the . These modifications contribute to mRNA half-lives ranging from hours to days, with stability further regulated by microRNAs (miRNAs) that bind to the 3' (UTR) to inhibit or promote decay via the . In contrast, exogenous RNA for transfection is typically synthesized via in vitro transcription (IVT) using bacteriophage polymerases like T7, which lacks the cellular processing machinery, resulting in uncapped, non-polyadenylated transcripts prone to rapid degradation by ubiquitous RNases in the extracellular and intracellular environments. To mitigate innate immune activation and enhance stability, synthetic RNAs incorporate modified nucleosides such as , which reduces recognition by Toll-like receptor 3 (TLR3) and other sensors by altering RNA secondary structure and evading endolysosomal processing. RNA transfection was pioneered in 1989 by et al., who demonstrated efficient delivery using cationic liposomes, though exogenous RNAs often trigger immune responses via the RIG-I pathway, leading to production upon detection of 5' triphosphate ends. Design strategies for exogenous RNA address these challenges by mimicking endogenous features: anti-reverse cap analogs (ARCA), which incorporate a modified 7-methylguanosine during IVT to ensure correct orientation and improve translation efficiency by approximately twofold (up to 100%), are commonly used alongside enzymatic poly(A) tail addition. Optimization of the 3' UTR sequences, such as incorporating stabilizing elements from highly expressed cellular mRNAs, further boosts by enhancing ribosome recruitment and reducing decay rates. Delivery methods like can achieve transfection efficiencies up to 90% in certain cell types by transiently permeabilizing membranes, though nucleoside modifications like —advanced post-2015 and critical for mRNA vaccines—remain essential to suppress immunogenicity and enable high-level protein expression.

Repeated Long-RNA Transfection Protocols

Repeated long-RNA transfection protocols involve iterative delivery of synthetic (mRNA) molecules, typically longer than 1,000 nucleotides, to achieve sustained protein expression over extended periods without genomic integration. These methods address the limitations of single-dose transient transfection, where expression typically declines within 24-72 hours due to mRNA degradation and dilution during . By administering multiple doses, protocols enable cumulative protein accumulation that can mimic stable transfection outcomes, lasting weeks to months depending on the frequency and RNA stability enhancements. Common protocols employ daily or every-48-hour dosing schedules using lipid-based carriers like MessengerMAX, with typical amounts ranging from 1-5 μg per 10^6 cells to balance efficiency and . For instance, in stem cell reprogramming applications refined during the 2010s, fibroblasts are transfected daily for 12-18 days with a cocktail of modified mRNAs encoding reprogramming factors such as Oct4, , , and c-Myc, at doses of approximately 100-400 ng per well in a 96-well plate format. This iterative approach yields high-efficiency generation, with pluripotency markers appearing after 10-14 doses. Weekly dosing variants have been explored for less frequent interventions in primary cell cultures, maintaining expression levels through optimized modifications like substitution to enhance stability. Key challenges in repeated long-RNA transfection include immune activation and cellular from cumulative carrier exposure, which can lead to reduced transfection efficiency or "immune fatigue" characterized by attenuated responses to subsequent doses due to pathway upregulation. strategies involve low-dose escalation—starting at 0.5-1 μg per 10^6 s and increasing gradually—to minimize innate immune sensing via Toll-like receptors, alongside chemical modifications such as 5-methylcytidine to suppress production. Off-target effects, such as unintended silencing of endogenous genes from repeated uptake, are monitored through quantitative (qPCR) assays that track transfected levels relative to genes like GAPDH, revealing peak accumulation after 3-5 doses before plateauing. In neural progenitor s, for example, repetitive daily transfections reduced when spaced 24 hours apart after sufficient , allowing sustained expression without significant cell loss. These protocols offer advantages such as avoidance of insertional mutagenesis risks associated with viral methods, making them ideal for therapeutic protein replacement in regenerative medicine, where repeated mRNA delivery (e.g., every 10 days for multiple cycles) sustains deficient enzymes like factor IX in hemophilia B models, with effects observed over intervals up to 3 months. However, disadvantages include the logistical burden of multiple interventions, potential for variable bioavailability in vivo, and higher costs compared to single-dose alternatives. Recent preclinical studies have explored repeated mRNA delivery for cardiac regeneration, demonstrating improved tissue function in animal models without genomic integration concerns.

