Next Article in Journal
Implication of Potential Differential Roles of the Two Phosphoglucomutase Isoforms in the Protozoan Parasite Cryptosporidium parvum
Next Article in Special Issue
A Modular Hepatitis E Virus Replicon System for Studies on the Role of ORF1-Encoded Polyprotein Domains
Previous Article in Journal
Distinct Schistosoma mansoni-Specific Immunoglobulin Subclasses Are Induced by Different Schistosoma mansoni Stages—A Tool to Decipher Schistosoma mansoni Infection Stages
Previous Article in Special Issue
From Screening to Therapy: Anti-HCV Screening and Linkage to Care in a Network of General Practitioners and a Private Gastroenterology Practice
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Comparison of Two Diagnostic Methods for the Detection of Hepatitis B Virus Genotypes in the Slovak Republic

by
Mariia Logoida
1,
Pavol Kristian
2,*,
Andrea Schreiberova
3,
Patrícia Denisa Lenártová
2,
Veronika Bednárová
1,
Elena Hatalová
1,
Ivana Hockicková
2,
Sylvia Dražilová
4,
Peter Jarčuška
4,
Martin Janičko
4,
Štefan Porhinčák
5 and
Monika Halánová
1
1
Department of Epidemiology, Faculty of Medicine, Pavol Jozef Šafárik University, 040 11 Kosice, Slovakia
2
Department of Infectology and Travel Medicine, Faculty of Medicine, Louis Pasteur University Hospital, Pavol Jozef Šafárik University, 041 90 Kosice, Slovakia
3
Department of Epizootiology, Parasitology and Protection of One Health, University of Veterinary Medicine and Pharmacy, 041 81 Kosice, Slovakia
4
2nd Department of Internal Medicine, Faculty of Medicine, Louis Pasteur University Hospital, Pavol Jozef Šafárik University, 040 11 Kosice, Slovakia
5
Centre of Applied Computer Science, P. J. Safarik University, 040 01 Kosice, Slovakia
*
Author to whom correspondence should be addressed.
Pathogens 2022, 11(1), 20; https://doi.org/10.3390/pathogens11010020
Submission received: 8 December 2021 / Revised: 22 December 2021 / Accepted: 22 December 2021 / Published: 24 December 2021

Abstract

:
The hepatitis B virus (HBV), belonging to the Hepadnaviridae family, is responsible for a global health concern still in the 21st century. The virus is divided into 10 genotypes, which differ in geographical distribution and in their effect on disease progression and transmission, susceptibility to mutations, and response to treatment. There are many methods for diagnostics of HBV and differentiating its genotypes. Various commercial kits based on real-time polymerase chain reaction (RT PCR) and hybridization available, as well as whole genome sequencing or the sequencing of only individual parts of the genomes. We compared a commercial kit AmpliSens HBV-genotype-FRT, based on RT PCR, with an adapted method of amplification of the surface genomic region combined with Sanger sequencing. In the examined samples we identified the A, B, C, D, and E genotypes. By PCR with Sanger sequencing, the genotypes were determined in all 103 samples, while by using the commercial kit we successfully genotyped only 95 samples, including combined genotypes, which we could not detect by sequencing.

1. Introduction

Viral hepatitis B is one of the most common viral infections in humans; it is spread worldwide and represents a global public health problem. The prevalence of hepatitis B varies worldwide and ranges from 0.7% of those infected in the adult population in low endemic regions to 6.2% in high endemic regions. In 2010, according to a European Centre for Disease Prevention and Control (ECDC) Technical report, the Slovak Republic was classified as a low endemic country for HBV infection [1]. However, there are still groups of the population in which the prevalence is higher. The cross-sectional population-based Hepa-Meta study, focused on the prevalence of viral hepatis, metabolic syndrome, and selected bacterial and parasitic infectious diseases in the Roma population living in segregated settlements, detected a 12.5% prevalence of HBV surface antigen (HBsAg) in these citizens [2,3]. Between years 2015 and 2020, in Slovak Republic there were on average 144 cases reported per year [4,5].
HBV is a partially double-stranded DNA virus roughly 3200 nucleotides in length and belonging to the Hepadnaviridae family [6]. The genome contains four partially or entirely overlapping open reading frames (C, P, S, and X), which encode seven proteins: pre-core and core protein (HBeAg and HBcAg); polymerase protein (reverse transcriptase, RT), preS1, preS2 and small hepatitis B surface proteins (SHB) (three forms of HBsAg) and X protein (transcriptional trans-activator protein) [7,8].
HBV is currently classified into 10 genotypes, A-J, according to differences in the complete genomic sequence, with the difference between the individual genotypes being about 8% and between subgenotypes about 4% [9,10]. The estimated mutation rate in HBV is determined to be approximately 10 −4 to 10 −6 nucleotide substitutions/site per year [11,12,13]. Significant differences between genotypes in geographical distribution have been observed. HBV genotype A is more common in Europe (A2), North America (A2), and Africa (A1, A3–A6), while genotype B and C is the most prevalent in Asia and the Pacific. HBV genotype D was detected in Middle East (D1), Central Asia (D1), Europe (D2), Japan (D2), Australia and Oceania (D4), India (D5), and South Africa (D6), and subgenotype D3 is spread worldwide. Genotype E is typical for Africa, while genotypes F and H for South, Central, and North America. HBV genotype G was detected in the USA, Germany, Italy, the UK, and France, and genotype I has been found in Asia and genotype J in Japan [7,14]. There is also a link between genotypes and their modes of transmission. In addition, different HBV genotypes can have variability in their clinical outcomes and response to treatment, including the development of drug resistance [15,16,17]. Therefore, data on the genotypes circulating in the population help to detect transmission pathways and serve as an epidemiological tool for monitoring the mode of transmission and clustering of the virus [18].
A lot of different techniques are available for HBV genotyping. Sequence and phylogenetic analysis of the entire HBV genome is still considered the gold standard for genotyping [19]. However, full genomic sequencing appears to be ineffective for regular use in clinical practice, particularly due to high costs, the time consumed, and the expertise required. Sequencing and phylogenetic analysis can be performed only on parts of the genome; most often the S region (S-surface) is used for genotyping. In general, the HBV S gene sequence is enough to assign genotypes [8,20]. In addition, to determine the HBV genotype, other methods can be used, such as restriction fragment length polymorphisms (RFLP), [21], PCR with specific primers and probes for single genotypes [22,23] or other methods based on hybridization technologies [14,24].
In this study, we compared the commercial kit AmpliSens HBV-genotype-FRT, based on real-time PCR using specific primers and probes for the A, B, C, and D genotypes with adapted direct and nested PCR, with primers which are used in INNO-LiPA HBV Genotyping kits. These primers target the HBV surface genomic region, which overlaps with the polymerase region, and this also allows us to detect the possible presence of an escape or resistance mutation [25]. The aim of this study was to compare two different diagnostic methods for the detection of Hepatitis B virus genotypes to better understand the molecular epidemiology of HBV in Slovakia, which can lead to better understanding of the origins and distribution patterns of HBV genotypes in patients in Slovakia, thus ensuring better patient management and appropriate treatment.

