Next Article in Journal
A Modified Protocol for the Isolation, Culture, and Characterization of Human Smooth Muscle Cells from the Umbilical Cord
Previous Article in Journal
Non-Antibiotic Prophylaxis for Recurrent UTIs in Neurogenic Lower Urinary Tract Dysfunction (NAPRUN): Study Protocol for a Prospective, Longitudinal Multi-Arm Observational Study
Previous Article in Special Issue
The Impact of Introducing Midwives and also Mentoring on the Quality of Sexual, Reproductive, Maternal, Newborn, and Adolescent Health Services in Low- and Middle-Income Countries: An Integrative Review Protocol
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Protocol

An Improved Technique for Genotyping the ABCB1 Gene Variant of Exon 21 †

by
Johanna Romina Zuccoli
1,*,
Priscila Ayelén Pagnotta
2,3,
Viviana Alicia Melito
1,2,
Jimena Verónica Lavandera
4,
Victoria Estela Parera
1 and
Ana María Buzaleh
1,2
1
Centro de Investigaciones sobre Porfirinas y Porfirias (CIPYP), CONICET, Hospital de Clínicas José de San Martín, Universidad de Buenos Aires, Buenos Aires 1120, Argentina
2
Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires 2610, Argentina
3
Institute of Biology and Experimental Medicine (IBYME), CONICET, Buenos Aires 2490, Argentina
4
Cátedra de Bromatología y Nutrición, Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Santa Fe 3000, Argentina
*
Author to whom correspondence should be addressed.
To the memory of Dr. Alcira Batlle and Dr. María Victoria Rossetti.
Methods Protoc. 2023, 6(3), 53; https://doi.org/10.3390/mps6030053
Submission received: 26 February 2023 / Revised: 21 April 2023 / Accepted: 2 May 2023 / Published: 26 May 2023
(This article belongs to the Special Issue Methods and Protocols 2023)

Abstract

:
The Multidrug Resistance protein (ABCB1, MDR1) is involved in the transport of xenobiotics and antiretroviral drugs. Some variants of the ABCB1 gene are of clinical importance; among them, exon 12 (c.1236C>T, rs1128503), 21 (c.2677G>T/A, rs2032582), and 26 (c.3435C>T, rs1045642) have a high incidence in Caucasians. Several protocols have been used for genotyping the exon 21 variants, such as allele-specific PCR-RFLP using adapted primer to generate a digestion site for several enzymes and automatic sequencing to detect the SNVs, TaqMan Allele Discrimination assay and High-Resolution Melter analysis (HRMA). The aim was to describe a new approach to genotype the three variants c.2677G>T/A for the exon 21 doing only one PCR with the corresponding primers and the digestion of the PCR product with two restriction enzymes: BrsI to identify A allele and BseYI to differentiate between G or T. An improvement of this methodology was also described. The proposal technique here described is demonstrated to be very efficient, easy, fast, reproducible, and cost-effective.