Applications and Advances

Research Applications

Transfection serves as a fundamental tool in gene function studies, enabling researchers to investigate protein roles through overexpression or knockdown techniques. For instance, transient overexpression of via plasmid transfection allows assessment of gain-of-function effects, while (siRNA) delivery facilitates targeted to elucidate loss-of-function phenotypes. These methods have been pivotal in dissecting cellular processes, such as the use of siRNA transfection to knockdown overexpressed genes in tumor progression models. In pathway mapping, transfection is employed to activate or inhibit specific signaling cascades, providing insights into regulatory networks. A representative example is the transient transfection of pathway components or reporters, which enables real-time monitoring of activation dynamics in response to stimuli, revealing interactions like those suppressed by Clara cell 10-kDa protein gene transfer. further leverages transfection for large-scale ; CRISPR libraries delivered via transfection into up to 10^6 cells per screen identify genetic modifiers of phenotypes, as demonstrated in imaging-based pooled assays that quantify multiplexed gene edits. Common model systems include adherent cell lines like HEK293, which achieve transfection efficiencies of approximately 70% with lipid-based reagents, making them ideal for rapid prototyping of gene effects. For more physiologically relevant contexts, 3D s are transfected using microfluidic encapsulation in microbeads, allowing clonal expansion and transgene expression with efficiencies exceeding 50% in organoid models. In animal models, transfects embryos, such as mouse neural progenitors, to study developmental gene functions with up to 80% targeting precision. Recent innovations highlight transfection's evolution in research precision. , advanced in the 2010s, relies on viral or non-viral transfection to express light-sensitive opsins in neurons, enabling millisecond control of neural circuits as reviewed in foundational studies. Single-cell transfection via nanopipette , developed around 2020, delivers precise cargos like components to individual cells with >90% viability, facilitating high-resolution functional assays. The 2006 for underscored transfection's role in siRNA delivery, transforming knockdown studies from model organisms to mammalian systems. Throughput has advanced with automated 384-well platforms, enabling parallel transfections of thousands of conditions for screens.

Therapeutic Applications

Transfection plays a pivotal role in therapeutic applications, particularly in , where nucleic acids are delivered to correct genetic defects or modulate cellular functions in patients. The first approved utilizing transfection was Glybera (), an (AAV)-based treatment for , authorized by the in 2012 but withdrawn in 2017 due to commercial reasons. This marked a milestone in clinical translation, demonstrating the feasibility of viral vector-mediated gene delivery for rare metabolic disorders. Subsequent approvals, such as Zolgensma () in 2019 by the U.S. Food and Drug Administration for (SMA) in children under two years, further advanced AAV-based therapies by delivering functional gene copies to motor neurons, achieving sustained motor function improvements in clinical trials. In 2023, the FDA approved Casgevy (exagamglogene autotemcel), the first CRISPR-based therapy for and transfusion-dependent beta-thalassemia, using —a non-viral physical method—to deliver CRISPR-Cas9 components to patient-derived hematopoietic stem cells , enabling high-efficiency editing with reduced risks. In vaccine development and , transfection enables rapid and targeted delivery. The Pfizer-BioNTech (BNT162b2), authorized in late 2020, relies on lipid nanoparticles to transfect mRNA encoding the into host cells, eliciting robust immune responses with 95% efficacy against symptomatic infection in phase 3 trials. Similarly, chimeric antigen receptor (CAR) T-cell engineering often incorporates transient transfection for enhanced safety, avoiding permanent genomic integration risks associated with viral methods; for instance, mRNA nanocarriers have been used to transiently express CARs in circulating T cells, reducing off-target effects and in preclinical models of solid tumors. Despite these successes, therapeutic transfection faces significant challenges, including efficient delivery and safety concerns. Hydrodynamic injection, a non-viral method for liver-targeted delivery, achieves 10-40% hepatocyte transfection efficiency in models but is limited by procedural invasiveness and lower in humans. Off-target effects, such as unintended genomic alterations from CRISPR-Cas9 editing, and the need for good manufacturing practice (GMP) production to ensure and purity remain hurdles, as evidenced by regulatory requirements for AAV therapies. Recent advances highlight non-viral nanoparticles for delivery in clinical trials, with lipid-based systems entering phase 1/2 studies in the 2020s for conditions like hereditary transthyretin-mediated , offering improved safety over vectors by minimizing . Post-2020 mRNA therapeutics have expanded to cancer vaccines, where personalized neoantigen-encoding mRNAs combined with checkpoint inhibitors have shown promising tumor regression in phase 1 trials for and . Ethical considerations distinguish therapies, which affect only the patient and are widely accepted, from germline editing, which risks heritable changes and is prohibited in clinical settings due to consent and equity issues for future generations.