2. Results

A total of 103 people positively diagnosed with viral hepatitis B were examined for the detection of HBV genotype.
From the 103 examined serum samples, 95 were successfully genotyped by real-time PCR using the commercial kit AmpliSens HBV-genotype-FRT. In all 103 samples, the genotypes were determined by amplification of the surface genomic region (using primers from the INNO-LiPA assays) and Sanger sequencing.
Among all the samples successfully analyzed by AmpliSens HBV-genotype-FRT commercial kit, the prevalent genotype was D with 44.7%; genotype A was second with 37.9%, and the most prevalent combination genotype was A/D, represented by 3.9%. Other genotypes had 1% to 1.9% occurrence, as shown in Figure 1a.
The AmpliSens kit could not detect viral DNA in 8 of our samples (7.8%), which were determined by amplification of the surface genomic region and Sanger sequencing. Of these 8 samples, 5 were determined as a genotype D and 2 as genotype A. One of these 8 samples was determined as genotype E, as the AmpliSens kit is limited only to genotypes A, B, C, and D (Appendix A Table A1).
By amplification of the surface genomic region (using primers from the INNO-LiPA assays) and Sanger sequencing, we determined the genotype in all 103 samples. Obtained sequences were analyzed by tools, NCBI, Geno2Pheno, and Phylogenetic analysis by Mega X Software. According to NCBI annotation, we found the most prevalent genotype was genotype D, with subgenotype D1—16.5%, D2—6.8%, D3—29.1%, followed by A2—43.7%, and genotypes C1, E, and B4, each with 1% or 1.9%, as show in Figure 1b. Genotyping by Geno2Pheno[hbv] tools provided us the similar results: The most prevalent genotype was D (D1—18.4%. D2—7.8%, D3—24.3%, D4—1.9%), next A2—43.7% and genotypes B3, B4, and C2, E, each with 1%, Figure 1c.
By phylogenetic analysis, we verified our genotypes and subgenotypes and detected genotypes D with 52.4% (D3 with 32%, D1 with 12.6%, D2 with 6.8%, and D4 with 1%), A2 with 43.7%, B4 with 1.9%, and C1 and E each with 1%, as shown in Figure 1d.
We discovered that all results of genotyping produced by different tools were the same on the genotype level, with differences only at the subgenotype level. The results of phylogenetic analysis were very similar to the results by manual annotation in the NCBI database, with difference only in 4 samples. Three of samples were determined by NCBI annotation and by Geno2Pheno[hbv] tools as subgenotype D1, but according to phylogenetic analysis they belonged to genotype D3.
One of these samples, which was determined by NCBI annotation as a subgenotype D1, was determined by Geno2Pheno[hbv] tools as subgenotype D4 and according to phylogenetic analysis belonged to genotype D4. Nevertheless, branches with reference sequences for subgenotype D4 were arranged as a subtree for the D1 branch (Figure 2). Such ambiguities in branching could be caused by relatively short and maybe insufficiently divergent sequences.
With commercial kit AmpliSens HBV-genotype-FRT, it was possible to detect the same genotypes as with other methods (plus mixed genotypes), except for 8 samples, in which we were unable to determine genotype with this kit (Appendix A Table A1). Thus, the success rate of the commercial kit compared to other methods was 92.23%. As for the comparison of, for example, manual annotation in the NCBI database and the Geno2Pheno[hbv] tools concurred on 89.32% (on the subgenotype levels), and results of phylogenetic analysis using MEGA X software matched with the Geno2Pheno[hbv] tools in 88.35% and the results of NCBI annotation were same as analysis using MEGA X in 94.17% of samples.
In addition, the obtained sequences were used for the detection of clinically important resistant and escape mutations through different online tools, such as the Geno2Pheno[hbv] tool (https://hbv.geno2pheno.org/; accessed on 5 December 2021) and the HBV-Resistance interpretation tool (http://www.hiv-grade.de/hbv_grade/deployed/grade; accessed on 5 December 2021) (Appendix A Table A2). The most common mutations were the HBsAg escape mutations—A128V and P127T, which were typical for genotypes D and A, followed by the HBsAg escape mutations—D144 E and K122R and the compensatory mutation—S202I, which can cause to resistance to Entecavir, Baraclude®. Other HBsAg escape mutations occurred only individually: V173M, P120T, S143L, G145R, P120P together with P120S, C121C together with G145R, A128V together with M133I, and P142L together with D144A, as shown in Figure 3. All the important mutations were detected by both instruments, except the P127T mutation, which was detected only by the HBV-Resistance interpretation tool. Regarding neutral mutations, some were detected by only one of two software. All mutation data should be interpreted with caution since it is only a prediction and the region of interest of the genes is relatively short.

