Abstract
Objectives
β-Thalassemias, caused by mutations in HBB, are hereditary blood disorders that impose a significant global health burden. Detecting these mutations through accurate genetic analysis is essential. This study aimed to create a panel of cell type reference materials for β-thalassemia genetic testing using prime editing (PE), a flexible and precise genomic editing method.
Methods
PE3 systems were designed to target specific HBB mutations, including single nucleotide variants (SNVs) (−32 (C>A), CD 17 (A>T)), insertions (CD 14/15 (+G), CD 71–72 (+A)), and deletions (CD 31 (−C), CD 41/42 (−TTCT)). HEK293T cells were edited to carry these mutations. Sanger sequencing was performed to confirm the accurate introduction of homozygous and heterozygous mutations. The cell lines were further verified using commercial reverse dot-blot hybridization and melting curve assays.
Results
We successfully constructed 12 stable HEK293T cell lines carrying the intended homozygous or heterozygous HBB mutations using PE3 systems. No off-target mutations in the HBB were detected by Sanger sequencing in these stable cell lines, even after more than 10 weeks of culturing. Additionally, all target mutations were accurately and consistently detected using two reverse dot-blot hybridization kits and one melting curve assay kit.
Conclusions
The 12 stable cell lines exhibited accurate and stable HBB mutations, making them valuable reference materials for β-thalassemia genetic testing. PE3 systems show potential for generating a comprehensive panel of HBB mutations, particularly those that are clinically rare.
Introduction
β-Thalassemias are a group of heterogeneous autosomal recessive hereditary blood disorders characterized by reduced (β+) or absent (β0) synthesis of the β-globin chain [1]. It is estimated that there are 80 to 90 million β-thalassemia carriers worldwide, and approximately 68,000 children born with various thalassemia syndromes each year [2], [3], [4]. Over 400 hemoglobin-β gene (HBB) mutations have been identified to date (Ithanet database, https://www.ithanet.eu/), including single-nucleotide variants (SNVs), small insertions, or deletions, which affect gene transcription, RNA processing, and mRNA translation [5]. Clinical severity could be affected by the mutation type. Patients who are homozygous or compound heterozygous for these mutations develop β-thalassemia major and require lifelong blood transfusions and iron chelation therapy [3]. Individuals with β-thalassemia traits have a 25 % risk of having children affected with thalassemia major if their partner is also a carrier. Nowadays, HBB mutations can be detected by laboratory molecular analysis based on reverse dot-blot hybridization (RDB) analysis, melting curve analysis, allele-specific amplification or sequence analysis [6], 7]. Accurate molecular analysis is essential to meet the high demand of population-based screening, genetic counseling, and prenatal diagnosis of β-thalassemia.
Reference materials are essential for validating and ensuring the accuracy and reliability of genetic testing across different assays, platforms, and laboratories in clinical genetic testing [8]. They also support external quality assessment programs, testing development and validation, as well as proficiency testing and training [9]. Therefore, the use of reference materials for laboratory quality assurance is mandated by regulatory requirements and professional guidelines. Traditionally, plasmids, pseudoviruses, residual patient samples or commercial cell lines are used as reference materials [10]. Ideally, reference materials should mimic patient samples and encompass all variant alleles or mutation types targeted by the assay [11]. Genomic DNA sourced from cell samples, preferably derived from residual patient specimens or immortalized B lymphoblastoid cell lines, can be used for clinical β-thalassemia molecular analysis [12]. However, collecting enough peripheral blood samples from corresponding mutation carriers can be challenging, especially for rare or homozygous HBB mutations. Additionally, immortalized B lymphoblastoid cell lines have greater production cost, longer production cycle, longer population doubling time and lower yield compared to conventional HEK293T cells [13]. To address the need for publicly available, renewable, and characterized genomic DNA reference materials for β-thalassemia, we proposed using programmable gene-editing tools to efficiently establish stable HEK293T cell lines with HBB mutations causing β-thalassemia.
Prime editing (PE), established by David Liu’s research group, introduces a novel two-component genome editing platform that enables targeted gene correction, precise substitution, insertion, or deletion, rather than gene disruption [14]. Unlike early gene editing techniques such as CRISPR-Cas9, PE operates without the requirement for double-strand breaks (DSBs) or homologous directed recombination (HDR). It achieves higher editing efficiency while avoiding adverse outcomes associated with DSBs, such as unnecessary mixtures of insertions and deletions (indels) at the target site and HDR-mediated correction inefficiency [15]. Additionally, compared to cytosine base editors (CBEs) and adenine base editors (ABEs), PE offers a broader range of editing types, including multiple-site editing, DNA fragment insertion, and deletion [16]. The prime editor 3 (PE3) system reduces the risk of cellular mismatch repair by incorporating an additional sgRNA that guides the editor to nick the unedited DNA strand, thus significantly improving editing efficiency [17]. Its applicability in gene editing has been validated in various cell lines including HEK293T, K562, HeLa cells [18], stem Leydig cells [19], and mice [20].