1. Introduction

The multidrug resistance gene (MDR1, ABCB1) encodes for the P-glycoprotein (P-gp), an integral membrane protein of 170 kDA that belongs to the adenosine triphosphate–binding cassette protein superfamily [1,2,3,4,5]. The substrates recognized and transported by P-gp include a variety of drugs. MDR1 is expressed in many normal tissues. P-gp acts as a cellular efflux pump to control the intracellular concentration of substances being an effective cellular protector against toxic substances. P-gp and other ABC transporters play a key role in absorption, distribution, metabolism, and elimination of drugs, working as a barrier to the entry of compounds into the body or controlling their transfer rate between tissues and compartments [6,7,8,9,10,11,12,13,14]. The discovery of genetic variations that influence the function or expression of P-gp may have a direct impact on the therapy of several diseases and in a near future in the personalized therapy.
There has been described more than 50 SNVs, three of high frequency in exons 12, 21 and 26 that affect the activity and/or expression of de P-gp protein and the toxicity of the drugs [3,4,9,11,15].
The c.2677G>T/A (rs2032582) SNV in exon 21 is a change in the 2677 position of a G for a T or A. Its sequence is CTAGAAGGT [G/T/A]GGGAAGGT. The wild type G variant encodes for Alanine at position 893, while T variant does so for Serine and A for Threonine. T and A variants alter the transporter function respect to G (wild type) variant, affecting drugs pharmacokinetics [16]. Several studies have linked this variant to a different response of pharmacological treatments for diseases such as epilepsy, HIV, coagulation disorders and different types of cancer [17,18,19,20]. Moreover, an exhaustive investigation about rs2032582 genotypes and haplotypes of the ABCB1 gene was reported [21,22,23,24].
Pagnotta et al. [25] showed the relevance of the T allele in c.2677G>T/A variant in the triggering of Porphyria Cutanea Tarda (PCT) in patients with HIV, possibly through a mechanism involving antiretroviral therapy that are substrates of P-gp. The study was also performed in patients with Acute Intermittent Porphyria (AIP), with a major frequency of A allele in AIP individuals [26]. A bioinformatic analysis was performed using different databases to establish the role of this variant in the PCT-HIV association and the relationship with drugs used in HIV therapy, or those avoided for patients with AIP [27].
In an Egyptian population of patients with chronic myeloid leukemia, the frequency of the rs2032582 TT genotype was fewer than that in healthy individuals; moreover, the authors reported that in individuals under therapy with imatinib, the TT genotype was associated with a complete hematological response and also, it has been assigned a role of producing protective factors against drug resistance [28].
Several protocols have been described to genotype c.2677G>T/A variant in exon 21, like allele specific PCR [29], RFLP using adapted primer to generate a digestion site for several enzymes [30] and automatic sequencing [31]. Another genotyping approach used for this variant is the TaqMan Allele Discrimination assay that involves real-time PCR and specific probes [32,33]. Moreover, High-Resolution Melter analysis (HRMA) is a method based in high-resolution melting, a novel, closed-tube post-PCR method to analyze genetic variations in PCR amplicons prior to or as an alternative to sequencing [34].
In our method, genotyping was performed using PCR-RFLP for amplifying the c.2677G>T/A variant in exon 21 with specific designed primers followed by the digestion with two restriction enzymes to characterize the SNVs in exon 21 from genomic DNA.
The technique proposed would be useful for researchers studying ABCB1 gene variants to underlying the mechanisms that lead to the effectiveness of a xenobiotic used as therapeutically drug for diseases.
This study was conducted to design a new approach that allows us to genotype the three variants of allele 21 by performing only one PCR with specific primers and the use of two restriction enzymes. This proposal technique was very efficient, easy, fast, reproducible, and cost-effective. Moreover, an improvement of this methodology was also suggested.

2. Experimental Design

2.1. Equipment

  • Thermal Cycler
  • Thermostatic digital dry bath
  • Electrophoresis Tank and Power supply

2.2. Biological Material

The recruited cohorts consisted of Caucasian individuals of both sexes. Samples were collected from patients attending the Research Center on Porphyrins and Porphyrias (CIPYP) (Buenos Aires, Argentina) between March 2010 and December 2018. All individuals provided signed consent for participation. The present study conformed with the guidelines stated in the Declaration of Helsinki (www.wma.net/policies-post/wma-declaration-of-helsinki-ethical-principles-for-medical-research-involving-human-subjects, 18th WMA General Assembly, Helsinki, Finland, June 1964 and amended by 64th WMA General Assembly, Fortaleza, Brazil, October 2013) and was approved (June 2013 and certified again in July 2015) by the Institutional Research Ethics Committee of the CIPYP, National Scientific and Technical Research Council.
DNA samples obtained from whole peripheral blood with EDTA were extracted, using the Illustra blood genomicPrep Mini Spin kit (Invitrogen; Thermo Fisher Scientific, Inc.; Waltham, Massachusetts, USA). The amount of genomic DNA obtained was fluorometrically quantified using Qubit Fluorometric Quantification System.

3. Procedure

Validation: This method was initially carried out by triplicate in 10 DNA samples and after that, it was performed in more than 450 DNA samples [26,27,35,36,37,38].

3.1. Primers Design

Use the DNASTAR software (LaserGene) to design the forward and reverse primers. This program led to determine in silico the primer length, length difference of primer pairs, PCR product length, GC proportion, melting temperature (Tm), GC clamp, dimer (including cross-dimers and self-dimers), hairpin structure, and specificity. All these properties are showed in Table 1. Moreover, a target SNV can be discriminated by digestion using available restriction enzymes. The BrsI restriction enzyme could differentiate the presence of the A allele from the G allele. If the A allele is present, the digestion product consists of three bands of 491 bp, 433 bp and 177 bp; while allele G/T shows two bands of 668 bp and 433 bp. BseYI restriction enzyme produced only two bands of 615 y 486 bp when the G allele is present or one band of 1101 bp for the T allele.