3. Discussion

Knowledge of the circulating genotypes in a community and data on existing mutations can lead to a better understanding of the molecular epidemiology of the hepatitis B virus and contribute to better patient management and treatment. HBV genotype can be confirmed by a variety of methods, such as sequence analysis of partial or whole genome, genotype-specific PCR assays, real-time PCR, RFLP, microarray (DNAChip), or fluorescence polarization assay [26]. The reference point for all methods is sequencing and phylogenetic analysis. While whole genome sequencing is the gold standard and the most reliable method, it is cumbersome to use in large scale studies and is expensive and requires expertise [19]; thus, sequencing of only a part of the genome can be a viable alternative. Phylogenetic analysis allows relative and evolutionary relatedness of sequences to be assessed and can also be performed on individual genes, especially on S gene [14].
Commercial kits make diagnostics easier, faster, and more convenient for the routine. One of the commercial methods to genotype HBV is the INNO-LIPA® HBV Genotyping (Fujirebio Europe, Tokyo, Japan) based on reverse hybridization. This method allows the identification of HBV genotypes A to G and shows high sensitivity [27], but is relatively expensive. The other available commercial kit is AmpliSens® HBV-genotype-FRT PCR kit (Federal Budget Institute of Science “Central Research Institute for Epidemiology”, Moscow, Russia), based on real-time PCR with specific hybridization probes. This kit used on qualitative detection and differentiation of hepatitis B virus (HBV) genotypes A, B, C, and D.
We compared a commercial kit AmpliSens HBV-genotype-FRT, based on RT PCR, with an adapted PCR method (using primers from INNO-LiPA assay), combined with Sanger sequencing (Appendix A Table A1). By commercial kits AmpliSens HBV-genotype-FRT we determined genotype in 95 from 103 samples, which represents 92.2% of samples. This method for genotyping appears to be useful for the rapid genotyping of HBV, as is quick and easy for preparing. In addition, genotyping using the commercial kit AmpliSens HBV-genotype-FRT allows us to detect combined genotypes. But this method is limited only to genotypes A, B, C, and D. Furthermore, a disadvantage is that the single nucleotide polymorphisms (SNP) at the primer site can affect the sensitivity of method [19].
Using the PCR method adapted by us (using primers from INNO-LiPA assay) combined with Sanger sequencing, we successfully determined genotype in all 103 samples. This method appears to be very sensitive, and it allows for further increase of sensitivity with nested PCR. PCR with sequencing also opens many possibilities, as well as sequence comparison in various databases or the use of various online tools, phylogenetic analysis, and in the case of a sequence that is coding polymerase and S protein, we also have the possibility of detecting important resistant or escape mutations. This method is cheaper than commercial methods but takes more time and is technically demanding and requires expertise with processing data. Another disadvantage of sequencing using a single region of the HBV genome is the inability to determine combined genotypes [8]. We also tried to detect not only the genotype but the subgenotype, too. To obtain subgenotype, we compared our sequencing to the reference sequences using the bioinformatics tool Blast from the NCBI database. These results were compared with results obtained by Geno2Pheno[hbv] tools (https://hbv.geno2pheno.org/; accessed on 5 December 2021) and with results from phylogenetic analysis by the MEGA X software. Only 14 samples have a different subgenotype using one of the methods, but they have the same results according to at least two other methods. All the other samples have the same subgenotype according to different methods. Overall, on the subgenotype level, the results of NCBI annotation, Geno2Pheno[hbv] tools and phylogenetic analysis using the MEGA X software matched in 86.41% of the samples, and these methods showed 80.58% consistency with RT PCR. However, the fact that this study is limited by the short lengths of gene sequences must be taken into consideration.
In addition, the obtained sequences were used for the detection of clinically important resistant and escape mutations through online tools Geno2Pheno[hbv] (https://hbv.geno2pheno.org/; accessed on 5 December 2021) and the HBV-Resistance interpretation tool (http://www.hiv-grade.de/hbv_grade/deployed/grade; accessed on 5 December 2021). From all 103 samples, the important clinical mutation was detected in 27 samples, which represents 26.21%. The most prevalent was the HBsAg escape mutations A128V and P127T, followed by the HBsAg escape mutations D144 E and K122R and the compensatory mutation S202I. Other mutations occurred individually, such as P120T, S143L, and G145R, while mutation M133I was together with A128V and P142L was together with D144A (Appendix A Table A2). The HBsAg vaccine escape mutation A128V was associated with occult HBV infection [28]. This is the most typical mutation for genotype D, but we found this mutation in genotype A, too. Of interest was the fact that two samples that had resistant mutation S202I also had the A128V escape mutation. Compensatory mutation S202I, which can cause resistance to Entecavir and Baraclude®, was among the most described mutations in the RT region [29,30]. We detected this mutation only in genotype D and only together with the A128V escape mutation. A mutation which affects the 144 or 145 amino acid position can be responsible for vaccine escape and failure of immunoglobulin (IG) therapy and detection [31,32]. Liver transplant patients infected with these escape mutations were described as having a worse clinical outcome compared to other patients [33]. Mutation P120T is also responsible for vaccine, therapy (IG) and detection failure [32,34]. The S143L immune escape mutation was also previously described in genotype D [35,36]. There is not sufficient data available on the K122R and P127T mutations to interpret these mutations, but they were also detected in other studies [29,37,38,39], though mutation K122R was detected only in genotype B. Mutation P127T could not be detected by the Geno2Pheno[hbv] tools as HBsAg escape; this variant was detected only by the HBV-resistance interpretation tool.
To our knowledge, this is the first published study based on data from HBV genotyping in the Slovak Republic. We compared two diagnostic methods for the detection of Hepatitis B Virus genotypes in the Slovak Republic, commercial kit AmpliSens HBV-genotype-FRT versus an adapted PCR method combine with Sanger sequencing. Both methods have advantages and disadvantages: Genotyping by commercial kit is quicker, and able to detect combined genotypes, while genotyping by PCR with Sanger sequencing appears to be a more informative and sensitive. The optimal approach could be first using the commercial kit for faster routine diagnostics and secondly, in case of ambiguous or unspecified results verify these results by sequencing and phylogenetic analysis. In addition, sequencing can be useful for following some clinically important mutations and prediction of response to treatment. We found that the most common genotype was D (49.5%), followed by genotype A (39.8%), genotype B with 1.9%, and genotypes C and E with 1%. We also detected a prevalence of the genotype combination A/D, represented by 3.9%, followed by the combination A/C and B/D, with 1.9% and 1% prevalence respectively (Figure 4).
Obtaining prevalence of HBV in Slovak Republic gives us an opportunity to compare our data with data from neighboring states. In the Czech Republic, for example, a prevalence of genotype A (n = 33; 73% and 67.1%) over D (n = 12; 27% and 28.4%) was described [40,41], and genotypes B and C (3.4% and 1.1%, respectively) were also found [41]. In Poland, the most common genotype determined was genotype A with 67%, followed by genotype D (20%), and genotype H (5%) and mixed A/D (5%). In addition, genotype F, combined genotypes D/G, A/C, and D/F, individually, were found in Poland [42]. In Ukraine, genotype D (52.4%), followed by A (14.2%) and C (4.7%), was the most prevalent [43]. In a report of the prevalence of HBV genotypes in Central and Eastern Europe as a whole, the prevalence of genotype D was 48% and genotype A was 42%, and only a few cases of genotype B, C, E, and F were detected [44]. According to that study, genotype A predominated in Poland (77%) and the Czech Republic (67%), as compared to Hungary (47%), Lithuania (41%), Croatia (8%) and Germany (32%), and genotype D was the most common for Lithuania (54%), Germany (58%), Romania (67%), Croatia (80%), and Russia (93%). About 8% of this European cohort’s patient had a mixed genotype mutation. Most of them were in Romania, where 27% of the samples proved to have more than one genotype and 82% of the combination genotypes took the form of the A/D genotype combination. Our results are comparable to the prevalence of HBV in neighboring countries.

4. Materials and Methods

4.1. Population Study

Between June 2019–October 2021, a total 103 serum samples were collected from HBsAg positive patients. To isolate serum from patients, a sample of approximately 5–7 mL of whole blood was collected into EDTA vacutainers. Serum was stored at −80 °C until testing. All patients were informed and provided written consent prior to examination. This study was approved by Ethics Committee of the L. Pasteur University Teaching Hospital, No. 2019/EK/4022.

4.2. DNA Isolation

HBV DNA was isolated from 400 μL of serum using the QIAamp® DNA Mini kit (QIAGEN GmbH, Hilden, Germany) in accordance with the manufacturer’s protocol and dissolved in 40 μL of elution buffer. Strict precaution was taken to prevent contamination. Subsequently, the DNA thus isolated was used for amplification and, if necessary, stored at −20 °C (for a short time) or at −80 °C (for a long time) for further use.