References

  1. [1]
    Transfection types, methods and strategies: a technical review - PMC
    Transfection is a modern and powerful method used to insert foreign nucleic acids into eukaryotic cells. The ability to modify host cells' genetic content ...
  2. [2]
    Transfection - an overview | ScienceDirect Topics
    Transfection is defined as the process of introducing foreign DNA into cells using either physical methods, such as electroporation, or chemical methods, ...
  3. [3]
  4. [4]
    Genetics - Medical Microbiology - NCBI Bookshelf - NIH
    Genetic exchanges among bacteria occur by several mechanisms. In transformation, the recipient bacterium takes up extracellular donor DNA. In transduction, ...
  5. [5]
  6. [6]
    Synthetic Approaches for Nucleic Acid Delivery: Choosing the Right ...
    Jul 9, 2019 · The complexes with different structures, formulations, and physical properties display different mechanisms of transfection through different ...
  7. [7]
    Progress and prospects: nuclear import of nonviral vectors - PMC
    The nuclear envelope represents a key barrier to successful nonviral transfection and gene therapy both in vitro and in vivo.
  8. [8]
    Translation of in vitro-transcribed RNA therapeutics - PMC
    Feb 8, 2023 · IVTmRNAs need to escape from the endosome into the cytoplasm to be translated, and this is thought to be one of the most limiting steps of ...
  9. [9]
    Green fluorescent protein is a quantitative reporter of gene ... - NIH
    We report that GFP is a reliable reporter of gene expression in individual eukaryotic cells when fluorescence is measured by flow cytometry.
  10. [10]
    a highly efficient, lipid-mediated DNA-transfection procedure. - PNAS
    The technique is simple, highly reproducible, and effective for both transient and stable expression of transfected DNA.
  11. [11]
    Approved Cellular and Gene Therapy Products - FDA
    Aug 15, 2025 · Approved Cellular and Gene Therapy Products. Below is a list of licensed products from the Office of Therapeutic Products (OTP).Abecma · Adstiladrin · Zolgensma · AmtagviMissing: transfection 2010s
  12. [12]
    Avanti Transfection Reagent I | 1:1 DOTAP, DOPE Cationic Lipid
    DOTAP is one of the most widely used cationic lipids for gene transfection applications. DOTAP is proven to be efficient for in vitro and in vivo transfection ...
  13. [13]
  14. [14]
    [PDF] Chemically-assisted DNA transfection methods – An overview
    Nov 10, 2023 · To obtain transfectants, host cells are usually treated with biocompatible DNA carriers such as calcium phosphate, cationic lipids, DEAE-dextran ...
  15. [15]
    Appraisal for the Potential of Viral and Nonviral Vectors in Gene ...
    Jul 30, 2022 · Viruses provide good transfection efficiency and sustainable gene expression, and they protect the gene from degradation; however, they are ...