3.2. Genotypic Evaluation

Genotyping is performed using PCR-RFLP (Figure 1), amplifying a PCR product that contains the c.2677G>T/A variant in exon 21 with specific designed primers using the SeqBuilder and PrimerSelect (LaserGene) programs: Primer Forward: 5′GCTTTAGTAATGTTGCCGTGAT3′, Primer Reverse: 5′ATACCCCTAGCATTTTTCCATA3′. Prepare the PCR reaction mixture in a total volume of 25 µL, containing 14 mM Tris-HCl; 2 mM MgCl2; 0.2 mM dNTPs; 0.52 μM of each primer, 2.5 units of Taq DNA polymerase and 10 μL of template DNA (100–500 ng). The genomic DNA sample must have good integrity, quality, and concentration. Make sure to quantify DNA concentration of the samples and perform a horizontal agarose gel electrophoresis to check the quality and integrity of the genomic DNA. Carry out PCR in a Thermal Cycler with an initial denaturation for 3 min at 95 °C followed by 35 cycles of denaturation at 94 °C for 1 min, annealing at 58 °C for 30 s, and extension at 72 °C for 2 min. Perform terminal elongation at 72 °C for 10 min. The total time needed in the performance of the PCR is 2:26 h. Evaluate the 1101 bp PCR product on 1.5% agarose gels using ethidium bromide (30 min, 100 V) as the stain and under the Gel Imaging system.
To identify the A allele, digest the PCR product with the BrsI restriction enzyme (New England Biolab, Ipswich, Massachusetts, USA) during 2 h at 65 °C. Analyze the resulted product in 2.5% agarose gels dotted with ethidium bromide (40 min at 80 V). The digestion pattern has three bands of 491 bp, 433 bp y 177 bp for the wild type (G/G). The pattern for G/T is about two bands of 668 bp and 433 bp. To identify the G/T alleles, digest the PCR product with BseYI restriction enzyme (Invitrogen) during 1 h at 37 °C, in parallel. Then, analyze the results on an agarose 2.5% gel electrophoresis stained with ethidium bromide (40 min, 80 V). The allele G pattern shows two bands of 615 bp and 486 bp and the T allele pattern shows only 1 band of 1101 bp.
To validate this method, the results were confirmed using Sanger’s automatic sequencing of the PCR product (Sequencer AB13730XL, Macrogen, Seul, South Korea), also known as Dideoxy chain termination sequencing. This methodology involves the incorporation of chain-terminating nucleotides into a complementary DNA strand to a single-stranded DNA template. The chain-terminating nucleotides are radioactive or fluorescent labelled; the detector of the equipment can discriminate the label to determine the nucleotide (A, T, G, C) in the DNA sequence.

3.3. Data Analysis

The bands obtained in agarose gels were classified as GG, GT and TT, +A or -A depending on the restriction enzyme used. The spherograms obtained via Sanger sequencing were manually analyzed. In order to setup the results of our technique, RFLP results were contrasted to that obtained with Sanger sequencing.

4. Expected Results

Figure 2 shows the band pattern of c.2677G>T/A in exon 21 of the ABCB1 gene after amplification and digestion with the two restriction enzymes.

4.1. Improvement of the PCR Technique

If DNA samples are of low quality, low concentration or even degraded DNA is present, you can solve these problems using MyTaq™ Extract-PCR Kit (Bioline, London, UK). This kit offers a rapid, easy, and safer alternative for the extraction and amplification of DNA with high specificity because this method incorporates a polymerase that exhibits increased affinity for DNA, thereby improving yield and that uses antibody hot-start technology to avoid non-specific amplification. Moreover, we facilitate PCR set-up because the kit includes all the reagents needed for PCR and a red dye that allows a direct gel loading when electrophoresis is performed.