4.3. HBV Genotyping by Real-Time PCR

HBV genotyping was done using 10 μL of extracted DNA and the commercial kit AmpliSens® HBV-genotype-FRT (AmpliSens, Federal Budget Institute of Science “Central Research Institute for Epidemiology”, Moscow, Russia). The AmpliSens® HBV-genotype-FRT PCR kit is a nucleic acid amplification test for qualitative detection and differentiation of HBV genotypes A, B, C, and D. Amplification was performed on a LightCycler ® 480 Real-Time PCR System (ROCHE Diagnostics, Mannheim, Germany).

4.4. HBV Amplification by PCR (Direct or Nested)

For PCR amplification, we adapted specific genotyping primers for the surface genomic region from INNO-LiPA HBV genotyping assay (Fujirebio US, Inc., Malvern, PA, USA), [24]. PCR amplification was performed using the primers: HBPr134 (5′-TGCTGCTATGCCTCATCTTC-3′) and HBPr135 (5′-CARAGACARAA-GAAAATTGG-3′) for direct PCR, HBPr75 (5′-CAAGGTATGTTGCCCGTTTGTCC-3′) and HBPr94 (5′-GGYAWAAAGGGACTCAMGATG-3′) for nested PCR, and 5 x HOT FIREPol® Blend Master Mix Ready to Load (Solis BioDyne, Tartu, Estonia) and 5 μL of DNA samples were also used for direct and 2 μL (products of the direct round PCR) for nested PCR. Amplification was performed in a standard thermocycler (T1Thermocycler, Biometra GmbH, Göttingen, Germany), and the thermal cycling parameters used were: initial denaturation −95 °C for 12 min and 30 cycles of 95 °C for 20 s, 52 °C for 40 s, 72 °C for 60 s, and a final elongation of 72 °C for 10 min. The thermal cycling parameters were the same for both the direct and nested PCR. PCR products were visualized using 1% agarose gel electrophoresis. Nested PCR was performed only if direct PCR was not sufficient. Generally, the first round of PCR was sufficient, and the nested (second round) PCR was needed only five time. We obtained 409 bp products from direct and 341 bp from nested PCR.

4.5. Determination of HBV Genotype and Phylogenetic Analysis

Amplicons were sent for Sanger sequencing (Microsynth AG, Wien, Austria). The obtained chromatograms were analyzed and edited using the MEGA X software [25]. If the sequences had a short length or poor sequence quality, those samples were sent for Sanger sequencing repeatedly. All sequences were assembled in GeneTool Lite 1.0 software (BioTools Inc., Edmonton, AB, Canada). The sequences were compared to the reference sequences using the bioinformatics tool Blast from the U.S. National Centre for Biotechnology Information (NCBI, Bethesda, MD, USA) (http://www.ncbi.nlm.nih.gov/; accessed on 5 December 2021). In addition, these sequences are deposited in the NCBI GenBank (Accession numbers: MZ130378-MZ130381, MZ130383-MZ130385, MZ148125-MZ148129, MZ148131-MZ148136, MZ166567-MZ166571, MZ230740-MZ230777, OL702789-OL702830). The samples were genotyped by annotation in the NCBI database to detect genotype and subgenotype, which we compared with results using the Geno2Pheno[hbv] tools from the Max-Planck Institute for Informatics (https://hbv.geno2pheno.org/; accessed on 5 December 2021). As our sequencing regions overlapped the surface and polymerase genes, this also allowed us to check our sequences for the presence of resistance and escape mutations. For this, we used the same Geno2Pheno[hbv] tools and compared the results with the HBV-Resistance interpretation tool algorithm available online and based on the Stanford HIValg Software (http://www.hiv-grade.de/hbv_grade/deployed/grade; accessed on 5 December 2021). The genotyping results were also confirmed by phylogenetic analysis using the Mega X software [45]. A phylogenetic tree was constructed using a Maximum likelihood tree with Kimura-2-parameter substitution methods, and bootstrap values were calculated from 1000 replicates. The gene bank reference sequences of the major genotypes and subgenotypes that were included in phylogenetic analysis are: KP322600, FJ904443, JF754594, EU594406, GQ477457, MN310710, EU594435, KY810018, KM524153, HE974373, KT749850, KT749832, GQ477469, MF674438, MF674499, KP341009, AP011085, GQ924658, GQ377617, KF849718, HM363594.

Author Contributions

Conceptualization, M.L., P.K., P.D.L., I.H., S.D., P.J., M.J. and M.H.; methodology, M.L., A.S., V.B., E.H. and M.H.; validation, P.K., P.D.L., I.H., S.D., P.J. and M.J.; investigation, M.L., A.S., V.B., E.H. and M.H.; data curation, M.L., P.K, A.S., P.D.L. and Š.P.; writing—original draft preparation, M.L., P.K., A.S. and M.H.; writing—review and editing, M.L., P.K, A.S., P.J. and M.H.; visualization, Š.P.; supervision, P.K., P.J. and M.H.; project administration, P.K.; funding acquisition, P.K. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by a grant from the Slovak Research and Development Agency of the Ministry of Education, Science, Research and Sport of the Slovak Republic, project ID: APVV-18-0171.

Institutional Review Board Statement

The study was conducted in accordance with the guidelines of the Declaration of Helsinki and approved by the Ethics Committee of the L. Pasteur University Teaching Hospital (protocol code 2019/EK/4022 from 3 May 2019).

Informed Consent Statement

Informed consent was obtained from all subjects involved in the study.

Data Availability Statement

The data presented in this study are available on request from the corresponding author.

Conflicts of Interest

The authors declare that they have no competing interest.