Since PE systems offer a remarkably versatile and precise gene editing platform capable of potentially generating all mutations of the HBB in β-thalassemia, we aimed to generate a panel of HEK293T cell lines with distinct HBB mutations using the PE3 system. We successfully established 12 HEK293T cell lines with various heterozygous or homozygous HBB mutations (including SNVs, insertions, and deletions). These new cell lines were thoroughly characterized by various methods, confirming their suitability as reference materials for β-thalassemia genetic testing.
Materials and methods
PE plasmid construction
pCMV-PE2-P2A-GFP (Addgene plasmid # 132776) and pU6-pegRNA-GG-acceptor (Addgene plasmid # 132777) were gifted from David Liu [14]. For prime editing, DNA oligonucleotides for pegRNA were subcloned into pU6-pegRNA-GG-acceptor plasmid. The nicking sgRNA was subcloned into pUC-U6-GW plasmid. The design of pegRNA and nicking sgRNA, and molecular cloning procedures were performed following published protocols [21]. The pegRNA sequences were designed using a website tool called pegFinder and then screened and verified in HEK293T cells [22]. The sequences of pegRNA and nicking sgRNA for each mutation were listed in Table S1 (Supporting information).
Cell culture
HEK293T cells were obtained from the American Type Culture Collection (ATCC # CRL-3216) and maintained in our laboratory. The cells were cultured in DMEM medium (Thermo Fisher Scientific) supplemented with 10 % fetal bovine serum (Thermo Fisher Scientific), 1 % penicillin-streptomycin (Thermo Fisher Scientific), and 1 % GlutaMAX Supplement (Thermo Fisher Scientific). Passaging of cells was performed using 0.25 % Trypsin-EDTA (Thermo Fisher Scientific) when confluence reached 70–80 %. The cells were cultured, incubated, and maintained at 37 °C in a humidified atmosphere containing 5 % CO2.
Cell transfection and establishment of new cell lines
HEK293T cells were seeded at a density of 13,000 cells per well in 96-well plates (Corning) and cultured overnight. Transfection was performed the next day using Lipofectamine 3000 (Thermo Fisher Scientific) according to the manufacturer’s instructions. Briefly, cells were cotransfected with 200 ng of the pCMV-PE2-P2A-GFP plasmid, 66 ng of the pU6-pegRNA-GG-acceptor plasmid, and 13 ng of the pUC-U6-GW plasmid. One day after transfection, the cells were subjected to puromycin selection (1 μg/mL) for 3 days to enrich for successfully transfected cells. Single-cell clones were obtained through the limited dilution method and cultured in standard DMEM to establish stable cell lines. The established cells were verified to be free from bacterial, fungal, and mycoplasma contamination, and their successful cryopreservation and recovery cycles were confirmed.
DNA extraction and sequencing analysis
Genomic DNA extraction was carried out by washing the cells with PBS (pH 7.4) followed by isolation using the ONE-4-ALL Genomic DNA Mini-Preps Kit (Sangon Biotech) as per the manufacturer’s protocol. PCR analysis targeting the HBB was conducted on the SLAN-96s PCR System (Hongshi Medical Technology) using 2 × Phanta Flash Master Mix (Vazyme, P510). The primers were designed to amplify the mutant regions, and the specific sequences of primers used were as follows: forward primer (F), TGAAGTCCAACTCCTAAG; reverse primer (R), AGAATCCAGATGCTCAAGG. Sanger sequencing was performed using Illumina HiSeq 2500 platform (Illumina) by Genewiz Biotech.
RDB testing assays
Two thalassemia gene diagnostic kits (βTHA-RDB and THA-RDB, Shenzhen Yaneng Bioscience) were used following the manufacturer’s protocol. The βTHA-RDB kit detects 17 types of HBB mutations, including 10 mutations with homozygous/heterozygous status (e.g., CD 17 (A>T), CD 31 (−C), CD 41/42 (−TTCT), CD 71–72 (+A), and −32 (C>A)), and 7 mutations without homozygous/heterozygous differentiation (e.g., CD 14/15 (+G)). The THA-RDB kit detects 6 α-thalassemia gene mutations and 17 β-thalassemia gene mutations. It can distinguish between homozygous and heterozygous status for CD 17 (A>T), CD 41/42 (−TTCT), CD 71–72 (+A), and −32 (C>A), but not for CD 31 (−C) or CD 14/15 (+G).
Genomic DNA was extracted using the ONE-4-ALL Genomic DNA Mini-Preps Kit. The DNA concentration was determined with a Nanodrop2000 spectrophotometer (Thermo Fisher Scientific). The DNA samples were prepared in 50 ng/μL and the OD260/OD280 values were 1.7–2. Subsequently, two technicians from different research groups received the DNA samples, with each technician performing one assay, without knowledge of the other’s actions. Additionally, the expected mutation in each sample was not disclosed to either technician. For the protocol, PCR was performed with 2 μL of DNA template and 23 μL of PCR MIX. The resulting amplicons were denatured and then selectively hybridized to test strips containing wild-type and mutant oligonucleotide probes immobilized as parallel lines. The hybridization reaction, washing, and color development were conducted using an automatic hybridization system (Shenzhen Yaneng Bioscience). A visible blue-purple precipitate at the probe dot indicated a positive result. The results were captured using a scanning machine.