  16. [16]
    Adenovirus Vector - an overview | ScienceDirect Topics
    They have been developed as gene delivery vehicles due to the ability to infect non-dividing cells. Adenoviral vectors do not integrate into the genome of ...
  17. [17]
    Adenovirus-mediated gene delivery: Potential applications for gene ...
    Adeno-associated virus (AAV) is a small, helper-dependent, single-stranded DNA virus capable of transducing both dividing and non-dividing cells by delivering a ...
  18. [18]
    Lentiviral Vector Guide - Addgene
    Tropism dictates which types of host cells the lentiviral vector will infect. This can be altered by changing the envelope gene, a process called pseudotyping.
  19. [19]
    Lentiviral Vector Pseudotypes: Precious Tools to Improve Gene ...
    The vesicular stomatitis virus glycoprotein (VSV-G) is commonly used for pseudotyping as it enhances gene transfer into multiple hematopoietic cell types.
  20. [20]
    Adeno-associated virus as a delivery vector for gene therapy of ...
    Apr 3, 2024 · Adeno-associated virus (AAV) has emerged as a pivotal delivery tool in clinical gene therapy owing to its minimal pathogenicity and ability to establish long- ...
  21. [21]
    Adeno-Associated Virus Vector Genomes Persist as Episomal ... - NIH
    Recombinant adeno-associated virus (rAAV) vectors are capable of mediating long-term gene expression following administration to skeletal muscle.
  22. [22]
    Adenovirus vector production using low-multiplicity infection of 293 ...
    Viral stocks propagated in 293 cells were purified using CsCl density-gradient centrifugation and were stored at −80 °C. Adenovirus vector production by ...Missing: packaging | Show results with:packaging
  23. [23]
    Rapid, scalable, and low-cost purification of recombinant adeno ...
    Our results demonstrate that simple, rapid, and relatively low-cost methods can easily be implemented for obtaining a high-quality grade of gene therapy ...
  24. [24]
    Viral Vectors in Gene Therapy: Advantages & Disadvantages
    This article discusses common viral vectors for gene therapy, including their production, limitations and future potential.Common Viral Vectors Used In... · Considerations In Viral... · Future Direction
  25. [25]
    Viral Vector Systems for Gene Therapy: A Comprehensive Literature ...
    The most significant advantage that retroviral vectors offer is their ability to transform their ssRNA genome into a dsDNA molecule that stably integrates into ...Viral Vectors · Adenovirus Vectors · Retrovirus Vectors
  26. [26]
    Biosafety Guidelines for Viral Vector-Based Gene Therapies
    May 15, 2025 · AAV vectors can trigger a robust immune response, especially if the patient has preexisting immunity from exposure to natural AAV infections.Biosafety Guidelines For... · Viral Vector Platforms · Oncolytic Viruses In Cancer...
  27. [27]
    A brief account of viral vectors and their promise for gene therapy
    Jan 26, 2017 · The demonstration by the Varmus/Bishop lab in the mid-1970s that γ-retroviruses can naturally acquire cellular genes provided a strong ...
  28. [28]
    CRISPR/Cas9 therapeutics: progress and prospects - Nature
    Jan 16, 2023 · The first clinical trial of CRISPR/Cas9 technology was conducted by Lu and colleagues at West China Hospital in Sichuan, China. In October 2016, ...
  29. [29]
    Construction and application of adenoviral vectors - Cell Press
    Sep 7, 2023 · Here, we review the construction methods of adenoviral vectors, including “gutless” adenovirus and discuss application of adenoviral vectors as prophylactic ...
  30. [30]
    Roles of mRNA poly(A) tails in regulation of eukaryotic gene ...
    Mar 13, 2023 · The interplay between poly(A) tails, poly(A)-binding protein, translation, and mRNA decay plays a major role in regulating gene expression.
  31. [31]
    Differential regulation of microRNA stability - RNA Journal
    ABSTRACT. MicroRNAs (miRNAs) are endogenous single-stranded RNA molecules of about 21 nucleotides in length that are fundamental post-transcriptional ...
  32. [32]
    MicroRNAs control translation initiation by inhibiting eukaryotic ...
    Nov 22, 2005 · We find that the 5′ cap structure and the 3′ poly(A) tail are each necessary but not sufficient for full miRNA-mediated repression of mRNA ...
  33. [33]
    mRNA vaccines for infectious diseases: principles, delivery ... - Nature
    Aug 25, 2021 · Both the Moderna and Pfizer–BioNTech SARS-CoV-2 vaccines, which produced >94% efficacy in phase III clinical trials25, contain nucleoside- ...
  34. [34]
    Pseudouridine RNA avoids immune detection through impaired ...
    Sep 4, 2025 · Here, we report that RNase T2 and PLD exonucleases do not adequately process pseudouridine-containing RNA to generate TLR-agonistic ligands.
  35. [35]
    Cationic liposome-mediated RNA transfection. - PNAS
    Cationic liposome-mediated RNA transfection. R W Malone, P L Felgner, and ... 1987. Lipofection: a highly efficient, lipid-mediated DNA-transfection ...<|separator|>
  36. [36]
    RIG-I-like receptors: their regulation and roles in RNA sensing - Nature
    Mar 13, 2020 · Accurate recognition of immunostimulatory RNA and rapid signal activation by RLRs are crucial steps in the initiation of innate immunity. As ...
  37. [37]
    mRNA-based vaccines and therapeutics: an in-depth survey of ...
    Oct 7, 2023 · The second method is to perform RNA capping during transcription by adding a cap analog like ARCA (anti-reverse cap analog), which carries a ...
  38. [38]
    Optimizing mRNA translation efficiency through rational 5'UTR and 3 ...
    This study adopted a combinatorial screening strategy to enhance exogenous mRNA translation efficiency by de novo designing 5'UTRs and combining them with ...
  39. [39]
    mRNA-based therapeutics: powerful and versatile tools to combat ...
    Different delivery strategies contribute to distinct mRNA transfection efficiency, namely, electroporation (90%), lipofection (5–50%) and sonporation (5–50%).<|separator|>
  40. [40]
    A Single Immunization with Nucleoside-Modified mRNA Vaccines ...
    Our findings suggest that the nucleoside-modified mRNA-LNP vaccine platform can induce robust immune responses and is a promising candidate to combat COVID-19.
  41. [41]
    Strategies for simultaneous and successive delivery of RNA - PMC
    Nov 4, 2020 · Here, we investigated different co- and successive transfection approaches, with particular focus on in vitro transcribed messenger RNA (IVT-mRNA).
  42. [42]
    mRNA-Based Genetic Reprogramming - PMC - NIH
    Dec 14, 2018 · mRNA reprogramming is the most unambiguously “footprint-free,” most productive, and perhaps the best suited to clinical production of stem cells.
  43. [43]
    Reduced Cytotoxicity by Repetitive mRNA Transfection in ... - NIH
    Daily mRNA transfection is considered as an unbearable level of damage in these undifferentiated NPCs, possibly resulting in cell death. When NPCs are subjected ...
  44. [44]
    mRNA in the Context of Protein Replacement Therapy - PMC - NIH
    In this review, we present preclinical and clinical applications of mRNA as a tool for protein replacement therapy.
  45. [45]
    Systemic delivery of factor IX messenger RNA for protein ... - PNAS
    Feb 15, 2017 · Safe and efficient delivery of messenger RNAs for protein replacement therapies offers great promise but remains challenging.
  46. [46]
    RNA interference as a key to knockdown overexpressed ... - Nature
    Apr 11, 2006 · The advancement in the field of RNA interference (RNAi) has opened up a completely new strategy to silence genes involved in tumour progression ...
  47. [47]
    Clara Cell 10-kDa Protein Gene Transfection Inhibits NF-κB Activity ...
    These results indicate that CC10 gene transfer may inhibit airway inflammation through suppressing the activation of NF-κB.