Appendix A

Table A1. HBV genotyping, comparison of different methods.
Table A1. HBV genotyping, comparison of different methods.
SampleAccession NumbersReal-Time PCRNCBI
Annotation
Genotype by hbv.geno2phenoMEGA X Software
Phylogenetic Tree
1001MZ130378DD1D1D1
1002MZ130379DD1D1D1
1003MZ130380BB4B4B4
1004MZ130381B/DD1D1D1
1005MZ130383AA2A2A2
1006MZ130384DD3D1D3
1007MZ130385AA2A2A2
1008MZ148125DD3D3D3
1009MZ148126DD1D1D1
1010MZ148127A/CA2A2A2
1011MZ148128AA2A2A2
1012MZ148129DD3D3D3
1014MZ148131DD2D2D2
1015OL702789DD1D1D1
1016MZ148132-EEE
1017MZ148133DD3D3D3
1018MZ148134AA2A2A2
1019MZ148135AA2A2A2
1020MZ148136AA2A2A2
1021MZ166567DD3D3D3
1022MZ166568AA2A2A2
1023MZ166569DD1D4D4
1024MZ166570DD1D1D1
1025MZ166571AA2A2A2
1026MZ230740DD3D3D3
1027MZ230741A/DD2D2D2
1028MZ230742DD3 D3D3
1029MZ230743DD3D1D3
1030MZ230744DD2D2D2
1031MZ230745AA2A2A2
1032MZ230746AA2A2A2
1033MZ230747-D3D3D3
1034MZ230748DD3D3D3
1035MZ230749DD1D1D1
1036MZ230750AA2A2A2
1037MZ230751BB4B3B4
1039MZ230752DD3D1D3
1040MZ230753DD1D1D1
1041MZ230754A/DA2A2A2
1042MZ230755AA2A2A2
1043MZ230756DD3D3D3
1044MZ230757DD2D2D2
1045MZ230758A/DD1D1D1
1046MZ230759DD3D3D3
1047MZ230760DD3D3D3
1048OL702827-A2A2A2
1049MZ230761A A2A2A2
1050MZ230762DD2D2D2
1052MZ230763AA2A2A2
1053MZ230764DD3D3D3
1054MZ230765AA2A2A2
1055MZ230766DD3D3D3
1056MZ230767AA2A2A2
1057MZ230768AA2A2A2
1058MZ230769AA2A2A2
1059MZ230770AA2A2A2
1060MZ230771DD3D3D3
1061MZ230772AA2A2A2
1063MZ230773DD1D1D1
1064MZ230774DD3D1D3
1065MZ230775CC1C2C1
1066MZ230776A/DA2A2A2
1067MZ230777-D3D3D3
1068OL702790DD1D1D1
1070OL702791AA2A2A2
1071OL702792AA2A2A2
1072OL702793AA2A2A2
1073OL702794DD3D3D3
1074OL702795C/AA2A2A2
1075OL702796AA2A2A2
1076OL702797AA2A2A2
1077OL702798AA2A2A2
1078OL702799AA2A2A2
1079OL702800AA2A2A2
1080OL702828DD1D2D2
1081OL702801DD2D2D2
1082OL702802DD3D3D3
1083OL702803DD3D3D3
1084OL702804-D1D4D1
1085OL702805DD1D1D3
1086OL702806AA2A2A2
1087OL702807AA2A2A2
1088OL702808-D3D3D3
1089OL702809DD3D3D3
1090OL702810-A2A2A2
1091OL702811DD3D3D3
1092OL702812AA2A2A2
1093OL702813AA2A2A2
1094OL702814AA2A2A2
1095OL702815DD1D1D3
1096OL702816DD3D3D3
1097OL702817AA2A2A2
1098OL702818DD2D2D3
1099OL702819DD3D3D3
1100OL702820AA2A2A2
1101OL702821AA2A2A2
1103OL702822DD1D1D1
1104OL702829DD3D3D3
1105OL702830DD3D1D3
2001OL702823DD3D3D3
2002OL702824AA2A2A2
2003OL702825AA2A2A2
2005OL702826AA2A2A2
Table A2. Detection of resistance and escape mutations.
Table A2. Detection of resistance and escape mutations.
SampleMutations RT DomainDrug Resistance
Mutation
Mutations SHB ProteinEscape Mutations
SHB Protein
1001G127R, M129L, N131D, Y135S, Q215S T127P, T189I, S207R
1002Y135S T127P
1003N124H, N134D, K149Q, V207M K122R, M198I, F200YK122R
1004G127R, Y135S T127P
1005V207I, L217R M198I, W199L, L209V
1006Q130P, Y135S T127P
1007W153GRW, L217R, S219A L209V, D144DE, S210RD144E-Vaccine,
Therapy (IG), Detection
1008F122L, Q130P, Y135S, F221Y T127P, L213I
1009Y135S, S202IS202I-compensatory
mutation, possible
resistance to
Entecavir, Baraclude®
T127P, A128AV, T189I, V194FILVA128V-Vaccine
1010L217R, S219A L209V, S210R
1011L217R L209V
1012F122L, T128INST, Q130P, Y135S P120PS, T127P, S207NP120P-Vaccine
P120S-Vaccine, Detection
1014F122V, H126R, Q149K T118A, P127T P127T
1015G127GR, M129L, Y135S, Q149KQ T127P
1016 T189I
1017F122L, Q130P, Y135S, Q215H T127P, S207T
1018S159T, L217R L209V, L216
1019W153R, L217R S207N, L209V
1020R120G, N124H, L217R L209V
1021F122L, Q130P, Y135S T127P, V177A, Y206C
1022 A128AV, S207N, V209LA128V-Vaccine
1023H126R, M129L, Y135S, Q149K, Q215P T118A, T127P, P142L, D144A, S204N, S207RP142L, D144A-Vaccine,
Therapy (IG), Detection
1024Y135S T127P
1025L217R P188L, L209V, P211L
1026F122L, Q130P, Y135S, Q215H T127P, S207T
1027H126R T118A, P127T P127T
1028F122L, H124N, Q130P, Y135S, V190M, Q215H, S219A T127P, S204N, S207T, I208T, S210R
1029F122L, Y135S T127P