4.2. Discussion

The developed method allowed us to genotype the three variants of exon 21 doing only one PCR with the corresponding primers and then digest the PCR product with two restriction enzymes. This technique demonstrated to be very efficient, easy, fast, reproducible, and cost-effective (Figure 2).
The main differences with other published methods are described below.
Cascorbi et al. [30] described an indirect method of genotyping for which it is necessary to use mismatch primers for the PCR generating two restriction sites for TaqII or BanI enzymes in the presence of the mutation.
Zoto et al. [29] used two different pair of primers with which two PCR products were obtained to identify the variants G or T.
Zhang et al. [31] used a specific allelic PCR to identify the three variants (G, T or A).
Other authors employed the TaqMan® PCR technique that consist of pre-optimized PCR primer pairs and two probes to amplify and detect specific alleles in genomic DNA (gDNA) [32,33]. This technique requires to perform a real-time PCR and the synthesis of specific probes, as well as a high-throughput real-time PCR instrument and also trained technical staff, which makes the determination more expensive and not accessible for all the laboratories.
Another technique used in the allelic discrimination of the rs2032582 variant is the HRMA assay, a post-PCR analysis method based on detecting small differences in PCR melting curves, which is specific and sensitive enough to distinguish nucleic acid variations [34]. Although this method is faster, simple, and less expensive than alternative approaches requiring labeled probes, it is necessary to have a high-resolution melting instrument and a master mix of reagents that increases the cost of each determination. Moreover, the performance and data analysis of HRMA results can be affected by type and quality of DNA source material, different isolation methods (DNA preparation), poor melting curve resolution and PCR optimization (primer design) and other issues [39].
The frequencies obtained using the described technique in Argentinean population (T allele: 0.52; A allele 0.03) [25] were similar to those reported for Caucasians (T allele: 0.39–0.43; A allele: 0.02–0.05) [1,29,30,40].
In conclusion, the main difference with the mentioned authors was that they used more complex and expensive methods, which need more time to achieve the result than our proposed technique.

5. Reagents Setup

Solutions: All reagents were of molecular grade.
Kit GFx Genomic blood DNA purification (Invitrogen: Waltham, Massachusetts, USA).
10× Tris-HCl buffer.
50 µM oligonucleotide primer forward: 50 pmol/µL in sterile H2O (store at −20 °C).
50 µM oligonucleotide primer reverse: 50 pmol/µL in sterile H2O (store at −20 °C).
Template DNA: 10 µL human genomic DNA.
25 mM 4 dNTP mix.
2 mM MgCl2.
2.5 U/µL Taq DNA polymerase (native or recombinant).
BrsI restriction enzyme (New England Biolab, Ipswich, MA, USA)
BseYI restriction enzyme (Invitrogen, Waltham, MA, USA)
Agarose 2.5% gel electrophoresis stained with ethidium bromide.
MyTaq™ Extract-PCR Kit (Bioline, London, UK) (optional).

Author Contributions

All authors have made substantial contributions to the paper about the design technique and the proposal method. Moreover, all authors contributed to the drafting and editing of the manuscript and approved the submitted version. In particular, J.R.Z., P.A.P., V.A.M. and J.V.L. carried out the experiments to optimize the methodology designing the primers and the experimental conditions for PCR; V.E.P. brought her expertise in molecular biology techniques and in the discussion of the results, and also facilitated the blood samples to perform the setup of the technique; A.M.B. supervised the project both in the development of the method and in the writing of the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This work has been supported by grants from the University of Buenos Aires (UBACYT 2014–2017, 01/Q839 and 01/Q287), Argentina.

Institutional Review Board Statement

The study was conducted in concordance to the guidelines stated in the Declaration of Helsinki. The study was approved by the Institutional Research Ethics Committee of the CIPYP, National Scientific and Technical Research Council (CONICET), University of Buenos Aires (UBA), Argentina (June 2013 and certified again in July 2015). All authors approved the submitted version of the manuscript for publication.

Informed Consent Statement

Patients signed the corresponding Informed Consent form.

Data Availability Statement

All data generated or analyzed during this study are included in this published article.

Acknowledgments

J. Lavandera, V. Parera, A.M. Buzaleh hold the post of Adjunct and Independent Scientific Researchers at the Argentine National Research Council (CONICET). V. Melito is a research assistant at the Technical Career from CONICET. P. Pagnotta is a fellow from CONICET and J. Zuccoli is a postdoctoral fellow from CONICET.

Conflicts of Interest

The authors declare that they have no competing interest.