Melting curve testing assay
The βTHA-Melt kit (Shenzhen Yaneng Bioscience) is based on two-tube reactions and four-color channels for probe-based melting curve analysis and can detect 23 HBB mutations that cause β-thalassemia. The analysis was performed using a standard real-time PCR instrument, where the melting curve and melting temperature (Tm) were automatically analyzed. Genotype information for each mutation was determined based on the fluorescence channel and difference in Tm between the wild-type and mutant (ΔTm). According to the manufacturer’s protocol, this kit detects both heterozygous and homozygous mutations including −32 (C>A), CD 17 (A>T), CD 71–72 (+A), CD 14/15 (+G), and CD 41/42 (−TTCT). Briefly, DNA samples prepared as described above were tested using the βTHA-Melt kit. Following PCR and melting curve analysis programs, Tm values in each channel were obtained and genotypes were determined based on the preset ΔTm provided by the manufacturer.
Results
Construction of novel HBB mutation HEK293T cell lines by PE3 system
We employed the PE3 system to establish HBB mutations in HEK293T cells. As Illustrated in Figure 1A, PE3 systems consist of a prime editor, combining a Cas9 nickase fused with an M-MLV reverse transcriptase (RT), along with a prime editing guide RNA (pegRNA) and an additional nicking sgRNA. The pegRNA comprises a spacer, scaffold, RT template (RTT), and primer-binding site (PBS). Upon delivery into cells, the pegRNA spacer guides the prime editor to the target site, where Cas9 nicks one DNA strand, exposing a 3′ end. The PBS then anneals to this end, serving as a substrate for RT to synthesize the desired edit encoded by the RTT. In the PE3 system, an additional sgRNA nicks the nonedited DNA strand, enhancing editing efficiency. Subsequently, the newly synthesized DNA strand is integrated into genomic DNA by cellular repair machinery to achieve correction.

Construction of HBB mutant cell lines using the PE3 system. (A) Architecture of pegRNA and the mechanism of the PE3 system. (B) Structure of HBB and the locations of HBB mutations. The blue areas denote exons. The numbers indicate the amino acid residues encoded by the three exons. (C) PegRNA sequences (RTT plus PBS) for each mutation. The desired edits are shown in red. (D) Construction workflow of HEK293T cell lines with heterozygous and homozygous mutations.
To demonstrate the editing capabilities of the PE3 system in HBB, we selected mutations including SNVs (−32 (C>A), CD 17 (A>T)), insertions (CD 14/15 (+G), CD 71–72 (+A)), and deletions (CD 31 (−C), CD 41/42 (−TTCT)). The locations of these mutations in HBB are shown in Figure 1B. For each site, we designed three pegRNAs to induce the desired HBB mutations, and successful editable pegRNA (RTT plus PBS) are presented (Figure 1C). To establish the stable HEK293T cells with HBB mutations, HEK293T cells were edited by transfecting plasmids containing the machineries of PE3 systems, followed by selection in culture medium supplemented with puromycin (Figure 1D). Correctly edited cells were verified by sequencing. The cells with heterozygous and homozygous HBB mutations were further cultured for at least 10 weeks. Finally, 12 HEK293T cell lines with various HBB mutations were obtained, and these cells were passaged and preserved accordingly.
Validation of HBB mutations in HEK293T cell lines
To verify the reliability of the genomic DNA sequences in these long-term cultured cell lines, the HBB region of both homozygous and heterozygous mutant cells was amplified for Sanger sequencing. Compared with their respective wild-type counterparts, cells with homozygous mutations had correct sequence variations, while cells with heterozygous mutations had both mutant and wild-type alleles (Figure 2). These results demonstrated accurate editing of HBB mutations, including SNVs, insertions, and deletions, at the desired sites, with no other mutation sites generated. The accuracy of the edits remained stable after 10 weeks of culturing.

Sanger sequencing verification of established HEK293T cell lines. Genomic DNA samples were extracted from HEK293T cells with wild-type, homozygous mutant or heterozygous mutant HBB alleles. The sense-strand sequencing results at six mutation sites in the HBB gene are shown. The specific HBB mutation for each cell line is listed on the left side. SNVs or insertions are indicated by underlines, while deletions are denoted by slashes.
Evaluation of HBB mutant HEK293T cell lines as reference materials
To validate the suitability of these novel cell lines as reference materials for β-thalassemia genetic testing, we conducted RDB assays (THA-RDB and βTHA-RDB kits) and a melting curve assay (βTHA-Melt kit) with genomic DNA extracted from cell lines with wild-type, heterozygous mutant and homozygous mutant HBB. The results from the THA-RDB and βTHA-RDB kits (Figure 3) showed that all the normal control dots in the wild-type samples were colored, while no mutant dots were observed. In the heterozygous and homozygous mutant samples, indicative mutant dots were observed on the chips, while no other mutant dots were detected. Furthermore, in the βTHA-RDB kits, the normal control dot for each mutation (−32 (C>A), CD 17 (A>T), CD 71–72 (+A), CD 31 (−C), or CD 41/42 (−TTCT)) was colored in heterozygous samples but disappeared in homozygous samples. Similar results, except for those for CD 31 (−C), were observed for the THA-RDB kits. These findings showed that the selected HBB mutations and the heterozygous and homozygous statuses of our novel cell lines were effectively distinguished in RDB assays.