1030H126R T118A, P127T P127T
1031L217R, S219A S207N, L209V, S210R
1032L217R, S219A L209V, S210R
1033N118T, F122L, Q130P, Y135S, V191L, Q215S I110L, T127P, G159A, W182C, V190A, Y206S, S207R, P214L
1034F122L, Q130P, Y135S T127P
1035Y135S T127P, A128AV A128V-Vaccine
1036V163I, S213T, S219A A159G, Y161F, A194V, S204K, S210R, P214L
1037N124H, N134D, L220I K122R, V168A, F200Y
1039F122L, Y135S, S202IS202I-compensatory
mutation, possible
resistance to
Entecavir, Baraclude®
T127P, A128AV, V194FVA128V-Vaccine
1040Y135S T127P
1041V112A, K212T S204R, V209L
1042S213T, L217R Y161F, S204R, Y206S, L209V
1043F122L, Q130P, Y135S, L164M T127P, S143L, S204N, S207NS143L-Vaccine, Detection
1044H126R T118A, P127T P127T
1045Y135S T127P, T189I
1046F122L, Q130P, Y135S T127P
1047N118T, F122L, Q130P, Y135S, V191L, Q215S I110L, T127P, G159A, W182C, V190A, Y206S, S207R, P214L
1048L217R L209V
1049L217R, S219A L209V, S210R
1050H126R T118A, P127TP127T
1052L217R, S219A L209V, S210R
1053F122L, Q130P, Y135S, R153QR T127P, G145GRG145R-Vaccine, Therapy (IG),
Detection
1054S159T A194V, S207N, V209L
1055S117T, F122L, Q130P, Y135S, I187L, K212N, Q215P T127P, S204T, Y206C, S207R
1056R138K, V142I A128AV, G130S, M133I, V209L A128V-Vaccine,
M133I-Therapy (IG), Detection
1057L217R, S219A L209V, S210R
1058I187L, V190M, L217R, S219A L209V, S210R
1059L217R L209V
1060F122L, Q130P, Y135S T127P
1061S159T, L217R A128AV, L209VA128V-Vaccine
1063M129L, Y135S T127P
1064F122L, T128N, Y135S, Q215P P120T, T127P, S207RP120T-Vaccine,
Therapy (IG), Detection
1065M129L, K149Q D144E, V159A, A177V, S210N, I213LD144E-Vaccine,
Therapy (IG), Detection
1066S159T, L217R, S219A L209V, S210R
1067F122L, Q130P, Y135S T127P
1068G127R, M129L, N131D, Y135S, Q215S T127P, T189I, S207R
1070R110G, L217R Q101R, V184A, L209V
1071W153R, L217R S204N, S207NS, L209V
1072S106AT, V163I, L199V, V207I, L217R S193L, A194V, M198I, W199L, L209V
1073N118T, F122L, Q130P, Y135S, V191L, Q215S I110L, T127P, G159A, W182C, V190A, Y206S, S207R, P214L
1074M129IM, W153QR, V191IV, K212T, L217R, L220I C121CY, G145GR, W182 *W, S204R, L209VG145R-Vaccine, Therapy (IG),
Detection; C121Y-Detection
1075W153R, K212R, L217R, S219A S204D, L209V, S210K
1076W153R, L217R, S219A L209V, S210R
1077K212T, L217R S204R, L209V
1078Y151FY, W153R, L179FL, L217R T143ST, L209V
1079L217R, S219A L209V, S210R
1080A200AV P127T, L192FLP127T
1081H126R T118A, P127T P127T
1082F122L, Q130P, Y135S T127P, V177AV
1083F122L, Q130P, Y135S, Q215H T127P, S207T
1084G127R, M129L, Y135S, Q149K, I187L, V190M T127P, S207N
1085M129L, Y135S, V173M, Q215H, L217R, S219A, F221YV173MT127P, S207T, L209V, S210R, L213I
1086L217R A194V, Y200FY, S204NS, L209V
1087I121N, L217R S113T, A194V, S207N, L209V
1088V103IV, F122L, Q130PQ, Y135S T127P
1089R110GR, F122L, Q130P, Y135S T127P
1090L217R, S219A L94LS, F158FS, S193L, P203L, S204N, L205P, Y206F, S207N, L209V, S210R
1091F122L, Q130P, Y135S T127P
1092L217R A194V, S204N, L209V
1093W153R, L217R, S219A, L220I S204N, S207N, L209V, S210K
1094V207IMV, L217LR, S219AS M198IM, W199LW, L209LV, S210RS
1095Y135S T127P, G159A, S207N
1096F122L, H124N, Q130PS, Y135S, V190M, Q215H, S219A T127P, S204N, S207T, I208T, S210R
1097R110G, W153R, S159T, K212R, L217R, S219A, Y221F S204D, S207N, L209V, S210R, I213L
1098H126R, L132LM, Y135HY, S213ST T118A, P127T, Y200FY, S204RS, S207N, P211HPP127T
1099F122L, Q130P, Q215S P127T, S136Y, S207RP127T
1100L217R, S219A V96G, A194V, S204N, Y206C, L209V, S210K
1101L217R, S219A Y161F, S204NS, L209V, S210R
1103F122V, M129L, Y135S T127P
1104F122L, Q130P, Y135S, F221Y T127P, L213I
1105F122L, Y135S, L199V T127P, V184A, Y200C, Y206S
2001F122L, Q130P, Y135S T127P, V184A, I208T
2002R138K, S219A L98Q, A128V, G130N, Y161F, V209L, S210RA128V-Vaccine
2003S159T, L217R L209V
2005W153R, L157M, S219A S204N, S207N, V209L, S210R