References

  1. Hoffmeyer, S.; Burk, O.; von Richter, O.; Arnold, H.; Brockmöller, J.; Johne, A.; Cascorbi, I.; Gerloff, T.; Roots, I.; Eichelbaum, M.; et al. Functional polymorphisms of the human multidrug-resistance gene: Multiple sequence variations and correlation of one allele with P-glycoprotein expression and activity in vivo. Proc. Natl. Acad. Sci. USA 2000, 97, 3473–3478. [Google Scholar] [CrossRef] [PubMed]
  2. Li, Y.; Wang, Y.; Li, Y.; Yang, L. MDR1 gene polymorphisms and clinical relevance. Acta Genet. Sin. 2006, 33, 93–104. [Google Scholar] [CrossRef] [PubMed]
  3. Fung, K.L.; Gottesman, M.M. A synonymous polymorphism in a common MDR1 (ABCB1) haplotype shapes protein function. Biochim. Biophys. Acta 2009, 1794, 860–871. [Google Scholar] [CrossRef] [PubMed]
  4. Zolnerciks, J.; Booth-Genthe, C.; Gupta, A.; Harris, J.; Unadkat, J. Substrate- and species-dependent inhibition of P-glycoprotein-mediated transport: Implications for predicting in vivo drug interactions. J. Pharm. Sci. 2011, 100, 3055–3061. [Google Scholar] [CrossRef] [PubMed]
  5. Thomas, C.; Tampé, R. Structural and Mechanistic Principles of ABC Transporters. Annu. Rev. Biochem. 2020, 89, 605–636. [Google Scholar] [CrossRef]
  6. Fromm, M.F. P-glycoprotein: A defence mechanism limiting oral bioavailability and CNS accumulation of drugs. Int. J. Clin. Pharmacol. Ther. 2000, 38, 69–74. [Google Scholar] [CrossRef]
  7. Tanigawara, Y. Role of P-glycoprotein in drug disposition. Ther. Drug Monit. 2000, 22, 137–140. [Google Scholar] [CrossRef]
  8. Schwab, M.; Eichelbaum, M.; Fromm, M. Genetic polymorphisms of the human MDR1 drug transporter. Ann. Rev. Pharmacol. Toxicol. 2003, 43, 285–307. [Google Scholar] [CrossRef]
  9. Thuerauf, N.; Fromm, M. The role of the transporter P-glycoprotein for disposition and effects of centrally acting drugs and for the pathogenesis of CNS diseases. Eur. Arch. Psychiatry Clin. Neurosci. 2006, 256, 281–286. [Google Scholar] [CrossRef]
  10. Taheri, M.; Mahjoubi, F.; Omranipour, R. Effect of MDR1 polymorphism on multidrug resistance expression in breast cancer patients. Gen. Mol. Res. 2010, 9, 34–40. [Google Scholar] [CrossRef]
  11. Brambila-Tapia, A.J. MDR1 (ABCB1) polymorphisms: Functional effects and clinical implications. Rev. Investig. Clin. 2013, 65, 445–454. [Google Scholar]
  12. Wolking, S.; Schaeffeler, E.; Lerche, H.; Schwab, M.; Nies, A. Impact of genetic polymorphisms of ABCB1 (MDR1, P-Glycoprotein) on drug disposition and potential clinical implications: Update of the literature. Clin. Pharmacokinet. 2015, 54, 709–735. [Google Scholar] [CrossRef]
  13. Jurkic, K.C.; Subramanian, N.; Mark, A.; O’Mara, M.L. The reliability of molecular dynamics simulations of the multidrug transporter P-glycoprotein in a membrane environment. PLoS ONE 2018, 13, e0191882. [Google Scholar]
  14. Moore, J.M.; Bell, E.; Hughes, R.O.; Garfield, A.S. ABC transporters: Human disease and pharmacotherapeutic potential. Trends Mol. Med. 2023, 29, 152–172. [Google Scholar] [CrossRef]
  15. Mollazadeh, S.; Sahebkar, A.; Hadizadeh, F.; Behravan, J.; Arabzadeh, S. Structural and functional aspects of P-glycoprotein and its inhibitors. Life Sci. 2018, 214, 118–123. [Google Scholar] [CrossRef]
  16. Brant, S.; Panhuysen, C.; Nicolae, D.; Reddy, D.; Bonen, D.; Karaliukas, R.; Zhang, L.; Swanson, E.; Datta, L.; Moran, T.; et al. Ala893 polymorphism is associated with inflammatory bowel disease. Am. J. Hum. Genet. 2003, 73, 1282–1292. [Google Scholar] [CrossRef]
  17. Bart, G.; Giang, L.; Yen, H.; Hodges, J.; Brundage, R. Effect of HIV, antiretrovirals, and genetics on methadone pharmacokinetics: Results from the methadone antiretroviral pharmacokinetics study. Drug Alcohol Depend. 2021, 227, 109025. [Google Scholar] [CrossRef]
  18. Lähteenmäki, J.; Vuorinen, A.L.; Pajula, J.; Harno, K.; Lehto, M.; Niemi, M.; van Gils, M. Pharmacogenetics of Bleeding and Thromboembolic Events in Direct Oral Anticoagulant Users. Clin. Pharmacol. Ther. 2021, 110, 768–776. [Google Scholar] [CrossRef]
  19. Vivaldi, C.; Crucitta, S.; Catanese, S.; Cucchiara, F.; Arrigoni, E.; Pecora, I.; Rofi, E.; Fornaro, L.; Salani, F.; Massa, V.; et al. Comprehensive pharmacogenetic analysis of DPYD, UGT, CDA, and ABCB1 polymorphisms in pancreatic cancer patients receiving mFOLFIRINOX or gemcitabine plus nab-paclitaxel. Pharmacogenom. J. 2021, 21, 233–242. [Google Scholar] [CrossRef]
  20. Song, C.; Li, X.; Zhang, H.; Bao, J.; Lu, W.; Peng, Q.; Zhang, Y. Impact of MDR1 and UGT Gene Polymorphisms on Sodium Valproate Plasma Concentration in Patients with Epilepsy. Clin. Lab. 2022, 1, 68. [Google Scholar] [CrossRef]
  21. Saiz-Rodríguez, M.; Valdez-Acosta, S.; Borobia, A.; Burgueño, M.; Gálvez-Múgica, M.; Acero, J.; Cabaleiro, T.; Muñoz-Guerra, M.; Puerro, M.; Llanos, L.; et al. Influence of Genetic Polymorphisms on the Response to Tramadol, Ibuprofen, and the Combination in Patients with Moderate to Severe Pain after Dental Surgery. Clin. Ther. 2021, 43, e86–e102. [Google Scholar] [CrossRef]
  22. Chen, Y.; Lu, J.; Huang, C.; Wu, W.; Lee, K.; Liu, H.; Wu, L.S.-H. Pharmacogenetic study of methadone treatment for heroin addiction: Associations between drug-metabolizing gene polymorphisms and treatment efficacy. Pharmacogenet. Genom. 2021, 32, 31–38. [Google Scholar] [CrossRef]
  23. Irwin, M.; Ellingrod, V.; Smith, M. Pharmacogenetics of Methadone for Pain Management in Palliative Care. J. Pain Symptom Manage. 2022, 63, e142–e145. [Google Scholar] [CrossRef]
  24. Maeda, A.; Ando, H.; Irie, K.; Hashimoto, N.; Morishige, J.I.; Fukushima, S.; Okada, A.; Ebi, H.; Matsuzaki, M.; Iwata, H.; et al. Effects of ABCB1 and ABCG2 polymorphisms on the pharmacokinetics of abemaciclib. Eur. J. Clin. Pharmacol. 2022, 78, 1239–1247. [Google Scholar] [CrossRef]
  25. Pagnotta, P.; Melito, V.; Lavandera, J.; Parera, V.; Rossetti, M.; Zuccoli, J.; Buzaleh, A. Role of ABCB1 and glutathione S-transferase gene variants in the association of porphyria cutanea tarda and human immunodeficiency virus infection. Biomed. Rep. 2021, 14, 22. [Google Scholar] [CrossRef]
  26. Manrique Bojórquez, N.Z.; Zuccoli, J.; Pagnotta, P.; Parera, V.; Rossetti, M.; Batlle, A.; Melito, V.; Buzaleh, A. MDR1 polymorphims and Acute Intermittent Porphyria. Medicina 2018, 78 (Suppl. III), 197. [Google Scholar]
  27. Pagnotta, P.; Buscalia, M.; Zuccoli, J.; Melito, V.; Parera, V.; Buzaleh, A. Bioinformatic complementation of the role of genetic variants in the manifestation of Porphyrias. Medicina 2022, 82 (Suppl. V), 92–93. [Google Scholar]
  28. Elghannam, D.; Ibrahim, L.; Ebrahim, M.; Azmy, E.; Hakem, H. Association of MDR1 gene polymorphism (G2677T) with imatinib response in Egyptian chronic myeloid leukemia patients. Hematology 2014, 19, 123–128. [Google Scholar] [CrossRef]
  29. Zoto, T.; Kilickap, S.; Yasar, U.; Celik, I.; Bozkurt, A.; Babaoglu, M. Improved anti-emetic efficacy of 5-HT3 receptor antagonists in cancer patients with genetic polymorphisms of ABCB1 (MDR1) drug transporter. Basic Clin. Pharmacol. Toxicol. 2015, 116, 354–360. [Google Scholar] [CrossRef]
  30. Cascorbi, I.; Gerloff, T.; Johne, A.; Meisel, C.; Hoffmeyer, S.; Schwab, M.; Schaeffeler, E.; Eichelbaum, M.; Brinkmann, U.; Roots, I. Frequency of single nucleotide polymorphisms in the P-glycoprotein drug transporter MDR1 gene in white subjects. Clin. Pharmacol. Ther. 2001, 69, 169–174. [Google Scholar] [CrossRef]
  31. Zhang, Y.; Jiang, X.; Hu, Y.; Li, Z.; Su, L.; Wang, Z.; Ma, G. MDR1 genotypes do not influence the absorption of a single oral dose of 600 mg valacyclovir in healthy Chinese Han ethnic males. Br. J. Clin. Pharmacol. 2008, 66, 247–254. [Google Scholar] [CrossRef] [PubMed]
  32. Cusinato, D.; Lacchini, R.; Romao, E.; Moysés-Neto, M.; Coelho, E. Relationship of CYP3A5 genotype and ABCB1 diplotype to tacrolimus disposition in Brazilian kidney transplant patients. Br. J. Clin Pharmacol. 2014, 78, 364–372. [Google Scholar] [CrossRef] [PubMed]
  33. Tovilla-Zárate, C.; Vargas, I.; Hernández, S.; Fresán, A.; Aguilar, A.; Escamilla, R.; Saracco, R.; Palacios, J.; Camarena, B. Association study between the MDR1 gene and clinical characteristics in schizophrenia. Braz. J. Psychiatry 2014, 36, 227–232. [Google Scholar] [CrossRef] [PubMed]
  34. Smolarz, B.; Skalski, D.; Rysz, A.; Marchel, A.; Romanowicz, H.; Makowska, M. Polymorphism of the multidrug resistance 1 gene MDR1 G2677T/A (rs2032582) and the risk of drug-resistant epilepsy in the Polish adult population. Acta Neurol. Belg. 2017, 117, 849–855. [Google Scholar] [CrossRef]
  35. Zuccoli, J.; Melito, V.; Ruspini, S.; Lavandera, J.; Abelleyro, M.; Parera, V.; Rossetti, M.; Batlle, A.; Buzaleh, A. Análisis de polimorfismos del exón 21 del gen MDR1 en la asociación Porfiria Cutánea Tardía-VIH. J. Basic Appl. Genet. 2014, XXV, 112. [Google Scholar]
  36. Pagnotta, P.; Zuccoli, J.; Parera, V.; Rossetti, M.; Lavandera, J.; Batlle, A.; Buzaleh, A.; Melito, V. MDR1 polymorphisms in HIV infected individuals. Its influence in Porphyria Cutanea Tarda triggering. Medicina 2017, 77 (Suppl. 1), 262. [Google Scholar]
  37. Pagnotta, P.A. Polimorfismos del Gen de Resistencia a Multidrogas y de la Glutatión S-Transferasa. Su Efecto en el Desencadenamiento de la Porfiria Cutánea Tardía en Individuos Infectados con el Virus de la Inmunodeficiencia Humana. Master’s Thesis, University of Buenos Aires, Buenos Aires, Argentina, 2018. [Google Scholar]
  38. Manrique Bojórquez, N.Z. Polimorfismos del Gen de Resistencia a Multidrogas “ABCB1” en Familias con Porfiria Aguda Intermitente. Master’s Thesis, University of Buenos Aires, Buenos Aires, Argentina, 2019. [Google Scholar]
  39. Słomka, M.; Sobalska-Kwapi, M.; Wachulec, M.; Bartosz, G.; Strapagiel, D. High Resolution Melting (HRM) for High-Throughput Genotyping-Limitations and Caveats in Practical Case Studies. Int. J. Mol. Sci. 2017, 18, 2316. [Google Scholar] [CrossRef]
  40. Milojkovic, M.; Stojnev, S.; Jovanovic, I.; Ljubisavljevic, S.; Stefanovic, V.; Sunder, R. Frequency of the C1236T, G2677T/A and C3435T MDR1 gene polymorphisms in the Serbian population. Pharmacol. Rep. 2011, 63, 808–814. [Google Scholar] [CrossRef]
Figure 1. Work scheme of the proposed method.
Figure 1. Work scheme of the proposed method.
Mps 06 00053 g001
Figure 2. Band pattern of c.2677G>T/A in exon 21 of ABCB1 gene after amplification and digestion with restriction enzymes. (1) PCR product. Line 1: Marker, line 2: Band corresponding to PCR product (1101 bp). (2) Digestion with BrSI restriction enzyme of PCR product: Line 1: Marker; line 2: Pattern band showing the absence of A allele; line 3: Pattern band for A allele in heterozygosis. (3) Digestion with BreYI restriction enzyme of PCR product: Line 1: Marker; line 2: Pattern band showing G/G allele in homozygosis; line 3: Pattern band showing T/T allele in homozygosis; line 4: Pattern band showing heterozygosis genotype. (4) Sanger’s automatic sequencing of the PCR product (A) and after digestion with BrSI (B) or BreYI (C) restriction enzymes.
Figure 2. Band pattern of c.2677G>T/A in exon 21 of ABCB1 gene after amplification and digestion with restriction enzymes. (1) PCR product. Line 1: Marker, line 2: Band corresponding to PCR product (1101 bp). (2) Digestion with BrSI restriction enzyme of PCR product: Line 1: Marker; line 2: Pattern band showing the absence of A allele; line 3: Pattern band for A allele in heterozygosis. (3) Digestion with BreYI restriction enzyme of PCR product: Line 1: Marker; line 2: Pattern band showing G/G allele in homozygosis; line 3: Pattern band showing T/T allele in homozygosis; line 4: Pattern band showing heterozygosis genotype. (4) Sanger’s automatic sequencing of the PCR product (A) and after digestion with BrSI (B) or BreYI (C) restriction enzymes.
Mps 06 00053 g002
Table 1. Main characteristics of the designed primers.
Table 1. Main characteristics of the designed primers.
Forward Primer: MDR9_New
Reverse Primer: MDR10_New
5’ATACCCCTAGCATTTTTCCATA3’
5’GCTTTAGTAATGTTGCCGTGAT3’
DNA 250 pM, Salt 50 mMForward PrimerReverse Primer
Primer Tm
Primer Overall Stability
Primer Location
50.7 °C
−40.0 kc/m
23.44
49.5 °C
−40.3 kc/m
1123.1102
Product Tm – Primer Tm
Primers Tm Difference
Optimal Annealing Temperature
23.6 °C
1.2 °C
51.1 °C
Product Length
Product Tm (%GC Method)
Product GC Content
Product Tm at 6xSSC
1101 bp
73.1 °C
33.6%
94.7 °C
Product Melting Temperature (%GC Method)
SaltFormamide
mMxSSCxSSPE0%10%20%50%
1
10
50
165
330
500
1000
0.005
0.051
0.256
0.846
1.692
2.564
5.128
0.006
0.062
0.312
1.031
2.062
3.125
6.250
44.9
61.5
73.1
81.7
86.7
89.7
94.7
38.4
55.0
66.6
75.2
80.2
83.2
88.2
31.9
48.5
60.1
68.7
73.7
76.7
81.7
12.4
29.0
40.6
49.2
54.2
57.2
62.2
Tm: Melting Temperature. SSC: Saline Sodium Citrate Buffer. SSPE: Saline Sodium Phosphate EDTA Buffer.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Zuccoli, J.R.; Pagnotta, P.A.; Melito, V.A.; Lavandera, J.V.; Parera, V.E.; Buzaleh, A.M. An Improved Technique for Genotyping the ABCB1 Gene Variant of Exon 21. Methods Protoc. 2023, 6, 53. https://doi.org/10.3390/mps6030053

AMA Style

Zuccoli JR, Pagnotta PA, Melito VA, Lavandera JV, Parera VE, Buzaleh AM. An Improved Technique for Genotyping the ABCB1 Gene Variant of Exon 21. Methods and Protocols. 2023; 6(3):53. https://doi.org/10.3390/mps6030053

Chicago/Turabian Style

Zuccoli, Johanna Romina, Priscila Ayelén Pagnotta, Viviana Alicia Melito, Jimena Verónica Lavandera, Victoria Estela Parera, and Ana María Buzaleh. 2023. "An Improved Technique for Genotyping the ABCB1 Gene Variant of Exon 21" Methods and Protocols 6, no. 3: 53. https://doi.org/10.3390/mps6030053

Article Metrics

Back to TopTop