  48. [48]
    High-content CRISPR screening | Nature Reviews Methods Primers
    Feb 10, 2022 · This Primer describes the basic and advanced concepts of CRISPR screening and its application as a flexible and reliable method for biological discovery.
  49. [49]
    High-content imaging-based pooled CRISPR screens in mammalian ...
    Jan 19, 2021 · CRISPR-based screening typically uses large genomic pools of single guide RNAs (sgRNAs). However, this approach is limited to phenotypes that ...
  50. [50]
    A Guide to Transient Expression of Membrane Proteins in HEK-293 ...
    Jul 18, 2016 · They also exhibit high transfection efficiency, faithful translation, and processing of proteins (Wurm, 2004) that will result in higher protein ...
  51. [51]
    Direct transfection of clonal organoids in Matrigel microbeads
    Jul 6, 2018 · We developed an innovative approach for transgene expression in 3D organoids by combining single-cell encapsulation in Matrigel microbeads using a microfluidic ...
  52. [52]
    Direct and efficient transfection of mouse neural stem cells and ...
    In vivo brain electroporation of DNA expression vectors is a widely used method for lineage and gene function studies in the developing and postnatal brain.Cre Mrna-Mediated... · Plasmids And Mrna... · Brdu Injections
  53. [53]
    Optogenetics: 10 years of microbial opsins in neuroscience - PMC
    Optogenetics is the combination of genetic and optical methods to cause or inhibit well-defined events in specific cells of living tissue and behaving animals.
  54. [54]
    Deep Learning‐Assisted Automated Single Cell Electroporation ...
    Mar 21, 2022 · A fully automated version of the nanofountain probe electroporation (NFP-E) system, a nanopipette-based single-cell electroporation method is presented
  55. [55]
    The Nobel Prize in Physiology or Medicine 2006 - Advanced ...
    The 2006 Nobel Prize was awarded for the discovery of RNA interference (RNAi), where double-stranded RNA suppresses gene activity.Missing: transfection | Show results with:transfection
  56. [56]
    Miniaturization of Gene Transfection Assays in 384 and 1536-Well ...
    The miniaturization of gene transfer assays to either 384 or 1536-well plates greatly economizes the expense and allows much higher throughput.
  57. [57]
    Glybera | European Medicines Agency (EMA)
    Glybera was granted marketing authorisation in the European Union (EU) on 25 October 2012 as a one time, single-administration gene therapy for adult patients ...Overview · Product information · Product details · Authorisation details
  58. [58]
    FDA approves innovative gene therapy to treat pediatric patients ...
    May 24, 2019 · Zolgensma is indicated for the treatment of children less than two years of age with SMA. The product is an adeno-associated virus vector-based ...
  59. [59]
    Safety and Efficacy of the BNT162b2 mRNA Covid-19 Vaccine
    Dec 10, 2020 · A two-dose regimen of BNT162b2 conferred 95% protection against Covid-19 in persons 16 years of age or older. Safety over a median of 2 months was similar to ...
  60. [60]
    In vitro-transcribed antigen receptor mRNA nanocarriers for transient ...
    Nov 27, 2020 · Here we report an injectable nanocarrier that delivers in vitro-transcribed (IVT) CAR or TCR mRNA for transiently reprograming of circulating T cells.
  61. [61]
    CRISPR Clinical Trials: A 2025 Update - Innovative Genomics Institute
    Jul 9, 2025 · An update on the progress of CRISPR clinical trials with the latest data and a survey of the CRISPR landscape in 2025.
  62. [62]
    mRNA cancer vaccines from bench to bedside - Biomarker Research
    Dec 18, 2024 · On the other hand, there are some challenges associated with their application, including mRNA instability, insufficient uptake by immune cells, ...
  63. [63]
    What are the ethical issues surrounding gene therapy? - MedlinePlus
    Feb 28, 2022 · Because of these ethical concerns, the U.S. Government does not allow federal funds to be used for research on germline gene therapy in people.