References

  1. Hepatitis B and C in the EU Neighbourhood: Prevalence, Burden of Disease and Screening Policies. Available online: https://www.ecdc.europa.eu/en/publications-data/hepatitis-b-and-c-eu-neighbourhood-prevalence-burden-disease-and-screening (accessed on 7 December 2021).
  2. Drazilova, S.; Janicko, M.; Kristian, P.; Schreter, I.; Halanova, M.; Urbancikova, I.; Madarasova-Geckova, A.; Marekova, M.; Pella, D.; Jarcuska, P.; et al. Prevalence and Risk Factors for Hepatitis B Virus Infection in Roma and Non-Roma People in Slovakia. Int. J. Environ. Res. Public Health 2018, 15, 1047. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Veselíny, E.; Janičko, M.; Dražilová, S.; Siegfried, L.; Pastvová, L.; Schréter, I.; Kristian, P.; Virág, L.; Jarčuška, P.; Valková, I.; et al. High Hepatitis B and Low Hepatitis C Prevalence in Roma Population in Eastern Slovakia. Cent. Eur. J. Public Health 2014, 22, S51–S56. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Hepatitis B—Annual Epidemiological Report 2019. Available online: https://www.ecdc.europa.eu/en/publications-data/hepatitis-b-annual-epidemiological-report-2019 (accessed on 7 December 2021).
  5. Public Health Authority of the Slovak Republic (PHA SR). Annual Report of Regional Public Health Authorities in Slovakia for the Year 2020; Public Health Authority of the Slovak Republic (PHA SR): Bratislava, Slovakia, 2021. (In Slovak) [Google Scholar]
  6. Summers, J.; Smolec, J.M.; Snyder, R. A Virus Similar to Human Hepatitis B Virus Associated with Hepatitis and Hepatoma in Woodchucks. Proc. Natl. Acad. Sci. USA 1978, 75, 4533–4537. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Kramvis, A. Genotypes and Genetic Variability of Hepatitis B Virus. Intervirology 2014, 57, 141–150. [Google Scholar] [CrossRef]
  8. Kramvis, A. The Clinical Implications of Hepatitis B Virus Genotypes and HBeAg in Pediatrics. Rev. Med. Virol. 2016, 26, 285–303. [Google Scholar] [CrossRef] [Green Version]
  9. Sunbul, M. Hepatitis B Virus Genotypes: Global Distribution and Clinical Importance. World J. Gastroenterol. 2014, 20, 5427–5434. [Google Scholar] [CrossRef]
  10. Tong, S.; Revill, P. Overview of Hepatitis B Viral Replication and Genetic Variability. J. Hepatol. 2016, 64, S4–S16. [Google Scholar] [CrossRef] [Green Version]
  11. Andernach, I.E.; Hunewald, O.E.; Muller, C.P. Bayesian Inference of the Evolution of HBV/E. PLoS ONE 2013, 8, 81690. [Google Scholar] [CrossRef] [Green Version]
  12. Littlejohn, M.; Locarnini, S.; Yuen, L. Origins and Evolution of Hepatitis B Virus and Hepatitis D Virus. Cold Spring Harb. Perspect. Med. 2016, 6, a021360. [Google Scholar] [CrossRef]
  13. Paraskevis, D.; Magiorkinis, G.; Magiorkinis, E.; Ho, S.Y.W.; Belshaw, R.; Allain, J.P.; Hatzakis, A. Dating the Origin and Dispersal of Hepatitis B Virus Infection in Humans and Primates. Hepatology 2013, 57, 908–916. [Google Scholar] [CrossRef]
  14. Bartholomeusz, A.; Schaefer, S. Hepatitis B Virus Genotypes: Comparison of Genotyping Methods. Rev. Med. Virol. 2004, 14, 3–16. [Google Scholar] [CrossRef] [PubMed]
  15. Rajoriya, N.; Combet, C.; Zoulim, F.; Janssen, H.L.A. How Viral Genetic Variants and Genotypes Influence Disease and Treatment Outcome of Chronic Hepatitis, B. Time for an Individualised Approach? J. Hepatol. 2017, 67, 1281–1297. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Guettouche, T.; Hnatyszyn, J. Chronic Hepatitis B and Viral Genotype: The Clinical Significance of Determining HBV Genotypes. Antivir. Ther. 2005, 10, 593–604. [Google Scholar] [PubMed]
  17. McNaughton, A.L.; D’Arienzo, V.; Ansari, M.A.; Lumley, S.F.; Littlejohn, M.; Revill, P.; McKeating, J.A.; Matthews, P.C. Insights from Deep Sequencing of the HBV Genome—Unique, Tiny, and Misunderstood. Gastroenterology 2019, 156, 384–399. [Google Scholar] [CrossRef] [Green Version]
  18. Lin, C.-L.; Kao, J.-H.; Chen, B.-F.; Chen, P.-J.; Lai, M.-Y.; Chen, D.-S. Application of Hepatitis B Virus Genotyping and Phylogenetic Analysis in Intrafamilial Transmission of Hepatitis B Virus. Clin. Infect. Dis. 2005, 41, 1576–1581. [Google Scholar] [CrossRef]
  19. Guirgis, B.S.S.; Abbas, R.O.; Azzazy, H.M.E. Hepatitis B Virus Genotyping: Current Methods and Clinical Implications. Int. J. Infect. Dis. 2010, 14, e941–e953. [Google Scholar] [CrossRef] [Green Version]
  20. Kramvis, A.; Arakawa, K.; Yu, M.C.; Nogueira, R.; Stram, D.O.; Kew, M.C. Relationship of Serological Subtype, Basic Core Promoter and Precore Mutations to Genotypes/Subgenotypes of Hepatitis B Virus. J. Med. Virol. 2008, 80, 27–46. [Google Scholar] [CrossRef]
  21. Mizokami, M.; Nakano, T.; Orito, E.; Tanaka, Y.; Sakugawa, H.; Mukaide, M.; Robertson, B.H. Hepatitis B Virus Genotype Assignment Using Restriction Fragment Length Polymorphism Patterns. FEBS Lett. 1999, 450, 66–71. [Google Scholar] [CrossRef] [Green Version]
  22. Kirschberg, O.; Schüttler, C.; Repp, R.; Schaefer, S. A Multiplex-PCR to Identify Hepatitis B Virus—Genotypes A–F. J. Clin. Virol. 2004, 29, 39–43. [Google Scholar] [CrossRef]
  23. Naito, H.; Hayashi, S.; Abe, K. Rapid and Specific Genotyping System for Hepatitis B Virus Corresponding to Six Major Genotypes by PCR Using Type-Specific Primers. J. Clin. Microbiol. 2001, 39, 362–364. [Google Scholar] [CrossRef] [Green Version]
  24. Teles, S.A.; Martins, R.M.B.; Silva, S.A.; Gomes, D.M.F.; Cardoso, D.D.P.; Vanderborght, B.O.M.; Yoshida, C.F.T. Hepatitis B Virus Infection Profile in Central Brazilian Hemodialysis Population. Rev. Inst. Med. Trop. São Paulo 1998, 40, 281–286. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Lau, K.C.; Osiowy, C.; Coffin, C.S. Hepatitis B Virus (HBV) Genome Detection and Genotyping in Virally Suppressed Patients Using Nested Polymerase Chain Reaction-Based Sanger Sequencing. Diagn. Microbiol. Infect. Dis. 2019, 93, 318–324. [Google Scholar] [CrossRef]
  26. Pourkarim, M.R.; Amini-Bavil-Olyaee, S.; Kurbanov, F.; van Ranst, M.; Tacke, F. Molecular Identification of Hepatitis B Virus Genotypes/Subgenotypes: Revised Classification Hurdles and Updated Resolutions. World J. Gastroenterol. 2014, 20, 7152–7168. [Google Scholar] [CrossRef]
  27. Osiowy, C.; Giles, E. Evaluation of the INNO-LiPA HBV Genotyping Assay for Determination of Hepatitis B Virus Genotype. J. Clin. Microbiol. 2003, 41, 5473–5477. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  28. Kumar, G.T.; Kazim, S.N.; Kumar, M.; Hissar, S.; Chauhan, R.; Basir, S.F.; Sarin, S.K. Hepatitis B Virus Genotypes and Hepatitis B Surface Antigen Mutations in Family Contacts of Hepatitis B Virus Infected Patients with Occult Hepatitis B Virus Infection. J. Gastroenterol. Hepatol. 2009, 24, 588–598. [Google Scholar] [CrossRef] [PubMed]
  29. Ababneh, N.A.; Sallam, M.; Kaddomi, D.; Attili, A.M.; Bsisu, I.; Khamees, N.; Khatib, A.; Mahafzah, A. Patterns of Hepatitis B Virus S Gene Escape Mutants and Reverse Transcriptase Mutations among Genotype D Isolates in Jordan. PeerJ 2019, 7, e6583. [Google Scholar] [CrossRef] [Green Version]