Reverse dot-blot assay results for established HEK293T cell lines with various HBB mutations. Genomic DNA samples extracted from HEK293T cells with wild type, homozygous mutant or heterozygous mutant HBB were analyzed using two commercial kits, named βTHA-RDB and THA-RDB. The specific HBB mutation for each cell line is listed on the left side. Strips after hybridization and color development are displayed. The blue dots represent the respective normal controls or mutations listed on each strip. The positions of the mutant genotype are indicated by solid red boxes, while those of the representative wild-type genotype are marked by red dashed boxes.
Further validation of these cell lines was conducted through a melting curve assay using the βTHA-Melt kit (Figure 4). Compared to the Tm value of the wild-type genotype, shifts in Tm values were accurately detected in homozygous mutations of −32 (C>A), CD 17 (A>T), CD 71–72 (+A), CD 14/15 (+G), and CD 41/42 (−TTCT). The melting curve analysis of heterozygous mutations revealed Tm values corresponding to both wild-type and mutant HBB simultaneously. These results affirm that the HBB heterozygous and homozygous mutations were accurately introduced into the HEK293T cells, facilitating genotyping via RDB or melting curve analysis.

Melting curve assay results for established HEK293T cell lines with various HBB mutations. Genomic DNA samples extracted from HEK293T cells with wild type, homozygous mutant or heterozygous mutant HBB were analyzed using the βTHA-Melt kit. The melting curves and corresponding Tm values for the mutant detection channels are presented. The Tm values of wild-type and mutant HBB genotypes are indicated by solid boxes and dashed boxes, respectively. The specific HBB mutation for each cell line is listed on the left side.
Discussion
β-Thalassaemia is one of the most common inherited blood disorders and is recognized as a notable public health concern in Asia, including China [23], 24]. The objective of this study was to develop a set of publicly accessible genomic DNA reference materials from stable cell lines for β-thalassaemia genetic testing. We successfully established 12 genetic stable HEK293T cell lines with HBB mutations using PE3 systems.
Different types of reference materials, such as residual patient samples, plasmids [25], and cell lines [12], 26], are utilized for β-thalassaemia and other heritable disorders. Each type has its own advantages and limitations. Cell lines, unlike plasmid samples, which are prone to contamination due to their high copy number, offer the advantage of mimicking both heterozygous and homozygous genotypes, as well as real sample extraction and testing processes [27]. Compared with lymphoblastoid cell lines (LCLs) immortalized from blood samples collected from mutation carriers, genetically modified cell lines are particularly suitable for β-thalassemia reference materials. Due to the extensive number of HBB mutations and the low frequency of most mutations [4], 28], it is challenging to collect clinical patient samples or establish LCLs from them to obtain reference materials with a wide range of mutations. This is evident in the fact that from China’s National Institutes for Food and Drug Control or Coriell Cell Repositories, only 10–18 types of β-thalassemia variations’ genomic DNA samples from LCLs can be obtained [28]. Moreover, some β-thalassemia testing products require both heterozygous and homozygous reference materials, which can be challenging to obtain due to the low frequency of homozygous carriers [29], 30]. This challenge is exacerbated by the fact that most homozygous carriers of the β+ phenotype are β-thalassemia major, with some cases even resulting in fatality [31]. For these reasons, we chose HEK293T cells to obtain homozygous and heterozygous cell lines with specific mutations through genetically modified methods. The mutations we selected included −32 (C>A), which is a mutation in the gene promoter that corresponds to the β+ phenotype. The other mutations result in the β0 phenotype: CD17 (A>T) leads to nonsense codons, while CD 14/15 (+G), CD 71–72 (+A), CD 31 (−C), and CD 41/42 (−TTCT) cause frameshift mutations [5], 32]. Mutations such as −32 (C>A), CD 14/15 (+G), CD 27/28 (+C), and CD 31 (−C) are considered rare in China, with a prevalence potentially lower than 0.05 % [28]. Our novel cell lines can encompass rare mutations and obtain clinically challenging homozygous genotypes, thereby addressing the limitations associated with plasmids and LCLs as β-thalassemia reference materials.
Gene editing technology, especially CRISPR-Cas systems, provides powerful tools for targeted manipulation of the genome in cells and animals [16]. PE systems are novel CRISPR-based genomic editing methods. As illustrated in Figure 1A, PE systems function independently of DSBs, enabling targeted gene correction rather than gene disruption [33]. The PE3 system has been demonstrated to achieve all 12 single-nucleotide substitutions, small insertions, small deletions and deletions up to 80 bp in HEK293T cells, with an editing efficiency of up to 20 % and avoiding excess indel formation [14]. Thus, the wide-ranging editing capabilities of PE are ideally suited for the diverse mutation types found in HBB. Our study successfully targeted and achieved three typical HBB mutations (SNVs, small insertions, and deletions). Notably, the successful edits of −32 (C>A) and CD 17 (A>T) also showed the expanded editing capabilities of PE compared to those of base editors, which are limited to installing the A to G or C to T transition [34], 35]. Moreover, in the PE system, the desired edits are encoded by the RTT within the pegRNA, minimizing off-target edits compared to CRISPR-Cas9 or base editing [14], 36]. Given the existence of potentially hundreds of β-thalassemia-related mutations in HBB, PE systems with minimal off-target effects provide an optimal strategy for generating mutations in HBB. The use of PE3 systems to generate HBB mutant cell lines in our study also paves the way for using subsequent versions of PEs, such as PE4, PE5 or twinPE [37], 38], to target more gene mutations in α- and β-thalassemia.
For genetic testing applications, reference materials based on cell lines with stable long-term cultivation characteristics and well-defined qualitative and quantitative properties are necessary [11]. Following initial sequencing verification, the cells were cultured for at least 10 weeks and characterized using Sanger sequencing, RDB analysis, or melting curve analysis in this study. The results of DNA sequencing (Figure 2) showed that our cell lines carrying HBB mutations did not exhibit nontarget mutations even after extended culture. Additionally, thalassemia diagnosis in the laboratory typically involves molecular analysis, with RDB and melting curve assays being widely used [6], 39]. The RDB kits used in this study were approved by the China National Medical Products Administration for detecting β-thalassemia mutations. The melting curve kit is also well-developed for β-thalassemia genotyping. Our novel HEK293T cell lines were successfully characterized using RDB and melting curve assays, demonstrating their suitability for quality assurance of β-thalassemia genotyping. It should be noted that CD14/15 (+G) and CD31 (−C), which are rare mutations in clinical practice [29], could not be distinguished for homozygous/heterozygous status using βTHA-RDB or THA-RDB kits. CD31 (−C) was also undetectable with the βTHA-Melt kit. These facts also highlight the current need for reference materials for rare mutations.
Various molecular testing methods, including quantitative polymerase chain reaction (qPCR), digital PCR, and next-generation sequencing (NGS), are also employed for β-thalassemia diagnosis [40]. These innovative technological platforms expand the detection range of HBB mutations causing β-thalassemia disease, increasing the demand for relevant reference materials. It is promising that our cell lines could serve as reliable reference materials for other molecular analysis approaches beyond RDB and melting curve assays.
In conclusion, we used PE3 systems to generate stable HEK293T cell lines with homozygous or heterozygous HBB mutations, providing valuable reference materials for genetic testing of β-thalassemia. Given the ease of manipulation, cost-effectiveness, and rapid proliferation of HEK293T cell lines, these cell lines, along with future ones that include rare mutations, can contribute to a comprehensive set of reference materials for clinically relevant HBB mutations.
-
Research ethics: Not applicable.
-
Informed consent: Not applicable.
-
Author contributions: The authors have accepted responsibility for the entire content of this manuscript and approved its submission. B.R. and X.X.: conceptualization and original draft preparation. J.L.: conducting laboratory experiments. W.T.: data extraction and analysis. K.L. and J.X.: review & editing. Y.Z.: data extraction. X.X. and Q.Z.: project supervision and review & editing.
-
Use of Large Language Models, AI and Machine Learning Tools: None declared.
-
Conflict of interest: The authors state no conflict of interest.
-
Research funding: None declared.
-
Data availability: The raw data can be obtained on request from the corresponding author.
References
1. Weatherall, DJ. Phenotype-genotype relationships in monogenic disease: lessons from the thalassaemias. Nat Rev Genet 2001;2:245–55. https://doi.org/10.1038/35066048.Search in Google Scholar PubMed
2. Taher, AT, Musallam, KM, Cappellini, MD. Beta-thalassemias. N Engl J Med 2021;384:727–43. https://doi.org/10.1056/nejmra2021838.Search in Google Scholar
3. Modell, B, Darlison, M. Global epidemiology of haemoglobin disorders and derived service indicators. Bull World Health Organ 2008;86:480–7. https://doi.org/10.2471/blt.06.036673.Search in Google Scholar PubMed PubMed Central
4. Colah, R, Gorakshakar, A, Nadkarni, A. Global burden, distribution and prevention of beta-thalassemias and hemoglobin E disorders. Expert Rev Hematol 2010;3:103–17. https://doi.org/10.1586/ehm.09.74.Search in Google Scholar PubMed
5. Thein, SL. The molecular basis of beta-thalassemia. Cold Spring Harb Perspect Med. 2013;3:a011700. https://doi.org/10.1101/cshperspect.a011700.Search in Google Scholar PubMed PubMed Central
6. Origa, R. Beta-thalassemia. Genet Med 2017;19:609–19. https://doi.org/10.1038/gim.2016.173.Search in Google Scholar PubMed
7. Xiong, F, Huang, Q, Chen, X, Zhou, Y, Zhang, X, Cai, R, et al.. A melting curve analysis – based PCR assay for one-step genotyping of beta-thalassemia mutations a multicenter validation. J Mol Diagn 2011;13:427–35. https://doi.org/10.1016/j.jmoldx.2011.03.005.Search in Google Scholar PubMed PubMed Central
8. Vesper, HW, Miller, WG, Myers, GL. Reference materials and commutability. Clin Biochem Rev 2007;28:139–47.Search in Google Scholar
9. Chen, B, Gagnon, M, Shahangian, S, Anderson, NL, Howerton, DA, Boone, JD, et al.. Good laboratory practices for molecular genetic testing for heritable diseases and conditions. MMWR Recomm Rep 2009;58:1–37. quiz CE-1-4.Search in Google Scholar
10. Scott, SA. The genetic testing reference materials coordination program: over 10 years of support for pharmacogenomic testing. J Mol Diagn 2023;25:630–3. https://doi.org/10.1016/j.jmoldx.2023.07.001.Search in Google Scholar PubMed PubMed Central
11. Kalman, LV, Datta, V, Williams, M, Zook, JM, Salit, ML, Han, JY. Development and characterization of reference materials for genetic testing: focus on public partnerships. Ann Lab Med 2016;36:513–20. https://doi.org/10.3343/alm.2016.36.6.513.Search in Google Scholar PubMed PubMed Central
12. Yin, Z, Qu, S, Huang, C, Chen, F, Li, J, Chen, S, et al.. Development of a genomic DNA reference material panel for thalassemia genetic testing. Int J Lab Hematol 2020;42:510–7. https://doi.org/10.1111/ijlh.13213.Search in Google Scholar PubMed
13. Hussain, T, Mulherkar, R. Lymphoblastoid cell lines: a continuous in vitro source of cells to study carcinogen sensitivity and DNA repair. Int J Mol Cell Med 2012;1:75–87.Search in Google Scholar
14. Anzalone, AV, Randolph, PB, Davis, JR, Sousa, AA, Koblan, LW, Levy, JM, et al.. Search-and-replace genome editing without double-strand breaks or donor DNA. Nature 2019;576:149–57. https://doi.org/10.1038/s41586-019-1711-4.Search in Google Scholar PubMed PubMed Central
15. Chen, PJ, Liu, DR. Prime editing for precise and highly versatile genome manipulation. Nat Rev Genet 2023;24:161–77. https://doi.org/10.1038/s41576-022-00541-1.Search in Google Scholar PubMed PubMed Central
16. Anzalone, AV, Koblan, LW, Liu, DR. Genome editing with CRISPR-Cas nucleases, base editors, transposases and prime editors. Nat Biotechnol 2020;38:824–44. https://doi.org/10.1038/s41587-020-0561-9.Search in Google Scholar PubMed
17. Kim, HK, Yu, G, Park, J, Min, S, Lee, S, Yoon, S, et al.. Predicting the efficiency of prime editing guide RNAs in human cells. Nat Biotechnol 2021;39:198–206. https://doi.org/10.1038/s41587-020-0677-y.Search in Google Scholar PubMed
18. Nelson, JW, Randolph, PB, Shen, SP, Everette, KA, Chen, PJ, Anzalone, AV, et al.. Engineered pegRNAs improve prime editing efficiency. Nat Biotechnol 2022;40:402–10. https://doi.org/10.1038/s41587-021-01039-7.Search in Google Scholar PubMed PubMed Central
19. Xia, K, Wang, F, Tan, Z, Zhang, S, Lai, X, Ou, W, et al.. Precise correction of Lhcgr mutation in stem Leydig cells by prime editing rescues hereditary primary hypogonadism in mice. Adv Sci 2023;10:e2300993. https://doi.org/10.1002/advs.202300993.Search in Google Scholar PubMed PubMed Central
20. Gao, P, Lyu, Q, Ghanam, AR, Lazzarotto, CR, Newby, GA, Zhang, W, et al.. Prime editing in mice reveals the essentiality of a single base in driving tissue-specific gene expression. Genome Biol 2021;22:83. https://doi.org/10.1186/s13059-021-02304-3.Search in Google Scholar PubMed PubMed Central
21. Doman, JL, Sousa, AA, Randolph, PB, Chen, PJ, Liu, DR. Designing and executing prime editing experiments in mammalian cells. Nat Protoc 2022;17:2431–68. https://doi.org/10.1038/s41596-022-00724-4.Search in Google Scholar PubMed PubMed Central
22. Chow, RD, Chen, JS, Shen, J, Chen, S. A web tool for the design of prime-editing guide RNAs. Nat Biomed Eng 2021;5:190–4. https://doi.org/10.1038/s41551-020-00622-8.Search in Google Scholar PubMed PubMed Central
23. Hokland, P, Daar, S, Khair, W, Sheth, S, Taher, AT, Torti, L, et al.. Thalassaemia-A global view. Br J Haematol 2023;201:199–214. https://doi.org/10.1111/bjh.18671.Search in Google Scholar PubMed
24. Wang, WD, Hu, F, Zhou, DH, Gale, RP, Lai, YR, Yao, HX, et al.. Thalassaemia in China. Blood Rev 2023;60:101074. https://doi.org/10.1016/j.blre.2023.101074.Search in Google Scholar PubMed
25. Munkongdee, T, Nualkaew, T, Buasuwan, N, Hinna, N, Paiboonsukwong, K, Sripichai, O, et al.. Development of DNA controls for detection of beta-thalassemia mutations commonly found in Asian. Int J Lab Hematol 2020;42:727–33. https://doi.org/10.1111/ijlh.13292.Search in Google Scholar PubMed
26. Kalman, L, Leonard, J, Gerry, N, Tarleton, J, Bridges, C, Gastier-Foster, JM, et al.. Quality assurance for Duchenne and Becker muscular dystrophy genetic testing: development of a genomic DNA reference material panel. J Mol Diagn 2011;13:167–74. https://doi.org/10.1016/j.jmoldx.2010.11.018.Search in Google Scholar PubMed PubMed Central
27. Baoutina, A, Bhat, S, Zheng, M, Partis, L, Dobeson, M, Alexander, IE, et al.. Synthetic certified DNA reference material for analysis of human erythropoietin transgene and transcript in gene doping and gene therapy. Gene Ther 2016;23:708–17. https://doi.org/10.1038/gt.2016.47.Search in Google Scholar PubMed
28. Shang, X, Peng, Z, Ye, Y, Asan, ZX, Chen, Y, et al.. Rapid targeted next-generation sequencing platform for molecular screening and clinical genotyping in subjects with hemoglobinopathies. EBioMedicine 2017;23:150–9. https://doi.org/10.1016/j.ebiom.2017.08.015.Search in Google Scholar PubMed PubMed Central
29. Yang, Z, Cui, Q, Zhou, W, Qiu, L, Han, B. Comparison of gene mutation spectrum of thalassemia in different regions of China and Southeast Asia. Mol Genet Genomic Med 2019;7:e680. https://doi.org/10.1002/mgg3.680.Search in Google Scholar PubMed PubMed Central
30. Zhang, J, He, J, Zeng, XH, Su, J, Chen, H, Xu, YM, et al.. Gene mutation spectrum of beta-thalassemia in dai ethinic population of two border region in Chinese yunnan province. Zhongguo Shi Yan Xue Ye Xue Za Zhi 2016;24:157–61. https://doi.org/10.7534/j.issn.1009-2137.2016.01.030.Search in Google Scholar PubMed
31. Tesio, N, Bauer, DE. Molecular basis and genetic modifiers of thalassemia. Hematol Oncol Clin N Am 2023;37:273–99. https://doi.org/10.1016/j.hoc.2022.12.001.Search in Google Scholar PubMed PubMed Central
32. Tamana, S, Xenophontos, M, Minaidou, A, Stephanou, C, Harteveld, CL, Bento, C, et al.. Evaluation of in silico predictors on short nucleotide variants in HBA1, HBA2, and HBB associated with haemoglobinopathies. Elife 2022;11. https://doi.org/10.7554/elife.79713.Search in Google Scholar PubMed PubMed Central
33. Jinek, M, Chylinski, K, Fonfara, I, Hauer, M, Doudna, JA, Charpentier, E. A programmable dual-RNA-guided DNA endonuclease in adaptive bacterial immunity. Science 2012;337:816–21. https://doi.org/10.1126/science.1225829.Search in Google Scholar PubMed PubMed Central
34. Komor, AC, Kim, YB, Packer, MS, Zuris, JA, Liu, DR. Programmable editing of a target base in genomic DNA without double-stranded DNA cleavage. Nature 2016;533:420–4. https://doi.org/10.1038/nature17946.Search in Google Scholar PubMed PubMed Central
35. Gaudelli, NM, Komor, AC, Rees, HA, Packer, MS, Badran, AH, Bryson, DI, et al.. Programmable base editing of A*T to G*C in genomic DNA without DNA cleavage. Nature 2017;551:464–71. https://doi.org/10.1038/nature24644.Search in Google Scholar PubMed PubMed Central
36. Kim, DY, Moon, SB, Ko, JH, Kim, YS, Kim, D. Unbiased investigation of specificities of prime editing systems in human cells. Nucleic Acids Res 2020;48:10576–89. https://doi.org/10.1093/nar/gkaa764.Search in Google Scholar PubMed PubMed Central
37. Anzalone, AV, Gao, XD, Podracky, CJ, Nelson, AT, Koblan, LW, Raguram, A, et al.. Programmable deletion, replacement, integration and inversion of large DNA sequences with twin prime editing. Nat Biotechnol 2022;40:731–40. https://doi.org/10.1038/s41587-021-01133-w.Search in Google Scholar PubMed PubMed Central
38. Chen, PJ, Hussmann, JA, Yan, J, Knipping, F, Ravisankar, P, Chen, PF, et al.. Enhanced prime editing systems by manipulating cellular determinants of editing outcomes. Cell 2021;184:5635–52 e29. https://doi.org/10.1016/j.cell.2021.09.018.Search in Google Scholar PubMed PubMed Central
39. Lin, M, Zhu, JJ, Wang, Q, Xie, LX, Lu, M, Wang, JL, et al.. Development and evaluation of a reverse dot blot assay for the simultaneous detection of common alpha and beta thalassemia in Chinese. Blood Cells Mol Dis 2012;48:86–90. https://doi.org/10.1016/j.bcmd.2011.12.001.Search in Google Scholar PubMed
40. Cousens, NE, Gaff, CL, Metcalfe, SA, Delatycki, MB. Carrier screening for beta-thalassaemia: a review of international practice. Eur J Hum Genet 2010;18:1077–83. https://doi.org/10.1038/ejhg.2010.90.Search in Google Scholar PubMed PubMed Central
© 2024 the author(s), published by De Gruyter, Berlin/Boston
This work is licensed under the Creative Commons Attribution 4.0 International License.
Articles in the same Issue
- Frontmatter
- Review
- Interaction of fetuin-A with obesity related insulin resistance and diabetes mellitus
- Research Articles
- Preanalytical errors in pediatric blood sampling: a systematic review of common challenges and risks
- The promotive role of reticulocalbin 3 (RCN3) in the pathogenesis of keloid via TGFβ1/Smad2/Smad7 signaling pathway in vitro
- Investigation into drug resistance to cisplatin in cancer stem cell-enriched population in non-small cell lung cancer
- The cytotoxic and antiproliferative effect of Polygala saponin XLIV on the human colorectal carcinoma cell line
- Development of HEK293T cell reference materials for β-thalassemia genetic testing using prime editing
- Investigation of propofol, fentanyl, and midazolam-related toxicity and the protective effect of midazolam on THLE-2 cell lines
- Leucine-rich α-2-glycoprotein 1 can be a novel angiogenic mediator in autosomal dominant polycystic kidney disease
- Asiaticoside reverses the inhibition effect of miR-184 on proliferation, migration and AKT phosphorylation of HTR-8/Svneo cells
- Perioperative D-dimer levels and head and neck cancer surgery: a prospective observational study
- Comprehensive analysis of KRT15 in pan-cancer and verification in lung adenocarcinoma
- Age and sex-dependent effects of nitrosative stress on the osmotic fragility of human red blood cells
- Evaluation of oxidative stress parameters in older patients with urinary incontinence
- Determination of reference change values for thyroid-related biomarkers: TSH, fT3, fT4, Tg, Anti-Tg, and Anti-TPO
- An evaluation of serum boron level in pregnancies with severe pre-eclampsia
- Involvement of SIRT3/FOXO1 and TLR4/NF-κB/NLRP3 pathways in protective effects of Δ(9)-tetrahydrocannabinol on diabetic cardiomyopathy in rats
- Evaluating the potential therapeutic effect of Rosa damascena Mill. essential oil on acetic acid-induced ulcerative colitis in rats
- Assessment of blood collection competency in phlebotomists: a survey study
Articles in the same Issue
- Frontmatter
- Review
- Interaction of fetuin-A with obesity related insulin resistance and diabetes mellitus
- Research Articles
- Preanalytical errors in pediatric blood sampling: a systematic review of common challenges and risks
- The promotive role of reticulocalbin 3 (RCN3) in the pathogenesis of keloid via TGFβ1/Smad2/Smad7 signaling pathway in vitro
- Investigation into drug resistance to cisplatin in cancer stem cell-enriched population in non-small cell lung cancer
- The cytotoxic and antiproliferative effect of Polygala saponin XLIV on the human colorectal carcinoma cell line
- Development of HEK293T cell reference materials for β-thalassemia genetic testing using prime editing
- Investigation of propofol, fentanyl, and midazolam-related toxicity and the protective effect of midazolam on THLE-2 cell lines
- Leucine-rich α-2-glycoprotein 1 can be a novel angiogenic mediator in autosomal dominant polycystic kidney disease
- Asiaticoside reverses the inhibition effect of miR-184 on proliferation, migration and AKT phosphorylation of HTR-8/Svneo cells
- Perioperative D-dimer levels and head and neck cancer surgery: a prospective observational study
- Comprehensive analysis of KRT15 in pan-cancer and verification in lung adenocarcinoma
- Age and sex-dependent effects of nitrosative stress on the osmotic fragility of human red blood cells
- Evaluation of oxidative stress parameters in older patients with urinary incontinence
- Determination of reference change values for thyroid-related biomarkers: TSH, fT3, fT4, Tg, Anti-Tg, and Anti-TPO
- An evaluation of serum boron level in pregnancies with severe pre-eclampsia
- Involvement of SIRT3/FOXO1 and TLR4/NF-κB/NLRP3 pathways in protective effects of Δ(9)-tetrahydrocannabinol on diabetic cardiomyopathy in rats
- Evaluating the potential therapeutic effect of Rosa damascena Mill. essential oil on acetic acid-induced ulcerative colitis in rats
- Assessment of blood collection competency in phlebotomists: a survey study