  30. Shaw, T.; Bartholomeusz, A.; Locarnini, S. HBV Drug Resistance: Mechanisms, Detection and Interpretation. J. Hepatol. 2006, 44, 593–606. [Google Scholar] [CrossRef]
  31. Westhoff, T.H.; Jochimsen, F.; Schmittel, A.; Stöffler-Meilicke, M.; Schäfer, J.H.; Zidek, W.; Gerlich, W.H.; Thiel, E. Fatal Hepatitis B Virus Reactivation by an Escape Mutant Following Rituximab Therapy. Blood 2003, 102, 1930. [Google Scholar] [CrossRef]
  32. Avellón, A.; Echevarria, J.M. Frequency of Hepatitis B Virus “a” Determinant Variants in Unselected Spanish Chronic Carriers. J. Med. Virol. 2006, 78, 24–36. [Google Scholar] [CrossRef]
  33. Protzer-Knolle, U.; Naumann, U.; Bartenschlager, R.; Berg, T.; Hopf, U.; zum Buschenfelde, K.H.M.; Neuhaus, P.; Gerken, G.; Locarnini, S.A. Hepatitis B Virus with Antigenically Altered Hepatitis B Surface Antigen Is Selected by High-Dose Hepatitis B Immune Globulin after Liver Transplantation. Hepatology 1998, 27, 254–263. [Google Scholar] [CrossRef]
  34. Hossain, M.G.; Ueda, K. A Meta-Analysis on Genetic Variability of RT/HBsAg Overlapping Region of Hepatitis B Virus (HBV) Isolates of Bangladesh. Infect. Agents Cancer 2019, 14, 33. [Google Scholar] [CrossRef] [Green Version]
  35. Salpini, R.; Colagrossi, L.; Bellocchi, M.C.; Surdo, M.; Becker, C.; Alteri, C.; Aragri, M.; Ricciardi, A.; Armenia, D.; Pollicita, M.; et al. Hepatitis B Surface Antigen Genetic Elements Critical for Immune Escape Correlate with Hepatitis B Virus Reactivation upon Immunosuppression. Hepatology 2015, 61, 823–833. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Sayiner, A.A.; Agca, H.; Sengonul, A.; Celik, A.; Akarsu, M. A New Hepatitis B Virus Vaccine Escape Mutation in a Renal Transplant Recipient. J. Clin. Virol. 2007, 38, 157–160. [Google Scholar] [CrossRef]
  37. Coppola, N.; Onorato, L.; Minichini, C.; di Caprio, G.; Starace, M.; Sagnelli, C.; Sagnelli, E. Clinical Significance of Hepatitis B Surface Antigen Mutants. World J. Hepatol. 2015, 7, 2729–2739. [Google Scholar] [CrossRef]
  38. Abu Zeid, W.M.; Ramadan, D.I.; Shemis, M.A. Prevalence of Mutations within Major Hydrophilic Region of Hepatitis B Virus and Their Correlation with Genotypes among Chronically Infected Patients in Egypt. Arab. J. Gastroenterol. 2016, 17, 34–40. [Google Scholar] [CrossRef] [PubMed]
  39. Capai, L.; Falchi, A.; Charrel, R. Meta-Analysis of Human IgG Anti-HEV Seroprevalence in Industrialized Countries and a Review of Literature. Viruses 2019, 11, 84. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  40. Krekulova, L.; Rehak, V.; da Silva Filho, H.P.; Zavoral, M.; Riley, L.W. Genotypic Distribution of Hepatitis B Virus in the Czech Republic: A Possible Association with Modes of Transmission and Clinical Outcome. Eur. J. Gastroenterol. Hepatol. 2003, 15, 1183–1188. [Google Scholar] [CrossRef] [PubMed]
  41. Němeček, V.; Strunecký, O. Genotypy Viru Hepatitidy B (HBV) v České Republice. Epidemiol. Mikrobiol. Imunol. 2004, 53, 55–61. [Google Scholar] [PubMed]
  42. Świderska, M.; Pawłowska, M.; Mazur, W.; Tomasiewicz, K.; Simon, K.; Piekarska, A.; Wawrzynowicz-Syczewska, M.; Jaroszewicz, J.; Rajewski, P.; Zasik, E.; et al. Distribution of HBV Genotypes in Poland. Clin. Exp. Hepatol. 2015, 1, 1–4. [Google Scholar] [CrossRef] [Green Version]
  43. Yurchenko, T.S.; Stepchenkova, T.; Karnets, I.; Ashworth, K.; Cheusova, T. The Results of a Study on the Prevalence of HIV, HCV and HBV Genotypes in Some Regions of Ukraine. Retrovirology 2012, 9, P55. [Google Scholar] [CrossRef] [Green Version]
  44. Deterding, K.; Constantinescu, I.; Nedelcu, F.D.; Gervain, J.; Némeček, V.; Srtunecky, O.; Vince, A.; Grgurevic, I.; Bielawski, K.P.; Zalewska, M.; et al. Prevalence of HBV Genotypes in Central and Eastern Europe. J. Med. Virol. 2008, 80, 1707–1711. [Google Scholar] [CrossRef] [PubMed]
  45. Kumar, S.; Stecher, G.; Li, M.; Knyaz, C.; Tamura, K. MEGA X: Molecular Evolutionary Genetics Analysis across Computing Platforms. Mol. Biol. Evol. 2018, 35, 1547–1549. [Google Scholar] [CrossRef] [PubMed]
Figure 1. (a) Genotyping by RT PCR; (b) genotyping by NCBI annotation; (c) genotyping by the Geno2pheno[hbv] tool; (d) genotyping by the MEGA X software Phylogenetic tree.
Figure 1. (a) Genotyping by RT PCR; (b) genotyping by NCBI annotation; (c) genotyping by the Geno2pheno[hbv] tool; (d) genotyping by the MEGA X software Phylogenetic tree.
Pathogens 11 00020 g001
Figure 2. Phylogenetic analysis of the HBV S-gene region sequences. The 103 specimens were aligned with 21 representative sequences of 5 genotype (including the relevant subgenotypes) available from GenBank. Reference sequences were marked by filled square labels. The final length was 337 bp. The alignment was analyzed using the Maximum Likelihood method and Kimura 2-parameter model with 1000 bootstrap replicates in the MEGA X software. Branch nodes with bootstrap values >50 are included next to the corresponding node.
Figure 2. Phylogenetic analysis of the HBV S-gene region sequences. The 103 specimens were aligned with 21 representative sequences of 5 genotype (including the relevant subgenotypes) available from GenBank. Reference sequences were marked by filled square labels. The final length was 337 bp. The alignment was analyzed using the Maximum Likelihood method and Kimura 2-parameter model with 1000 bootstrap replicates in the MEGA X software. Branch nodes with bootstrap values >50 are included next to the corresponding node.
Pathogens 11 00020 g002
Figure 3. Occurrence of resistance and escape mutation divided per genotype (genotypes are marked by MEGA X phylogenetic analysis).
Figure 3. Occurrence of resistance and escape mutation divided per genotype (genotypes are marked by MEGA X phylogenetic analysis).
Pathogens 11 00020 g003
Figure 4. Prevalence of the HBV genotypes among patients in the Slovak Republic.
Figure 4. Prevalence of the HBV genotypes among patients in the Slovak Republic.
Pathogens 11 00020 g004
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Logoida, M.; Kristian, P.; Schreiberova, A.; Lenártová, P.D.; Bednárová, V.; Hatalová, E.; Hockicková, I.; Dražilová, S.; Jarčuška, P.; Janičko, M.; et al. Comparison of Two Diagnostic Methods for the Detection of Hepatitis B Virus Genotypes in the Slovak Republic. Pathogens 2022, 11, 20. https://doi.org/10.3390/pathogens11010020

AMA Style

Logoida M, Kristian P, Schreiberova A, Lenártová PD, Bednárová V, Hatalová E, Hockicková I, Dražilová S, Jarčuška P, Janičko M, et al. Comparison of Two Diagnostic Methods for the Detection of Hepatitis B Virus Genotypes in the Slovak Republic. Pathogens. 2022; 11(1):20. https://doi.org/10.3390/pathogens11010020

Chicago/Turabian Style

Logoida, Mariia, Pavol Kristian, Andrea Schreiberova, Patrícia Denisa Lenártová, Veronika Bednárová, Elena Hatalová, Ivana Hockicková, Sylvia Dražilová, Peter Jarčuška, Martin Janičko, and et al. 2022. "Comparison of Two Diagnostic Methods for the Detection of Hepatitis B Virus Genotypes in the Slovak Republic" Pathogens 11, no. 1: 20. https://doi.org/10.3390/pathogens11010020

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop